Skip to main content
Erschienen in: Infection 2/2024

Open Access 02.11.2023 | Review

Helicobacter pylori and oral–gut microbiome: clinical implications

verfasst von: Maged T. Elghannam, Moataz H. Hassanien, Yosry A. Ameen, Emad A. Turky, Gamal M. ELattar, Ahmed A. ELRay, Mohammed D. ELTalkawy

Erschienen in: Infection | Ausgabe 2/2024

Abstract

More than half of the world’s population are colonized with H. pylori; however, the prevalence varies geographically with the highest incidence in Africa. H. pylori is probably a commensal organism that has been associated with the development of gastritis, ulcers, and gastric cancer. H. pylori alone is most probably not enough for the development of gastric carcinoma, but evidence for its association with the disease is high and has, therefore, been classified by the International Agency for Research on Cancer as a Class 1 carcinogen. Bacteroidetes and Fusobacteria positively coexisted during H. pylori infection along the oral–gut axis. The eradication therapy required to treat H. pylori infection can also have detrimental consequences for the gut microbiota, leading to a decreased alpha diversity. Therefore, therapy regimens integrated with probiotics may abolish the negative effects of antibiotic therapy on the gut microbiota. These eradication therapies combined with probiotics have also higher rates of eradication, when compared to standard treatments, and are associated with reduced side effects, improving the patient’s compliance. The eradication therapy not only affects gut microbiome but also affects the oral microbiome with robust predominance of harmful bacteria. However, there have been reports of a protective role of H. pylori in Barrett’s esophagus, esophageal adenocarcinoma, eosinophilic esophagitis, IBD, asthma, and even multiple sclerosis. Therefore, eradication therapy should be carefully considered, and test to treat policy should be tailored to specific communities especially in highly endemic areas. Supplementation of probiotics, prebiotics, herbals, and microbial metabolites to reduce the negative effects of eradication therapy should be considered. After failure of many eradication attempts, the benefits of H. pylori eradication should be carefully balanced against the risk of adverse effects especially in the elderly, persons with frailty, and intolerance to antibiotics.

Background

H. pylori is a commensal organism associated with the development of gastritis, ulcers, and gastric cancer. The organism as well as eradication remedies can modulate gut microbiota in humans. Other non-H. pylori microbial species may colonize the same milieu, but H. pylori are regarded as a human pathogen [1]. Interactions between H. pylori and other members of the microbiome, the host, and the environment influence the clinical consequence and may lead to either disease or possible protective effects. Considering the beneficial effects on the host by regulating gastrointestinal microbiota, eradication of H. pylori can produce various adverse effects and alter the gastrointestinal microbiota. Gastrointestinal microbiota are defined as the entire community of microorganisms dwelling in the gastrointestinal tract, and it is dominated substantially by bacteria [2]. Gut microbiota composition varies between ethnic groups due to the different dietary, hygienic, and genetic factors in addition to the use of antibiotics. Its homeostasis plays a critical role in maintaining host health. Dysbiosis of the gut microbiome may produce multiple diseases and bacterial infections in addition to compromising human alimentation [3]. This review aims to discuss the relationships between H. pylori alone and in combination with oral and gut microbiota in the development of GI disease.

General characteristics of H. pylori

H. pylori colonization affects more than half of the population worldwide [4] with the highest incidence in Africa (79.1%) [5]. Despite this high prevalence, the majority of the infected population are asymptomatic. Acquisition of H. pylori occurs in early childhood (30%–50%), while during early adolescence, it reaches over 90% in developing countries [6]. The consequence of infection varies either from no clinical symptoms or continuing throughout his life with superficial chronic gastritis [7, 8] or developing peptic ulcers, 25% even experience ulcer complications, and 1% will advance to gastric cancers (GC) [9].
The main transmission route of H. pylori is not known. However, the intrafamilial transmission of the pathogen is the most significant route. This may be facilitated by close personal contacts, the unified socioeconomic status of the family members, and the genetic predisposition to H. pylori persistence [10, 11].
Children < 5 years of age have high infection rates, after which infection declines at school age when less time is spent at home. In Egypt, 33% of children < 6 years are infected [12]. The disease development is influenced by several factors such as host genetics, environmental factors related to diets, lifestyle habits, and pathogens [13].
Once entering the stomach, H. pylori produces urease to convert urea to ammonia which neutralizes hydrochloric acid, then after, uses mobile flagella to spread over the surface of the gastric epithelium forming microbial biofilms. Gastric persistence is determined mainly by bacterial adhesion [14]. The adhesion of H. pylori is facilitated by the gastric epithelium α1,2-Fucosylated glycans [15]. Both virulence factors; cytotoxic-associated gene, (CagA) and vacuolating cytotoxin A (vacA), have direct damaging impact on the gastric mucosal epithelium [16, 17]. The initial host Th1 cell immune response intended to eradicate the microorganisms is opposed by H. pylori vacA immunosuppressive effect [18]. A Th2 cellular pathway facilitates H. pylori colonization in infancy and leads to the development of immune tolerance resulting in a symbiotic relationship between the microbe and the host [19]. H. pylori influence host immune responses and the microbiota of both the stomach and distal organs [20].
The persistence of H. pylori in the stomach is associated with the development of gastroduodenal diseases, such as chronic gastritis, peptic ulcer disease (PUD), gastric adenocarcinoma, and gastric MALT lymphoma, and colorectal carcinoma [2123]. A significant relationship between H. pylori detection and pancreatic cancer has also been reported [2325]. H. Pylori had been implicated in the pathogenesis of extra-gastric diseases such as ischemic brain injury [26], Alzheimer’s disease [27, 28], Parkinson’s disease [29], atherosclerotic vascular lesions, a higher risk of coronary heart disease [3033], hypertension [34], endothelial dysfunction [34], vitamin B12 and folic acid malabsorption [35, 36]. Psoriasis [37, 38], lichen ruber planus, scabies, rosacea, Sweet’s syndrome, Behcet’s disease, and Schönlein-Henoch purpura [39] The mechanism of this extra gastric affection is not confirmed; however, H. pylori generates local inflammation in the stomach and can spread systemically by the release of cytokines establishing low-grade and chronic inflammation throughout the body [40].
In contrast to the harmful effects, H. pylori found to have a protective effect against many pathological conditions such as IBD [41, 42], asthma [4347], esophageal adenocarcinoma [48], eosinophilic esophagitis [49], and GERD and Barrett’s esophagus [50, 51].

Helicobacter pylori and oral microbiota

H. pylori detected in both the mouth and gut. The oral–gut axis microbiota has a dominant effect in H. pylori’s colonization, infection, and pathogenicity [52]. The number of H. pylori in the mouth is lower than in the stomach. It constitutes 42%–97% of the total gastric bacterial community [53]. The oral and gastric milieus are affected by saliva and digested food. The oral microbiome is the dominant source of gastric microbes, so, it is accused for the infection and transmission of H. pylori [54, 55].
The interplay between H. pylori and oral microbiome may take one or further of three main forms: co-aggregation, symbiotic biofilm formation, and endosymbiosis [56]. Fusobacterium nucleatum and Porphyromonas gingivalis are crucial bacteria in periodontal infection. The aggregation with H. pylori promotes oral to gastric colonization by oral bacteria [57]. Biofilms are surface bacterial communities embedded within an extracellular matrix. They contribute to an infection becoming chronic or recurrent, promote inflammation, and can make bacterial colonies resistant to antibiotics and the immune system [58].
The major cariogenic bacterium, Streptococcus mutans, forms a symbiotic biofilm with H. pylori prolonged its survival in the unsuitable atmosphere of the mouth [59]. H. pylori can anchor on the surface and/or enter C. albicans to form a mixed biofilm in the oral cavity and vagina [60, 61].
The interaction between and H. pylori members of the oral microbial community yields different results according to oral or gastrointestinal complaints in H. pylori-positive people. P. gingivalis has been established as a pathogenic agent of periodontitis and positively associated with H. pylori, indicating that H. pylori infection may promote periodontal disease [62]. The inter-transmission between oral and gut microorganisms can affect the ecosystem in both territories and hence regulate the pathogenesis of different diseases [63].

Gastric bacterial microbiome profile

The gastric core microbiome is mainly formed from five major phyla, including Firmicutes, Bacteroidetes, Actinobacteria, Fusobacteria, and Proteobacteria [64, 65]. Numerous oral bacteria such as Neisseria, Veillonella, Fusobacterium, Streptococcus, and Hemophilus, are enriched in the lower digestive tract and can be significantly found in gastric biopsy too [66]. A healthy gut microbiome is characterized by high gut microbial diversity [67]. The co-occurrence interactions were stronger in intestinal metaplasia (IM) than superficial gastritis (SG) [68] and among genera in IM which then decreased in intraepithelial neoplasia (IN) of gastric biopsies with gastritis progression suggesting that the bacteria tend to co-occur to form a specific micro ecology before the occurrence of neoplasia [69]. Several factors can affect the gastric microbiome such as diet, aging, geographic area of residence, and medications like PPI and antibiotics [70, 71]. A reduced number of Bacteroidetes and elevated numbers of Firmicutes and Proteobacteria were observed in patients with gastritis as compared with healthy individuals [72] (Fig. 1).

Gut microbiota and Helicobacter pylori infection

H. pylori infection disturbs commensal bacterium equilibrium in the gastric mucosa in addition to the disturbance of microbial changes in the human gut [7376]. H. pylori mainly influences the microbial composition and diversity in gastric mucosa rather than both gastric juice and stool [69] H. pylori infection results concerning bacterial diversity have been controversial as has been found with other specific groups of gut bacteria [77, 78]. Lactobacillus species abundance was higher in H. pylori-infected patients than in non-infected persons [79] , protecting the human gut from bacterial colonization through gut barrier preservation [80]. Iino et al. in 2018 [79] found that H. pylori-positive patients displayed reduced amounts of L. acidophilus and an increased proportion of L. salivarius in comparison with non-infected subjects due to the suppression of gastric acid secretion by H. pylori infection. He reported a higher abundance of Lactobacillus in H. pylori-positive patients with severe atrophic gastritis compared to infected patients with mild atrophic gastritis or without gastritis denoting affection of gastric microbiota according to symptom severity. On the other hand, gut bacteria might also influence the bacterial colonization of other gastrointestinal regions, including H. pylori in the stomach. Nitrospirae phylum can be seen only in H. pylori-negative personnel with minimal values in patients with duodenal ulcer and H. pylori infection as nitrite has a bactericidal effect against H. pylori [80, 81]. H. pylori infection alters the gut microbiota in asymptomatic patients by increasing Proteobacteria, Clostridium, Firmicutes, and Prevotella in a pediatric population [78] and members belonging to Succinivibrio, Coriobacteriaceae, Enterococcaceae, and Rikenellaceae in adults [75] compared to non-infected subjects. Gao et al. in 2018 [72] reported a disturbance of fecal microbiota, mainly the phyla Bacteroidetes, Firmicutes, and Proteobacteria in H. pylori-induced gastric diseases. The relationship between H. pylori and gastric microbiota could be mediated through multiple mechanisms, such as virulence factors, the modification of gastric acidity, host immune responses, and competition [82] (Fig. 2).
H. pylori-resistant strains showed a higher trend of diversity and evenness than the sensitive samples. The abundance of resistant strains decreased with increasing cohabitation of pathogenic bacteria. There is an increase in the α-diversity index among the MDR. The resistance status of H. pylori was correlated with the enriched diversity of the gastric microbiome composition, where the abundance of non-pylori pathogens increased, especially in triple-resistant strains [83].

Gut microbiota and Helicobacter pylori eradication therapy

Antibiotic administration decreases bacterial diversity [84, 85]. Bacterial diversity was restored in the short and the long term after treatment conclusion [8689]. Still, not all studies reported enhancement in bacterial diversity after treatment conclusion [90, 91]. Generally, gut microbiota composition is restored in most cases at 2 months post-treatment. Proteobacteria phylum is proposed to be partially responsible for the development of adverse effects during eradication therapy [92]. Probiotic supplementation and the antibiotic impose a beneficial gut microbiota profile after eradication therapy [93]. Niu et al. in 2021 [94] reported a success rate of H. pylori eradication 95.5% using the quadruple remedy. The majority of phyla in the two groups were the same and included Proteobacteria, Bacteroides, Firmicutes, Actinomycetes, and Fusobacteria. The microbial diversity in the failure group had a lowering fashion and the species abundance became extensively reduced compared with the success group. The presence of Rhodococcus, Lactobacillus, and Sphingomonas was associated with high rate of H. pylori eradication in the successful group. Veronococcus and Cilium were enriched in the mucosa of chronic atrophic gastritis cases compared to chronic superficial gastritis cases. In both study groups, H. pylori were negatively identified with other bacterial groups. They concluded that gastric microbiota is the corner stone in the effect of quadruple H. pylori eradication therapy. Tawfik et al. in 2023 [95] found oral microbiomes more diverse than the gut microbiomes. The eradication of H. pylori was associated with a significant reduction in the bacterial diversity along the orointestinal axis. H. pylori positive patients showed positive correlation between Proteobacteria and Fusobacteria. After eradication therapy, Fusobacterium, Veillonella, Catenibacterium, Neisseria and Prevotella enriched significantly. They stress the importance of eradication therapy on certain genera especially, in the oral microbiota.

Helicobacter pylori-associated diseases

Gastritis and ulcer disease

Only 10% of the population develop clinical manifestations latterly in their lives when getting elderly [96]. Seventy percent of people who are established to have the bacterium are healthy bacterial carriers, and 5%–10% of those infected develop symptoms of gastritis or PUD [97101]. Absence of H. pylori-gastritis had been reported [102, 103] , and indeed in severe cases and premalignant conditions, a low abundance of H. pylori had been reported [104]. H. pylori-negative gastritis was found to be 21% in the United States [105] and 27% of all cases of gastritis in Indonesia [103]. Araújo et al. in 2014 [106] reported that the discovery rate of H. pylori infection in cases diagnosed with PUD is the same as in the general population and 20–50% of PUD patients had idiopathic etiology. The high prevalence rate and low incidence of pathological diseases indicate that H. pylori are more likely to be an opportunistic or latent pathogen rather than a truly pathogenic bacterium. The development of PUD is multifactorial and depends on endogenous and exogenous factors, which means that the presence of H. pylori infection may be only one of many factors involved in the genesis of ulcerative disorders. The genotype of H. pylori is a determinant factor in producing ulcer disease. Cases with a verified diagnosis of PUD had vacA-positive and CagA-positive genotypes [107, 108]. Our group [109] reported a low prevalence of CagA (26.5%). Western type CagA is the fundamental kind (62.5%) while the East Asian type was not detected and others (37.5%) remain uncharacterized. Western-genotype CagA was found in 80% of patients with peptic ulcer disease and 40% of patients with gastritis. The primary genotype mixture in the studied Egyptian sufferers were; vacAs2m2/iceA1, vacAs1m1/cagA, mostly related to gastritis, and vacAs1/cagA/icA, mainly in PUD. The much less virulent (s2, s2m2) H. pylori genotypes were found in cases over the age of 43 years [110]. Lately, there has been a progressive increase in the idiopathic forms of PUD with a drop in the global frequencies of H. pylori infection. A further study demonstrated a significant correlation between the isolation of Streptococci and peptic ulcer disease [111]. Iijima and his associates [112] reported that 45.9% of cases of peptic ulcers of the stomach and 29.6% of those of the duodenum were idiopathic. H. pylori-positive ulcer had better convalescence rates, better course, more positive prognosis, less hospital stay, less 30 days readmission, and fewer recurrence rates [113, 114].

Gastric carcinoma

Gastric carcinoma (GC) develops in H. pylori-infected people 1.4–4.2 times more often than within the general population [115117]. Even so, only 1–2% of cases develop GC in 50% or more of H. pylori-infected patients [118]. In spite of the superiority of H. pylori in Africa and India than in the West, the incidence of GC is less frequent than in the West [119], which is known as an epidemiologic paradox [120]. This decreasing trend of bacterial richness going from the normal tissue to peritumoral and tumoral tissues indicated that as the microenvironment of a tumor is altered, it becomes unsuitable for colonization with specific bacteria. The low microbial diversity of the upper digestive tract was associated with a low serum pepsinogen I/pepsinogen II ratio, which has also been associated with gastric carcinogenesis [121]. H. pylori virulence factors have not been reported to be essential for cancer development [1]. In an Egyptian study of the prevalence of H. pylori CagA among patients with gastric carcinoma, a total of 34 (56.67%) patients have been CagA + ve and 26 (43.33%) patients were CagA − ve, with no statistically significant difference regarding sex or age [122]. It is well known that the persistence of H. pylori infection is linked to the development of only non-cardiac carcinoma, while it had a protective effect against cancer development in the cardiac area and lower esophagus [48]. The decrease in H. pylori infections in Japan is believed to have contributed to a decline in gastric cancer cases [123]. However, the cause of an increase in gastric cancer in the young population in the USA (notably, young Hispanic men), where overall the incidence of H. pylori infection is also waning, is unexplained [124]. Thus, unknown factors likely unrelated to H. pylori infection may be contributing to a rise in gastric cancer in specific populations.
Recently, it is accepted that cancer pathogenesis is precipitated by confounding factors such as high-salt diets and other carcinogenic substances that promote the carcinogenic pathway in addition to bacterial agents [125]. The multistep processes involved in the development of GC are initiated by the transition of the mucosa into chronic non-atrophic gastritis, which is primarily triggered by infection with H. pylori. This gastritis then progresses into atrophic gastritis and intestinal metaplasia, and then to dysplasia, and following Correa’s cascade, to adenocarcinoma [119].
During the transition from gastritis to GC, a significant difference in the gastric microbial community was observed. There is an increase in the abundance of non-H. pylori proteobacteria [126] (Fig. 3).
An analysis of gastric microbial communities from different stages of gastric cancer development revealed the significance of Peptostreptococcus stomatis, S. anginosus, Parvimonas micra, Slackia exigua, and Dialister pneumosintes in the progression of gastric cancer, as they were found in the precancerous stage [68]. Gastric cancer prevalence varies among different regions of the stomach, with cancers arising in the corpus potentially caused by mechanisms distinct from the other regions [127]. Alternatively, physiological factors that vary along the length of the stomach and pylorus such as differences in oxygen concentration, pH, mucus, and nutrient availability could play a part in determining regional cancer susceptibility [128].
Ralser and his colleagues in 2023 identified a unique H. pylori-driven immune alteration signature characterized by a reduction in regulatory T cells in addition to H. pylori induction of pro-carcinogenic STAT3 signaling and a loss of goblet cells in colonic epithelium, changes that have been shown to contribute; in combination with pro-inflammatory and mucus degrading microbial signatures, to tumor development in the intestinal and colonic epithelium [129].
Metabolites and their interactions with microbiota may be involved in Helicobacter pylori-associated gastric lesion development. Negative correlations between Helicobacter and glycerophospholipids, glycosylceramide, and triacylglycerol, which were altered by eradication. The characteristic negative correlations between glycosylceramides and Fusobacterium, Streptococcus, and Gemella in H. pylori-positive baseline biopsy specimens were further noticed in active gastritis and intestinal metaplasia. This helps discriminate high-risk subjects for progression from mild lesions to advanced precancerous lesions in short-term and long-term follow-up [130].
Antibiotic treatment in H. pylori-infected patients can reduce GC progression even if H. pylori is not eradicated, suggesting that suppression of other bacteria may serve a protective function [131]. Niikura and his colleagues in 2023 [132] identified potential pathogens; abnormally colonized gastric bacteria, particularly Fusobacterium and Neisseria spp., play an additional fundamental role in the later stages of gastric carcinogenesis. Testing for Fusobacterium and Neisseria spp. in gastric mucosal samples as a surrogate for gastric dysbiosis could be a next-generation approach for screening high-risk patients for GC. In addition, eradication of these oncogenic bacteria and/or inhibition of carcinogenic bacteria-derived molecules may be a future strategy for preventing GC development, particularly in patients with severe atrophic gastritis and intestinal metaplasia.

Clinical implications

H. pylori infection is frequent in developing countries and represents an annoying health problem. Eradication remedies had been recommended by all societies for fear of malignancy. Eradication remedies are complicated by the requirement for several agents such as the use of 2 antibiotics and a PPI. These strategies can be complicated by antibiotic resistance, high cost on the national level, PPI-related complications, and not the least microbiota dysbiosis. Despite the high frequency of H. pylori infection, there is a low prevalence of gastric malignancy. H. pylori alone is most probably not enough for the development of GC. Considering the protective role of H. pylori against numerous conditions such as IBD, asthma, multiple sclerosis, Barrett’s esophagus, esophageal adenocarcinoma, and eosinophilic esophagitis, H. pylori is now considered one of the bacteria in the healthy microbiome for the majority of the human population. Therefore, not every case should be treated for eradication. A personalized approach according to the H. pylori indigenous region, the presence of gastrointestinal malignancies among relatives, or the impossibility of banning non-modifiable threat factors is needed. It should include relatives with high familial risk or living in high-risk areas/populations where eradication effectively reduces the threat of gastric carcinoma as in South East Asia and cases with cancer who are on therapy with immune checkpoint inhibitors or vaccine-grounded immunotherapy [133].
The supplementation of probiotics, prebiotics, and microbial metabolites to reduce the negative effects of eradication should be considered.
Probiotics reduce H. pylori-induced gastric pathology in mice, with reduced inflammatory infiltration and precancerous lesion incidence [134], enhance H. pylori eradication rates, and reduce side effects in humans [135].
Autoprobiotics refer to indigenous bifidobacteria, lactobacilli, or enterococci isolated from a specific individual, intended to restore microbiota and improve health. The advantages of autoprobiotics include its safety, high survival rate, its unique individual composition and, extended duration in the gut [136]. Both the quadruple therapy group and the H. pylori-negative subjects after probiotic-supplemented eradication treatment had nearly the same microbial diversity [137]. The most effective types belong to the Firmicutes (Enterococcus and Lactobacillus) and Actinobacteria (Bifidobacterium genus) phyla and Saccharomyces boulardii [138, 139]. However, Yang and his colleagues in 2021 [140] reported failure to improve the eradication rate of H. pylori after supplementation with lactobacillus, but it helped build up a beneficial microbial profile and reduced the frequencies of abdominal distention and diarrhea.
The potential mechanisms of probiotic action against H. pylori include correction of the gut microbiota, immunological effects such as enhancement of humoral and cellular immunity, and reduction of oxidative stress, direct antagonistic effects against H. pylori such as colonization resistance and bacteriocin synthesis, and stimulation of local immunological protection such as strengthening of the mucous protective barrier and reduction of gastric mucosa inflammation [141]. As a double-edged sword, the use of probiotic-induced adverse effects include higher risk of systemic neonates infections throughout their life span [142], long-term gut dysbiosis [143], and risk to develop Parkinson’s disease mostly due to Desulfovibrio bacteria [144, 145]. The oral administration of multi-strain probiotics and paraprobiotics were more than single-strain probiotics, reducing the incidence of developing metabolic disorders [146].
Washing microflora transfer (WMT) is a modified FMT method that uses washed preparations. Ye et al. in 2020 [147] reported that WMT has an overall H. pylori eradication of 40.6%. No H. pylori transmission was recorded from healthy, asymptomatic donors to recipients by oral capsule-based FMT [148].
H. pylori living in both oral cavity and gut looks to be a commensal, occasionally pathogenic. It is not surprising for a case to have more than an eradication course. This is harmful to both oral and gut microbiota and may lead to different diseases. After multiple failed eradication attempts, the implicit benefits of H. pylori eradication should be weighed against the liability of adverse effects with repeated high-dose acid suppression and antibiotic exposure, particularly among individuals who are not at an identifiably advanced threat of complications from persistent H. pylori infection such as, GC or peptic ulcer disease. Similarly, a careful decision-making approach should be seriously considered, especially in the senior, those with frailty, and those with intolerance to antibiotics [149] (Best Practice Advice #9).

Declarations

Conflict of interest

All the authors contribute equally like the corresponding author in creating the idea of the article, gathering the information, drafting and writing, reviewing and editing the manuscript in the final shape. They are in agreement with the content of the manuscript. There is no conflict of interest and no financial support. This research did not receive any specific grant from funding agencies in the public, commercial, or not-for-profit sectors. No ethical committee approval is needed for this article. All datasets used can be accessed through the internet and no special requests are needed.
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​.

Unsere Produktempfehlungen

Neuer Inhalt

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

Weitere Produktempfehlungen anzeigen
Literatur
1.
Zurück zum Zitat Sitkin S, Lazebnik L, Avalueva E, Kononova S, Vakhitov T. Gastrointestinal microbiome and Helicobacter pylori: eradicate, leave it as it is, or take a personalized benefit–risk approach? World J Gastroenterol. 2022;28(7):766–74.CrossRefPubMedPubMedCentral Sitkin S, Lazebnik L, Avalueva E, Kononova S, Vakhitov T. Gastrointestinal microbiome and Helicobacter pylori: eradicate, leave it as it is, or take a personalized benefit–risk approach? World J Gastroenterol. 2022;28(7):766–74.CrossRefPubMedPubMedCentral
5.
Zurück zum Zitat Hooi JKY, Lai WY, Ng WK, Suen MMY, Underwood FE, Tanyingoh D, et al. Global prevalence of Helicobacter pylori infection: systematic review and meta-analysis. Gastroenterology. 2017;153:4209.CrossRef Hooi JKY, Lai WY, Ng WK, Suen MMY, Underwood FE, Tanyingoh D, et al. Global prevalence of Helicobacter pylori infection: systematic review and meta-analysis. Gastroenterology. 2017;153:4209.CrossRef
6.
Zurück zum Zitat Go MF. Review article: natural history and epidemiology of Helicobacter pylori infection. Aliment Pharmacol Ther. 2002;16:3–15.CrossRefPubMed Go MF. Review article: natural history and epidemiology of Helicobacter pylori infection. Aliment Pharmacol Ther. 2002;16:3–15.CrossRefPubMed
7.
Zurück zum Zitat Hunt RH. The role of H. pylori in pathogenesis: the spectrum of clinical outcomes. Scand J Gastroenterol. 1996;31:3–9.CrossRef Hunt RH. The role of H. pylori in pathogenesis: the spectrum of clinical outcomes. Scand J Gastroenterol. 1996;31:3–9.CrossRef
8.
Zurück zum Zitat Rothenbacher D, Brenner H. Burden of Helicobacter pylori and H. pylori-related diseases in developed countries: recent developments and future implications. Microbes Infect. 2003;5:693–703.CrossRefPubMed Rothenbacher D, Brenner H. Burden of Helicobacter pylori and H. pylori-related diseases in developed countries: recent developments and future implications. Microbes Infect. 2003;5:693–703.CrossRefPubMed
9.
Zurück zum Zitat Malaty HM. Epidemiology of Helicobacter pylori infection. Best Prac Res Clin Gastroenterol. 2007;21:205–14.CrossRef Malaty HM. Epidemiology of Helicobacter pylori infection. Best Prac Res Clin Gastroenterol. 2007;21:205–14.CrossRef
12.
Zurück zum Zitat Frenck RW Jr, Fathy HM, Sherif M, Mohran Z, El Mohammedy H, Francis W, et al. Sensitivity and specificity of various tests for the diagnosis of Helicobacter pylori in Egyptian children. Pediatrics. 2006;118:1195–202.CrossRef Frenck RW Jr, Fathy HM, Sherif M, Mohran Z, El Mohammedy H, Francis W, et al. Sensitivity and specificity of various tests for the diagnosis of Helicobacter pylori in Egyptian children. Pediatrics. 2006;118:1195–202.CrossRef
47.
Zurück zum Zitat Ierardi E, Losurdo G, Giorgio F, Di Leo A. Might Helicobacter pylori play a role in allergic or cross-reaction-related disorders? Expert Rev Gastroenterol Hepatol. 2020;14:643–6 (PMID: 32510247).CrossRefPubMed Ierardi E, Losurdo G, Giorgio F, Di Leo A. Might Helicobacter pylori play a role in allergic or cross-reaction-related disorders? Expert Rev Gastroenterol Hepatol. 2020;14:643–6 (PMID: 32510247).CrossRefPubMed
61.
Zurück zum Zitat 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 Iranian Med. 2013;16:705–10. 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 Iranian Med. 2013;16:705–10.
64.
Zurück zum Zitat Monstein HJ, Tiveljung A, Kraft CH, Borch K, Jonasson J. Profiling of bacterial flora in gastric biopsies from patients with Helicobacter pylori-associated gastritis and histologically normal control individuals by temperature gradient gel electrophoresis and 16S rDNA sequence analysis. J Med Microbiol. 2000;49:817–22.CrossRefPubMed Monstein HJ, Tiveljung A, Kraft CH, Borch K, Jonasson J. Profiling of bacterial flora in gastric biopsies from patients with Helicobacter pylori-associated gastritis and histologically normal control individuals by temperature gradient gel electrophoresis and 16S rDNA sequence analysis. J Med Microbiol. 2000;49:817–22.CrossRefPubMed
65.
Zurück zum Zitat Bik EM, Eckburg PB, Gill SR, Nelson KE, Purdom EA, Francois F, et al. Molecular analysis of the bacterial microbiota in the human stomach. Proc Natl Acad Sci USA. 2006;103:732–7.CrossRefPubMedPubMedCentral Bik EM, Eckburg PB, Gill SR, Nelson KE, Purdom EA, Francois F, et al. Molecular analysis of the bacterial microbiota in the human stomach. Proc Natl Acad Sci USA. 2006;103:732–7.CrossRefPubMedPubMedCentral
70.
Zurück zum Zitat Nardone G, Compare D, Rocco A. A microbiota-centric view of diseases of the upper gastrointestinal tract. Lancet Gastroenterol Hepatol. 2017;2:298–312.CrossRefPubMed Nardone G, Compare D, Rocco A. A microbiota-centric view of diseases of the upper gastrointestinal tract. Lancet Gastroenterol Hepatol. 2017;2:298–312.CrossRefPubMed
71.
Zurück zum Zitat Hojo M, Asahara T, Nagahara A, Takeda T, Matsumoto K, Ueyama H, et al. Gut microbiota composition before and after use of proton pump inhibitors. Dig Dis Sci. 2018;63:2940–9.CrossRefPubMedPubMedCentral Hojo M, Asahara T, Nagahara A, Takeda T, Matsumoto K, Ueyama H, et al. Gut microbiota composition before and after use of proton pump inhibitors. Dig Dis Sci. 2018;63:2940–9.CrossRefPubMedPubMedCentral
72.
Zurück zum Zitat Gao J, Zhang Y, Gerhard M, Mejias-Luque R, Zhang L, Vieth M, et al. Association between gut microbiota and Helicobacter pylori-related gastric lesions in a high-risk population of gastric cancer. Front Cell Infect Microbiol. 2018;8:202.CrossRefPubMedPubMedCentral Gao J, Zhang Y, Gerhard M, Mejias-Luque R, Zhang L, Vieth M, et al. Association between gut microbiota and Helicobacter pylori-related gastric lesions in a high-risk population of gastric cancer. Front Cell Infect Microbiol. 2018;8:202.CrossRefPubMedPubMedCentral
87.
Zurück zum Zitat Gotoda T, Takano C, Kusano C, Suzuki S, Ikehara H, Hayakawa S, et al. Gut microbiome can be restored without adverse events after Helicobacter pylori eradication therapy in teenagers. Helicobacter. 2018;23: e12541.CrossRefPubMed Gotoda T, Takano C, Kusano C, Suzuki S, Ikehara H, Hayakawa S, et al. Gut microbiome can be restored without adverse events after Helicobacter pylori eradication therapy in teenagers. Helicobacter. 2018;23: e12541.CrossRefPubMed
91.
92.
Zurück zum Zitat Hsu P-II, Pan C-YY, Kao JY, Tsay F-WW, Peng N-JJ, Kao S-SS, et al. Helicobacter pylori eradication with bismuth quadruple therapy leads to dysbiosis of gut microbiota with an increased relative abundance of proteobacteria and decreased relative abundances of bacteroidetes and actinobacteria. Helicobacter. 2018;10:e12498. https://doi.org/10.1111/hel.12498.CrossRef Hsu P-II, Pan C-YY, Kao JY, Tsay F-WW, Peng N-JJ, Kao S-SS, et al. Helicobacter pylori eradication with bismuth quadruple therapy leads to dysbiosis of gut microbiota with an increased relative abundance of proteobacteria and decreased relative abundances of bacteroidetes and actinobacteria. Helicobacter. 2018;10:e12498. https://​doi.​org/​10.​1111/​hel.​12498.CrossRef
94.
Zurück zum Zitat Niu ZY, Li SZ, Shi YY, Xue Y. Effect of gastric microbiota on quadruple Helicobacter pylori eradication therapy containing bismuth. World J Gastroenterol. 2021;27(25):3913–24.CrossRefPubMedPubMedCentral Niu ZY, Li SZ, Shi YY, Xue Y. Effect of gastric microbiota on quadruple Helicobacter pylori eradication therapy containing bismuth. World J Gastroenterol. 2021;27(25):3913–24.CrossRefPubMedPubMedCentral
101.
Zurück zum Zitat Tsimmerman YS. Critical analysis of the Helicobacter pylori-infection leading role in the development of gastroduodenal diseases. Clin Pharmacol Ther. 2019;28:19–27.CrossRef Tsimmerman YS. Critical analysis of the Helicobacter pylori-infection leading role in the development of gastroduodenal diseases. Clin Pharmacol Ther. 2019;28:19–27.CrossRef
102.
Zurück zum Zitat Shiota S, Thrift AP, Green L, Shah R, Verstovsek G, Rugge M, Graham DY, El-Serag HB. Clinical manifestations of Helicobacter pylori-negative gastritis. Clin Gastroenterol Hepatol. 2017;15:1037-46.e3.CrossRefPubMed Shiota S, Thrift AP, Green L, Shah R, Verstovsek G, Rugge M, Graham DY, El-Serag HB. Clinical manifestations of Helicobacter pylori-negative gastritis. Clin Gastroenterol Hepatol. 2017;15:1037-46.e3.CrossRefPubMed
103.
Zurück zum Zitat Miftahussurur M, Waskito LA, Syam AF, Nusi IA, Wibawa ID, Rezkitha YA, Siregar G, Yulizal OK, Akil F, Uwan WB, Simanjuntak D. Analysis of risks of gastric cancer by gastric mucosa among Indonesian ethnic groups. PLoS ONE. 2019;14: e0216670.CrossRefPubMedPubMedCentral Miftahussurur M, Waskito LA, Syam AF, Nusi IA, Wibawa ID, Rezkitha YA, Siregar G, Yulizal OK, Akil F, Uwan WB, Simanjuntak D. Analysis of risks of gastric cancer by gastric mucosa among Indonesian ethnic groups. PLoS ONE. 2019;14: e0216670.CrossRefPubMedPubMedCentral
104.
Zurück zum Zitat Pereira-Marques J, Ferreira RM, Pinto-Ribeiro I, Figueiredo C. Helicobacter pylori infection, the gastric microbiome and gastric cancer. Adv Exp Med Biol. 2019;1149:195–210.CrossRefPubMed Pereira-Marques J, Ferreira RM, Pinto-Ribeiro I, Figueiredo C. Helicobacter pylori infection, the gastric microbiome and gastric cancer. Adv Exp Med Biol. 2019;1149:195–210.CrossRefPubMed
105.
Zurück zum Zitat Nordenstedt H, Graham DY, Kramer JR, Rugge M, Verstovsek G, Fitzgerald S, Alsarraj A, Shaib Y, Velez ME, Abraham N, Anand B, Cole R, El-Serag HB. Helicobacter pylori-negative gastritis: prevalence and risk factors. Am J Gastroenterol. 2013;108:65–71.CrossRefPubMed Nordenstedt H, Graham DY, Kramer JR, Rugge M, Verstovsek G, Fitzgerald S, Alsarraj A, Shaib Y, Velez ME, Abraham N, Anand B, Cole R, El-Serag HB. Helicobacter pylori-negative gastritis: prevalence and risk factors. Am J Gastroenterol. 2013;108:65–71.CrossRefPubMed
109.
Zurück zum Zitat Diab M, Shemis M, Gamal D, El-Shenawy A, El-Ghannam M, El-Sherbini E, et al. Helicobacter pylori western cagA genotype in Egyptian patients with upper gastrointestinal disease. Egypt J Med Human Genetics. 2018;19:297–300.CrossRef Diab M, Shemis M, Gamal D, El-Shenawy A, El-Ghannam M, El-Sherbini E, et al. Helicobacter pylori western cagA genotype in Egyptian patients with upper gastrointestinal disease. Egypt J Med Human Genetics. 2018;19:297–300.CrossRef
110.
Zurück zum Zitat El-Shenawy A, Diab M, Shemis M, ElGhannam M, Salem D, Abdelnasser M, et al. Detection of Helicobacter pylori vacA, cagA and iceA1 virulence genes associated with gastric diseases in Egyptian patients. Egypt J Medi Human Genetics. 2017;18(4):365–71.CrossRef El-Shenawy A, Diab M, Shemis M, ElGhannam M, Salem D, Abdelnasser M, et al. Detection of Helicobacter pylori vacA, cagA and iceA1 virulence genes associated with gastric diseases in Egyptian patients. Egypt J Medi Human Genetics. 2017;18(4):365–71.CrossRef
113.
Zurück zum Zitat Kanno T, Iijima K, Abe Y, Yagi M, Asonuma S, Ohyauchi M, Ito H, Koike T, Shimosegawa T. Helicobacter pylori-negative and non-steroidal anti-inflammatory drugs-negative idiopathic peptic ulcers show refractoriness and high recurrence incidence: multicenter follow-up study of peptic ulcers in Japan. Dig Endosc. 2016;28:556–63. https://doi.org/10.1111/den.12635.CrossRefPubMed Kanno T, Iijima K, Abe Y, Yagi M, Asonuma S, Ohyauchi M, Ito H, Koike T, Shimosegawa T. Helicobacter pylori-negative and non-steroidal anti-inflammatory drugs-negative idiopathic peptic ulcers show refractoriness and high recurrence incidence: multicenter follow-up study of peptic ulcers in Japan. Dig Endosc. 2016;28:556–63. https://​doi.​org/​10.​1111/​den.​12635.CrossRefPubMed
114.
116.
Zurück zum Zitat Tsimmerman YS. Gastric cancers: a modern approach to the problem. Vestnik chirurgicheskoj gastroenterologii. 2011;2:77–88. Tsimmerman YS. Gastric cancers: a modern approach to the problem. Vestnik chirurgicheskoj gastroenterologii. 2011;2:77–88.
119.
Zurück zum Zitat Correa P. Human gastric carcinogenesis: a multistep and multifactorial process–first American cancer society award lecture on cancer epidemiology and prevention. Cancer Res. 1992;52:6735–40.PubMed Correa P. Human gastric carcinogenesis: a multistep and multifactorial process–first American cancer society award lecture on cancer epidemiology and prevention. Cancer Res. 1992;52:6735–40.PubMed
120.
Zurück zum Zitat Rokkas F. Helicobacter pylori infection as risk factor of carcinoma of the stomach: current evidence. Russ J Gastroenterol Hepatol Coloproctol. 2002;3:66–70. Rokkas F. Helicobacter pylori infection as risk factor of carcinoma of the stomach: current evidence. Russ J Gastroenterol Hepatol Coloproctol. 2002;3:66–70.
122.
Zurück zum Zitat El Maznya A, Hishmata T, Husseina A, Gaithb D. The prevalence of Helicobacter pylori cagA (+ve) among patients with gastric cancer: an Egyptian study. Egypt J Intern Med. 2019;31:522–8.CrossRef El Maznya A, Hishmata T, Husseina A, Gaithb D. The prevalence of Helicobacter pylori cagA (+ve) among patients with gastric cancer: an Egyptian study. Egypt J Intern Med. 2019;31:522–8.CrossRef
126.
Zurück zum Zitat Ferreira RM, Pereira-Marques J, Pinto-Ribeiro I, Costa JL, Carneiro F, MacHado JC, Figueiredo C. Gastric microbial community profiling reveals a dysbiotic cancer-associated microbiota. Gut. 2018;67:226–36.CrossRefPubMed Ferreira RM, Pereira-Marques J, Pinto-Ribeiro I, Costa JL, Carneiro F, MacHado JC, Figueiredo C. Gastric microbial community profiling reveals a dysbiotic cancer-associated microbiota. Gut. 2018;67:226–36.CrossRefPubMed
129.
Zurück zum Zitat Ralser A, Dietl A, Jarosch S, et al. Helicobacter pylori promotes colorectal carcinogenesis by deregulating intestinal immunity and inducing a mucus-degrading microbiota signature. Gut. 2023;72:1258–70.CrossRefPubMed Ralser A, Dietl A, Jarosch S, et al. Helicobacter pylori promotes colorectal carcinogenesis by deregulating intestinal immunity and inducing a mucus-degrading microbiota signature. Gut. 2023;72:1258–70.CrossRefPubMed
131.
Zurück zum Zitat Arai J, Niikura R, Hayakawa Y, et al. Use of antibiotics and probiotics reduces the risk of metachronous gastric cancer after endoscopic resection. Biology (Basel). 2021;10:455.PubMed Arai J, Niikura R, Hayakawa Y, et al. Use of antibiotics and probiotics reduces the risk of metachronous gastric cancer after endoscopic resection. Biology (Basel). 2021;10:455.PubMed
132.
Zurück zum Zitat Niikura R, Hayakawa Y, Nagata N, Miyoshi-Akiayama T, Miyabayashi K, Tsuboi M, et al. Non-Helicobacter pylori gastric microbiome modulates prooncogenic responses and is associated with gastric cancer risk. Gastro Hep Adv. 2023;2:684–700.CrossRef Niikura R, Hayakawa Y, Nagata N, Miyoshi-Akiayama T, Miyabayashi K, Tsuboi M, et al. Non-Helicobacter pylori gastric microbiome modulates prooncogenic responses and is associated with gastric cancer risk. Gastro Hep Adv. 2023;2:684–700.CrossRef
140.
Zurück zum Zitat Yang C, Liang L, Lv P, Liu L, Wang S, Wang Z, Chen Y. Effects of non-viable Lactobacillus reuteri combining with 14-day standard triple therapy on Helicobacter pylori eradication: a randomized double-blind placebo-controlled trial. Helicobacter. 2021;26: e12856. https://doi.org/10.1111/hel.12856.CrossRefPubMed Yang C, Liang L, Lv P, Liu L, Wang S, Wang Z, Chen Y. Effects of non-viable Lactobacillus reuteri combining with 14-day standard triple therapy on Helicobacter pylori eradication: a randomized double-blind placebo-controlled trial. Helicobacter. 2021;26: e12856. https://​doi.​org/​10.​1111/​hel.​12856.CrossRefPubMed
141.
Zurück zum Zitat Kamiya S, Yonezawa H, Osaki T. Role of probiotics in eradication therapy for Helicobacter pylori infection. Adv Exp Med Biol. 2019;1149:243–55.CrossRefPubMed Kamiya S, Yonezawa H, Osaki T. Role of probiotics in eradication therapy for Helicobacter pylori infection. Adv Exp Med Biol. 2019;1149:243–55.CrossRefPubMed
148.
Zurück zum Zitat Grosen A, Mikkelsen S, Baunwall S, Dahlerup J, Erikstrup L, Hvas C, et al. Risk of Helicobacter pylori transmission by faecal microbiota transplantation via oral capsules. Clin Microbiol Infect. 2023;29:799e1–4.CrossRef Grosen A, Mikkelsen S, Baunwall S, Dahlerup J, Erikstrup L, Hvas C, et al. Risk of Helicobacter pylori transmission by faecal microbiota transplantation via oral capsules. Clin Microbiol Infect. 2023;29:799e1–4.CrossRef
Metadaten
Titel
Helicobacter pylori and oral–gut microbiome: clinical implications
verfasst von
Maged T. Elghannam
Moataz H. Hassanien
Yosry A. Ameen
Emad A. Turky
Gamal M. ELattar
Ahmed A. ELRay
Mohammed D. ELTalkawy
Publikationsdatum
02.11.2023
Verlag
Springer Berlin Heidelberg
Erschienen in
Infection / Ausgabe 2/2024
Print ISSN: 0300-8126
Elektronische ISSN: 1439-0973
DOI
https://doi.org/10.1007/s15010-023-02115-7

Weitere Artikel der Ausgabe 2/2024

Infection 2/2024 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Erhöhte Mortalität bei postpartalem Brustkrebs

07.05.2024 Mammakarzinom Nachrichten

Auch für Trägerinnen von BRCA-Varianten gilt: Erkranken sie fünf bis zehn Jahre nach der letzten Schwangerschaft an Brustkrebs, ist das Sterberisiko besonders hoch.

Hypertherme Chemotherapie bietet Chance auf Blasenerhalt

07.05.2024 Harnblasenkarzinom Nachrichten

Eine hypertherme intravesikale Chemotherapie mit Mitomycin kann für Patienten mit hochriskantem nicht muskelinvasivem Blasenkrebs eine Alternative zur radikalen Zystektomie darstellen. Kölner Urologen berichten über ihre Erfahrungen.

Ein Drittel der jungen Ärztinnen und Ärzte erwägt abzuwandern

07.05.2024 Medizinstudium Nachrichten

Extreme Arbeitsverdichtung und kaum Supervision: Dr. Andrea Martini, Sprecherin des Bündnisses Junge Ärztinnen und Ärzte (BJÄ) über den Frust des ärztlichen Nachwuchses und die Vorteile des Rucksack-Modells.

Vorhofflimmern bei Jüngeren gefährlicher als gedacht

06.05.2024 Vorhofflimmern Nachrichten

Immer mehr jüngere Menschen leiden unter Vorhofflimmern. Betroffene unter 65 Jahren haben viele Risikofaktoren und ein signifikant erhöhtes Sterberisiko verglichen mit Gleichaltrigen ohne die Erkrankung.

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.