Open access peer-reviewed chapter

Biofilm and Quorum Sensing in Helicobacter pylori

Written By

Tarik Aanniz, Wissal Bakri, Safae El Mazouri, Hajar Wakrim, Ilham Kandoussi, Lahcen Belyamani, Mouna Ouadghiri and Azeddine Ibrahimi

Submitted: 17 January 2022 Reviewed: 18 March 2022 Published: 31 May 2022

DOI: 10.5772/intechopen.104568

From the Edited Volume

Focus on Bacterial Biofilms

Edited by Theerthankar Das

Chapter metrics overview

186 Chapter Downloads

View Full Metrics

Abstract

Helicobacter pylori (H. pylori) is a gram-negative bacterium living in the human gastrointestinal tract considered as the most common cause of gastritis. H. pylori was listed as the main risk factor for gastric cancer. Triple therapy consisting of a proton pump inhibitor and combinations of antibiotics is the main treatment used. However, this line of therapy has proven less effective mainly due to biofilm formation. Bacteria can regulate and synchronize the expression of multiple genes involved in virulence, toxin production, motility, chemotaxis, and biofilm formation by quorum sensing (QS), thus contributing to antimicrobial resistance. Henceforth, the inhibition of QS called quorum quenching (QQ) is a promising target and alternative to fight H. pylori resistance to antimicrobials. Many phytochemicals as well as synthetic compounds acting as quorum quenchers in H. pylori were described in vitro and in vivo. Otherwise, many other compounds known as quorum quenchers in other species and inhibitors of biofilm formation in H. pylori could act as quorum quenchers in H. pylori. Here, we summarize and discuss the latest findings on H. pylori’s biofilm formation, QS sensing, and QQ mechanisms.

Keywords

  • biofilm
  • Helicobacter pylori
  • quorum sensing
  • bacterial resistance
  • chemoreceptor
  • quorum quenching

1. Introduction

Helicobacter pylori (H. pylori) is a microaerophilic, spiral-shaped, gram-negative bacterium that belongs to Epsilonproteobacteria [1]. H. pylori establishes about 50% life-long infections. While it is asymptomatic in 85% of cases, individuals with chronic gastritis linked to H. pylori have a 10–20% chance to develop peptic ulcers and 1% chance to develop gastric carcinoma [2]. Barry Marshall and Robin Warren were the first to successfully isolate and culture H. pylori from the human stomach in 1983 [3]. The pair later conducted self-ingestion experiments that confirmed H. pylori’s colonization of the human stomach, thereby inducing inflammation of the gastric mucosa. Marshall first reported the development of persistent gastritis after ingestion, which was treated with doxycycline and bismuth subsalicylate [4]. These findings promoted more research, which ended up showing that high amount of H. pylori in the stomach promotes multiple gastrointestinal troubles, including chronic gastritis, peptic ulcer disease, gastric mucosa-associated lymphoid tissue (MALT) lymphoma, and gastric cancer [3].

In the early 1980s, Robin Warren and Barry Marshall showed for the first time that a bacterium named H. pylori could be associated with cancer development. In 2005, the Nobel Prize in Physiology or Medicine was awarded to R. Warren and B. Marshall for the “discovery of the bacterium H. pylori and its role in gastritis and peptic ulcer disease.”

Furthermore, the International Agency for Research on Cancer classified H. pylori in group 1 of carcinogens [5]. It has been shown that H. pylori infection may as well be correlated with insulin resistance, the increase of total and low-density lipoprotein cholesterol, and the decrease of high-density lipoprotein [6]. Due to differences in socioeconomic and hygienic conditions, H. pylori prevalence varies between and within countries. In general, it is estimated to range from 85–95% in developing countries and between 30% and 50% in developed countries [7]. The prevalence of the infection cannot be summarized in a single figure due to unreliable diagnostic methods in some regions, poor representation of some countries, and differences in data quality [8].

Currently, the first line therapy used to treat H. pylori infection is a combination of proton pump inhibitors (PPIs) with amoxicillin or metronidazole and clarithromycin. This triple therapy fails in about 20–30% of cases, requiring the use of a quadruple therapy consisting of a PPI, bismuth, tetracycline, and metronidazole [9, 10]. Nevertheless, an alarming increase in multidrug-resistant strains of H. pylori to ampicillin, penicillin, co-amoxiclav, amoxicillin, clarithromycin, metronidazole, tetracycline, doxycycline, erythromycin, and doxycycline has been reported [11, 12, 13]. This is ascribed to antibiotic abuse, therapeutic failures, and phenotypical mechanisms promoting resistance and/or tolerance to antimicrobials, notably, biofilm formation [14, 15]. Biofilm formation is a process in which organisms firmly adhere to abiotic, and/or biotic surfaces then grow together to form a complex community that often forms a special structure through four stages: (i) reversible bacterial adhesion; (ii) irreversible adhesion; (iii) formation and maturation of matrix; and (iv) dispersal of cells [16]. Biofilms mainly consist of extracellular polymeric substances composed of polysaccharides, proteins, nucleic acids, and lipids forming a protective barrier against adverse conditions and decreasing the penetration of antibiotics [17]. In H. pylori, flagella play a major role in biofilm formation in the gastrointestinal tract [18].

Most bacteria use quorum sensing (QS) as a communication system, relying on the secretion and perception of small molecules called auto-inducers (AIs) [19, 20]. The QS system can activate and/or regulate gene expression of many phenotypes that can be problematic for humans, i.e., biofilm formation, so that bacteria as a group can jointly cope with changes in the surrounding environment, resulting in adverse consequences such as drug resistance and virulence [21, 22]. A new tactic for outsmarting bacteria called quorum quenching (QQ) is currently explored to reduce their virulence without interfering with their growth, causing less Darwinian selection pressure for bacterial resistance [23]. This paradigm shift has become a promising antibacterial strategy, which not only prevents the development of antimicrobial resistance but also the disturbance of human gastrointestinal microflora, as well as the prevention of adverse side effects commonly associated with the available treatment [24]. Since the main steps of QS are the production and detection of signal molecules, QQ can interfere with this system in different ways, either intracellularly or extracellularly by application of inhibitors of AI biosynthesis and perception [25], application of AI antagonists (mimicking AIs), chemical inactivation of AI, sequestering antibodies [26] or macromolecules such as cyclodextrins [27], and degrading enzymes [28]. This strategy showed promising effect in vitro and in vivo, as well as synergistic effects with antibiotics by increasing bacterial susceptibility to antibiotics [29].

Here, we summarize the biofilm formation regulated by the QS system involved in the antimicrobial resistance in H. pylori. Meanwhile, we also provide the latest development of QS inhibitors (QSIs) or QQ enzymes (QQEs) as a potential strategy for the design of new antimicrobial agents to manage H. pylori infections.

Advertisement

2. Biofilm formation in H. pylori

Biofilms have been recognized as a microbial sessile community, irreversibly attached to either animate and inanimate objects [30]. Biofilms are contained in a self-produced extracellular polysaccharide (EPS) layer. This matrix is commonly rich in proteins including enzymes, polysaccharides (1–2%), nucleic acids (<1%), and water (up to 97%) [31]. Temperature, pH, osmolarity, UV radiation, desiccation, oxygen tension, and nutrient availability are all environmental stressors that directly affect the phenotype of biofilms [16, 32]. In vitro analyses have further confirmed that H. pylori biofilms reduce drug permeability and decrease the susceptibility to antibiotics. In fact, cells in the bacterial biofilm are 10–100 times more resistant toward antimicrobial agents than cells in a planktonic state [33, 34]. H. pylori colonizing the stomach has developed three patterns of drug resistance, including single drug resistance (SDR), heteroresistance (HR), and multidrug resistance (MDR), which probably overlap and are linked in their molecular mechanisms and their clinical implications [35, 36, 37, 38, 39, 40, 41, 42].

In the human stomach, H. pylori biofilms are found on the surface of gastric mucosa. Once introduced into the stomach, H. pylori appears in a spiral form, which is very mobile and associated with the colonization of new niches [43, 44, 45, 46]. Subsequently, it comes into contact with the mucin layer that covers the epithelial cells, resulting in tension-dependent adhesion between the mucin and H. pylori [47]. After an efficient adhesion and multiplication, a morphological transformation occurs, which is accompanied by the creation of multiple shapes (spiral, rod, curved, coccoid, and filamentous forms) to establish a biofilm [48]. However, in the case of prolonged colonization, all biofilm cells eventually transform into a coccoid form involved in survival and greater tolerance to adverse environmental factors [49, 50]. Biofilm formation in H. pylori involves many factors shown in Table 1.

FactorsReferences
Flagella and pili[18]
Outer membrane vesicles (OMV)[43]
Extracellular DNA (e-ADN)[43]
Adhesin (outer membrane proteins namely Hop & Hom)[51]
Lipopolysaccharides (LPS)[52]
Flagellar proteins[52]
Efflux pumps[53]
Enzymes regulating pH (urease and arginase)[54]
luxS gene[54]
Chemoreceptors[54]
Toxin-antitoxin system proteins[55]
H. pylori neutrophil-activating protein (HP-NAP)[55, 56]
Mannose-related proteoglycans (proteomannans)[57]

Table 1.

Factors involved in the formation of biofilms in Helicobacter pylori.

Advertisement

3. Biofilm formation and QS in H. pylori

The discovery of QS in Vibrio fischeri and Vibrio harveyi, two species that achieve bioluminescence using QS signaling molecules, sparked research into this complex signaling system [58]. The regulation of gene expression under QS control was investigated in multiple gram-negative bacteria species, including H. pylori [52, 59, 60]. For H. pylori, QS is involved in motility, biofilm development, and antibiotic resistance [32, 47, 59, 61]. Once biofilm formation is elicited from planktonic cells, the aggregated cells surrounded with extracellular polymeric substances (EPS) modify their phenotype, exchange genetic material, produce AI, and provide physical protection [33]. Owing to the formation of biofilms, H. pylori infections became typically persistent and rarely resolved by traditional antimicrobial therapies [34].

Overall, the QS system includes the following steps: (i) AI production; (ii) excretion of AI to the surrounding environment; (iii) sensing and binding of the AI to receptors at high cell density; (iv) retrieval of the receptor-signal complex from the cell and its binding to the promoter region; and (v) activation of genes expression [62, 63]. There are four different signals involved in QS. The most common are N-acyl homoserine lactones (AHLs), also known as autoinducer-1 (AI-1), which are fatty acid derivatives produced and used by gram-negative bacteria [64], while gram-positive bacteria use peptides or modified peptides. Furanosyl borate diesters or autoinducer-2 (AI-2) are derived from the recycling of S-adenosyl-homocysteine and used by both gram-positive and gram-negative bacteria [64]. There is also the autoinducer-3 (AI-3), which allows the cross-talking with mammalian epinephrine host cell signaling systems [65].

H. pylori, when located in the gastric mucosa, responds to several specific chemical signals. The chemotactic response is mediated by chemoreceptors called chemotaxis proteins [59]. H. pylori genome encodes four chemoreceptors: TlpA (effector; arginine, bicarbonate), TlpB (effector; AI2, urea, hydroxyurea, formamide acetamide.), TlpC (effector; unknown), and TlpD (effector; hydrogen peroxide) [66]. The H. pylori QS network involves the chemoreceptor TlpB responding to the AI-2 signaling molecule, a class of furanosyl borate diesters synthesized by the LuxS protein [59, 66] (Figure 1). The 4,5-dihydroxy-2,3-pentanedione (DPD), which is the precursor of AI-2 in H. pylori, is produced by LuxS protein [67]. First, LuxS produces the homocysteine through the cleavage of S-ribosylhomocysteine (SRH), which is a part of the S-adenosylmethionine (SAM) pathway. The process involves two main enzymes, i.e., 5′-methylthioadenosine/adenosylhomocysteine nucleosidase (MTAN) and metalloenzyme [68]. The DPD generated is rearranged into an assortment of chemically related molecules known as AI-2 through a process of dehydration and cyclization [69]. Usually, there are two types of chemoreceptor binding to their AIs, either through direct binding with AI or through interactions with AI binding proteins that transduce signals to the chemoreceptor [70]. In H. pylori, TlpB does not bind to AI-2 in vitro with high affinity and requires two periplasmic binding proteins, AibA and AibB, which bind to AI-2 independently. AibA and AibB are conserved at greater than 95% identity at the amino acid sequence level in all species of H. pylori [32]. The structures of AibA and AibB are not yet elucidated. However, protein sequence homology identifies AibA as homologous to dipeptide binding proteins (39% identity to E. coli dipeptide binding protein (PDB ID: 1DPP) and AibB as homologous to proteins of E. coli molybdate binding (36% identity to the periplasmic molybdate binding protein of Azotobacter vinelandii (PDB ID: 1ATG) [32].

Figure 1.

QS in Helicobacter pylori: LuxS produces AI-2 from the methyl cycle. At high cell density, high concentration of AI-2 in the environment bind to TlpB to active chemotaxis. The binding to the periplasmic proteins AibA and AibB active chemorepulsion. Moreover, AI-2 signals upstream of FlhA manages the branching pathways of gene expression under control of FlgS, FlgM, and σ28 proteins.

The QS system regulates several mechanisms to assure H. pylori colonization in the harsh conditions of the stomach. These include flagellar motility, chemotaxis, and the cag pathogenicity island (Cag PAI) expression, which are all involved in biofilm formation [18, 32, 60]. This indicates that the QS system regulates the various stages of biofilm development from the initial adhesion to the final detachment of the cells [46, 69]. The deletion of luxS gene altered the expression of flagellar genes, i.e., flaA, flaE, flhA, and fliI [69]. Otherwise, the addition of AI-2 or DPD restored the altered phenotype and transcription of these genes. This evidenced that AI-2 is involved in flagellar morphology in H. pylori as it influences the first steps of the flagellar gene expression (Figure 1) [69]. The presence of flagella provides motility that enhances the recruitment of planktonic cells to the biofilm, a crucial step in biofilm formation [18].

CagA protein, encoded by cag PAI, has been identified to be induced in H. pylori biofilms [54]. A significant decrease in biofilm biomass was observed following mutations in cagA and cag PAI, confirming its important role in biofilm formation [52]. The QS system regulates the cag PAI through its repression by AI-2, which, in turn, attenuates inflammatory response [60]. The type IV secretion system (T4SS), also encoded by cag PAI, is essential in direct cell–cell contact [71]. It is believed that this direct cell–cell contact can also control the biofilm behavior in H. pylori [33]. While cag PAI is involved in bacteria-host interaction, it could also be involved in H. pylori bacteria-bacteria interaction, as well as biofilm formation. Besides, bacterial outer membrane proteins (OMPs) are crucial for ion transport, osmotic stability, bacterial virulence, and adherence. Adhesion to gastric cell mediated by Omp18, a peptidoglycan-associated lipoprotein precursor, was reported in H. pylori [72]. After adhesion, the cell envelope gene (lpxD) is upregulated [73]. H. pylori urease enzyme (ureA) is important for pH regulation; it prevents the acidification of the biofilm, increasing its stability [74, 75]. Thus, omp18, lpxD, and ureA genes could be directly involved in H. pylori biofilm formation [76].

Advertisement

4. QQ in H. pylori

In H. pylori, AI-2 has been involved in the regulation of motility, type IV secretion, and, most importantly, biofilm formation [32]. The QS plays a critical role in multidrug resistance of H. pylori by upregulating both biofilm-associated matrix and efflux pump genes to improve bacterial resistance [77]. Cells in the bacterial biofilm are 100–1000 times more resistant toward antimicrobials than cells in a planktonic state [34]. The inhibition of QS results in a decrease in biofilm formation, making bacteria more susceptible [78].

Since the main component of QS is the production and detection of signal molecules, QQ can interfere with this system in different ways, either intracellularly or extracellularly. It includes: (i) the inhibition of signal synthesis; (ii) the inhibition of signal transmission; (iii) the enzymatic degradation of AI; and (iv) the inhibition of signal detection [25, 28] (Figure 2). These strategies showed promising effect in vitro and in vivo, as well as synergistic effects with traditional antibacterial treatments by increasing bacterial susceptibility to antibiotics [79].

Figure 2.

Different ways to inhibit QS in Helicobacter pylori.

To date, few H. pylori QSIs were described, whether synthetic or produced by living organisms, such as plants, animals, and bacteria [80, 81, 82]. Flavonoids, i.e., naringenin, quercetin, myricetin, baicalein, catechin, flavone, and turmeric, exhibited promising antibiofilm and antiadhesive properties against H. pylori [83, 84, 85, 86, 87, 88] (Table 2). Notably, a study conducted to assess the effect of Acorus calamus on H. pylori cultures demonstrated strong antibiofilm and antiadhesive properties [89]. Molecular interaction studies were later performed by the same group of researchers through molecular docking of β-sitosterol, a phytobioactive component of A. calamus, toward QS proteins ToxB, DnaA, PhnB, and Sip. Exceptionally high binding affinity and molecular interaction were exhibited, linking the antibiofilm properties of A. calamus to the inhibition of QS proteins by β-sitosterol [89]. The most direct and effective way to inhibit the QS system is the enzymatic degradation of the QS molecules, which stops signal transduction [93]. In gram-negative bacteria, two types of hydrolases were described, namely, AHL-lactonase and AHL-acylase. Today, few studies investigated the enzymatic lysis of QS signals in H. pylori. By degrading AHL produced by H. pylori, N-acylhomoserine lactonase produced by Bacillus licheniformis inhibited the biofilm formation and attenuate virulence [90].

QuencherEffect on H. pyloriTestMechanism of QQReference
β-sitosterol (Acorus calamus)Antibiofilm, Antibacterialin silico & in vitroAI-2 antagonist[89]
N-acylhomoserine lactonase (Bacillus licheniformis)Antibiofilm & antibacterialin vitroDegradation of AHL (Ais)[90]
Methylthio-DADMe-immucillin-AMTAN inhibitorin silicoBinding to the MTAN target[91]
Parachlorophenylthio-DADMe-immucillin-AMTAN inhibitorin silicoBinding to the MTAN target[91]
-SH Furanosyl Borate DiesterAntibiofilm, Antibacterial2in silicoAI-2 antagonist[92]

Table 2.

QSIs and QQEs in Helicobacter pylori.

Another effective way to inhibit QS is the blockage of signaling cascade through the inactivation of downstream response regulators. The precursor SRH of AI-2 results from the action of MTAN on SAH. The inhibition of MTAN induces an accumulation of 5-methylthioadenosine (MTA) and SAH, which, in turn, inhibits AI-2 production [91, 94]. In silico testing of DADMe-ImmA derivatives further confirmed this as a viable QQ technique, since it displayed MTAN inhibition by tight binding to the receptor [95]. More in silico studies investigated the possibility of designing furanosyl borate diester derivatives from its pharmacophore modeling by substituting the –OH groups of AI-2 and DPD by -SH making it a potent competitive inhibitor to AI-2 [92].

Based on previous studies, various phytochemicals from medicinal plants with known antibiofilm activity could act via inhibition of QS in H. pylori (Table 3). Baicalin from medicinal plants exhibited, in vivo, bactericidal and antiadhesive activities as well as limited urease production and reduced vacA gene expression, leading to virulence reduction. Baicalin limited the bacterial adhesion and colonization and enhanced bacterial sensitivity via suppression of urease and blockage of the sulfhydryl group. This makes Baicalin a potential quorum quencher in H. pylori [83, 88]. Quercetin from Vitis rotundifolia inhibited the growth of H. pylori [84], while in P. aeruginosa, quercetin inhibited AHL production suggesting its action through QQ against H. pylori. In parallel, catechin was described as a quorum quencher in P. fluorescens suggesting its potential inhibition of QS in H. pylori. Catechin from Chamomilla recutita inhibited the growth of H. pylori and urease production in H. pylori (which increases bacterial sensitivity) as well as caused membrane disruption [86]. Naringenin produced by Hibiscus rosa sinensis showed a potent bactericidal effect to MDR bacteria and also the inhibition of growth and biofilm formation in H. pylori [96]. Moreover, naringenin exhibited a potent competition with AHL for binding in P. aeruginosa. Taken together, it seems that naringenin inhibits biofilm formation in H. pylori by acting as quorum quencher. Turmeric (Curcuma longa) exhibited a good antibiofilm effect toward H. pylori [97, 101]. Besides, turmeric decreased AHL production in Aeromonas sobria and limited interaction with LuxI-type synthases and downregulated LuxI-type and LuxR-type genes in various bacterial species. This makes turmeric a potential quencher toward H. pylori. Vaccinium oxycoccus produces proanthocyanidins with antibiofilm and bacteriostatic activities against H. pylori [98]. Proanthocyanidins also limited the siallylactose-specific (S-fimbriae) adhesion of H. pylori to human mucus, erythrocytes, and gastric epithelial cells. In P. aeruginosa, proanthocyanidins was shown to inhibit AI production and to limit the activation of QS transcriptional regulators. Taken together, proanthocyanidins could be considered as a potent quorum quencher in H. pylori.

MoleculeEffect on H. pyloriTestPossible mechanismReference
BaicalinAntibiofilm
Adhesion inhibition
Bactericidal
Virulence reduction
Urease inhibition
in vivoReduction of binding and colonization
Suppression urease and blockade of sulfhydryl group.
[83, 88]
Quercetin
(V. rotundifolia)
Antibiofilm
Growth inhibition
in vitroQSI in P. aeruginosa[84]
Catechin
(Chamomilla recutita)
Antibiofilm
Growth inhibition Urease inhibition Membrane disruption
in vivoQSI in P. fluorescens[86]
Naringenin
(H. rosa sinensis)
Antibiofilm Bactericidalin vitroQSI in P. aeruginosa[96]
Turmeric
(C. longa)
Antibiofilm Antiadhesive Immunostimulant (igG toward H. pylori)in vitroInhibition of AHL production in A. sobria
Interaction with LuxI
Down-regulation of LuxI-type & LuxR
[97, 98]
Proantho-cyanidins
(Vaccinium oxycoccus)
Antibiofilm, Bacteriostatic,
Inhibits siallylactose-specific (S-fimbriae)
in vitro & in vivoInhibition of AHL production
Anti-QS regulators in P. aeruginosa
[98]
Emodin
(A. vera)
Antibiofilm
Antiadhesion
Affects n-acetyl transferase
in vitroInhibition of the HefA gene[99]
NiclosamideAntibiofilm
Bacteriostatic, Decreasing the secretion of IL-8,
Disruption of H. pylori proton motive force.
in vitro &
in vivo
QSI in P. aeruginosa
Affects transcription of QS genes in P. aeruginosa
[100]

Table 3.

Inhibitors of biofilm formation potentially via inhibition of QS in Helicobacter pylori.

Advertisement

5. Conclusion

Despite the advancements in the medical field, the treatment of H. pylori infections has lost its efficacy. H. pylori QS-mediated behavior is the main contributor to bacterial survival and pathogenicity. The significance of bacterial communication in the expression of pathogenic factors makes QS a great target to treat H. pylori infection or increase antibiotic efficacy by synergy. In the past two decades, researchers have discovered plenty of QSI agents that can prevent biofilm formation and decrease virulence. The development of new QSI/QQE that can be combined with antibiotics has been a hot topic in the antibacterial research field. More studies are required to demonstrate their mechanisms of action and the optimal doses of the QS inhibitory compounds that are safe and effective.

Advertisement

Conflict of interest

The authors declare no conflict of interest.

References

  1. 1. Pohl D, Peter MK, Valentine B, Karoline W. Review of current diagnostic methods and advances in Helicobacter pylori diagnostics in the era of next generation sequencing. World Journal of Gastroenterology. 2019;25(32):4629-4660. DOI: 10.3748/wjg.v25.i32.4629
  2. 2. Peek RM Jr, Jean EC. Helicobacter infection and gastric neoplasia. The Journal of Pathology. 2006;208(2):233-248. DOI: 0.1002/path.1868
  3. 3. Pincock S. Nobel prize winners robin warren and Barry Marshall. The Lancet. 2005;366(9495):1429. DOI: 10.1016/S0140-6736(05)67587-3
  4. 4. Marshall BJ. One hundred years of discovery and rediscovery of Helicobacter pylori and its association with peptic ulcer disease. In: Mobley HLT, Mendz GL, Hazell SL, editors. Helicobacter pylori: Physiology and Genetics. 1st ed. Washington, DC: ASM Press; 2001. pp. 19-26. DOI: 10.1128/9781555818005
  5. 5. “Schistosomes, Liver Flukes and Helicobacter pylori. IARC Working Group on the Evaluation of Carcinogenic Risks to Humans. Lyon, 7-14 June 1994.” IARC Monographs on the Evaluation of Carcinogenic Risks to Humans. 1994;61:1-241
  6. 6. Buzás GM. Metabolic consequences of Helicobacter pylori infection and eradication. World Journal of Gastroenterology. 2014;20(18):5226-5234. DOI: 10.3748/wjg.v20.i18.5226
  7. 7. Burucoa C, Axon A. Epidemiology of Helicobacter pylori infection. Helicobacter. 2017;22:e12403. DOI: 10.1111/hel.12403
  8. 8. Hooi JKY, Lai WY, Ng WK, Suen MMY, Underwood FE, Tanyingoh D, et al. Global prevalence of Helicobacter pylori infection: Systematic review and meta-analysis. Gastroenterology. 2017;153(2):420-429. DOI: 10.1053/j.gastro.2017.04.022
  9. 9. Jaka H, Andreas M, Christa K, Stephen EM. Predictors of triple therapy treatment failure among H. pylori infected patients attending at a tertiary Hospital in Northwest Tanzania: A prospective study. BMC Infectious Diseases. 2019;19(1):447. DOI: 10.1186/s12879-019-4085-1
  10. 10. Malfertheiner P, Megraud F, O’Morain CA, Atherton J, Axon ATR, Bazzoli F, et al. Management of Helicobacter pylori infection--the Maastricht IV/Florence consensus report. Gut. 2012;61(5):646-564. DOI: 10.1136/gutjnl-2012-302084
  11. 11. Almeida NJMR, Donato MM, Luxo C, Cardoso O, Cipriano MA, Marinho C, et al. Helicobacter pylori antimicrobial resistance rates in the central region of Portugal. Clinical Microbiology and Infection: The Official Publication of the European Society of Clinical Microbiology and Infectious Diseases. 2014;20(11):1127-1133. DOI: 10.1111/1469-0691.12701
  12. 12. De Palma GZ, Mendiondo N, Wonaga A, Viola L, Ibarra D, Campitelli E, et al. Occurrence of mutations in the antimicrobial target genes related to levofloxacin, clarithromycin, and amoxicillin resistance in Helicobacter pylori Isolates from Buenos Aires City. Microbial Drug Resistance. 2017;23(3):351-358. DOI: 10.1089/mdr.2015.0361
  13. 13. Mabeku LBK, Bille BE, Zemnou CT, Nguefack LDT, Leundji H. Broad Spectrum resistance in Helicobacter pylori isolated from gastric biopsies of patients with dyspepsia in Cameroon and efflux-mediated multiresistance detection in MDR isolates. BMC Infectious Diseases. 2019;19(1):880. DOI: 10.1186/s12879-019-4536-8
  14. 14. Apolinaria G, Salas-jara MJ, Herrera C, González C. Biofilm and Helicobacter pylori: From environment to human host. World Journal of Gastroenterology. 2014;20(19):5632-5638. DOI: 10.3748/wjg.v20.i19.5632
  15. 15. Cammarota G, Branca G, Ardito F, Sanguinetti M, Ianiro G, Cianci R, et al. Biofilm demolition and antibiotic treatment to eradicate resistant Helicobacter pylori: A clinical trial. Clinical Gastroenterology and Hepatology. 2010;8(9):817-820. DOI: 10.1016/j.cgh.2010.05.006
  16. 16. Wen Y, Wang Y, Liu L, He J. Biofilms: The microbial ‘protective clothing’ in extreme environments. International Journal of Molecular Sciences. 2019;20(14):3423. DOI: 10.3390/ijms20143423
  17. 17. Percival SL, Suleman L. Biofilms and Helicobacter pylori: Dissemination and persistence within the environment and host. World Journal of Gastrointestinal Pathophysiology. 2014;5(3):122. DOI: 10.4291/wjgp.v5.i3.122
  18. 18. Hathroubi S, Zerebinski J, Ottemann KM. Helicobacter pylori biofilm involves a multigene stress-biased response, including a structural role for flagella. MBio. 2018;9(5):e01973-18. DOI: 10.1128/mBio.01973-18
  19. 19. Diggle SP, Griffin AS, Campbell GS, West SA. Cooperation and conflict in quorum-sensing bacterial populations. Nature. 2007;450(7168):411-414. DOI: 10.1038/nature06279
  20. 20. Li YH, Tian X. Quorum sensing and bacterial social interactions in biofilms. Sensors. 2012;12(3):2519-2538. DOI: 10.3390/s120302519
  21. 21. Dong YH, Wang LH, Xu JL, Zhang HB, Zhang XF, Zhang LH. Quenching quorum-sensing-dependent bacterial infection by an N-acyl homoserine lactonase. Nature. 2001;411(6839):813-817. DOI: 10.1038/35081101
  22. 22. Miller BL, Bassler B. Quorum sensing in bacteria. Annual Review of Microbiology. 2001;55:165-199. DOI: 10.1146/annurev.micro.55.1.165
  23. 23. Maeda T, García-Contreras R, Pu M, et al. Quorum quenching quandary: Resistance to antivirulence compounds. The ISME Journal. 2012;6:493-501. DOI: 10.1038/ismej.2011.122
  24. 24. Huedo P, Coves X, Daura X, Gibert I, Yero D. Quorum sensing Signaling and quenching in the multidrug-resistant pathogen Stenotrophomonas Maltophilia. Frontiers in Cellular and Infection Microbiology. 2018;8(122):1-8. DOI: 10.3389/fcimb.2018.00122
  25. 25. Tang K, Zhang XH. Quorum quenching agents: Resources for Antivirulence therapy. Marine Drugs. 2014;12(6):3245-3282. DOI: 10.3390/md12063245
  26. 26. Park J, Jagasia R, Kaufmann GF, Mathison JC, Ruiz DI, Moss JA, et al. Infection control by antibody disruption of bacterial quorum sensing Signaling. Chemistry & Biology. 2007;14(10):1119-1127. DOI: 10.1016/j.chembiol.2007.08.013
  27. 27. Kato N, Morohoshi T, Nozawa T, Matsumoto H, Ikeda T. Control of gram-negative bacterial quorum sensing with Cyclodextrin immobilized cellulose ether gel. Journal of Inclusion Phenomena and Macrocyclic Chemistry. 2006;56(1):55-59. DOI: 10.1007/s10847-006-9060-y
  28. 28. Fetzner S. Quorum Quenching Enzymes. Journal of Biotechnology. 2015;201:2-14. DOI: 10.1016/j.jbiotec.2014.09.001
  29. 29. Ilk S, Sağlam N, Özgen M, Korkusuz F. Chitosan nanoparticles enhances the anti-quorum sensing activity of kaempferol. International Journal of Biological Macromolecules. 2017;94:653-662. DOI: 10.1016/j.ijbiomac.2016.10.068
  30. 30. Irie Y, Parsek MR. Quorum sensing and microbial biofilms. Current Topics in Microbiology and Immunology. 2008;322:67-84. DOI: 10.1007/978-3-540-75418-3_4
  31. 31. Cammarota G, Sanguinetti M, Gallo A, Posteraro B. Biofilm formation by Helicobacter pylori as a target for eradication of resistant infection. Alimentary Pharmacology & Therapeutics. 2012;36(3):222-230. DOI: 10.1111/j.1365-2036.2012.05165.x
  32. 32. Yonezawa H, Osaki T, Kamiya S. Biofilm formation by Helicobacter pylori and its involvement for antibiotic resistance. BioMed Research International. 2015;2015:914791. DOI: 10.1155/2015/914791
  33. 33. Binkowska A, Biernat M, Dus I, Gosciniak G. The role of biofilm formation in pathogenesis of Helicobacter pylori infections. Przeglad Gastroenterologiczny. 2013;8(1):27-30. DOI: 10.5114/pg.2013.34179
  34. 34. Mah TF, O’Toole GA. Mechanisms of biofilm resistance to antimicrobial agents. Trends in Microbiology. 2001;9(1):34-39. DOI: 10.1016/s0966-842x(00)01913-2
  35. 35. Savarino V, Zentilin P, Bisso G, Pivari M, Bilardi C, Biagini R, et al. Optimal duration of therapy combining ranitidine bismuth citrate with clarithromycin and metronidazole in the eradication of Helicobacter pylori infection. Alimentary Pharmacology & Therapeutics. 1999;13(1):43-47. DOI: 10.1046/j.1365-2036.1999.00436.x
  36. 36. Torres J, Camorlinga-Ponce M, Pérez-Pérez G, Madrazo-De la Garza A, Dehesa M, González-Valencia G, et al. Increasing multidrug resistance in Helicobacter pylori strains isolated from children and adults in Mexico. Journal of Clinical Microbiology. 2001;39(7):2677-2680. DOI: 10.1128/JCM.39.7.2677-2680.2001
  37. 37. Bardhan KD, Morton D, Perry MJ, Sanders DS, Morris P, Rowland A, et al. Ranitidine bismuth citrate with clarithromycin alone or with metronidazole for the eradication of Helicobacter pylori. Alimentary Pharmacology & Therapeutics. 2001;15(8):1199-1204. DOI: 10.1046/j.1365-2036.2001.01040.x
  38. 38. Zullo A, De Francesco V, Hassan C, Morini S, Vaira D. The sequential therapy regimen for Helicobacter pylori eradication: A pooled-data analysis. Gut. 2007;56(10):1353-1357. DOI: 10.1136/gut.2007.125658
  39. 39. Tanih NF, Ndip LM, Clarke AM, Ndip RN. An overview of pathogenesis and epidemiology of Helicobacter pylori infection. African Journal of Microbiology Research. 2010;4(6):426-436
  40. 40. Boyanova L, Davidkov L, Gergova G, Kandilarov N, Evstatiev I, Panteleeva E, et al. Helicobacter pylori susceptibility to fosfomycin, rifampin, and 5 usual antibiotics for H. pylori eradication. Diagnostic Microbiology and Infectious Disease. 2014;79(3):358-361. DOI: 10.1016/j.diagmicrobio.2014.03.028
  41. 41. Wueppenhorst N, Stueger HP, Kist M, Glocker EO. High secondary resistance to quinolones in German Helicobacter pylori clinical isolates. The Journal of Antimicrobial Chemotherapy. 2013;68(7):1562-1566. DOI: 10.1093/jac/dkt061
  42. 42. Bolor-Erdene M, Namdag B, Yamaoka Y, Jav S. Antibiotic resistance of Helicobacter pylori in Mongolia. The Journal of Infection in Developing Countries. 2017;11(11):887-894. DOI: 10.3855/jidc.8619
  43. 43. Azevedo NF, Pinto AR, Reis NM, Vieira MJ, Keevil CW. Shear stress, temperature, and inoculation concentration influence the adhesion of water-stressed Helicobacter pylori to stainless steel 304 and polypropylene. Applied and Environmental Microbiology. 2006;72(4):2936-2941. DOI: 10.1128/AEM.72.4.2936-2941.2006
  44. 44. Yang JC, Wang HL, Chern HD, Shun CT, Lin BR, Lin CJ, et al. Role of omeprazole dosage and cytochrome P450 2C19 genotype in patients receiving omeprazole-amoxicillin dual therapy for Helicobacter pylori eradication. Pharmacotherapy: The Journal of Human Pharmacology and Drug Therapy. 2011;31(3):227-238. DOI: 10.1592/phco.31.3.227
  45. 45. Rhee KH, Park JS, Cho MJ. Helicobacter pylori: Bacterial strategy for incipient stage and persistent colonization in human gastric niches. Yonsei Medical Journal. 2014;55(6):1453-1466. DOI: 10.3349/ymj.2014.55.6.1453
  46. 46. Krzyzek P, Gosciniak G. A proposed role for diffusible signal factors in the biofilm formation and morphological transformation of Helicobacter pylori. The Turkish Journal of Gastroenterology. 2018;29(1):7-13. DOI: 10.5152/tjg.2017.17349
  47. 47. Anderson JK, Huang JY, Wreden C, Goers Sweeney E, Goers J, Remington SJ, et al. Chemorepulsion from the quorum signal Autoinducer-2 promotes Helicobacter pylori biofilm dispersal. MBio. 2015;6(4):e00379-15. DOI: 10.1128/mBio.00379-15
  48. 48. Bavington C, Page C. Stopping bacterial adhesion: A novel approach to treating infections. Respiration. 2005;72(4):335-344. DOI: 10.1159/000086243
  49. 49. Krzyżek P, Paluch E, Gościniak G. Synergistic therapies as a promising option for the treatment of antibiotic-resistant Helicobacter pylori. Antibiotics. 2020;9(10):658. DOI: 10.3390/antibiotics9100658
  50. 50. Krzyżek P, Biernat MM, Gościniak G. Intensive formation of coccoid forms as a feature strongly associated with highly pathogenic Helicobacter pylori strains. Folia Microbiologica. 2019;64(3):273-281. DOI: 10.1007/s12223-018-0665-5
  51. 51. Servetas SL, Doster RS, Kim A, Windham IH, Cha JH, Gaddy JA, et al. ArsRS-dependent regulation of homB contributes to Helicobacter pylori biofilm formation. Frontiers in Microbiology. 2018;9:1497. DOI: DOI. 10.3389/fmicb.2018.01497
  52. 52. Wong EHJ, Ng CG, Chua EG, Tay ACY, Peters F, Marshall BJ, et al. Comparative genomics revealed multiple Helicobacter pylori genes associated with biofilm formation in vitro. PLoS One. 2016;11(11):1-16. DOI: 10.1371/journal.pone.0166835
  53. 53. Ge X, Cai Y, Chen Z, Gao S, Geng X, Li Y, et al. Bifunctional enzyme SpoT is involved in biofilm formation of Helicobacter pylori with multidrug resistance by upregulating efflux pump Hp1174 (gluP). Antimicrobial Agents and Chemotherapy. 2018;62(11):e00957-18. DOI: 10.1128/AAC.00957-18
  54. 54. Shao C, Sun Y, Wang N, Yu H, Zhou Y, Chen C, et al. Changes of proteome components of Helicobacter pylori biofilms induced by serum starvation. Molecular Medicine Reports. 2013;8(6):1761-1766. DOI: 10.3892/mmr.2013.1712
  55. 55. Cárdenas-Mondragón MG, Ares MA, Panunzi LG, Pacheco S, Camorlinga-Ponce M, Girón JA. De la Cruz MA. Transcriptional profiling of type II toxin–antitoxin genes of Helicobacter pylori under different environmental conditions: Identification of HP0967–HP0968 system. Frontiers in Microbiology. 2016;7:1872. DOI: 10.3389/fmicb.2016.01872
  56. 56. Satin B, Del Giudice G, Della Bianca V, Dusi S, Laudanna C, Tonello F, et al. The neutrophil-activating protein (HP-NAP) of Helicobacter pylori is a protective antigen and a major virulence factor. The Journal of Experimental Medicine. 2000;191(9):1467-1476. DOI: 10.1084/jem.191.9.1467
  57. 57. Bessa LJ, Grande R, Iorio DD, Giulio MD, Campli ED, Cellini L. Helicobacter pylori free-living and biofilm modes of growth: Behavior in response to different culture media. APMIS. 2013;121(6):549-560. DOI: 10.1111/apm.12020
  58. 58. Nealson KH, Hastings JW. Bacterial bioluminescence: Its control and ecological significance. Microbiological Reviews. 1979;43(4):496-518. DOI: 10.1128/MMBR.43.4.496-518.1979
  59. 59. Rader BA, Wreden C, Hicks KG, Sweeney EG, Ottemann KM, Guillemin K. Helicobacter pylori perceives the quorum-sensing molecule AI-2 as a Chemorepellent via the chemoreceptor TlpB. Microbiology. 2011;157(9):2445-2455. DOI: 10.1099/mic.0.049353-0
  60. 60. Wen Y, Huang H, Tang T, Yang H, Wang X, Huang X, et al. AI-2 represses CagA expression and bacterial adhesion, attenuating the Helicobacter pylori-induced inflammatory response of gastric epithelial cells. Helicobacter. 2021;26(2):e12778. DOI: 10.1111/hel.12778
  61. 61. Challa S, Neelapu NRR. Quorum sensing in Helicobacter pylori: Role of biofilm and its implications for antibiotic resistance and immune evasion. In: Implication of Quorum Sensing System in Biofilm Formation and Virulence. 1st ed. Singapore: Springer; 2019. pp. 277-286. DOI: 10.1007/978-981-13-2429-1_18
  62. 62. Deng Y, Wu J, Tao F, Zhang LH. Listening to a new language: DSF-based quorum sensing in gram-negative bacteria. Chemical Reviews. 2011;111:160-173. DOI: 10.1021/cr100354f M.B
  63. 63. Duran N, Justo GZ, Duran M, Brocchi M, Cordi L, Tasic L, et al. Advances in Chromobacterium violaceum and properties of violacein-its main secondary metabolite: A review. Biotechnology Advances. 2016;34:1030-1045. DOI: 10.1016/j.biotechadv.2016.06.003
  64. 64. Hemmati F, Salehi R, Ghotaslou R, Kafil HS, Hasani A, Gholizadeh P, et al. Quorum quenching: A potential target for antipseudomonal therapy. Infection and Drug Resistance. 2020;13:2989-3005. DOI: 10.2147/IDR.S263196
  65. 65. Asfour HZ. Anti - quorum sensing natural compounds. Journal of Microscopy and Ultrastructure. 2018;6(1):1-10. DOI: 10.4103/JMAU.JMAU
  66. 66. Ortega Á, Zhulin IB, Krell T. Sensory repertoire of bacterial chemoreceptors. Microbiology and Molecular Biology Reviews. 2017;81(4):e00033-e00017. DOI: 10.1128/MMBR.00033-17
  67. 67. Bassler BL, Greenberg EP, Stevens AM. Cross-species induction of luminescence in the quorum-sensing bacterium Vibrio harveyi. Journal of Bacteriology. 1997;179(12):4043-4045. DOI: 10.1128/jb.179.12.4043-4045.1997
  68. 68. Forsyth MH, Cover TL. Intercellular communication in Helicobacter pylori: LuxS is essential for the production of an extracellular Signaling molecule. Infection and Immunity. 2000;68(6):3193-3199. DOI: 10.1128/IAI.68.6.3193-3199.2000
  69. 69. Rader BA, Campagna SR, Semmelhack MF, Bassler BL, Guillemin K. The quorum-sensing molecule autoinducer 2 regulates motility and Flagellar morphogenesis in Helicobacter pylori. Journal of Bacteriology. 2007;189(17):6109-6117. DOI: 10.1128/JB.00246-07
  70. 70. Miller ST, Xavier KB, Campagna SR, Taga ME, Semmelhack MF, Bassler BL, et al. Salmonella typhimurium recognizes a chemically distinct form of the bacterial quorum-sensing signal AI-2. Molecular Cell. 2004;15:677-687. DOI: 10.1016/j.molcel.2004.07.020
  71. 71. Mattar R, Marques SB, Monteiro MDS, Dos Santos AF, Iriya K, Carrilho FJ. Helicobacter pylori cag Pathogenicity Island genes: Clinical relevance for peptic ulcer disease development in Brazil. Journal of Medical Microbiology. 2007;56(1):9-14. DOI: 10.1099/jmm.0.46824-0
  72. 72. Voland P, Hafsi N, Zeitner M, Laforsch S, Wagner H, Prinz C. Antigenic properties of HpaA and Omp18, two outer membrane proteins of Helicobacter pylori. Infection and Immunity. 2003;71(7):3837-3843. DOI: 10.1128/IAI.71.7.3837-3843.2003
  73. 73. Kim N, Marcus EA, Wen Y, Weeks DL, Scott DR, Jung HC, et al. Genes of Helicobacter pylori regulated by attachment to AGS cells. Infection and Immunity. 2004;72(4):2358-2368. DOI: 10.1128/IAI.72.4.2358-2368.2004
  74. 74. Dunne C, Dolan B, Clyne M. Factors that mediate colonization of the human stomach by Helicobacter pylori. World Journal of Gastroenterology. 2014;20(19):5610-5624. DOI: 10.3748/wjg.v20.i19.5610
  75. 75. Coticchia JM, Sugawa C, Tran VR, Gurrola J, Kowalski E, Carron MA. Presence and density of Helicobacter pylori biofilms in human gastric mucosa in patients with peptic ulcer disease. Journal of Gastrointestinal Surgery. 2006;10(6):883-889. DOI: 10.1016/j.gassur.2005.12.009
  76. 76. Lucio-Sauceda DG, Urrutia-Baca VH, Gomez-Flores R, La Garza-Ramos D, Angélica M, Tamez-Guerra P, et al. Antimicrobial and anti-biofilm effect of an electrolyzed Superoxidized solution at neutral-PH against Helicobacter pylori. BioMed Research International. 2019;2019:6154867. DOI: 10.1155/2019/6154867
  77. 77. De Francesco V, Zullo A, Hassan C, Giorgio F, Rosania R, Ierardi E. Mechanisms of Helicobacter pylori antibiotic resistance: An updated appraisal. World Journal of Gastrointestinal Pathophysiology. 2011;2(3):35-41. DOI: /10.4291/wjgp.v2.i3.35
  78. 78. Cushnie TT, Lamb AJ. Recent advances in understanding the antibacterial properties of flavonoids. International Journal of Antimicrobial Agents. 2011;38(2):99-107. DOI: 10.1016/j.ijantimicag.2011.02.014
  79. 79. Ning Q , Wang D, You J. Joint effects of antibiotics and quorum sensing inhibitors on resistance development in bacteria. Environmental Science: Processes & Impacts. 2021;23:995-1005. DOI: 10.1039/D1EM00047K
  80. 80. Ahumedo M, Velásquez M, Reyes RV. Virtual screening: Identification of compounds with possible quorum sensing agonistic activity in Pseudomonas aeruginosa. Vitae. 2017;24(2):89-101. DOI: 10.17533/udea.vitae.v24n2a02
  81. 81. Miró-Canturri A, Ayerbe-Algaba R, Smani Y. Drug repurposing for the treatment of bacterial and fungal infections. Frontiers in Microbiology. 2019;10:41-52. DOI: 10.3389/fmicb.2019.00041
  82. 82. O’Mahony R, Al-Khtheeri H, Weerasekera D, Fernando N, Vaira D, Holton J, et al. Bactericidal and anti-adhesive properties of culinary and medicinal plants against Helicobacter pylori. World Journal of Gastroenterology. 2005;11(47):7499-7507. DOI: 10.3748/wjg.v11.i47.7499
  83. 83. Chen ME, Su CH, Yang JS, Lu CC, Hou YC, Wu JB, et al. Baicalin, Baicalein, and lactobacillus Rhamnosus JB3 alleviated Helicobacter pylori infections in vitro and in vivo. Journal of Food Science. 2018;83(12):3118-3125. DOI: 10.1111/1750-3841.14372
  84. 84. Brown JC, Wang J, Kasman L, Jiang X, Haley-Zitlin V. Activities of Muscadine grape skin and quercetin against Helicobacter pylori infection in mice. Journal of Applied Microbiology. 2011;110(1):139-146. DOI: 10.1111/j.1365-2672.2010.04870.x
  85. 85. Krzyżek P, Junka A, Słupski W, Dołowacka-Jóźwiak A, Płachno BJ, Sobiecka A, et al. Antibiofilm and antimicrobial-enhancing activity of Chelidonium majus and Corydalis cheilanthifolia extracts against multidrug-resistant Helicobacter pylori. Pathogens. 2021;10(8):1033. DOI: 10.3390/pathogens10081033
  86. 86. Shin JE, Kim JM, Bae EA, Hyun YJ, Kim DH. In vitro inhibitory effect of flavonoids on growth, infection and Vacuolation of Helicobacter pylori. Planta Medica. 2005;71(3):197-201. DOI: 10.1055/s-2005-837816
  87. 87. Jiang X, Jiang C, Huang C, Chen G, Jiang K, Huang B, et al. Berberine combined with triple therapy versus triple therapy for Helicobacter pylori eradication: A Meta-analysis of randomized controlled trials. Evidence-based Complementary and Alternative Medicine. 2018;2018:8716910. DOI: 10.1155/2018/8716910
  88. 88. Yu XD, Zheng RB, Xie JH, Su JY, Huang XQ , Wang YH, et al. Biological evaluation and molecular docking of Baicalin and Scutellarin as Helicobacter pylori urease inhibitors. Journal of Ethnopharmacology. 2015;162:69-78. DOI: 10.1016/j.jep.2014.12.041
  89. 89. Prasad A, Devi AT, Prasad MN, Zameer F, Shruthi G, Shivamallu C. Phyto anti-biofilm elicitors as potential inhibitors of Helicobacter pylori. 3 Biotech. 2019;9(2):53. DOI: 10.1007/s13205-019-1582-2
  90. 90. Gopalakrishnan V, Masanam E, Ramkumar VS, Baskaraligam V, Selvaraj G. Influence of N-Acylhomoserine lactonase silver nanoparticles on the quorum sensing system of Helicobacter pylori: A potential strategy to combat biofilm formation. Journal of Basic Microbiology. 2020;60(3):207-215. DOI: 10.1002/jobm.201900537
  91. 91. Harijan RK, Hoff O, Ducati RG, Firestone RS, Hirsch BM, Evans GB, et al. Selective inhibitors of Helicobacter pylori Methylthioadenosine Nucleosidase and human Methylthioadenosine phosphorylase. Journal of Medicinal Chemistry. 2019;62(7):3286-3296. DOI: 10.1021/acs.jmedchem.8b01642
  92. 92. Kumar M, Naaz A, Sharma A, Teotia D, Nandi S. Pharmacophore Modeling of Furanosyl borate Diester: An attempt to combat antibiotic resistance induced Helicobacter pylori. Quorum sensing mediated peptic and duodenal ulcer. Journal of Bioanalysis & Biomedicine. 2018;10(6):114-117. DOI: 10.4172/1948-593x.1000218
  93. 93. Zhou L, Zhang Y, Ge Y, Zhu X, Pan J. Regulatory mechanisms and promising applications of quorum sensing-inhibiting agents in control of bacterial biofilm formation. Frontiers in Microbiology. 2020;11:1-11. DOI: 10.3389/fmicb.2020.589640
  94. 94. Ronning DR, Iacopelli NM, Mishra V. Enzyme-ligand interactions that drive active site rearrangements in the Helicobacter pylori 5′-Methylthioadenosine/S-Adenosylhomocysteine Nucleosidase. Protein Science. 2010;19(12):2498-2510. DOI: 10.1002/pro.524
  95. 95. Wang S, Haapalainen AM, Yan F, Du Q , Tyler PC, Evans GB, et al. A picomolar transition state analogue inhibitor of MTAN as a specific antibiotic for Helicobacter pylori. Biochemistry. 2012;51(35):6892-6894. DOI: 10.1021/bi3009664
  96. 96. Tran TH, Huynh TTH, Thuy NTL, Minh NVH, Nguyen TMN, Thi LMN. Growth-inhibiting, bactericidal, Antibiofilm, and urease inhibitory activities of Hibiscus rosa sinensis L. flower constituents toward antibiotic sensitive-and resistant-strains of Helicobacter pylori. ACS Omega. 2020;5(32):20080-20089. DOI: 10.1021/acsomega.0c01640
  97. 97. Vetvicka V, Vetvickova J, Fernandez-Botran R. Effects of curcumin on Helicobacter pylori infection. Annals of Translational Medicine. 2016;4(24):1-7. DOI: 10.21037/atm.2016.12.52
  98. 98. Shmuely H, Ofek I, Weiss EI, Rones Z, Houri-Haddad Y. Cranberry components for the therapy of infectious disease. Current Opinion in Biotechnology. 2012;23(2):148-152. DOI: 10.1016/j.copbio.2011.10.009
  99. 99. Cellini L, Di Bartolomeo S, Di Campli E, Genovese S, Locatelli M, Di Giulio M. In vitro activity of aloe Vera inner gel against Helicobacter pylori strains. Letters in Applied Microbiology. 2014;59(1):43-48. DOI: 10.1111/lam.12241
  100. 100. Tharmalingam N, Port J, Castillo D, Mylonakis E. Repurposing the anthelmintic drug Niclosamide to combat Helicobacter pylori. Scientific Reports. 2018;8(1):1-12. DOI: 10.1038/s41598-018-22037-x
  101. 101. Pattiyathanee P, Vilaichone RK, Chaichanawongsaroj N. Effect of curcumin on Helicobacter pylori biofilm formation. African Journal of Biotechnology. 2009;8(19):5106-5115. DOI: 10.4314/ajb.v8i19.65231

Written By

Tarik Aanniz, Wissal Bakri, Safae El Mazouri, Hajar Wakrim, Ilham Kandoussi, Lahcen Belyamani, Mouna Ouadghiri and Azeddine Ibrahimi

Submitted: 17 January 2022 Reviewed: 18 March 2022 Published: 31 May 2022