Next Article in Journal
Genetic Inactivation of Free Fatty Acid Receptor 3 Impedes Behavioral Deficits and Pathological Hallmarks in the APPswe Alzheimer’s Disease Mouse Model
Next Article in Special Issue
Accumulation of Deleterious Effects in Gastric Epithelial Cells and Vascular Endothelial Cells In Vitro in the Milieu of Helicobacter pylori Components, 7-Ketocholesterol and Acetylsalicylic Acid
Previous Article in Journal
Expression of Irisin/FNDC5 in Breast Cancer
Previous Article in Special Issue
Epigenetics and Helicobacter pylori
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Helicobacter pylori Pathogen-Associated Molecular Patterns: Friends or Foes?

1
Department of Pharmacy, University of Salerno, 84084 Fisciano, Italy
2
Ph.D. Program in Drug Discovery and Development, University of Salerno, 84084 Fisciano, Italy
*
Authors to whom correspondence should be addressed.
Int. J. Mol. Sci. 2022, 23(7), 3531; https://doi.org/10.3390/ijms23073531
Submission received: 2 March 2022 / Revised: 18 March 2022 / Accepted: 21 March 2022 / Published: 24 March 2022
(This article belongs to the Special Issue Helicobacter: Infection, Diagnosis and Treatment 2.0)

Abstract

:
Microbial infections are sensed by the host immune system by recognizing signature molecules called Pathogen-Associated Molecular Patterns—PAMPs. The binding of these biomolecules to innate immune receptors, called Pattern Recognition Receptors (PRRs), alerts the host cell, activating microbicidal and pro-inflammatory responses. The outcome of the inflammatory cascade depends on the subtle balance between the bacterial burn and the host immune response. The role of PRRs is to promote the clearance of the pathogen and to limit the infection by bumping inflammatory response. However, many bacteria, including Helicobacter pylori, evolved to escape PRRs’ recognition through different camouflages in their molecular pattern. This review examines all the different types of H. pylori PAMPs, their roles during the infection, and the mechanisms they evolved to escape the host recognition.
Keywords:
PAMP; LPS; ADP-heptose; OMV

1. Introduction

In 1989, Janeway proposed the pattern recognition theory defining PAMPs (Pathogen-Associated Molecular Patterns), the microbial components not found in eukaryotic hosts recognized as non-self by several innate immune receptors (Pattern Recognition Receptors: PRRs) during an infection. The host cell, warned of the presence of potentially dangerous microorganisms by the PAMPs, responds by producing inflammatory mediators and recruiting immune cells to fight infection [1].
This conceptual framework for understanding innate immune recognition was later validated experimentally and has become a paradigm [2]. To be recognized by the innate immune system, PAMPs must have three main features. First, within a given class of microorganisms, they should be invariant, so that they can be detected by a limited number of germ-line encoded receptors. Second, they should only be present in microorganisms and not in eukaryotic cells in order to allow for discrimination between self and non-self. Third, they should have essential roles in microbial physiology, so that their detection cannot be easily eliminated via mutation. Indeed, bacterial PAMPs are often cell wall constituents, such as lipopolysaccharide, peptidoglycan, lipoteichoic acids, and cell wall lipoproteins.
Examples of PAMPs of particular importance include lipid A, which is a constituent of the Gram-negative lipopolysaccharide (LPS), recognized by Toll-like receptor 4 (TLR4); flagellin, a protein that polymerizes to form the flagellum, sensed by TLR5; lipoproteins from both Gram-positive and Gram-negative bacteria, recognized by TLR2; bacterial DNA containing CpG motifs that stimulate TLR9; and fragments of bacterial peptidoglycan that are detected in the host cell cytosol by NOD1 and NOD2 (Nucleotide-binding Oligomerization Domain-containing 1 and 2) receptors.
Helicobacter pylori is a highly successful human pathogen infecting approximately 50% of the world’s population [3]. It can successfully colonize the human stomach for a long time, escaping the host immune system and causing a prolonged infection with subsequent chronic gastritis, one of the most common risk factors for gastric malignancy. The pathological outcomes of H. pylori chronic colonization depend on the host–pathogen interactions, which are heavily influenced by bacterial and host genetics, and environmental factors.
The first line of defense for H. pylori is the gastric mucosal layer, localized in the gastric lamina propria, which consists of gastric epithelial and innate immune cells (dendritic, natural killer, mast cells, etc.). These cells are activated during the pre-infection stage of the H. pylori gastric infection, causing a significant rise in the expressions of IL-1, IL-6, and IL-8 in the antrum region and CD4+ and CD8+ T cell recruitment. Immune cells are then prone to secreting cytokines such as interferon γ (IFN-γ) and tumor necrosis factor-α (TNF-α), and interleukins such as IL-1, IL-6, IL-7, IL-8, IL-10, IL-17, and IL-17A [4,5,6]. H. pylori penetration into the epithelial mucus layer induces inflammation and produces a phagocytic-rich environment. These phagocytic cells produce their chemical responses, including reactive oxygen species (ROS) and nitric oxide (NO) [7]. All H. pylori strains are able to detoxify ROS, producing catalase and superoxide dismutase. Furthermore, H. pylori arginase limits NO production by macrophage, neutrophils, and epithelial cells [8,9]. It was demonstrated that non-opsonized H. pylori can resist phagocytosis, targeting the NADPH oxidase and disrupting it; this leads to phagosome dysfunction, since superoxide ions diffuse into the extracellular space rather than being located in the phagosome [10]. Finally, the bacterium manages to survive, inducing mitochondria-dependent apoptosis, particularly of macrophages (Figure 1) [11,12].
The recognition of PAMPs is gained through PRRs; H. pylori has an efficient process to escape those receptors through different camouflages of its molecular pattern. Moreover, PAMPs do not always induce a pro-inflammatory response and/or pathogen clearance but can also dampen immune responses [13]. Here, we summarize some of the most common bacterial PAMPs and how they are modified from H. pylori to make their recognition inefficient.

2. Lipopolysaccharide

LPS consists of three different domains [14]: the more or less acylated and phosphorylated lipid A of the bacterial outer membrane; the core oligosaccharide (inner and outer) linked by 3-deoxy-d-manno-octulosonic acid (Kdo) to lipid A; and the so-called O-antigen or O-specific polysaccharide, with the latter two facing the aqueous environment. Lipopolysaccharides encompassing all three regions are called smooth (S)-form LPS, while LPS lacking the O-antigen are indicated as rough (R)-form LPS or lipooligosaccharide (LOS).
The H. pylori LPS structure plays an essential role in the bacterial immune escape. Compared to other bacterial LPS, such as those of Escherichia coli, H. pylori LPS has ~1000 fold less endotoxicity [15], and this reduction is attributed to three significant modifications of the lipid A. The first modification is a hypo-acylation pattern; indeed, H. pylori Lipid A is tetra-acylated compared to hexa- or penta-acylated chains of other bacteria. Interestingly, H. pylori synthesizes a hexa-acylated lipid A that is subsequently deacylated [16]. Second, hypo-acylated fatty acids have longer carbon chain lengths (18-carbon and 16-carbon chains) compared to the optimal chain lengths (12- and 14-carbon chains) required for robust TLR4 activation [17]. Lastly, H. pylori LPS is hypo-phosphorylated, since phosphate groups from the 1ʹ- and 4ʹ-positions of the lipid A backbone are removed [18]. This modification reduces the binding with cationic antimicrobial peptides (CAMPs), such as polymyxin B, and escapes detection by TLRs [15,19].
The role of TLR4 in immune activation is controversial; the TLR (or TLRs) involved in the residual detection of H. pylori LPS remains a matter of debate. On the one hand, several studies enrolling purified LPS consider TLR4 as a classical LPS sensor [20,21], while on the other hand, further studies suggest that TLR2 is the primary receptor of H. pylori LPS [22,23].
A unique feature of the O-chain of the LPS of most H. pylori strains is the expression of Lewis (Le) antigens, mimicking the ones expressed by gastric epithelium [24]. This camouflage assists bacteria in escaping from the host’s immune responses and may also induce autoimmune disorders, contributing to the severity and chronicity of infection [25].
In most strains, both Lewis X (LeX) and Lewis Y (LeY) can be found in certain regions of the O-antigen. Some strains also display Lewis A (LeA) and B (LeB) antigens or can have alternative O-antigen structures [26]. H. pylori takes advantage of this molecular mimicry, as LeX and LeY interact with the C-type lectin DC-SIGN (Dendritic Cell-Specific Intercellular adhesion molecule-3-Grabbing Non-integrin), leading the immune system to down-regulate the inflammatory response [27].
C-type lectin receptors (CLRs) are members of the PRR family and induce many immune system genes. DC-SIGN is a member of the CLR family and is primarily expressed in DCs. DC-SIGN recognizes predominantly N-linked high-mannose oligosaccharides and branched fucosylated structures and may have two different roles. Fucose residues on H. pylori activate the DC-SIGN, stimulating its response. Fucosylated ligand binding can activate NF-kB, inducing T cell maturation and Th1/Th2 polarization [28]. H. pylori can also target these receptors to limit cell proliferation and LeX/LeY strains can escape all these recognition processes [29].
Li et al. established the first complete LPS biosynthesis pathway using H. pylori strain G27 as a model and redefined the core oligosaccharide domain as a hexasaccharide (Glc-Gal-DD-HepIII-LD-HepII-LD-HepI-KDO), which is decorated with a long O-antigen encompassing the trisaccharide (DD-Hep-Fuc-GlcNAc) termed as Trio; a variable glucan (homopolymer of Glc); a DD-heptane (homopolymer of Hep); and terminal Lewis antigens [30]. H. pylori LPS lacks the canonical inner and outer core organization. Instead, it displays a short core and a longer O-antigen encompassing residues previously assigned as the outer core domain (Figure 2).
Analyzing this pathway in a large panel of H. pylori strains from different ethnic origins, they found that the genes involved in LPS heptane incorporation are lacking in East Asian strains [31]. Given the high gastric cancer (GC) rate in East Asia, this observation opens exciting questions on a potential role of LPS heptane in H. pylori pathogenesis.

3. Metabolites

3.1. HBP and ADP-Heptose

It has recently been demonstrated that heptose metabolites of LPS inner core biosynthesis can be considered novel PAMPs of Gram-negative bacteria as they activate human epithelial cells at early times of infection via the ALPK1-TIFA axis.
The inner core of LPS typically consists of 3-deoxy-D-manno-octulosonic acid and L,D-heptose units. The precursor of the L,D-heptose units is the ADP heptose (ADP-L-glycero-b-D-manno-heptose), synthesized in a five-step pathway starting from D-sedoheptulose 7-phosphate [32]. The latter is converted into D-glycero-a,b-D-manno-heptose-7-phosphate by the sedoheptulose-7-phosphate isomerase GmhA; phosphorylated to form D-glycero-b-D-manno-heptose-1,7-bisphosphate (HBP) by the bifunctional kinase or adenylyltransferase HldE; then dephosphorylated to D-glycero-b-D-manno-heptose-1-phosphate by the D-glycero-b-D-manno-heptose-1,7-bisphosphate-7-phosphatase (GmhB); followed by nucleotide activation by the formation of ADP-D-glycero-b-D-manno-heptose by HldE; and finally, epimerization to ADP heptose by the epimerase HldD (Figure 3). The resulting activated heptose unit is then integrated into the LPS core region.
The intermediate metabolite HBP was first identified as a potent mediator of NF-kB activation by Neisseria species; during the infection, it is released into the cytosol of infected epithelial cells via phagocytosis, activating TRAF-interacting protein with forkhead-associated domain (TIFA) dependent immunity [33].
Subsequently, Stein and co-workers demonstrated that the Cag type-IV secretion system (CagT4SS) is involved in HBP translocation into host cells, inducing the secretion of the pro-inflammatory cytokine IL-8 and activation of NF-kB [34]. Indeed, it was also demonstrated that, in gastric epithelial cells infected with H. pylori, NF-kB activation was stimulated by HBP upon transfection, presumably mimicking HBP translocation to the cytosol via the CagT4SS with the involvement of Alpha-kinase 1 (ALPK1) and TIFA [35].
Pfannuck and co-workers recently found that a derivative of HBP, ADP-heptose, is produced by H. pylori at concentrations more than 10 times higher than HBP. At the same time, it is far more potent, activating ALPK1-TIFA-controlled innate immune responses in epithelial cells at 100-fold lower concentrations [36]. Furthermore, ADP heptose was demonstrated to be a ligand of ALPK1, suggesting that HBP is converted inside the host cell to ADP-heptose-7-phosphate, which can also activate ALPK1, but to a lesser extent [37].
ALPK1 belongs to the family of alpha kinases. These serine/threonine kinases display no homology with conventional kinases in their catalytic domains and are implicated in the apical protein transport to maintain polarity in epithelial cells [38]. ALPK1 was then identified as a PRR in infections by Gram-negative bacteria, since its catalytic activity is essential for the self-oligomerization of TIFA [35,39]. The latter was first described as a TRAF2-binding protein for its ability to activate NF-kB and AP-1 [40]. Through the involvement of the FHA domain and the phosphorylation of threonine at position 9 [41], TIFA undergoes self-oligomerization, thus promoting the oligomerization and ubiquitinylation of TRAF6, leading to the activation of the IKK complex (Figure 3) [42].
The classical activation of NF-kB depends on the formation of oligomeric TIFA-TRAF6 complexes [39], and TAK1 (TGFβ-activated kinase 1) binding to TRAF6 upon H. pylori infection seems to be dependent on TIFA. At the same time, TRAF3, TRAF2, and cIAP1 (cellular inhibitor of apoptosis 1) are probably involved in the formation of TIFAsomes. Therefore, H. pylori infection can induce different TIFAsome complexes that activate the classical and alternative NF-kB pathways [43]. This dual possibility could trigger various cellular events, leading to the induction of inflammatory cytokines by the classical NF-kB pathway and more complex physiological changes due to the alternative NF-kB pathway that can lead to gastric pathologies [44].
Hence, H. pylori heptose metabolites play a significant role in early phagocytic cell activation, including NF-kB activation and IL-8 production. Bacterial purified lysates and pure ADP-heptose on its own, in the absence of other bacterial PAMPs, are able to strongly activate Thp-1 cells and human primary monocytes/macrophages, differentiating monocytes into macrophages of predominantly M1 type, and this activation was significantly reduced upon TIFA knock-down. In this cell model, the CagT4SS was less necessary than in epithelial cells. In contrast, active heptose biosynthesis or pure ADP-heptose was required and sufficient for an early innate response and NF-kB activation [45].

3.2. Cholesterol Derivatives

H. pylori produces different lipids, mainly various cholesterol metabolites [46]. Indeed, H. pylori extracts cholesterol from the host and converts it to specific cholesteryl glucosides, such as cholesteryl acyl α-glucosides (αCAGs) [47,48]. One of the hypothesized roles of these cholesterol metabolites is to suppress host immunity [49], although their immunomodulatory properties and host pattern recognition receptors have not yet been determined.
Recently, αCAG and cholesteryl phosphatidyl α-glucoside (αCPG) were identified as noncanonical ligands for Mincle (Clec4e) and DCAR (Clec4b1). During chronic infection, H. pylori-specific T cell responses and gastritis were ameliorated in Mincle-deficient mice, although bacterial burdens remained unchanged. Furthermore, a mutant H. pylori strain lacking αCAG and αCPG exhibited an impaired ability to cause gastritis. Thus, H. pylori-specific modification of host cholesterol plays a pathophysiological role that exacerbates gastric inflammation by triggering C-type lectin receptors [50].

4. Bacterial Proteins and Peptides

4.1. Flagellin

The natural ligand of TLR5 is flagellin, specifically the highly conserved N-terminus of the D1 domain [51]. Since H. pylori is a flagellated bacterium, TLR5, expressed in the gastric epithelium, should be able to recognize this pathogen and to mediate the pro-inflammatory signaling cascades. However, H. pylori flagellin is not recognized by TLR5 [52] due to a mutation in the conserved domain of FlaA. This mutation, which occurs in the D0-D1 domain between amino acids 89 and 96, masks H. pylori flagella from TLR5 binding and prevents its activation. The importance of these residues was demonstrated by Andersen-Nissen and co-workers, who substituted them into the corresponding region of the Salmonella enterica serovar Typhimurium FliC, making this flagellin unable to activate TLR5 similar to that of H. pylori [53]. The low intrinsic activity of its mutated FlaA confers to H. pylori the opportunity to avoid permanent TLR5 activation. These data suggest an essential role for H. pylori FlaA in maintaining persistence within the gastric niche by dampening the innate immune response. Bacterium persistence is also due to the formation of H. pylori aggregates in the mucus layer, which do not affect flaA gene transcription [54].
Indeed, H. pylori evolved a CagT4SS-dependent but flagellin-independent ability to activate TLR5. CagY and CagL, exposed at the tip of the secretion system, can directly bind to TLR5. Since the T4SS-pilus is induced upon host cell contact, the TLR5 activation can be turned on and off by exposing or hiding CagY and CagL, deregulating or hyper-regulating host immunity [55,56].
Recently, new polymorphisms of TLR5 have been associated with a high risk for gastric carcinoma consequent to H. pylori infection, likely due to an acquired ability to recognize H. pylori flagellin [57].

4.2. Hp(2-20) Peptide

Formyl Peptide Receptors (FPRs), comprising FPR1, FPR2, and FPR3, can be considered another class of PRRs. They are highly expressed by innate immune cells but are also presented by epithelial cells of the mucosal systems, including the gastrointestinal tract [58]. FPRs were discovered as receptors with a high affinity for N-formylated peptides, either derived from bacterial signal peptides or released from mitochondria of damaged host cells. Indeed, these receptors interact with several different ligands other than the N-formyl-methionine-leucyl-phenylalanine (fMLF), the prototype of N-formylated peptides [59].
Hp(2-20) is a non-formylated, cecropin-like peptide released during H. pylori growth with a high affinity for formyl peptide receptor like-1 (FPR2). Hp(2-20) shows different functional features. When H. pylori colonizes the human stomach, Hp(2-20) contributes to activating the innate immune response by interacting with the FPR2 expressed on gastric epithelial cells. Indeed, this interaction favors gastric mucosa inflammation by initiating a cell signaling cascade, which leads to cytokine release, recruitment of inflammatory cells, and stimulation of NADPH oxidase-dependent superoxide generation [60]. Hp(2-20) also stimulates gastric epithelial cell migration and proliferation, inducing the expression of vascular endothelial growth factor (VEGF), which aids in repairing the gastric mucosa after induced injury [61]. These events favor the host defense and the restoration of homeostasis. On the other hand, Hp(2-20) is also an antimicrobial peptide (AMP), which plays an essential role in H. pylori gastric mucosa persistence by acting as a bactericidal molecule, thus conferring an advantage over other microorganisms [62].

5. Extracellular DNA and RNA

During microbial infection, DNA can be actively secreted or released due to degradation from invading microbes or damaged host cells. TLR9, an endosome-bound transmembrane receptor, detects these aberrant DNAs and promotes an immune response [63]. TLR9 was initially reported to recognize pathogenic DNAs based upon the presence of hypo-methylated CpG DNA motifs; however, accumulating evidence suggests that TLR9 can also detect DNA in a sequence-independent manner by recognition of the saccharide backbone [64,65]. Since TLR9 is constrained to the endosome, cells of the immune system must internalize pathogens or pathogenic DNA before TLR9 can detect it. Most immune cells accomplish this task through receptor-mediated endocytosis in response to scavenger receptor binding, phagocytosis of complement-mediated opsonized material, or Fc receptor-mediated uptake of antibody opsonized material or through a combination of these processes [66].
Regarding the gastric compartment, the expression of this receptor differs between healthy individuals and individuals who are infected with H. pylori. TLR9 is located in the apical compartment of the gastric epithelial cells in healthy controls and in the basolateral compartment in individuals with infections [67]. Additionally, it seems mandatory for TLR9 activation that H. pylori translocates its DNA into the cells by cancer-associated CagT4SS, allowing for its engagement by the receptor [68].
In the context of dendritic cells, the scenario changes, as the intracellular delivery of H. pylori DNA by lipofection efficiently activates endosomal localized TLR9, promoting an anti-inflammatory rather than a pro-inflammatory response [69]. In the same way, in the early stages of a mouse model infection, TLR9 activation induced an anti-inflammatory cascade [13]. H. pylori colonization is inversely correlated with the risk of developing inflammatory bowel diseases (IBD), and this seems to be caused by a specific immunoregulatory sequence (TTTAGGG) unique to the H. pylori genome. Therefore, H. pylori DNA can be used therapeutically to treat experimentally induced IBD in mice [69,70].
Therefore, the hypothetical scenario is that H. pylori may utilize TLR9 signaling to dampen the inflammatory response during the acute phase to establish infection; however, at a certain point, when cells have lost their polarity due to inflammatory micro-environment, TLR9 may increase pro-inflammatory cascades and further favor the progression toward gastric cancer.
While H. pylori DNA is sensed by TLR9, the detection of the bacterial RNA in dendritic cells seems to be mediated by endosomal localized TLR8 and by a cytoplasmic nucleic acid sensor, RIG-I, which belongs to the RIG-like helicase receptor family (RLR). RIG-I is able to recognize 5ʹ triphosphorylated H. pylori RNA and to promote the following IRF3- and IRF7-dependent induction of type I interferons (IFN α and β) by dendritic cells. It is not yet clear whether H. pylori–RNA sensing has mainly pro-inflammatory or anti-inflammatory effects [71].

6. Outer Membrane Vesicles

Gram-negative cell surfaces constantly release outer membrane vesicles (OMVs), which contain different components with various functions: increasing virulence, mediating bacterial cell–cell communication, modulating host immune response, and more.
The vesicle diameter ranges between 10 and 300 nm, consisting of lipids, proteins, LPS, phospholipids, DNA, RNA, proteins, inner membrane (IM), periplasm, outer membrane (OM), and other molecules. Several studies have provided that OMVs have opposite effects, both harmful and defensive [72].
Since they deliver many pathogenic bacterial proteins, they induce pro-inflammatory responses in the host. Indeed, OMVs from the Gram-negative pathogens H. pylori, Neisseria, Pseudomonas, Campylobacter, and Vibrio promote the secretion of interleukin-8 (IL-8) by non-phagocytic epithelial cells [73].
H. pylori takes advantage of OMVs to cross the cellular barrier and efficiently translocate antigens through the gastric mucosa up to immune cells in tissues or even to circulation. Blebbing of H. pylori OM was detected in bacteria cultures during the late stationary growth phase and biofilm formation and in gastric biopsies of individuals with infection [74,75].
Regarding their content, Olofsson and co-workers showed that vesicles of this bacterium contain OM but not IM molecules [76]. The content of OMVs might vary among H. pylori strains. H. pylori OMVs contain different phospholipids: phosphatidylglycerol (PG); phosphatidylethanolamine (PE); lyso PE (LPE); phosphatidylcholine (PC); lyso PC (LPC); and cardiolipin [76]. LPS is another critical component of H. pylori OMVs; in particular, the O-chain of H. pylori LPS displays LewisXY antigens in iron-reach conditions, whereas the presence of LewisY antigens is reduced in iron-limited conditions [77].
Recent proteomic analyses of small and large OMVs revealed that smaller OMVs contained significantly fewer proteins than larger ones. Moreover, larger H. pylori OMVs contain bacterial adhesion proteins absent from smaller OMVs, which may facilitate their entry into host cells via receptor-mediated endocytosis. Twenty-four proteins were found to be common to both small and large OMVs; these were mainly associated with virulence, including vacuolating cytotoxin (VacA); CagA; blood group antigen-binding adhesin (BabA); sialic acid-binding adhesin (SabA); outer inflammatory protein A (OipA); H. pylori neutrophil-activating protein (HP-NAP); adherence associated lipoprotein (AlpA); urease; and catalase (KatA) [9,78].
Regarding internalization mechanism, H. pylori OMVs can enter epithelial cells via macropinocytosis, clathrin, and caveolin-dependent endocytosis. Indeed, smaller OMVs with sizes ranging from 20 to 100 nm preferentially use caveolin-mediated endocytosis. In contrast, larger OMVs ranging between 90 and 450 nm use macropinocytosis and endocytosis [78].
One of the first PRRs found to become activated on H. pylori infection was NOD1, able to detect H. pylori peptidoglycan. Initially, it seemed that peptidoglycan could be delivered into the cytoplasm of host epithelial cells only by H. pylori strains with a functional CagT4SS [79]. It is now clear that peptidoglycan can also be delivered to NOD1 from outer-membrane vesicles (OMVs) of Cag PAI-negative strains of H. pylori. Intragastric delivery of OMVs in mice induces innate and adaptive immune responses through a NOD1-dependent but TLR-independent mechanism [80]. The delivery of peptidoglycan by both OMVs and the T4SS occurs at cholesterol-rich lipid rafts [80,81].
NOD1 activates NF-kB signaling pathways and the transcription factor AP1 via ERK- and p38-dependent pathways [82]. A direct effect of NOD1 signaling activation in gastric epithelial cells is the production of the antimicrobial peptide β-defensin 2, with consequent H. pylori killing [83]. It has recently been described an alternative NOD1-dependent signaling pathway, which activates the IRF3 and IRF7 transcription factors suggested to induce type I IFNs essential for H. pylori-specific inflammatory response [84].
Hence, H. pylori OMVs can activate NF-kB, MAPK, and ERK pathways, thus modulating the expression of genes involved in the inflammatory response, proliferation, and carcinogenesis [85]. Moreover, it was shown that H. pylori OMVs can induce IL-8 production by gastric epithelial cells independently of VacA [86].
This pro-inflammatory potential suggests that they can participate in the carcinogenesis process. This hypothesis was assessed by the observation that H. pylori OMVs induce inflammation in a mice model of gastric cancer and that their presence is significantly higher in the gastric juices of gastric cancer patients than in normal controls [87].
AGS human gastric epithelial cells treated with H. pylori OMVs showed increased micronuclei formation, alteration in iron metabolism, and glutathione loss [88].
However, some H. pylori OMV components, such as LPS, may stimulate opposite outcomes. The endotoxic activity of H. pylori LPS is lower than LPS from other bacteria due to the different structure of lipid A, as already discussed. OMVs decorated with H. pylori LPS bearing Lewis antigens in O-chains may chronically stimulate the host immune cells to produce autoantibodies, playing an essential role in autoimmune processes related to H. pylori infection [89].

7. Conclusions

H. pylori, like all Gram-negative bacteria, stimulates the innate immune response at the onset of the infection by PAMP signals detected by host PRRs. Many of these patterns (LPS, flagellin) have been extensively analyzed in Helicobacter pylori and have shown structural features that differentiate them from other bacteria’s more efficient systems. These mutated H. pylori PAMPs are less recognized by the host receptors, contributing to the establishment of a silent and persistent infection. Some others (ADP-heptose, nucleic acids, OMVs) are not yet fully explored and deserve a more detailed characterization in order to definitely assess their role during the infection: host friends or foes?

Author Contributions

Conceptualization, D.E., A.T.; writing—original draft preparation, D.E., F.M., A.V.; writing—review and editing, A.T., A.P. (Amalia Porta); visualization, A.P. (Amalia Porta), A.T.; supervision, A.P. (Antonello Petrella); funding acquisition, A.P. (Amalia Porta), A.T. All authors have read and agreed to the published version of the manuscript.

Funding

Project GENOBU, PON01_00486, D.M. 0001332.07-08-2020.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Janeway, C.A., Jr. Approaching the asymptote? Evolution and revolution in immunology. Cold Spring Harb. Symp. Quant. Biol. 1989, 54 Pt 1, 1–13. [Google Scholar] [CrossRef] [PubMed]
  2. Medzhitov, R. Pattern recognition theory and the launch of modern innate immunity. J. Immunol. 2013, 191, 4473–4474. [Google Scholar] [CrossRef] [PubMed]
  3. Mezmale, L.; Coelho, L.G.; Bordin, D.; Leja, M. Review: Epidemiology of Helicobacter pylori. Helicobacter 2020, 25 (Suppl. S1), e12734. [Google Scholar] [CrossRef] [PubMed]
  4. Algood, H.M.; Cover, T.L. Helicobacter pylori persistence: An overview of interactions between H. pylori and host immune defenses. Clin. Microbiol. Rev. 2006, 19, 597–613. [Google Scholar] [CrossRef] [Green Version]
  5. Bagheri, N.; Azadegan-Dehkordi, F.; Shirzad, H.; Rafieian-Kopaei, M.; Rahimian, G.; Razavi, A. The biological functions of IL-17 in different clinical expressions of Helicobacter pylori-infection. Microb. Pathog. 2015, 81, 33–38. [Google Scholar] [CrossRef]
  6. Adamsson, J.; Ottsjö, L.S.; Lundin, S.B.; Svennerholm, A.M.; Raghavan, S. Gastric expression of IL-17A and IFNγ in Helicobacter pylori infected individuals is related to symptoms. Cytokine 2017, 99, 30–34. [Google Scholar] [CrossRef]
  7. Abadi, A.T.B. Strategies used by Helicobacter pylori to establish persistent infection. World J. Gastroenterol. 2017, 23, 2870–2882. [Google Scholar] [CrossRef]
  8. Gobert, A.P.; McGee, D.J.; Akhtar, M.; Mendz, G.L.; Newton, J.C.; Cheng, Y.; Mobley, H.L.; Wilson, K.T. Helicobacter pylori arginase inhibits nitric oxide production by eukaryotic cells: A strategy for bacterial survival. Proc. Natl. Acad. Sci. USA 2001, 98, 13844–13849. [Google Scholar] [CrossRef] [Green Version]
  9. Lekmeechai, S.; Su, Y.C.; Brant, M.; Alvarado-Kristensson, M.; Vallström, A.; Obi, I.; Arnqvist, A.; Riesbeck, K. Helicobacter pylori outer membrane vesicles protect the pathogen from reactive oxygen species of the respiratory burst. Front. Microbiol. 2018, 9, 1837. [Google Scholar] [CrossRef] [Green Version]
  10. Nguyen, G.T.; Green, E.R.; Mecsas, J. Neutrophils to the ROScue: Mechanisms of NADPH oxidase activation and bacterial resistance. Front. Cell Infect. Microbiol. 2017, 7, 373. [Google Scholar] [CrossRef]
  11. Chaturvedi, R.; Cheng, Y.; Asim, M.; Bussière, F.I.; Xu, H.; Gobert, A.P.; Hacker, A.; Casero, R.A., Jr.; Wilson, K.T. Induction of polyamine oxidase 1 by Helicobacter pylori causes macrophage apoptosis by hydrogen peroxide release and mitochondrial membrane depolarization. J. Biol. Chem. 2004, 279, 40161–40173. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Calvino-Fernández, M.; Benito-Martínez, S.; Parra-Cid, T. Oxidative stress by Helicobacter pylori causes apoptosis through mitochondrial pathway in gastric epithelial cells. Apoptosis 2008, 13, 1267–1280. [Google Scholar] [CrossRef] [PubMed]
  13. Otani, K.; Tanigawa, T.; Watanabe, T.; Nadatani, Y.; Sogawa, M.; Yamagami, H.; Shiba, M.; Watanabe, K.; Tominaga, K.; Fujiwara, Y.; et al. Toll-like receptor 9 signaling has anti-inflammatory effects on the early phase of Helicobacter pylori-induced gastritis. Biochem. Biophys. Res. Commun. 2012, 426, 342–349. [Google Scholar] [CrossRef] [PubMed]
  14. Caroff, M.; Novikov, A. LPS Structure, Function, and Heterogeneity. In Endotoxin Detection and Control in Pharma, Limulus, and Mammalian Systems; Williams, K., Ed.; Springer: Cham, Switzerland, 2019; pp. 53–93. [Google Scholar] [CrossRef]
  15. Needham, B.D.; Trent, M.S. Fortifying the barrier: The impact of lipid A remodelling on bacterial pathogenesis. Nat. Rev. Microbiol. 2013, 11, 467–481. [Google Scholar] [CrossRef] [PubMed]
  16. Stead, C.M.; Beasley, A.; Cotter, R.J.; Trent, M.S. Deciphering the unusual acylation pattern of Helicobacter pylori lipid A. J. Bacteriol. 2008, 190, 7012–7021. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Facchini, F.A.; Zaffaroni, L.; Minotti, A.; Rapisarda, S.; Calabrese, V.; Forcella, M.; Fusi, P.; Airoldi, C.; Ciaramelli, C.; Billod, J.M.; et al. Structure-Activity Relationship in Monosaccharide-Based Toll-Like Receptor 4 (TLR4) Antagonists. J. Med. Chem. 2018, 61, 2895–2909. [Google Scholar] [CrossRef] [Green Version]
  18. Tran, A.X.; Whittimore, J.D.; Wyrick, P.B.; McGrath, S.C.; Cotter, R.J.; Trent, M.S. The lipid A 1-phosphatase of Helicobacter pylori is required for resistance to the antimicrobial peptide polymyxin. J. Bacteriol. 2006, 188, 4531–4541. [Google Scholar] [CrossRef] [Green Version]
  19. Cullen, T.W.; Giles, D.K.; Wolf, L.N.; Ecobichon, C.; Boneca, I.G.; Trent, M.S. Helicobacter pylori versus the host: Remodeling of the bacterial outer membrane is required for survival in the gastric mucosa. PLoS Pathog. 2011, 7, e1002454. [Google Scholar] [CrossRef] [Green Version]
  20. Ishihara, S.; Rumi, M.A.K.; Kadowaki, Y.; Ortega-Cava, C.F.; Yuki, T.; Yoshino, N.; Miyaoka, Y.; Kazumori, H.; Ishimura, N.; Amano, Y.; et al. Essential role of MD-2 in TLR4-dependent signaling during Helicobacter pylori-associated gastritis. J. Immunol. 2004, 173, 1406–1416. [Google Scholar] [CrossRef] [Green Version]
  21. Sangwan, V.; Al-Marzouki, L.; Pal, S.; Stavrakos, V.; Alzahrani, M.; Antonatos, D.; Nevo, Y.; Camilleri-Broët, S.; Rayes, R.; Bourdeau, F.; et al. Inhibition of LPS-mediated TLR4 activation abrogates gastric adenocarcinoma-associated peritoneal metastasis. Clin. Exp. Metastasis 2021. [Google Scholar] [CrossRef]
  22. Yokota, S.; Ohnishi, T.; Muroi, M.; Tanamoto, K.; Fujii, N.; Amano, K. Highly-purified Helicobacter pylori LPS preparations induce weak inflammatory reactions and utilize Toll-like receptor 2 complex but not Toll-like receptor 4 complex. FEMS Immunol. Med. Microbiol. 2007, 51, 140–148. [Google Scholar] [CrossRef] [Green Version]
  23. Smith, S.M.; Moran, A.P.; Duggan, S.P.; Ahmed, S.E.; Mohamed, A.S.; Windle, H.J.; O’Neill, L.A.; Kelleher, D.P. Tribbles 3: A novel regulator of TLR2-mediated signaling in response to Helicobacter pylori lipopolysaccharide. J. Immunol. 2011, 186, 2462–2471. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Monteiro, M.A. Helicobacter pylori: A wolf in sheep’s clothing: The glycotype families of Helicobacter pylori lipopolysaccharides expressing histo-blood groups: Structure, biosynthesis, and role in pathogenesis. Adv. Carbohydr. Chem. Biochem. 2001, 57, 99–158. [Google Scholar] [CrossRef] [PubMed]
  25. Moran, A.P.; Prendergast, M.M. Molecular mimicry in Campylobacter jejuni and Helicobacter pylori lipopolysaccharides: Contribution of gastrointestinal infections to autoimmunity. J. Autoimmun. 2001, 16, 241–256. [Google Scholar] [CrossRef] [PubMed]
  26. Moran, A.P. Relevance of fucosylation and Lewis antigen expression in the bacterial gastroduodenal pathogen Helicobacter pylori. Carbohydr. Res. 2008, 343, 1952–1965. [Google Scholar] [CrossRef]
  27. Bergman, M.P.; Engering, A.; Smits, H.H.; van Vliet, S.J.; van Bodegraven, A.A.; Wirth, H.P.; Kapsenberg, M.L.; Vandenbroucke-Grauls, C.M.J.E.; van Kooyk, Y.; Appelmelk, B.J. Helicobacter pylori modulates the T helper cell 1/T helper cell 2 balance through phase-variable interaction between lipopolysaccharide and DC-SIGN. J. Exp. Med. 2004, 200, 979–990. [Google Scholar] [CrossRef]
  28. Lina, T.T.; Alzahrani, S.; Gonzalez, J.; Pinchuk, I.V.; Beswick, E.J.; Reyes, V.E. Immune evasion strategies used by Helicobacter pylori. World J. Gastroenterol. 2014, 20, 12753–12766. [Google Scholar] [CrossRef]
  29. Miszczyk, E.; Rudnicka, K.; Moran, A.P.; Fol, M.; Kowalewicz-Kulbat, M.; Druszczyńska, M.; Matusiak, A.; Walencka, M.; Rudnicka, W.; Chmiela, M. Interaction of Helicobacter pylori with C-type lectin dendritic cell-specific ICAM grabbing nonintegrin. J. Biomed. Biotechnol. 2012, 2012, 206463. [Google Scholar] [CrossRef] [Green Version]
  30. Li, H.; Yang, T.; Liao, T.; Debowski, A.W.; Nilsson, H.O.; Fulurija, A.; Haslam, S.M.; Mulloy, B.; Dell, A.; Stubbs, K.A.; et al. The redefinition of Helicobacter pylori lipopolysaccharide O-antigen and core-oligosaccharide domains. PLoS Pathog. 2017, 13, e1006280. [Google Scholar] [CrossRef]
  31. Li, H.; Marceau, M.; Yang, T.; Liao, T.; Tang, X.; Hu, R.; Xie, Y.; Tang, H.; Tay, A.; Shi, Y.; et al. East-Asian Helicobacter pylori strains synthesize heptan-deficient lipopolysaccharide. PLoS Genet. 2019, 15, e1008497. [Google Scholar] [CrossRef]
  32. Kneidinger, B.; Marolda, C.; Graninger, M.; Zamyatina, A.; McArthur, F.; Kosma, P.; Valvano, M.A.; Messner, P. Biosynthesis pathway of ADP-L-glycero-beta-D-manno-heptose in Escherichia coli. J. Bacteriol. 2002, 184, 363–369. [Google Scholar] [CrossRef] [Green Version]
  33. Gaudet, R.G.; Sintsova, A.; Buckwalter, C.M.; Leung, N.; Cochrane, A.; Li, J.; Cox, A.D.; Moffat, J.; Gray-Owen, S.D. INNATE IMMUNITY. Cytosolic detection of the bacterial metabolite HBP activates TIFA-dependent innate immunity. Science 2015, 348, 1251–1255. [Google Scholar] [CrossRef] [PubMed]
  34. Stein, S.C.; Faber, E.; Bats, S.H.; Murillo, T.; Speidel, Y.; Coombs, N.; Josenhans, C. Helicobacter pylori modulates host cell responses by CagT4SS-dependent translocation of an intermediate metabolite of LPS inner core heptose biosynthesis. PLoS Pathog. 2017, 13, e1006514. [Google Scholar] [CrossRef] [Green Version]
  35. Zimmermann, S.; Pfannkuch, L.; Al-Zeer, M.A.; Bartfeld, S.; Koch, M.; Liu, J.; Rechner, C.; Soerensen, M.; Sokolova, O.; Zamyatina, A.; et al. ALPK1- and TIFA-Dependent Innate Immune Response Triggered by the Helicobacter pylori Type IV Secretion System. Cell. Rep. 2017, 20, 2384–2395. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Pfannkuch, L.; Hurwitz, R.; Traulsen, J.; Sigulla, J.; Poeschke, M.; Matzner, L.; Kosma, P.; Schmid, M.; Meyer, T.F. ADP heptose, a novel pathogen-associated molecular pattern identified in Helicobacter pylori. FASEB J. 2019, 33, 9087–9099. [Google Scholar] [CrossRef] [Green Version]
  37. Zhou, P.; She, Y.; Dong, N.; Li, P.; He, H.; Borio, A.; Wu, Q.; Lu, S.; Ding, X.; Cao, Y.; et al. Alpha-kinase 1 is a cytosolic innate immune receptor for bacterial ADP-heptose. Nature 2018, 561, 122–126. [Google Scholar] [CrossRef]
  38. Heine, M.; Cramm-Behrens, C.I.; Ansari, A.; Chu, H.P.; Ryazanov, A.G.; Naim, H.Y.; Jacob, R. Alpha-kinase 1, a new component in apical protein transport. J. Biol. Chem. 2005, 280, 25637–25643. [Google Scholar] [CrossRef] [Green Version]
  39. Milivojevic, M.; Dangeard, A.S.; Kasper, C.A.; Tschon, T.; Emmenlauer, M.; Pique, C.; Schnupf, P.; Guignot, J.; Arrieumerlou, C. ALPK1 controls TIFA/TRAF6-dependent innate immunity against heptose-1,7-bisphosphate of gram-negative bacteria. PLoS Pathog. 2017, 13, e1006224. [Google Scholar] [CrossRef]
  40. Kanamori, M.; Suzuki, H.; Saito, R.; Muramatsu, M.; Hayashizaki, Y. T2BP, a novel TRAF2 binding protein, can activate NF-kappaB and AP-1 without TNF stimulation. Biochem. Biophys. Res. Commun. 2002, 290, 1108–1113. [Google Scholar] [CrossRef]
  41. Huang, C.C.F.; Weng, J.H.; Wei, T.Y.W.; Wu, P.Y.G.; Hsu, P.H.; Chen, Y.H.; Wang, S.C.; Qin, D.; Hung, C.C.; Chen, S.T.; et al. Intermolecular binding between TIFA-FHA and TIFA-pT mediates tumor necrosis factor alpha stimulation and NF-κB activation. Mol. Cell. Biol. 2012, 32, 2664–2673. [Google Scholar] [CrossRef] [Green Version]
  42. Ea, C.K.; Sun, L.; Inoue, J.; Chen, Z.J. TIFA activates IkappaB kinase (IKK) by promoting oligomerization and ubiquitination of TRAF6. Proc. Natl. Acad. Sci. USA. 2004, 101, 15318–15323. [Google Scholar] [CrossRef] [Green Version]
  43. Maubach, G.; Lim, M.C.C.; Sokolova, O.; Backert, S.; Meyer, T.F.; Naumann, M. TIFA has dual functions in Helicobacter pylori-induced classical and alternative NF-κB pathways. EMBO Rep. 2021, 22, e52878. [Google Scholar] [CrossRef] [PubMed]
  44. Merga, Y.J.; O’Hara, A.; Burkitt, M.D.; Duckworth, C.A.; Probert, C.S.; Campbell, B.J.; Pritchard, D.M. Importance of the alternative NF-κB activation pathway in inflammation-associated gastrointestinal carcinogenesis. Am. J. Physiol. Gastrointest. Liver. Physiol. 2016, 310, G1081–G1090. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Faass, L.; Stein, S.C.; Hauke, M.; Gapp, M.; Albanese, M.; Josenhans, C. Contribution of heptose metabolites and the cag pathogenicity island to the activation of monocytes/macrophages by Helicobacter pylori. Front. Immunol. 2021, 12, 632154. [Google Scholar] [CrossRef] [PubMed]
  46. Jan, H.M.; Chen, Y.C.; Shih, Y.Y.; Huang, Y.C.; Tu, Z.; Ingle, A.B.; Liu, S.W.; Wu, M.S.; Gervay-Hague, J.; Mong, K.K.T.; et al. Metabolic labelling of cholesteryl glucosides in Helicobacter pylori reveals how the uptake of human lipids enhances bacterial virulence. Chem. Sci. 2016, 7, 6208–6216. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Haque, M.; Hirai, Y.; Yokota, K.; Mori, N.; Jahan, I.; Ito, H.; Hotta, H.; Yano, I.; Kanemasa, Y.; Oguma, K. Lipid profile of Helicobacter spp.: Presence of cholesteryl glucoside as a characteristic feature. J. Bacteriol. 1996, 178, 2065–2070. [Google Scholar] [CrossRef] [Green Version]
  48. Lebrun, A.H.; Wunder, C.; Hildebrand, J.; Churin, Y.; Zähringer, U.; Lindner, B.; Meyer, T.F.; Heinz, E.; Warnecke, D. Cloning of a cholesterol-alpha-glucosyltransferase from Helicobacter pylori. J. Biol. Chem. 2006, 281, 27765–27772. [Google Scholar] [CrossRef] [Green Version]
  49. Wunder, C.; Churin, Y.; Winau, F.; Warnecke, D.; Vieth, M.; Lindner, B.; Zähringer, U.; Mollenkopf, H.J.; Heinz, E.; Meyer, T.F. Cholesterol glucosylation promotes immune evasion by Helicobacter pylori. Nat. Med. 2006, 12, 1030–1038. [Google Scholar] [CrossRef]
  50. Nagata, M.; Toyonaga, K.; Ishikawa, E.; Haji, S.; Okahashi, N.; Takahashi, M.; Izumi, Y.; Imamura, A.; Takato, K.; Ishida, H.; et al. Helicobacter pylori metabolites exacerbate gastritis through C-type lectin receptors. J. Exp. Med. 2021, 218, e20200815. [Google Scholar] [CrossRef]
  51. Smith, K.D.; Andersen-Nissen, E.; Hayashi, F.; Strobe, K.; Bergman, M.A.; Barrett, S.L.R.; Cookson, B.T.; Aderem, A. Toll-like receptor 5 recognizes a conserved site on flagellin required for protofilament formation and bacterial motility. Nat. Immunol. 2003, 4, 1247–1253. [Google Scholar] [CrossRef]
  52. Gewirtz, A.T.; Yu, Y.; Krishna, U.S.; Israel, D.A.; Lyons, S.L.; Peek, R.M., Jr. Helicobacter pylori flagellin evades toll-like receptor 5-mediated innate immunity. J. Infect. Dis. 2004, 189, 1914–1920. [Google Scholar] [CrossRef] [Green Version]
  53. Andersen-Nissen, E.; Smith, K.D.; Strobe, K.L.; Barrett, S.L.R.; Cookson, B.T.; Logan, S.M.; Aderem, A. Evasion of Toll-like receptor 5 by flagellated bacteria. Proc. Natl. Acad. Sci. USA 2005, 102, 9247–9252. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Eletto, D.; Vllahu, M.; Mentucci, F.; Del Gaudio, P.; Petrella, A.; Porta, A.; Tosco, A. TFF1 induces aggregation and reduces motility of Helicobacter pylori. Int. J. Mol. Sci. 2021, 22, 1851. [Google Scholar] [CrossRef] [PubMed]
  55. Tegtmeyer, N.; Neddermann, M.; Lind, J.; Pachathundikandi, S.K.; Sharafutdinov, I.; Gutiérrez-Escobar, A.J.; Brönstrup, M.; Tegge, W.; Hong, M.; Rohde, M.; et al. Toll-like Receptor 5 activation by the cagy repeat domains of Helicobacter pylori. Cell Rep. 2020, 32, 108159. [Google Scholar] [CrossRef] [PubMed]
  56. Pachathundikandi, S.K.; Tegtmeyer, N.; Arnold, I.C.; Lind, J.; Neddermann, M.; Falkeis-Veits, C.; Chattopadhyay, S.; Brönstrup, M.; Tegge, W.; Hong, M.; et al. T4SS-dependent TLR5 activation by Helicobacter pylori infection. Nat. Commun. 2019, 10, 5717. [Google Scholar] [CrossRef] [Green Version]
  57. Xu, T.; Fu, D.; Ren, Y.; Dai, Y.; Lin, J.; Tang, L.; Ji, J. Genetic variations of TLR5 gene interacted with Helicobacter pylori infection among carcinogenesis of gastric cancer. Oncotarget 2017, 8, 31016–31022. [Google Scholar] [CrossRef] [Green Version]
  58. Jeong, Y.S.; Bae, Y.S. Formyl peptide receptors in the mucosal immune system. Exp. Mol. Med. 2020, 52, 1694–1704. [Google Scholar] [CrossRef]
  59. He, H.Q.; Ye, R.D. The Formyl Peptide Receptors: Diversity of Ligands and Mechanism for Recognition. Molecules 2017, 22, 455. [Google Scholar] [CrossRef]
  60. Bylund, J.; Christophe, T.; Boulay, F.; Nyström, T.; Karlsson, A.; Dahlgren, C. Proinflammatory activity of a cecropin-like antibacterial peptide from Helicobacter pylori. Antimicrob. Agents Chemother. 2001, 45, 1700–1704. [Google Scholar] [CrossRef] [Green Version]
  61. De Paulis, A.; Prevete, N.; Rossi, F.W.; Rivellese, F.; Salerno, F.; Delfino, G.; Liccardo, B.; Avilla, E.; Montuori, N.; Mascolo, M.; et al. Helicobacter pylori Hp(2–20) promotes migration and proliferation of gastric epithelial cells by interacting with formyl peptide receptors in vitro and accelerates gastric mucosal healing in vivo. J. Immunol. 2009, 183, 3761–3769. [Google Scholar]
  62. Capparelli, R.; De Chiara, F.; Nocerino, N.; Montella, R.C.; Iannaccone, M.; Fulgione, A.; Romanelli, A.; Avitabile, C.; Blaiotta, G.; Capuano, F. New perspectives for natural antimicrobial peptides: Application as antinflammatory drugs in a murine model. BMC Immunol. 2012, 13, 61. [Google Scholar]
  63. Hemmi, H.; Takeuchi, O.; Kawai, T.; Kaisho, T.; Sato, S.; Sanjo, H.; Matsumoto, M.; Hoshino, K.; Wagner, H.; Takeda, K.; et al. A Toll-like receptor recognizes bacterial DNA. Nature 2000, 408, 740–745. [Google Scholar] [CrossRef] [PubMed]
  64. Wagner, H. The sweetness of the DNA backbone drives Toll-like receptor 9. Curr. Opin. Immunol. 2008, 20, 396–400. [Google Scholar] [CrossRef] [PubMed]
  65. Ohto, U.; Shibata, T.; Tanji, H.; Ishida, H.; Krayukhina, E.; Uchiyama, S.; Miyake, K.; Shimizu, T. Structural basis of CpG and inhibitory DNA recognition by Toll-like receptor 9. Nature 2015, 520, 702–705. [Google Scholar] [CrossRef] [PubMed]
  66. Brencicova, E.; Diebold, S.S. Nucleic acids and endosomal pattern recognition: How to tell friend from foe? Front. Cell. Infect. Microbiol. 2013, 3, 37. [Google Scholar] [CrossRef] [Green Version]
  67. Schmausser, B.; Andrulis, M.; Endrich, S.; Lee, S.K.; Josenhans, C.; Müller-Hermelink, H.K.; Eck, M. Expression and subcellular distribution of toll-like receptors TLR4, TLR5 and TLR9 on the gastric epithelium in Helicobacter pylori infection. Clin. Exp. Immunol. 2004, 136, 521–526. [Google Scholar] [CrossRef]
  68. Varga, M.G.; Shaffer, C.L.; Sierra, J.C.; Suarez, G.; Piazuelo, M.B.; Whitaker, M.E.; Romero-Gallo, J.; Krishna, U.S.; Delgado, A.; Gomez, M.A.; et al. Pathogenic Helicobacter pylori strains translocate DNA and activate TLR9 via the cancer-associated cag type IV secretion system. Oncogene 2016, 35, 6262–6269. [Google Scholar] [CrossRef] [Green Version]
  69. Luther, J.; Owyang, S.Y.; Takeuchi, T.; Cole, T.S.; Zhang, M.; Liu, M.; Erb-Downward, J.; Rubenstein, J.H.; Chen, C.C.; Pierzchala, A.V.; et al. Helicobacter pylori DNA decreases pro-inflammatory cytokine production by dendritic cells and attenuates dextran sodium sulphate-induced colitis. Gut 2011, 60, 1479–1486. [Google Scholar] [CrossRef]
  70. Owyang, S.Y.; Luther, J.; Owyang, C.C.; Zhang, M.; Kao, J.Y. Helicobacter pylori DNA’s anti-inflammatory effect on experimental colitis. Gut. Microbes. 2012, 3, 168–171. [Google Scholar] [CrossRef] [Green Version]
  71. Rad, R.; Ballhorn, W.; Voland, P.; Eisenächer, K.; Mages, J.; Rad, L.; Ferstl, R.; Lang, R.; Wagner, H.; Schmid, R.M.; et al. Extracellular and intracellular pattern recognition receptors cooperate in the recognition of Helicobacter pylori. Gastroenterology 2009, 136, 2247–2257. [Google Scholar] [CrossRef]
  72. Guerrero-Mandujano, A.; Hernández-Cortez, C.; Ibarra, J.A.; Castro-Escarpulli, G. The outer membrane vesicles: Secretion system type zero. Traffic 2017, 18, 425–432. [Google Scholar] [CrossRef] [Green Version]
  73. Tiku, V.; Tan, M.W. Host immunity and cellular responses to bacterial outer membrane vesicles. Trends Immunol. 2021, 42, 1024–1036. [Google Scholar] [CrossRef] [PubMed]
  74. Grande, R.; Di Marcantonio, M.C.; Robuffo, I.; Pompilio, A.; Celia, C.; Di Marzio, L.; Paolino, D.; Codagnone, M.; Muraro, R.; Stoodley, P.; et al. Helicobacter pylori ATCC 43629/NCTC 11639 Outer Membrane Vesicles (OMVs) from biofilm and planktonic phase associated with extracellular DNA (eDNA). Front. Microbiol. 2015, 6, 1369. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Yonezawa, H.; Osaki, T.; Woo, T.; Kurata, S.; Zaman, C.; Hojo, F.; Hanawa, T.; Kato, S.; Kamiya, S. Analysis of outer membrane vesicle protein involved in biofilm formation of Helicobacter pylori. Anaerobe 2011, 17, 388–390. [Google Scholar] [CrossRef] [PubMed]
  76. Olofsson, A.; Vallström, A.; Petzold, K.; Tegtmeyer, N.; Schleucher, J.; Carlsson, S.; Haas, R.; Backert, S.; Wai, S.N.; Gröbner, G.; et al. Biochemical and functional characterization of Helicobacter pylori vesicles. Mol. Microbiol. 2012, 77, 1539–1555. [Google Scholar] [CrossRef] [Green Version]
  77. Keenan, J.I.; Davis, K.A.; Beaugie, C.R.; McGovern, J.J.; Moran, A.P. Alterations in Helicobacter pylori outer membrane and outer membrane vesicle-associated lipopolysaccharides under iron-limiting growth conditions. Innate Immun. 2008, 14, 279–290. [Google Scholar] [CrossRef]
  78. Turner, L.; Bitto, N.J.; Steer, D.L.; Lo, C.; D’Costa, K.; Ramm, G.; Shambrook, M.; Hill, A.F.; Ferrero, R.L.; Kaparakis-Liaskos, M. Helicobacter pylori outer membrane vesicle size determines their mechanisms of host cell entry and protein content. Front. Immunol. 2018, 9, 1466. [Google Scholar] [CrossRef] [Green Version]
  79. Viala, J.; Chaput, C.; Boneca, I.G.; Cardona, A.; Girardin, S.E.; Moran, A.P.; Athman, R.; Mémet, S.; Huerre, M.R.; Coyle, A.J.; et al. Nod1 responds to peptidoglycan delivered by the Helicobacter pylori cag pathogenicity island. Nat. Immunol. 2004, 5, 1166–1174. [Google Scholar] [CrossRef]
  80. Kaparakis, M.; Turnbull, L.; Carneiro, L.; Firth, S.; Coleman, H.A.; Parkington, H.C.; Le Bourhis, L.; Karrar, A.; Viala, J.; Mak, J.; et al. Bacterial membrane vesicles deliver peptidoglycan to NOD1 in epithelial cells. Cell Microbiol. 2010, 12, 372–385. [Google Scholar] [CrossRef] [Green Version]
  81. Hutton, M.L.; Kaparakis-Liaskos, M.; Turner, L.; Cardona, A.; Kwok, T.; Ferrero, R.L. Helicobacter pylori exploits cholesterol-rich microdomains for induction of NF-kappaB-dependent responses and peptidoglycan delivery in epithelial cells. Infect. Immun. 2010, 78, 4523–4531. [Google Scholar] [CrossRef] [Green Version]
  82. Allison, C.C.; Kufer, T.A.; Kremmer, E.; Kaparakis, M.; Ferrero, R.L. Helicobacter pylori induces MAPK phosphorylation and AP-1 activation via a NOD1-dependent mechanism. J. Immunol. 2009, 183, 8099–8109. [Google Scholar] [CrossRef] [Green Version]
  83. Grubman, A.; Kaparakis, M.; Viala, J.; Allison, C.; Badea, L.; Karrar, A.; Boneca, I.G.; Le Bourhis, L.; Reeve, S.; Smith, I.A.; et al. The innate immune molecule, NOD1, regulates direct killing of Helicobacter pylori by antimicrobial peptides. Cell. Microbiol. 2010, 12, 626–639. [Google Scholar] [CrossRef] [PubMed]
  84. Watanabe, T.; Asano, N.; Fichtner-Feigl, S.; Gorelick, P.L.; Tsuji, Y.; Matsumoto, Y.; Chiba, T.; Fuss, I.J.; Kitani, A.; Strober, W. NOD1 contributes to mouse host defense against Helicobacter pylori via induction of type I IFN and activation of the ISGF3 signaling pathway. J. Clin. Investig. 2010, 120, 1645–1662. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Jarzab, M.; Posselt, G.; Meisner-Kober, N.; Wessler, S. Helicobacter pylori-Derived Outer Membrane Vesicles (OMVs): Role in Bacterial Pathogenesis? Microorganisms 2020, 8, 1328. [Google Scholar] [CrossRef]
  86. Ismail, S.; Hampton, M.B.; Keenan, J.I. Helicobacter pylori outer membrane vesicles modulate proliferation and interleukin-8 production by gastric epithelial cells. Infect. Immun. 2003, 71, 5670–5675. [Google Scholar] [CrossRef] [Green Version]
  87. Choi, H.I.; Choi, J.P.; Seo, J.; Kim, B.J.; Rho, M.; Han, J.K.; Kim, J.G. Helicobacter pylori-derived extracellular vesicles increased in the gastric juices of gastric adenocarcinoma patients and induced inflammation mainly via specific targeting of gastric epithelial cells. Exp. Mol. Med. 2017, 49, e330. [Google Scholar] [CrossRef]
  88. Chitcholtan, K.; Hampton, M.B.; Keenan, J.I. Outer membrane vesicles enhance the carcinogenic potential of Helicobacter pylori. Carcinogenesis 2008, 29, 2400–2405. [Google Scholar] [CrossRef] [Green Version]
  89. Chmiela, M.; Walczak, N.; Rudnicka, K. Helicobacter pylori outer membrane vesicles involvement in the infection development and Helicobacter pylori-related diseases. J. Biomed. Sci. 2018, 25, 78. [Google Scholar] [CrossRef]
Figure 1. H. pylori survival strategies against phagocytes. The bacterium secretes catalase and SOD to reduce ROS toxicity, synthetizes arginase to limit NO production, reduces host NADPH oxidase response, and induces macrophage apoptosis.
Figure 1. H. pylori survival strategies against phagocytes. The bacterium secretes catalase and SOD to reduce ROS toxicity, synthetizes arginase to limit NO production, reduces host NADPH oxidase response, and induces macrophage apoptosis.
Ijms 23 03531 g001
Figure 2. H. pylori LPS structure. Lacking the canonical inner and outer core organization, H. pylori LPS shows a short core and a longer O-antigen with a terminal Lewis antigen.
Figure 2. H. pylori LPS structure. Lacking the canonical inner and outer core organization, H. pylori LPS shows a short core and a longer O-antigen with a terminal Lewis antigen.
Ijms 23 03531 g002
Figure 3. Schematic release and action of H. pylori heptose metabolites. HBP and ADP-heptose enter epithelial cells to induce TIFAsome formation and NF-kB activation.
Figure 3. Schematic release and action of H. pylori heptose metabolites. HBP and ADP-heptose enter epithelial cells to induce TIFAsome formation and NF-kB activation.
Ijms 23 03531 g003
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Eletto, D.; Mentucci, F.; Voli, A.; Petrella, A.; Porta, A.; Tosco, A. Helicobacter pylori Pathogen-Associated Molecular Patterns: Friends or Foes? Int. J. Mol. Sci. 2022, 23, 3531. https://doi.org/10.3390/ijms23073531

AMA Style

Eletto D, Mentucci F, Voli A, Petrella A, Porta A, Tosco A. Helicobacter pylori Pathogen-Associated Molecular Patterns: Friends or Foes? International Journal of Molecular Sciences. 2022; 23(7):3531. https://doi.org/10.3390/ijms23073531

Chicago/Turabian Style

Eletto, Daniela, Fatima Mentucci, Antonia Voli, Antonello Petrella, Amalia Porta, and Alessandra Tosco. 2022. "Helicobacter pylori Pathogen-Associated Molecular Patterns: Friends or Foes?" International Journal of Molecular Sciences 23, no. 7: 3531. https://doi.org/10.3390/ijms23073531

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop