Open access peer-reviewed chapter

Gastric Microbiota: Between Health and Disease

Written By

Hristo Ilianov Iliev, Mila Dimitrova Kovacheva-Slavova, Todor Asenov Angelov, Hristo Yankov Valkov, Ali Bedran and Borislav Georgiev Vladimirov

Submitted: 28 January 2019 Reviewed: 18 May 2019 Published: 10 August 2019

DOI: 10.5772/intechopen.86926

From the Edited Volume

Gastrointestinal Stomas

Edited by Vincenzo Neri

Chapter metrics overview

969 Chapter Downloads

View Full Metrics

Abstract

The etiologic link between H. pylori infection and gastric chronic inflammation and related complications has been well established, but pathogenic pathways are still widely discussed and not sufficiently clear. The introduction of culture-independent molecular techniques has allowed better understanding of the gastric microbiota and has revealed that, when present, H. pylori represents the main colonizer but is part of a far more complex and dynamic microbiota than previously thought. This conceptual shift has made way for new pathogenic theories, focused on the interrelations between H. pylori and other gastric microbiota. Main factors that affect the gastric microbiota are gastric acidity, inflammation, and environmental factors, such as diet and drugs. Previous studies have made progress in explaining the complex interactions between gastric microorganisms in healthy individuals and their role in the development of related gastroduodenal (peptic ulcers and gastric cancer (GC)) and extraintestinal diseases, but more scientific proof is needed. This review presents current knowledge on gastric microbiota and its role in health and in the development of gastroduodenal diseases.

Keywords

  • gastric microbiota
  • H. pylori
  • pathogenesis
  • chronic inflammation
  • peptic ulcers
  • gastric cancer

1. Introduction

The first isolation of Helicobacter pylori (H. pylori) in 1982 [1] changed the traditional misconception that the stomach, with its naturally hostile environment, cannot be a reservoir for microbial species. Since then H. pylori has been well established as a key factor in gastric pathology, being the main cause of chronic active gastritis. This inflammation can progress to often interlinked severe complications such as atrophic gastritis, peptic ulcer disease, and gastric malignancies [2]. The assumption of the uniqueness of H. pylori as the only bacteria able to survive in the gastric hostile environment has also fast been shattered with the advances in culture-dependent and culture-independent techniques that revealed that H. pylori is part of a complex gastric microbiota. The gastric microbial density is now estimated at around 102 to 104 colony-forming units (CFU)/mL [3], with variations related to local pH, site of isolation, and environmental factors, such as food ingestion and medication intake. Furthermore, the pool of species and subspecies varies related to H. pylori status and related pathologic complications. Due to this multitude of factors, the differentiation between resident and transient microbiota and its role in health and disease remains controversial. Nevertheless, the potential of understanding the structure and dynamics of the gastric microbiota for the pathogenesis, diagnosis, and treatment of gastroduodenal diseases remains. Hence, this review aims to underline current knowledge on gastric microbiota and its relation to gastroduodenal pathology.

Advertisement

2. The hostile gastric environment

Compared to other gastrointestinal (GI) segments, the stomach has a physiological environment that is significantly more hostile to bacterial colonization and is a crucial part of the dynamics of the gastric microbiota. Primary reason for this is the gastric juice, which is composed of two main components—proteolytic enzymes and hydrochloric acid (HCl). The hydrochloric acid creates a strong acidic environment by maintaining a pH of 1–2 in the gastric lumen, which together with the proteolytic features of the gastric enzymes creates an intragastric environment that serves both digestive and protective roles. This environment facilitates the denaturation of proteins and nutrient absorption but also severely limits bacterial colonization and survival, preventing infection by pathogens [4]. The low pH value is the main restrictive component of the gastric juice [5]. To prevent damage to the mucosa from the acid and enzymes, neck cells of the gastric glands secrete mucus on the surface of the gastric epithelium. This mucus layer establishes a pH gradient that increases the pH up to 6–7 at the surface of the mucosa [6]. This is due to the unique properties of the mucus which permit acid to flow from parietal cells into crypts which communicate with the lumen, but do not allow acid at pH <4 from penetrating the mucus layer [6]. The mucus layer consists of several different mucin molecules, including MUC1, MUC5AC, MUC5AB, and MUC6, and forms two sublayers, an inner mucus layer that is firmly attached to the epithelia and a loose mucus layer, which is in direct contact with the lumen [7, 8]. Additional factors that contribute to the strong antimicrobial environment of the stomach are the accidental bile reflux and the gastric peristalsis.

Advertisement

3. H. pylori

In 1982 Helicobacter pylori was isolated by Barry Marshall and Robin Warren [1]. Their research changed the long-standing view that the stomach is as a sterile organ, being naturally hostile to bacterial survival. Today, it is known that H. pylori infects more than 50% of the world’s population with the only significant reservoir for the infection appearing to be humans. Possible routes of infection include oral-oral, fecal-oral, and iatrogenic spread (e.g., by unsterile endoscopic interventions). In developing countries, infection is usually acquired early in childhood, unlike in industrialized countries, where it develops more commonly in adulthood [9].

H. pylori is a Gram-negative, spiral-shaped, motile, and flagellated bacteria that is uniquely adapted to colonizing the gastric niche. Hence, it comes with no surprise that when present, H. pylori has the highest relative abundance among all gastric microbial communities in both adults and children [10, 11, 12]. Upon infection, H. pylori utilizes urease and α-carbonic anhydrase to generate ammonia and HCO3. This neutralizes H+ and locally increases the pH, facilitating the bacteria’s passage through the acidic gastric fluid and the pH-sensitive mucous layer. Using chemotaxis, the bacteria navigates the pH gradient to their niche near the host’s epithelium [13, 14]. Once established in the inner mucus layer, H. pylori can utilize diverse adhesins (e.g., SabA and BabA) to attach to epithelial cells. Once attached, bacterial effector molecules, such as the vacuolating cytotoxin (VacA) and the cytotoxin-associated gene A (CagA), modulate the gastric epithelial cell behavior, leading to loss of cell polarity, release of nutrients and chemokines, and regulation of acid secretion via control of gastrin and H+/K+ ATPase [9, 15, 16]. In response to the H. pylori infection, the host mounts a complex inflammatory response, which ultimately leads to active chronic gastritis and subsequent gastroduodenal diseases. Therefore, the host’s attempts to eradicate H. pylori increase gastric immunopathology (gastritis, epithelial damage such as atrophy and intestinal metaplasia), which alters the gastric compartment and its microbiota and may subsequently progress to gastric cancer.

Advertisement

4. Other non-H. pylori microbiota

4.1 Culture-dependent identification of gastric microbiota

Initial studies on the bacteria present in the stomach, using culture-based techniques, such as gastric juice cultures and mucosal biopsies were reported even before the isolation of H. pylori. In 1977 Savage DC [17] isolated bacteria from the stomach estimated at >103 CFU/g. The predominant phyla were Firmicutes (genera Lactobacillus, Streptococcus, Clostridium, and Veillonella), Actinobacteria (genus Bifidobacterium), and Proteobacteria (coliforms). However, due to the fact that these bacteria are prevalent along the whole GI tract, they were considered transient bacteria, which form small colonies that exist for short periods of time, rather than true gastric colonizers. Later culture-based studies [18, 19, 20, 21, 22, 23] find that the most prevalent phylum, regardless of H. pylori status, is Firmicutes, followed by Proteobacteria and Bacteroidetes. Actinobacteria varies in studies as the second or third most prevalent phylum. The most commonly found genera were Streptococcus, Lactobacillus, Bacteroides, Staphylococcus, Veillonella, Corynebacterium, and Neisseria. However, given that culturing conditions for the majority of microbes colonizing the GI tract are not established, culture-based methods are considered to underestimate the gastric microbial diversity and are largely replaced by culture-independent methods.

4.2 Culture-independent identification of gastric microbiota

Culture-independent studies use a variety of molecular methods based on 16S rRNA gene sequencing. A multitude of reasons define these methods as far superior to those which are culture-dependent. These include:

  • 16S rRNA is present in almost all bacteria.

  • The function of the 16S rRNA gene has remained unchanged over time, suggesting that random sequence changes are a more accurate measure of time (evolution).

  • The 16S rRNA gene is large enough for computational purposes [24].

A variety of 16S rRNA based methods exist, including:

  • Fluorescent in situ hybridization (FISH) [25]

  • Dot-blot hybridization with rRNA-targeted probes [26]

  • Targeted qPCR [27]

  • Traditional or sequence-aided community fingerprinting [28]

  • Temperature gradient gel electrophoresis (TGGE) [29]

  • Terminal restriction fragment length polymorphism (T-RFLP) [30]

  • Sequencing of cloned 16S rDNA [29]

  • Microarrays (PhyloChip) [31]

  • Next-generation sequencing (NGS) [32]

In an extensive review of eight culture-independent studies, Sheh A. and Fox J. concluded that the most prominent phyla in the stomach are Proteobacteria, Firmicutes, Bacteroidetes, Actinobacteria, and Fusobacteria [33]. Furthermore, the most abundant phyla in H. pylori-positive subjects are Proteobacteria, Firmicutes, and Actinobacteria. In the absence of H. pylori, the most abundant phyla are Firmicutes, Bacteroidetes, and Actinobacteria. However, H. pylori remains the most dominant species in the stomach, comprising 72–99% of sequencing reads [10, 34]. In the absence of H. pylori, analysis consistently shows the presence of Streptococcus spp., which seem to be the most abundant genus in H. pylori-negative subjects [3, 30, 35, 36]. In the gastric microbiota, the non-Helicobacter genera commonly detected are Streptococcus, Prevotella, Veillonella, and Rothia.

Advertisement

5. Interrelations between H. pylori and other gastric microbiota

Numerous studies have shown significant variability of the gastric microbial communities with respect to H. pylori status. One such study was carried out by Osaki et al. [37], who examined the gastric microbiota of H. pylori-positive and H. pylori-negative Mongolian gerbils. The study showed a larger number of Bifidobacterium spp. in H. pylori-positive gerbils, compared to the H. pylori-negative, while Eubacterium cylindroides and Prevotella spp. were only found in the H. pylori-negative group.

Several mouse model studies have also shown clear differences in the composition of the gastric microbiota with respect to H. pylori status. Infection by H. pylori of pathogen-free female BALB/c mice has been shown to reduce the Lactobacillus spp. in the gastric microflora [26]. In transgenic, insulin-gastrin (INS-GAS) mice, the H. pylori-infected male mice show a significantly different phyla compared to the non-infected control group, with an increase in Firmicutes and a decrease in Bacteroidetes [26]. Findings in H. pylori-colonized C57BL/6 N female mice included reductions in Firmicutes (class Bacilli), Bacteroidetes, and Proteobacteria and an increase of Firmicutes (class Clostridia), Proteobacteria (genus Helicobacter), and Verrucomicrobia. However, other published data on the murine gastric microbiota suggest that neither acute nor chronic H. pylori infection substantially modifies the gastric microbial ecosystem [38].

A few studies have also examined H. pylori-related microbial differences in humans. One study found relative abundances of Proteobacteria, Spirochetes, and Acidobacteria in H. pylori-positive patients, compared to the control H. pylori-negative group. Another study demonstrated that patients positive for H. pylori culture showed significantly increased colonization of Proteobacteria and a decrease in Actinobacteria [39].

A few studies have given insight on how other microbial species can affect H. pylori by modulating H. pylori-induced gastric inflammatory responses. Two studies have indicated that the presence of intestinal Helicobacter (H. bilis, H. hepaticus, and H. muridarum) can both increase and decrease the severity of H. pylori-induced gastric inflammation by altering Treg cell responses [40, 41]. Another study demonstrated that H. pylori is present within the intestine in a coccoid form and that its interaction with phagocytes within the intestinal Peyer’s patches modifies the intensity of H. pylori-induced gastritis [42]. However, other studies have shown that the gastric microbiota can accelerate gastric cancer progression in the presence of H. pylori and does so with no differences detected in the composition of the intestinal microbiota [27, 32].

As previously described H. pylori infection is associated with a multitude of changes in the gastric physiology and immunology, e.g., reduced gastric acidity, disturbed nutrient availability, and local inflammatory responses. These changes might be one explanation for the shift in the gastric microbial communities described, but the relation between H. pylori and non-H. pylori microbiota seems to be far more complex and remains to be further clarified. One problem is that so far, most studies are focused only on the effects of H. pylori on other microbiota, but little is known of how H. pylori is affected by other resident bacteria. Another problem is that different studies cannot be objectively compared, since they highly vary in methods and models used and the results depend on numerous other factors such as the time of H. pylori infection and the degree of mucosal inflammation. Therefore, further experiments are needed to give a more extensive understanding of these complex microbial interrelations.

Advertisement

6. Factors affecting the gastric microbiota

As described, H. pylori is the most significant species that colonizes the stomach and a key factor for the gastric microbial diversity but is far from sufficient to provide a wholesome understanding of the factors that determine its dynamics. Major factors that influence and define the dynamics of the gastric microbiota include gastric acidity, inflammation of the gastric mucosa, dietary habits, and use of medications.

6.1 Gastric acidity

The human gastric juice has an interprandial pH of between 1 and 2 in the gastric lumen, whereas with food ingestion it can reach up to pH 5. pH also varies in the different anatomical regions of the stomach, with most acidic being the fundus and the least being the antrum. The mucus lining the gastric mucosa establishes also a pH gradient from the lumen to the surface of the epithelium. This mucus consists of two sublayers—an inner mucus layer that is firmly attached to the epithelium and a variable mucus layer directly interacting with the lumen [7, 8]. Thus, across the mucus layer, the pH ranges from about 5.5 to 6.8 or even 7 at the surface of the gastric epithelial cells [5, 6]. It was already discussed that the low pH, caused by the hydrochloric acid, restricts the quantity of microorganisms and reduces the risk of infection by pathogens. Hence, sites with higher pH are significantly more hospitable to colonization and have a higher microbial density. Considerable fluctuations in the microbial density have been described with respect to the pH in the stomach, whereby both the quantity and the proportion of genera also fluctuate [43, 44]. Bacteria and bacterial DNA, which are isolated from gastric juice, differ from bacterial isolates adhering to the mucosa. During abnormal conditions, this balance may be different.

6.2 Dietary habits

While many studies document the effects of diet on the gut microbiota composition in humans, [45, 46, 47, 48, 49] there are only a few, mainly animal model studies, addressing the influence of diet on the gastric microbiota. An example is an in vivo study that compared the gastric microbiota of mice fed a non-purified diet (natural source-derived food) to mice fed a purified diet (refined food) and found higher levels of total aerobes, total anaerobes, and Lactobacillus in the stomach of the mice on a non-purified diet [50]. Nevertheless, it is well established how dietary factors affect the gastric microclimate, and since the microbiota is an inseparable part of this microclimate, it is not farfetched to suspect that diet affects the gastric microbial communities. However, more research is needed.

6.3 Use of medications

The long-term use of proton pump inhibitors (PPIs) and H2 antagonists affects the composition of the gastric microbiota by inducing a non-H. pylori bacterial overgrowth [51]. This is not surprising, considering that normally the non-H. pylori gastric microbiota is suppressed by the significantly acidic gastric environment. Suppression of gastric acidity will alter the bacterial flora of the upper GI tract, and studies have confirmed that PPIs do alter the bacterial population in the stomach [52]. This is mainly due to oral bacteria that survive instead of being killed in the normally acidic stomach. It has also been suspected that by causing alteration and overgrowth of the microbiota, acid-suppressive treatments may increase the risk of gastric cancer [52]. It has also been shown that a previously antrum-dominant H. pylori infection after treatment with acid inhibitors changed to a more corpus predominant infection [51]. The less acidic corpus allows H. pylori to penetrate deeper in the crypts and increase the inflammation, which causes faster progression to atrophy [53]. Treatment with acid inhibitors has by culture-dependent methods been shown to affect the survival of bacteria in the stomach. However, no significant differences have been found regarding diversity and composition of the microbiota by using culture methods [10].

Antibiotics are well known to have suppressive effects on the gastrointestinal microflora. H. pylori eradication is dependent on combined antibiotic treatment. However, certain antibiotic treatments can have negative effects on the “healthy” gastric microbiota. Animal studies indicate that treatment with penicillins reduces Lactobacillus populations and promotes yeast colonization of the gastric epithelium. Furthermore, Mason et al. [54] showed that cefoperazone treatment in humans causes long-term alteration of the gastric microbiota, such as a significant reduction in the number of Lactobacillus and overgrowth of Enterococcus.

Advertisement

7. Gastric microbiota and gastroduodenal diseases

7.1 Chronic gastritis and peptic ulcer disease

The isolation of H. pylori was a real breakthrough, not because it declared the stomach as a non-sterile organ but because subsequent research established it as a main etiological factor for the development of gastroduodenal diseases [2, 55, 56]. It is well known that long-term H. pylori infection causes various degrees of chronic inflammation of the underlying gastric mucosa. A subset of patients develops clinical symptoms, and a further subset will develop complications including peptic ulcer, gastric mucosa-associated lymphoid tissue (MALT) lymphoma, and gastric cancer. The cascade leading from chronic gastritis to neoplasm is known as Correa’s cascade and involves the progression to glandular atrophy with intestinal metaplasia and dysplasia and eventually to invasive carcinoma [57]. Peptic ulcer disease is the most common complication of chronic H. pylori infection, with 95% of duodenal and 70% of gastric ulcers being linked to it [58]. All of this serves to prove that H. pyloriis by far the most important microbial species that can colonize the stomach, with an enormous impact on the pathogenesis and development of gastroduodenal diseases.

Nevertheless, there is arising evidence that non-H. pylori bacteria may also play an important role in the pathogenesis of chronic gastritis and peptic ulcers. One study suggests that different gastric microbial communities, such as the overrepresentation of the Streptococcus genus within the Firmicutes phylum, can lead to gastritis as well, even in the absence of H. pylori [35]. Another study has found an increase of Streptococcus and a decrease of Prevotella in patients with atrophic gastritis, versus healthy subjects [59]. There are also several studies that assess the role of non-H. pylori species in peptic ulcer disease. The non-H. pylori bacteria seems to be more prevalent in patients with non-ulcer dyspepsia than in those with gastric ulcer as shown by a culture-based study by Hu et al. [19]. Another study in Malaysian patients showed significant correlation between the isolation of Streptococcus and patients with peptic ulcers [60]. Given that most studies find that the Streptococcus genus is one of the most abundant non-H. pylori species in the stomach, a possible pathogenic relation can be a subject of further research. However, by far no other species have an established pathogenic role except H. pylori.

7.2 Gastric cancer

Each year approximately 990,000 people are diagnosed with gastric cancer (GC) worldwide, of whom about 738,000 die from this disease, making GC the fourth most common incident cancer and the second most common cause of cancer death. Both incidence and mortality rates are about twice as high in males as in females. Over 70% of cases occur in developing nations, concentrated in Eastern Asia, Eastern Europe, and Central and South America. Approximately 90% of gastric cancers are adenocarcinomas, with the other 10% shared between mucosa-associated lymphoid tissue (MALT) lymphomas, gastrointestinal stromal tumors (GIST), leiomyosarcomas, and other more rare types of cancer. Adenocarcinomas are histologically classified into two major types: diffuse and intestinal. These two types not only look different under the microscope but also differ in gender ratio, age at diagnosis, and other epidemiologic features. Anatomically, gastric cancers are categorized as proximal and distal. Proximal adenocarcinomas are more similar to esophageal adenocarcinomas and may be associated with the absence of H. pylori, while distal adenocarcinomas originate in the antrum, with approximately 90% of such cases related to H. pylori infection [61]. Today the correlation between H. pylori and the development of gastric cancer is undeniable as shown in several prospective studies [62, 63, 64]. Moreover, the eradication of H. pylori is proven to significantly reduce the risk of gastric cancer development, according to several international consensuses [65, 66]. H. pylori was recognized as a “definite carcinogen” by the World Health Organization in 1994, and this fact was reconfirmed in 2009.

However, H. pylori coevolved with humans for millennia, and only 1–2% of people infected with this bacterium actually develop gastric cancer or MALT lymphoma. Similar to most cancers, pathogenetic mechanisms remain unclear with a multitude of other factors to influence the final carcinogenesis [61]. Although, H. pylori is clearly the most relevant microbial risk factor for the development of gastric cancer, an increasing pool of evidence suggests that other microbial communities play a causative role in the pathophysiology of gastric cancer. To date, several animal and human studies have supported this theory.

Studies with INS-GAS mice have revealed that male mice with intestinal microbiota developed gastric pathology from chronic gastritis to atrophy and dysplasia independent of H. pylori infection. Furthermore, the presence of commensal microbiota accelerated the progression to gastric intraepithelial neoplasia, and gastric intraepithelial neoplasia became invasive in H. pylori-infected INS-GAS mice. Male INS-GAS mice with H. pylori infection, colonized with artificial mouse intestinal microbiota, have shown increased incidence of gastric intraepithelial neoplasia by 69% [27, 32]. On the other hand, antibiotic treatments significantly delayed the onset of gastric neoplasia in Helicobacter-free and specific pathogen-free INS-GAS mice [67].

A study by Wang et al. found a similar number of bacterial species in the microbiota between gastric cancer and chronic gastritis, but by using a method to explore and visualize similarities or dissimilarities of the data, a pattern suggesting the presence of a diversified microbiota in gastric cancer was found [68]. Moreover, a 16S rRNA gene sequencing analysis of gastric mucosa of patients with gastric cancer showed a prevalence of the genera Lactobacillus, Streptococcus (among which the most common species were S. mitis and Streptococcus parasanguinis), Prevotella, and Veillonella [30]. Two other studies evaluated the gastric microbiota of subjects with non-atrophic gastritis, intestinal metaplasia, and gastric cancer. The first one showed a significantly lower diversity and a higher abundance of the genus Pseudomonas in the microbiota of neoplastic patients compared to patients with simple gastritis. Moreover, both the progressive decrease of six taxa and the progressive increase of two taxa were observed from the gastritis group to the neoplastic group, via the metaplastic group [69]. In the second study, a high-throughput sequencing platform was used for the assessment of gastric microbiota in the three groups, showing definitely different results: a greater bacterial diversity, a relative rise of Bacilli and Streptococcaceae, and a relative reduction of Helicobacteraceae were found in the cancer group compared to other groups [70].

It is possible that non-H. pylori species potentiate carcinogenesis through various mechanisms, such as promoting inflammation, stimulating cell proliferation, modifying stem cell dynamics, and producing toxic metabolites [71]. However, it is still unsure whether the different microbial structure is causative for the carcinogenesis or carcinogenesis itself causes a shift in the microbial communities, which subsequently promotes carcinogenesis further.

Advertisement

8. Gastric microbiota and extra-gastric diseases

The stomach is part of the GI tract, and as such, possible relations between the gastric microbiota’s composition and diseases of other parts of the GI tract, such as the esophagus (esophagitis and esophageal cancer), small intestines, and colon cannot be overlooked [72]. One study, using 16S rDNA analyses of duodenal aspirates, demonstrated lower diversity in irritable bowel syndrome patients compared to controls with significant alterations in 12 genera [73]. An increased risk for colorectal neoplasia in H. pylori-infected patients has been confirmed by many large-scale studies [74, 75].

Other studies have addressed the association between autoimmune hepatitis and altered microbiome of the upper GI tract and found this to be linked to increased intestinal permeability [76].

Regarding the extra-gastrointestinal involvement of gastric microbiota (especially H. pylori), studies find possible associations with hematological diseases like idiopathic thrombocytopenic purpura [77] and anemia [78] and cardiovascular [79], neurological [80], and endocrine [81] diseases.

Nevertheless, research in this field is far from sufficient to be conclusive.

Advertisement

9. Discussion

It is undeniable that H. pylori is by far the most unique and important species that can colonize the gastric niche with clear pathogenic significance to gastroduodenal diseases. However, culture-dependent and later culture-independent studies prove that the stomach harbors a complex microbiota with many other phyla (Proteobacteria, Firmicutes, Bacteroidetes, Actinobacteria, and Fusobacteria) and genera (Lactobacillus, Streptococcus, Clostridium, Prevotella, Veillonella, Bifidobacterium, and Rothia) being identified. By far, it is arguable which genera can be considered resident since results depend on a multitude of factors, among which gastric acidity stands out as the most significant. However, it is worth noting that Streptococcus, Prevotella, Veillonella, and Rothia seem to be the most abundant genera in H. pylori-negative subjects. Nevertheless, the significance of both transient and resident non-H. pylori microbiota lies in its possible role in the development and progression of gastroduodenal diseases, such as gastritis, peptic ulcer disease, and gastric cancer. So far, only a few and far from enough studies suggest that non-H. pylori microbiota can lead to gastric pathology in the absence of H. pylori. However, findings are convincing that the non-H. pylori microbiota can play an important role in modulating H. pylori-induced gastric inflammation, influencing an individual’s risk of gastric diseases and consequently the severity of the resulting disease. Suspected pathophysiologic mechanisms involved in this include modulating immune cell responses, stimulating cell proliferation, modifying stem cell dynamics, and producing toxic metabolites. Although, substantial advancements in unrevealing the complexity of the gastric microbiota and its’ role in health and disease have been made, studies are far from sufficient to suggest new strategies for prevention, diagnosis, and treatment of gastroduodenal diseases. However, this potential remains and undoubtedly should be further explored.

Advertisement

Acknowledgments

The publication of this work was supported by KRKA.

Conflict of interest

There are no conflicts of interest.

References

  1. 1. Warren JR, Marshall BJ. Unidentified curved bacilli on gastric epithelium in active chronic gastritis. Lancet. 1983;1:1273-1275. DOI: 10.1016/S0140-6736(83)92719-8
  2. 2. Wang F, Meng W, Wang B, Qiao L. Helicobacter pylori-induced gastric inflammation and gastric cancer. Cancer Letters. 2014;345(2):196-202. DOI: 10.1016/j.canlet.2013.08.016
  3. 3. Delgado S, Cabrera-Rubio R, Mira A, Suárez A, Mayo B. Microbiological survey of the human gastric ecosystem using culturing and pyrosequencing methods. Microbial Ecology. 2013;65(3):763-772. DOI: 10.1007/s00248-013-0192-5
  4. 4. Martinsen TC, Bergh K, Waldum HL. Gastric juice: A barrier against infectious diseases. Basic & Clinical Pharmacology & Toxicology. 2005;96:94-102. DOI: 10.1111/j.1742-7843.2005.pto960202.x
  5. 5. Manson JM, Rauch M, Gilmore MS. The commensal microbiology of the gastrointestinal tract. Advances in Experimental Medicine and Biology. 2008;635:15-28. DOI: 10.1007/978-0-387-09550-9_2
  6. 6. Bhaskar KR, Garik P, Turner BS, Bradley JD, Bansil R, Stanley HE, et al. Viscous fingering of HCl through gastric mucin. Nature. 1992;360:458-461. DOI: 10.1038/360458a0
  7. 7. Corfield AP, Carroll D, Myerscough N, Probert CS. Mucins in the gastrointestinal tract in health and disease. Frontiers in Bioscience. 2001;6:D1321-D1357. DOI: 10.2741/Corfield
  8. 8. Atuma C, Strugala V, Allen A, Holm L. The adherent gastrointestinal mucus gel layer: Thickness and physical state in vivo. American Journal of Physiology. Gastrointestinal and Liver Physiology. 2001;280:G922-G929. DOI: 10.1152/ajpgi.2001.280.5.G922
  9. 9. Kusters JG, van Vliet AH, Kuipers EJ. Pathogenesis of Helicobacter pylori infection. Clinical Microbiology Reviews. 2006;19(3):449-490. DOI: 10.1128/CMR.00054-05
  10. 10. Bik EM, Eckburg PB, Gill SR, et al. Molecular analysis of the bacterial microbiota in the human stomach. Proceedings of the National Academy of Sciences of the United States of America. 2006;103(3):732-737. DOI: 10.1073/pnas.0506655103
  11. 11. Schulz C, Schütte K, Koch N, et al. The active bacterial assemblages of the upper GI tract in individuals with and without Helicobacter infection. Gut. 2016;67(2):216-225. DOI: 10.1136/gutjnl-2016-312904
  12. 12. Llorca L, Pérez-Pérez G, Urruzuno P, et al. Characterization of the gastric microbiota in a pediatric population according to Helicobacter pylori status. The Pediatric Infectious Disease Journal. 2017;36(2):173-178. DOI: 10.1097/INF.0000000000001383
  13. 13. Williams SM, Chen YT, Andermann TM, Carter JE, McGee DJ, Ottemann KM. Helicobacter pylori chemotaxis modulates inflammation and bacterium-gastric epithelium interactions in infected mice. Infection and Immunity. 2007;75:3747-3757. DOI: 10.1128/IAI.00082-07
  14. 14. Croxen MA, Sisson G, Melano R, Hoffman PS. The Helicobacter pylori chemotaxis receptor TlpB (HP0103) is required for pH taxis and for colonization of the gastric mucosa. Journal of Bacteriology. 2006;188:2656-2665. DOI: 10.1128/JB.188.7.2656-2665.2006
  15. 15. Saha A, Backert S, Hammond CE, Gooz M, Smolka AJ. Helicobacter pylori CagL activates ADAM17 to induce repression of the gastric H, K-ATPase alpha subunit. Gastroenterology. 2010;139:239-248. DOI: 10.1053/j.gastro.2010.03.036
  16. 16. O’Keeffe J, Moran AP. Conventional, regulatory, and unconventional T cells in the immunologic response to Helicobacter pylori. Helicobacter. 2008;13:1-19. DOI: 10.1111/j.1523-5378.2008.00559.x
  17. 17. Savage DC. Microbial ecology of the gastrointestinal tract. Annual Review of Microbiology. 1977;31:107-133. DOI: 10.1146/annurev.mi.31.100177.000543
  18. 18. Stockbruegger RW. Bacterial overgrowth as a consequence of reduced gastric acidity. Scandinavian Journal of Gastroenterology. Supplement. 1985;111:7-16. DOI: 10.3109/00365528509093749
  19. 19. Hu Y, He LH, Xiao D, Liu GD, Gu YX, Tao XX, et al. Bacterial flora concurrent with Helicobacter pylori in the stomach of patients with upper gastrointestinal diseases. World Journal of Gastroenterology. 2012;18:1257-1261. DOI: 10.3748/wjg.v18.i11.1257
  20. 20. Zilberstein B, Quintanilha AG, Santos MA, Pajecki D, Moura EG, Alves PR, et al. Clinics (São Paulo, Brazil). 2007;62:47-54. DOI: 10.1590/S1807-59322007000100008
  21. 21. Sharma BK, Santana IA, Wood EC, Walt RP, Pereira M, Noone P, et al. Intragastric bacterial activity and nitrosation before, during, and after treatment with omeprazole. British Medical Journal. 1984;289:717-719. DOI: 10.1136/bmj.289.6447.717
  22. 22. Adamsson I, Nord CE, Lundquist P, Sjöstedt S, Edlund C. Comparative effects of omeprazole, amoxycillin plus metronidazole versus omeprazole, clarithromycin plus metronidazole on the oral, gastric and intestinal microflora in Helicobacter pylori-infected patients. The Journal of Antimicrobial Chemotherapy. 1999;44:629-640. DOI: 10.1093/jac/44.5.629
  23. 23. Sjöstedt S, Heimdahl A, Kager L, Nord CE. Microbial colonization of the oropharynx, esophagus and stomach in patients with gastric diseases. European Journal of Clinical Microbiology. 1985;4:49-51. DOI: 10.1007/BF02148660
  24. 24. Janda JM, Abbott SL. 16S rRNA gene sequencing for bacterial identification in the diagnostic laboratory: Pluses, perils, and pitfalls. Journal of Clinical Microbiology. 2007;45(9):2761-2764. DOI: 10.1128/JCM.01228-07
  25. 25. Langendijk PS, Schut F, Jansen GJ, Raangs GC, Kamphuis GR, Wilkinson MH, et al. Quantitative fluorescence in situ hybridization of Bifidobacterium spp. with genus-specific 16S rRNAtargeted probes and its application in fecal samples. Applied and Environmental Microbiology. 1995;61:3069-3075
  26. 26. Aebischer T, Fischer A, Walduck A, Schlötelburg C, Lindig M, Schreiber S, et al. Vaccination prevents Helicobacter pylori-induced alterations of the gastric flora in mice. FEMS Immunology and Medical Microbiology. 2006;46:221-229. DOI: 10.1111/rp10.1016-j.femsim.2004.05.008
  27. 27. Lertpiriyapong K, Whary MT, Muthupalani S, Lofgren JL, Gamazon ER, Feng Y, et al. Gastric colonisation with a restricted commensal microbiota replicates the promotion of neoplastic lesions by diverse intestinal microbiota in the Helicobacter pylori INS-GAS mouse model of gastric carcinogenesis. Gut. 2013;63:54-63. DOI: 10.1136/gutjnl-2013-305178
  28. 28. Heilig HG, Zoetendal EG, Vaughan EE, Marteau P, Akkermans AD, de Vos WM. Molecular diversity of Lactobacillus spp. and other lactic acid bacteria in the human intestine as determined by specific amplification of 16S ribosomal DNA. Applied and Environmental Microbiology. 2002;68:114-123. DOI: 10.1128/AEM.68.1.114-123.2002
  29. 29. 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 16SrDNA sequence analysis. Journal of Medical Microbiology. 2000;49:817-822. DOI: 10.1099/0022-1317-49-9-817
  30. 30. Dicksved J, Lindberg M, Rosenquist M, Enroth H, Jansson JK, Engstrand L. Molecular characterization of the stomach microbiota in patients with gastric cancer and in controls. Journal of Medical Microbiology. 2009;58:509-516. DOI: 10.1099/jmm.0.007302-0
  31. 31. Rolig AS, Cech C, Ahler E, Carter JE, Ottemann KM. The degree of Helicobacter pylori-triggered inflammation is manipulated by preinfection host microbiota. Infection and Immunity. 2013;81:1382-1389. DOI: 10.1128/IAI.00044-13
  32. 32. Lofgren JL, Whary MT, Ge Z, Muthupalani S, et al. Lack of commensal flora in Helicobacter pylori-infected INS-GAS mice reduces gastritis and delays intraepithelial neoplasia. Gastroenterology. 2011;140:210-220. DOI: 10.1053/j.gastro.2010.09.048
  33. 33. Sheh A, Fox JG. The role of the gastrointestinal microbiome in Helicobacter pylori pathogenesis. Gut Microbes. 2013;4(6):505-531. DOI: 10.4161/gmic.26205
  34. 34. Andersson AF, Lindberg M, Jakobsson H, Bäckhed F, Nyrén P, Engstrand L. Comparative analysis of human gut microbiota by barcoded pyrosequencing. PLoS One. 2008;3:e2836. DOI: 10.1371/journal.pone.0002836
  35. 35. Li XX, Wong GL, To KF, Wong VW, Lai LH, Chow DK, et al. Bacterial microbiota profiling in gastritis without Helicobacter pylori infection or non-steroidal anti-inflammatory drug use. PLoS One. 2009;4:e7985. DOI: 10.1371/journal.pone.0007985
  36. 36. Stearns JC, Lynch MD, Senadheera DB, Tenenbaum HC, Goldberg MB, Cvitkovitch DG, et al. Bacterial biogeography of the human digestive tract. Scientific Reports. 2011;1:170. DOI: 10.1038/srep00170
  37. 37. Osaki T, Matsuki T, Asahara T, Zaman C, Hanawa T, Yonezawa H, et al. Comparative analysis of gastric bacterial microbiota in Mongolian gerbils after long-term infection with Helicobacter pylori. Microbial Pathogenesis. 2012;53(1):12-18
  38. 38. Tan MP, Kaparakis M, Galic M, Pedersen J, Pearse M, Wijburg OL, et al. Chronic Helicobacter pylori infection does dot significantly alter the microbiota of the murine stomach. Applied and Environmental Microbiology. 2007;73(3):1010-1013
  39. 39. Maldonado-Contreras A, Goldfarb KC, Godoy-Vitorino F, et al. Structure of the human gastric bacterial community in relation to Helicobacter pylori status. The ISME Journal. 2010;5(4):574-579. DOI: 10.1038/ismej.2010.149
  40. 40. Lemke LB, Ge Z, Whary MT, et al. Concurrent Helicobacter bilis infection in C57BL/6 mice attenuates proinflammatory H. pylori-induced gastric pathology. Infection and Immunity. 2009;77(5):2147-2158. DOI: 10.1128/IAI.01395-08
  41. 41. Ge Z, Feng Y, Muthupalani S, et al. Infection and Immunity. 2011;79(10):3861-3871. DOI: 10.1128/IAI.05357-11
  42. 42. Nagai S, Mimuro H, Yamada T, et al. Role of Peyer's patches in the induction of Helicobacter pylori-induced gastritis. Proceedings of the National Academy of Sciences of the United States of America. 2007;104(21):8971-8976. DOI: 10.1073/pnas.0609014104
  43. 43. Draser BS, Shiner M, McLeod GM. Studies of the intestinal flora 1. The bacterial flora of the gastrointestinal tract in healthy and achlorhydric patients. Gastroenterology. 1969;56:71-79
  44. 44. Milton-Thompson GJ, Lightfoot NF, Ahmet Z, et al. Intragastric acidity, bacteria, nitrite, and N-nitroso compounds before, during, and after cimetidine treatment. Lancet. 1982;1(8281):1091-1095
  45. 45. Goldsmith JR, Sartor RB. The role of diet on intestinal microbiota metabolism: Downstream impacts on host immune function and health, and therapeutic implications. Journal of Gastroenterology. 2014;49(5):785-798. DOI: 10.1007/s00535-014-0953-z
  46. 46. Korpela K, Flint HJ, Johnstone AM, et al. Gut microbiota signatures predict host and microbiota responses to dietary interventions in obese individuals. PLoS One. 2014;9(6):e90702. DOI: 10.1371/journal.pone.0090702
  47. 47. David LA, Maurice CF, Carmody RN, et al. Diet rapidly and reproducibly alters the human gut microbiome. Nature. 2013;505(7484):559-563. DOI: 10.1038/nature12820
  48. 48. Chan YK, Estaki M, Gibson DL. Clinical consequences of diet-induced dysbiosis. Annals of Nutrition and Metabolism. 2013;63(suppl 2):28-40. DOI: 10.1159/000354902
  49. 49. Fan W, Huo G, Li X, et al. Impact of diet in shaping gut microbiota revealed by a comparative study in infants during the six months of life. Journal of Microbiology and Biotechnology. 2014;24(2):133-143
  50. 50. Wu GD, Chen J, Hoffmann C, et al. Linking long-term dietary patterns with gut microbial enterotypes. Science. 2011;334:105-108. DOI: 10.1126/science. 1208344
  51. 51. Sanduleanu S, Jonkers D, De Bruine A, Hameeteman W, Stockbrügger RW. Non-Helicobacter pylori bacterial flora during acid-suppressive therapy: Differential findings in gastric juice and gastric mucosa. Alimentary Pharmacology & Therapeutics. 2001;15(3):379-388. DOI: 10.1046/j.1365-2036.2001.00888.x
  52. 52. Williams C, McColl KEL. Review article: Proton pump inhibitors and bacterial overgrowth. Alimentary Pharmacology & Therapeutics. 2006;23(1):3-10. DOI: 10.1111/j.1365-2036.2006.02707.x
  53. 53. Meuwissen SG, Craanen ME, Kuipers EJ. Gastric mucosal morphological consequences of acid suppression: A balanced view. Best practice & research. Clinical Gastroenterology. 2001;15(3):497-510. DOI: 10.1053/bega.2001.0189
  54. 54. Mason KL, Erb Downward JR, Falkowski NR, Young VB, Kao JY, Huffnagle GB. Interplay between the gastric bacterial microbiota and Candida albicans during postantibiotic recolonization and gastritis. Infection and Immunity. 2012;80(1):150-158. DOI: 10.1128/IAI.05162-11
  55. 55. Zhang X, Zhang J, Lin Y, Xu K, Li N, Chen H, et al. Analysis of the relationship between invasive capability of Helicobacter pylori and gastroduodenal diseases. Journal of Medical Microbiology. 2015 May;64(Pt 5):498-506. DOI: 10.1099/jmm.0.000049
  56. 56. Blaser MJ, Atherton JC. Helicobacter pylori persistence: Biology and disease. The Journal of Clinical Investigation. 2004;113(3):321-333. DOI: 10.1172/JCI20925
  57. 57. Correa P, Piazuelo MB. Helicobacter pylori infection and gastric adenocarcinoma. US Gastroenterology and Hepatology Review. 2011;7(1):59-64
  58. 58. Ford AC, Gurusamy KS, Delaney B, Forman D, Moayyedi P. Eradication therapy for peptic ulcer disease in Helicobacter pylori-positive people. Cochrane Database of Systematic Reviews. 2016;4:CD003840. DOI: 10.1002/14651858.CD003840.pub5
  59. 59. Engstrand L, Lindberg M. Helicobacter pylori and the gastric microbiota. Best Practice & Research. Clinical Gastroenterology. 2013;27:39-45. DOI: 10.1016/j.bpg.2013.03.016
  60. 60. Khosravi Y, Dieye Y, Poh BH, et al. Culturable bacterial microbiota of the stomach of Helicobacter pylori positive and negative gastric disease patients. The Scientific World Journal. 2014;2014:610421. DOI: 10.1155/2014/610421
  61. 61. Karimi P, Islami F, Anandasabapathy S, Freedman ND, Kamangar F. Gastric cancer: Descriptive epidemiology, risk factors, screening, and prevention. Cancer Epidemiology, Biomarkers & Prevention. 2014;23(5):700-713. DOI: 10.1158/1055-9965.EPI-13-1057
  62. 62. Forman D, Newell DG, Fullerton F, et al. Association between infection with Helicobacter pylori and risk of gastric cancer: Evidence from a prospective investigation. BMJ. 1991;302(6788):1302-1305
  63. 63. Nomura A, Stemmermann GN, Chyou PH, Kato I, Perez-Perez GI, Blaser MJ. Helicobacter pylori infection and gastric carcinoma among Japanese Americans in Hawaii. The New England Journal of Medicine. 1991;325(16):1132-1136. DOI: 10.1056/NEJM199110173251604
  64. 64. Hsu PI, Lai KH, Hsu PN, et al. Helicobacter pylori infection and the risk of gastric malignancy. The American Journal of Gastroenterology. 2007;102(4):725-730. DOI: 10.1111/j.1572-0241.2006.01109.x
  65. 65. Malfertheiner P, Megraud F, O’Morain CA, et al. Management of Helicobacter pylori infection--the Maastricht IV/ Florence consensus report. Gut. 2012;61(5):646-664. DOI: 10.1136/gutjnl-2012-302084
  66. 66. Sheu BS, Wu MS, Chiu CT, et al. Consensus on the clinical management, screening-to-treat, and surveillance of Helicobacter pylori infection to improve gastric cancer control on a nationwide scale. Helicobacter. 2017;22(3):e12368. DOI: 10.1111/hel.12368
  67. 67. Lee CW, Rickman B, Rogers AB, et al. Combination of sulindac and antimicrobial eradication of Helicobacter pylori prevents progression of gastric cancer in hypergastrinemic INS-GAS mice. Cancer Research. 2009;69(20):8166-8174. DOI: 10.1158/0008-5472.CAN-08-3856
  68. 68. Wang L, Zhou J, Xin Y, Geng C, Tian Z, Yu X, et al. Bacterial overgrowth and diversification of microbiota in gastric cancer. European Journal of Gastroenterology & Hepatology. 2016;28(3):261-266. DOI: 10.1097/MEG.0000000000000542
  69. 69. Eun CS, Kim BK, Han DS, Kim SY, Kim KM, Choi BY, et al. Differences in gastric mucosal microbiota profiling in patients with chronic gastritis, intestinal metaplasia, and gastric cancer using pyrosequencing methods. Helicobacter. 2014;19:407-416. DOI: 10.1111/hel.12145
  70. 70. Walker MM, Talley NJ. Review article: Bacteria and pathogenesis of disease in the upper gastrointestinal tract–beyond the era of Helicobacter pylori. Alimentary Pharmacology & Therapeutics. 2014;39(8):767-779. DOI: 10.1111/apt.12666
  71. 71. Alarcon T, Lorca L, Pereze-Perez G. Impact of microbiota and gastric disease development by Helicobacter pylori. Current Topics in Microbiology and Immunology. 2017;400:253-275. DOI: 10.1007/978-3-319-50520-6_11
  72. 72. Ianiro G, Molina-Infante J, Gasbarrini A. Gastric microbiota. Helicobacter. 2015;20(Suppl. 1):68-71. DOI: 10.1111/hel.12260
  73. 73. Giamarellos-Bourboulis E, Tang J, Pyleris E, Pistiki A, Barbatzas C, Brown J, et al. Molecular assessment of differences in the duodenal microbiome in subjects with irritable bowel syndrome. Scandinavian Journal of Gastroenterology. 2015;50(9):1076-1087. DOI: 10.3109/00365521.2015.1027261
  74. 74. Zhang Y, Hoffmeister M, Weck MN, Chang-Claude J, Brenner H. Helicobacter pylori infection and colorectal cancer risk: Evidence from a large population-based case-control study in Germany. American Journal of Epidemiology. 2012;175(5):441-450. DOI: 10.1093/aje/kwr331
  75. 75. Kim TJ, Kim ER, Chang DK, et al. Helicobacter pylori infection is an independent risk factor of early and advanced colorectal neoplasm. Helicobacter. 2017;22(3):e12377. DOI: 10.1111/hel.12377
  76. 76. Lin R, Zhou L, Zhang J, Wang B. Abnormal intestinal permeability and microbiota in patients with autoimmune hepatitis. International Journal of Clinical and Experimental Pathology. 2015;8(5):5153-5160
  77. 77. Franchini M, Vescovi PP, Garofano M, Veneri D. Helicobacter pylori-associated idiopathic thrombocytopenic purpura: A narrative review. Seminars in Thrombosis and Hemostasis. 2012;38(5):463-468. DOI: 10.1055/s-0032-1305781
  78. 78. Cardenas VM, Mulla ZD, Ortiz M, Graham DY. Iron deficiency and Helicobacter pylori infection in the United States. American Journal of Epidemiology. 2006;163(2):127-134. DOI: 10.1093/aje/kwj018
  79. 79. Nam SY, Park BJ, Ryu KH, Nam JH. Effect of Helicobacter pylori eradication on the regression of gastric polyps in National Cancer Screening Program. The Korean Journal of Internal Medicine. 2017;33(3):506-511. DOI: 10.3904/kjim.2016.286
  80. 80. Suwarnalata G, Tan AH, Isa H, et al. Augmentation of autoantibodies by Helicobacter pylori in Parkinson's disease patients may be linked to greater severity. PLoS One. 2016;11(4):e0153725. DOI: 10.1371/journal.pone.0153725
  81. 81. Wang F, Liu J, Lv Z. Association of Helicobacter pylori infection with diabetes mellitus and diabetic nephropathy: A meta-analysis of 39 studies involving more than 20,000 participants. Scandinavian Journal of Infectious Diseases. 2013;45(12):930-938. DOI: 10.3109/00365548.2013.844351

Written By

Hristo Ilianov Iliev, Mila Dimitrova Kovacheva-Slavova, Todor Asenov Angelov, Hristo Yankov Valkov, Ali Bedran and Borislav Georgiev Vladimirov

Submitted: 28 January 2019 Reviewed: 18 May 2019 Published: 10 August 2019