Next Article in Journal
Preliminary Expression Analysis of the OSCA Gene Family in Maize and Their Involvement in Temperature Stress
Next Article in Special Issue
Bamboo Salt and Triple Therapy Synergistically Inhibit Helicobacter pylori-Induced Gastritis In Vivo: A Preliminary Study
Previous Article in Journal
Fine Mapping and Candidate Gene Analysis of Pm36, a Wild Emmer-Derived Powdery Mildew Resistance Locus in Durum Wheat
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Helicobacter pylori in the Oral Cavity: Current Evidence and Potential Survival Strategies

1
State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
2
Department of Operative Dentistry and Endodontics, West China School of Stomatology, Sichuan University, Chengdu 610041, China
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Int. J. Mol. Sci. 2022, 23(21), 13646; https://doi.org/10.3390/ijms232113646
Submission received: 12 September 2022 / Revised: 3 November 2022 / Accepted: 5 November 2022 / Published: 7 November 2022
(This article belongs to the Special Issue New Advances on Helicobacter pylori Research)

Abstract

:
Helicobacter pylori (H. pylori) is transmitted primarily through the oral–oral route and fecal–oral route. The oral cavity had therefore been hypothesized as an extragastric reservoir of H. pylori, owing to the presence of H. pylori DNA and particular antigens in distinct niches of the oral cavity. This bacterium in the oral cavity may contribute to the progression of periodontitis and is associated with a variety of oral diseases, gastric eradication failure, and reinfection. However, the conditions in the oral cavity do not appear to be ideal for H. pylori survival, and little is known about its biological function in the oral cavity. It is critical to clarify the survival strategies of H. pylori to better comprehend the role and function of this bacterium in the oral cavity. In this review, we attempt to analyze the evidence indicating the existence of living oral H. pylori, as well as potential survival strategies, including the formation of a favorable microenvironment, the interaction between H. pylori and oral microorganisms, and the transition to a non-growing state. Further research on oral H. pylori is necessary to develop improved therapies for the prevention and treatment of H. pylori infection.

1. Introduction

H. pylori is a microaerophilic pathogen with a typical spiral or arc form that infects about 50% of the world’s population [1]. Although most H. pylori-positive individuals are asymptomatic, long-term infection with H. pylori has been associated with the development of gastric malignancies, particularly gastric cancer and gastric mucosa-associated lymphoid tissue lymphoma [2]. Hence, this organism was identified as a class I carcinogen by the World Health Organization in 1994. A combination of at least two diagnostic methods is often required to detect H. pylori infection and related diseases. Moreover, the emergence of drug-resistant strains poses considerable challenges to current treatment [3]. Therefore, it is beyond dispute that H. pylori is associated with a heavy disease burden worldwide.
It is generally accepted that H. pylori transmits between the human population through the oral–oral route, the fecal–oral route, and the gastro–oral route [4,5]. The oral cavity is the first channel for H. pylori into the human body, and its function in H. pylori infection of the human body generates considerable concerns among researchers. Many scholars have employed molecular techniques, immunological or biochemical approaches, and classic culture techniques to identify oral samples as H. pylori-positive [6]. Accordingly, studies have been carried out to detect H. pylori infection using saliva samples. Such a non-invasive test might be more acceptable to individuals [3]. In addition, some studies have shown that H. pylori in the oral cavity could adversely affect the clinical outcome of eradication therapy [7,8], and oral H. pylori is considered a risk factor for the recrudescence of gastric H. pylori infection [9]. Consequently, it is hypothesized that the oral cavity might be a potential source for H. pylori gastric reinfection [10].
The environment of the oral cavity differs considerably from that of the stomach. It is generally accepted that H. pylori is a microaerophilic microorganism that requires high CO2 tension to thrive and survive for long periods of time [11,12]. However, owing to the communication with the outside environment, a lower CO2 concentration and a higher O2 concentration are detected in the oral cavity than in the stomach. The microbial composition, temperature, and mechanical scouring within the oral cavity differ considerably from those of the stomach. For instance, Streptococcus mitis (S. mitis) and Streptococcus mutans (S. mutans) may inhibit the growth of H. pylori in vitro [13,14]. Furthermore, eating can cause an unstable oral temperature, and the mechanical flushing of saliva might be a barrier to the long-term oral survival of H. pylori (Figure 1). Therefore, the inconsistencies between clinical detections and unfavorable conditions represent a challenging question concerning how H. pylori adapt to the environment of the oral cavity.
In this review, we analyze various lines of evidence regarding the persistent survival of H. pylori in the oral cavity and discuss three potential strategies favoring its survival in order to better comprehend the role of this bacterium in the oral cavity and to inspire the development of adjunctive treatment for improved control of H. pylori.

2. The Evidence for the Persistent Survival of Oral H. pylori

2.1. Various Samples from the Oral Cavity Can Be Detected as H. pylori-Positive

H. pylori need to reach and continue to survive in the oral cavity, which is considered the first channel of H. pylori transmission into the stomach. Many scholars have applied molecular biology and immunology methods to detect oral H. pylori in dental plaque [15], saliva [16,17], the tongue coating [18], and dental pulp [19,20,21]. Recently, Sruthi et al. [22] suggested that H. pylori can also be detected on deep carious surfaces in the oral cavity of children, with a positivity rate of 70%. In comparison, dental plaque is more likely than saliva to be detected by PCR as H. pylori-positive [6], possibly due to the continuous flow of saliva reducing the bacterial load [17]. In a study detecting H. pylori in dental pulp, most H. pylori-positive specimens from the same teeth remained positive after an interval of one to two weeks, indicating that H. pylori colonized the inflamed pulp tissue [20].
Although most studies have reported positive detection of oral H. pylori by molecular biology, such results are unsatisfactory because they cannot reflect the viability of H. pylori in the oral cavity. Several studies have attempted to culture oral H. pylori, indicating that H. pylori appears to be an organism that lives in the oral cavity for the long term. In 1989, Krajden et al. [23] successfully isolated and cultured H. pylori in dental plaque for the first time; saliva cultures from all 71 patients included in the study failed to show any positive results. Since then, a small number of published studies [6,21] have described the successful isolation and culture of H. pylori from dental plaque, saliva, or pulp samples. However, most of these studies confirmed the isolate as H. pylori by oxidase test, catalase test, urease tests, or microscopic observations, which cannot rule out the influence of other H. pylori-like microorganisms. Therefore, further study with a focus on whole-genome sequencing of the isolated bacterial strain is suggested [6].

2.2. The Association between Gastric H. pylori Infection and Oral H. pylori Positivity

Several clinical studies in recent years have reported a close association between gastric H. pylori infection and oral H. pylori positivity (Table 1). A meta-analysis of 23 studies (1861 patients) found that the rate of coinfection with H. pylori in gastric and dental plaque was 49.7% [24]. In theory, the high coexistence rate of H. pylori in oral and gastric samples may imply that the oral cavity serves as a reservoir of H. pylori. Some scholars further used H. pylori virulence marker genes for genotyping and found that the vacuolating cytotoxin A (vacA) genotype was concordant in 51.1~58% of saliva and biopsy from the same patient, suggested that H. pylori strains in that oral cavity and stomach are likely to be homologous [4,25]. However, some scholars found that H. pylori was commonly present in the oral cavity with no clear relation to H. pylori infection of the stomach [16,26,27], indicating that H. pylori in the oral cavity exhibited a degree of independence.

2.3. H. pylori in the Oral Cavity Is Associated with Oral Diseases and Gastric Infection

The survival of H. pylori in the oral cavity threatens oral health. In recent years, the pathogenic role of oral H. pylori has attracted the attention of many researchers. A meta-analysis by Liu et al. [39] showed that oral H. pylori, especially in supragingival plaque, is a risk factor for periodontitis. The total proportion of periodontal pathogens in oral H. pylori-positive subgingival plaque samples was higher than that in H. pylori-negative samples [40], indicating that oral H. pylori infection may promote periodontal disease by altering the microecology. Preincubation of Porphyromonas gingivalis (P. gingivalis) with H. pylori affects P. gingivalis virulence, including biofilm formation, bacterial internalization into oral keratinocytes, and hemagglutination, indicating that the direct interaction between P. gingivalis and H. pylori in subgingival plaque may increase the severity or progression of periodontitis [41]. In addition, the expression of periodontitis-related protein Wnt5a and cytokines IL-8, IL-6, and INF-γ was significantly increased after cagA + H. pylori stimulated the human leukemia mononuclear cell line, suggesting that H. pylori can aggravate inflammation progression [40]. In addition to periodontitis, oral H. pylori has been reported to be associated with erosive oral lichen planus (OLP) [42,43] and oral squamous cell carcinoma (OSCC) [44]. However, wing to the heterogeneity in interstudy design, conflicting findings have been reported [44,45,46], and the pathogenic role of oral H. pylori in these diseases remains unclear. In the next step, detection methods with high sensitivity and specificity should be used to clarify the role and specific mechanism of oral H. pylori in the development of oral diseases.
Oral H. pylori increases the severity of gastric infections and the difficulty of eradication. On the one hand, oral H. pylori was linked to an increased incidence of grade II gastroesophageal reflux, esophageal sphincter relaxation, and duodenitis in a case–control study including 567 patients [7]. On the other hand, the gastric H. pylori eradication success rate was significantly lower in oral H. pylori-positive patients than in oral H. pylori-negative patients (52.2% vs. 91.6%, respectively, p = 0.0028) four weeks after eradication therapy [8]. The association between oral H. pylori and gastric infection suggests that the oral cavity may be the source of gastric reinfection.

2.4. Effects of Oral Hygiene Management on H. pylori Infection

Given the disparity in eradication success rates between gastric H. pylori and oral H. pylori eradication therapy (85.8% vs. 5.7%, respectively, OR 55.59, p < 0.00001) [47], eradicating oral H. pylori with systemic therapy remains difficult in ordinary clinical applications [48]. In such cases, adjuvant topical treatment to eradicate oral H. pylori seems necessary (Table 2). For instance, eradication therapy combined with periodontal therapy can increase the eradication rate of gastric H. pylori [49,50]. Consistently, the OR for the unsuccessful gastric eradication increased 64-fold if periodontal treatment failed to eliminate oral H. pylori [51]. Oral hygiene management can reduce H. pylori in the oral cavity and help to control its migration from to the stomach and can be used as an adjuvant treatment option for gastric H. pylori eradication therapy. Attention should be paid to the development of oral healthcare products with anti-H. pylori effect to contribute to oral defense against H. pylori.
The evidence presented thus far supports the idea that H. pylori is likely to survive in the oral cavity and be involved in gastric infection. However, currently, we can only reduce the negative effects of oral H. pylori by some oral hygiene treatments. Therefore, further investigation of the details of oral H. pylori survival is required to achieve the effective eradication of H. pylori infection.

3. Pangenome and Virulence Factors

The prevalence of H. pylori infection is not always correlated with the incidence of gastric diseases. Only a small percentage of those infected progress to peptic ulcers or even gastric cancer. Some countries in Africa and Asia have lower rates of gastric cancer and higher rates of H. pylori infection, whereas others exhibit the opposite trend. Such a phenomenon of clinical diversity is determined by the genetic variability of the infecting H. pylori strains; the genetic background and immunity of different ethnic groups; gastric and intestinal microbiota; and environmental factors, such as geographic location and dietary habits [55,56,57,58]. For example, H. pylori infection status can change oral microorganism composition and alter the interactions between microorganisms, whereas the oral host–microbial interactome could provide signals to impact health and disease [59]. The oral microbiota of the host may cause carcinogenesis via various potential mechanisms, including the induction of chronic inflammation, the inhibition of the host’s immune system, antiapoptotic activity, and the production of carcinogenic substances that may fuel the progression of cancer [60].
Geographically stratified H. pylori subpopulations have emerged as a result of the mode of intrafamilial transmission and long-term coevolution with human hosts [61]. The whole-genome sequencing of H. pylori indicated a high rate of gene recombination and unusual genetic flexibility; these traits enable the bacteria adapt to the dynamic environment [62,63]. Therefore, systematic analysis of the whole-gene repertoire, termed the pan-genome, is important for understanding bacterial intraspecies diversity, population genetics, and evolution. Sequence-alignment-based multilocus sequence typing (MLST) of H. pylori grouped H. pylori isolates into seven types connected to geographic information [64], and the conserved regions and genes among H. pylori genomes were potentially associated with H. pylori pathogenicity and adaptation, such as cag pathogenicity island (cagPAI) [62,63]. A pangenome Fst analysis showed that variation in virulence genes was more common in the Americas than other regions [65]. A total of 22 of the 35 genes with the highest Fst values encode recognized virulence factors and membrane proteins, suggesting that virulence plays a strong role in H. pylori adaption to specific human populations [65].
H. pylori virulence factors can be categorized to be related with three main pathogenic steps: bacterial colonization, immune evasion, and disease induction [66]. The helical shape and flagella enable H. pylori to penetrate the mucous layer and subsequently adhere to cellular surface receptors via adhesins [67]. Urease hydrolyses urea, thus neutralizing acidic pH and forming a neutral layer favorable to H. pylori survival [67]. The blood group antigen-binding adhesion (BabA) and sialic acid-binding adherence (SabA) are two of the most extensively studied H. pylori outer-membrane proteins, which function as adhesins that mediate H. pylori binding to gastric epithelial cells [68]. The surface features and various virulence proteins have been identified to contribute to immune evasion and disease induction.
CagA and VacA are the main H. pylori virulence factors involved in immune evasion and disease induction and are likely to cause disease in the oral cavity [69]. cagPAI is a 35–40 kb DNA segment located on the H. pylori chromosome and carries more than 30 genes. The variability of H. pylori is reflected in the frequency of possession of the cagPAI, as the carriage of cagPAI varies from almost universal presence in hpEastAsia and hpAfrica1 through intermediate presence (hpEurope) to complete absence (hpAfrica2) [70]. cagPAI encodes an antigenic effector protein (CagA). The odds ratio for dysplasia was reported to be higher in cagA-positive individuals compared with cagA-negative individuals (15.4 vs. 0.90, respectively), suggesting that cagA is associated with increased gastric cancer risk [71]. The Western-type CagA and East Asian-type CagA were further described based on the repeat sequence Glu-Pro-Ile-Tyr-Ala (EPIYA) motifs at the N-terminus of CagA and their binding activity to Src homology 2 (SH2)-containing tyrosine phosphatase SHP-2, whereas East Asian-type CagA was reported to confer stronger SHP-2 binding and morphologically transforming activities compared to Western CagA, making East Asian-type CagA more virulent [67,72]. In addition, genes encoding proteins of type IV secretion systems (T4SS) are located in cagPAI. CagA can be delivered into the cytoplasm via the T4SS and can subsequently interact with at least 10 host cell components in both a phosphorylation-dependent and phosphorylation-independent manner, altering the cellular signal transduction system [73]. cag+ H. pylori strains also showed diversity in terms of levels of CagA production, whereas strains producing higher levels of CagA were associated with increased risk of premalignant lesions [74,75].
VacA is another extensively studied virulence factor that can be produced by all H. pylori strains. VacA can induce the formation of large cytoplasmic vacuoles in host cells and is involved in immune evasion of H. pylori. However, there are differences between strains at the level of VacA production or secretion and in the amino acid sequence among VacA proteins [76]. Several vacA subtypes can be divided according to combinations of the signal sequence (s1a, s1b, s1c, and s2), mid-region (m1, m1T, and m2), and the intermediate region (i1, i2, and i3) [67]. As a result, infection with strains carrying combinations of these hypervirulence genes (e.g., vacAs1m1cagA +) is associated with a higher risk of invasive disease than infection with strains carrying combinations of less virulent alleles (e.g., vacAs2m2cagA−) [57].
The genetic diversity of H. pylori may be associated with their adaptation to the host and disease progression. Attention should be paid to the virulence characteristics and genetic background of oral H. pylori. Further pangenomic analysis of the genes related to H. pylori colonization can facilitate an improved understanding of the survival strategies of H. pylori in the oral cavity.

4. Interactions between H. pylori and Microenvironments in the Oral Cavity

The oral cavity might be the prime habitat for H. pylori colonization and gastric re-infection; however, how H. pylori adapt to the environment of the oral cavity is still unclear. The mutual interaction of H. pylori with the local oral environment can be considered on two levels. The first focuses on biofilm formation; the biofilm matrix plays a synergistic role in protecting H. pylori against unfavorable factors. The second concerns the physical contact with host cells, whereby the adhesion and invading capability of H. pylori may favor its oral survival.

4.1. Oral Microenvironment and H. pylori

As mentioned above, H. pylori has been detected in plaque samples collected from carious cavities and periodontal pockets, suggesting that it can survive in the microenvironments of these niches, where biofilm provides various benefits for bacterial reproduction, metabolism, and defense and is the favored method of long-term survival for many colonizers. Therefore, H. pylori is often found in dental plaque, where the detection rate is often higher than that in saliva. The layered morphology of biofilm suggests that there can be gradients in nutrients, gas concentration, and pH value. When observed at the micron scale, supragingival plaque has a complex microbial consortium called “hedgehog” composed primarily of Corynebacterium, a multitaxon filament-rich annulus, and a periphery of Streptococcus-containing corncob structures [77,78]. In corncob structures, Streptococcus cells consume carbohydrates and O2 to generate CO2, thus generating a CO2-rich environment for survival of microaerophilic and anaerobic microbes, which might be favorable to H. pylori survival. Typically, cariogenic microorganisms in dental plaque produce acids upon exposure to carbohydrates, resulting in a decrease in pH value, which challenges the survival of H. pylori. Oral H. pylori also needs to adapt to the changing pH value of cariogenic dental plaque. In this case, urea-metabolizing capacity is important for the survival of alkali-producing symbionts in dental plaque [79], and the same may also hold true for H. pylori, which can use urease to convert urea into ammonia and CO2 with a certain buffering capacity, leading to medium alkalization around itself [57]. The formation of biofilms in these niches enables the long-term adhesion of H. pylori without being affected by saliva flushing or food chewing. Moreover, bacteria surviving in biofilms are usually resistant to host defense systems and antimicrobial medicines, protecting them from the adverse effects of drugs during systemic treatment [80,81], which may be among the reasons why it is so difficult to eliminate oral H. pylori.

4.2. Oral Host Cells and H. pylori

Interaction with the host is also an important strategy for H. pylori survival in dental pulp. H. pylori surviving in biofilm may reach pulp through caries cavities and survive in dental pulp, as pulp H. pylori-positive teeth often have deep cavities, whereas those with milder caries are rarely detected with pulp H. pylori positivity [21,82]. For instance, H. pylori has the ability to adhere to host cells in dental pulp. H. pylori ATCC 51932, H. pylori 26695, and H. pylori J99 all displayed adhesion capabilities in human dental pulp fibroblast cells (HDPFs), whereas the other two cagA-positive strains showed higher adhesion rates than cagA-negative H. pylori ATCC 51932 [20]. In studies involving gastric epithelial cells, CagA (cytotoxin associated antigen) was reported to disrupt host cell polarity, enabling adherent H. pylori to replicate and grow on the cell surface [83,84].
Helicobacter outer-membrane protein Q (HopQ) was recently found to bind to the receptor carcinoembryonic antigen-related cell adhesion molecule family (CEACAMs) exposed on the host cell surface [85,86]. Consequently, this interaction allows bacteria to adhere to host cells and is required for the injection of CagA into host cells via the type IV secretion system (T4SS) [85,86]. However, unlike gastric epithelial cells, oral epithelial cell-derived cell lines HN, CAL-27, and BHY were reported to be resistant to CagA injection due to the lack of CEACAM expression, suggesting that healthy oral epithelium cells may lack an H. pylori docking site [87]. However, the ceacam1 gene is highly upregulated during palate development [88]. Moreover, CEACAM expression in the oral cavity is upregulated in patients with oral cancer, periodontitis, and oral lichen planus, as well as in smokers [89,90,91,92]. Therefore, in some pathological conditions, the increased expression of oral CEACAMs may favor H. pylori adhesion to oral host cells and even create favorable conditions for CagA injection into oral cells, inducing inflammatory factors. Unfortunately, no in vivo evidence exists to support these ideas.
In addition, CEACAM1 functions as an inhibitory receptor on various immune cells, including T and NK cells [93]. H. pylori HopQ can inhibit interferon-gamma (IFN-γ) secretion of CD4 cells and suppress T or NK cell cytotoxicity by interacting with CEACAM1, and the inhibition of immune cells may help protect developing gastric tumors from immune cell attack [94]. For H. pylori survival, this inhibitory effect may also benefit the peaceful coexistence of the H. pylori immune system, which is consistent with the fact that the majority of H. pylori-positive individuals are asymptomatic. The interaction of H. pylori with human immunity is discussed further in Section 4.3.
H. pylori is able to invade gastric epithelial cells and complete the entire biological cycle, including proliferation and apoptosis, within the cells [95,96,97]; a similar phenomenon has been observed in the oral cavity. Coccoid forms of H. pylori SS1 were localized and surrounded by vacuoles in the cytoplasm of human periodontal ligament fibroblasts (hPDLFs), and the invasion of H. pylori SS1 can adversely affect basic cellular functions of hPDLFs, resulting in G2 phase arrest and inhibition of cell proliferation [98]. Interestingly, H. pylori can also invade immune cells and survive within them by affecting autophagy [99,100]. This intracellular location could facilitate H. pylori evasion of host immune surveillance and antibiotic pressure, allowing H. pylori to intracellularly persist, proliferate, and spread to adjacent tissues. At present, it is still unclear whether invading H. pylori can be released from the cells, and more studies are necessary to establish the effects on cell functions.

4.3. Human Host Immunity and H. pylori

The host immune system can produce antimicrobial peptides, activate the cellular autophagy pathway, and enhance oxidative stress against H. pylori infection [73]. The survival tactics of H. pylori in stomach mucosa have been well-studied; one of the strategies of H. pylori is to modulate surface features that interfere with host immune system recognition [101]. For example, the negatively charged group on H. pylori lipopolysaccharide (LPS) is replaced, which reduces the surface negative charge and thus resists cationic antimicrobial peptide action [102]. LPS from H. pylori is less able to activate the Toll-like receptor (TLR), and a recent study revealed that H. pylori specifically binds human annexins via lipid A and strongly inhibits LPS-mediated TLR4 signal transduction to avoid its recognition by the innate immune system [103].
As mentioned previously, HopQ-CEACAM interaction suppresses immune cell function. In addition, H. pylori controls the production and secretion of chemokines in immune cells via HopQ-CEACAM interaction and survives within neutrophils in a HopQ-dependent manner [104]. The classically activated macrophages (M1 macrophages) highly express proinflammatory cytokines IL-1β, transforming growth factor-β (TNF-β), and nitric oxide synthase (iNOS), leading to the reduction in bacterial load and enhanced pathology [105]. To achieve long-term survival in the host, H. pylori has been found to inhibit macrophage phagocytosis and suppress T-cell activation by hindering expression of human leukocyte antigen-II (HLA-II) and IFN-γ production from macrophages [106]. The expression of programmed cell death ligand 1 (PD-L1) has been suggested as an immune modulatory mechanism for persistent infection of H pylori, and dendritic cells expressing higher levels of PD-L1 have been found to impede H. pylori-induced inflammation but allow persistent H. pylori colonization in mice [107].
H. pylori virulence factors are often considered to contribute to the progression of gastric pathology. However, from the bacterial perspective, the virulence factors may be mechanisms supporting escape from host immune clearance and maintenance of chronic infection [108]. For example, VacA is able to suppress phagocytosis, induce tolerogenic dendritic cells, and block effector T-cell response, thus inhibiting the function of various immune cells [66]. In addition, H. pylori can affect cell autophagy via VacA to benefit its intracellular survival [73]. The interaction between H. pylori and host immunity may influence the success of H. pylori colonization. Because H. pylori is often detected in infected dental pulp and periodontal pockets, the interaction between H. pylori and local immunity warrants further research.

5. The Synergistic Interactions of Oral H. pylori with Oral Microorganisms

Clinically, people with caries or periodontitis are more likely to be infected with oral H. pylori [26,109,110]. Survival of H. pylori in the oral niches may benefit from interactions with oral microorganisms in these lesions, such as S. mutans [111,112], Candida albicans (C. albicans) [113], P. gingivalis [114], and Fusobacterium nucleatum (F. nucleatum) [115] (Table 3).
S. mutans is the most common cariogenic microorganism; there may be a synergistic relationship between S. mutans and H. pylori. H. pylori can grow throughout the biofilm formed by S. mutans in vitro, and the location of H. pylori in biofilms was reported to be dependent upon the presence or absence of S. mutans [111]. Streptococcus in biofilm contributes to the generation of a CO2-rich environment, which might be related to H. pylori benefiting from S. mutans in biofilm coculture. Moreover, the production of mutant proteins and the acid of S. mutans could be induced by H. pylori supernatant, with S. mutans showing a competitive advantage over S. sanguinis, indicating that H. pylori could create favorable conditions for S. mutans [112]. Based on these existing studies reporting positive feedback between S. mutans and H. pylori, S. mutans could provide a colonization environment for oral H. pylori, whereas oral H. pylori can create a competitive advantage for S. mutans.
P. gingivalis and F. nucleatum are the main anaerobic pathogens of periodontal disease. H. pylori is known to have the ability to coaggregate with P. gingivalis, which can facilitate the long-term persistence of H. pylori in periodontal pockets [14]. H. pylori may promote the severity of periodontitis. Preincubation of P. gingivalis with H. pylori enhanced P. gingivalis virulence, including biofilm formation, bacterial internalization into oral keratinocytes, and hemagglutination [41]. In addition, the role of F. nucleatum in the survival of H. pylori is worthy of further investigation, as H. pylori-negative chronic gastritis patients were reported to have lower levels of F. nucleatum in saliva than healthy subjects [124]. Generally, F. nucleatum plays a critical role in the formation and maturation of dental plaque biofilms, owing to its long and narrow rod-like structure and the expression of a variety of adhesins [125,126]. H. pylori adheres to F. nucleatum and thus colonizes dental plaque through coaggregation [14,115]. Therefore, F. nucleatum may act as a bridge and make an important contribution to the long-term survival of H. pylori in oral biofilms.
However, oral organisms such as S. mutans and Prevotella intermedia (P. intermedia) can also inhibit the growth of H. pylori during coculture in vitro [14,123]. The intricate and dynamic interactions between H. pylori and oral microorganisms might make in vitro cultivation of H. pylori difficult.
Fungus is another important component of the oral microbiome. C. albicans is one of the most prevalent fungi in humans involved in oral infectious diseases. Clinical studies have reported the coexistence of C. albicans and H. pylori in the vagina and stomach [127,128]. Complex cross-kingdom interactions occur between the two organisms [113,116]. Sánchez-Alonzo et al. [116] observed an accumulation of coccoid and bacillary bacteria on yeast pseudohyphae cocultured with H. pylori. Furthermore, several surface interaction mechanisms, including hydrophobic interactions between non-polar peptide chains and lipid structures, hydrogen bonds, and thiol-mediated surface interactions, occur between H. pylori and C. albicans, eventually contributing to the formation of polymicrobial biofilms [117]. These synergistic interactions may be related to C. albicans in mixed-species biofilm consuming oxygen to support the milieu changes from aerobic to anaerobic, favoring the growth of anaerobes [129].
H. pylori can invade the C. albicans yeast cells and was strained green by live/dead BacLight staining, indicating that H. pylori can survive within C. albicans [118,119]. Consistent with this phenomenon, fast-moving bacteriolar-like bodies (BLBs) were observed within the vacuoles of the C. albicans yeast cells and were subsequently identified as H. pylori using PCR and fluorescence in situ hybridization (FISH) techniques [122,130]. Some non-adaptive conditions, such as nutrient deprivation [116], acidic pH [121], amoxicillin [120], and other stress factors, can further induce H. pylori to invade yeast cells. As a consequence, the invading H. pylori can survive unfavorable factors, such as high temperature, desiccation, and antibiotic exposure while expressing proteins [131] and showing an active motility state in vacuoles [120]. H. pylori-carrying C. albicans has been found in food [132] and in many other ecological niches, such as the human oral cavity [133] and vagina [134]. Therefore, yeast cells may provide H. pylori with an intracellular niche that protects H. pylori from unfavorable conditions.
Interestingly, Candida not only harbors intracellular H. pylori but also contributes to the transmission of H. pylori. Invading H. pylori can propagate vertically to the vacuoles of daughter cells of yeasts in consecutive subcultures of yeasts [118]. Moreover, vesicle-encased or free H. pylori can be released by C. albicans, and the released H. pylori may invade new C. albicans yeast cells [122]. In addition to transmission between yeast cells, the H. pylori can spread within yeast cells to various human bodies and niches. For instance, normally born babies had a higher frequency of H. pylori-invaded C. albicans in the oral cavity than babied born by cesarean birth [128,134], indicating that H. pylori-carrying C. albicans in the vagina may support H. pylori transmission to newborns. As a result, reducing the yeast content of the oral cavity might be beneficial in terms of controlling the infection and transmission of oral H. pylori [122].
In conclusion, H. pylori interacts with various oral microorganisms to survive in the oral cavity for a long time and can even invade C. albicans yeast cells to protect itself from harsh conditions. The antagonistic relationship and the invasion of H. pylori into yeast cells may lead to difficulties associated with isolating and culturing H. pylori in vitro.

6. Non-Growing State of H. pylori: Viable but Non-Culturable State and Dormant State

Despite the existence of some synergistic factors, the oral environment is complex and hostile. Non-sporulating bacteria can transmit to the viable but non-culturable (VBNC) state when exposed to harmful stimuli. In the VBNC state, bacterial cells are reduced in size and metabolic activity and become unculturable in vitro [135]. However, the transformation to the VBNC state is reversible, and several VBNC bacterial cells were able to recover to a culturable state under specific conditions [135].
When cultured in the laboratory, H. pylori transform into a spherical shape and lose of culturability under various adverse conditions, such as anaerobic culture [136], nutrient deprivation [137,138], and long-term liquid culture [139,140]. However, there is evidence that these unculturable H. pylori are capable of active transcription and translation processes [136,138,141,142,143,144], possibly cells in the VBNC state. VBNC H. pylori cells have been found to be distributed in freshwater and seawater [139,143,145,146], but no resuscitation technology has been found for H. pylori in vitro [139].
H. pylori in the VBNC state also has certain pathogenic abilities. H. pylori and physiological changes were detected in mouse stomachs after inoculation of VBNC H. pylori suspension, suggesting that VBNC H. pylori can colonize the gastric wall of mice and induce mucosal tissue damage, although less virulently than helical H. pylori [147]. However, there appear to be differences in the infectivity of VBNC H. pylori of different strains. For instance, H. pylori SS1 in the VBNC state in drinking water was unable to infect mice [148]. In another study, H. pylori strain 553/93 in the VBNC state produced more severe inflammation than the other two tested VBNC strains [149].
Another common survival strategy under adverse conditions is dormancy. In a stressful environment (e.g., pH and temperature changes, nutrient deficiencies, and antimicrobial drugs), microorganisms can escape threats by reversibly transitioning from an active state to an inactive (dormant) state [150,151]. In the dormant state, bacteria exhibit little or low metabolic activity [152], remain unreplicated for long periods of time, and have increased resistance to extreme stress [150]. Dormant and VBNC states are similar but differ in terms of performance, which has led to some controversy in distinguishing the two. Some scholars believe that in both the VBNC state and the dormant state, bacteria can survive under adverse conditions but cannot be cultured. Therefore, the two terms can be used to describe the same physiological state; alternatively, the VBNC state is a type of dormant state [150]. Other scholars believe that bacteria still exhibits a certain metabolic activity in the VBNC state, which should be distinguished from the dormant state that does not exhibit obvious metabolic activity [153].
Some bacteria in the oral cavity may have an antagonistic relationship with H. pylori [154], and parts of biofilm have suboptimal growth conditions (e.g., reduced nutrient concentrations or acidity, hypoxia) [79,155], thereby promoting the transition of H. pylori to a VBNC or dormant state. VBNC or dormant H. pylori are highly resistant to adverse environments, which may contribute to the survival of oral H. pylori. Few isolated cultures of oral H. pylori can be associated with these non-growing states. Although H. pylori coccoid, similar to dormant or VBNC states, have been found in oral samples [21,98], the exact status of this coccoid form of H. pylori and its role in survival are still unclear. At present, little is known about the physiological changes and survival status of oral H. pylori.

7. Conclusions

Numerous studies have shown the existence of H. pylori in a variety of oral niches, including dental plaque, infected pulp, and periodontal pockets, implying that H. pylori may be able to survive in the oral environment through certain survival strategies. The formation of biofilms in these niches enables the long-term adhesion of H. pylori without being affected by saliva washing or food chewing. In addition, H. pylori can adhere to and invade host cells in the oral cavity. Furthermore, H. pylori can coaggregate with a variety of oral bacteria and yeast cells, and the invading H. pylori is able to escape from some extracellular pressure. Additionally, transitioning to a non-growing state may be another important strategy for H. pylori to adapt to unfavorable oral cavity conditions, which, together with invading cells and growth inhibition by other microorganisms, explains why oral H. pylori is difficult to culture (Figure 2).

Author Contributions

Conceptualization, L.Z., X.C., B.R., X.Z. and L.C.; writing—original draft preparation, L.Z., X.C. and L.C.; writing—review and editing, B.R., X.Z. and L.C.; visualization, L.Z. and X.C.; supervision, L.C.; funding acquisition, L.C. All authors have read and agreed to the published version of the manuscript.

Funding

National Natural Science Foundation of China 82271033 (L.C.). Chengdu Technological Innovation and R&D Project, grant number: 2022-YF05-01415-SN (L.C.). Research Funding from the West China School/Hospital of Stomatology Sichuan University, grant number: RCDWJS2021-19 (L.C.) SKLOD202113 (L.C.). The authors declare no potential conflicts of interest with respect to the authorship and/or publication of this article.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors have no conflict of interest to declare. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript; or in the decision to publish the results.

References

  1. Schulz, C.; Kalali, B.; Link, A.; Gerhard, M.; Malfertheiner, P. New Rapid Helicobacter pylori Blood Test Based on Dual Detection of FliD and CagA Antibodies for On-Site Testing. Clin. Gastroenterol. Hepatol. 2021. [Google Scholar] [CrossRef] [PubMed]
  2. Mladenova, I. Clinical Relevance of Helicobacter pylori Infection. J. Clin. Med. 2021, 10, 3473. [Google Scholar] [CrossRef] [PubMed]
  3. Ranjbar, R.; Behzadi, P.; Farshad, S. Advances in diagnosis and treatment of Helicobacter pylori infection. Acta Microbiol. Immunol. Hung. 2017, 64, 273–292. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Momtaz, H.; Souod, N.; Dabiri, H.; Sarshar, M. Study of Helicobacter pylori genotype status in saliva, dental plaques, stool and gastric biopsy samples. World J. Gastroenterol. 2012, 18, 2105–2111. [Google Scholar] [CrossRef]
  5. Burucoa, C.; Axon, A. Epidemiology of Helicobacter pylori infection. Helicobacter 2017, 22 (Suppl. S1), e12403. [Google Scholar] [CrossRef] [PubMed]
  6. Mao, X.; Jakubovics, N.S.; Bachle, M.; Buchalla, W.; Hiller, K.A.; Maisch, T.; Hellwig, E.; Kirschneck, C.; Gessner, A.; Al-Ahmad, A.; et al. Colonization of Helicobacter pylori in the oral cavity—An endless controversy? Crit. Rev. Microbiol. 2021, 47, 612–629. [Google Scholar] [CrossRef]
  7. Ansari, S.A.; Iqbal, M.U.N.; Khan, T.A.; Kazmi, S.U. Association of oral Helicobacter pylori with gastric complications. Life Sci. 2018, 205, 125–130. [Google Scholar] [CrossRef]
  8. Miyabayashi, H.; Furihata, K.; Shimizu, T.; Ueno, I.; Akamatsu, T. Influence of oral Helicobacter pylori on the success of eradication therapy against gastric Helicobacter pylori. Helicobacter 2000, 5, 30–37. [Google Scholar] [CrossRef]
  9. Sun, Y.; Zhang, J. Helicobacter pylori recrudescence and its influencing factors. J. Cell Mol. Med. 2019, 23, 7919–7925. [Google Scholar] [CrossRef] [Green Version]
  10. Gisbert, J.P. The recurrence of Helicobacter pylori infection: Incidence and variables influencing it. A critical review. Am. J. Gastroenterol. 2005, 100, 2083–2099. [Google Scholar] [CrossRef]
  11. Park, S.A.; Ko, A.; Lee, N.G. Stimulation of growth of the human gastric pathogen Helicobacter pylori by atmospheric level of oxygen under high carbon dioxide tension. BMC Microbiol. 2011, 11, 96. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Bury-Mone, S.; Kaakoush, N.O.; Asencio, C.; Megraud, F.; Thibonnier, M.; De Reuse, H.; Mendz, G.L. Is Helicobacter pylori a true microaerophile? Helicobacter 2006, 11, 296–303. [Google Scholar] [CrossRef] [PubMed]
  13. Khosravi, Y.; Dieye, Y.; Loke, M.F.; Goh, K.L.; Vadivelu, J. Streptococcus mitis induces conversion of Helicobacter pylori to coccoid cells during co-culture in vitro. PLoS ONE 2014, 9, e112214. [Google Scholar] [CrossRef] [PubMed]
  14. Ishihara, K.; Miura, T.; Kimizuka, R.; Ebihara, Y.; Mizuno, Y.; Okuda, K. Oral bacteria inhibit Helicobacter pylori growth. FEMS Microbiol. Lett. 1997, 152, 355–361. [Google Scholar] [CrossRef] [PubMed]
  15. Mendoza-Cantu, A.; Urrutia-Baca, V.H.; Urbina-Rios, C.S.; De la Garza-Ramos, M.A.; Garcia-Martinez, M.E.; Torre-Martinez, H.H.H. Prevalence of Helicobacter pylori vacA Genotypes and cagA Gene in Dental Plaque of Asymptomatic Mexican Children. Biomed. Res. Int. 2017, 2017, 4923640. [Google Scholar] [CrossRef] [Green Version]
  16. Ji, Y.; Liang, X.; Lu, H. Analysis of by high-throughput sequencing: Helicobacter pylori infection and salivary microbiome. BMC Oral. Health 2020, 20, 84. [Google Scholar] [CrossRef] [Green Version]
  17. Seligova, B.; Lukac, L.; Babelova, M.; Vavrova, S.; Sulo, P. Diagnostic reliability of nested PCR depends on the primer design and threshold abundance of Helicobacter pylori in biopsy, stool, and saliva samples. Helicobacter 2020, 25, e12680. [Google Scholar] [CrossRef]
  18. Zhao, Y.; Gao, X.; Guo, J.; Yu, D.; Xiao, Y.; Wang, H.; Li, Y. Helicobacter pylori infection alters gastric and tongue coating microbial communities. Helicobacter 2019, 24, e12567. [Google Scholar] [CrossRef] [Green Version]
  19. Ogaya, Y.; Nomura, R.; Watanabe, Y.; Nakano, K. Detection of Helicobacter pylori DNA in inflamed dental pulp specimens from Japanese children and adolescents. J. Med. Microbiol. 2015, 64, 117–123. [Google Scholar] [CrossRef] [Green Version]
  20. Nomura, R.; Ogaya, Y.; Matayoshi, S.; Morita, Y.; Nakano, K. Molecular and clinical analyses of Helicobacter pylori colonization in inflamed dental pulp. BMC Oral Health 2018, 18, 64. [Google Scholar] [CrossRef]
  21. Hirsch, C.; Tegtmeyer, N.; Rohde, M.; Rowland, M.; Oyarzabal, O.A.; Backert, S. Live Helicobacter pylori in the root canal of endodontic-infected deciduous teeth. J. Gastroenterol. 2012, 47, 936–940. [Google Scholar] [CrossRef]
  22. Sruthi, M.A.; Mani, G.; Ramakrishnan, M.; Selvaraj, J. Dental caries as a source of Helicobacter pylori infection in children. Int. J. Paediatr. Dent. 2022. [Google Scholar] [CrossRef]
  23. Krajden, S.; Fuksa, M.; Anderson, J.; Kempston, J.; Boccia, A.; Petrea, C.; Babida, C.; Karmali, M.; Penner, J.L. Examination of human stomach biopsies, saliva, and dental plaque for Campylobacter pylori. J. Clin. Microbiol. 1989, 27, 1397–1398. [Google Scholar] [CrossRef] [Green Version]
  24. Navabi, N.; Aramon, M.; Mirzazadeh, A. Does the presence of the Helicobacter pylori in the dental plaque associate with its gastric infection? A meta-analysis and systematic review. Dent. Res. J. 2011, 8, 178–182. [Google Scholar] [CrossRef]
  25. Roman-Roman, A.; Giono-Cerezo, S.; Camorlinga-Ponce, M.; Martinez-Carrillo, D.N.; Loaiza-Loeza, S.; Fernandez-Tilapa, G. vacA genotypes of Helicobacter pylori in the oral cavity and stomach of patients with chronic gastritis and gastric ulcer. Enferm Infecc. Microbiol. Clin. 2013, 31, 130–135. [Google Scholar] [CrossRef]
  26. Aksit Bicak, D.; Akyuz, S.; Kiratli, B.; Usta, M.; Urganci, N.; Alev, B.; Yarat, A.; Sahin, F. The investigation of Helicobacter pylori in the dental biofilm and saliva samples of children with dyspeptic complaints. BMC Oral Health 2017, 17, 67. [Google Scholar] [CrossRef] [Green Version]
  27. Song, Q.; Lange, T.; Spahr, A.; Adler, G.; Bode, G. Characteristic distribution pattern of Helicobacter pylori in dental plaque and saliva detected with nested PCR. J. Med. Microbiol. 2000, 49, 349–353. [Google Scholar] [CrossRef]
  28. Tongtawee, T.; Wattanawongdon, W.; Simawaranon, T. Effects of periodontal therapy on eradication and recurrence of Helicobacter pylori infection after successful treatment. J. Int. Med. Res. 2019, 47, 875–883. [Google Scholar] [CrossRef] [Green Version]
  29. Cai, H.; Li, W.; Shu, X.; Peng, K.; Zhang, Y.; Jiang, M. Genetic variation of Helicobacter pylori in the oral cavity and stomach detected using thymine adenine cloning in children with chronic gastritis. Pediatr. Infect. Dis. J. 2014, 33, e1–e6. [Google Scholar] [CrossRef]
  30. Leszczynska, K.; Namiot, D.B.; Namiot, Z.; Leszczynska, J.K.; Jakoniuk, P.; Kemona, A. Application of immunoassay for detection of Helicobacter pylori antigens in the dental plaque. Adv. Med. Sci. 2009, 54, 194–198. [Google Scholar] [CrossRef]
  31. Anand, P.S.; Nandakumar, K.; Shenoy, K.T. Are dental plaque, poor oral hygiene, and periodontal disease associated with Helicobacter pylori infection? J. Periodontol. 2006, 77, 692–698. [Google Scholar] [CrossRef] [PubMed]
  32. Al Asqah, M.; Al Hamoudi, N.; Anil, S.; Al Jebreen, A.; Al-Hamoudi, W.K. Is the presence of Helicobacter pylori in dental plaque of patients with chronic periodontitis a risk factor for gastric infection? Can. J. Gastroenterol. 2009, 23, 177–179. [Google Scholar] [CrossRef] [Green Version]
  33. Liu, Y.; Yue, H.; Li, A.; Wang, J.; Jiang, B.; Zhang, Y.; Bai, Y. An epidemiologic study on the correlation between oral Helicobacter pylori and gastric H. pylori. Curr. Microbiol. 2009, 58, 449–453. [Google Scholar] [CrossRef]
  34. Teoman, I.; Ozmeric, N.; Ozcan, G.; Alaaddinoglu, E.; Dumlu, S.; Akyon, Y.; Balos, K. Comparison of different methods to detect Helicobacter pylori in the dental plaque of dyspeptic patients. Clin. Oral. Investig. 2007, 11, 201–205. [Google Scholar] [CrossRef]
  35. Medina, M.L.; Medina, M.G.; Merino, L.A. Correlation between virulence markers of Helicobacter pylori in the oral cavity and gastric biopsies. Arq. Gastroenterol. 2017, 54, 217–221. [Google Scholar] [CrossRef] [Green Version]
  36. Rasmussen, L.T.; de Labio, R.W.; Neto, A.C.; Silva, L.C.; Queiroz, V.F.; Smith, M.A.C.; Payao, S.L.M. Detection of Helicobacter pylori in gastric biopsies, saliva and dental plaques of dyspeptic patients from Marilia, Sao Paulo, Brazil: Presence of vacA and cagA genes. J. Venom. Anim. Toxins Incl. Trop. Dis. 2012, 18, 180–187. [Google Scholar] [CrossRef] [Green Version]
  37. Silva, D.G.; Stevens, R.H.; Macedo, J.M.; Albano, R.M.; Falabella, M.E.; Veerman, E.C.; Tinoco, E.M. Detection of cytotoxin genotypes of Helicobacter pylori in stomach, saliva and dental plaque. Arch. Oral. Biol. 2009, 54, 684–688. [Google Scholar] [CrossRef]
  38. Bago, I.; Bago, J.; Plecko, V.; Aurer, A.; Majstorovic, K.; Budimir, A. The effectiveness of systemic eradication therapy against oral Helicobacter pylori. J. Oral. Pathol. Med. 2011, 40, 428–432. [Google Scholar] [CrossRef]
  39. Liu, Y.; Li, R.; Xue, X.; Xu, T.; Luo, Y.; Dong, Q.; Liu, J.; Liu, J.; Pan, Y.; Zhang, D. Periodontal disease and Helicobacter pylori infection in oral cavity: A meta-analysis of 2727 participants mainly based on Asian studies. Clin. Oral. Investig. 2020, 24, 2175–2188. [Google Scholar] [CrossRef]
  40. Hu, Z.; Zhang, Y.; Li, Z.; Yu, Y.; Kang, W.; Han, Y.; Geng, X.; Ge, S.; Sun, Y. Effect of Helicobacter pylori infection on chronic periodontitis by the change of microecology and inflammation. Oncotarget 2016, 7, 66700–66712. [Google Scholar] [CrossRef]
  41. Soto, C.; Rojas, V.; Yáñez, L.; Hidalgo, A.; Olivera, M.; Pacheco, M.; Venegas, D.; Salinas, D.; Bravo, D.; Quest, A.F.G. Porphyromonas gingivalis-Helicobacter pylori co-incubation enhances Porphyromonas gingivalis virulence and increases migration of infected human oral keratinocytes. J. Oral. Microbiol. 2022, 14, 2107691. [Google Scholar] [CrossRef] [PubMed]
  42. Li, S.; Zhang, Y.; Yang, Z.; Li, J.; Li, Y.; Li, H.; Li, W.; Jia, J.; Ge, S.; Sun, Y. Helicobacter pylori infection is correlated with the incidence of erosive oral lichen planus and the alteration of the oral microbiome composition. BMC Microbiol. 2021, 21, 122. [Google Scholar] [CrossRef] [PubMed]
  43. Kazanowska-Dygdala, M.; Dus, I.; Radwan-Oczko, M. The presence of Helicobacter pylori in oral cavities of patients with leukoplakia and oral lichen planus. J. Appl. Oral. Sci. 2016, 24, 18–23. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Gupta, A.A.; Kheur, S.; Raj, A.T.; Mahajan, P. Association of Helicobacter pylori with oral potentially malignant disorders and oral squamous cell carcinoma-a systematic review and meta-analysis. Clin. Oral. Investig. 2020, 24, 13–23. [Google Scholar] [CrossRef] [PubMed]
  45. Meng, X.; Wang, Q.; He, C.; Chen, M.; Liu, J.; Liu, W.; Yuan, Y. An inverse association of Helicobacter pylori infection with oral squamous cell carcinoma. J. Oral. Pathol. Med. 2016, 45, 17–22. [Google Scholar] [CrossRef]
  46. Hulimavu, S.R.; Mohanty, L.; Tondikulam, N.V.; Shenoy, S.; Jamadar, S.; Bhadranna, A. No evidence for Helicobacter pylori in oral lichen planus. J. Oral. Pathol. Med. 2014, 43, 576–578. [Google Scholar] [CrossRef]
  47. Zou, Q.H.; Li, R.Q. Helicobacter pylori in the oral cavity and gastric mucosa: A meta-analysis. J. Oral. Pathol. Med. 2011, 40, 317–324. [Google Scholar] [CrossRef]
  48. Gebara, E.C.; Faria, C.M.; Pannuti, C.; Chehter, L.; Mayer, M.P.; Lima, L.A. Persistence of Helicobacter pylori in the oral cavity after systemic eradication therapy. J. Clin. Periodontol. 2006, 33, 329–333. [Google Scholar] [CrossRef]
  49. Ren, Q.; Yan, X.; Zhou, Y.; Li, W.X. Periodontal therapy as adjunctive treatment for gastric Helicobacter pylori infection. Cochrane Database Syst. Rev. 2016, 2, CD009477. [Google Scholar] [CrossRef]
  50. Ozturk, A. Periodontal Treatment Is Associated With Improvement in Gastric Helicobacter pylori Eradication: An Updated Meta-analysis of Clinical Trials. Int. Dent. J. 2021, 71, 188–196. [Google Scholar] [CrossRef]
  51. Yuksel Sert, S.; Ozturk, A.; Bektas, A.; Cengiz, M.I. Periodontal treatment is more effective in gastric Helicobacter pylori eradication in those patients who maintain good oral hygiene. Int. Dent. J. 2019, 69, 392–399. [Google Scholar] [CrossRef]
  52. Song, H.Y.; Li, Y. Can eradication rate of gastric Helicobacter pylori be improved by killing oral Helicobacter pylori? World J. Gastroenterol. 2013, 19, 6645–6650. [Google Scholar] [CrossRef]
  53. Zaric, S.; Bojic, B.; Jankovic, L.; Dapcevic, B.; Popovic, B.; Cakic, S.; Milasin, J. Periodontal therapy improves gastric Helicobacter pylori eradication. J. Dent. Res. 2009, 88, 946–950. [Google Scholar] [CrossRef]
  54. Gao, J.; Li, Y.; Wang, Q.; Qi, C.; Zhu, S. Correlation between distribution of Helicobacter pylori in oral cavity and chronic stomach conditions. J. Huazhong Univ. Sci. Technol. Med. Sci. 2011, 31, 409–412. [Google Scholar] [CrossRef]
  55. Miller, A.K.; Williams, S.M. Helicobacter pylori infection causes both protective and deleterious effects in human health and disease. Genes Immun. 2021, 22, 218–226. [Google Scholar] [CrossRef]
  56. Yamaoka, Y. Mechanisms of disease: Helicobacter pylori virulence factors. Nat. Rev. Gastroenterol. Hepatol. 2010, 7, 629–641. [Google Scholar] [CrossRef] [Green Version]
  57. Alexander, S.M.; Retnakumar, R.J.; Chouhan, D.; Devi, T.N.B.; Dharmaseelan, S.; Devadas, K.; Thapa, N.; Tamang, J.P.; Lamtha, S.C.; Chattopadhyay, S. Helicobacter pylori in Human Stomach: The Inconsistencies in Clinical Outcomes and the Probable Causes. Front. Microbiol. 2021, 12, 713955. [Google Scholar] [CrossRef]
  58. Wen, J.; Lau, H.C.; Peppelenbosch, M.; Yu, J. Gastric Microbiota beyond H. pylori: An Emerging Critical Character in Gastric Carcinogenesis. Biomedicines 2021, 9, 1680. [Google Scholar] [CrossRef]
  59. Freire, M.; Nelson, K.E.; Edlund, A. The Oral Host-Microbial Interactome: An Ecological Chronometer of Health? Trends Microbiol. 2021, 29, 551–561. [Google Scholar] [CrossRef]
  60. Bakhti, S.Z.; Latifi-Navid, S. Oral microbiota and Helicobacter pylori in gastric carcinogenesis: What do we know and where next? BMC Microbiol. 2021, 21, 71. [Google Scholar] [CrossRef]
  61. Maixner, F.; Krause-Kyora, B.; Turaev, D.; Herbig, A.; Hoopmann, M.R.; Hallows, J.L.; Kusebauch, U.; Vigl, E.E.; Malfertheiner, P.; Megraud, F.; et al. The 5300-year-old Helicobacter pylori genome of the Iceman. Science 2016, 351, 162–165. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Cao, D.M.; Lu, Q.F.; Li, S.B.; Wang, J.P.; Chen, Y.L.; Huang, Y.Q.; Bi, H.K. Comparative Genomics of H. pylori and Non-Pylori Helicobacter Species to Identify New Regions Associated with Its Pathogenicity and Adaptability. Biomed. Res. Int. 2016, 2016, 6106029. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Noto, J.M.; Chopra, A.; Loh, J.T.; Romero-Gallo, J.; Piazuelo, M.B.; Watson, M.; Leary, S.; Beckett, A.C.; Wilson, K.T.; Cover, T.L.; et al. Pan-genomic analyses identify key Helicobacter pylori pathogenic loci modified by carcinogenic host microenvironments. Gut 2018, 67, 1793–1804. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Falush, D.; Wirth, T.; Linz, B.; Pritchard, J.K.; Stephens, M.; Kidd, M.; Blaser, M.J.; Graham, D.Y.; Vacher, S.; Perez-Perez, G.I.; et al. Traces of human migrations in Helicobacter pylori populations. Science 2003, 299, 1582–1585. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Munoz-Ramirez, Z.Y.; Pascoe, B.; Mendez-Tenorio, A.; Mourkas, E.; Sandoval-Motta, S.; Perez-Perez, G.; Morgan, D.R.; Dominguez, R.L.; Ortiz-Princz, D.; Cavazza, M.E.; et al. A 500-year tale of co-evolution, adaptation, and virulence: Helicobacter pylori in the Americas. ISME J. 2021, 15, 78–92. [Google Scholar] [CrossRef]
  66. Chang, W.L.; Yeh, Y.C.; Sheu, B.S. The impacts of H. pylori virulence factors on the development of gastroduodenal diseases. J. Biomed. Sci. 2018, 25, 68. [Google Scholar] [CrossRef] [Green Version]
  67. Kao, C.Y.; Sheu, B.S.; Wu, J.J. Helicobacter pylori infection: An overview of bacterial virulence factors and pathogenesis. Biomed. J. 2016, 39, 14–23. [Google Scholar] [CrossRef] [Green Version]
  68. Doohan, D.; Rezkitha, Y.A.A.; Waskito, L.A.; Yamaoka, Y.; Miftahussurur, M. Helicobacter pylori BabA-SabA Key Roles in the Adherence Phase: The Synergic Mechanism for Successful Colonization and Disease Development. Toxins 2021, 13, 485. [Google Scholar] [CrossRef]
  69. Sekar, R.; Murali, P.; Junaid, M. Quantification of Helicobacter pylori and its oncoproteins in the oral cavity: A cross-sectional study. Oral. Dis. 2022. [Google Scholar] [CrossRef]
  70. Olbermann, P.; Josenhans, C.; Moodley, Y.; Uhr, M.; Stamer, C.; Vauterin, M.; Suerbaum, S.; Achtman, M.; Linz, B. A global overview of the genetic and functional diversity in the Helicobacter pylori cag pathogenicity island. PLoS Genet. 2010, 6, e1001069. [Google Scholar] [CrossRef]
  71. Plummer, M.; van Doorn, L.J.; Franceschi, S.; Kleter, B.; Canzian, F.; Vivas, J.; Lopez, G.; Colin, D.; Munoz, N.; Kato, I. Helicobacter pylori cytotoxin-associated genotype and gastric precancerous lesions. J. Natl. Cancer Inst. 2007, 99, 1328–1334. [Google Scholar] [CrossRef]
  72. Higashi, H.; Tsutsumi, R.; Fujita, A.; Yamazaki, S.; Asaka, M.; Azuma, T.; Hatakeyama, M. Biological activity of the Helicobacter pylori virulence factor CagA is determined by variation in the tyrosine phosphorylation sites. Proc. Natl. Acad. Sci. USA 2002, 99, 14428–14433. [Google Scholar] [CrossRef] [Green Version]
  73. Zeng, J.; Xie, C.; Zhang, L.; Liu, X.; Chan, M.T.V.; Wu, W.K.K.; Chen, H. Host Cell Antimicrobial Responses against Helicobacter pylori Infection: From Biological Aspects to Therapeutic Strategies. Int. J. Mol. Sci. 2022, 23, 941. [Google Scholar] [CrossRef]
  74. Loh, J.T.; Shaffer, C.L.; Piazuelo, M.B.; Bravo, L.E.; McClain, M.S.; Correa, P.; Cover, T.L. Analysis of cagA in Helicobacter pylori strains from Colombian populations with contrasting gastric cancer risk reveals a biomarker for disease severity. Cancer Epidemiol. Biomark. Prev. 2011, 20, 2237–2249. [Google Scholar] [CrossRef] [Green Version]
  75. Ferreira, R.M.; Pinto-Ribeiro, I.; Wen, X.; Marcos-Pinto, R.; Dinis-Ribeiro, M.; Carneiro, F.; Figueiredo, C. Helicobacter pylori cagA Promoter Region Sequences Influence CagA Expression and Interleukin 8 Secretion. J. Infect. Dis. 2016, 213, 669–673. [Google Scholar] [CrossRef] [Green Version]
  76. Forsyth, M.H.; Atherton, J.C.; Blaser, M.J.; Cover, T.L. Heterogeneity in levels of vacuolating cytotoxin gene (vacA) transcription among Helicobacter pylori strains. Infect. Immun. 1998, 66, 3088–3094. [Google Scholar] [CrossRef] [Green Version]
  77. Mark Welch, J.L.; Rossetti, B.J.; Rieken, C.W.; Dewhirst, F.E.; Borisy, G.G. Biogeography of a human oral microbiome at the micron scale. Proc. Natl. Acad. Sci. USA 2016, 113, E791–E800. [Google Scholar] [CrossRef] [Green Version]
  78. Zijnge, V.; van Leeuwen, M.B.; Degener, J.E.; Abbas, F.; Thurnheer, T.; Gmur, R.; Harmsen, H.J. Oral biofilm architecture on natural teeth. PLoS ONE 2010, 5, e9321. [Google Scholar] [CrossRef] [Green Version]
  79. Bowen, W.H.; Burne, R.A.; Wu, H.; Koo, H. Oral Biofilms: Pathogens, Matrix, and Polymicrobial Interactions in Microenvironments. Trends Microbiol. 2018, 26, 229–242. [Google Scholar] [CrossRef]
  80. Huang, R.; Li, M.; Gregory, R.L. Bacterial interactions in dental biofilm. Virulence 2011, 2, 435–444. [Google Scholar] [CrossRef]
  81. Donlan, R.M.; Costerton, J.W. Biofilms: Survival mechanisms of clinically relevant microorganisms. Clin. Microbiol. Rev. 2002, 15, 167–193. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Iwai, K.; Watanabe, I.; Yamamoto, T.; Kuriyama, N.; Matsui, D.; Nomura, R.; Ogaya, Y.; Oseko, F.; Adachi, K.; Takizawa, S.; et al. Association between Helicobacter pylori infection and dental pulp reservoirs in Japanese adults. BMC Oral Health 2019, 19, 267. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Tan, S.; Noto, J.M.; Romero-Gallo, J.; Peek, R.M., Jr.; Amieva, M.R. Helicobacter pylori perturbs iron trafficking in the epithelium to grow on the cell surface. PLoS Pathog. 2011, 7, e1002050. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Tan, S.; Tompkins, L.S.; Amieva, M.R. Helicobacter pylori usurps cell polarity to turn the cell surface into a replicative niche. PLoS Pathog. 2009, 5, e1000407. [Google Scholar] [CrossRef] [Green Version]
  85. Koniger, V.; Holsten, L.; Harrison, U.; Busch, B.; Loell, E.; Zhao, Q.; Bonsor, D.A.; Roth, A.; Kengmo-Tchoupa, A.; Smith, S.I.; et al. Helicobacter pylori exploits human CEACAMs via HopQ for adherence and translocation of CagA. Nat. Microbiol. 2016, 2, 16188. [Google Scholar] [CrossRef]
  86. Bonsor, D.A.; Zhao, Q.; Schmidinger, B.; Weiss, E.; Wang, J.; Deredge, D.; Beadenkopf, R.; Dow, B.; Fischer, W.; Beckett, D.; et al. The Helicobacter pylori adhesin protein HopQ exploits the dimer interface of human CEACAMs to facilitate translocation of the oncoprotein CagA. EMBO J. 2018, 37, e98664. [Google Scholar] [CrossRef]
  87. Tegtmeyer, N.; Ghete, T.D.; Schmitt, V.; Remmerbach, T.; Cortes, M.C.C.; Bondoc, E.M.; Graf, H.L.; Singer, B.B.; Hirsch, C.; Backert, S. Type IV secretion of Helicobacter pylori CagA into oral epithelial cells is prevented by the absence of CEACAM receptor expression. Gut. Pathog. 2020, 12, 25. [Google Scholar] [CrossRef]
  88. Mima, J.; Koshino, A.; Oka, K.; Uchida, H.; Hieda, Y.; Nohara, K.; Kogo, M.; Chai, Y.; Sakai, T. Regulation of the epithelial adhesion molecule CEACAM1 is important for palate formation. PLoS ONE 2013, 8, e61653. [Google Scholar] [CrossRef] [Green Version]
  89. Wang, F.F.; Guan, B.X.; Yang, J.Y.; Wang, H.T.; Zhou, C.J. CEACAM1 is overexpressed in oral tumors and related to tumorigenesis. Med. Mol. Morphol. 2017, 50, 42–51. [Google Scholar] [CrossRef]
  90. Huynh-Torlakovic, H.; Bjerkan, L.; Schenck, K.; Blix, I.J. Distribution of carcinoembryonic antigen-related cellular adhesion molecules in human gingiva. Eur. J. Oral Sci. 2012, 120, 395–401. [Google Scholar] [CrossRef]
  91. Liu, G.X.; Xie, Q.; Zhou, C.J.; Zhang, X.Y.; Ma, B.L.; Wang, C.Q.; Wei, F.C.; Qu, X.; Sun, S.Z. The possible roles of OPN-regulated CEACAM1 expression in promoting the survival of activated T cells and the apoptosis of oral keratinocytes in oral lichen planus patients. J. Clin. Immunol. 2011, 31, 827–839. [Google Scholar] [CrossRef]
  92. Boyle, J.O.; Gumus, Z.H.; Kacker, A.; Choksi, V.L.; Bocker, J.M.; Zhou, X.K.; Yantiss, R.K.; Hughes, D.B.; Du, B.; Judson, B.L.; et al. Effects of cigarette smoke on the human oral mucosal transcriptome. Cancer Prev. Res. 2010, 3, 266–278. [Google Scholar] [CrossRef] [Green Version]
  93. Gray-Owen, S.D.; Blumberg, R.S. CEACAM1: Contact-dependent control of immunity. Nat. Rev. Immunol. 2006, 6, 433–446. [Google Scholar] [CrossRef]
  94. Gur, C.; Maalouf, N.; Gerhard, M.; Singer, B.B.; Emgard, J.; Temper, V.; Neuman, T.; Mandelboim, O.; Bachrach, G. The Helicobacter pylori HopQ outermembrane protein inhibits immune cell activities. Oncoimmunology 2019, 8, e1553487. [Google Scholar] [CrossRef]
  95. Petersen, A.M.; Krogfelt, K.A. Helicobacter pylori: An invading microorganism? A review. FEMS Immunol. Med. Microbiol. 2003, 36, 117–126. [Google Scholar] [CrossRef] [Green Version]
  96. Dubois, A.; Boren, T. Helicobacter pylori is invasive and it may be a facultative intracellular organism. Cell Microbiol. 2007, 9, 1108–1116. [Google Scholar] [CrossRef] [Green Version]
  97. Chu, Y.T.; Wang, Y.H.; Wu, J.J.; Lei, H.Y. Invasion and multiplication of Helicobacter pylori in gastric epithelial cells and implications for antibiotic resistance. Infect. Immun. 2010, 78, 4157–4165. [Google Scholar] [CrossRef] [Green Version]
  98. Li, H.; Liang, D.; Hu, N.; Dai, X.; He, J.; Zhuang, H.; Zhao, W. Helicobacter pylori inhibited cell proliferation in human periodontal ligament fibroblasts through the Cdc25C/CDK1/cyclinB1 signaling cascade. J. Periodontal. Implant. Sci. 2019, 49, 138–147. [Google Scholar] [CrossRef]
  99. Sit, W.Y.; Chen, Y.A.; Chen, Y.L.; Lai, C.H.; Wang, W.C. Cellular evasion strategies of Helicobacter pylori in regulating its intracellular fate. Semin. Cell. Dev. Biol. 2020, 101, 59–67. [Google Scholar] [CrossRef]
  100. Huang, Y.; Wang, Q.L.; Cheng, D.D.; Xu, W.T.; Lu, N.H. Adhesion and Invasion of Gastric Mucosa Epithelial Cells by Helicobacter pylori. Front. Cell Infect. Microbiol. 2016, 6, 159. [Google Scholar] [CrossRef]
  101. Cheok, Y.Y.; Lee, C.Y.Q.; Cheong, H.C.; Vadivelu, J.; Looi, C.Y.; Abdullah, S.; Wong, W.F. An Overview of Helicobacter pylori Survival Tactics in the Hostile Human Stomach Environment. Microorganisms 2021, 9, 2502. [Google Scholar] [CrossRef] [PubMed]
  102. 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] [PubMed] [Green Version]
  103. Schmidinger, B.; Petri, K.; Lettl, C.; Li, H.; Namineni, S.; Ishikawa-Ankerhold, H.; Jimenez-Soto, L.F.; Haas, R. Helicobacter pylori binds human Annexins via Lipopolysaccharide to interfere with Toll-like Receptor 4 signaling. PLoS Pathog. 2022, 18, e1010326. [Google Scholar] [CrossRef] [PubMed]
  104. Behrens, I.K.; Busch, B.; Ishikawa-Ankerhold, H.; Palamides, P.; Shively, J.E.; Stanners, C.; Chan, C.; Leung, N.; Gray-Owen, S.; Haas, R. The HopQ-CEACAM Interaction Controls CagA Translocation, Phosphorylation, and Phagocytosis of Helicobacter pylori in Neutrophils. mBio 2020, 11. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Martinez, F.O.; Gordon, S. The M1 and M2 paradigm of macrophage activation: Time for reassessment. F1000Prime Rep. 2014, 6, 13. [Google Scholar] [CrossRef] [Green Version]
  106. Cheok, Y.Y.; Tan, G.M.Y.; Lee, C.Y.Q.; Abdullah, S.; Looi, C.Y.; Wong, W.F. Innate Immunity Crosstalk with Helicobacter pylori: Pattern Recognition Receptors and Cellular Responses. Int. J. Mol. Sci. 2022, 23, 7561. [Google Scholar] [CrossRef]
  107. Go, D.M.; Lee, S.H.; Lee, S.H.; Woo, S.H.; Kim, K.; Kim, K.; Park, K.S.; Park, J.H.; Ha, S.J.; Kim, W.H.; et al. Programmed Death Ligand 1-Expressing Classical Dendritic Cells Mitigate Helicobacter-Induced Gastritis. Cell Mol. Gastroenterol. Hepatol. 2021, 12, 715–739. [Google Scholar] [CrossRef]
  108. Javed, S.; Skoog, E.C.; Solnick, J.V. Impact of Helicobacter pylori Virulence Factors on the Host Immune Response and Gastric Pathology. Curr. Top. Microbiol. Immunol. 2019, 421, 21–52. [Google Scholar] [CrossRef]
  109. Liu, Y.; Lin, H.; Bai, Y.; Qin, X.; Zheng, X.; Sun, Y.; Zhang, Y. Study on the relationship between Helicobacter pylori in the dental plaque and the occurrence of dental caries or oral hygiene index. Helicobacter 2008, 13, 256–260. [Google Scholar] [CrossRef]
  110. Souto, R.; Colombo, A.P. Detection of Helicobacter pylori by polymerase chain reaction in the subgingival biofilm and saliva of non-dyspeptic periodontal patients. J. Periodontol. 2008, 79, 97–103. [Google Scholar] [CrossRef]
  111. Nomura, R.; Kadota, T.; Ogaya, Y.; Matayoshi, S.; Iwashita, N.; Okawa, R.; Nakano, K. Contribution of Streptococcus mutans to Helicobacter pylori colonisation in oral cavity and gastric tissue. Sci. Rep. 2020, 10, 12540. [Google Scholar] [CrossRef]
  112. Zhang, W.; Deng, X.; Zhou, X.; Hao, Y.; Li, Y. Influence of Helicobacter pylori culture supernatant on the ecological balance of a dual-species oral biofilm. J. Appl. Oral. Sci. 2018, 26, e20170113. [Google Scholar] [CrossRef] [Green Version]
  113. Chen, X.; Zhou, X.; Liao, B.; Zhou, Y.; Cheng, L.; Ren, B. The cross-kingdom interaction between Helicobacter pylori and Candida albicans. PLoS Pathog. 2021, 17, e1009515. [Google Scholar] [CrossRef]
  114. Kadota, T.; Hamada, M.; Nomura, R.; Ogaya, Y.; Okawa, R.; Uzawa, N.; Nakano, K. Distribution of Helicobacter pylori and Periodontopathic Bacterial Species in the Oral Cavity. Biomedicines 2020, 8, 161. [Google Scholar] [CrossRef]
  115. Andersen, R.N.; Ganeshkumar, N.; Kolenbrander, P.E. Helicobacter pylori adheres selectively to Fusobacterium spp. Oral Microbiol. Immunol. 1998, 13, 51–54. [Google Scholar] [CrossRef]
  116. Sanchez-Alonzo, K.; Silva-Mieres, F.; Arellano-Arriagada, L.; Parra-Sepulveda, C.; Bernasconi, H.; Smith, C.T.; Campos, V.L.; Garcia-Cancino, A. Nutrient Deficiency Promotes the Entry of Helicobacter pylori Cells into Candida Yeast Cells. Biology 2021, 10, 426. [Google Scholar] [CrossRef]
  117. Palencia, S.L.; Garcia, A.; Palencia, M. Multiple surface interaction mechanisms direct the anchoring, co-aggregation and formation of dual-species biofilm between Candida albicans and Helicobacter pylori. J. Adv. Res. 2022, 35, 169–185. [Google Scholar] [CrossRef]
  118. Saniee, P.; Siavoshi, F.; Nikbakht Broujeni, G.; Khormali, M.; Sarrafnejad, A.; Malekzadeh, R. Localization of H.pylori within the vacuole of Candida yeast by direct immunofluorescence technique. Arch. Iran. Med. 2013, 16, 705–710. [Google Scholar]
  119. Saniee, P.; Siavoshi, F. Endocytotic uptake of FITC-labeled anti-H. pylori egg yolk immunoglobulin Y in Candida yeast for detection of intracellular H. pylori. Front. Microbiol. 2015, 6, 113. [Google Scholar] [CrossRef] [Green Version]
  120. Sanchez-Alonzo, K.; Belmar, L.; Parra-Sepulveda, C.; Bernasconi, H.; Campos, V.L.; Smith, C.T.; Saez, K.; Garcia-Cancino, A. Antibiotics as a Stressing Factor Triggering the Harboring of Helicobacter pylori J99 within Candida albicans ATCC10231. Pathogens 2021, 10, 382. [Google Scholar] [CrossRef]
  121. Sanchez-Alonzo, K.; Parra-Sepulveda, C.; Vega, S.; Bernasconi, H.; Campos, V.L.; Smith, C.T.; Saez, K.; Garcia-Cancino, A. In Vitro Incorporation of Helicobacter pylori into Candida albicans Caused by Acidic pH Stress. Pathogens 2020, 9, 489. [Google Scholar] [CrossRef] [PubMed]
  122. Heydari, S.; Siavoshi, F.; Jazayeri, M.H.; Sarrafnejad, A.; Saniee, P. Helicobacter pylori release from yeast as a vesicle-encased or free bacterium. Helicobacter 2020, 25, e12725. [Google Scholar] [CrossRef] [PubMed]
  123. Okuda, K.; Ishihara, K.; Miura, T.; Katakura, A.; Noma, H.; Ebihara, Y. Helicobacter pylori may have only a transient presence in the oral cavity and on the surface of oral cancer. Microbiol. Immunol. 2000, 44, 385–388. [Google Scholar] [CrossRef] [PubMed]
  124. Contaldo, M.; Fusco, A.; Stiuso, P.; Lama, S.; Gravina, A.G.; Itro, A.; Federico, A.; Itro, A.; Dipalma, G.; Inchingolo, F.; et al. Oral Microbiota and Salivary Levels of Oral Pathogens in Gastro-Intestinal Diseases: Current Knowledge and Exploratory Study. Microorganisms 2021, 9, 1064. [Google Scholar] [CrossRef] [PubMed]
  125. Tefiku, U.; Popovska, M.; Cana, A.; Zendeli-Bedxeti, L.; Recica, B.; Spasovska-Gjorgovska, A.; Spasovski, S. Determination of the Role of Fusobacterium Nucleatum in the Pathogenesis in and Out the Mouth. Prilozi 2020, 41, 87–99. [Google Scholar] [CrossRef] [PubMed]
  126. Lima, B.P.; Shi, W.; Lux, R. Identification and characterization of a novel Fusobacterium nucleatum adhesin involved in physical interaction and biofilm formation with Streptococcus gordonii. Microbiologyopen 2017, 6, e00444. [Google Scholar] [CrossRef]
  127. Massarrat, S.; Saniee, P.; Siavoshi, F.; Mokhtari, R.; Mansour-Ghanaei, F.; Khalili-Samani, S. The Effect of Helicobacter pylori Infection, Aging, and Consumption of Proton Pump Inhibitor on Fungal Colonization in the Stomach of Dyspeptic Patients. Front. Microbiol. 2016, 7, 801. [Google Scholar] [CrossRef]
  128. Siavoshi, F.; Taghikhani, A.; Malekzadeh, R.; Sarrafnejad, A.; Kashanian, M.; Jamal, A.S.; Saniee, P.; Sadeghi, S.; Sharifi, A.H. The role of mother’s oral and vaginal yeasts in transmission of Helicobacter pylori to neonates. Arch. Iran. Med. 2013, 16, 288–294. [Google Scholar]
  129. Fox, E.P.; Cowley, E.S.; Nobile, C.J.; Hartooni, N.; Newman, D.K.; Johnson, A.D. Anaerobic bacteria grow within Candida albicans biofilms and induce biofilm formation in suspension cultures. Curr. Biol. 2014, 24, 2411–2416. [Google Scholar] [CrossRef] [Green Version]
  130. Siavoshi, F.; Heydari, S.; Shafiee, M.; Ahmadi, S.; Saniee, P.; Sarrafnejad, A.; Kolahdoozan, S. Sequestration inside the yeast vacuole may enhance Helicobacter pylori survival against stressful condition. Infect. Genet. Evol. 2019, 69, 127–133. [Google Scholar] [CrossRef]
  131. Saniee, P.; Siavoshi, F.; Nikbakht Broujeni, G.; Khormali, M.; Sarrafnejad, A.; Malekzadeh, R. Immunodetection of Helicobacter pylori-specific proteins in oral and gastric Candida yeasts. Arch. Iran. Med. 2013, 16, 624–630. [Google Scholar]
  132. Siavoshi, F.; Sahraee, M.; Ebrahimi, H.; Sarrafnejad, A.; Saniee, P. Natural fruits, flowers, honey, and honeybees harbor Helicobacter pylori-positive yeasts. Helicobacter 2018, 23, e12471. [Google Scholar] [CrossRef]
  133. Siavoshi, F.; Salmanian, A.H.; Akbari, F.; Malekzadeh, R.; Massarrat, S. Detection of Helicobacter pylori-specific genes in the oral yeast. Helicobacter 2005, 10, 318–322. [Google Scholar] [CrossRef]
  134. Sanchez-Alonzo, K.; Matamala-Valdes, L.; Parra-Sepulveda, C.; Bernasconi, H.; Campos, V.L.; Smith, C.T.; Saez, K.; Garcia-Cancino, A. Intracellular Presence of Helicobacter pylori and Its Virulence-Associated Genotypes within the Vaginal Yeast of Term Pregnant Women. Microorganisms 2021, 9, 131. [Google Scholar] [CrossRef]
  135. Dong, K.; Pan, H.; Yang, D.; Rao, L.; Zhao, L.; Wang, Y.; Liao, X. Induction, detection, formation, and resuscitation of viable but non-culturable state microorganisms. Compr. Rev. Food Sci. Food Saf. 2020, 19, 149–183. [Google Scholar] [CrossRef] [Green Version]
  136. Hirukawa, S.; Sagara, H.; Kaneto, S.; Kondo, T.; Kiga, K.; Sanada, T.; Kiyono, H.; Mimuro, H. Characterization of morphological conversion of Helicobacter pylori under anaerobic conditions. Microbiol. Immunol. 2018, 62, 221–228. [Google Scholar] [CrossRef] [Green Version]
  137. Nilsson, H.O.; Blom, J.; Abu-Al-Soud, W.; Ljungh, A.A.; Andersen, L.P.; Wadstrom, T. Effect of cold starvation, acid stress, and nutrients on metabolic activity of Helicobacter pylori. Appl. Environ. Microbiol. 2002, 68, 11–19. [Google Scholar] [CrossRef] [Green Version]
  138. Mizoguchi, H.; Fujioka, T.; Kishi, K.; Nishizono, A.; Kodama, R.; Nasu, M. Diversity in protein synthesis and viability of Helicobacter pylori coccoid forms in response to various stimuli. Infect. Immun. 1998, 66, 5555–5560. [Google Scholar] [CrossRef] [Green Version]
  139. Adams, B.L.; Bates, T.C.; Oliver, J.D. Survival of Helicobacter pylori in a natural freshwater environment. Appl. Environ. Microbiol. 2003, 69, 7462–7466. [Google Scholar] [CrossRef] [Green Version]
  140. Saito, N.; Konishi, K.; Sato, F.; Kato, M.; Takeda, H.; Sugiyama, T.; Asaka, M. Plural transformation-processes from spiral to coccoid Helicobacter pylori and its viability. J. Infect. 2003, 46, 49–55. [Google Scholar] [CrossRef]
  141. Sisto, F.; Brenciaglia, M.I.; Scaltrito, M.M.; Dubini, F. Helicobacter pylori: ureA, cagA and vacA expression during conversion to the coccoid form. Int. J. Antimicrob. Agents 2000, 15, 277–282. [Google Scholar] [CrossRef]
  142. Poursina, F.; Fagri, J.; Mirzaei, N.; Safaei, H.G. Overexpression of spoT gene in coccoid forms of clinical Helicobacter pylori isolates. Folia Microbiol. 2018, 63, 459–465. [Google Scholar] [CrossRef] [PubMed]
  143. Moreno, Y.; Piqueres, P.; Alonso, J.L.; Jimenez, A.; Gonzalez, A.; Ferrus, M.A. Survival and viability of Helicobacter pylori after inoculation into chlorinated drinking water. Water Res. 2007, 41, 3490–3496. [Google Scholar] [CrossRef] [PubMed]
  144. Poursina, F.; Faghri, J.; Moghim, S.; Zarkesh-Esfahani, H.; Nasr-Esfahani, B.; Fazeli, H.; Hasanzadeh, A.; Safaei, H.G. Assessment of cagE and babA mRNA expression during morphological conversion of Helicobacter pylori from spiral to coccoid. Curr. Microbiol. 2013, 66, 406–413. [Google Scholar] [CrossRef] [PubMed]
  145. Piqueres, P.; Moreno, Y.; Alonso, J.L.; Ferrus, M.A. A combination of direct viable count and fluorescent in situ hybridization for estimating Helicobacter pylori cell viability. Res. Microbiol. 2006, 157, 345–349. [Google Scholar] [CrossRef] [PubMed]
  146. Quaglia, N.C.; Storelli, M.M.; Scardocchia, T.; Lattanzi, A.; Celano, G.V.; Monno, R.; Dambrosio, A. Helicobacter pylori: Survival in cultivable and non-cultivable form in artificially contaminated Mytilus galloprovincialis. Int. J. Food Microbiol. 2020, 312, 108363. [Google Scholar] [CrossRef]
  147. Cellini, L.; Allocati, N.; Angelucci, D.; Iezzi, T.; Di Campli, E.; Marzio, L.; Dainelli, B. Coccoid Helicobacter pylori not culturable in vitro reverts in mice. Microbiol. Immunol. 1994, 38, 843–850. [Google Scholar] [CrossRef]
  148. Boehnke, K.F.; Eaton, K.A.; Fontaine, C.; Brewster, R.; Wu, J.; Eisenberg, J.N.S.; Valdivieso, M.; Baker, L.H.; Xi, C. Reduced infectivity of waterborne viable but nonculturable Helicobacter pylori strain SS1 in mice. Helicobacter 2017, 22, e12391. [Google Scholar] [CrossRef] [Green Version]
  149. Wang, X.; Sturegard, E.; Rupar, R.; Nilsson, H.O.; Aleljung, P.A.; Carlen, B.; Willen, R.; Wadstrom, T. Infection of BALB/c A mice by spiral and coccoid forms of Helicobacter pylori. J. Med. Microbiol. 1997, 46, 657–663. [Google Scholar] [CrossRef] [Green Version]
  150. Kaprelyants, A.S.; Gottschal, J.C.; Kell, D.B. Dormancy in non-sporulating bacteria. FEMS Microbiol. Rev. 1993, 10, 271–285. [Google Scholar] [CrossRef]
  151. Sachidanandham, R.; Yew-Hoong Gin, K. A dormancy state in nonspore-forming bacteria. Appl. Microbiol. Biotechnol. 2009, 81, 927–941. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  152. Kell, D.B.; Kaprelyants, A.S.; Weichart, D.H.; Harwood, C.R.; Barer, M.R. Viability and activity in readily culturable bacteria: A review and discussion of the practical issues. Antonie Van Leeuwenhoek 1998, 73, 169–187. [Google Scholar] [CrossRef] [PubMed]
  153. Mukamolova, G.V.; Kaprelyants, A.S.; Kell, D.B.; Young, M. Adoption of the transiently non-culturable state--a bacterial survival strategy? Adv. Microb. Physiol. 2003, 47, 65–129. [Google Scholar] [CrossRef] [PubMed]
  154. Lopez-Brea, M.; Alarcon, T.; Domingo, D.; Diaz-Reganon, J. Inhibitory effect of Gram-negative and Gram-positive microorganisms against Helicobacter pylori clinical isolates. J. Antimicrob. Chemother. 2008, 61, 139–142. [Google Scholar] [CrossRef]
  155. Krzyzek, P.; Grande, R.; Migdal, P.; Paluch, E.; Gosciniak, G. Biofilm Formation as a Complex Result of Virulence and Adaptive Responses of Helicobacter pylori. Pathogens 2020, 9, 1062. [Google Scholar] [CrossRef]
Figure 1. The oral cavity is a potential source for H. pylori gastric reinfection. However, the oral cavity seems not to be an ideal habitat for H. pylori, owing to the unstable temperature, high O2 tension, and varied bacterial composition [13,14]. Therefore, survival strategies of living H. pylori in the oral cavity remain to be investigated. The green arrows symbolize the common transmission pathway of H. pylori. PCR, polymerase chain reaction; RUT, rapid urease test.
Figure 1. The oral cavity is a potential source for H. pylori gastric reinfection. However, the oral cavity seems not to be an ideal habitat for H. pylori, owing to the unstable temperature, high O2 tension, and varied bacterial composition [13,14]. Therefore, survival strategies of living H. pylori in the oral cavity remain to be investigated. The green arrows symbolize the common transmission pathway of H. pylori. PCR, polymerase chain reaction; RUT, rapid urease test.
Ijms 23 13646 g001
Figure 2. Potential survival strategies of H. pylori in the oral cavity. H. pylori can hide within dental plaque biofilm in caries cavities and periodontal pockets. Moreover, this organism has the ability to adhere to and invade host oral cells in these niches. Synergistic interaction with oral microorganisms and transition to a VBNC or dormant state may also help H. pylori adapt to adverse conditions in the oral cavity.
Figure 2. Potential survival strategies of H. pylori in the oral cavity. H. pylori can hide within dental plaque biofilm in caries cavities and periodontal pockets. Moreover, this organism has the ability to adhere to and invade host oral cells in these niches. Synergistic interaction with oral microorganisms and transition to a VBNC or dormant state may also help H. pylori adapt to adverse conditions in the oral cavity.
Ijms 23 13646 g002
Table 1. Summary of data regarding H. pylori coinfection in gastric biopsy and oral samples.
Table 1. Summary of data regarding H. pylori coinfection in gastric biopsy and oral samples.
Oral SampleSample SizeH. pylori Detection Method in Oral SamplesCoinfection RateRef.
Saliva689 H. pylori-associated gastritis patientsPCR-16S rRNA and ureA gene79.7%[28]
Saliva162 patients with
gastric disease
PCR24%; 51.1% agreement in vacA genotype in saliva and biopsy from the same patients[25]
Saliva300 patients with
gastric disease
PCR-ureC, cagA, and vacA gene10.72%; high homology (58%) in vacA genotype in saliva and gastric samples from the same patients[4]
Dental plaque235 patients with chronic gastritisPCR-16S rRNA56.52%[29]
Dental plaque164 dyspeptic patientsEnzyme Immunoassay82.1%[30]
Dental plaque65 patients with gastric H. pylori infection among 134 dyspeptic patientsRUT89.2% among gastric H. pylori-positive patients[31]
Subgingival plaque101 dyspeptic patientsRUT66%[32]
Subgingival plaque443 dyspeptic patientsNested PCR- 860bp fragment71.86%[33]
Subgingival plaque67 dyspeptic patientsPCR-ureA gene25.4%[34]
Dental plaque and saliva70 children with dyspepsiaPCR-16S rRNA and 23S rRNADental plaque (77.6%); saliva (75.9%)[26]
Dental plaque and saliva61 patients with dyspepsiaPCR-ureA gene50.8%; 38.7% genotype concordance between oral and gastric samples from the same patients[35]
Dental plaque and saliva62 patients with dyspepsiaPCR-16S-rRNAAll oral samples (68%)[36]
Dental plaque and saliva30 patients with gastric diseasePCR-cagA gene60%; 98% agreement between gastric DNA H. pylori sequence and their corresponding saliva or dental plaque DNA[37]
Supragingival plaque, subgingival plaque, and saliva56 gastric H. pylori-positive patients with periodontitisPCR-16S rDNAAll oral samples (41.1%); supragingival plaque (26.8%); subgingival plaque (30.4%); saliva (21.4%);[38]
ureA, α-subunit of the urease gene; cagA, cytotoxin-associated gene A; vacA, vacuolating cytotoxin A.
Table 2. Oral hygiene management strategies against H. pylori infection.
Table 2. Oral hygiene management strategies against H. pylori infection.
Oral Hygiene ManagementEradication TherapyEffects on Gastric H. pylori InfectionRef.
Scaling and/or combined with root planing and oral hygiene instructions on brushing with the modified Bass technique10-day course of triple therapy consisted of a PPI combined with amoxicillin (2 × 1 g daily) and clarithromycin (2 × 500 mg daily)The eradication rate in the combined therapy group was higher than that in the triple therapy only group (64.71% vs. 51.06%, respectively, p = 0.17).[51]
Scaling and root planing; oral hygiene instruction1-week triple therapy (esomeprazole 20 mg twice per day, clarithromycin 500 mg twice per day, or metronidazole 400 mg three times per day (if clarithromycin-resistant), as well as amoxicillin 1000 mg twice per day)The recurrence rate of gastric H. pylori in the combined therapy group was lower than that in the triple therapy only group (2.04% vs. 15.27%, respectively; OR 0.69; 95% CI 0.52 to 0.99; p = 0.001).[28]
Mouth rinse (0.02% tinidazole and 0.12% chlorhexidine) with 20 mL held in the mouth for 5 min for 10 d; ultrasonic periodontal scaling twice a monthTriple therapy consisted of amoxicillin (1.0 g) and esomeprazole (20 mg) twice a day and levofloxacin (0.5 g) once a day for 10 dThe eradication rate in the combined therapy group was higher than that in the triple therapy only group (94.7% vs. 78.4%, respectively, p = 0.012).[52]
Basic periodontal therapy during triple therapy7-day course of triple therapy consisted of amoxicillin 2 g/day (g/d), clarithromycin 1 g/d, and pantoprazole 80 mg/dThe eradication rate in the combined therapy group was higher than that in the triple therapy only group (77.3% vs. 47.6%, respectively, p = 0.044).[53]
Oral hygiene education, dental cleaning, and scaling14-day PPI or triple therapyThe eradication rate in the combined therapy group was higher than that in triple therapy only group (62.8% vs. 32.4%, respectively, p < 0. 05).[54]
Table 3. Interaction between H. pylori and oral microorganisms.
Table 3. Interaction between H. pylori and oral microorganisms.
Interaction TypeOral MicroorganismsInteraction between H. pylori and Oral MicroorganismsRef.
Mutualistic relationshipS. mutansH. pylori can penetrate the biofilm formed by S. mutans.[111]
S. mutans contributes to the formation of “hedgehog” in the dental plaque, which could generate a CO2-rich environment.[77]
F. nucleatumH. pylori can adhere to F. nucleatum and might help to colonize the dental plaque.[15,115]
P. gingivalisH. pylori has the ability to coaggregate with P. gingivalis.[14]
P. gingivalis with a specific filamentous appendage (fimA) genotype may be involved in the colonization by H. pylori.[114]
C. albicansH. pylori can adhere to yeast pseudohyphae.[116]
H. pylori can anchor on C. albicans and form polymicrobial biofilms.[117]
H. pylori can invade yeast cells.[118,119]
Nutrient deprivation, acidic pH, and amoxicillin may stimulate the entry of H. pylori into Candida.[116,120,121]
H. pylori entering yeast cells can propagate vertically to the vacuoles of progeny yeast cells.[118]
C. albicans releases H. pylori as a vesicle-encased or free bacterium, which may facilitate H. pylori invasion of new yeast cells.[122]
Antagonistic relationshipS. mitisThe diffusible factors released by S. mitis can inhibit the growth and induce the coccoid conversion of H. pylori during coculture in vitro.[13]
S. mutans, etc.Bacteriocin-like inhibitory proteins against H. pylori could be produced by oral bacteria.[14]
S. mutans and
Prevotella intermedia
Culture supernatants of these bacteria showed growth inhibitory activity against H. pylori and caused the formation of the coccoid form of H. pylori in vitro.[123]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Zhang, L.; Chen, X.; Ren, B.; Zhou, X.; Cheng, L. Helicobacter pylori in the Oral Cavity: Current Evidence and Potential Survival Strategies. Int. J. Mol. Sci. 2022, 23, 13646. https://doi.org/10.3390/ijms232113646

AMA Style

Zhang L, Chen X, Ren B, Zhou X, Cheng L. Helicobacter pylori in the Oral Cavity: Current Evidence and Potential Survival Strategies. International Journal of Molecular Sciences. 2022; 23(21):13646. https://doi.org/10.3390/ijms232113646

Chicago/Turabian Style

Zhang, Lin, Xi Chen, Biao Ren, Xuedong Zhou, and Lei Cheng. 2022. "Helicobacter pylori in the Oral Cavity: Current Evidence and Potential Survival Strategies" International Journal of Molecular Sciences 23, no. 21: 13646. https://doi.org/10.3390/ijms232113646

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