Open access peer-reviewed chapter

Helicobacter pylori Gastric Infection: Pathogenesis and Clinical Management

Written By

Neha Bisht and Amar P. Garg

Submitted: 30 June 2022 Reviewed: 26 July 2022 Published: 27 October 2022

DOI: 10.5772/intechopen.106783

Chapter metrics overview

446 Chapter Downloads

View Full Metrics

Abstract

Helicobacter pylori (H. pylori) is a Gram-negative bacterium that infects approximately 50% of the world population, and currently, no treatment is satisfactory for its management. Understanding the pathophysiology and pathogenesis mechanisms of H. pylori has increased over the years. Proper adherence and colonization of H. pylori induce genetic alterations, express numerous virulence factors, and trigger diverse adaptive mechanisms, making possible the colonization of an organ with a highly acidic lumen. The mode for the transmission of infection can be oral-oral or fecal-oral. Various effector proteins or toxins are released by the organism for successful colonization and infection. For the virulence and pathogenicity of H. pylori, the virulence factors, host, and environmental factors interplay a very important role. Virulence factors for H. pylori enhanced the pathogenicity of cytotoxin-associated antigen A, vacuolating cytotoxin, duodenal ulcer promoting gene A protein, outer inflammatory proteins, and gamma-glutamyl transpeptidase. The host immune system through Th1-polarized response plays a crucial role in the course of infection. The most common symptoms in H. pylori-positive individuals are peptic ulcers, gastric adenocarcinomas, and mucosa-associated lymphoid tissue lymphomas, whereas some positive individuals remain asymptomatic. Detection of H. pylori infection can be through invasive and noninvasive diagnostic methods. We critically reflect on the infection of H. pylori and the virulence and pathogenesis mechanisms of H. pylori.

Keywords

  • pathogenesis
  • virulence factor
  • colonization
  • transmission
  • diagnosis

1. Introduction

Helicobacter pylori (H. pylori) is a gram-negative, motile, helical, and micro-aerophilic (5% O2, 15% CO2 & 80% N, required low concentration of O2 than in the atmosphere) microorganism that is considered as one of the most successful pathogens due to its persistent infection in the human stomach, that inhabits the gastric environment of more than half of the world population of 4.4 billion people worldwide [1, 2, 3]. Various shreds of evidence suggest that H. pylori are the etiological agent of both extra-gastric and gastric diseases (gastric malignancy, peptic ulcer, chronic gastritis) [4, 5, 6]. The main reason for the occurrence of infection among the members of the same family (parents and children) is the route of transmission of H. pylori through oral–oral transmission. In this way, during feeding sharing utensils seems to be important for the establishment of infection [7]. Another route for the transmission of infection is fecal-oral, due to the ingestion of contaminated water and unsatisfactory basic sanitation conditions [8]. Improvement of living conditions and socioeconomic status are the two factors that greatly influence the reduction in H. pylori infection [9]. It was believed that the gastric environment was sterile because of its high acidity until the discovery of H. pylori infection from gastric mucosa by Warren and Marshall’s [10, 11]. The bacteria use various mechanisms that provide mobility improvement, robust adherence to epithelial cells, and an enzymatic apparatus that allows the establishment of an appropriate microenvironment for the perpetuation of infection [12, 13, 14, 15]. Certain virulence factors are responsible for the potential pathogenicity of infection such as vacuolating cytotoxin (VacA), cytotoxin-associated antigen A (CagA), duodenal ulcer promoting gene A protein (DupA), outer inflammatory protein (OipA) and gamma-glutamyl transpeptidase (GGT) and genetic substrates of the host [e.g., IL1B gene cluster and tumor necrosis factor-α (TNFα) gene polymorphism] play an important role [16, 17, 18, 19, 20] shown in Figure 1. During the course of infection host immune system play a very pivotal role shown in Figure 1. Flagella-mediated motility is used by H. pylori for movement toward the epithelial cells of stomachs and penetrating the mucus lining [21]. With all the suitable conditions, organism crosses the acidic environment. At the mucus layer, the coccoid form enabled its colonization [22, 23]. Attachment to the host epithelial cells is through the production of an adhesin [21, 24]. So far colonization of H. pylori could be negatively and positively associated with the induction and progression of several diseases [23, 25, 26]. It has been reported to be linked to gastric and duodenal ulcers, gastric carcinoma, and gastric mucosa-associated lymphoid tissue (MALT) lymphoma [62728]. Several other studies are also directing a positive correlation between gastrointestinal diseases and H. pylori. Many research shows a positive association between H. pylori, duodenal ulcer and gastric ulcer [29], gastritis [30], and esophageal cancer [31]. Moreover, evidence shows a positive association between H. pylori and non-gastrointestinal diseases such as diabetes mellitus [32], coronary artery disease [33], and anemia [34].

Figure 1.

Representation of characteristics of H. pylori infection.

Advertisement

2. Clinical manifestations and diagnosis

The discovery of H. pylori in the early 1980s caused peptic ulcer diseases [35]. By International Agency for Research on Cancer in 1994 H. pylori was first classified as a carcinogen [36]. In 2001, an epidemiological study stated that patients with H. pylori were nearly six times more potent to develop gastric cancer compared with uninfected people [36]. In 2015, a community of gastroenterologists meticulously recognized H. pylori gastritis as an infectious disease and recommended its eradication [37]. This recommendation has repeatedly been confirmed by more recent consensus statements [35, 38, 39]. The most recent consensus conferences on the proactive testing and eradication of H. pylori infection [35, 38, 39]. Patients with uninvestigated dyspepsia, having a history of past or current gastric or duodenal ulcer, with a diagnosis of gastric mucosa-associated lymphoid tissue lymphoma, having a family history with peptic ulcer or gastric cancer, and a person with constant use of non-steroidal anti-inflammatory drugs should be recommended for H. pylori testing [35, 38]. The strengths and weaknesses of endoscopic and noninvasive tests are shown in Table 1.

TestsMechanismStrengthsWeaknesses
Endoscopic
CultureCan be kept in a transport medium, like Portagerm pylori or Stuart’s transport medium up to 24 h at 4°C. The commonly used media include Pylori agar, Skirrow agar, Columbia blood agar, Brucella agar, Brain heart infusion or Trypticase soy agar, supplemented with sheep or horse blood.Allows testing of antibiotic susceptibilitiesPoor availability in some countries
Molecular-basedSeveral target genes like UreA, glmM, UreC, 16S rRNA, 23S rRNA, HSP60, and VacA genes detected for the detection of H. pyloriDetects infection and can assess susceptibility/resistance for all six commonly used antibiotics. Stool can be used. It gives Rapid results (days)not to be covered by insurance
HistologyAccuracy of histology, such as the site, size and number of biopsies, the staining methods, proton pump inhibitor (PPI), antibiotics of the examining pathologist.Can be used for the testing of infection and evaluate for eradication. Provides additional information such as degree of inflammation and associated pathologyAccurate results require interested pathologist and use of special stain, preferably immunohistochemical
Rapid ureaseConvert the urea test reagent to ammonia, resulting in increase of pH and color change during pH monitorIt is a Rapid, Inexpensive. Good sensitivity and specificityRequires prior cessation of antibiotics, bismuth. products, or proton pump inhibitors to reduce risk of false negative results
Noninvasive
SerologyDetection of Specific anti-H. pylori IgG antibodies in the patient's serumWidely available, Least expensive. Does not require medication modifications prior to testingDoes not reliably delineate between active and previous infection. Cannot confirm eradication
Stool antigenassesses the presence of bacterial antigens in stool.High sensitivity and specificity. Can be used to test for active infection and evaluate for eradicationStool sample needed, patient aversion. Requires prior cessation of antibiotics, bismuth products, or proton pump inhibitors to reduce risk of false negative results
Urea breathEnzyme splits urea into ammonia and CO2Patients ingest urea labeled with either 13C or 14C, and the labeled urea comes into contact with the mucosa and diffuses through the
mucus towards the H. pylori and the mucosal blood supply
High sensitivity and specificity
Can be used to test for active infection and evaluate for eradication
Resources and trained personnel needed to reliably reproduce test
Requires prior cessation of antibiotics, bismuth products, or proton pump inhibitors to reduce risk of false negative results

Table 1.

Strengths and weakness of endoscopic and noninvasive tests for the detection of H. pylori infection.

First-generation immigrants from high-prevalence countries and potential high-risk populations were targeted for testing. Once the presence of the infection has been diagnosed and documented outreach to family members is suggested because transmission from person to person occurs within families [40]. Testing and treating of H. pylori-infected individuals can protect other members from infection as well as re-infection from its related diseases [41]. This approach may engage those who test positive to concur with the eradication treatment [41, 42]. To detect H. pylori infection a wide variety of methods is available [43]. Because the organism is trophic for gastric epithelium, H. pylori are primarily found in the stomach which causes an acute-on-chronic inflammation [44]. Using special stains, organisms can be detected the most accurate and popularly used is immunohistochemistry with H. pylori-specific antibodies [45, 46]. Wide range of other tests such as serologic tests for anti–H. pylori IgG antibodies and by using molecular testing such as next-generation sequencing [47]. Other tests are noninvasive while some require endoscopy to sample gastric contents, amongst them noninvasive testing is more preferred [48, 49].

The diagnostic strategy utilized clinical indication as well as the local availability and costs of the different tests followed by patient preferences [35]. The presence of the infection gives rise to a serum immune response. Recently, the most commonly used diagnostic test was serology [38]. By Medicare serology is generally neither recommended currently. Tests for the detection of H. pylori infection are shown in Table 1. IgA and IgM anti–H. pylori tests available in some laboratories are not recommended or trusted because of their low specificity and sensitivity generally not approved by the US Food and Drug Administration (FDA). Another important diagnostic method for H. pylori is the rapid urease test (RUT). RUT detects an increase in pH of the reagent after the addition of a biopsy specimen that contains H. pylori to the reagent [35, 38]. The sort of pH variation is caused by the transformation of the urea test reagent into ammonia. As compared to other tests, RUT is quick, cheap, easy, and specific and is a widely available test [50]. The urea breadth test (UBT) and the stool antigen test (SAT) are the main non-invasive tests for the diagnosis of active infection [51, 52]. UBT is based on the mechanism of degradation of 13C or 14C-labeled urea into CO2 that can be measured by an infrared spectrometer [3852]. The false positive tests are commonly associated with serology but can occur in the UBT. For example presence of achlorhydria promotes excessive growth of non–H. pylori organisms that produce urease result in false positive UBTs. The apparent failure of repeated treatment is the main cause of false positive tests [3850, 51]. The apparent failure of repeated treatment results in False positive tests. When false positive tests are suspected, a less expensive option for UBT is with a stool antigen test or endoscopy for the diagnosis of H. pylori [51, 53]. For the diagnosis of H. pylori infection urine test has been a new promising non-invasive largely recommended as an alternative [51, 54].

A meta-analysis reported that testing for antibodies from urine samples might be a good diagnostic option [54, 55]. For the confirmation of the accuracy of this method, further studies are necessary. Through the discovery of specific serological markers for the diagnosis of H. pylori, the infection has been developed. A recent study confirmed the accuracy of the “hook-associated protein 2 homologs”, FliD, as a marker of the diagnosis of H. pylori infection [54]. The use of Flid ELISA method for the detection of H. pylori infection provides up to 99% high specificity and 97% of sensibility at a low cost [35, 41, 51, 56].

Advertisement

3. Pathogenesis

3.1 Colonization

A special mechanism is required by H. pylori for successful colonization in the gastric environment. After entering the gastric environment H. pylori uses crucial flagellar motility for swimming in gastric content which allows the bacteria to get into the gastric mucus layer [38, 40, 51]. Flagella movements are differ in different media “spreading” in solid media and “swarming” movements, or “swimming motility” in liquid media [57]. Various mutations in genes that encode specific flagellar proteins such as fliD, FlaA, and FlaB disrupts the motility of H. pylori resulting in reduce colonization in the gastric mucosal layer [58]. Apart from flagella, the mobility of H. pylori lies in the chemotaxis actions of various molecules like mucin, urea, sodium bicarbonate, sodium chloride, and specific amino acids [59]. In recent years various colonization factors like urease, GGT, Flolitin-like protein (FLOT), and RhpA [60]. Several H. pylori chemoreceptors like T1pA, B, C, and D, CheA kinase, and various coupling proteins are all important for bacterium colonization [61].

Apart from them, various transition metals are crucial for living organisms, as they serve as cofactors for enzymatic reactions that enhanced the rate of reaction which carries out genetic material replication, transcription, attenuation of oxidative stress, and cellular energy production [54]. In bacteria for survival and successful infection, these metals are crucial [62]. Nickel is an essential metal for H. pylori and cofactors for two important enzymes like urease and hydrogenase, both enzymes play a vital role in the process of infection [14]. H. pylori urease catalyzes the hydrolysis of urea to ammonia and carbon dioxide which acts as a buffer medium that diminished the acidity of the stomach [63, 64]. Hydrogenase is a signaling cascade that allows Hydrogen as a source of energy for metabolism to be used by H. pylori [65]. In the interaction between bacteria and host Adhesion molecules and surface receptors of gastric cells play a very important role [1465]. The well-characterized molecule is blood group antigen binding adhesin A (BabA) that carries specific binding to b and H-1 Lewis antigens from the surface of epithelial cells of gastric [66, 67]. The adhesion of the outer membrane Hp HopQ depicts the bacterial-host interaction. These adhesins bind to the cell adhesion molecules related to the carcinoembryonic antigen 1,3,5 and 6 (CEACAMs which give rise to cell signaling mediated by the HopQ-CEACAM interaction that allows the translocation of CagA which is the most crucial and virulence factor of H. pylori that increase pro-inflammatory mediators [63, 64, 68, 69, 70]. 64 outer membrane proteins (OMPs) of H. pylori are grouped into five gene families. Hop and hor genes are part of family 1, which encodes BabA/B/C, SabA/B, and AlpA/B [64]. In the virulence of H. pylori, various OMPs have been identified [64].

3.2 Cytotoxin-associated gene product (CagA)

Cytotoxin-associated gene product (CagA) is one of the most important and studied virulence factors of H. pylori. Expression of cagA is always induced whenever the pathogen adhered to the epithelial cells [71]. The cag-pathogenicity island (cagPAI) is approximately 40kb that encodes CagA [72], type (IV) secretion system (T4SS) [73], and oncoproteins [74]. CagPA1 is a 40kb chromosomal DNA region of which 31 genes form a type IV secretion system (T4SS) [65, 73]. Seven different secretion systems from I-VII are present, amongst them penetration in the plasma membrane and delivery of bacterial molecules directly to the cytoplasm of the target cells is done by Type III and IV. Flagellum-like tube to translocate effector proteins into eukaryotic host cells performed by type III secretion system (T3SS) whereas a pilus-based structure to mediate the delivery of DNA or proteins into target cells employed by type IV secretion system (T4SS). Cytotoxic effect on the host cell, induced by oncoprotein that is injected into the cell via a pilus formed by the T4SS [74, 75]. This results in the induction of cellular alterations like cellular proliferation, impairing cell motility, apoptosis, and change in the pattern of the cytoskeleton [76].

Transport of CagA protein from gastric mucosal surface to endothelial cells for the tyrosine phosphorylation, carried out by T4SS which directly induces an immune response [75, 77]. The specific interaction between the H. pylori HopQ adhesin and the CEACAM (a cellular adhesion molecule) is used for translocating CagA into gastric epithelial cells [77]. Some studies showed this interaction using CEACAM-humanized (hCEACAM) mouse PMNs and humans resulting that H. pylori Hop Q-dependent interaction greatly eases the translocation and phosphorylation of CagA. The PMNs positively enhanced the expression of pro-inflammatory chemokines MIP-1-alpha whereas chronic mouse model infection showed downregulation of hCEACAM and -6 receptors on neutrophils [38, 78, 79]. Once CagA, the effector protein of H. pylori outreach the host cell it interacts with a diversity of SH2 domains where tyrosine phosphorylation took place (Table 2). Site of phosphorylation in H. pylori coerce with Glu-Pro-Ile-Tyr-Ala (EPIYA) sequence simultaneously or with closely related sequence located at the N-terminal region and a C-terminal tail [87]. The classification of different subtypes of CagA such as CagA-AB, CagA-ABC, CagA-ABD, or CagA-BD totally based on the composition of the EPIYA-A, EPIYA-B, EPIYA-C, and EPIYA-D motifs [89]. Binding of the host SH2 domain with different EPIYA segments also differs, Among all, the D segment is more harmonious than the C segment and releases the maximum amount of IL-8. [77, 89]. The cytotoxin-associated gene A (cagA)-positive and cagA-negative are the two subclasses of H. pylori [88]. The cagA-positive is more motile as compared to cagA negative [88]. A more severe clinical outcome of H. pylori infection, associated with mucosal inflammation is due to the presence of two segments cag I and cag II [87]. The level of expression of cagA differs in H. pylori strains. Yeh et al, 2019 demonstrate that the H. pylori strains which possess Y58/E59 polymorphism in the cagL are at higher risk of facilitating gastric cancer [87, 88]. Mutations in genes present in the cagPAI totally influence virulence [87].

Virulence factorsFunctionsReferences
ArginasePrevents bacterial killing & T-cell proliferation, Stimulate apoptosis, Impairing of immune responses, Help the H. pylori to withstand the acidic environment[80]
Heat shock proteinsEnhancing adherence to epithelial surfaces, engaged in urease activation, apoptosis and autophagy Control, To maintain the structure and properties of the effector proteins, Protection of cell from reactive oxygen species (ROS),Promotes the production and release of IL-8, TNF-α, and COX-2[81, 82]
Superoxide dismutase (SOD)Protection of cell from reactive oxygen species (ROS), colonization enhancement, Inhibiting the production of cytokines, Stimulating the activation of macrophage[83]
γ-glutamyl-transferaseFacilitating the apoptosis and necrosis, Inducing the release of pro-inflammatory proteins and release of ROS, Stimulate DNA damage[84]
LipopolysaccharideTriggering of signaling pathways, Induction of several inflammatory responses, Induce immune responses, Disrupts the mucus secretion[85]
Cholesteryl α-glucosyltransferase (αCgT)Shield H. pylori from immunological attack, Stimulates the production of pro-inflammatory proteins (e.g. IL-8),Enhancing the bacterial growth and resistance to antibiotics[86]
PhospholipaseActivating the signaling pathways (e.g. ERK1/2)and Trigger chronic inflammation, Enhancing the bacterial colonization, Involved in the degradation of lipids and damage to mucus layer[87, 88]

Table 2.

Important virulence factors in H. pylori.

3.2.1 Non-CagA virulence factors

Impairing gastric homeostasis is the main capacity by other various virulence factors. Vacuolating cytotoxins (VacA) protein and its genes are presents in almost all the strains, promotes pathogenicity of H. pylori (Table 2). Vacuolation is known to be the creation of vacuoles on a host cell which promotes its pathogenicity by inducing cytotoxicity and apoptosis [90]. It promotes endocytic trafficking disruption, perturbations in mitochondrial, depolarization in plasma membrane potential and various ions like chloride, bicarbonate and urea efflux, and results in the activation of MAP kinases [91, 92]. The activation and suppression of the immune response is prompted by VacA that induce immune tolerance through T cells and antigen-presenting cells. Moreover, this protein also enhances the activation and suppression of the immune response resulting in immune tolerance and persistent infection through its activities on T-cells and antigen-presenting cells [93]. The 140 kDa precursor is produced by VacA which undergoes proteolytic process to produce a toxin of 88 kDa in mass [90]. VacA bind to the epithelial cells with the help of receptor-like protein tyrosine phosphatase alpha and beta (RPTP-α, RPTP-β), sphingomyelin, and density lipoprotein receptor-related protein-1 (LRP-1) [90, 94] also binds to β2 integrin (CD18) receptors [90]. The VacA toxins in amino-terminal is 33KDa and carboxy-terminal 55 kDa that transport into extracellular space by type V secretion pathway, subsequently internalized into endosomal compartments [90, 93, 94]. Several clinical outcomes are due to the different polymorphic forms [94].

The virulence factor promotes gastritis, ulcer, and prolonged infection lead to the cancer development [17, 92]. Duodenal ulcer-promoting gene (DupA) gives a higher acid resistance to the bacterium which promote increase in the production of IL-8 in the gastric mucosa [95]. DupA belongs to the T4SS which integrate conjugative element (ICEHptfs4). VirB4 ATPase homolog is encoded by dupA ,linked with gastric ulceration [90, 93] .Pro-inflammatory cytokine secreted by mononuclear cells are induced by dupA [96]. The secretions of IL-8 and IL-12 are induced by DupA in the gastric mucosa by gastric epithelial cells in vitro [97]. The biomarkers for peptic ulcers disease is due to dupA [98]. Recently a study didn’t show any positive correlation between dupA and peptic ulcer disease [99]. The increased development of peptic ulcers and gastric cancer totally correlated with the discovery of OipA [96]. OipA is one of the important factor in the outcome of the infection is regulated by “slipped strand mispairing”, that depends on the quantity of CT dinucleotide repeats in the5’ region of oipA [100]. This process determines the functionality and nonfunctionality of OipA that enhance the gastric pathogenicity [100]. OipA change the signaling of β-catenin, proliferation of the cell and decreased the cell-cell junctions [100, 101]. H. pylori produce the enzyme GGT which catalyzes the conversion of glutamine into glutamate and finally to ammonia; glutathione into glutamate and cysteinylglycine [102]. It lead to the production of reactive oxygen species (ROS) that induce apoptosis and necrosis [20, 21]. The enzyme inhibits T cell proliferation and dendritic cell differentiation [25, 51, 52]. Patients with peptic ulcers have high GGT activity as compared to the other gastroduodenal disease [90, 96]. Other important virulence factors in H. pylori shown in Table 2.

3.3 Treatment

No universal regimen for the treatment of H. pylori infection. Mostly target the regressing symptomatology and mucosa healing [102]. Since 1997 Maarstricht consensus, proton pump inhibitors (PPI) as standard triple therapy by using a standard dose of 500 mg of clarithromycin and 1g of amoxicillin twice for 7 days as a first-line regimen for the eradication of H. pylori. Triple therapy is still recommended as the first line of treatment for H. pylori infection in areas with a low rate of clarithromycin according to the European Helicobacter and Microbiota Study Group (EHMSG) [49]. A high dose of clarithromycin resistance lowers the rate for eradication rate of clarithromycin-containing triple therapy like 75% cure rate in Argentina is estimated [103]. A recent study showed that in type 2 diabetic patients the rate of infection eradication is up to 74% [103]. The PPIs, bismuth salt, tetracycline, and metronidazole are complex regimens of Bismuth quadruple therapy, recommended as the second-line or even first-line [104]. According to some previous studies it is stated that the multicenter randomized controlled trials (RCTs) rate of curing of bismuth quadruple therapy is notably high in the standard triple therapy from 90.4% to 83.7% for 14 days [103, 104]. In a meta-analysis study, nine RCTs were evaluated and the eradication of infection by bismuth quadruple therapy was the same as those who had received clarithromycin triple therapy [104, 105]. The bismuth quadruple and Levofloxacin triple therapy are the two best therapeutic against infection of H. pylori [103]. Levofloxacin regime contains PPIs plus levofloxacin and amoxicillin [104]. Eradication rate of levofloxacin triple therapy (74.5%) and bismuth quadruple therapy (78%) [102105]. The third-line therapy is prescribed by using antibiotic susceptibility testing (AST) and prescribed only when the first and second lines of treatment failed [70]. Therapeutic protocols like bismuth-based levofloxacin quadruple therapy or rifabutin triple therapy are used [106]. All three treatment lines are summarized in Figure 2.

Figure 2.

Treatment regimens of H. pylori infection.

Probiotics are ―live microorganisms that, when administered in adequate amounts, confer health benefits on the host and they favorably alter the balance in intestinal microflora [107]. Probiotics prevent the adhesion of pathogens by competing for the binding site on intestinal epithelial cells, reducing the colonization of pathogens and thereby preventing the onset of infection. The probiotic's clinical benefits are widely accepted such as diarrhea, antibiotic-associated diarrhea, functional digestive involvements, inflammatory bowel disease, cardiovascular diseases, allergic reactions, and cancer [104, 107]. Anti -H. pylori properties proved by spp. of Lactobacillus [108]. Lactobacillus strains prevent the colonization of H. pylori due to their specific adhesins [109] shown in Figure 3. The adjuvant therapies with probiotics cure infections caused by H. pylori according to the European Helicobacter Pylori Study Group (EHPSG) [104, 105]. Lactobacillus spp. and Bifidobacterium spp are used more in clinical trials than other probiotics [110]. Studies showed that the yogurt containing bacteria could improve the eradication rate of H. pylori infection in the fifth week of treatment [107, 110]. Probiotics act as antibiotic-producing bacteria as they produce antimicrobial substances that inhibit the growth of H. pylori [87]. Probiotics' short-chain fatty acids products such as acetic acid, propionic acid, and lactic acid lower the pH of the environment totally unfavorable conditions for the survival of H. pylori [111]. Bacteriocins produced by probiotics showed antagonistic activity against H. pylori [112]. Lactobacillus acidophilus significantly reduced the viability of H. pylori [112]. Inhibition of H. pylori is due to the antimicrobial Nisin A [113]. The heterogeneous group of antimicrobial proteins is mostly produced by all LAB lactic acid bacteria [114]. Inducing pores in the membrane, activating autolytic enzymes, and downregulating the expression of vacA, cagA, luxS, and flaA genes are several mechanisms by bacteriocin to inhibit the growth of H. pylori [112, 114].

Figure 3.

Mechanism of probiotics against H. pylori infection.

References

  1. 1. Alzahrani S, Lina TT, Gonzalez J, Pinchuk IV, Beswick EJ, Reyes VE. Effect of Helicobacter pylori on gastric epithelial cells. World Journal of Gastroenterology. 2014;20:12767-12780
  2. 2. Karbalaei M, Khorshidi M, Sisakht-pour B, Ghazvini K, Farsiani H, Youssefi M, et al. What are the effects of IL-1β (rs1143634), IL-17A promoter (rs2275913) and TLR4 (rs4986790) gene polymorphism on the outcomes of infection with H. pylori within as Iranian population. Gene Report. 2020;20:100735
  3. 3. Hooi JK, Lai WY, Ng WK, Suen MM, Underwood FE, Tanyingoh D, et al. Global prevalence of Helicobacter pylori infection: Systematic review and meta-analysis. Gastroenterology. 2017;153(2):420-429
  4. 4. Keikha M. Is there a relationship between Helicobacter pylori vacA i1 or i2 alleles and development into peptic ulcer and gastric cancer? A meta-analysis study on an Iranian population. New Microbes New Infection. 2020;36:100726
  5. 5. Youssefi M, Tafaghodi M, Farsiani H, Ghazvini K, Keikha M. Helicobacter pylori infection and autoimmune diseases; Is there an association with systemic lupus erythematosus, rheumatoid arthritis, autoimmune atrophy gastritis and autoimmune pancreatitis? A systematic review and meta-analysis study. Journal of Microbiology, Immunology, and Infection. 2021;54(3):359-369
  6. 6. Gravina AG, Zagari RM, De Musis C, Romano L, Loguercio C, Romano M. Helicobacter pylori and extragastric diseases: A review. World Journal of Gastroenterology. 2018;24(29):3204
  7. 7. Urita Y, Watanabe T, Kawagoe N, Takemoto I, Tanaka H, Kijima S, et al. Role of infected grandmothers in transmission of Helicobacter pylori to children in a Japanese rural town. Pediatrician Child Health. 2013;49:394
  8. 8. Goh KL, Chan WK, Shiota S, Yamaoka Y. Epidemiology of Helicobacter pylori infection and public health implications. Helicobacter. 2011;16:1
  9. 9. Laszewicz W, Iwańczak F, Iwańczak B, Task Force of the Polish Society of Gastroenterology; Task Force of the Polish Society of Gastroenterology. Seroprevalence of Helicobacter pylori infection in Polish children and adults depending on socioeconomic status and living conditions. Advanced Medical Science. 2014;59:147
  10. 10. Warren JR, Marshall B. Unidentified curved bacilli on gastric epithelium in active chronic gastritis. Lancet. 1983;1:1273-1275
  11. 11. Wroblewski LE, Peek RM. Helicobacter pylori, cancer, and the gastric microbiota. Advanced Experiment in Medical Biology. 2016;908:393-408
  12. 12. Eaton KA, Morgan DR, Krakowka S. Motility as a factor in the colonisation of gnotobiotic piglets by Helicobacter pylori. Journal of Medical Microbiology. 1992;37:123-127
  13. 13. Alm RA, Bina J, Andrews BM, Doig P, Hancock RE, Trust TJ. Comparative genomics of Helicobacter pylori: Analysis of the outer membrane protein families. Infectious Immunity. 2000;68:4155-4168
  14. 14. Camilo V, Sugiyama T, Touati E. Pathogenesis of Helicobacter pylori infection. Helicobacter. 2017;22:1
  15. 15. Fischer W. Assembly and molecular mode of action of the Helicobacter pylori Cag type IV secretion apparatus. FEBS Journal. 2011;278:1203-1212
  16. 16. El-Omar EM, Carrington M, Chow WH, McColl KE, Bream JH, Young HA, et al. Interleukin-1 polymorphisms associated with increased risk of gastric cancer. Nature. 2000;404:398-402
  17. 17. Boquet P, Ricci V. Intoxication strategy of Helicobacter pylori VacA toxin. Trends in Microbiology. 2012;20:165-174
  18. 18. Lu H, Hsu PI, Graham DY, Yamaoka Y. Duodenal ulcer promoting gene of Helicobacter pylori. Gastroenterology. 2005;128:833-848
  19. 19. Yamaoka Y, Kwon DH, Graham DY. A M(r) 34,000 proinflammatory outer membrane protein (oipA) of Helicobacter pylori. Proceedings of the National Academy Science USA. 2000;97:7533-7538
  20. 20. Shibayama K, Wachino J, Arakawa Y, Saidijam M, Rutherford NG, Henderson PJ. Metabolism of glutamine and glutathione via gamma-glutamyltranspeptidase and glutamate transport in Helicobacter pylori: Possible significance in the pathophysiology of the organism. Molecular Microbiology. 2007;64:396-406
  21. 21. Kao C-Y, Sheu B-S, Wu J-J. Helicobacter pylori infection: An overview of bacterial virulence factors and pathogenesis. Biomedical Journal. 2016;39(1):14-23
  22. 22. Saxena A, Mukhopadhyar AK, Nandi SP. Helicobacter pylori: Perturbation and restoration of gut microbiome. Journal of Biosciences. 2020;45(1):1-15
  23. 23. Reshetnyak VI, Burnistrov AI, Maev IV. Helicobacter pylori: Commensals, symbiont or pathogens? World Journal of Gastroenterology. 2021;27(7):545
  24. 24. Huang Y, Wang Q-L, Cheng D-d. Adhesin and invasion of gastric mucosa epithelial cells by Helicobacter pylori. Frontiers in Cellular and Infection Microbiology. 2016;6:159
  25. 25. Alexander SM, Retnakumar RJ, Chouhan D, Devi TNB, Dharmaseelan S, Devadas K, et al. Helicobacter pylori in human stomach: The inconsistencies in clinical outcomes and the probable causes. Frontiers in Microbiology. 2021;12:713955
  26. 26. Goni E, Franceschi F. Helicobacter pylori and extragastric diseases. Helicobacter. 2016;1:45-48
  27. 27. Crowe SE. Helicobacter pylori infection. The New England Journal of Medicine. 2019;380:1158-1165
  28. 28. McColl KE. Helicobacter pylori infection. The New England Journal of Medicine. 2010;362(17):1597-1604
  29. 29. Narayanan M, Reddy KM, Marsicano E. Peptic ulcer disease and Helicobacter pylori infection. Missouri Medicine. 2018;115:219
  30. 30. Watari J, Chen N, Amenta PS, Fukui H, Oshima T, Tomita T, et al. Helicobacter pylori associated chronic gastritis, clinical syndromes, precancerous lesions, and pathogenesis of gastric cancer development. World Journal of Gastroenterology: WJG. 2014;20:5461
  31. 31. Polyzos SA, Zeglinas C, Artemaki F, Doulberis M, Kazakos E, Katsinelos P, et al. Helicobacter pylori infection and esophageal adenocarcinoma: A review and a personal view. Annals of Gastroenterology. 2018;31:8
  32. 32. Chen TP, Hung HF, Chen MK, Lai HH, Hsu WF, Huang KC, et al. Helicobacter pylori infection is positively associated with metabolic syndrome in Taiwanese adults: A cross-sectional study. Helicobacter. 2015;20:184-191
  33. 33. Yu XJ, Yang X, Feng L, Wang LL, Dong QJ. Association between Helicobacter pylori infection and angiographically demonstrated coronary artery disease: A meta-analysis. Experimental and Therapeutic Medicine. 2017;13:787-793
  34. 34. Sato Y, Yoneyama O, Azumaya M, Takeuchi M, Sasaki SY, Yokoyama J, et al. The relationship between iron deficiency in patients with Helicobacter pylori-infected nodular gastritis and the serum prohepcidin level. Helicobacter. 2015;20:11-18
  35. 35. Lee YC, Dore MP, Graham D. Diagnosis and Treatment of Helicobacter pylori Infection. Annual Review of Medicine. 2022;73:183-195
  36. 36. Aziz F, Chakraborty A, Liu K, Zhang T, Li X, Du R, et al. Gastric tumorigenesis induced by combining Helicobacter pylori infection and chronic alcohol through IL-10 inhibition. Carcinogenesis. 2022;43:126-139
  37. 37. Sugano K, Tack J, Kuipers EJ, et al. Kyoto global consensus report on Helicobacter pylori gastritis. Gut. 2015;64:1353-1367
  38. 38. El-Serag HB, Kao JY, Kanwal F, et al. Houston Consensus Conference on testing for Helicobacter pylori infection in the United States. Clinical Gastroenterology and Hepatology. 2018;16:992-1002
  39. 39. Liou JM, Malfertheiner P, Lee YC, et al. Screening and eradication of Helicobacter pylori for gastric cancer prevention: The Taipei global consensus. Gut. 2020;69:2093-2112
  40. 40. Bair M-J, Chuang S-L, Lei W-Y, et al. Planning mass eradication of Helicobacter pylori infection for indigenous Taiwanese peoples to reduce gastric cancer. Journal of Gastroenterology and Hepatology. 2020;35:609-616
  41. 41. Ding S-Z. Global whole family based-Helicobacter pylori eradication strategy to prevent its related diseases and gastric cance. World Journal of Gastroenterologia. 2020;26:995-1004
  42. 42. Zhao J-B, Yuan L, Yu X-C, et al. Whole family–based Helicobacter pylori eradication is a superior strategy to single-infected patient treatment approach: A systematic review and meta-analysis. Helicobacter. 2021;26:e12793
  43. 43. Dore MP, Pes GM. What is new in Helicobacter pylori diagnosis. An overview. Journal of Clinical Medicine. 2021;10:2091
  44. 44. Dixon MF, Genta RM, Yardley JH, Correa P. Classification and grading of gastritis. The updated Sydney System. International Workshop on the Histopathology of Gastritis, Houston 1994. The American Journal of Surgical Pathology. 1996;20:1161-1181
  45. 45. Glickman JN, Noffsinger A, Nevin DT, et al. Helicobacter infections with rare bacteria or minimal gastritis: Expecting the unexpected. Digestive and Liver Disease. 2015;47:549-555
  46. 46. Lash RH, Genta RM. Routine anti-Helicobacter immunohistochemical staining is significantly superior to reflex staining protocols for the detection of Helicobacter in gastric biopsy specimens. Helicobacter. 2016;21:581-585
  47. 47. Pohl D, Keller PM, Bordier V, Wagner K. Review of current diagnostic methods and advances in Helicobacter pylori diagnostics in the era of next generation sequencing. World Journal of Gastroenterologia. 2019;25:4629-4660
  48. 48. Talley NJ. American Gastroenterological Associationmedical position statement: Evaluation of dyspepsia. Gastroenterology. 2005;129:1753-1755
  49. 49. Malfertheiner P, Megraud F, O’Morain CA, et al. Management of Helicobacter pylori infection—the Maastricht V/Florence Consensus Report. Gut. 2017;66:6-30
  50. 50. Wang YK, Kuo FC, Liu CJ, Wu MC, Shih HY, Wang SS, et al. Diagnosis of Helicobacter pylori infection: Current options and developments. World Journal of Gastroenterology. 2015;21:11221-11235
  51. 51. Keller J, Hammer HF, Afolabi PR, et al. European guideline on indications, performance and clinical impact of 13C-breath tests in adult and pediatric patients: An EAGEN, ESNM, and ESPGHAN consensus, supported by EPC. United European Gastroenterology Journal. 2021;9:598-625
  52. 52. Best LM, Takwoingi Y, Siddique S, et al. Non-invasive diagnostic tests for Helicobacter pylori infection. Cochrane Database System Review. 2018;3:CD012080
  53. 53. Shimoyama T. Stool antigen tests for the management of Helicobacter pylori infection. World Journal of Gastroenterology. 2013;19:8188-8191
  54. 54. de Brito BB, da Silva FAF, Soares AS, Pereira VA, Santos MLC, Sampaio MM, et al. Pathogenesis and clinical management of Helicobacter pylori gastric infection. World Journal of Gastroenterology. 2019;25(37):5578-5589
  55. 55. Gong Y, Li Q, Yuan Y. Accuracy of testing for anti-Helicobacter pylori IgG in urine for H. pylori infection diagnosis: A systematic review and meta-analysis. BMJ Open. 2017;7:e013248
  56. 56. Khalifeh Gholi M, Kalali B, Formichella L, Göttner G, Shamsipour F, Zarnani AH, et al. Helicobacter pylori FliD protein is a highly sensitive and specific marker for serologic diagnosis of H. pylori infection. International Journal of Medical Microbiology. 2013;303:618-623
  57. 57. Gu H. Role of flagella in the pathogenesis of Helicobacter pylori. Current Microbiology. 2017;74:863-869
  58. 58. Clyne M, Ocroinin T, Suerbaum S, Josenhans C, Drumm B. Adherence of isogenic flagellum-negative mutants of Helicobacter pylori and Helicobacter mustelae to human and ferret gastric epithelial cells. Infection Immunity. 2000;68:4335-4339
  59. 59. Worku ML, Karim QN, Spencer J, Sidebotham RL. Chemotactic response of Helicobacter pylori to human plasma and bile. Journal of Medical Microbiology. 2004;53:807-811
  60. 60. Waskito LA, Salama NR, Yamaoka Y. Pathogenesis of Helicobacter pylori infection. Helicobacter. 2018;23(Suppl. 1):e12516
  61. 61. Aizawa SI, Harwood CS, Kadner RJ. Signaling components in bacterial locomotion and sensory reception. Journal of Bacteriology. 2000;182:1459-1471
  62. 62. Becker KW, Skaar EP. Metal limitation and toxicity at the interface between host and pathogen. FEMS Microbiological Review. 2014;38:1235-1249
  63. 63. Javaheri A, Kruse T, Moonens K, Mejías-Luque R, Debraekeleer A, Asche CI, et al. Helicobacter pylori adhesin HopQ engages in a virulence-enhancing interaction with human CEACAMs. Nature Microbiology. 2016;2:16189
  64. 64. Imkamp F, Lauener FN, Pohl D, Lehours P, Vale FF, Jehanne Q, et al. Rapid characterization of virulence determinants in Helicobacter pylori isolated from non-atrophic gastritis patients by next-generation sequencing. Journal of Clinical Medicine. 2019;8(7):1030
  65. 65. Kalali B, Mejías-Luque R, Javaheri A, Gerhard M. H. pylori virulence factors: Influence on immune system and pathology. Mediators Inflammation. 2014;2014:426309
  66. 66. Kable ME, Hansen LM, Styer CM, et al. Host determinants of expression of the Helicobacter pylori BabA adhesin. Scientific Reports. 2017;7:46499
  67. 67. Hansen LM, Gideonsson P, Canfield DR, Boren T, Solnick JV. Dynamic expression of the BabA adhesin and its BabB paralog during Helicobacter pylori infection in rhesus macaques. Infection and Immunity. 2017;85(6):e00094-e00017
  68. 68. Königer V, Holsten L, Harrison U, Busch B, Loell E, Zhao Q, et al. Helicobacter pylori exploits human CEACAMs via HopQ for adherence and translocation of CagA. Nature Microbiology. 2016;2:16188
  69. 69. Zhao Q, Busch B, Jiménez-Soto LF, Ishikawa-Ankerhold H, Massberg S, Terradot L, et al. Integrin but not CEACAM receptors are dispensable for Helicobacter pylori CagA translocation. PLoS Pathogens. 2018;14:e1007359
  70. 70. Chey WD, Leontiadis GI, Howden CW, Moss SF. ACG clinical guideline: Treatment of Helicobacter pylori infection. The American Journal of Gastroenterology. 2017;112(2):212-239
  71. 71. Chowdhury R. Host cell contact induces fur-dependent expression of virulence factors CagA and VacA in Helicobacter pylori. Helicobacter. 2014;19(1):17-25
  72. 72. Noto JM Jr, Peek RM. The Helicobacter pylori:cag pathogenicity island. Methods in Molecular Biology. 2012;921:41-50
  73. 73. Odenbreit S, Puls J, Sedlmaier B, et al. Translocation of Helicobacter pylori CagA into gastric epithelial cells by type IV secretion. Science. 2000;287:1497-1500
  74. 74. Baj J, Forma A, Sitarz M, Portincasa P, Garruti G, Krasowska D, et al. Helicobacter pylori virulence factorsmechanisms of bacterial pathogenicity in the gastric microenvironment. Cell. 2021;10(1):27
  75. 75. Altobelli A, Bauer M, Valez K, Cover TL, Muller A. Helicobacter pylori VacA targets myeloid cells in the gastric lamina propria to promote peripherally induced regulatory T-cell differentiation and persistent infection. mBio. 2019;10:e00261
  76. 76. Machlowska J, Baj J, Sitarz M, Maciejewski R, Sitarz R. Gastric cancer: Epidemiology, risk factors, classification, genomic characteristics and treatment strategies. International Journal of Molecular Sciences. 2020;21:4012
  77. 77. Behrens I-K, Busch B, Ishikawa-Ankerhold H, et al. The HopQ-CEACAM interaction controls CagA translocation, phosphorylation, and phagocytosis of Helicobacter pylori in neutrophils. MBio. 2020;11(1):e03256-ee3319
  78. 78. Bridge DR, Blum FC, Jang S, Kim J, Cha JH, Merrell DS. Creation and initial characterization of isogenic Helicobacter pylori CagA EPIYA variants reveals differential activation of host cell signaling pathways. Scientific Reports. 2017;7(1):1-14
  79. 79. Suzuki H, Warren R, Marshal B. Helicobacter pylori. Springer. 2016;1:267. DOI: 10.1007/978-4-431-55705-0
  80. 80. Zhang X, Zhang J, Zhang R, Guo Y, Wu C, Mao X, et al. Structural, enzymatic and biochemical studies on Helicobacter pylori arginase. The International Journal of Biochemistry & Cell Biology. 2013;45(5):995-1002
  81. 81. Dara M, Khashei R, Dehghani B. High frequency of hopQ genotypes among Iranian Helicobacter pylori clinical isolates. Le Infezioni in Medicina. 2017;25(2):123-126
  82. 82. Feige MH, Sokolova O, Pickenhahn A, Maubach G, Naumann M. HopQ impacts the integrin α5β1-independent NF-κB activation by Helicobacter pylori in CEACAM expressing cells. International Journal of Medical Microbiology. 2018;308(5):527-533
  83. 83. Švagelj D, Terzić V, Dovhanj J, Švagelj M, Cvrković M, Švagelj I. Superoxide dismutases in chronic gastritis. APMIS. 2016;124(4):252-256
  84. 84. Morey P, Meyer TF. The sweeping role of cholesterol depletion in the persistence of Helicobacter pylori infections. In: Backert S, editor. Molecular Mechanisms of Inflammation: Induction, Resolution and Escape by Helicobacter pylori. Cham: Springer; 2019. pp. 209-227
  85. 85. Zimmermann S, Pfannkuch L, Al-Zeer MA, Bartfeld S, Koch M, Liu J, et al. ALPK1-and TIFA-dependent innate immune response triggered by the Helicobacter pylori type IV secretion system. Cell Reports. 2017;20(10):2384-2395
  86. 86. Morey P, Pfannkuch L, Pang E, Boccellato F, Sigal M. Helicobacter pylori depletes cholesterol in gastric glands to prevent interferon gamma signaling and escape the inflammatory response. Gastroenterology;154:1391
  87. 87. Palrasu M, Zaika E, El-Rifai W, et al. Bacterial CagA protein compromises tumor suppressor mechanisms in gastric epithelial cells. The Journal of Clinical Investigation. 2020;130(5):2422-2434
  88. 88. Yeh YC, Kuo HY, Chang WL, et al. H. pylori isolates with amino acid sequence polymorphisms as presence of both HtrA-L171 & CagL-Y58/E59 increase the risk of gastric cancer. Journal of Biomedical Science. 2019;26:4
  89. 89. Hatakeyama M. Anthropological and clinical implications for the structural diversity of the Helicobacter pylori CagA oncoprotein. Cancer Science. 2011;102(1):36-43
  90. 90. McClain MS, Beckett AC, Cover TL. Helicobacter pylori vacuolating toxin and gastric cancer. Toxins. 2017;9(10):316
  91. 91. Foegeding NJ, Caston RR, McClain MS, Ohi MD, Cover TL. An overview of Helicobacter pylori VacA toxin biology. Toxins. 2016;8(6):173
  92. 92. Ricci V. Relationship between VacA toxin and host cell autophagy in Helicobacter pylori infection of the human stomach: A few answers, many questions. Toxins. 2016;8(7):203
  93. 93. Djekic A, Müller A. The Immunomodulator VacA Promotes Immune Tolerance and Persistent Helicobacter pylori Infection through Its Activities on T-Cells and Antigen-Presenting Cells. Toxins (Basel). 2016;8:2
  94. 94. Yahiro K, Hirayama T, Moss J, Noda M. New insights into VacA intoxication mediated through its cell surface receptors. Toxins. 2016;8(5):152
  95. 95. Muzahhed M. Helicobacter pylori oncogenicity: Mechanism, prevention and risk factors. Science World Journal. 2020
  96. 96. Jiang X, Xu Z, Zhang T, Li Y, Li W, Tan H. Wholegenome- based Helicobacter pylori Geographic surveillance: Visualized and expanded webtool. Frontiers in Microbiology. 2019;12:687259
  97. 97. Dadashzadeh K. Relevance of Helicobacter pylori dupA and oipA genotypes and development of gastric disease. Biomedical Research. 2017;28(19):8179-8183
  98. 98. Pereira WN, Ferraz MA, Zabaglia LM, de Labio RW, Orcini WA, Bianchi Ximenez JP, et al. Association among H. pylori virulence markers dupA, cagA and vacA in Brazilian patients. Journal of Venomous Animals and Toxins Including Tropical Diseases. 2014;20(1):1
  99. 99. Idowu A, Mzukwa A, Harrison U, Palamides P, Haas R, Mbao M, et al. Detection of Helicobacter pylori and its virulence genes (cagA, dupA, and vacA) among patients with gastroduodenal diseases in Chris Hani Baragwanath Academic Hospital. South Africa BMC gastroenterology. 2019;19(1):1-10
  100. 100. Sallas ML, Dos Santos MP, Orcini WA, David ÉB, Peruquetti RL, Payão SLM, et al. Status (on/off) of oipA gene: Their associations with gastritis and gastric cancer and geographic origins. Archives in Microbiology. 2019;201:93-97
  101. 101. Farzi N, Yadegar A, Aghdaei HA, Yamaoka Y, Zali MR. Genetic diversity and functional analysis of oipA gene in association with other virulence factors among Helicobacter pylori isolates from Iranian patients with different gastric diseases. Infectious Genetic Evolution. 2018;60:26-34
  102. 102. Safavi M, Sabourian R, Foroumadi A. Treatment of Helicobacter pylori infection: Current and future insights. World Journal of Clinical Cases. 2016;4:5-19
  103. 103. Yao C-C, Kuo C-M, Hsu C-N, Yang S-C, Wu C-K, Tai W-C, et al. First-line Helicobacter pylori eradication rates are significantly lower in patients with than those without type 2 diabetes mellitus. Infectious Drug Resistant. 2019;12:1425
  104. 104. Zagari RM, Rabitti S, Eusebi LH, Bazzoli F. Treatment of Helicobacter pylori infection: A clinical practice update. European Journal of Clinical Investigation. 2018;48(1):e12857
  105. 105. Chen PY, Wu MS, Chen CY, Bair MJ, Chou CK, Lin JT, et al. Systematic review with meta-analysis: The efficacy of levofloxacin triple therapy as the first-or second-line treatments of Helicobacter pylori infection. Alimentary Pharmacology & Therapeutics. 2016;44(5):427-437
  106. 106. Flores-Trevino S, Mendoza-Olazaran S, Bocanegra-Ibarias P. Helicobacter pylori drug resistance: Therapy changes and challenges. Expert Review of Gastroenterology & Hepatology. 2018;12(8):819-827
  107. 107. Bisht N, Garg AP. Isolation, characterization and probiotic value of lactic acid bacteria from milk and milk products. An International Journal of Biological Sciences. 2019;9:54
  108. 108. Yousefi B, Eslami M, Ghasemian A, Kokhaei P, Sadeghnejhad A. Probiotics can really cure an autoimmune disease? Gene Report. 2019;15:100364
  109. 109. Iqbal U, Khara HS, Akhtar D, Hu Y, Anwar H, Haq KF, et al. Safety and efficacy of Nitazoxanide-based regimen for the eradication of Helicobacter pylori infection: A systematic review and meta-analysis. Gastroenterology Research. 2020;13(6):260
  110. 110. Keikha M, Karbalaei M. Probiotics as the live microscopic fighters against Helicobacter pylori gastric infections. BMC Gastroenterology. 2021;21:388
  111. 111. Boyanova L, Gergova G, Markovska R, Yordanov D, Mitov I. Bacteriocinlike inhibitory activities of seven Lactobacillus delbrueckii subsp. Bulgaricus strains against antibiotic susceptible and resistant Helicobacter pylori strains. Letters in Applied Microbiology. 2017;65(6):469-474
  112. 112. Negash AW, Tsehai BA. Current applications of Bacteriocin. Int J Microbiol. 2020;2020:1-7
  113. 113. Lim E-S. Purification and characterization of two bacteriocins from Lactobacillus brevis BK11 and Enterococcus faecalis BK61 showing anti-Helicobacter pylori activity. Journal of Korean Society for Applied Biological Chemistry 2015;58(5):703-714.
  114. 114. Rezaee P, Kermanshahi RK, Falsafi T. Antibacterial activity of lactobacilli probiotics on clinical strains of Helicobacter pylori. Iranian Journal of Basic Medical Sciences. 2019;22(10):1118

Written By

Neha Bisht and Amar P. Garg

Submitted: 30 June 2022 Reviewed: 26 July 2022 Published: 27 October 2022