Helicobacter pylori is a Gram-negative bacterium that infects the gastric epithelia of its human host. Everyone who is colonized with these pathogenic bacteria can develop gastric inflammation, termed gastritis. Additionally, a small proportion of colonized people develop more adverse outcomes, including gastric ulcer disease, gastric adenocarcinoma, or gastric mucosa-associated lymphoid tissue lymphoma.
KEYWORDS: gastritis, Helicobacter pylori, IL-17, IL-21, inflammation, Th17 cells, carcinogenesis
ABSTRACT
Helicobacter pylori is a Gram-negative bacterium that infects the gastric epithelia of its human host. Everyone who is colonized with these pathogenic bacteria can develop gastric inflammation, termed gastritis. Additionally, a small proportion of colonized people develop more adverse outcomes, including gastric ulcer disease, gastric adenocarcinoma, or gastric mucosa-associated lymphoid tissue lymphoma. The development of these adverse outcomes is dependent on the establishment of a chronic inflammatory response. The development and control of this chronic inflammatory response are significantly impacted by CD4+ T helper cell activity. Noteworthy, T helper 17 (Th17) cells, a proinflammatory subset of CD4+ T cells, produce several proinflammatory cytokines that activate innate immune cell antimicrobial activity, drive a pathogenic immune response, regulate B cell responses, and participate in wound healing. Therefore, this review was written to take an intricate look at the involvement of Th17 cells and their affiliated cytokines (interleukin-17A [IL-17A], IL-17F, IL-21, IL-22, and IL-26) in regulating the immune response to H. pylori colonization and carcinogenesis.
INTRODUCTION
Helicobacter pylori is the predominant member of the gastric microbiota in a most of the world’s population (1–3). Depending on the region, an estimated 30 to 80% of the population is colonized with the Gram-negative bacterium. Remarkably, H. pylori colonization can have dichotomous impacts on the host immune response; the impact displayed will depend on the timing of colonization and the environment. H. pylori colonization can lead to protection from some proinflammatory diseases (4–10) or to detrimental outcomes, including gastritis, peptic ulcer disease, and gastric cancer (11, 12). H. pylori-infected individuals are all believed to develop some level of gastritis, but many colonized persons may be asymptomatic or not seek medical care (11).
Detrimental outcomes of H. pylori colonization range from symptomatic gastritis to gastric cancers, including gastric adenocarcinoma and gastric mucosa-associated lymphoid tissue (MALT) lymphoma (11, 12) (Fig. 1). Infection with H. pylori is the single most common risk factor for gastric cancer and, for this reason, was defined by the World Health Organization (WHO) as a class I carcinogen. The Cancer Statistics Center of the American Cancer Society estimated that in 2019 there will have been 27,510 new cases of gastric cancer in the United States with over 11,140 estimated deaths (13). Gastric cancer is the 3rd most common cause of cancer-related deaths in the world, accounting for upwards of 783,000 deaths in 2018, according to the WHO (14).
There is now evidence that H. pylori colonization protects against pathologies of the esophagus and gastric cardia (8, 15–17), childhood asthma (8, 9, 18), and childhood allergies (19, 20). Moreover, a recent review of the literature and a meta-analysis suggest that there is a protective effect of H. pylori infection on the incidence of inflammatory bowel disease (21, 22). While H. pylori has persistently colonized humans since the origin of the species, studies have found that the outcomes of H. pylori colonization depend on several factors, including, but not limited to, the presence of specific H. pylori virulence factors, diet, and/or host genetics (23–25). Specifically, CD4+ T cell responses, including expression of gamma interferon (IFN-γ) and interleukin-17 (IL-17) and regulatory T (Treg) cell development, impact the pathology elicited in response to H. pylori colonization. This review is designed to take an intricate look at the involvement of T helper 17 (Th17) cells and the Th17 cytokines in the immunopathogenesis of H. pylori infection.
INNATE RESPONSE: THE EARLY RESPONSE TO H. PYLORI
The development of an immune response to H. pylori infection has largely been investigated in mouse models of disease. In humans, since early infection is likely asymptomatic or possibly mistaken for a short-lived gastrointestinal infection, knowing when infection occurs in specific individuals is difficult. In some areas where H. pylori colonization is endemic, there is evidence that H. pylori colonization occurs early in childhood (26–30). The mouse model facilitates tractable immunological studies and the use of key technologies to investigate cellular infiltration (and gastritis) in the mouse model.
The course of infection and the development of pathology have been traced using serial evaluations in mice. Flow cytometry was used to characterize the early inflammatory response to H. pylori. In the first 2 to 3 days postinfection, there is an influx of neutrophils and macrophages (31). Then, by 4 days postinfection, the number of macrophages and neutrophils drops significantly. H. pylori induces macrophage apoptosis by the generation of polyamines from ornithine decarboxylase (32, 33), but it is not understood why neutrophil numbers drop so significantly. Subsequently, the H. pylori infection seems to be somewhat quiescent (in terms of gastric immune cell infiltration) for a few more weeks. During this early response, when neutrophil infiltration is reduced, differences in acute inflammation are observed depending on the H. pylori strain’s virulence factor expression. Most notably, the cag type IV secretion system (T4SS) has been shown to modulate the immune response. Strains which harbor a functional cag type IV secretion system activate gastric epithelial cells to produce IL-8 (or IL-8 homologs), a chemokine which recruits neutrophils, and therefore, the cag T4SS can directly induce inflammation through this pathway. Priming of the adaptive response is likely happening during this quiescent stage; the earliest adaptive response is detectable by 2 weeks postinfection, when H. pylori-specific T cells are generated. T cell priming in response to H. pylori infection was demonstrated using paragastric lymph node cells in an antigen-specific enzyme-linked immunosorbent spot assay (31). Unfortunately, the paragastric lymph nodes are difficult to locate in some strains of mice, and while one might expect the cell numbers to expand and lead to the enlargement of local nodes, this does not seem to be the case. Peyer’s patches have become a feasible location to identify H. pylori-specific T cells (34). Identifying sites of T cell priming and activation during H. pylori infection, even in the mouse model, would facilitate more detailed studies about cell differentiation, polarization, migration, and even T cell exhaustion. Nevertheless, it is clear from these studies and others that the gastritis response to H. pylori leads to the activation of dendritic cells (DCs); recruitment of CD4+ T cells, including Th1, Th17, and Treg cells; and recruitment and activation of neutrophils and macrophages in the tissue (Fig. 2).
CYTOKINE DRIVERS OF THE T CELL RESPONSE
From in vitro assays using macrophages, monocytes, and dendritic cells (DCs), studies have indicated that these innate antigen-presenting cells play a significant role in priming and activation of the immune responses. Activation of these cells with live H. pylori or H. pylori lysates results in strong cytokine production, including the production of cytokines involved in the differentiation of Th1, Th17, and Treg cell responses. There is an upregulation of Th1-polarizing cytokines, including IL-12 (35); Treg cell-activating cytokines, including IL-2 and transforming growth factor β (TGF-β); and Th17-polarizing cytokines, including IL-1β, IL-23, and IL-6 (36, 37). For Th17 cells, IL-23 has been shown to be vital for the development of the Th17 response to H. pylori. Specifically, IL-23 plays a key role in the activation of IL-17A and IL-17F expression (38). IL-23 is a heterodimer of IL-23(p19) and IL-12(p40) and signals through the IL-23 receptor (IL-23R)/IL-12Rβ1 (having a common p40 subunit and a common shared receptor subunit with IL-12). IL-23 expression has been reported in the gastric mucosa of uninfected persons, IL-23 expression increases in those infected with H. pylori (39). The role of IL-23 was addressed in a chronic H. pylori infection model. In that study, the level of H. pylori colonization in IL-23(p19)−/− mice was significantly higher than that in wild-type (WT) mice. Moreover, the expression of IL-17A was reduced in IL-23(p19)−/− mice, and this reduction correlated with reduced inflammation (36, 40). In relationship to Th1 and Th17 cell development and H. pylori, it has been demonstrated that IL-21, which is required for the expansion and stabilization of Th17 responses, is also important for the maintenance of both Th1 and Th17 responses in the mouse model of H. pylori-induced gastritis (41).
While there is evidence that macrophages and dendritic cells (DCs) respond to H. pylori or H. pylori lysates by the production of proinflammatory cytokines, other studies provide evidence that DCs may play a vital role in Treg cell development. In vivo, H. pylori-DC interactions skew the T cell response toward a Treg cell response rather than a Th17 response (42). In vitro, some bone marrow-derived dendritic cell (BMDC) experiments have demonstrated that H. pylori not only survives within DCs but also alters the DC function by inhibiting antigen presentation, altering cytokine production, and therefore ultimately affecting T cell activation (43). The local cytokine environment may significantly impact the cytokine profile of H. pylori-infected DCs. Moreover, there is a single study which indicates that the phosphorylation of H. pylori’s cytotoxin A (CagA) within DCs can lead to the downregulation of DC cytokine production (44).
HELICOBACTER PYLORI ANTIGENS INVOLVED IN T CELL ACTIVATION AND DIFFERENTIATION
There is evidence in the literature that specific H. pylori proteins, including several virulence factors, influence the T cell response. The influence of these antigens on the T cell response has been investigated from two different perspectives. In some studies, the activation of innate antigen-presenting cells has been measured with different H. pylori proteins, and in other studies, the T cell response to specific antigens has been measured. Only a few studies have simultaneously examined both the activation of antigen-presenting cells and antigen-specific T cell activation. For example, the H. pylori protein urease has been carefully examined in coculture with macrophages. Recombinant urease B (rUreB) induces the expression of IL-6 and IL-23(p19) (which drive Th17 differentiation) in cultured macrophages (45). Moreover, in the same report, the authors provide evidence that when splenic lymphocytes from H. pylori-infected mice are cocultured with rUreB, there was an increase in the number of Th17 cells as well as an increase in the levels of IL-17A. Finally, when using rUreB as an immunization antigen, it elicited increased UreB-specific Th17 cells. Combined, these data indicate that UreB is an important protein which is able to elicit Th17 cell responses both in vivo and in vitro (45). Interestingly, when CD4+ T cell epitopes of UreB were combined with CD4+ T cell epitopes of H. pylori adhesion A (HpaA) and CagA to create the epitope-based vaccine Epivac, immunizations elicited a Th1 cell-biased immune response which inhibited H. pylori colonization. Unfortunately, this 2012 study did not measure Th17 responses to the vaccination (46).
There is also evidence that CagA influences T cell activation or T cell skewing. Using CagA isogenic mutants, CagA was shown to influence phytohemagglutinin-induced T cell proliferation (47). In human H. pylori-infected patients, the polarization of the T helper cell response was influenced by both the CagA status of the infecting strain and the stage of progression to gastric carcinogenesis. Th1 or Th2 polarization was not skewed in patients infected with CagA-negative H. pylori strains. On the other hand, patients infected with CagA-positive (CagA+) strains had Th1-mediated cellular immunity in earlier stages of gastric carcinogenesis, while the advanced stages were dominated by Th2 cell-mediated humoral immunity. Further, these advanced stages and CagA+ strains were negatively associated with an abundance of Treg cells (48).
H. pylori affects costimulatory markers associated with T cell responses likely through CagA-dependent mechanisms. For example, H. pylori infection downregulates B7-H2 expression by gastric epithelial cells (GECs) in a CagA-dependent manner (49), and this downregulation is correlated with a decrease in Th17 cell responses in vitro and in a mouse model. B7-H1 is another member of the B7 family of proteins, but unlike B7-H2, which is proinflammatory, B7-H1 (better known as programmed death 1 ligand 1 [PD-L1]) is associated with T cell inhibition on GECs. It has been demonstrated that H. pylori coculture with GECs leads to increased PD-L1 expression; moreover, this induction of PD-L1 influences T cell activation and IL-2 production (50). Using H. pylori WT and isogenic mutant strains, Lina et al. showed that H. pylori requires CagA and peptidoglycan for B7-H1 upregulation in GECs (51). Taken together, these data indicate that a functional cag pathogenicity island (cagPAI) can impact the host response.
Another H. pylori virulence factor which influences T cell responses is H. pylori’s gamma-glutamyl transpeptidase (GGT). GGT converts glutamine into glutamate and ammonia and converts glutathione into glutamate and cysteinylglycine. H. pylori GGT induces immune tolerance through the inhibition of T cell-mediated immunity and dendritic cell differentiation (52). In many of these studies, it is difficult to separate the functions of VacA and GGT, for they seem to have similar effects on dendritic cell activity (toward T cell skewing) and on T cell proliferation.
IDENTIFICATION OF THE FUNDAMENTAL FUNCTIONS OF Th17 CELLS
Classically, proinflammatory activated T helper cells were functionally divided into two subsets, Th1 and Th2, based on the cytokines that they produce (reviewed in reference 19), with an understanding that there were also CD4+ T regulatory cells playing an immune-modulatory role in inflammation. The cytokines that Th1 and Th2 cells produce, IFN-γ and IL-4, respectively, are necessary for the control of specific types of pathogens based on their lifestyle (40). Th1 responses are key for the control of intracellular pathogens, while Th2 responses are vital for the control of extracellular pathogens, especially helminths (42). Th1 and Th2 responses can also contribute to immune pathogenesis. Th1 cells play an active role under inflammatory conditions, such as inflammatory bowel disease and arthritis, while Th2 cells can contribute to the inflammatory responses in allergy and asthma (43, 44).
Specifically, murine models deficient in IL-12(p35) and IFN-γ demonstrate that cytokine and receptor knockout mice still develop inflammatory conditions, such as experimental autoimmune encephalomyelitis (EAE), a mouse model of multiple sclerosis (53–55). This suggests that another subset of T cells, in addition to Th1 cells, may drive inflammation in this model. Further research demonstrated that IL-23(p19/p40) and another proinflammatory CD4+-derived cytokine, IL-17A, are associated with some of the same inflammatory processes as Th1 cell-associated cytokines, including EAE (35, 38, 56). Henceforth, this newly identified lineage of CD4+ T cells, involved in immune pathogenesis and in the production of IL-17A, became known as Th17 cells (57). Th17 cells are distinct from Th1 and Th2 cells on the basis of their unique differentiation and transcription factor expression (58). IL-12(p35/p40) and IL-4 have been shown to be the major differentiation factors for Th1 and Th2 cells, respectively. The differentiation of Th1 and Th2 cells is associated with induction of the transcription factors Tbet and GATA-3, respectively, whereas TGF-β, IL-6, and IL-23(p19/p40) are required for the differentiation of Th17 cells through the upregulation of RORγt (18, 59).
Subsequently, it has also been demonstrated that Th17 cells and, specifically, Th17 cytokines are critical for neutrophil recruitment as well as the production of various antimicrobial peptides, matrix metalloproteinases, chemokines, and cytokines, such as IL-6, IL-8, and granulocyte-macrophage colony-stimulating factor (GM-CSF), all of which are essential factors in the control of extracellular pathogens (40, 48, 60–64). Th17 production of IL-17A and IL-17F in murine models plays a similar role in neutrophil and cytokine recruitment. However, while they share a significant degree of sequence homology, studies have suggested that IL-17F may play a more central role in innate responses than IL-17A (48). Th17 also regulates the production of IL-21, which amplifies the Th17 response in an autocrine manner (65), and plays a critical role in the regulation of B cell function (66). A fourth cytokine produced by Th17 cells, IL-22, is important for mucosal immunity against pathogens which infect the intestines, and it enhances the expression of antimicrobial peptides on epithelial cells, especially in skin inflammation and infection (67). Unlike the other cytokines produced by Th17 cells, IL-22 receptor expression is not present on immune cells. Instead, it provides direct communication between the immune system and the tissues, since the receptor is solely present in tissues (reviewed in reference 68). One Th17 cytokine, IL-26, is produced only in humans. This proinflammatory cytokine has been associated with inflammation and may have direct antimicrobial activities. The role(s) of each of these cytokines in the context of controlling H. pylori colonization and immunopathology is discussed below.
ROLES OF Th17 CYTOKINES IN H. PYLORI-DRIVEN IMMUNOPATHOGENESIS
In this section, we summarize the data on the mechanisms by which Th17 cytokines contribute to pathology and the antimicrobial response to H. pylori infection.
IL-17A.
IL-17A has been shown to play a role in inflammation as a result of H. pylori colonization (69). Studies from murine models have demonstrated a significant increase in neutrophil infiltration within the submucosa and lamina propria of the stomach in WT mice infected with H. pylori compared to H. pylori-infected IL-17A−/− mice (60). The longer that the WT mice are infected with H. pylori, the greater that the level of neutrophil infiltration is. However, the results from H. pylori infection in IL-17A−/− mice or IL-17RA−/− mice have shown that at no point during infection is there a significant rise in the level of neutrophils (69, 70). All of these studies suggest that IL-17A is essential for neutrophil infiltration. This is likely through the activation of IL-8 homolog expression in epithelial cells, which subsequently recruits neutrophils. Mizuno et al. investigated IL-17A and IL-8 expression in gastric ulcer and nonulcer patients and found increased levels of IL-17A and IL-8 in the gastric mucosa of H. pylori-infected gastric ulcer and nonulcer patients compared to uninfected nonulcer patients (71). Histologically, they found that both IL-17A and IL-8 were strongly correlated with an increase in neutrophil infiltration in infected patients (71). Prior to that study, Luzza et al. analyzed IL-17A RNA transcripts taken from homogenized gastric mucosa from human subjects complaining of dyspeptic symptoms and found that there was increased IL-17A gene expression in H. pylori-infected patients compared to uninfected patients (72). Luzza et al. also demonstrated that IL-17A specifically induced the production of IL-8 in mucosa by isolating lamina propria mononuclear cells (LMPC) from the gastric mucosa and coculturing them with anti-IL-17A antibodies (Ab). This study found that the levels of IL-8 were downregulated in cultures treated with anti-IL-17A Ab in a dose-dependent manner, with greater concentrations of anti-IL-17A Ab resulting in the release of lower levels of IL-8 (73). Therefore, as IL-17A activity heavily stimulates proinflammatory effects, such as neutrophil infiltration through IL-8 stimulation, interest in targeting IL-17A to reduce inflammation has developed. Since neutrophils are significant antimicrobial effector cells, controlling inflammation through IL-17A in light of its antimicrobial role has proven to be a complex task. In some models, a reduction in IL-17 activity may lift a repression of IFN-γ expression, which can also drive inflammation and lead to the recruitment of innate immune cells. When anti-IL-17 antibody was administered to chronically infected mice (74), there was an increase in inflammation, but the authors reported an increase in IFN-γ, and they hypothesized that this was driven by the Th1 response. Therefore, as IL-17A activity heavily stimulates proinflammatory effects, such as neutrophil infiltration through IL-8 stimulation, this provides a promising therapeutic target to reduce this acute inflammation but may not reduce all chronic inflammation, especially if it is driven by Th1 responses.
Expression of IL-17A and control of H. pylori colonization also appear to be linked. It is difficult to separate the role of IL-17A in recruiting polymorphonuclear leukocytes (PMNs) for their antimicrobial functions from the role of IL-17A in activating antimicrobial product expression by epithelial cells of the gastric mucosa. Studies on murine epithelial cells and human gastric epithelial cell lines suggest that IL-17A only minimally activates antimicrobial responses in these cells but, rather, synergizes with IL-22 to activate the expression of S100A8 and S100A9 (both of which are components of calprotectin [CP], lipocalin [LCN], and some β-defensin proteins [75]). Interestingly, however, IL-17A−/− mice controlled H. pylori colonization as well as WT mice and arguably did so better than WT mice in the study of Shiomi et al. (69). On the other hand, in IL-17 receptor A knockout (IL-17RA−/−) mice, the bacterial burden was significantly higher than that in WT mice (70), suggesting that signaling through IL-17RA is important for antimicrobial responses. This is not just evidenced in H. pylori infection models; impaired IL-17 signaling was linked to host susceptibility to a variety of pathogens, including Salmonella enterica, Streptococcus pneumoniae, Listeria monocytogenes, Staphylococcus aureus, Citrobacter rodentium, herpes simplex virus, Trypanosoma cruzi, and Candida albicans (reviewed in reference 76).
Another indication that IL-17A plays a role in the control of bacterial proliferation is through the vaccination studies that have been performed. IL-17 has been identified to be a critical cytokine in vaccine-induced Helicobacter clearance in some studies (77–79), but not in all studies (80, 81). Antibody-mediated depletion of neutrophils abrogated vaccine efficacy, suggesting that PMNs are required for the observed reduced bacterial burden (78). Moreover, treatment of WT immunized mice with anti-IL17A antibodies led to reduced inflammation and increased bacterial colonization after Helicobacter infection compared to the inflammation and colonization seen after treatment of immunized mice with control antibodies. Vaccine-induced reductions in the bacterial load comparable to those in WT mice were observed in IL-12(p35)−/−, IL-23(p19)−/−, and IL-17A−/− mice (80, 82), potentially contradicting the reported requirement for IL-17A activity. This may be explained by the fact that vaccination protocols vary significantly in the mouse models with a variety of antigen types, different adjuvants, and varied routes for immunization. More research is required to interpret correlates of protection. While both Th1 and Th17 responses contribute to the control of extracellular bacterial clearance, only the induced, immunodominant IFN-γ-positive Th1 lymphocytes are required for actual protection against H. pylori (81). An alternative explanation is that Th1 responses may compensate for the absence of a Th17 response if Th1 responses are amplified by administering recombinant cytokines. It has been demonstrated that the use of recombinant IL-12 increased IFN-γ responses to vaccination (83). A review written by D’Elios and Czinn argues that there is “considerable evidence that the key to inducing protection is to pre-empt or override regulatory T-cell activity by promoting Th1 and/or Th17 mediated immunity” (84). This evidence may be worth considering further as vaccines move to clinical trials (ClinicalTrials.gov registration numbers NCT00736476 and NCT02302170).
The role of IL-17A and Th17 cells in activating the inflammatory response and controlling bacterial proliferation may be quite different from the role that IL-17A might play if gastric cancer (GC) develops. The tumor microenvironment (TME) can be immune suppressive, which leads to a bourgeoning interest in exploring whether a proinflammatory environment in the tumor would slow the progression of GC. A recent publication investigating the expression of several cytokines and their relationship with clinicopathological characteristics in GC revealed that IL-17A expression (measured by immunohistochemistry) was associated with decreased survival (85). Further supporting the idea that IL-17A is procarcinogenic, a single study performed on a gastric cancer cell line (AGS cells) found that IL-17A could promote the migration and invasion of GC cells. The data from that study suggest that the upregulated expression and activity of matrix metalloproteinase 2 (MMP-2) and MMP-9 and the downregulated expression of TIMP-1 and TIMP-2 may be the mechanism by which IL-17A promotes invasiveness (86).
On the other hand, the role of IL-17A in different TMEs is not well understood, and there is also some suggestion that increased Th17 responses may lead to better survival. In a herculean effort, Thorsson et al. subtyped more than 10,000 tumors from 33 different cancers by the tumor’s immunological signature (87). The six immune subtypes were type 1 (wound healing), type 2 (IFN-γ), type 3 (inflammatory), type 4 (lymphocyte depleted), type 5 (immunologically quiet), and type 6 (TGF-β dominant) (87). Close to 80% of gastric adenocarcinomas were subtyped into the first two classifications. Type 1 was simply defined as a wound-healing microenvironment, and type 2 was defined as IFN-γ dominant. Both type 1 and type 2 TMEs had high proliferation rates, but type 1 TMEs exhibited elevated angiogenic gene expression and a strong Th2 response, whereas type 2 TMEs not only expressed high levels of IFN-γ but also showed bias toward proinflammatory macrophages and a strong cytotoxic T cell signal. Combining all cancer types, patients with TME defined as immune subtype 3 (elevated Th17 and Th1 signatures with low to moderate proliferation rates) had the best overall survival rates, while patients with the more common subtypes associated with gastric adenocarcinomas (types 1 and 2) had less favorable outcomes (87). It is unclear how commonly immune subtype 3 occurs in gastric cancer and H. pylori infection, indicating a need for a deeper understanding of Th17 cytokines in the development of cancer and in the tumor microenvironment in GC.
IL-17F.
IL-17F is another cytokine that is within the IL-17 family and that is produced by Th17 cells. It has also been implicated in driving inflammation and may contribute to the control of infection. Like IL-17A, the production of IL-17F is regulated by IL-23 (88). IL-17F has been shown to upregulate various proinflammatory cytokines and chemokines, such as IL-2, TGF-β, IL-6, and GM-CSF (89, 90). Research studying the exact role that IL-17F plays in H. pylori infection is minimal in the mouse model. In the gastric mucosa, a compensatory mechanism and/or a redundancy in the immune systems can be found, and these result in similar levels of control of H. pylori when one of these Th17 cytokines is missing.
Expression of IL-17F has been measured in human subjects with H. pylori infection and H. pylori-associated diseases. A study examining the role of IL-17F in functional dyspepsia using human gastric biopsy samples found that an allele of IL-17F, IL-17F 7488T, is positively correlated with the development of functional dyspepsia in H. pylori-infected patients with upper abdominal symptoms who did not show any sign of ulceration of the gastric mucosa. This study also found that the same subjects with the IL-17F 7488T allele had an increase in inflammation in their gastric biopsy samples (91). A meta-analysis of studies focusing on three IL-17 polymorphisms found that these polymorphisms significantly increased the risk of gastric cancer, especially in Chinese participants (92). Another study looked at the expression of IL-17F in H. pylori-infected individuals from Kenya and Germany (93). Kimang’a et al. took stomach biopsy samples from both H. pylori-positive and -negative individuals with dyspeptic symptoms and found that both IL-17A and IL-17F expression was significantly increased in the H. pylori-infected individuals, suggesting a potential link between these two related cytokines (93).
While research on the mechanisms by which IL-17F acts during H. pylori infection has been minimal, some studies have looked at these mechanisms and found that IL-17F is associated with various pathologies in the gastrointestinal tract. Distinct roles for IL-17F and IL-17A in intestinal inflammation have been identified (94). Inhibition of IL-17F, in contrast to inhibition of IL-17A, greatly augmented protection from T cell-induced colitis. This appears to be the result of different regulatory transcription factors and epigenetic modifications between IL-17F and IL-17A; specifically, IL-17F, as opposed to IL-17A, is found to be continuously expressed in naive T cells in colonic cells. Thus, IL-17F can be found in abundance in the intestine (in inflammatory and noninflammatory settings) and has an important function in regulating the microenvironment of the intestinal mucosa. In fact, IL-17F−/− Rag−/− mice given IL-17F−/− T cells showed increased protection against colitis compared with the Rag−/− mouse recipients (94). These studies also demonstrated that IL-17F plays an important role in activating antimicrobial peptides (AMPs) secreted by Paneth cells. While anti-IL17A treatments showed no benefits in their colitis model, IL-17F, indeed, played a vital role in inflammation of the intestines, specifically, in the colonization of the normal flora and functioning as a target for the treatment of intestinal inflammation (94). Research studying the exact role that IL-17F plays in H. pylori infection is minimal in the mouse model. In the gastric mucosa, there may be some compensatory mechanism and/or redundancy in the immune systems which results in similar levels of control of H. pylori when one of these Th17 cytokines is missing. This is an area ripe for research since the role of IL-17F is undefined and its activities independent of IL-17A in the gastric mucosa are not well understood.
Evidence for regulatory roles of IL-17 signaling.
The Th17 cytokines IL-17A, IL-17F, and heterodimer IL-17A/F signal through a shared receptor. These three cytokines, IL-17A, IL-17F, and IL-17A/F, utilize the IL-17R, which is a multimeric receptor with an unknown stoichiometry. While IL-17RA is expressed constitutively on most cells, the signaling through IL-17 has been best characterized in nonhematopoietic cells, including stromal cells, fibroblasts, and epithelial cells. In the mouse model, IL-17RA−/− mice do not respond to H. pylori infection like the single-cytokine-deficient (IL-17A−/−) mice (69, 70, 77). As expected, PMN recruitment to the gastric mucosa was reduced in IL-17RA−/− mice, and the mice did not control H. pylori levels as well as WT mice. Loss of IL-17 receptor signaling led to increased chronic inflammation in the gastric mucosa concomitant with the increased infiltration of CD4+ T cells and a very significant B cell infiltration into the tissue. The B lymphocytes organized into lymphoid follicles with germinal centers. These data indicate that IL-17 signaling, while having proinflammatory activity to drive PMN infiltration and antimicrobial production, may exhibit some regulatory activity. This suggests that some ligation of the receptor by either IL-17A or IL-17F may provide a negative feedback loop within T lymphocytes to inhibit Th17 cytokine production. While there is evidence in the literature for this negative feedback loop, the mechanism by which it functions has not been elucidated. Garg and Gaffen have identified at least two pathways which restrict IL-17 signaling in fibroblasts (95). The investigators demonstrated that the ubiquitin-editing enzyme A20 inhibits IL-17 signaling by deubiquitinating tumor necrosis factor (TNF) receptor-associated factor 6 (TRAF6), thereby limiting NF-κB and mitogen-activated protein kinase activation (95). Since IL-17 upregulates A20 expression, this demonstrates that this pathway of regulation is an established negative feedback loop that restricts IL-17-driven inflammation (96). Moreover, they also reported that the A20-binding inhibitor of NF-κB activation 1 (ABIN-1) negatively regulates the IL-17-induced production of Il6, Lcn2, and a few other NF-κB-dependent genes in fibroblasts and stromal cells (97). We speculate that similar signaling pathways may regulate the downstream activation of T cell cytokine production.
Interestingly, in an Aspergillus airway inflammation model, the IL-17F/IL-17RC axis is functional under conditions of an IL-17RA deficiency model (98). Aspergillus-infected IL-17RA−/− mice had an elevated inflammatory response after infection. However, in this model, in mice lacking IL-17RA, the interaction of IL-17F with IL-17RC on epithelial cells in the upper airways contributed to inflammatory allergy, despite providing protection against local colonizers. It was reported that IL-17F is produced by the lung epithelium, which would be an unusual source of this Th17 cytokine. It has yet to be determined if IL-17F could function in this way in the absence of IL-17RA during H. pylori infection.
These data raise the question: is IL-17 signaling protective for people infected with H. pylori? Despite its reputation for driving inflammation, in the context of H. pylori infection, IL-17 expression may help regulate antimicrobial functions and chronic inflammation. Overexpression or uncontrolled IL-17A expression may contribute to cancer, but there is conflicting evidence as to whether expression of IL-17A or IL-17RA is associated with a better prognosis (87, 99).
IL-17RA can also complex with the IL-17RB subunit. When IL-17RA and IL-17RB complex, they form the receptor for IL-17E (also known as IL-25). The IL-25 signaling pathway has been reported to play a role in Th2 cell-driven inflammation. Furthermore, IL-17B shares the receptor IL-17RB with IL-17E, and the function of IL-17B has not been fully determined. In mouse models of H. pylori infection, the role of IL-17RB has been investigated in both the early and the late stages. During acute infection, H. pylori infection can lead to the decreased expression of IL-17RB on gastric epithelial cells in a cag-dependent fashion. This decreased expression impaired CD11b+ CD11c− myeloid cell accumulation in the gastric mucosa and disrupted the early activation of antimicrobial RegIIIα (100). In a separate study, at the chronic stage of H. pylori infection, IL-17RB−/− mice controlled H. pylori colonization as well as WT mice did, but IL-17RB−/− mice did not develop H. pylori-specific antibodies (101). These data suggest that IL-17E/IL-25 is required for the development of the Th2 cell response to support B cell activation and antibody production.
IL-21.
IL-21 is another cytokine produced by Th17 cells. It is also produced by T follicular helper (Tfh) cells and may be produced in smaller quantities from NK T cells or Th1 cells. IL-21 deficiency in the mouse model has very profound effects on H. pylori-induced gastritis compared to those in WT mice (41). This is likely due to its pleotropic activities in various tissues (Fig. 3 and 4). At 3 months postinfection, IL-21−/− mice do not control H. pylori colonization as well as WT mice, and the bacteria localize more frequently in the gastric glands. IL-21−/− mice exhibit less inflammation than H. pylori-infected WT mice (41). The data also suggest that IL-21 is a key cytokine for maintenance of both the Th1 and Th17 responses during H. pylori infection in the gastric mucosa (Fig. 3). To further investigate T helper cell responses and to delineate the molecular mechanisms behind IL-21-driven signaling during chronic H. pylori infection, computational modeling and mechanistic experimental studies were used (see the data in the supplemental material associated with the published paper for more details [41]). Starting with a published model of CD4+ T cell differentiation (102), the computational model was calibrated and IL-21-specific pathways were investigated in situ. The CD4+ T cell model predicted that proinflammatory markers were positively correlated with IL-21, whereas regulatory T cell responses were negatively correlated. The computational model’s predictions were validated using the IL-21−/− animal model. CD4+ T cells from H. pylori-infected IL-21−/− mice had lower phosphoactivation of STAT1 and STAT3 than CD4+ T cells from H. pylori-infected WT mice. The STAT1 and STAT3 pathways are important for Th1 and Th17 responses, respectively (41). Also, H. pylori infection in IL-21−/− mice led to lower levels of expression of tbx21 and rorc, which encode Tbet and RORγt, respectively, than infection in WT mice (41). Moreover, as predicted, CD4+ T cells from H. pylori-infected IL-21−/− mice expressed higher levels of IL-10 than CD4+ T cells from H. pylori-infected WT mice. These data demonstrate not only that IL-21 plays a critical role in the development or maintenance of Th1 and Th17 responses to H. pylori but also that IL-21 may affect Treg cell responses.
The expression of IL-21 has been associated with H. pylori infection in studies of infected humans (103, 104). An expression analysis of Th17-associated markers during H. pylori infection and gastric cancer demonstrated a strong positive correlation of RORγt and IL-17A with IL-21 in both H. pylori-infected tissues and cancer tissues (105). These data suggest that IL-21 may be a key T cell-derived cytokine promoting a more pathogenic T cell response by driving strong Th1 and Th17 responses and associated pathologies and potentially inhibit Treg cell responses. The concentration of IL-21 in peripheral blood, in fact, is higher in gastric cancer patients than in healthy controls and is associated with the development, metastasis, and maturation of gastric cancer in later stages of disease (106).
IL-21 does signal B lymphocytes to proliferate and switch isotypes; therefore, IL-21 plays a critical role in activation of antigen-specific antibody responses. H. pylori infection of WT mice does induce H. pylori-specific antibody responses, but in H. pylori-infected IL-21−/− mice, H. pylori-specific antibodies were not detected in the serum (41). This could be due to the direct effects of IL-21 on the B cell response or could possibly be due to IL-21 promoting the differentiation of Tfh cells, which then subsequently support B cell activity (107, 108). Although the data are correlative, chronically infected IL-17RA−/− mice, which express very high levels of IL-21, also have significantly higher levels of H. pylori-specific antibodies in their serum than WT mice (70), suggesting that IL-21 may drive the antibody response in IL-17RA−/− mice. These H. pylori-infected IL-17RA−/− mice also develop very significant B cell infiltration and lymphoid follicles with germinal centers in their gastric tissue, a pathological outcome which is rare in WT C57BL/6 mice. The significance of high or low H. pylori-specific antibodies is not well understood. Using B cell-deficient mice, it was demonstrated that H. pylori-specific antibodies inhibit the development of gastritis, may enable bacterial colonization, and actually lead to increased resistance against H. pylori infection (109). As such, the data demonstrating that higher levels of H. pylori-specific antibodies in IL-17RA−/− mice correlate with increased inflammation do not contradict the finding that B cell-deficient mice which lack antigen-specific responses had less inflammation. Rather, the data suggest that the correlation between antibodies and gastritis may support the hypothesis that high H. pylori-specific antibody responses may be a marker of increased inflammation, and there may be a way to use this correlative biomarker to predict when a patient may experience a more detrimental outcome of H. pylori infection. In fact, although potentially looking at it from a different perspective, Epplein et al. have found that seropositivity to a number of specific H. pylori antigens was associated with an increase in the odds of gastric cancer in several prospective cohorts (110–112). The relationship between the inflammatory response, IL-21 expression, IL-21 polymorphisms, and antibody levels in these cohorts has not been investigated.
IL-21 is expressed in lymph nodes or in Peyer’s patches (107, 108) and functions to maintain Tfh cells and B cell function (Fig. 4). Interestingly, while IL-21−/− mice infected with H. pylori had reduced Ifng and reduced Il17 expression in their gastric mucosa compared to WT mice (41), the T cells in the Peyer’s patches and mesenteric lymph nodes of the H. pylori-infected IL-21−/− mice produced significantly more IL-17A (113). Moreover, there was an increase in Th17-directing cytokines, including IL-1β and IL-6, in the IL-21−/− mice than in the WT mice. In vitro assays performed on dendritic cells demonstrated that recombinant IL-21 significantly reduces the H. pylori-induced expression of many proinflammatory cytokines, including IL-1β, IL-6, IL-12, and IL-23, but not IL-10 (113). These data suggest that IL-21 has an immune-modulatory role in the context of dendritic cell responses.
The proinflammatory cytokine IL-21, being pleiotropic, may also signal through gastric epithelial cells. There is only one group thus far which has addressed the ability of IL-21 to activate gastric epithelial cells. Caruso et al. have reported that IL-21 upregulates expression of matrix metalloprotease 1 (MMP-1) and MMP-3 on the AGS cell line (104). In other chronic inflammation settings, such as rheumatoid arthritis, IL-21 induced the migration and invasion of fibroblast-like synoviocytes (114), as well as induced MMP-1 and MMP-3 production in these cells (115). Additional studies utilizing other epithelial cells (i.e., intestinal epithelial cells) have shown that IL-21 can activate the expression of CCL20 (macrophage inflammatory protein 3a), a T cell-recruiting chemokine (116). While not proven in gastric cancer, some cancer cell lines are more invasive and migratory when MMP expression is up and when the cells are treated with IL-21, including some breast cancer cell lines (117). IL-21 binding to its receptor activates the JAK/STAT pathway; in fact, in gastric cancer patients, increased IL-21 receptor expression is correlated with survival and the recurrence of disease. A decrease in IL-21R expression is associated with the reduced growth/spread of gastric cancer, while an increase in IL-21R expression inactivates antitumor medications, such as oxymatrine (OMT), which work through the inactivation of JAK/STAT signaling (118). It has been suggested that since the development of gastric cancer relies upon inflammation, IL-21 could increase the risk of gastric cancer, but more studies of this key cytokine are needed (119). Despite the concern that increased IL-21 signaling may activate inflammation and drive carcinogenesis, there is interest in boosting IL-21 signaling during immune therapies because IL-21 also augments CD8+ T cell functions and could improve antitumor cytotoxic T cell responses. Moreover, one study designed to address how IL-21 directly impacts gastric cancer cells used IL-21 alone and in combination with 5-fluorouracil on gastric cancer cell line SGC-7901 (120). The data from that study demonstrated that IL-21 inhibited the proliferation and growth of the cell line (120).
IL-22.
Interleukin-22, a member of the IL-10 cytokine family (121), was first described to be a Th1 CD4+ T cell-derived cytokine. Later studies attributed IL-22’s expression to predominantly that of the Th17 cell subset; and then Th22 cells, innate lymphoid cells (ILCs), and γδ cells were added to the list of IL-22 producers (122–124). All of these cell types produce IL-22 primarily in response to IL-23. In 2015, Zhuang et al. suggested that an H. pylori infection could elicit a Th22 cell response and IL-22 expression (113). While they also attributed the IL-22 expression to IL-23, they did not rule out Th17 cells as the source of IL-22 in that instance.
IL-22 acts primarily on nonhematopoietic cells. It acts through a heterodimeric receptor of IL-22R1 and IL-10R2, which is present primarily on nonhematopoietic cells of the skin, respiratory system, liver, and gastrointestinal tract (126–130). At mucosal surfaces, its functions include antimicrobial defense, wound healing, and protection against tissue damage (127, 131–133). Specifically, when IL-22 was overexpressed using adenovirus, IL-22 induced a systematic acute-phase response, activating fibrinogen and increasing platelet counts and PMN responses in the blood (134). In the skin lesions of psoriasis patients, higher levels of IL-22 were associated with increased expression of some antimicrobials, namely, S100 proteins (A7, A8, and A9) and increased expression of matrix metalloprotease 1 (135). IL-22 acts independently and synergistically with a number of cytokines, including TNF, IL-1β, and IL-17A (136, 137). It is its synergistic activity with IL-17A that has been a major focus of research, which indicates that, together, these cytokines activate antimicrobial responses in the mucosa, including lipocalin-2, S100A8, S100A9 (and calprotectin A8/A9), and serum amyloid A-3 (SAA3) (134–138).
In H. pylori-infected humans and H. pylori-infected mice, IL-22 expression correlates with increased gastritis (75, 125). Many proinflammatory cytokines are activated on the same time course as IL-22, and several even activate STAT3; therefore, identifying an independent role for IL-22 that is not compensated for has been difficult. The role of IL-22 in the activation of antimicrobial responses was examined in models of H. pylori infection using human gastric epithelial cell lines and the mouse model of H. pylori infection (75). Recapitulating what had been observed in several epithelial cell lines, when both IL-17A and IL-22 were cultured with human cell lines, primary mouse gastric epithelial cells (GEC), or gastroids, and antimicrobials, including components of calprotectin (CP), lipocalin (LCN), and some β-defensins, were induced (75). In vivo IL-22 deficiency alone did not render mice more susceptible to H. pylori infection or significantly change the pathological consequences of the infection (75). These data conflict with data that were published by Zhuang et al., where they attributed IL-22 expression to Th22 cells rather than Th17 cells (125). These experimental design differences are worth noting. WT BALB/c mice and IL-22−/− mice in the BALB/c background were used for the studies of Zhuang et al. (125), whereas in the study of Dixon et al. (75), C57BL/6 mice and IL-22-/- in the C57BL/6 background were used. This is relevant because C57BL/6 and BALB/c mice respond differently to infection (139). Therefore, mice on the C57BL/6 background may have produced higher levels of Th1 cytokines, such as IFN-γ and IL-21, and compensated for the IL-22 deficiency in the experiments performed by Dixon et al. (75). Another difference between the experimental designs was the choice of H. pylori strains. While both studies used cagPAI-positive strains (strain NCTC 11637 [125] versus strain PMSS1 [75]), there may be undefined differences between these strains that could activate epithelial cells directly and induce inflammation differently. A key signaling pathway that may need to be activated to compensate for IL-22 deficiency is the phosphorylation of STAT3. Since several proinflammatory cytokines can activate STAT3 (140), expression of these cytokines may be enough to compensate for IL-22 deficiency. Also, IL-22 or IL-17A may enhance cytokine receptor expression for other proinflammatory cytokines, therefore enhancing epithelial cell sensitivity. Thus, in vivo IL-22 deficiency may be compensated for by several other activation signals that appear to converge on similar signaling components.
IL-21 is not the only Th17 cytokine to impact matrix metalloprotease expression. IL-22 impacts matrix metalloprotease 10 expression and, therefore, impacts gastritis in humans and mouse models. MMP-10 expression was found to be positively correlated with IL-22 expression in the gastric mucosa of H. pylori-infected patients. Moreover, in vitro H. pylori and IL-22 synergistically induce MMP-10 expression in a cagA-dependent pathway (141), suggesting that IL-22 impacts angiogenesis and/or tissue repair.
In a study out of Taiwan, polymorphisms in IL22 were found to be significantly associated with gastric MALT lymphoma (142). Interestingly, H. pylori-infected patients with higher IL-22 expression were more likely to respond to therapy. Consistent with data that IL-22 activates antimicrobial protein expression (75), that study demonstrated that IL-22 expression increased the expression of RegIIIα and LCN2. On the other hand, several recent studies suggest that IL-22 can promote carcinogenesis. IL-22 increased gastric cancer cell invasion through activation of the STAT3/Erk pathway (143) and increased the metastatic activity of human gastric cancer cells through increased AKT activation and MMP-9 production (138). Interestingly, in the tumor microenvironment, IL-22 production may be not only from T lymphocytes but also from cancer-associated fibroblasts (143).
IL-26.
The human IL-26 gene, IL26, maps to chromosome region 12q15 between two other cytokine genes, IFNG and IL22 (144). IL26 is often coexpressed with IL22 at high levels by human Th17 cells (145, 146). High IL-26 expression is strongly associated with inflammatory activity in the synovia of individuals with rheumatoid arthritis, in psoriatic skin lesions, and in the colonic lesions of individuals with inflammatory bowel disease (147–150). A risk locus containing IL26 and single-nucleotide polymorphisms within the IL26 gene region have been associated with several of these inflammatory diseases (151, 152). The IL-26 gene is conserved in most vertebrate species but, curiously, is absent in mice. At the time of this review, our literature review was unable to find any studies addressing whether H. pylori infection enhances the expression of IL-26 in humans. A recent article published results which supported the hypothesis that IL-26 promotes gastric cancer. Immunohistochemical staining of human gastric tissues revealed increased levels of IL-26 and activation of STAT3 (153). Moreover, ex vivo analysis of tumor-infiltrating lymphocytes (TILs) indicated that IL-26 is expressed by both Th17 cells and NKT cells in the tumor. Furthermore, that study demonstrated that IL-26 promotes the proliferation and survival of human gastric cancer cells (MKN45 and SGC-7901 cells) by regulating the balance of STAT1 and STAT3 activation.
IL-26 has also received some attention as an antimicrobial protein. The cytokine has cationic amphipathic properties, and at concentrations greater than 2 mM, IL-26 can kill extracellular bacteria (including Pseudomonas aeruginosa, Escherichia coli, S. aureus, and Klebsiella pneumoniae) (148) via membrane pore formation. Moreover, recent reports suggest that IL-26 may act in a manner similar to that of LL-37 and inhibit S. aureus growth and biofilm formation (154). There is no evidence to date that IL-26 can inhibit H. pylori growth.
PLASTICITY OF T CELL RESPONSES
Defining T helper subsets is helpful for the field to describe cell types, the cytokines that they produce, and their downstream responses, but in reality, there is plenty of evidence to suggest that T helper cells have the ability to switch the cytokine production patterns under some circumstances (reviewed in reference 155). Moreover, the activation of T cell subsets in vivo likely leads to significant heterogeneity within a population of cells. The heterogeneity may be a result of cells being at different points along the pathway to terminal differentiation (if terminal differentiation even exists). Fate- mapping experiments in mouse models have been used to address plasticity, but few fate-mapping experiments have been performed with H. pylori. In the Helicobacter hepaticus infection model, it was found to be the loss of IL-17A expression—in IFN-γ-producing ex-Th17 cells—which plays a pathogenic role in the disease (156). It is common in intestinal inflammation models that IFN-γ and IL-17 dual-producing Tbet-positive RORγt-positive T cells are associated with more severe disease. Adoptive transfer models utilizing both single and double knockout T cells have demonstrated that both IFN-γ and IL-17 contribute to H. pylori gastritis (157).
TOLERANCE THROUGH Treg CELL RESPONSES
While the focus of this review is on the dichotomous roles of Th17 cells in H. pylori infection and inflammation, the impact of Th17 responses on H. pylori cannot be fully considered without some discussion of the other heterogeneous CD4+ T cell populations, including the T regulatory (Treg) cell response. Entire reviews have certainly been dedicated to the potential roles of Treg cells in reducing inflammation, potentially facilitating the persistence of H. pylori and potentially inhibiting anticancer immune responses (158–160). Studies have suggested that it is not the level of the Th17 or Treg cell response in patients which may dictate their prognosis but, rather, the imbalance of Th17 cells and Treg cells which drives chronic inflammation. Treg cells are induced during H. pylori infection. In fact, a few groups have demonstrated that DCs not only activate cytokines which can drive Th1 or Th17 responses but also produce IL-10 and IL-18, which can induce Foxp3 expression and Treg differentiation (161, 162). To characterize whether Treg cells may prevent pathological outcomes, experiments were performed on athymic C57BL/6 nu/nu mice. The C57BL/6 nu/nu mice were reconstituted with either all lymph node cells or lymph node cells depleted of CD25+ T cells. These reconstituted mice were compared to mice that were not reconstituted at all (163). The result of this adoptive transfer experiment demonstrates that in the absence of Treg cells (cells depleted of CD25+ cells), H. pylori colonization is reduced, but at the cost of inducing an earlier, increased severity of gastritis. Moreover, using a different model, depletion of CD25+ Treg cells also led to a reduced H. pylori bacterial burden, which may be due to the enhanced peripheral H. pylori-specific Th17 cells observed in those mice (161). The literature suggests that this balance of Th17 and Treg cells may change as people age. In several studies focusing on H. pylori-infected children, there was evidence that Treg cell responses positively correlate with bacterial density, while Th17 responses negatively correlate with bacterial density (164). In a study which compared adults and children, Serrano et al. reported that children had more IL-10+ cells than adults, and, accordingly, it was reported that they had reduced gastric Th17 responses, which correlated with reduced gastritis (165).
Interestingly, Treg cells may be more plastic than other CD4+ T cell subsets and under some conditions have been shown to become proinflammatory. In vitro, when culturing Treg cells with IL-6, especially in the absence of TGF-β, they upregulate RORγt and produce IL-17 (166). In addition, Foxp3+ IL-17+ T cells have been identified in vivo (167).
CONCLUSIONS
These studies led us to recognize that the field needs a clearer understanding of the dynamic interactions of T cells with the gastric mucosa. For example, Th17 responses (especially IL-22) are important for maintaining the mucosal barrier and repairing the intestinal mucosa; how the Th17 cell-produced cytokines contribute to repair of the gastric mucosa during H. pylori-induced ulcer disease or injury has not been investigated. Moreover, the roles of IL-17F and IL-26 are virtually unexplored and may be underappreciated in this complex microenvironment. There is a lack of understanding of how common dual cytokine (IFN-γ and IL-17)-producing T cells, which are pathogenic in some inflammatory diseases, are activated in response to H. pylori infection, and whether there is a differential role for these dual cytokine-producing cells from that of Th1 and Th17 cells has not been studied in the context of infection. Further, since the IL-17 signaling pathway is being targeted by immunotherapeutics, a better comprehension of the immune-modulatory mechanisms regulating IL-17RA signaling is required. In psoriasis patients, there has been great success with reducing disease pathology through inhibition of the IL-17 pathway, but in Crohn’s disease patients, the data suggest that anti-IL-17 therapies worsen the disease. With up to 50% of the world’s population being colonized by H. pylori, we need a better understanding of how these therapies would impact the outcomes of H. pylori colonization both in the short term (gastritis) and in the long term (carcinogenesis). Finally, an area ripe for research investigation is the impact of Th17 cells in the gastric tumor microenvironment and how the role for Th17 and IL-17 might shift as carcinogenesis develops. Th17 cells may contribute significantly to many of these processes, as they are central to inducing inflammation, activating antimicrobial responses, and impacting cell infiltration in tumor environments.
ACKNOWLEDGMENTS
This work was supported by Office of Medical Research, Veterans Affairs Merit Review, grant IBX000915A (to H.M.S.A.) and the Vanderbilt University Digestive Disease Research Center, supported by NIH grant P30DK058404.
We are very grateful to Sebastian Joyce at the Vanderbilt University Medical Center (Department of Pathology, Microbiology and Immunology, Vanderbilt Institute of Infection, Immunity and Inflammation [VI4]) for the brilliant watercolor art incorporated in Fig. 2 to 4.
Biographies
Beverly R. E. A. Dixon began her journey in biological research at Fisk University, where her academic interest shifted from Computer Information Systems and International Management to Biology. Upon completing her bachelor’s degree at Fisk, she enrolled in the Masters of Agricultural Science program at Tennessee State University. There, she focused on the gut microbiota in poultry, developing a deep appreciation for the astounding capabilities of microbes―especially those that provide significant benefits to the host. This interest in microbe-host interaction led Dixon to the Algood laboratory at Vanderbilt University Medical Center where she serves as a Sr. Research Scientist. Currently, she champions the goals of the Algood laboratory in seeking to answer questions that arise from the interaction between Helicobacter pylori and its human host.
Rafat Hossain graduated from Vanderbilt University in 2017, where he majored in psychology on a pre-medicine track. His interest in medicine, especially the aspects of immunity and microbiology, led him back to Vanderbilt in the summer of 2018, to the Algood laboratory in the Department of Medicine, Division of Infectious Diseases. During this research internship, he worked alongside Dr. Algood and other research assistants exploring the body's immune reaction to inflammation. Rafat is currently a 3rd year medical student at LMU-DeBusk College of Osteopathic Medicine, where he is starting rotations and exploring the various medical fields.
Rachna V. Patel is currently an Assistant Professor of Hospitalist Medicine at Temple University Hospital in Philadelphia, PA. She completed medical school and earned her M.D. at Vanderbilt University Medical School. While at Vanderbilt, she worked under Dr. Holly Algood and helped investigate the role of IL-17 cytokines in the stimulation of antimicrobial responses during H. pylori infection in a mouse model. She completed her residency in internal medicine at Hahnemann University Hospital/Drexel University School of Medicine and then went on to do a sleep medicine fellowship at Thomas Jefferson University Hospital. She will be starting a pulmonary critical care fellowship at Cooper University Hospital.
Holly M. Scott Algood is currently an Associate Professor of Medicine, and Pathology, Microbiology and Immunology at Vanderbilt University Medical Center. She holds a dual appointment with the Department of Veteran’s Affairs Hospital. Dr. Algood established her long-standing interest in microbe-host interactions as an undergraduate student at Mount Union College in Alliance, Ohio. It was there that she did her first experiments on the pathogenic yeast Cryptococcus neoformans. Holly completed her Ph.D. at the University of Pittsburgh in the laboratory of JoAnne Flynn working on animal models of Mycobacterium tuberculosis and the role of TNF on granuloma formation. This training piqued her curiosity and she specifically started investigating how chronic infections challenge the immune system to balance its need for immunity with its need to limit inflammation. Dr. Algood has been working on Helicobacter pylori and mucosal inflammation models since her post-doctoral fellowship began in 2004.
REFERENCES
- 1.Mane SP, Dominguez-Bello MG, Blaser MJ, Sobral BW, Hontecillas R, Skoneczka J, Mohapatra SK, Crasta OR, Evans C, Modise T, Shallom S, Shukla M, Varon C, Megraud F, Maldonado-Contreras AL, Williams KP, Bassaganya-Riera J. 2010. Host-interactive genes in Amerindian Helicobacter pylori diverge from their Old World homologs and mediate inflammatory responses. J Bacteriol 192:3078–3092. doi: 10.1128/JB.00063-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Bik EM, Eckburg PB, Gill SR, Nelson KE, Purdom EA, Francois F, Perez-Perez G, Blaser MJ, Relman DA. 2006. Molecular analysis of the bacterial microbiota in the human stomach. Proc Natl Acad Sci U S A 103:732–737. doi: 10.1073/pnas.0506655103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Ghose C, Perez-Perez GI, van Doorn LJ, Domínguez-Bello MG, Blaser MJ. 2005. High frequency of gastric colonization with multiple Helicobacter pylori strains in Venezuelan subjects. J Clin Microbiol 43:2635–2641. doi: 10.1128/JCM.43.6.2635-2641.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Sonnenberg A, Lash RH, Genta RM. 2010. A national study of Helicobactor [sic] pylori infection in gastric biopsy specimens. Gastroenterology 139:1894–1901.e2. doi: 10.1053/j.gastro.2010.08.018. [DOI] [PubMed] [Google Scholar]
- 5.Vieth M, Masoud B, Meining A, Stolte M. 2000. Helicobacter pylori infection: protection against Barrett’s mucosa and neoplasia? Digestion 62:225–231. doi: 10.1159/000007820. [DOI] [PubMed] [Google Scholar]
- 6.Perry S, de Jong BC, Solnick JV, de la Luz Sanchez M, Yang S, Lin PL, Hansen LM, Talat N, Hill PC, Hussain R, Adegbola RA, Flynn J, Canfield D, Parsonnet J. 2010. Infection with Helicobacter pylori is associated with protection against tuberculosis. PLoS One 5:e8804. doi: 10.1371/journal.pone.0008804. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Reibman J, Marmor M, Filner J, Fernandez-Beros ME, Rogers L, Perez-Perez GI, Blaser MJ. 2008. Asthma is inversely associated with Helicobacter pylori status in an urban population. PLoS One 3:e4060. doi: 10.1371/journal.pone.0004060. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Blaser MJ, Chen Y, Reibman J. 2008. Does Helicobacter pylori protect against asthma and allergy? Gut 57:561–567. doi: 10.1136/gut.2007.133462. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Chen Y, Blaser MJ. 2007. Inverse associations of Helicobacter pylori with asthma and allergy. Arch Intern Med 167:821–827. doi: 10.1001/archinte.167.8.821. [DOI] [PubMed] [Google Scholar]
- 10.Konturek PC, Rienecker H, Hahn EG, Raithel M. 2008. Helicobacter pylori as a protective factor against food allergy. Med Sci Monit 14:CR452–CR458. [PubMed] [Google Scholar]
- 11.Noto JM, Peek RM Jr.. 2012. Helicobacter pylori: an overview. Methods Mol Biol 921:7–10. doi: 10.1007/978-1-62703-005-2_2. [DOI] [PubMed] [Google Scholar]
- 12.Bauer B, Meyer TF. 2011. The human gastric pathogen Helicobacter pylori and its association with gastric cancer and ulcer disease. Ulcers 2011:340157. doi: 10.1155/2011/340157. [DOI] [Google Scholar]
- 13.National Cancer Institute. 2019. Cancer stat facts: stomach cancer, SEER cancer statistics review 1975–2016. https://seer.cancer.gov/csr/1975_2016/.
- 14.World Health Organization. 2019. Cancer: key facts. http://www.who.int/news-room/fact-sheets/detail/cancer. Accessed 20 July 2019.
- 15.Vieth M, Stolte M. 2000. Barrett’s mucosa, Barrett’s dysplasia and Barrett’s carcinoma: diagnostic endoscopy without biopsy-taking does not suffice. Dis Esophagus 13:23–27. doi: 10.1046/j.1442-2050.2000.00068.x. [DOI] [PubMed] [Google Scholar]
- 16.Vaezi MF, Falk GW, Peek RM, Vicari JJ, Goldblum JR, Perez-Perez GI, Rice TW, Blaser MJ, Richter JE. 2000. CagA-positive strains of Helicobacter pylori may protect against Barrett’s esophagus. Am J Gastroenterol 95:2206–2211. doi: 10.1111/j.1572-0241.2000.02305.x. [DOI] [PubMed] [Google Scholar]
- 17.Chow WH, Blaser MJ, Blot WJ, Gammon MD, Vaughan TL, Risch HA, Perez-Perez GI, Schoenberg JB, Stanford JL, Rotterdam H, West AB, Fraumeni JF Jr.. 1998. An inverse relation between cagA+ strains of Helicobacter pylori infection and risk of esophageal and gastric cardia adenocarcinoma. Cancer Res 58:588–590. [PubMed] [Google Scholar]
- 18.Bettelli E, Carrier Y, Gao W, Korn T, Strom TB, Oukka M, Weiner HL, Kuchroo VK. 2006. Reciprocal developmental pathways for the generation of pathogenic effector TH17 and regulatory T cells. Nature 441:235–238. doi: 10.1038/nature04753. [DOI] [PubMed] [Google Scholar]
- 19.Mosmann TR, Coffman RL. 1989. TH1 and TH2 cells: different patterns of lymphokine secretion lead to different functional properties. Annu Rev Immunol 7:145–173. doi: 10.1146/annurev.iy.07.040189.001045. [DOI] [PubMed] [Google Scholar]
- 20.Komiyama Y, Nakae S, Matsuki T, Nambu A, Ishigame H, Kakuta S, Sudo K, Iwakura Y. 2006. IL-17 plays an important role in the development of experimental autoimmune encephalomyelitis. J Immunol 177:566–573. doi: 10.4049/jimmunol.177.1.566. [DOI] [PubMed] [Google Scholar]
- 21.Luther J, Dave M, Higgins PD, Kao JY. 2010. Association between Helicobacter pylori infection and inflammatory bowel disease: a meta-analysis and systematic review of the literature. Inflamm Bowel Dis 16:1077–1084. doi: 10.1002/ibd.21116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Koloski NA, Bret L, Radford-Smith G. 2008. Hygiene hypothesis in inflammatory bowel disease: a critical review of the literature. World J Gastroenterol 14:165–173. doi: 10.3748/wjg.14.165. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Crabtree JE, Wessler S. 2018. Special issue “H. pylori virulence factors in the induction of gastric cancer.” Toxins (Basel) 10:176. doi: 10.3390/toxins10050176. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Jia ZF, Zhang SL, Cao XY, Zhou BS, Jiang J. 2016. Interaction between Helicobacter pylori and host genetic variants in gastric carcinogenesis. Future Oncol 12:2127–2134. doi: 10.2217/fon-2016-0233. [DOI] [PubMed] [Google Scholar]
- 25.Prepin J, Gibello-Kervran C, Charpentier G, Jost A. 1985. Number of germ cells and meiotic prophase stages in fetal rat ovaries cultured in vitro. J Reprod Fertil 73:579–583. doi: 10.1530/jrf.0.0730579. [DOI] [PubMed] [Google Scholar]
- 26.Bhuiyan TR, Qadri F, Saha A, Svennerholm AM. 2009. Infection by Helicobacter pylori in Bangladeshi children from birth to two years: relation to blood group, nutritional status, and seasonality. Pediatr Infect Dis J 28:79–85. doi: 10.1097/INF.0b013e31818a5d9d. [DOI] [PubMed] [Google Scholar]
- 27.Bhuiyan TR, Saha A, Lundgren A, Qadri F, Svennerholm AM. 2010. Immune responses to Helicobacter pylori infection in Bangladeshi children during their first two years of life and the association between maternal antibodies and onset of infection. J Infect Dis 202:1676–1684. doi: 10.1086/657085. [DOI] [PubMed] [Google Scholar]
- 28.Daugule I, Rowland M. 2008. Helicobacter pylori infection in children. Helicobacter 13(Suppl 1):41–46. doi: 10.1111/j.1523-5378.2008.00632.x. [DOI] [PubMed] [Google Scholar]
- 29.de Carvalho Costa Cardinali L, Rocha GA, Rocha AM, de Moura SB, de Figueiredo Soares T, Esteves AM, Nogueira AM, Cabral MM, de Carvalho AS, Bitencourt P, Ferreira A, Queiroz DM. 2003. Evaluation of [13C]urea breath test and Helicobacter pylori stool antigen test for diagnosis of H. pylori infection in children from a developing country. J Clin Microbiol 41:3334–3335. doi: 10.1128/jcm.41.7.3334-3335.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Shaikh S, Khaled MA, Islam A, Kurpad AV, Mahalanabis D. 2005. Evaluation of stool antigen test for Helicobacter pylori infection in asymptomatic children from a developing country using 13C-urea breath test as a standard. J Pediatr Gastroenterol Nutr 40:552–554. doi: 10.1097/01.MPG.0000150093.12457.0D. [DOI] [PubMed] [Google Scholar]
- 31.Algood HM, Gallo-Romero J, Wilson KT, Peek RM Jr, Cover TL. 2007. Host response to Helicobacter pylori infection before initiation of the adaptive immune response. FEMS Immunol Med Microbiol 51:577–586. doi: 10.1111/j.1574-695X.2007.00338.x. [DOI] [PubMed] [Google Scholar]
- 32.Gobert AP, Cheng Y, Wang JY, Boucher JL, Iyer RK, Cederbaum SD, Casero RA Jr, Newton JC, Wilson KT. 2002. Helicobacter pylori induces macrophage apoptosis by activation of arginase II. J Immunol 168:4692–4700. doi: 10.4049/jimmunol.168.9.4692. [DOI] [PubMed] [Google Scholar]
- 33.Chaturvedi R, Cheng Y, Asim M, Bussiere FI, Xu H, Gobert AP, Hacker A, Casero RA Jr, Wilson KT. 2004. Induction of polyamine oxidase 1 by Helicobacter pylori causes macrophage apoptosis by hydrogen peroxide release and mitochondrial membrane depolarization. J Biol Chem 279:40161–40173. doi: 10.1074/jbc.M401370200. [DOI] [PubMed] [Google Scholar]
- 34.Nagai S, Mimuro H, Yamada T, Baba Y, Moro K, Nochi T, Kiyono H, Suzuki T, Sasakawa C, Koyasu S. 2007. Role of Peyer’s patches in the induction of Helicobacter pylori-induced gastritis. Proc Natl Acad Sci U S A 104:8971–8976. doi: 10.1073/pnas.0609014104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Bimczok D, Clements RH, Waites KB, Novak L, Eckhoff DE, Mannon PJ, Smith PD, Smythies LE. 2010. Human primary gastric dendritic cells induce a Th1 response to H. pylori. Mucosal Immunol 3:260–269. doi: 10.1038/mi.2010.10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Horvath DJ Jr, Washington MK, Cope VA, Algood HM. 2012. IL-23 contributes to control of chronic Helicobacter pylori infection and the development of T helper responses in a mouse model. Front Immunol 3:56. doi: 10.3389/fimmu.2012.00056. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Zhuang Y, Shi Y, Liu XF, Zhang JY, Liu T, Fan X, Luo J, Wu C, Yu S, Chen L, Luo P, Guo G, Liu Z, Tang B, Mao XH, Guo Y, Zou QM. 2011. Helicobacter pylori-infected macrophages induce Th17 cell differentiation. Immunobiology 216:200–207. doi: 10.1016/j.imbio.2010.05.005. [DOI] [PubMed] [Google Scholar]
- 38.Cheung PF, Wong CK, Lam CW. 2008. Molecular mechanisms of cytokine and chemokine release from eosinophils activated by IL-17A, IL-17F, and IL-23: implication for Th17 lymphocytes-mediated allergic inflammation. J Immunol 180:5625–5635. doi: 10.4049/jimmunol.180.8.5625. [DOI] [PubMed] [Google Scholar]
- 39.Caruso R, Fina D, Paoluzi OA, Del Vecchio Blanco G, Stolfi C, Rizzo A, Caprioli F, Sarra M, Andrei F, Fantini MC, MacDonald TT, Pallone F, Monteleone G. 2008. IL-23-mediated regulation of IL-17 production in Helicobacter pylori-infected gastric mucosa. Eur J Immunol 38:470–478. doi: 10.1002/eji.200737635. [DOI] [PubMed] [Google Scholar]
- 40.Kabir S. 2011. The role of interleukin-17 in the Helicobacter pylori induced infection and immunity. Helicobacter 16:1–8. doi: 10.1111/j.1523-5378.2010.00812.x. [DOI] [PubMed] [Google Scholar]
- 41.Carbo A, Olivares-Villagomez D, Hontecillas R, Bassaganya-Riera J, Chaturvedi R, Piazuelo MB, Delgado A, Washington MK, Wilson KT, Algood HM. 2014. Systems modeling of the role of interleukin-21 in the maintenance of effector CD4+ T cell responses during chronic Helicobacter pylori infection. mBio 5:e01243-14. doi: 10.1128/mBio.01243-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Kao JY, Zhang M, Miller MJ, Mills JC, Wang B, Liu M, Eaton KA, Zou W, Berndt BE, Cole TS, Takeuchi T, Owyang SY, Luther J. 2010. Helicobacter pylori immune escape is mediated by dendritic cell-induced Treg skewing and Th17 suppression in mice. Gastroenterology 138:1046–1054. doi: 10.1053/j.gastro.2009.11.043. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Wang YH, Gorvel JP, Chu YT, Wu JJ, Lei HY. 2010. Helicobacter pylori impairs murine dendritic cell responses to infection. PLoS One 5:e10844. doi: 10.1371/journal.pone.0010844. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Tanaka H, Yoshida M, Nishiumi S, Ohnishi N, Kobayashi K, Yamamoto K, Fujita T, Hatakeyama M, Azuma T. 2010. The CagA protein of Helicobacter pylori suppresses the functions of dendritic cell in mice. Arch Biochem Biophys 498:35–42. doi: 10.1016/j.abb.2010.03.021. [DOI] [PubMed] [Google Scholar]
- 45.Zhang JY, Liu T, Guo H, Liu XF, Zhuang Y, Yu S, Chen L, Wu C, Zhao Z, Tang B, Luo P, Mao XH, Guo G, Shi Y, Zou QM. 2011. Induction of a Th17 cell response by Helicobacter pylori urease subunit B. Immunobiology 216:803–810. doi: 10.1016/j.imbio.2010.12.006. [DOI] [PubMed] [Google Scholar]
- 46.Li HB, Zhang JY, He YF, Chen L, Li B, Liu KY, Yang WC, Zhao Z, Zou QM, Wu C. 2012. Systemic immunization with an epitope-based vaccine elicits a Th1-biased response and provides protection against Helicobacter pylori in mice. Vaccine 31:120–126. doi: 10.1016/j.vaccine.2012.10.091. [DOI] [PubMed] [Google Scholar]
- 47.Paziak-Domanska B, Chmiela M, Jarosinska A, Rudnicka W. 2000. Potential role of CagA in the inhibition of T cell reactivity in Helicobacter pylori infections. Cell Immunol 202:136–139. doi: 10.1006/cimm.2000.1654. [DOI] [PubMed] [Google Scholar]
- 48.Wang SK, Zhu HF, He BS, Zhang ZY, Chen ZT, Wang ZZ, Wu GL. 2007. CagA+ H. pylori infection is associated with polarization of T helper cell immune responses in gastric carcinogenesis. World J Gastroenterol 13:2923–2931. doi: 10.3748/wjg.v13.i21.2923. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Lina TT, Pinchuk IV, House J, Yamaoka Y, Graham DY, Beswick EJ, Reyes VE. 2013. CagA-dependent downregulation of B7-H2 expression on gastric mucosa and inhibition of Th17 responses during Helicobacter pylori infection. J Immunol 191:3838–3846. doi: 10.4049/jimmunol.1300524. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Das S, Suarez G, Beswick EJ, Sierra JC, Graham DY, Reyes VE. 2006. Expression of B7-H1 on gastric epithelial cells: its potential role in regulating T cells during Helicobacter pylori infection. J Immunol 176:3000–3009. doi: 10.4049/jimmunol.176.5.3000. [DOI] [PubMed] [Google Scholar]
- 51.Lina TT, Alzahrani S, House J, Yamaoka Y, Sharpe AH, Rampy BA, Pinchuk IV, Reyes VE. 2015. Helicobacter pylori cag pathogenicity island’s role in B7-H1 induction and immune evasion. PLoS One 10:e0121841. doi: 10.1371/journal.pone.0121841. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Ricci V, Giannouli M, Romano M, Zarrilli R. 2014. Helicobacter pylori gamma-glutamyl transpeptidase and its pathogenic role. World J Gastroenterol 20:630–638. doi: 10.3748/wjg.v20.i3.630. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Becher B, Durell BG, Noelle RJ. 2002. Experimental autoimmune encephalitis and inflammation in the absence of interleukin-12. J Clin Invest 110:493–497. doi: 10.1172/JCI15751. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Gran B, Zhang GX, Yu S, Li J, Chen XH, Ventura ES, Kamoun M, Rostami A. 2002. IL-12p35-deficient mice are susceptible to experimental autoimmune encephalomyelitis: evidence for redundancy in the IL-12 system in the induction of central nervous system autoimmune demyelination. J Immunol 169:7104–7110. doi: 10.4049/jimmunol.169.12.7104. [DOI] [PubMed] [Google Scholar]
- 55.Tran EH, Prince EN, Owens T. 2000. IFN-gamma shapes immune invasion of the central nervous system via regulation of chemokines. J Immunol 164:2759–2768. doi: 10.4049/jimmunol.164.5.2759. [DOI] [PubMed] [Google Scholar]
- 56.McCune A, Lane A, Murray L, Harvey I, Nair P, Donovan J, Harvey R. 2003. Reduced risk of atopic disorders in adults with Helicobacter pylori infection. Eur J Gastroenterol Hepatol 15:637–640. doi: 10.1097/01.meg.0000059127.68845.57. [DOI] [PubMed] [Google Scholar]
- 57.Bettelli E, Korn T, Oukka M, Kuchroo VK. 2008. Induction and effector functions of T(H)17 cells. Nature 453:1051–1057. doi: 10.1038/nature07036. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Ivanov II, McKenzie BS, Zhou L, Tadokoro CE, Lepelley A, Lafaille JJ, Cua DJ, Littman DR. 2006. The orphan nuclear receptor RORgammat directs the differentiation program of proinflammatory IL-17+ T helper cells. Cell 126:1121–1133. doi: 10.1016/j.cell.2006.07.035. [DOI] [PubMed] [Google Scholar]
- 59.Zhou L, Ivanov II, Spolski R, Min R, Shenderov K, Egawa T, Levy DE, Leonard WJ, Littman DR. 2007. IL-6 programs T(H)-17 cell differentiation by promoting sequential engagement of the IL-21 and IL-23 pathways. Nat Immunol 8:967–974. doi: 10.1038/ni1488. [DOI] [PubMed] [Google Scholar]
- 60.Conti HR, Bruno VM, Childs EE, Daugherty S, Hunter JP, Mengesha BG, Saevig DL, Hendricks MR, Coleman BM, Brane L, Solis N, Cruz JA, Verma AH, Garg AV, Hise AG, Richardson JP, Naglik JR, Filler SG, Kolls JK, Sinha S, Gaffen SL. 2016. IL-17 receptor signaling in oral epithelial cells is critical for protection against oropharyngeal candidiasis. Cell Host Microbe 20:606–617. doi: 10.1016/j.chom.2016.10.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Kimizuka Y, Kimura S, Saga T, Ishii M, Hasegawa N, Betsuyaku T, Iwakura Y, Tateda K, Yamaguchi K. 2012. Roles of interleukin-17 in an experimental Legionella pneumophila pneumonia model. Infect Immun 80:1121–1127. doi: 10.1128/IAI.05544-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Kawaguchi M, Kokubu F, Kuga H, Matsukura S, Hoshino H, Ieki K, Imai T, Adachi M, Huang SK. 2001. Modulation of bronchial epithelial cells by IL-17. J Allergy Clin Immunol 108:804–809. doi: 10.1067/mai.2001.119027. [DOI] [PubMed] [Google Scholar]
- 63.Iwakura Y, Ishigame H. 2006. The IL-23/IL-17 axis in inflammation. J Clin Invest 116:1218–1222. doi: 10.1172/JCI28508. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Ye P, Rodriguez FH, Kanaly S, Stocking KL, Schurr J, Schwarzenberger P, Oliver P, Huang W, Zhang P, Zhang J, Shellito JE, Bagby GJ, Nelson S, Charrier K, Peschon JJ, Kolls JK. 2001. Requirement of interleukin 17 receptor signaling for lung CXC chemokine and granulocyte colony-stimulating factor expression, neutrophil recruitment, and host defense. J Exp Med 194:519–527. doi: 10.1084/jem.194.4.519. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Korn T, Bettelli E, Gao W, Awasthi A, Jager A, Strom TB, Oukka M, Kuchroo VK. 2007. IL-21 initiates an alternative pathway to induce proinflammatory T(H)17 cells. Nature 448:484–487. doi: 10.1038/nature05970. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Ozaki K, Spolski R, Feng CG, Qi CF, Cheng J, Sher A, Morse HC III, Liu C, Schwartzberg PL, Leonard WJ. 2002. A critical role for IL-21 in regulating immunoglobulin production. Science 298:1630–1634. doi: 10.1126/science.1077002. [DOI] [PubMed] [Google Scholar]
- 67.Liang SC, Tan XY, Luxenberg DP, Karim R, Dunussi-Joannopoulos K, Collins M, Fouser LA. 2006. Interleukin (IL)-22 and IL-17 are coexpressed by Th17 cells and cooperatively enhance expression of antimicrobial peptides. J Exp Med 203:2271–2279. doi: 10.1084/jem.20061308. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Zenewicz LA, Flavell RA. 2011. Recent advances in IL-22 biology. Int Immunol 23:159–163. doi: 10.1093/intimm/dxr001. [DOI] [PubMed] [Google Scholar]
- 69.Shiomi S, Toriie A, Imamura S, Konishi H, Mitsufuji S, Iwakura Y, Yamaoka Y, Ota H, Yamamoto T, Imanishi J, Kita M. 2008. IL-17 is involved in Helicobacter pylori-induced gastric inflammatory responses in a mouse model. Helicobacter 13:518–524. doi: 10.1111/j.1523-5378.2008.00629.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Algood HM, Allen SS, Washington MK, Peek RM Jr, Miller GG, Cover TL. 2009. Regulation of gastric B cell recruitment is dependent on IL-17 receptor A signaling in a model of chronic bacterial infection. J Immunol 183:5837–5846. doi: 10.4049/jimmunol.0901206. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Mizuno T, Ando T, Nobata K, Tsuzuki T, Maeda O, Watanabe O, Minami M, Ina K, Kusugami K, Peek RM, Goto H. 2005. Interleukin-17 levels in Helicobacter pylori-infected gastric mucosa and pathologic sequelae of colonization. World J Gastroenterol 11:6305–6311. doi: 10.3748/wjg.v11.i40.6305. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Luzza F, Parrello T, Sebkova L, Pensabene L, Imeneo M, Mancuso M, La Vecchia AM, Monteleone G, Strisciuglio P, Pallone F. 2001. Expression of proinflammatory and Th1 but not Th2 cytokines is enhanced in gastric mucosa of Helicobacter pylori infected children. Dig Liver Dis 33:14–20. doi: 10.1016/S1590-8658(01)80130-4. [DOI] [PubMed] [Google Scholar]
- 73.Luzza F, Parrello T, Monteleone G, Sebkova L, Romano M, Zarrilli R, Imeneo M, Pallone F. 2000. Up-regulation of IL-17 is associated with bioactive IL-8 expression in Helicobacter pylori-infected human gastric mucosa. J Immunol 165:5332–5337. doi: 10.4049/jimmunol.165.9.5332. [DOI] [PubMed] [Google Scholar]
- 74.Shi Y, Liu XF, Zhuang Y, Zhang JY, Liu T, Yin Z, Wu C, Mao XH, Jia KR, Wang FJ, Guo H, Flavell RA, Zhao Z, Liu KY, Xiao B, Guo Y, Zhang WJ, Zhou WY, Guo G, Zou QM. 2010. Helicobacter pylori-induced Th17 responses modulate Th1 cell responses, benefit bacterial growth, and contribute to pathology in mice. J Immunol 184:5121–5129. doi: 10.4049/jimmunol.0901115. [DOI] [PubMed] [Google Scholar]
- 75.Dixon BR, Radin JN, Piazuelo MB, Contreras DC, 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: 10.1371/journal.pone.0148514. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Gu C, Wu L, Li X. 2013. IL-17 family: cytokines, receptors and signaling. Cytokine 64:477–485. doi: 10.1016/j.cyto.2013.07.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Velin D, Favre L, Bernasconi E, Bachmann D, Pythoud C, Saiji E, Bouzourene H, Michetti P. 2009. Interleukin-17 is a critical mediator of vaccine-induced reduction of Helicobacter infection in the mouse model. Gastroenterology 136:2237–2246.e1. doi: 10.1053/j.gastro.2009.02.077. [DOI] [PubMed] [Google Scholar]
- 78.DeLyria ES, Redline RW, Blanchard TG. 2009. Vaccination of mice against H. pylori induces a strong Th-17 response and immunity that is neutrophil dependent. Gastroenterology 136:247–256. doi: 10.1053/j.gastro.2008.09.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Flach C-F, Östberg AK, Nilsson A-T, Malefyt RDW, Raghavan S. 2011. Proinflammatory cytokine gene expression in the stomach correlates with vaccine-induced protection against Helicobacter pylori infection in mice: an important role for interleukin-17 during the effector phase. Infect Immun 79:879–886. doi: 10.1128/IAI.00756-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.DeLyria ES, Nedrud JG, Ernst PB, Alam MS, Redline RW, Ding H, Czinn SJ, Xu J, Blanchard TG. 2011. Vaccine-induced immunity against Helicobacter pylori in the absence of IL-17A. Helicobacter 16:169–178. doi: 10.1111/j.1523-5378.2011.00839.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Li B, Chen L, Sun H, Yang W, Hu J, He Y, Wei S, Zhao Z, Zhang J, Li H, Zou Q, Wu C. 2015. Immunodominant epitope-specific Th1 but not Th17 responses mediate protection against Helicobacter pylori infection following UreB vaccination of BALB/c mice. Sci Rep 5:14793. doi: 10.1038/srep14793. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Ding H, Nedrud JG, Blanchard TG, Zagorski BM, Li G, Shiu J, Xu J, Czinn SJ. 2013. Th1-mediated immunity against Helicobacter pylori can compensate for lack of Th17 cells and can protect mice in the absence of immunization. PLoS One 8:e69384. doi: 10.1371/journal.pone.0069384. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Akhiani AA, Pappo J, Kabok Z, Schon K, Gao W, Franzen LE, Lycke N. 2002. Protection against Helicobacter pylori infection following immunization is IL-12-dependent and mediated by Th1 cells. J Immunol 169:6977–6984. doi: 10.4049/jimmunol.169.12.6977. [DOI] [PubMed] [Google Scholar]
- 84.D’Elios MM, Czinn SJ. 2014. Immunity, inflammation, and vaccines for Helicobacter pylori. Helicobacter 19(Suppl 1):19–26. doi: 10.1111/hel.12156. [DOI] [PubMed] [Google Scholar]
- 85.Kim JY, Bae BN, Kang G, Kim HJ, Park K. 2017. Cytokine expression associated with Helicobacter pylori and Epstein-Barr virus infection in gastric carcinogenesis. APMIS 125:808–815. doi: 10.1111/apm.12725. [DOI] [PubMed] [Google Scholar]
- 86.Wang Y, Wu H, Wu X, Bian Z, Gao Q. 2014. Interleukin 17A promotes gastric cancer invasiveness via NF-kappaB mediated matrix metalloproteinases 2 and 9 expression. PLoS One 9:e96678. doi: 10.1371/journal.pone.0096678. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Thorsson V, Gibbs DL, Brown SD, Wolf D, Bortone DS, Ou Yang T-H, Porta-Pardo E, Gao GF, Plaisier CL, Eddy JA, Ziv E, Culhane AC, Paull EO, Sivakumar IKA, Gentles AJ, Malhotra R, Farshidfar F, Colaprico A, Parker JS, Mose LE, Vo NS, Liu J, Liu Y, Rader J, Dhankani V, Reynolds SM, Bowlby R, Califano A, Cherniack AD, Anastassiou D, Bedognetti D, Mokrab Y, Newman AM, Rao A, Chen K, Krasnitz A, Hu H, Malta TM, Noushmehr H, Pedamallu CS, Bullman S, Ojesina AI, Lamb A, Zhou W, Shen H, Choueiri TK, Weinstein JN, Guinney J, Saltz J, Holt RA, et al. . 2018. The immune landscape of cancer. Immunity 48:812–830.e14. doi: 10.1016/j.immuni.2018.03.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Langrish CL, Chen Y, Blumenschein WM, Mattson J, Basham B, Sedgwick JD, McClanahan T, Kastelein RA, Cua DJ. 2005. IL-23 drives a pathogenic T cell population that induces autoimmune inflammation. J Exp Med 201:233–240. doi: 10.1084/jem.20041257. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Yang XO, Chang SH, Park H, Nurieva R, Shah B, Acero L, Wang YH, Schluns KS, Broaddus RR, Zhu Z, Dong C. 2008. Regulation of inflammatory responses by IL-17F. J Exp Med 205:1063–1075. doi: 10.1084/jem.20071978. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Starnes T, Robertson MJ, Sledge G, Kelich S, Nakshatri H, Broxmeyer HE, Hromas R. 2001. Cutting edge: IL-17F, a novel cytokine selectively expressed in activated T cells and monocytes, regulates angiogenesis and endothelial cell cytokine production. J Immunol 167:4137–4140. doi: 10.4049/jimmunol.167.8.4137. [DOI] [PubMed] [Google Scholar]
- 91.Arisawa T, Tahara T, Shibata T, Nagasaka M, Nakamura M, Kamiya Y, Fujita H, Yoshioka D, Arima Y, Okubo M, Hirata I, Nakano H. 2007. Genetic polymorphisms of molecules associated with inflammation and immune response in Japanese subjects with functional dyspepsia. Int J Mol Med 20:717–723. doi: 10.3892/ijmm.20.5.717. [DOI] [PubMed] [Google Scholar]
- 92.Gagala C, Kawalski A, Klukowski M. 1980. Fistulas for decompression and parenteral feeding after operations on the gastrointestinal tract. Pol Przegl Chir 52:55–58. (In Polish.) [PubMed] [Google Scholar]
- 93.Kimang’a A, Revathi G, Kariuki S, Sayed S, Devani S, Vivienne M, Kuester D, Monkemuller K, Malfertheiner P, Wex T. 2010. IL-17A and IL-17F gene expression is strongly induced in the mucosa of H. pylori-infected subjects from Kenya and Germany. Scand J Immunol 72:522–528. doi: 10.1111/j.1365-3083.2010.02469.x. [DOI] [PubMed] [Google Scholar]
- 94.Tang C, Kakuta S, Shimizu K, Kadoki M, Kamiya T, Shimazu T, Kubo S, Saijo S, Ishigame H, Nakae S, Iwakura Y. 2018. Suppression of IL-17F, but not of IL-17A, provides protection against colitis by inducing Treg cells through modification of the intestinal microbiota. Nat Immunol 19:755–765. doi: 10.1038/s41590-018-0134-y. [DOI] [PubMed] [Google Scholar]
- 95.Garg AV, Gaffen SL. 2013. IL-17 signaling and A20: a balancing act. Cell Cycle 12:3459–3460. doi: 10.4161/cc.26699. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Garg AV, Ahmed M, Vallejo AN, Ma A, Gaffen SL. 2013. The deubiquitinase A20 mediates feedback inhibition of interleukin-17 receptor signaling. Sci Signal 6:ra44. doi: 10.1126/scisignal.2003699. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Cruz JA, Childs EE, Amatya N, Garg AV, Beyaert R, Kane LP, Aneskievich BJ, Ma A, Gaffen SL. 2017. IL-17 signaling triggers degradation of the constitutive NF-κB inhibitor ABIN-1. Immunohorizons 1:133–141. doi: 10.4049/immunohorizons.1700035. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.De Luca A, Pariano M, Cellini B, Costantini C, Villella VR, Jose SS, Palmieri M, Borghi M, Galosi C, Paolicelli G, Maiuri L, Fric J, Zelante T. 2017. The IL-17F/IL-17RC axis promotes respiratory allergy in the proximal airways. Cell Rep 20:1667–1680. doi: 10.1016/j.celrep.2017.07.063. [DOI] [PubMed] [Google Scholar]
- 99.Uhlen M, Bjorling E, Agaton C, Szigyarto CA, Amini B, Andersen E, Andersson AC, Angelidou P, Asplund A, Asplund C, Berglund L, Bergstrom K, Brumer H, Cerjan D, Ekstrom M, Elobeid A, Eriksson C, Fagerberg L, Falk R, Fall J, Forsberg M, Bjorklund MG, Gumbel K, Halimi A, Hallin I, Hamsten C, Hansson M, Hedhammar M, Hercules G, Kampf C, Larsson K, Lindskog M, Lodewyckx W, Lund J, Lundeberg J, Magnusson K, Malm E, Nilsson P, Odling J, Oksvold P, Olsson I, Oster E, Ottosson J, Paavilainen L, Persson A, Rimini R, Rockberg J, Runeson M, Sivertsson A, Skollermo A, et al. . 2005. A human protein atlas for normal and cancer tissues based on antibody proteomics. Mol Cell Proteomics 4:1920–1932. doi: 10.1074/mcp.M500279-MCP200. [DOI] [PubMed] [Google Scholar]
- 100.Teng YS, Liu YG, Chen XH, Wang TT, Cheng P, Lv YP, Kong H, Mao FY, Hao CJ, Yang SM, Chen W, Zhang JY, Peng LS, Han B, Ma Q, Han J, Zou QM, Zhuang Y. 2019. Decreased IL-17RB expression impairs CD11b(+)CD11c(−) myeloid cell accumulation in gastric mucosa and host defense during the early-phase of Helicobacter pylori infection. Cell Death Dis 10:79. doi: 10.1038/s41419-019-1312-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Horvath DJ Jr, Radin JN, Cho SH, Washington MK, Algood HM. 2013. The interleukin-17 receptor B subunit is essential for the Th2 response to Helicobacter pylori, but not for control of bacterial burden. PLoS One 8:e60363. doi: 10.1371/journal.pone.0060363. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Carbo A, Hontecillas R, Kronsteiner B, Viladomiu M, Pedragosa M, Lu P, Philipson CW, Hoops S, Marathe M, Eubank S, Bisset K, Wendelsdorf K, Jarrah A, Mei Y, Bassaganya-Riera J. 2013. Systems modeling of molecular mechanisms controlling cytokine-driven CD4+ T cell differentiation and phenotype plasticity. PLoS Comput Biol 9:e1003027. doi: 10.1371/journal.pcbi.1003027. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Bagheri N, Azadegan-Dehkordi F, Shirzad M, Zamanzad B, Rahimian G, Taghikhani A, Rafieian-Kopaei M, Shirzad H. 2015. Mucosal interleukin-21 mRNA expression level is high in patients with Helicobacter pylori and is associated with the severity of gastritis. Cent Eur J Immunol 40:61–67. doi: 10.5114/ceji.2015.50835. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Caruso R, Fina D, Peluso I, Fantini MC, Tosti C, Del Vecchio Blanco G, Paoluzi OA, Caprioli F, Andrei F, Stolfi C, Romano M, Ricci V, MacDonald TT, Pallone F, Monteleone G. 2007. IL-21 is highly produced in Helicobacter pylori-infected gastric mucosa and promotes gelatinases synthesis. J Immunol 178:5957–5965. doi: 10.4049/jimmunol.178.9.5957. [DOI] [PubMed] [Google Scholar]
- 105.Pinchuk IV, Morris KT, Nofchissey RA, Earley RB, Wu JY, Ma TY, Beswick EJ. 2013. Stromal cells induce Th17 during Helicobacter pylori infection and in the gastric tumor microenvironment. PLoS One 8:e53798. doi: 10.1371/journal.pone.0053798. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Meng X, Yu X, Dong Q, Xu X, Li J, Xu Q, Ma J, Zhou C. 2018. Distribution of circulating follicular helper T cells and expression of interleukin-21 and chemokine C-X-C ligand 13 in gastric cancer. Oncol Lett 16:3917–3922. doi: 10.3892/ol.2018.9112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Vogelzang A, McGuire HM, Yu D, Sprent J, Mackay CR, King C. 2008. A fundamental role for interleukin-21 in the generation of T follicular helper cells. Immunity 29:127–137. doi: 10.1016/j.immuni.2008.06.001. [DOI] [PubMed] [Google Scholar]
- 108.Chtanova T, Tangye SG, Newton R, Frank N, Hodge MR, Rolph MS, Mackay CR. 2004. T follicular helper cells express a distinctive transcriptional profile, reflecting their role as non-Th1/Th2 effector cells that provide help for B cells. J Immunol 173:68–78. doi: 10.4049/jimmunol.173.1.68. [DOI] [PubMed] [Google Scholar]
- 109.Akhiani AA, Schon K, Franzen LE, Pappo J, Lycke N. 2004. Helicobacter pylori-specific antibodies impair the development of gastritis, facilitate bacterial colonization, and counteract resistance against infection. J Immunol 172:5024–5033. doi: 10.4049/jimmunol.172.8.5024. [DOI] [PubMed] [Google Scholar]
- 110.Cai H, Ye F, Michel A, Murphy G, Sasazuki S, Taylor PR, Qiao YL, Park SK, Yoo KY, Jee SH, Cho ER, Kim J, Chen SC, Abnet CC, Tsugane S, Cai Q, Shu XO, Zheng W, Pawlita M, Epplein M. 2016. Helicobacter pylori blood biomarker for gastric cancer risk in East Asia. Int J Epidemiol 45:774–781. doi: 10.1093/ije/dyw078. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Epplein M, Pawlita M, Michel A, Peek RM Jr, Cai Q, Blot WJ. 2013. Helicobacter pylori protein-specific antibodies and risk of colorectal cancer. Cancer Epidemiol Biomarkers Prev 22:1964–1974. doi: 10.1158/1055-9965.EPI-13-0702. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Epplein M, Zheng W, Xiang YB, Peek RM Jr, Li H, Correa P, Gao J, Michel A, Pawlita M, Cai Q, Shu XO. 2012. Prospective study of Helicobacter pylori biomarkers for gastric cancer risk among Chinese men. Cancer Epidemiol Biomarkers Prev 21:2185–2192. doi: 10.1158/1055-9965.EPI-12-0792-T. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Yasmin S, Dixon BREA, Olivares-Villagomez D, Algood HMS. 2019. Interleukin-21 (IL-21) downregulates dendritic cell cytokine responses to Helicobacter pylori and modulates T lymphocyte IL-17A expression in the Peyer’s patches during infection. Infect Immun 11:e00237-19. doi: 10.1128/IAI.00237-19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Xing R, Jin Y, Sun L, Yang L, Li C, Li Z, Liu X, Zhao J. 2016. Interleukin-21 induces migration and invasion of fibroblast-like synoviocytes from patients with rheumatoid arthritis. Clin Exp Immunol 184:147–158. doi: 10.1111/cei.12751. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Lebre MC, Vieira PL, Tang MW, Aarrass S, Helder B, Newsom-Davis T, Tak PP, Screaton GR. 2017. Synovial IL-21/TNF-producing CD4(+) T cells induce joint destruction in rheumatoid arthritis by inducing matrix metalloproteinase production by fibroblast-like synoviocytes. J Leukoc Biol 101:775–783. doi: 10.1189/jlb.5A0516-217RR. [DOI] [PubMed] [Google Scholar]
- 116.Caruso R, Fina D, Peluso I, Stolfi C, Fantini MC, Gioia V, Caprioli F, Del Vecchio Blanco G, Paoluzi OA, Macdonald TT, Pallone F, Monteleone G. 2007. A functional role for interleukin-21 in promoting the synthesis of the T-cell chemoattractant, MIP-3alpha, by gut epithelial cells. Gastroenterology 132:166–175. doi: 10.1053/j.gastro.2006.09.053. [DOI] [PubMed] [Google Scholar]
- 117.Wang LN, Cui YX, Ruge F, Jiang WG. 2015. Interleukin 21 and its receptor play a role in proliferation, migration and invasion of breast cancer cells. Cancer Genomics Proteomics 12:211–221. [PubMed] [Google Scholar]
- 118.Huang Y, Zhang J, Wang G, Chen X, Zhang R, Liu H, Zhu J. 2018. Oxymatrine exhibits anti-tumor activity in gastric cancer through inhibition of IL-21R-mediated JAK2/STAT3 pathway. Int J Immunopathol Pharmacol 32:2058738418781634. doi: 10.1177/2058738418781634. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Stolfi C, Pallone F, Macdonald TT, Monteleone G. 2012. Interleukin-21 in cancer immunotherapy: friend or foe? Oncoimmunology 1:351–354. doi: 10.4161/onci.19122. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Fu ZQ, Zhou Q, Zhu S, Liu W. 2018. Anti-tumor mechanism of IL-21 used alone and in combination with 5-fluorouracil in vitro on human gastric cancer cells. J Biol Regul Homeost Agents 32:619–625. [PubMed] [Google Scholar]
- 121.Xie MH, Aggarwal S, Ho WH, Foster J, Zhang Z, Stinson J, Wood WI, Goddard AD, Gurney AL. 2000. Interleukin (IL)-22, a novel human cytokine that signals through the interferon receptor-related proteins CRF2-4 and IL-22R. J Biol Chem 275:31335–31339. doi: 10.1074/jbc.M005304200. [DOI] [PubMed] [Google Scholar]
- 122.Eyerich S, Eyerich K, Pennino D, Carbone T, Nasorri F, Pallotta S, Cianfarani F, Odorisio T, Traidl-Hoffmann C, Behrendt H, Durham SR, Schmidt-Weber CB, Cavani A. 2009. Th22 cells represent a distinct human T cell subset involved in epidermal immunity and remodeling. J Clin Invest 119:3573–3585. doi: 10.1172/JCI40202. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Lee JS, Cella M, Colonna M. 2012. AHR and the transcriptional regulation of type-17/22 ILC. Front Immunol 3:10. doi: 10.3389/fimmu.2012.00010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Sutton CE, Lalor SJ, Sweeney CM, Brereton CF, Lavelle EC, Mills KH. 2009. Interleukin-1 and IL-23 induce innate IL-17 production from gammadelta T cells, amplifying Th17 responses and autoimmunity. Immunity 31:331–341. doi: 10.1016/j.immuni.2009.08.001. [DOI] [PubMed] [Google Scholar]
- 125.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, Zou QM. 2015. A pro-inflammatory role for Th22 cells in Helicobacter pylori-associated gastritis. Gut 64:1368–1378. doi: 10.1136/gutjnl-2014-307020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Wolk K, Kunz S, Witte E, Friedrich M, Asadullah K, Sabat R. 2004. IL-22 increases the innate immunity of tissues. Immunity 21:241–254. doi: 10.1016/j.immuni.2004.07.007. [DOI] [PubMed] [Google Scholar]
- 127.McGee HM, Schmidt BA, Booth CJ, Yancopoulos GD, Valenzuela DM, Murphy AJ, Stevens S, Flavell RA, Horsley V. 2013. IL-22 promotes fibroblast-mediated wound repair in the skin. J Invest Dermatol 133:1321–1329. doi: 10.1038/jid.2012.463. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Xiang X, Gui H, King NJ, Cole L, Wang H, Xie Q, Bao S. 2012. IL-22 and non-ELR-CXC chemokine expression in chronic hepatitis B virus-infected liver. Immunol Cell Biol 90:611–619. doi: 10.1038/icb.2011.79. [DOI] [PubMed] [Google Scholar]
- 129.Boniface K, Bernard FX, Garcia M, Gurney AL, Lecron JC, Morel F. 2005. IL-22 inhibits epidermal differentiation and induces proinflammatory gene expression and migration of human keratinocytes. J Immunol 174:3695–3702. doi: 10.4049/jimmunol.174.6.3695. [DOI] [PubMed] [Google Scholar]
- 130.Lejeune D, Dumoutier L, Constantinescu S, Kruijer W, Schuringa JJ, Renauld JC. 2002. Interleukin-22 (IL-22) activates the JAK/STAT, ERK, JNK, and p38 MAP kinase pathways in a rat hepatoma cell line. Pathways that are shared with and distinct from IL-10. J Biol Chem 277:33676–33682. doi: 10.1074/jbc.M204204200. [DOI] [PubMed] [Google Scholar]
- 131.Pickert G, Neufert C, Leppkes M, Zheng Y, Wittkopf N, Warntjen M, Lehr HA, Hirth S, Weigmann B, Wirtz S, Ouyang W, Neurath MF, Becker C. 2009. STAT3 links IL-22 signaling in intestinal epithelial cells to mucosal wound healing. J Exp Med 206:1465–1472. doi: 10.1084/jem.20082683. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Zheng Y, Valdez PA, Danilenko DM, Hu Y, Sa SM, Gong Q, Abbas AR, Modrusan Z, Ghilardi N, de Sauvage FJ, Ouyang W. 2008. Interleukin-22 mediates early host defense against attaching and effacing bacterial pathogens. Nat Med 14:282–289. doi: 10.1038/nm1720. [DOI] [PubMed] [Google Scholar]
- 133.Raffatellu M, George MD, Akiyama Y, Hornsby MJ, Nuccio SP, Paixao TA, Butler BP, Chu H, Santos RL, Berger T, Mak TW, Tsolis RM, Bevins CL, Solnick JV, Dandekar S, Baumler AJ. 2009. Lipocalin-2 resistance confers an advantage to Salmonella enterica serotype Typhimurium for growth and survival in the inflamed intestine. Cell Host Microbe 5:476–486. doi: 10.1016/j.chom.2009.03.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Liang SC, Nickerson-Nutter C, Pittman DD, Carrier Y, Goodwin DG, Shields KM, Lambert AJ, Schelling SH, Medley QG, Ma HL, Collins M, Dunussi-Joannopoulos K, Fouser LA. 2010. IL-22 induces an acute-phase response. J Immunol 185:5531–5538. doi: 10.4049/jimmunol.0904091. [DOI] [PubMed] [Google Scholar]
- 135.Wolk K, Witte E, Wallace E, Docke WD, Kunz S, Asadullah K, Volk HD, Sterry W, Sabat R. 2006. IL-22 regulates the expression of genes responsible for antimicrobial defense, cellular differentiation, and mobility in keratinocytes: a potential role in psoriasis. Eur J Immunol 36:1309–1323. doi: 10.1002/eji.200535503. [DOI] [PubMed] [Google Scholar]
- 136.Liu JZ, Pezeshki M, Raffatellu M. 2009. Th17 cytokines and host-pathogen interactions at the mucosa: dichotomies of help and harm. Cytokine 48:156–160. doi: 10.1016/j.cyto.2009.07.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Wozniak KL, Hole CR, Yano J, Fidel PL Jr, Wormley FL Jr.. 2014. Characterization of IL-22 and antimicrobial peptide production in mice protected against pulmonary Cryptococcus neoformans infection. Microbiology 160:1440–1452. doi: 10.1099/mic.0.073445-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Ji Y, Yang X, Li J, Lu Z, Li X, Yu J, Li N. 2014. IL-22 promotes the migration and invasion of gastric cancer cells via IL-22R1/AKT/MMP-9 signaling. Int J Clin Exp Pathol 7:3694–3703. [PMC free article] [PubMed] [Google Scholar]
- 139.Kamradt AE, Greiner M, Ghiara P, Kaufmann SH. 2000. Helicobacter pylori infection in wild-type and cytokine-deficient C57BL/6 and BALB/c mouse mutants. Microbes Infect 2:593–597. doi: 10.1016/S1286-4579(00)00367-1. [DOI] [PubMed] [Google Scholar]
- 140.Li LJ, Gong C, Zhao MH, Feng BS. 2014. Role of interleukin-22 in inflammatory bowel disease. World J Gastroenterol 20:18177–18188. doi: 10.3748/wjg.v20.i48.18177. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.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, Zhuang Y. 2019. Helicobacter pylori-induced matrix metallopeptidase-10 promotes gastric bacterial colonization and gastritis. Sci Adv 5:eaau6547. doi: 10.1126/sciadv.aau6547. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Liao F, Hsu YC, Kuo SH, Yang YC, Chen JP, Hsu PN, Lin CW, Chen LT, Cheng AL, Fann CS, Lin JT, Wu MS. 2014. Genetic polymorphisms and tissue expression of interleukin-22 associated with risk and therapeutic response of gastric mucosa-associated lymphoid tissue lymphoma. Blood Cancer J 4:eXX. doi: 10.1038/bcj.2014.70. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Fukui H, Zhang X, Sun C, Hara K, Kikuchi S, Yamasaki T, Kondo T, Tomita T, Oshima T, Watari J, Imura J, Fujimori T, Sasako M, Miwa H. 2014. IL-22 produced by cancer-associated fibroblasts promotes gastric cancer cell invasion via STAT3 and ERK signaling. Br J Cancer 111:763–771. doi: 10.1038/bjc.2014.336. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Collins PL, Henderson MA, Aune TM. 2012. Lineage-specific adjacent IFNG and IL26 genes share a common distal enhancer element. Genes Immun 13:481–488. doi: 10.1038/gene.2012.22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Wilson NJ, Boniface K, Chan JR, McKenzie BS, Blumenschein WM, Mattson JD, Basham B, Smith K, Chen T, Morel F, Lecron JC, Kastelein RA, Cua DJ, McClanahan TK, Bowman EP, de Waal Malefyt R. 2007. Development, cytokine profile and function of human interleukin 17-producing helper T cells. Nat Immunol 8:950–957. doi: 10.1038/ni1497. [DOI] [PubMed] [Google Scholar]
- 146.Manel N, Unutmaz D, Littman DR. 2008. The differentiation of human T(H)-17 cells requires transforming growth factor-beta and induction of the nuclear receptor RORgammat. Nat Immunol 9:641–649. doi: 10.1038/ni.1610. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Pene J, Chevalier S, Preisser L, Venereau E, Guilleux MH, Ghannam S, Moles JP, Danger Y, Ravon E, Lesaux S, Yssel H, Gascan H. 2008. Chronically inflamed human tissues are infiltrated by highly differentiated Th17 lymphocytes. J Immunol 180:7423–7430. doi: 10.4049/jimmunol.180.11.7423. [DOI] [PubMed] [Google Scholar]
- 148.Meller S, Di Domizio J, Voo KS, Friedrich HC, Chamilos G, Ganguly D, Conrad C, Gregorio J, Le Roy D, Roger T, Ladbury JE, Homey B, Watowich S, Modlin RL, Kontoyiannis DP, Liu YJ, Arold ST, Gilliet M. 2015. T(H)17 cells promote microbial killing and innate immune sensing of DNA via interleukin 26. Nat Immunol 16:970–979. doi: 10.1038/ni.3211. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Corvaisier M, Delneste Y, Jeanvoine H, Preisser L, Blanchard S, Garo E, Hoppe E, Barré B, Audran M, Bouvard B, Saint-André J-P, Jeannin P. 2012. IL-26 is overexpressed in rheumatoid arthritis and induces proinflammatory cytokine production and Th17 cell generation. PLoS Biol 10:e1001395. doi: 10.1371/journal.pbio.1001395. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Dambacher J, Beigel F, Zitzmann K, De Toni EN, Goke B, Diepolder HM, Auernhammer CJ, Brand S. 2009. The role of the novel Th17 cytokine IL-26 in intestinal inflammation. Gut 58:1207–1217. doi: 10.1136/gut.2007.130112. [DOI] [PubMed] [Google Scholar]
- 151.Vandenbroeck K, Cunningham S, Goris A, Alloza I, Heggarty S, Graham C, Bell A, Rooney M. 2003. Polymorphisms in the interferon-gamma/interleukin-26 gene region contribute to sex bias in susceptibility to rheumatoid arthritis. Arthritis Rheum 48:2773–2778. doi: 10.1002/art.11236. [DOI] [PubMed] [Google Scholar]
- 152.Silverberg MS, Cho JH, Rioux JD, McGovern DP, Wu J, Annese V, Achkar JP, Goyette P, Scott R, Xu W, Barmada MM, Klei L, Daly MJ, Abraham C, Bayless TM, Bossa F, Griffiths AM, Ippoliti AF, Lahaie RG, Latiano A, Pare P, Proctor DD, Regueiro MD, Steinhart AH, Targan SR, Schumm LP, Kistner EO, Lee AT, Gregersen PK, Rotter JI, Brant SR, Taylor KD, Roeder K, Duerr RH. 2009. Ulcerative colitis-risk loci on chromosomes 1p36 and 12q15 found by genome-wide association study. Nat Genet 41:216–220. doi: 10.1038/ng.275. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.You W, Tang Q, Zhang C, Wu J, Gu C, Wu Z, Li X. 2013. IL-26 promotes the proliferation and survival of human gastric cancer cells by regulating the balance of STAT1 and STAT3 activation. PLoS One 8:e63588. doi: 10.1371/journal.pone.0063588. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 154.Woetmann A, Alhede M, Dabelsteen S, Bjarnsholt T, Rybtke M, Nastasi C, Krejsgaard T, Andersen MH, Bonefeld CM, Geisler C, Givskov M, Odum N. 2018. Interleukin-26 (IL-26) is a novel anti-microbial peptide produced by T cells in response to staphylococcal enterotoxin. Oncotarget 9:19481–19489. doi: 10.18632/oncotarget.24603. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155.Zhu J, Paul WE. 2010. Heterogeneity and plasticity of T helper cells. Cell Res 20:4–12. doi: 10.1038/cr.2009.138. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Morrison PJ, Bending D, Fouser LA, Wright JF, Stockinger B, Cooke A, Kullberg MC. 2013. Th17-cell plasticity in Helicobacter hepaticus-induced intestinal inflammation. Mucosal Immunol 6:1143–1156. doi: 10.1038/mi.2013.11. [DOI] [PubMed] [Google Scholar]
- 157.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]
- 158.Bagheri N, Azadegan-Dehkordi F, Rahimian G, Rafieian-Kopaei M, Shirzad H. 2016. Role of regulatory T-cells in different clinical expressions of Helicobacter pylori infection. Arch Med Res 47:245–254. doi: 10.1016/j.arcmed.2016.07.013. [DOI] [PubMed] [Google Scholar]
- 159.Chaudhary B, Elkord E. 2016. Regulatory T cells in the tumor microenvironment and cancer progression: role and therapeutic targeting. Vaccines (Basel) 4:E28. doi: 10.3390/vaccines4030028. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160.Halvorsen EC, Mahmoud SM, Bennewith KL. 2014. Emerging roles of regulatory T cells in tumour progression and metastasis. Cancer Metastasis Rev 33:1025–1041. doi: 10.1007/s10555-014-9529-x. [DOI] [PubMed] [Google Scholar]
- 161.Abbas AK, Murphy KM, Sher A. 1996. Functional diversity of helper T lymphocytes. Nature 383:787–793. doi: 10.1038/383787a0. [DOI] [PubMed] [Google Scholar]
- 162.Oertli M, Sundquist M, Hitzler I, Engler DB, Arnold IC, Reuter S, Maxeiner J, Hansson M, Taube C, Quiding-Järbrink M, Müller A. 2012. DC-derived IL-18 drives Treg differentiation, murine Helicobacter pylori-specific immune tolerance, and asthma protection. J Clin Invest 122:1082–1096. doi: 10.1172/JCI61029. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163.Raghavan S, Fredriksson M, Svennerholm AM, Holmgren J, 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. Clin Exp Immunol 132:393–400. doi: 10.1046/j.1365-2249.2003.02177.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164.Gil JH, Seo JW, Cho MS, Ahn JH, Sung HY. 2014. Role of Treg and TH17 cells of the gastric mucosa in children with Helicobacter pylori gastritis. J Pediatr Gastroenterol Nutr 58:245–251. doi: 10.1097/MPG.0000000000000194. [DOI] [PubMed] [Google Scholar]
- 165.Serrano C, Wright SW, Bimczok D, Shaffer CL, Cover TL, Venegas A, Salazar MG, Smythies LE, Harris PR, Smith PD. 2013. Downregulated Th17 responses are associated with reduced gastritis in Helicobacter pylori-infected children. Mucosal Immunol 6:950–959. doi: 10.1038/mi.2012.133. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166.Voo KS, Wang YH, Santori FR, Boggiano C, Wang YH, Arima K, Bover L, Hanabuchi S, Khalili J, Marinova E, Zheng B, Littman DR, Liu YJ. 2009. Identification of IL-17-producing FOXP3+ regulatory T cells in humans. Proc Natl Acad Sci U S A 106:4793–4798. doi: 10.1073/pnas.0900408106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167.Lochner M, Peduto L, Cherrier M, Sawa S, Langa F, Varona R, Riethmacher D, Si-Tahar M, Di Santo JP, Eberl G. 2008. In vivo equilibrium of proinflammatory IL-17+ and regulatory IL-10+ Foxp3+ RORγt+ T cells. J Exp Med 205:1381–1393. doi: 10.1084/jem.20080034. [DOI] [PMC free article] [PubMed] [Google Scholar]