Đánh giá pan strep 3 ic

Gastric cancer is the fifth most common and fourth type to cause the highest mortality rates worldwide. The leading cause is related to Helicobacter pylori [H. pylori] infection. Unfortunately, current treatments have low success rates, highlighting the need for alternative treatments against carcinogenic agents, specifically H. pylori. Noteworthy, natural origin products contain pharmacologically active metabolites such as flavonoids, with potential antimicrobial applications.

Objective: This article overviews flavonoid-rich extracts’ biological and pharmacological activities. It focuses on using these substances against Helicobacter pylori infection to prevent gastric cancer. For this, PubMed and Science Direct databases were searched for studies that reported the activity of flavonoids against H. pylori, published within a 10-year time frame [2010 to August 2020]. It resulted in 1,773 publications, of which 44 were selected according to the search criteria. The plant family primarily found in publications was Fabaceae [9.61%]. Among the flavonoids identified after extraction, the most prevalent were quercetin [19.61%], catechin [13.72], epicatechin [11.76], and rutin [11.76]. The potential mechanisms associated with anti-H. pylori activity to the extracts were: inhibition of urease, damage to genetic material, inhibition of protein synthesis, and adhesion of the microorganism to host cells.

Conclusion: Plant extracts rich in flavonoids with anti-H. pylori potential proved to be a promising alternative therapy source, reinforcing the relevance of studies with natural products.

Keywords: phytotherapy, stomach neoplasms, anti-infective agents, ethnopharmacology, preventive medicine

Graphical Abstract

![An external file that holds a picture, illustration, etc. Object name is FPHAR_fphar-2022-951125_wc_abs.jpg][////i0.wp.com/www.ncbi.nlm.nih.gov/pmc/articles/PMC9470917/bin/FPHAR_fphar-2022-951125_wc_abs.jpg]

Introduction

Gastric cancer represents the fifth most common type of cancer [1.08 million cases in 2020]. It is the fourth most common cause of death among cancers worldwide [768.000 deaths in 2020] due to the advanced stage at diagnosis [; ]. Incidence and mortality vary considerably between regions, although it is more prevalent in developing countries, in which the percentage of cases is equivalent to 70%, especially in East Asia. In addition, the incidence of gastric cancer is proportional to population age, with an average of 68 years, and is more common in men [1 in each 96] than in women [1 in each 152] [; medical and editorial content Team, 2018].

According to the topology, gastric cancer can be classified into cardia, usually associated with gastroesophageal reflux, and non-cardia or distal gastric cancer, caused by the interaction with different factors [; ]. The latter is histologically subdivided according to Laurén’s classification in diffuse and intestinal []. The diffuse type consists of individually infiltrated neoplastic cells without glandular structures. In contrast, the intestinal type mimics the intestinal glands. It progresses through a series of histological changes that begin with transitioning from normal mucosa to chronic superficial gastritis, followed by atrophic gastritis, intestinal metaplasia, dysplasia, and adenocarcinoma [; ]. For both types of non-cardiac gastric cancer, the main factor associated with developing approximately 90% of adenocarcinomas is the bacterium Helicobacter pylori [H. pylori] [; , ; ; ].

  1. pylori is a gram-negative, flagellated, microaerophilic bacteria that infects the epithelial lining of the stomach [; ]. The infection is prevalent in approximately 50% of the world population, varying according to geographic region, age, socioeconomic status, education level, environment, and occupation. It is usually contracted in the first years of life and tends to persist until the completion of the appropriate treatment [; ; ; ].

The inflammatory process developed by H. pylori infection involves a variety of pathways induced in both gastric epithelial cells and circulating immune cells recruited to the infection site. Activated pathways involve mitogen-activated protein kinase [MAPK], nuclear factor-κB [NF-kB], activating protein [AP]-1, Wnt/β-catenin, PI3K pathways, signal transducers and transcription activators 3 [STAT3]. These alterations cause an increase in the production of inflammatory cytokines, such as interleukin 1 [IL-1], IL-6, IL-8, and tumor necrosis factor-alpha [TNF-α]. Also, they alter the apoptosis rate and proliferation and differentiation of epithelial cells. These phenomena result in the oncogenic transformation of epithelial cells and gastric cancer formation [; ; ]. In addition, virulence factors contribute to determining the pattern of immune defense performed in response to the infection. It includes the factors named vacuolating cytotoxin [VacA], cytotoxin-associated antigen A [CagA], the Cag pathogenicity island [PAI], HP-NAP, oipA, and dupA [; ; ; , ].

The clinical condition produced by the microorganism in question is usually asymptomatic. However, the infection caused is associated with gastrointestinal diseases, such as chronic gastritis, peptic ulcer disease, gastric B-cell mucosa-associated lymphoid tissue lymphoma, and, as aforementioned, gastric adenocarcinoma. Thereby, H. pylori was recognized and classified as a definite [group 1] carcinogen by the World Health Organization’s International Agency for Research on Cancer in 1994 [; ; ; ]. Additionally, studies demonstrate that eradicating H. pylori decreases the risk of developing cancer in individuals without pre-malignant lesions. It reinforces that this infection influences the early stages of gastric carcinogenesis [].

Currently, the treatment for gastric cancer consists of surgical intervention associated with chemotherapy using 5-fluorouracil [5-FU], platinum, taxane, irinotecan, and anthracycline []. The treatment options for tumors associated with H. pylori are antibiotics [clarithromycin and amoxicillin or metronidazole], proton pump inhibitors, and bismuth []. However, surgery remains the only curative therapy. At the same time, chemotherapy can improve the outcome, thus emphasizing the importance of employing preventive recourses [].

Alternatively, there is an increase in the use of natural products. One of the most investigated sources is the plants, representing 25% of the medical industry [; ; ]. Botanical drugs contain active metabolites with pharmacological activities capable of relieving symptoms or curing diseases [; ]. Thus, the application of ethnopharmacology has collaborated in discovering new chemical entities, mainly through the bioprospecting of secondary metabolites [].

Among the most known and studied ethnobotanical constituents are flavonoids. They possess a polyphenolic benzo-γ-pyrone structure of low molecular weight [; ]. Moreover, these compounds present antioxidant [; ; ], hepatoprotective [; ; ], anti-inflammatory [; ; ], anticancer [; ; ], antiviral [; ], and antibacterial properties [; ; ; ]. Concerning the antibacterial activity, flavonoids can inhibit the synthesis of nucleic acids, the function of the cytoplasmic membrane, and energy metabolism, among others. These activities prompt their application as antibacterial drugs with a scope of possible mechanisms of action [; ].

The antimicrobial potential of flavonoids against H. pylori has been described in the literature by several studies [; ; ; ; ; ; ]. In addition to demonstrating an in vitro action against this bacterium, these compounds could also promote synergistic interactions with antibiotics commonly used in treatments against H. pylori infections []. The pathways of action of flavonoids can be diverse. Some mechanisms in H. pylori have already been described, such as inhibition of the essential function of HsrA [], mediation of the response to oxidative stress [, ; ], interactions with virulence factors [], recognition of molecular targets including secretion systems [] and enzymes [; ] acting on pathways that lead to changes in cell morphology of H. pylori []. In turn, the broad pharmacotherapeutic and biochemical spectrum of flavonoids and the possible contribution of these compounds to improving human health make such substances increasingly explored []. Still, these findings support the valuable potential of flavonoids as candidate botanical drugs for novel antibacterial and anticancer strategies. The most promising flavonoid compounds are Catechin, Epicatechin, Kaempferol, Luteolin, Morin, Myricetin, Naringenin, Naringin, Quercetin, Hyperoside, and Rutin, In this way, this article provides an overview of the biological and pharmacological activities of flavonoid-rich plant extracts with a focus on the use of these substances against H. pylori infection in the prevention of gastric cancer.

Materials and Methods

The Question Under Analysis

This review was guided by the question: “Are flavonoids-rich plant extracts a promising alternative in treatments against Helicobacter pylori infection and preventing gastric cancer?”

Search Strategy and Articles Selection

PRISMA guidelines were followed [; ]. In addition, an electronic search was performed in the PubMed and Science Direct databases from studies published between 2010 to August 2020, with the keywords “flavonoids” and “Helicobacter pylori.”

Studies Selection

Eligible studies followed the criteria: 1] pre-clinical in vitro and in vivo; 2] studies with rodents and cells; 3] any type of treatment that used plant extracts containing flavonoids in its composition; 4] studies with positive or negative control; 5] no language restriction. Clinical research, studies with other than flavonoids that did not determine the value of the tested dose, with flavonoids in their isolated form, and studies that used flavonoids as control compounds, were excluded

Two independent reviewers selected the studies. In the first screening, titles and abstracts were evaluated, and studies considered irrelevant were excluded. The two reviewers read the articles for each potential manuscript and evaluated them based on the inclusion criteria. Duplicate studies between the bases were excluded. A third reviewer was contacted in the presence of inconsistency between the two examiners. Thus, 44 articles were selected for this review that reports the activity of plant species extracts containing flavonoids against H. pylori.

Results

The initial search of the databases [with the strategies presented in Table 1] allowed the identification of 1,773 publications. Review studies, meta-analyzes, encyclopedias, book chapters, abstracts, conference proceedings, editorials/letters, and case reports were excluded. The 567 articles left were screened based on titles and abstracts for the inclusion criteria mentioned above. At this stage, 128 articles remained, following the exclusion of 439 articles. Subsequently to the removal of 25 repeated articles, 103 remained. These studies were afterward entirely read. Finally, 44 articles were selected, as 59 articles did not meet all inclusion criteria [Figure 1]. The selected studies were concentrated between 2010 and 2020 and are considered current.

TABLE 1

Pharmacobotanical information, extracts and tests involving activity against H. pylori of flavonoids contained in plant species.

SpeciesFamilyExtracted partIdentified flavonoidsStudy modelPharmacological evaluationCountryReferences Qualea parviflora VochysiaceaeBark— In vitro Agar well diffusionBrazil Camellia sinensis TheaceaeLeavesCatechin; Epicatechin Epigallocatechin; Quercetin In vitro Agar diffusionUSA Hypericum erectum HypericaceaeWhole plantQuercetin-3′-O-β-D-galactopyranoside In vitro MICSouth Korea Bridelia micranta PhyllanthaceaeStem bark— In vitro Agar well diffusion, MIC, rate of killSouth Africa Byrsonima intermedi MalpighiaceaeLeavesCatechin; Epicatechin; Quercetin Quercetin-3-[2″-O-galloyl]-O-α-galactopyranoside; Quercetin-3-O-[2″-O-galloyl]-α- arabinopyranoside; Quercetin-3′-O-[2″-acetyl]-β-D-glucopyranoside; Quercetin-3-O-α-arabinopyranoside; Quercetin-3′-O-β-D-galactopyranoside 7,3′-di-O- methyleriodictyol In vitro MICBrazil Glycyrrhiza Glabra FabaceaeRootsGlabridin; Glabrol In vitro MICIndia Amygdalus communis RosaceaeFruitEpicatechin; Naringenin In vitro MICItaly Vitis rotundifolia VitaceaeFruit— In vitro Disk diffusionUSA Polygala cyparissias PolygalaceaeWhole plant— In vitro MICBrazil Lythrum salicaria LythraceaeLeaves, flowers and stem— In vitro Disk diffusionIran Caesalpinia pyramidalis LeguminosaeInner bark— In vitro MIC, MBCBrazil Hippocratea celastroides HippocrateaceaeLeaves, stems, and root bark— In vitro MICMexico Theobroma cacao MalvaceaeSeeds— In vitro MICNigeria Lippia integrifolia LythraceaeLeaves and flowersSalvagenin; 6-Hydroxyluteolin 7-hexoside; 6-Methoxyluteolin-hexoside 6-Methylscutellarein 7-hexoside B-ring-dimethoxylated Flavone- hexoside; Methoxylated apigenin-hexoside In vitro Agar diffusionArgentina Cuphea aequipetala LythraceaeLeaves and flowers— In vitro MICBrazil Peumus boldus MonimiaceaeLeavesCatechin; Epicatechin In vitro MICChile Solanum cernuum SolanaceaeLeavesAfzelin; Quercitrin In vitro MIC, MBCBrazil Copaifera malmei FabaceaeLeavesRutin; Catechin; Quercetin In vitro MICBrazil Parthenium hysterophorus AsteraceaeRoots— In vitro MICMexico Lithraea molleoides AnacardaceaeLeavesRutin In vitro MICArgentina Syzygium aromaticum; Piper nigrum; Cuminum cyminum; Salvia officinalis; Punica granatum; Zingiber officinale; Commiphora myrrha; Glycyrrhiza glabra Myrtaceae; Piperaceae; Apiaceae; Lamiaceae; Punicaceae; Zingiberaceae; Burseraceae FabaceaeFlowers; Fruit; Seeds; Leaves; Peel; Roots; Resin; RootsCatechin In vitro MICEgypt Maytenus robusta CelastraceaeLeaves— In vitro MICBrazil Leonotis nepetifolia LamiaceaeWhole plantKaempferol; Morin; Myricetin; Naringin; Naringenin; Quercetin; Rutin In vitro MIC, MBCBrazil Piper umbellatum PiperaceaeLeaves— In vitro MICBrazil Euphorbia umbellata EuphorbiaceaeBark— In vitro Disk diffusionBrazil Rosa hybrida RosaceaeFlowers— In vitro MICKorea Agrimonia eupatoria; Fragaria vesca RosaceaeLeaves and stems; flowers and fruit— In vitro MICPortugal Heterotheca inuloides AsteraceaeLeaves, stems and flowersQuercetin; 7,3′-di-O- methyleriodictyol In vitro MICMexico Anoda cristata; Cnidoscolus aconitifolius; Crotalaria pumila Malvaceae; Euphorbiaceae; FabaceaeLeavesAcacetin; Diosmetin In vitro MICMexico Desmostachya bipinnata PoaceaeLeaves and flowers— In vitro MICSaudi Arabia Oryza sativa PoaceaeGrain— In vitro Western blottingSouth Korea Ixeris chinensis AsteraceaeWhole plantKaempferol; Luteolin; Myricetin; Naringenin; Naringin; Rutin In vitro Disk diffusionTaiwan Physalis alkekengi SolanaceaeLeaves and flowersKaempferol; Quercetin In vitro MICChina Cannabis sativa CannabaceaeFlowersCatechin; Epicatechin; Naringenin; Naringin; Quercetin; Rutin In vitro MIC, MBCItaly Cochlospermum regium CochlospermaceaeLeavesKaempferol; Morin; Myricetin; Rutin In vitro MICBrazil Azadirachta indica MeliaceaeFruit and seeds— In vitro MICItaly Virola elongata MyristicaceaeStems— In vitro MICBrazil Byrsonima intermedia MalpighiaceaeLeavesCatechin; Epicatechin; Quercetin In vitro MICBrazil Diospyros virginiana EbenaceaePedicels— In vitro MICSouth Korea Casearia sylvestris SalicaceaeLeaves— In vitro MICBrazil Plectranthus barbatus LamiaceaeLeavesLuteolin; Quercetin In vitro MIC, MBCBrazil Berberis aristata BerberidaceaeStems— In vitro Disk diffusionIndia Erythrina speciosa FabaceaeLeaves— In vitro MICEgypt Alpinia Officinarum ZingiberaceaeRhizomesApigenin; Galangin; Galangin-3-methylether; Kaempferol; Kaempferide; Pinobaksin; Ponocembrin; Quercetin; Quercetin-3-methylether; Salvagenin In vivo H. pylori-associated gastritis [HAG] model [mice]China

![An external file that holds a picture, illustration, etc. Object name is fphar-13-951125-g001.jpg][////i0.wp.com/www.ncbi.nlm.nih.gov/pmc/articles/PMC9470917/bin/fphar-13-951125-g001.jpg]

Flowchart of article selection for the systematic review. The bibliographic study started with 1.773 articles. After applying the eligibility criteria, 567 remained. Four hundred thirty-nine were excluded after reading the title, 25 excluded by repetition, and 59 excluded after full reading. Forty-four articles fit the purpose and were selected for this review.

There was variability in the study regions for the selected manuscripts, with 25% of the papers coming from Asia [; ; ; ; ; ; ; ; ; ; ], 56.82% from America, of which 40.91% were from South America [; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ] and 15.91% from North America [; ; ; ; ; ; ], 9.09% of the articles originated from the Africa [; ; ; ], and 9,09% from the Europe [; ; ; ].

Among the Asian countries, South Korea represented 9.09% of the publications [; ; ; ], India [; ] and China [; ] both 4.55%, Iran [], Saudi Arabia [], and Taiwan [] 2.27%. In South America, Brazil [; ; ; ; ; ; ; ; ; ; ; ; ; ; ] represented 34.09%, Argentina [; ] 4.55%, and Chile [] 2.27%. In North America, Mexico [; ; ; ; ] represented 11.36% and the United States [; ] 4.55%. In the Africa, Egypt [; ] represented 4.55%, Nigeria [] and South Africa [] both 2.27%. In the Europe, Italy [; ; ] represented 6.82% and Portugal [] 2.27% of publications.

The plant families used in the studies were Anacardaceae [], Apiaceae [], Asteraceae [; ; ], Berberidaceae [], Burseraceae [], Cannabaceae [], Celastraceae [], Cochlospermaceae [], Ebenaceae [], Euphorbiaceae [; ], Fabaceae [; ; ; ; ], Hippocrateaceae [], Hypericaceae [], Lamiaceae [], Leguminosae [], Lythraceae [; ; ], Malpighiaceae [; ], Malvaceae [; ], Meliaceae [], Monimiaceae [], Myristicaceae [], Myrtaceae [], Phyllanthaceae [], Piperaceae [; ], Poaceae [; ], Polygalaceae [], Rosaceae [; ; ], Salicaceae [], Solanaceae [; ], Theaceae [], Vitaceae [], Vochysiaceae [] and Zingiberaceae [; ]. The most prominent family in these studies was the Fabaceae.

Between the flavonoids identified in the extracts [Table 2], quercetin [; ; ; ; ; ; ; ; ; ], catechin [; ; ; ; ; ; ], epicatechin [; ; ; ; ; ], rutin [; ; ; ; ; ], kaempferol [; ; ; ; ], naringenin [; ; ; ], naringin [; ; ], luteolin [; ; ], myricetin [; ; ], morin [; ], and quercetin-3′-O-β-D-galactopyranoside [; ] stood out.

TABLE 2

Main flavonoids identified in the articles included in this review.

The tests for determination of antimicrobial activity included the evaluation of minimum inhibitory concentration [MIC] [; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ; ], minimum bactericidal concentration [MBC] [; ; ; ; ], disk diffusion [; ; ; ; ], well agar diffusion [; ], agar diffusion [; ], rate of kill [], and H. pylori-associated gastritis [HAG] in vivo model [mice] [].

Discussion

The use of natural products and synthetic variations of their structures is the primary source of novel chemical entities approved as drugs by federal regulatory agencies. Despite the significant advance in combinatorial chemistry, discovering new active compounds through exclusively synthetic routes does not fulfill the role of presenting itself as a primary source of therapeutic innovation. In silico analysis has been used as an optimization tool to identify natural compounds as a valuable alternative to the pharmaceutical industry [].

Natural products and their derivatives represent more than a third of all newly discovered molecular entities approved by the FDA [Food and Drugs Administration]. Notably, about 25% are of plant origin []. Sixty-four percent of these compounds have been used to treat neoplastic diseases. For example, of the 126 drugs discovered between 1981 and 2019, 78 [48%] are natural products for antibacterials.

Flavonoids

The pharmacological potential of medicinal plants is given by the chemical structures produced by secondary plant metabolism. It presents several biosynthesis mechanisms capable of supplying substances with complex chemical structures. In addition, these structures are responsible for specialized intrinsic functions, favoring the activity in biological environments. They generally possess pharmacophoric regions, which are intricate to create or reproduce through organic synthesis [].

Flavonoids are among the main classes of secondary metabolites with pharmacological relevance []. These phenolic compounds act on plants as adaptive agents, playing a crucial role in the survival of species against environmental stresses and in response to invasions by microorganisms. This natural function of flavonoids explains the growing interest in studying these compounds in searching for new drugs with antimicrobial activity, mainly due to the lack of effective therapies in the current clinical scenario [].

Moreover, flavonoids represent one of the most important and diversified phenolic groups among natural metabolites. Its occurrence is often associated with the color of plants, and it is frequently found in flowers, fruits, leaves, stems, and seeds []. The word “Flavonoid” derives from the Latin “Flavus,” which means light yellow. However, the color of the flavonoid often varies according to the species. The basic skeleton of the flavonoids [Figure 2] has a tricyclic structure with 15 carbon atoms, with a chromatic ring [A] fused to a pinane ring [C] connected to an aromatic ring [B], leading to the subcategories of flavonoids [; ; ].

![An external file that holds a picture, illustration, etc. Object name is fphar-13-951125-g002.jpg][////i0.wp.com/www.ncbi.nlm.nih.gov/pmc/articles/PMC9470917/bin/fphar-13-951125-g002.jpg]

A basic chemical skeleton of flavonoids. General structural representation of a flavonoid, characteristic of 15 carbon atoms in its basic skeleton, corresponding to two benzene rings [A and B] linked by a pyran ring [C].

Literature reports have shown that flavonoids play a role in plant survival by preventing the spread of fungal and bacterial pathogens [; ]. Furthermore, flavonoid biosynthesis is closely related to defense responses in plant tissues. Significantly, it interferes in the vascular strands of leaves, which are most exposed and susceptible to contamination []. These findings support the hypothesis that flavonoids are potential antimicrobial agents that may be effective against human pathogens.

Due to the great diversity of compounds within the flavonoid group, the highly concentrated extracts exhibit more diverse mechanisms of antimicrobial activity. In other words, they target more components and functions of bacterial cells than other plant secondary metabolites [; ]. Other advantages of flavonoid-rich extracts with antimicrobial action compared to others are summarized in Table 3.

TABLE 3

Advantages of flavonoid-rich extracts with antimicrobial activities compared to other extracts.

Catechin and Epicatechin

Catechins, such as catechin and epicatechin, are flavonoids in plants, fruits [e.g., apple, strawberry, kiwi], black and green tea, red wine, beer, chocolate, and cocoa, among others []. For example, green tea has a high catechin concentration [], about 1 g/ml in a teacup []. Approximately 5–7% of this concentration is epicatechin [].

Catechins’ antimicrobial activity is related to the interaction of this compound with the cell wall and inner membrane of bacteria and hydrogen peroxide production. One of the proposed mechanisms of action is related to the formation of high molecular mass complexes between this compound and proteins on the surface of the bacterial cell wall. It interrupts substrate transit between the intra and extracellular environment, inhibiting bacterial cell activity [].

Some studies report catechin’s antimicrobial activity against gram-positive and gram-negative bacteria. It includes species like Escherichia coli [E. coli] [], H. pylori [], Staphylococcus aureus [S. aureus] [; ], and Bacillus subtilis [B. subtilis] [].

Kaempferol

Kaempferol is a natural flavonol present in many edible plants [e.g., broccoli, cabbage, beans, tomatoes, and strawberries] as well as in traditional medicine [e.g., Ginkgo biloba L. [Ginkgoaceae]; Moringa oleífera Lamarck [Moringaceae] []. This compound and its glycosides possess several pharmacological properties, such as antioxidant, anti-inflammatory, antidiabetic, anticancer, cardioprotective, neuroprotective, anti-steroidal, anxiolytic, estrogenic/anti-estrogenic, analgesic, anti-allergic and antimicrobial [; ].

Kaempferol’s antimicrobial activity may be associated with its ability to form complexes with the bacterial cell wall, which causes the inhibition of microbial growth []. In addition, the compound blocked the formation of S. aureus biofilm in the initial adhesion stage []. This inhibition probably occurs due to the inhibition of enzymes responsible for the beginning of biofilm formation and promoting suppression of the expression of genes of some surface proteins involved in adhesion.

In addition to S. aureus, kaempferol and its glycosides have reported activity against H. pylori [], E. coli [], Pseudomonas aeruginosa [P. aeruginosa] [], Vibrio parahaemolyticus [V. parahaemolyticus], Bacillus cereus [B. cereus], Bacillus licheniformis [B. licheniformis] [], and Enterococcus faecalis [E. faecalis] [].

Luteolin

Luteolin is a flavone naturally found in its glycosylated structure in many edible plant species [e.g., carrot, pepper, peppermint, and oregano] []. Its pharmacological activities include antioxidant, anti-inflammatory, neuroprotective, anticancer, antidiabetic, and antimicrobial [; ].

Luteolin demonstrates antimicrobial activity against the uropathogenic E. coli [UPEC] strain []. The compound reduces UPEC adhesion to urinary epithelium cells by decreasing the expression of adhesion proteins in the microorganism’s fimbriae. Furthermore, Luteolin reduced the expression of adhesion-related genes and increased the hydrophilicity, inhibiting biofilm formation.

Other studies also describe the activities of luteolin against H. pylori [], S. aureus [; ; ], Listeria monocytogenes [L. monocytogenes] [], P. aeruginosa, B. cereus, and Salmonella typhimurium [S. typhimurium] [].

Morin

Morin is a flavone in many fruits and plants of the Moraceae and Myrtaceae families, such as Maclura pomifera [Rafinesque] C. K. Schneider [Moraceae]; Maclura tinctoria L. D. Don ex Steudel. [Moraceae]; Psidium guajava L. [Myrtaceae]; and Morus alba L. [Moraceae] [; ; ]. This flavonoid is attributed to antioxidant, anti-inflammatory, antidiabetic, antihistamine, antitumor, antihypertensive, antiuricemic, neuroprotective, antiviral, and antimicrobial activities [; ].

The antimicrobial activity of morin was demonstrated against Listeria monocytogenes can be explained by two mechanisms []. Firstly, it reduces biofilm formation by inhibiting microbial motility and adhesion and compromising cell-surface and cell-cell interactions. The second mechanism is the interruption of listeriolysin O secretion. It reduces the pathogenicity of L. monocytogenes in epithelial cells and macrophages.

Morin acts against S. aureus [], B. cereus, Salmonella enteritidis [S. enteritidis] [], E. coli [], and H. pylori [].

Myricetin

Myricetin is a flavone encountered in many fruits, vegetables, teas, berries, and red wine []. Biological activities attributed to this flavone include hypoglycemic [], anti-inflammatory [], anticarcinogenic, and antiviral [], and antimicrobial [].

This compound reduces the expression of genes that encode some virulence factors responsible for bacterial colonization. Moreover, it inactivates host defenses, tissue damage, and nutrient uptake genes in pathogenic strains of Porphyromonas gingivalis [P. gingivalis] []. Moreover, myricetin activity was demonstrated against E. coli [], S. aureus, and Proteus vulgaris [P. vulgaris] [], and H. pylori [].

Naringenin and Naringin

The flavanone naringenin and its glycoside [naringin] are abundant in the peels of citrus fruits, mainly grapefruit and orange []. They present antioxidant, antidiabetic [], anti-inflammatory [], hypolipemic, antihypertensive, and antifibrotic [], and antimicrobial [] activities.

Naringenin and naringin act against Salmonella enteritidis []. The study reported the synergism of these grapefruit juice components with the acidic pH generated. In combination, these factors reduced the adhesion of S. enteritidis by inhibiting the bacterium’s acid tolerating response mechanism.

Moreover, other studies reported the activity of naringenin and naringin against E. coli, P. aeruginosa [], Proteus mirabilis [P. mirabilis], Acinetobacter baumannii [A. baumannii], Klebsiella pneumonia [K. pneumoniae], S. aureus, B. subtilis, E. faecalis [], and H. pylori [].

Quercetin

Quercetin is a bioflavonoid obtained from various plant sources [e.g., apple, onion, citrus fruits, and vegetables] [; ]. It has several biological activities, such as antioxidant, anti-inflammatory, anticancer [], antiviral [], and antimicrobial [].

Feasible antimicrobial mechanisms of action of quercetin have been described [] in S. aureus and E. coli. Quercetin can initiate the peroxidation of the outer lipid membrane in gram-negative bacteria, such as E. coli. It compromises the integrity of the bacterial cell barrier, leading to cell lysis. Additionally, in gram-positive bacteria, quercetin causes oxidative stress and activates the kynurenine pathway. It depletes L-tryptophan reserves, leading to a reduction in bacterial growth.

Studies also report quercetin activity against P. aeruginosa, P. vulgaris [], P. mirabilis, A. baumannii, K. pneumoniae, E. faecalis, B. subtilis [] and H. pylori [].

Hyperoside and Rutin

Hyperoside [quercetin-3′-O-β-D-galactopyranoside] and rutin [quercetin-3-O-rutinoside] are quercetin glycosides found in vegetables, citrus fruits, and berries. They exhibit several biological activities, such as anti-inflammatory, antithrombotic, antidiabetic, hepatoprotective, antioxidant, antihistamine, antitumor, antiplatelet, antihypertensive, antispasmodic, antiprotozoal, and antimicrobial [; ].

Rutin can inhibit biofilm formation in E. coli and S. aureus []. The probable mechanism of this activity is related to reducing the production of exopolysaccharides. These molecules are responsible for biofilms’ higher resistance to antimicrobials than planktonic cultures and protect the biofilm by forming multiple layers on its surface that aid in adhesion.

Additionally, hyperoside and rutin activities against P. aeruginosa [; ], Serratia marcescens [S. marcescens] [], E. faecalis [], Actinomyces viscosus [A. viscosus], Actinomyces naeslundii [A. naeslundii] [], S. pyogenes [] and H. pylori [] are also described.

Use of Extracts Containing Flavonoids Against H. pylori and Its Advantages

Defined as a group I carcinogen since 1994 by the International Agency for Research on Cancer, H. pylori represents about 5% of the total burden of all cancers worldwide [; ]. In addition, H. pylori is associated with diseases such as chronic gastritis, peptic ulcer, and gastric mucosa-associated lymphoid tissue lymphoma. This bacterium has a urease enzyme capable of converting the urea present in gastric acid into ammonia, increasing the stomach pH, thus allowing its colonization. Its silent permanence results in chronic inflammation and, consequently, in the appearance of gastritis and ulcers that can lead to gastric perforation. Thereupon, the epithelial tissue begins to undergo metaplasia. The metaplastic cells start the process of uncontrolled division, which undergoes gene mutation and culminate in the formation of malignant neoplastic tissue [Figure 3] [; ].

![An external file that holds a picture, illustration, etc. Object name is fphar-13-951125-g003.jpg][////i0.wp.com/www.ncbi.nlm.nih.gov/pmc/articles/PMC9470917/bin/fphar-13-951125-g003.jpg]

General overview of H. pylori-associated gastric cancer development. According to its topology, gastric cancer can be classified into adenocarcinoma, lymphoma, carcinoid tumor, and leiomyosarcoma. Adenocarcinoma is the type of gastric cancer most affecting the population, subdivided into [1] intestinal and [2] diffuse. Type [1] is mainly elicited by an acute immune response induced by H. pylori infection. The silent persistence of the bacteria provides a picture of chronic inflammation, consequently inducing gastritis and ulcers that can lead to gastric perforation. As a result, epithelial tissue undergoes metaplasia [cell differentiation] and behaves like intestinal cells, losing function. Metaplastic cells begin a process of disordered division by undergoing gene mutation that ends up in the formation of malignant neoplastic tissue. Type [2] adenocarcinoma is caused by genetic factors that affect the expression of the intercellular adhesion proteins. For example, E-cadherin is responsible for keeping gastric epithelial cells interconnected and controls the cell cycle.

Among the current treatment strategies for patients with H. pylori-associated gastritis and peptic ulcer disease, triple therapy is based on combinations of multiple agents, including bismuth subsalicylate, proton pump inhibitors, H2 blockers, and antibiotics, mainly clarithromycin. Additionally, the eradication of H. pylori is indicated for treating lymphoma. Also, other regimens used as adjunctive therapy include probiotics, bovine lactoferrin, and curcumin [].

As expected, the application of multiple drugs is associated with several side effects, making it impossible for the patient to adhere to and abandon the course of treatment. In addition, these interferents can lead to the emergence of resistant strains [].

Therefore, the search for new anti-H. pylori therapy alternatives promoted the exploration in the field of medicinal plants. As a result, several studies have been conducted, and many natural products have anti-H. pylori mechanisms of action proven, such as urease inhibition, DNA damage, protein synthesis inhibition, and anti-inflammatory effects. Moreover, they inhibit some enzymes, such as dihydrofolate reductase and myeloperoxidase N-acetyltransferase [Figure 4] [].

![An external file that holds a picture, illustration, etc. Object name is fphar-13-951125-g004.jpg][////i0.wp.com/www.ncbi.nlm.nih.gov/pmc/articles/PMC9470917/bin/fphar-13-951125-g004.jpg]

Mechanisms of action of flavonoids against H. pylori. The pathogenesis of H. pylori depends on several virulence factors, including urease. This enzyme neutralizes the acidic pH of the gastric medium and facilitates bacteria adhesion to the epithelium. Flavonoids can block this process once they inhibit urease activity. Flavonoids also act by inhibiting enzymes crucial for the reproduction and survival of H. pylori, such as DNA gyrase and ATPase. In addition, it induces ionic and metabolic imbalances at the cytoplasmic level, causing bacterial cell wall disruption, which leads to cell death or latency. Once adhered to the gastric epithelium, H. pylori induces the release of proinflammatory cytokines and the generation of specific genetic mutations which promote cell apoptosis. Flavonoids have anti-inflammatory activities. They inhibit MAPK and NF-kB pathways and regulate the oxidative stress response in phagocytes and other cells. In addition, some flavonoids act as antioxidant agents, scavenging free radicals and reestablishing the ionic balance. The Establishment of adenocarcinoma from successive gene mutations caused by chronic inflammation and the inhibition of any stage of this process can attenuate, delay or prevent the onset of gastric cancer.

Preparations containing physiologically active constituents in folk medicine support the therapeutic use of flavonoids. Several pharmacological actions include antimicrobial, antitumor, anti-inflammatory, and antioxidant []. For example, flavonoids are effective against different microorganisms by different mechanisms of action. They cause membrane disruption, antibiofilm action, cell envelope synthesis, nucleic acid synthesis, electron transport chain and ATP synthesis, formation of flavonoid-metal complexes, inhibition of bacterial toxins, and others [].

As mentioned above, pathogenic bacteria may resist antibiotic drugs through different mechanisms. For example, mechanisms might be the prevention of interaction of the drug with the target, efflux of the antibiotic from the cell, and direct destruction or modification of the drug compound. Moreover, bacteria can interchange resistance genes with surrounding colonies. For example, the β-lactamase gene encodes an enzyme that hydrolyzes the amide bond in the β-lactam ring through transformation [incorporation of naked DNA], transduction [phage-mediated], and conjugation. Gram-negative bacteria interfere with β-lactam ring hydrolysis, whereas Gram-positive bacteria modify the target site of antibiotics [; ].

Occasionally the resistance to antimicrobial agents can be obtained via combined mechanisms. For instance, although gentamicin resistance does not rely on antibiotic modification, it is executed by altering the membrane potential and efflux and 16S rRNA methylation [].

Cistus laurifolius L. [Cistaceae] extract has proved anti-H. pylori, including against resistant strains []. Moreover, the extract of Glycyrrhiza glabra L. [Fabaceae] maintained its action against H. pylori even after being clinically used without any drug resistance []. Such findings infer how flavonoids may be helpful as lead compounds in the development of a new class of anti-H. pylori regimens.

Possible Mechanisms of Action of Flavonoids Against H. pylori

The urease enzyme is considered a potent H. pylori virulence factor. It comprises 6% of the synthesized proteins, representing a significant energy investment in colonization. Moreover, its inhibition hinders the survival of H. pylori in the gastric environment, which becomes hostile due to high acidity []. In vitro experiments demonstrated that the flavonoid quercetin, present in Heterotheca inuloides Cassini [Asteraceae] extract, promotes high enzyme inhibition [IC50 = 132.4 μg/ml] []. Additionally, a complimentary in silico study of molecular docking has identified that quercetin interacts with the catalytic site, forming ionic bonds with the zinc cation. Ionic bonds are among the strongest, supporting and inducing high enzyme inhibition activity. Catechin and epicatechin isolated from the Peumus boldus extract inhibit urease activity, with IC50 values of 66 and 112 µg GAE/ml, respectively []. However, due to molecules’ sizes and high polarity, they seem to neutralize only the most external pool of urease. It means they could not inhibit the urease cytoplasmic pool, suggesting a more preventive application.

Glabridin and glabrol present in the extract of Glycyrrhiza glabra L. [GutGard®] inhibit the dihydrofolate reductase [DHFR] []. DHFR consists of a ubiquitous enzyme in every eukaryotic and prokaryotic cell that plays a crucial role in the synthesis of thymidine. It catalyzes the reduction of 7,8-dihydrofolate to 5,6,7,8-tetrahydrofolate, using NADPH as a cofactor. This reaction is an essential step in the biosynthesis of DNA nucleotide bases and, therefore, plays an essential role in bacteria survival, such as H. pylori in the human body. It is interesting to note that in addition to the anti-H. pylori activity, GutGard® also has anti-inflammatory [] and antioxidant [].

The potential activity of the luteolin-rich extract from Plectranthus barbatus [Andrews] Benth. ex G.Don [Lamiaceae] has been proved []. The subinhibitory dose of ethyl acetate [EAF] fraction [128 μg/ml] produced similar morphological changes as did the subinhibitory dose of amoxicillin [0.25 μg/ml]. The filamentous cells found and the production of protrusions indicate a possible action on the bacterial cell wall. These data suggest a possible action on PBPs [Penicillin-Binding Proteins], which are involved in the cell septation process, especially the 63 kDa PBP [PBP63]. Besides, observations in producing protrusions and blebs suggest a possible action on other PBPs involved in the peptidoglycan wall.

When assessing the intracellular accumulation of quercetin from Vitis rotundifolia Michaux [Vitaceae], it was observed a behavior possibly interspersed in the hydrophobic region of the lipid bilayers of the cell envelope, passively diffused through the cell membrane into the cytosol []. It may be an interaction with cell membrane proteins or active import into the cytosol, leading to metabolic imbalance, followed by cell death.

Catechin, epicatechin, epigallocatechin, and quercetin flavonoids in white, green, oolong, and black teas deplete membrane electrons responsible for the transport chain []. They disrupt oxidative phosphorylation and inhibit the proton efflux linked to dehydrogenase. Thereby, they interfere with the flow of electrons at the cytochrome level. However, low hydrophobic and simple soluble phenolic compounds may not be effective. The outer lipopolysaccharide layer of H. pylori avoids oxidative phosphorylation. As a result, the membrane creates a hydrophobic microenvironment along the bacterial surface. Also, soluble phenols interrupt the H+-adenosine triphosphatase required to synthesize adenosine triphosphate. It causes hyperacidification via proton donation in the plasma membrane or in the intracellular cytosolic pathway. Another explanation is that the hydrophobic portion of polyphenols adheres to the cell wall. It would cause destabilization and rupture of the membrane and inhibition of transmembrane transport. These mechanisms can act synergistically: the disturbing and destabilizing effect of polyphenols can make it easier for simple soluble phenols to exercise their hyperacidification.

Alpinia officinarum Hance [Zingiberaceae] extracts possess the flavonoids apigenin, galangin, galangin-3-methyl ether, kaempferol, kaempferide, pinobaksin, pinocembrin, quercetin, quercetin-3-methyl ether, and salvagenin []. To such metabolites has been attributed the anti-H. pylori activity by inhibiting the synthesis of the proinflammatory cytokine interleukin-8 [IL-8] via the MAPK pathway, whose gene shows a significant increase in expression in the entire genome of gastric epithelial cells after infection by H. pylori. This reduction would result in decreased inflammation and adhesion of the bacteria to the epithelium.

Moreover, the Lippia integrifolia [Griseb.] Hieronymus [Verbenaceae] extract demonstrated the flavonoids salvagenin, 6-Hydroxyluteolin 7-hexoside, 6-Methoxyluteolin-hexoside, 6-Methylscutellarein 7-hexoside, B-ring-dimethoxylated Flavone-hexoside and Methoxylated apigenin-hexoside []. As a result, it exhibited strong antioxidant capacity in vitro. Furthermore, it inhibited H. pylori adhesion to stomach cells by up to 40%. In comparison, the ethanol-soluble fraction showed up to 60% inhibition rates. Furthermore, the decoction significantly increased the gastric adenocarcinoma cell line [AGS] cell viability at> 10 μg/ml without influencing the proliferation rate. Besides, H. pylori-induced IL-8 secretion was significantly reduced by the coincubation of AGS cells with extracts.

Study Relevance

Here, the activity of flavonoid extracts was investigated against H. pylori for gastric cancer prevention and treatment. These studies provided a basis for further investigations that may lead to new clinical trials for proper drug administration. In this perspective, the use of botanical drugs might present as a viable and safe alternative.

Administration of botanic products is a complementary treatment for acute and chronic diseases and preventive care. GutGard® [] is an example currently used to help control indigestion and heartburn and manage H. pylori infection at a dosage of 150 mg/day []. So, using extracts containing flavonoid compounds might be highly relevant to eradicating H. pylori as a curative and preventive therapeutic strategy.

Furthermore, botanical drugs are cost-effective compared to synthetic medicines and processed plant derivates []. The focus on the herbal products research encourages the investigation of alternative therapeutic modalities for long-standing and persistent health problems.

Limitations

Studies using pure isolated flavonoids were rare, which required the inclusion of additional extracts in our protocol. Few publications mentioned the mechanism of action of flavonoids on H. pylori. Additionally, many articles identified their flavonoids but not their concentration. Furthermore, some studies did not use full botanical taxonomic names, limiting comparative analysis and data translation. Finally, only one in vivo study was included in this review, limiting the analytical power of the effectiveness of the flavonoid in the live organism.

Future Directions

This review on flavonoids’ antimicrobial and anticancer action is in line with recent literature [; ; ]. Altogether, data demonstrate the great potential of these compounds in combating H. pylori infection and gastric cancer protection. The immense variety of plants that serve as a source of flavonoids can ensure the sustainable production of drugs containing the compounds. In addition, the great potential of these compounds for various health applications is observed. New flavonoids’ extraction methods might allow for better profitability and sustainability. Moreover, dosage control and compound stability offered by smart drug delivery systems can further expand the understanding of the activities of flavonoid-rich extracts against various pathogens [; ].

Furthermore, a broader understanding of the interactions of flavonoids with drugs of choice for the treatment of H. pylori may help determine the use of the compounds as adjuvant therapy in the fight against the development of gastric cancer. Also, it is imperative to evaluate the effect of flavonoids on the human microbiome and how the metabolic process of the colonizing microbes might interfere with the activities of the compounds against pathogenic strains. Clinical trials testing the action of these compounds in patients with H. pylori infection deserve attention.

Conclusion

There is no single therapy for H. pylori eradication. Instead, an association of an antiulcerogenic drug and two antibiotics with 70–85% success rates. Our data demonstrated the relevance of new studies involving natural products, specifically extracts of plants rich in flavonoids. These compounds have shown promising results in anti-H. pylori targeting different mechanisms of action.

Acknowledgments

Thanks to CAPES, UFPB, and UEPB for the most valuable support given to this research.

Author Contributions

All authors contributed to the development of the article. RI, DF, GG, and WR held the bibliographic search and selection of articles. MP and DF performed the methodology of the article. RI, DF, SA, and GG discussed the articles. RI and GG were in charge of generating the tables and figures. BG, SC, SW, and LC were responsible for the general review of the content.

Funding

Funds supported this study from the Brazilian Federal Foundation for Support and Evaluation of Graduate Education [CAPES] - Finance Code 001 [fellowships to SC and LC].

Conflict of Interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Publisher’s Note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

References

  • Abreu Miranda M., Lemos M., Alves Cowart K., Rodenburg D., D McChesney J., Radwan M. M., et al. [2015]. Gastroprotective Activity of the Hydroethanolic Extract and Isolated Compounds from the Leaves of Solanum Cernuum Vell. J. Ethnopharmacol. 172, 421–429. 10.1016/j.jep.2015.06.047 [PubMed] [CrossRef] [Google Scholar]
  • Abreu Miranda M., Lemos M., Alves Cowart K., Rodenburg D., D. McChesney J., Radwan M. M., et al. [2015]. Gastroprotective Activity of the Hydroethanolic Extract and Isolated Compounds from the Leaves of Solanum Cernuum Vell. J. Ethnopharmacol. 172, 421–429. 10.1016/j.jep.2015.06.047 [PubMed] [CrossRef] [Google Scholar]
  • Adamczak A., Ożarowski M., Karpiński T. M. [2019]. Antibacterial Activity of Some Flavonoids and Organic Acids Widely Distributed in Plants. J. Clin. Med. 9, 109. 10.3390/jcm9010109[PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Adeyemi O. S., Ebugosi C., Akpor O. B., Hetta H. F., Al-Rashed S., Otohinoyi D. A., et al. [2020]. Quercetin Caused Redox Homeostasis Imbalance and Activated the Kynurenine Pathway [Running Title: Quercetin Caused Oxidative Stress]. Biol. [Basel] 9, 1–9. 10.3390/biology9080219[PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Adzu B., Balogun S. O., Pavan E., Ascêncio S. D., Soares I. M., Aguiar R. W., et al. [2015]. Evaluation of the Safety, Gastroprotective Activity and Mechanism of Action of Standardised Leaves Infusion Extract of Copaifera Malmei Harms. J. Ethnopharmacol. 175, 378–389. 10.1016/j.jep.2015.09.027 [PubMed] [CrossRef] [Google Scholar]
  • Ahmad A., Kaleem M., Ahmed Z., Shafiq H. [2015]. Therapeutic Potential of Flavonoids and Their Mechanism of Action against Microbial and Viral Infections-A Review. Food Res. Int. 77, 221–235. 10.1016/j.foodres.2015.06.021 [CrossRef] [Google Scholar]
  • Al-Ishaq R. K., Liskova A., Kubatka P., Büsselberg D. [2021]. Enzymatic Metabolism of Flavonoids by Gut Microbiota and its Impact on Gastrointestinal Cancer. Cancers [Basel] 13. 10.3390/cancers13163934[PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Al-Numair K. S., Chandramohan G., Alsaif M. A., Veeramani C., El Newehy A. S. [2014]. Morin, a Flavonoid, on Lipid Peroxidation and Antioxidant Status in Experimental Myocardial Ischemic Rats. Afr. J. Tradit. Complement. Altern. Med. 11, 14–20. 10.4314/ajtcam.v11i3.3[PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Al-Shabib N. A., Husain F. M., Ahmad I., Khan M. S., Khan R. A., Khan J. M. [2017]. Rutin Inhibits Mono and Multi-Species Biofilm Formation by Foodborne Drug Resistant Escherichia coli and Staphylococcus aureus. Food control. 79, 325–332. 10.1016/j.foodcont.2017.03.004 [CrossRef] [Google Scholar]
  • Albino S. L., da Silva J. M., de C NobreNobre M. S. M. S., de M E Silvae Silva Y. M. S. Y. M. S., Santos M. B., de Araújo R. S. A., et al. [2020]. Bioprospecting of Nitrogenous Heterocyclic Scaffolds with Potential Action for Neglected Parasitosis: A Review. Curr. Pharm. Des. 26, 4112–4150. 10.2174/1381612826666200701160904 [PubMed] [CrossRef] [Google Scholar]
  • Almeida G. V. B., Arunachalam K., Balogun S. O., Pavan E., Ascêncio S. D., Soares I. M., et al. [2019]. Chemical Characterization and Evaluation of Gastric Antiulcer Properties of the Hydroethanolic Extract of the Stem Bark of Virola Elongata [Benth.] Warb. J. Ethnopharmacol. 231, 113–124. 10.1016/j.jep.2018.11.011 [PubMed] [CrossRef] [Google Scholar]
  • Almeida G. V. B., Arunachalam K., Balogun S. O., Pavan E., Ascêncio S. D., Soares I. M., et al. [2019]. Chemical Characterization and Evaluation of Gastric Antiulcer Properties of the Hydroethanolic Extract of the Stem Bark of Virola Elongata [Benth.] Warb. J. Ethnopharmacol. 231, 113–124. 10.1016/j.jep.2018.11.011 [PubMed] [CrossRef] [Google Scholar]
  • Amin M. U., Khurram M., Khattak B., Khan J. [2015]. Antibiotic Additive and Synergistic Action of Rutin, Morin and Quercetin against Methicillin Resistant Staphylococcus aureus. BMC Complement. Altern. Med. 15, 59–12. 10.1186/s12906-015-0580-0[PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Ankolekar C., Johnson D., Pinto Mda S., Johnson K., Labbe R., Shetty K. [2011]. Inhibitory Potential of Tea Polyphenolics and Influence of Extraction Time against helicobacter Pylori and Lack of Inhibition of Beneficial Lactic Acid Bacteria. J. Med. Food 14, 1321–1329. 10.1089/jmf.2010.0237 [PubMed] [CrossRef] [Google Scholar]
  • Arima H., Danno G. [2002]. Isolation of Antimicrobial Compounds from Guava [Psidium Guajava l.] and Their Structural Elucidation. Biosci. Biotechnol. Biochem. 66, 1727–1730. 10.1271/bbb.66.1727 [PubMed] [CrossRef] [Google Scholar]
  • Arunachalam K., Damazo A. S., Pavan E., Oliveira D. M., Figueiredo F. F., Machado M. T. M., et al. [2019]. Cochlospermum Regium [Mart. Ex Schrank] Pilg.: Evaluation of Chemical Profile, Gastroprotective Activity and Mechanism of Action of Hydroethanolic Extract of its Xylopodium in Acute and Chronic Experimental Models. J. Ethnopharmacol. 233, 101–114. 10.1016/j.jep.2019.01.002 [PubMed] [CrossRef] [Google Scholar]
  • Asha M. K., Debraj D., Prashanth D., Edwin J. R., Srikanth H. S., Muruganantham N., et al. [2013]. In Vitro anti-Helicobacter pylori Activity of a Flavonoid Rich Extract of Glycyrrhiza Glabra and its Probable Mechanisms of Action. J. Ethnopharmacol. 145, 581–586. 10.1016/j.jep.2012.11.033 [PubMed] [CrossRef] [Google Scholar]
  • Ay M., Charli A., Jin H., Kanthasamy A., Kanthasamy A. G. [2016]. Muhammet Ay, Adhithiya Charli, Huajun Jin, Vellareddy Anantharam, Arthi Kanthasamy and Anumantha G. Kanthasamy. 447–452. [Google Scholar]
  • Babiaka S. B., Nia R., Abuga K. O., Mbah J. A., Nziko V. d. P. N., Paper D. H., et al. [2020]. Antioxidant Potential of Flavonoid Glycosides from Manniophyton Fulvum Müll. [Euphorbiaceae]: Identification and Molecular Modeling. Sci. Afr. 8, e00423. 10.1016/j.sciaf.2020.e00423 [CrossRef] [Google Scholar]
  • Bae E. A., Han M. J., Kim D. H. [1999]. In Vitro Anti-Helicobacter pylori Activity of Some Flavonoids and Their Metabolites. Planta Med. 65, 442–443. 10.1055/s-2006-960805 [PubMed] [CrossRef] [Google Scholar]
  • Bailly C. [2020]. Molecular and Cellular Basis of the Anticancer Activity of the Prenylated Flavonoid Icaritin in Hepatocellular Carcinoma. Chem. Biol. Interact. 325, 109124. 10.1016/j.cbi.2020.109124 [PubMed] [CrossRef] [Google Scholar]
  • Baker D. A. [2020]. Plants against Helicobacter pylori to Combat Resistance: An Ethnopharmacological Review. Biotechnol. Rep. [Amst] 26, e00470. 10.1016/j.btre.2020.e00470[PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Baliga M. S., Shivashankara A. R., Venkatesh S., Bhat H. P., Palatty P. L., Bhandari G., et al. [2019]. Phytochemicals in the Prevention of Ethanol-Induced Hepatotoxicity. Diet. Interv. Liver Dis. Foods, Nutr. Diet. Suppl., 79–89. 10.1016/B978-0-12-814466-4.00007-0 [CrossRef] [Google Scholar]
  • Batra P., Sharma A. K. [2013]. Anti-cancer Potential of Flavonoids: Recent Trends and Future Perspectives. 3 Biotech. 3, 439–459. 10.1007/s13205-013-0117-5[PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Beck S., Stengel J. [2016]. Mass Spectrometric Imaging of Flavonoid Glycosides and Biflavonoids in Ginkgo Biloba L. Phytochemistry 130, 201–206. 10.1016/j.phytochem.2016.05.005 [PubMed] [CrossRef] [Google Scholar]
  • Bernal-Mercado A. T., Vazquez-Armenta F. J., Tapia-Rodriguez M. R., Islas-Osuna M. A., Mata-Haro V., Gonzalez-Aguilar G. A., et al. [2018]. Comparison of Single and Combined Use of Catechin, Protocatechuic, and Vanillic Acids as Antioxidant and Antibacterial Agents against Uropathogenic Escherichia coli at Planktonic and Biofilm Levels. Molecules 23. 10.3390/molecules23112813[PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Biharee A., Sharma A., Kumar A., Jaitak V. [2020]. Antimicrobial Flavonoids as a Potential Substitute for Overcoming Antimicrobial Resistance. Fitoterapia 146, 104720. 10.1016/j.fitote.2020.104720 [PubMed] [CrossRef] [Google Scholar]
  • Bisignano C., Filocamo A., La Camera E., Zummo S., Fera M. T., Mandalari G. [2013]. Antibacterial Activities of Almond Skins on cagA-Positive And-Negative Clinical Isolates of Helicobacter pylori. BMC Microbiol. 13, 103. 10.1186/1471-2180-13-103[PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Borges A. S., Minozzo B. R., Santos H., Ardisson J. S., Rodrigues R. P., Romão W., et al. [2020]. Plectranthus Barbatus Andrews as Anti-Helicobacter pylori Agent with Activity against Adenocarcinoma Gastric Cells. Industrial Crops Prod. 146, 112207. 10.1016/j.indcrop.2020.112207 [CrossRef] [Google Scholar]
  • Botten D., Fugallo G., Fraternali F., Molteni C. [2015]. Structural Properties of Green Tea Catechins. J. Phys. Chem. B 119, 12860–12867. 10.1021/acs.jpcb.5b08737 [PubMed] [CrossRef] [Google Scholar]
  • Braicu C., Ladomery M. R., Chedea V. S., Irimie A., Berindan-Neagoe I. [2013]. The Relationship between the Structure and Biological Actions of Green Tea Catechins. Food Chem. 141, 3282–3289. 10.1016/j.foodchem.2013.05.122 [PubMed] [CrossRef] [Google Scholar]
  • Brown J. C., Jiang X. [2013]. Activities of Muscadine Grape Skin and Polyphenolic Constituents against Helicobacter pylori. J. Appl. Microbiol. 114, 982–991. 10.1111/jam.12129 [PubMed] [CrossRef] [Google Scholar]
  • Bush K., Fisher J. F. [2011]. Epidemiological Expansion, Structural Studies, and Clinical Challenges of New β-lactamases from Gram-Negative Bacteria. Annu. Rev. Microbiol. 65, 455–478. 10.1146/annurev-micro-090110-102911 [PubMed] [CrossRef] [Google Scholar]
  • Bush K. [2013]. The ABCD's of β-lactamase Nomenclature. J. Infect. Chemother. 19, 549–559. 10.1007/s10156-013-0640-7 [PubMed] [CrossRef] [Google Scholar]
  • Calixto J. B. [2019]. The Role of Natural Products in Modern Drug Discovery. An. Acad. Bras. Cienc. 91 Suppl 3, e20190105–7. 10.1590/0001-3765201920190105 [PubMed] [CrossRef] [Google Scholar]
  • Cardoso O., Donato M. M., Luxo C., Almeida N., Liberal J., Figueirinha A., et al. [2018]. Anti- Helicobacter pylori potential of Agrimonia eupatoria L. and Fragaria vesca. J. Funct. Foods 44, 299–303. 10.1016/j.jff.2018.03.027 [CrossRef] [Google Scholar]
  • Casas-Grajales S., Muriel P. [2017]. The Liver, Oxidative Stress, and Antioxidants. Liver Pathophysiol. Ther. Antioxidants, 583–604. 10.1016/B978-0-12-804274-8.00043-6 [CrossRef] [Google Scholar]
  • Caselli A., Cirri P., Santi A., Paoli P. [2016]. Morin: A Promising Natural Drug. Curr. Med. Chem. 23, 774–791. 10.2174/0929867323666160106150821 [PubMed] [CrossRef] [Google Scholar]
  • Céliz G., Daz M., Audisio M. C. [2011]. Antibacterial activity of naringin derivatives against pathogenic strains. J. Appl. Microbiol. 111, 731–738. 10.1111/j.1365-2672.2011.05070.x [PubMed] [CrossRef] [Google Scholar]
  • Cesa S., Sisto F., Zengin G., Scaccabarozzi D., Kokolakis A. K., Scaltrito M. M., et al. [2019]. Phytochemical analyses and pharmacological screening of Neem oil. South Afr. J. Bot. 120, 331–337. 10.1016/j.sajb.2018.10.019 [CrossRef] [Google Scholar]
  • Chandrasekaran C. V., Deepak H. B., Thiyagarajan P., Kathiresan S., Sangli G. K., Deepak M., et al. [2011]. Dual inhibitory effect of Glycyrrhiza glabra [GutGard™] on COX and LOX products. Phytomedicine 18, 278–284. 10.1016/j.phymed.2010.08.001 [PubMed] [CrossRef] [Google Scholar]
  • Cho M. H., Lee S. W. [2015]. Phenolic phytoalexins in rice: Biological functions and Biosynthesis. Int. J. Mol. Sci. 16, 29120–29133. 10.3390/ijms161226152[PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Cui Y., Chen J., He Z., Xiao Y. [2013]. SUZ12 depletion suppresses the proliferation of gastric cancer cells. Cell. Physiol. biochem. 31, 778–784. 10.1159/000350095 [PubMed] [CrossRef] [Google Scholar]
  • Cushnie T. P., Lamb A. J. [2005]. Antimicrobial activity of flavonoids. Int. J. Antimicrob. Agents 26, 343–356. 10.1016/j.ijantimicag.2005.09.002[PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Cushnie T. P., Lamb A. J. [2011]. Recent advances in understanding the antibacterial properties of flavonoids. Int. J. Antimicrob. Agents 38, 99–107. 10.1016/j.ijantimicag.2011.02.014 [PubMed] [CrossRef] [Google Scholar]
  • da Silva Junior I. F., Balogun S. O., de Oliveira R. G., Damazo A. S., Martins D. T. O. [2016]. Piper umbellatum L.: A medicinal plant with gastric-ulcer protective and ulcer healing effects in experimental rodent models. J. Ethnopharmacol. 192, 123–131. 10.1016/j.jep.2016.07.011 [PubMed] [CrossRef] [Google Scholar]
  • da Silva L. M., Boeing T., Somensi L. B., Cury B. J., Steimbach V. M., Silveria A. C., et al. [2015]. Evidence of gastric ulcer healing activity of Maytenus robusta Reissek: In Vitro and In Vivo studies. J. Ethnopharmacol. 175, 75–85. 10.1016/j.jep.2015.09.006 [PubMed] [CrossRef] [Google Scholar]
  • Das S., Das M. K., Das R., Gehlot V., Mahant S., Mazumder P. M., et al. [2020]. Isolation, characterization of Berberine from Berberis aristata DC for eradication of resistant Helicobacter pylori. Biocatal. Agric. Biotechnol. 26, 101622. 10.1016/j.bcab.2020.101622 [CrossRef] [Google Scholar]
  • de Cássia dos Santos R., Bonamin F., Périco L. L., Rodrigues V. P., Zanatta A. C., Rodrigues C. M., et al. [2019]. Byrsonima intermedia A. Juss partitions promote gastroprotection against peptic ulcers and improve healing through antioxidant and anti-inflammatory activities. Biomed. Pharmacother. 111, 1112–1123. 10.1016/j.biopha.2018.12.132 [PubMed] [CrossRef] [Google Scholar]
  • de Lima Nascimento T. R., de Amoêdo Campos Velo M. M., Silva C. F., Costa Cruz S. B. S., Gondim B. L. C., Mondelli R. F. L., et al. [2019]. Current Applications of Biopolymer-based Scaffolds and Nanofibers as Drug Delivery Systems. Curr. Pharm. Des. 25, 3997–4012. 10.2174/1381612825666191108162948 [PubMed] [CrossRef] [Google Scholar]
  • de Martel C., Forman D., Plummer M. [2013]. Gastric cancer: epidemiology and risk factors. Gastroenterol. Clin. North Am. 42, 219–240. 10.1016/j.gtc.2013.01.003 [PubMed] [CrossRef] [Google Scholar]
  • del Valle P., García-Armesto M. R., de Arriaga D., González-Donquiles C., Rodríguez-Fernández P., Rúa J. [2016]. Antimicrobial activity of kaempferol and resveratrol in binary combinations with parabens or propyl gallate against Enterococcus faecalis. Food control. 61, 213–220. 10.1016/j.foodcont.2015.10.001 [CrossRef] [Google Scholar]
  • Díaz-Gómez R., López-Solís R., Obreque-Slier E., Toledo-Araya H. [2013]. Comparative antibacterial effect of gallic acid and catechin against Helicobacter pylori. LWT - Food Sci. Technol. 54, 331–335. 10.1016/j.lwt.2013.07.012 [CrossRef] [Google Scholar]
  • Ding S. Z., Goldberg J. B., Hatakeyama M. [2010]. Helicobacter pylori infection, oncogenic pathways and epigenetic mechanisms in gastric carcinogenesis. Future Oncol. 6, 851–862. 10.2217/fon.10.37[PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Dong H., Yang X., He J., Cai S., Xiao K., Zhu L. [2017]. Enhanced antioxidant activity, antibacterial activity and hypoglycemic effect of luteolin by complexation with Manganese[II] and its inhibition kinetics on xanthine oxidase. RSC Adv. 7, 53385–53395. 10.1039/c7ra11036g [CrossRef] [Google Scholar]
  • Dormán G., Flachner B., Hajdú I., András C. D. [2016]. Target identification and polypharmacology of nutraceuticals. Nutraceuticals Effic. Saf. Toxic., 263–286. 10.1016/B978-0-12-802147-7.00021-8 [CrossRef] [Google Scholar]
  • Eddouks M., Bidi A., Bouhali B. E. L., Zeggwagh N. A. [2014]. Insulin resistance as a target of some plant-derived phytocompounds. Stud. Nat. Prod. Chem. 43, 351–373. 10.1016/B978-0-444-63430-6.00011-4 [CrossRef] [Google Scholar]
  • Egas V., Salazar-Cervantes G., Romero I., Méndez-Cuesta C. A., Rodríguez-Chávez J. L., Delgado G. [2018]. Anti-Helicobacter pylori metabolites from Heterotheca inuloides [Mexican arnica]. Fitoterapia 127, 314–321. 10.1016/j.fitote.2018.03.001 [PubMed] [CrossRef] [Google Scholar]
  • Escandón R. A., del Campo M., López-Solis R., Obreque-Slier E., Toledo H. [2016]. Antibacterial effect of kaempferol and [−]-epicatechin on Helicobacter pylori. Eur. Food Res. Technol. 242, 1495–1502. 10.1007/s00217-016-2650-z [CrossRef] [Google Scholar]
  • Espinosa-Rivero J., Rendón-Huerta E., Romero I. [2015]. Inhibition of Helicobacter pylori growth and its colonization factors by Parthenium hysterophorus extracts. J. Ethnopharmacol. 174, 253–260. 10.1016/j.jep.2015.08.021 [PubMed] [CrossRef] [Google Scholar]
  • Fahmy N. M., Al-Sayed E., Michel H. E., El-Shazly M., Singab A. N. B. [2020]. Gastroprotective effects of Erythrina speciosa [Fabaceae] leaves cultivated in Egypt against ethanol-induced gastric ulcer in rats. J. Ethnopharmacol. 248, 112297. 10.1016/j.jep.2019.112297 [PubMed] [CrossRef] [Google Scholar]
  • Fathima A., Rao J. R. [2016]. Selective toxicity of Catechin-a natural flavonoid towards bacteria. Appl. Microbiol. Biotechnol. 100, 6395–6402. 10.1007/s00253-016-7492-x [PubMed] [CrossRef] [Google Scholar]
  • Ferreira T. S., Moreira C. Z., Cária N. Z., Victoriano G., Silva Jr W. F., Magalhães J. C. [2014]. Phytotherapy: an introduction to its history, use and application. Rev. Bras. Plantas Med. 16, 290–298. 10.1590/S1516-05722014000200019 [CrossRef] [Google Scholar]
  • Fukai T., Marumo A., Kaitou K., Kanda T., Terada S., Nomura T. [2002]. Anti-Helicobacter pylori flavonoids from licorice extract. Life Sci. 71, 1449–1463. 10.1016/S0024-3205[02]01864-7 [PubMed] [CrossRef] [Google Scholar]
  • Garro M. F., Salinas Ibáñez A. G., Vega A. E., Arismendi Sosa A. C., Pelzer L., Saad J. R., et al. [2015]. Gastroprotective effects and antimicrobial activity of Lithraea molleoides and isolated compounds against Helicobacter pylori. J. Ethnopharmacol. 176, 469–474. 10.1016/j.jep.2015.11.009 [PubMed] [CrossRef] [Google Scholar]
  • Ge L., Li J., Wan H., Zhang K., Wu W., Zou X., et al. [2018]. NMR data for novel flavonoids from Lonicera japonica flower buds. Data Brief. 21, 2192–2207. 10.1016/j.indcrop.2018.08.07310.1016/j.dib.2018.11.021[PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Gomez-Chang E., Uribe-Estanislao G. V., Martinez-Martinez M., Gálvez-Mariscal A., Romero I. [2018]. Anti-helicobacter pylori potential of three edible plants known as quelites in Mexico. J. Med. Food 21, 1150–1157. 10.1089/jmf.2017.0137 [PubMed] [CrossRef] [Google Scholar]
  • Gondim B. L. C., Oshiro-Júnior J. A., Fernanandes F. H. A., Nóbrega F. P., Castellano L. R. C., Medeiros A. C. D. [2019]. Plant Extracts Loaded in Nanostructured Drug Delivery Systems for Treating Parasitic and Antimicrobial Diseases. Curr. Pharm. Des. 25, 1604–1615. 10.2174/1381612825666190628153755 [PubMed] [CrossRef] [Google Scholar]
  • González A., Casado J., Lanas Á. [2021]. Fighting the Antibiotic Crisis: Flavonoids as Promising Antibacterial Drugs Against Helicobacter pylori Infection. Front. Cell. Infect. Microbiol. 11, 709749. 10.3389/fcimb.2021.709749[PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • González A., Salillas S., Velázquez-Campoy A., Espinosa Angarica V., Fillat M. F., Sancho J., et al. [2019]. Identifying potential novel drugs against Helicobacter pylori by targeting the essential response regulator HsrA. Sci. Rep. 9, 11294. 10.1038/s41598-019-47746-9[PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Górniak I., Bartoszewski R., Króliczewski J. [2019]. Comprehensive Review of Antimicrobial Activities of Plant Flavonoids, 18, 241–272. 10.1007/s11101-018-9591-zComprehensive review of antimicrobial activities of plant flavonoids Phytochem. Rev. [CrossRef] [Google Scholar]
  • Grenier D., Chen H., Ben Lagha A., Fournier-Larente M. P. [2015]. Dual action of myricetin on Porphyromonas gingivalis and the inflammatory response of host cells: A promising therapeutic molecule for periodontal diseases. PLoS One 10, e0131758–15. 10.1371/journal.pone.0131758[PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Grzesik M., Naparło K., Bartosz G., Sadowska-Bartosz I. [2018]. Antioxidant properties of catechins: Comparison with other antioxidants. Food Chem. 241, 480–492. 10.1016/j.foodchem.2017.08.117 [PubMed] [CrossRef] [Google Scholar]
  • Gutiérrez-Venegas G., Gómez-Mora J. A., Meraz-Rodríguez M. A., Flores-Sánchez M. A., Ortiz-Miranda L. F. [2019]. Effect of flavonoids on antimicrobial activity of microorganisms present in dental plaque. Heliyon 5, e03013. 10.1016/j.heliyon.2019.e03013[PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Hamad G. M., Taha T. H., El-Deeb N. M., Alshehri A. M. [2015]. Advanced trends in controlling Helicobacter pylori infections using functional and therapeutically supplements in baby milk. J. Food Sci. Technol. 52, 8156–8163. 10.1007/s13197-015-1875-3[PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Herrero R., Park J. Y., Forman D. [2014]. The fight against gastric cancer - The IARC Working Group report. Best. Pract. Res. Clin. Gastroenterol. 28, 1107–1114. 10.1016/j.bpg.2014.10.003 [PubMed] [CrossRef] [Google Scholar]
  • Hinojosa W. I., Quiróz M. A., Álvarez I. R., Castañeda P. E., Villarreal M. L., Taketa A. C. [2014]. Anti-Helicobacter pylori, gastroprotective, anti-inflammatory, and cytotoxic activities of methanolic extracts of five different populations of Hippocratea celastroides collected in Mexico. J. Ethnopharmacol. 155, 1156–1163. 10.1016/j.jep.2014.06.044 [PubMed] [CrossRef] [Google Scholar]
  • Hooi J. K. Y., Lai W. Y., Ng W. K., Suen M. M. Y., Underwood F. E., Tanyingoh D., et al. [2017]. Global Prevalence of Helicobacter pylori Infection: Systematic Review and Meta-Analysis. Gastroenterology 153, 420–429. 10.1053/j.gastro.2017.04.022 [PubMed] [CrossRef] [Google Scholar]
  • Horwitz R. J. [2018]. “Chapter 30 - The Allergic Patient,” in Integrative Medicine [Fourth Edition]. Editor Rakel D. [Amsterdam, Netherlands: Elsevier; ], 300–300.e2. [Google Scholar]
  • Ibrahim N. H., Awaad A. S., Alnafisah R. A., Alqasoumi S. I., El-Meligy R. M., Mahmoud A. Z. [2018]. In - Vitro activity of Desmostachya bipinnata [L.] Stapf successive extracts against Helicobacter pylori clinical isolates. Saudi Pharm. J. 26, 535–540. 10.1016/j.jsps.2018.02.002[PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Imran M., Rauf A., Abu-Izneid T., Nadeem M., Shariati M. A., Khan I. A., et al. [2019]. Luteolin, a flavonoid, as an anticancer agent: A review. Biomed. Pharmacother. 112, 108612. 10.1016/j.biopha.2019.108612 [PubMed] [CrossRef] [Google Scholar]
  • Jadeja R. N., Devkar R. V. [2014]. Polyphenols and Flavonoids in Controlling Non-Alcoholic Steatohepatitis. Polyphenols Hum. Heal. Dis. 1, 615–623. 10.1016/B978-0-12-398456-2.00047-5 [CrossRef] [Google Scholar]
  • Jaisinghani R. N. [2017]. Antibacterial properties of quercetin. Microbiol. Res. [Pavia] 8. 10.4081/mr.2017.6877 [CrossRef] [Google Scholar]
  • Jeong C.-S. [2009]. Evaluation for protective effect of rutin, a natural flavonoid, against Hcl/ethanol-induced gastric lesions. Biomol. Ther. 17, 199–204. 10.4062/biomolther.2009.17.2.199 [CrossRef] [Google Scholar]
  • Jesus R. S., Piana M., Freitas R. B., Brum T. F., Alves C. F. S., Belke B. V., et al. [2018]. In Vitro antimicrobial and antimycobacterial activity and HPLC-DAD screening of phenolics from Chenopodium ambrosioides L. Braz J. Microbiol. 49, 296–302. 10.1016/j.bjm.2017.02.012[PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Ji H. F., Li X. J., Zhang H. Y. [2009]. Natural products and drug discovery. Can thousands of years of ancient medical knowledge lead us to new and powerful drug combinations in the fight against cancer and dementia? EMBO Rep. 10, 194–200. 10.1038/embor.2009.12[PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Joseph Sahayarayan J., Udayakumar R., Arun M., Ganapathi A., Alwahibi M. S., Aldosari N. S., et al. [2020]. Effect of different Agrobacterium rhizogenes strains for in-vitro hairy root induction, total phenolic, flavonoids contents, antibacterial and antioxidant activity of [Cucumis anguria L.]. Saudi J. Biol. Sci. 27, 2972–2979. 10.1016/j.sjbs.2020.08.050[PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Joung D. K., Lee Y. S., Han S. H., Lee S. W., Cha S. W., Mun S. H., et al. [2016]. Potentiating activity of luteolin on membrane permeabilizing agent and ATPase inhibitor against methicillin-resistant Staphylococcus aureus. Asian pac. J. Trop. Med. 9, 19–22. 10.1016/j.apjtm.2015.12.004 [PubMed] [CrossRef] [Google Scholar]
  • Jucá M. M., Cysne Filho F. M. S., de Almeida J. C., Mesquita D. D. S., Barriga J. R. M., Dias K. C. F., et al. [2020]. Flavonoids: biological activities and therapeutic potential. Nat. Prod. Res. 34, 692–705. 10.1080/14786419.2018.1493588 [PubMed] [CrossRef] [Google Scholar]
  • Kim H. W., Woo H. J., Yang J. Y., Kim J.-B., Kim S.-H. [2021]. Hesperetin Inhibits Expression of Virulence Factors and Growth of Helicobacter pylori. Ijms 22, 10035. 10.3390/ijms221810035[PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Kim S.-H., Lee M. H., Park M., Woo H. J., Kim Y. S., Tharmalingam N., et al. [2018]. Regulatory Effects of Black Rice Extract on Helicobacter pylori Infection-Induced Apoptosis. Mol. Nutr. Food Res. 62, 1700586. 10.1002/mnfr.201700586 [PubMed] [CrossRef] [Google Scholar]
  • Klein-Júnior L. C., Santin J. R., Lemos M., Silveira A. C., Rocha J. A., Beber A. P., et al. [2013]. Role of gastric mucus secretion, oxinitrergic system and sulfhydryl groups on the gastroprotection elicited by Polygala cyparissias [Polygalaceae] in mice. J. Pharm. Pharmacol. 65, 767–776. 10.1111/jphp.12038 [PubMed] [CrossRef] [Google Scholar]
  • Kopacz M., Woźnicka E., Gruszecka J. [2016]. Antibacterial activity of morin and its complexes with La[III], Gd[III] and Lu[III] ions. Acta Pol. Pharm. 62, 65–67. [PubMed] [Google Scholar]
  • Krzyżek P., Migdał P., Paluch E., Karwańska M., Wieliczko A., Gościniak G. [2021]. Myricetin as an Antivirulence Compound Interfering with a Morphological Transformation into Coccoid Forms and Potentiating Activity of Antibiotics against Helicobacter pylori. Int. J. Mol. Sci. 22, 2695. 10.3390/ijms22052695[PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Kumar S., Pandey A. K. [2013]. Chemistry and biological activities of flavonoids: An overview. ScientificWorldJournal 2013, 162750. 10.1155/2013/162750[PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Ladeira M. S. P., Salvadori D. M. F., Rodrigues M. A. M. [2003]. Biopatologia Do Helicobacter pylori. J. Bras. Patol. Med. Lab. 39, 335–342. 10.1590/s1676-24442003000400011 [CrossRef] [Google Scholar]
  • Lahlou M. [2013]. The Success of Natural Products in Drug Discovery. Pp 04, 17–31. 10.4236/pp.2013.43a003 [CrossRef] [Google Scholar]
  • Lamb A., Chen L. F. [2013]. Role of the Helicobacter pylori-Induced inflammatory response in the development of gastric cancer. J. Cell.Biochem. 114, 491–497. 10.1002/jcb.24389[PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Lauren P. [1965]. THE TWO HISTOLOGICAL MAIN TYPES OF GASTRIC CARCINOMA: DIFFUSE AND SO-CALLED INTESTINAL-TYPE CARCINOMA. AN ATTEMPT AT A HISTO-CLINICAL CLASSIFICATION. Acta Pathol. Microbiol. Scand. 64, 31–49. 10.1111/apm.1965.64.1.31 [PubMed] [CrossRef] [Google Scholar]
  • Lawal T. O., Olorunnipa T. A., Adeniyi B. A. [2014]. Susceptibility testing and bactericidal activities of Theobroma cacao Linn. [cocoa] on Helicobacter pylori in an In Vitro study. J. Herb. Med. 4, 201–207. 10.1016/j.hermed.2014.09.004 [CrossRef] [Google Scholar]
  • Lee H. M., Hong E., Jeon B. Y., Kim D. U., Byun J. S., Lee W., et al. [2006]. Crystallization and preliminary X-ray crystallographic study of HP1043, a Helicobacter pylori orphan response regulator. Biochim. Biophys. Acta 1764, 989–991. 10.1016/j.bbapap.2005.10.024 [PubMed] [CrossRef] [Google Scholar]
  • Li C., Li X., Jiang Z., Wang D., Sun L., Li J., et al. [2022]. Flavonoids Inhibit Cancer by Regulating the Competing Endogenous RNA Network. Front. Oncol. 12, 842790. 10.3389/fonc.2022.842790[PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Liberati A., Altman D. G., Tetzlaff J., Mulrow C., Gøtzsche P. C., Ioannidis J. P., et al. [2009]. The PRISMA statement for reporting systematic reviews and meta-analyses of studies that evaluate health care interventions: Explanation and elaboration. PLoS Med. 6, e1000100. 10.1371/journal.pmed.1000100[PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Liu C., Huang H., Zhou Q., Liu B., Wang Y., Li P., et al. [2020]. Pithecellobium clypearia extract enriched in gallic acid and luteolin has antibacterial activity against MRSA and reduces resistance to erythromycin, ceftriaxone sodium and levofloxacin. J. Appl. Microbiol. 129, 848–859. 10.1111/jam.14668 [PubMed] [CrossRef] [Google Scholar]
  • Loo V. G., Fallone C. A., De Souza E., Lavallée J., Barkun A. N. [1997]. In-vitro susceptibility of Helicobacter pylori to ampicillin, clarithromycin, metronidazole and omeprazole. J. Antimicrob. Chemother. 40, 881–883. 10.1093/jac/40.6.881 [PubMed] [CrossRef] [Google Scholar]
  • Lu M.-C., Chiu H.-F., Lin C.-P., Shen Y.-C., Venkatakrishnan K., Wang C.-K. [2018]. Anti- Helicobacter pylori effect of various extracts of ixeris chinensis on inflammatory markers in human gastric epithelial AGS cells. J. Herb. Med. 11, 60–70. 10.1016/j.hermed.2017.08.002 [CrossRef] [Google Scholar]
  • Ma J., Shen H., Kapesa L., Zeng S. [2016]. Lauren classification and individualized chemotherapy in gastric cancer. Oncol. Lett. 11, 2959–2964. 10.3892/ol.2016.4337[PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Ma Q., Wei R., Sang Z., Dong J. [2020a]. Structural characterization, neuroprotective and hepatoprotective activities of flavonoids from the bulbs of Heleocharis dulcis. Bioorg. Chem. 96, 103630. 10.1016/j.bioorg.2020.103630 [PubMed] [CrossRef] [Google Scholar]
  • Ma X., You P., Xu Y., Ye X., Tu Y., Liu Y., et al. [2020b]. Anti-Helicobacter pylori-associated gastritis effect of the ethyl acetate extract of Alpinia officinarum Hance through MAPK signaling pathway. J. Ethnopharmacol. 260, 113100. 10.1016/j.jep.2020.113100 [PubMed] [CrossRef] [Google Scholar]
  • Maleki S. J., Crespo J. F., Cabanillas B. [2019]. Anti-inflammatory effects of flavonoids. Food Chem. 299, 125124. 10.1016/j.foodchem.2019.125124 [PubMed] [CrossRef] [Google Scholar]
  • Manayi A., Khanavi M., Saeidnia S., Saiednia S., Azizi E., Mahmoodpour M. R., et al. [2013]. Biological activity and microscopic characterization of Lythrum salicaria L. Daru 21, 61–67. 10.1186/2008-2231-21-61[PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Maqbool M., Amin Dar M., Gani I., Ahmad Mir S., Khan M. [2019]. Herbal Medicines As An Alternative Source of Therapy: A Review. World J. Pharm. Pharm. Sci. 8, 374–380. 10.20959/wjpps20192-13108 [CrossRef] [Google Scholar]
  • Marcial G., Sendker J., Brandt S., De Lampasona M. P., Catalán C. A., De Valdez G. F., et al. [2014]. Gastroprotection as an example: Antiadhesion against Helicobacter pylori, anti-inflammatory and antioxidant activities of aqueous extracts from the aerial parts of Lippia integrifolia Hieron. J. Ethnopharmacol. 155, 1125–1133. 10.1016/j.jep.2014.06.039 [PubMed] [CrossRef] [Google Scholar]
  • Martins A., Vasas A., Viveiros M., Molnár J., Hohmann J., Amaral L. [2011]. Antibacterial properties of compounds isolated from Carpobrotus edulis. Int. J. Antimicrob. Agents 37, 438–444. 10.1016/j.ijantimicag.2011.01.016 [PubMed] [CrossRef] [Google Scholar]
  • Mazzolin L. P., Nasser A. L., Moraes T. M., Santos R. C., Nishijima C. M., Santos F. V., et al. [2010]. Qualea parviflora Mart.: An integrative study to validate the gastroprotective, antidiarrheal, antihemorragic and mutagenic action. J. Ethnopharmacol. 127, 508–514. 10.1016/j.jep.2009.10.005 [PubMed] [CrossRef] [Google Scholar]
  • Mbaveng A. T., Hamm R., Kuete V. [2014]. Harmful and Protective Effects of Terpenoids from African Medicinal Plants, 557–576. 10.1016/B978-0-12-800018-2.00019-4 [CrossRef] [Google Scholar]
  • McColl K. E. [2010]. Clinical practice. Helicobacter pylori infection. N. Engl. J. Med. 362, 1597–1604. 10.1056/NEJMcp1001110 [PubMed] [CrossRef] [Google Scholar]
  • medical, and editorial content Team [2018]. About Stomach Cancer. Am. Cancer Soc., 1–14. Available at: //www.cancer.org/content/dam/CRC/PDF/Public/8838.00.pdf . [Google Scholar]
  • Mendes D. B., Lemes L. S., Carlos R., Cruz S., Lúcia M., Castro L. De, et al. [2012]. Metabólitos secundários e hipertireodismo. J. Biotechnol. Biodivers. 3, 1–69. Available at: //www.infoteca.cnptia.embrapa.br/infoteca/handle/doc/951393 . [Google Scholar]
  • Ming D., Wang D., Cao F., Xiang H., Mu D., Cao J., et al. [2017]. Kaempferol inhibits the primary attachment phase of biofilm formation in Staphylococcus aureus. Front. Microbiol. 8, 1–11. 10.3389/fmicb.2017.02263[PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Minozzo B. R., Lemes B. M., Justo A. D. S., Lara J. E., Petry V. E. K., Fernandes D., et al. [2016]. Anti-ulcer mechanisms of polyphenols extract of Euphorbia umbellata [Pax] Bruyns [Euphorbiaceae]. J. Ethnopharmacol. 191, 29–40. 10.1016/j.jep.2016.06.032 [PubMed] [CrossRef] [Google Scholar]
  • Moher D., Liberati A., Tetzlaff J., Altman D. G., Altman D., Antes G., et al. [2009]. Preferred reporting items for systematic reviews and meta-analyses: The PRISMA statement. PLoS Med. 6, e1000097. 10.1371/journal.pmed.1000097[PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Moon H. I., Lee Y. C., Lee J. H. [2011]. Phenol glycosides with In Vitro anti-helicobacter pylori activity from hypericum erectum Thunb. Phytother. Res. 25, 1389–1391. 10.1002/ptr.3453 [PubMed] [CrossRef] [Google Scholar]
  • Mori A., Nishino C., Enoki N., Tawata S. [1987]. Antibacterial activity and mode of action of plant flavonoids against Proteus vulgaris and Staphylococcus aureus. Phytochemistry 26, 2231–2234. 10.1016/S0031-9422[00]84689-0 [CrossRef] [Google Scholar]
  • Moss S. F. [2017]. The Clinical Evidence Linking Helicobacter pylori to Gastric Cancer. Cell. Mol. Gastroenterol. Hepatol. 3, 183–191. 10.1016/j.jcmgh.2016.12.001[PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Mukherjee M., Bhaskaran N., Srinath R., Shivaprasad H. N., Allan J. J., Shekhar D., et al. [2010]. Anti-ulcer and antioxidant activity of GutGard. Indian J. Exp. Biol. 48, 269–274. [PubMed] [Google Scholar]
  • Nakayama M., Shimatani K., Ozawa T., Shigemune N., Tsugukuni T., Tomiyama D., et al. [2013]. A study of the antibacterial mechanism of catechins: Isolation and identification of Escherichia coli cell surface proteins that interact with epigallocatechin gallate. Food control. 33, 433–439. 10.1016/j.foodcont.2013.03.016 [CrossRef] [Google Scholar]
  • Nardone G. [2006]. “16 Role of Helicobacter pylori in Gastric Cancer,” in Handbook of Immunohistochemistry and in Situ Hybridization of Human Carcinomas, 205–220. 10.1016/S1874-5784[05]80078-9 [CrossRef] [Google Scholar]
  • Newman D. J., Cragg G. M. [2020]. Natural Products as Sources of New Drugs over the Nearly Four Decades from 01/1981 to 09/2019. J. Nat. Prod. 83, 770–803. 10.1021/acs.jnatprod.9b01285 [PubMed] [CrossRef] [Google Scholar]
  • Okeleye B. I., Bessong P. O., Ndip R. N. [2011]. Preliminary phytochemical screening and In Vitro anti-helicobacter pylori activity of extracts of the stem bark of bridelia micrantha [Hochst., Baill., Euphorbiaceae]. Molecules 16, 6193–6205. 10.3390/molecules16086193[PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Olekhnovich I. N., Vitko S., Chertihin O., Hontecillas R., Viladomiu M., Bassaganya-Riera J., et al. [2013]. Mutations to Essential Orphan Response Regulator HP1043 of Helicobacter pylori Result in Growth-Stage Regulatory Defects. Infect. Immun. 81, 1439–1449. 10.1128/IAI.01193-12[PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Olekhnovich I. N., Vitko S., Valliere M., Hoffman P. S. [2014]. Response to Metronidazole and Oxidative Stress Is Mediated through Homeostatic Regulator HsrA [HP1043] in Helicobacter pylori. J. Bacteriol. 196, 729–739. 10.1128/JB.01047-13[PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Oliveira D. M., Melo F. G., Balogun S. O., Flach A., De Souza E. C., De Souza G. P., et al. [2015]. Antibacterial mode of action of the hydroethanolic extract of Leonotis nepetifolia [L.] R. Br. involves bacterial membrane perturbations. J. Ethnopharmacol. 172, 356–363. 10.1016/j.jep.2015.06.027 [PubMed] [CrossRef] [Google Scholar]
  • Omar S. H. [2018]. Biophenols. Biophenols Impacts Prospects Anti-Alzheimer Drug Discov., 103–148. 10.1016/B978-0-12-809593-5.00004-5 [CrossRef] [Google Scholar]
  • Orditura M., Galizia G., Sforza V., Gambardella V., Fabozzi A., Laterza M. M., et al. [2014]. Treatment of gastric cancer. World J. Gastroenterol. 20, 1635–1649. 10.3748/wjg.v20.i7.1635[PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Özçelik B., Kartal M., Orhan I. [2011]. Cytotoxicity, antiviral and antimicrobial activities of alkaloids, flavonoids, and phenolic acids. Pharm. Biol. 49, 396–402. 10.3109/13880209.2010.519390 [PubMed] [CrossRef] [Google Scholar]
  • Palacios-Espinosa J. F., Arroyo-García O., García-Valencia G., Linares E., Bye R., Romero I. [2014]. Evidence of the anti-Helicobacter pylori, gastroprotective and anti-inflammatory activities of Cuphea aequipetala infusion. J. Ethnopharmacol. 151, 990–998. 10.1016/j.jep.2013.12.012 [PubMed] [CrossRef] [Google Scholar]
  • Pandey A. K., Kumar S. [2013]. Perspective on Plant Products as Antimicrobials Agents: A Review. Pharmacologia 4, 469–480. 10.5567/pharmacologia.2013.469.480 [CrossRef] [Google Scholar]
  • Park D., Shin K., Choi Y., Guo H., Cha Y., Kim S. H., et al. [2016]. Antimicrobial activities of ethanol and butanol fractions of white rose petal extract. Regul. Toxicol. Pharmacol. 76, 57–62. 10.1016/j.yrtph.2016.01.011 [PubMed] [CrossRef] [Google Scholar]
  • Pastene E., Parada V., Avello M., Ruiz A., García A. [2014]. Catechin-based Procyanidins from peumus boldus mol. aqueous extract inhibit helicobacter pylori urease and adherence to adenocarcinoma gastric cells. Phytother. Res. 28, 1637–1645. 10.1002/ptr.5176 [PubMed] [CrossRef] [Google Scholar]
  • Patel K., Patel D. K. [2019]. The Beneficial Role of Rutin, A Naturally Occurring Flavonoid in Health Promotion and Disease Prevention: A Systematic Review and Update. Bioact. Food as Diet. Interv. Arthritis Relat. Inflamm. Dis., 457–479. 10.1016/b978-0-12-813820-5.00026-x [CrossRef] [Google Scholar]
  • Patridge E., Gareiss P., Kinch M. S., Hoyer D. [2016]. An analysis of FDA-approved drugs: Natural products and their derivatives. Drug Discov. Today 21, 204–207. 10.1016/j.drudis.2015.01.009 [PubMed] [CrossRef] [Google Scholar]
  • Peek R. M. [20052005]. Orchestration of aberrant epithelial signaling by Helicobacter pylori CagA. Sci. STKE 2005, pe14–280. 10.1126/stke.2772005pe14 [PubMed] [CrossRef] [Google Scholar]
  • Pelliciari S., Pinatel E., Vannini A., Peano C., Puccio S., De Bellis G., et al. [2017]. Insight into the essential role of the Helicobacter pylori HP1043 orphan response regulator: genome-wide identification and characterization of the DNA-binding sites. Sci. Rep. 7, 41063. 10.1038/srep41063[PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Piasecka A., Jedrzejczak-Rey N., Bednarek P. [2015]. Secondary metabolites in plant innate immunity: Conserved function of divergent chemicals. New Phytol. 206, 948–964. 10.1111/nph.13325 [PubMed] [CrossRef] [Google Scholar]
  • Plummer M., de Martel C., Vignat J., Ferlay J., Bray F., Franceschi S. [2016]. Global burden of cancers attributable to infections in 2012: a synthetic analysis. Lancet Glob. Health 4, e609–16. 10.1016/S2214-109X[16]30143-7 [PubMed] [CrossRef] [Google Scholar]
  • Plummer M., Franceschi S., Vignat J., Forman D., De Martel C. [2015]. Global burden of gastric cancer attributable to Helicobacter pylori. Int. J. Cancer 136, 487–490. 10.1002/ijc.28999 [PubMed] [CrossRef] [Google Scholar]
  • Polk D. B., Peek R. M. [2010]. Helicobacter pylori: Gastric cancer and beyond. Nat. Rev. Cancer 10, 403–414. 10.1038/nrc2857[PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Puupponen-Pimia R., Nohynek L., Meier C., Kahkonen M., Heinonen M., Hopia A., et al. [2001]. Antimicrobial properties of phenolic compounds from berries. J. Appl. Microbiol. 90, 494–507. 10.1046/j.1365-2672.2001.01271.x [PubMed] [CrossRef] [Google Scholar]
  • Qian W., Liu M., Fu Y., Zhang J., Liu W., Li J., et al. [2020]. Antimicrobial mechanism of luteolin against Staphylococcus aureus and Listeria monocytogenes and its antibiofilm properties. Microb. Pathog. 142, 104056. 10.1016/j.micpath.2020.104056 [PubMed] [CrossRef] [Google Scholar]
  • Qiu J., Li H., Meng H., Hu C., Li J., Luo M., et al. [2011]. Impact of luteolin on the production of alpha-toxin by Staphylococcus aureus. Lett. Appl. Microbiol. 53, 238–243. 10.1111/j.1472-765X.2011.03098.x [PubMed] [CrossRef] [Google Scholar]
  • Rahardiyan D. [2019]. Antibacterial potential of catechin of tea [Camellia sinensis] and its applications. Food Res. 3, 1–6. 10.26656/fr.2017.3[1].097 [CrossRef] [Google Scholar]
  • Rashed K., Ćirić A., Glamočlija J., Soković M. [2014]. Antibacterial and antifungal activities of methanol extract and phenolic compounds from Diospyros virginiana L. Industrial Crops Prod. 59, 210–215. 10.1016/j.indcrop.2014.05.021 [CrossRef] [Google Scholar]
  • Ribeiro A. R., Diniz P. B., Estevam C. S., Pinheiro M. S., Albuquerque-Júnior R. L., Thomazzi S. M. [2013]. Gastroprotective activity of the ethanol extract from the inner bark of Caesalpinia pyramidalis in rats. J. Ethnopharmacol. 147, 383–388. 10.1016/j.jep.2013.03.023 [PubMed] [CrossRef] [Google Scholar]
  • Ribeiro L. H. L. [2019]. Análise dos programas de plantas medicinais e fitoterápicos no Sistema Único de Saúde [SUS] sob a perspectiva territorial. Ciênc. saúde coletiva 24, 1733–1742. 10.1590/1413-81232018245.15842017 [PubMed] [CrossRef] [Google Scholar]
  • Russo M., Moccia S., Spagnuolo C., Tedesco I., Russo G. L. [2020]. Roles of flavonoids against coronavirus infection. Chem. Biol. Interact. 328, 109211. 10.1016/j.cbi.2020.109211[PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Saldanha E., Saxena A., Kaur K., Kalekhan F., Venkatesh P., Fayad R., et al. [2019]. Polyphenols in the Prevention of Ulcerative Colitis. Diet. Interv. Gastrointest. Dis. Foods, Nutr. Diet. 2, 277–287. 10.1016/B978-0-12-814468-8.00023-5 [CrossRef] [Google Scholar]
  • Santos R. C., Kushima H., Rodrigues C. M., Sannomiya M., Rocha L. R., Bauab T. M., et al. [2012]. Byrsonima intermedia A. Juss.: Gastric and duodenal anti-ulcer, antimicrobial and antidiarrheal effects in experimental rodent models. J. Ethnopharmacol. 140, 203–212. 10.1016/j.jep.2011.12.008 [PubMed] [CrossRef] [Google Scholar]
  • Saravanakumar K., Chellia R., Hu X., Kathiresan K., Oh D. H., Wang M. H. [2019]. Eradication of Helicobacter pylori through the inhibition of urease and peptide deformylase: Computational and biological studies. Microb. Pathog. 128, 236–244. 10.1016/j.micpath.2019.01.001 [PubMed] [CrossRef] [Google Scholar]
  • Sathya S., Pandima Devi K. [2018]. The Use of Polyphenols for the Treatment of Alzheimer's Disease. Role Mediterr. Diet. Brain Neurodegener. Dis., 239–252. 10.1016/B978-0-12-811959-4.00015-8 [CrossRef] [Google Scholar]
  • Schaalan M., Mohamed W., Fathy S. [2020]. MiRNA-200c, MiRNA-139 and ln RNA H19; new predictors of treatment response in H-pylori- induced gastric ulcer or progression to gastric cancer. Microb. Pathog. 149, 104442. 10.1016/j.micpath.2020.104442 [PubMed] [CrossRef] [Google Scholar]
  • Sepulveda A. R. [2013]. Helicobacter, Inflammation, and Gastric Cancer. Curr. Pathobiol. Rep. 1, 9–18. 10.1007/s40139-013-0009-8[PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Shankar G. M., Antony J., Anto R. J. [2015]. Quercetin and Tryptanthrin: Two Broad Spectrum Anticancer Agents for Future Chemotherapeutic Interventions. Enzymes 37, 43–72. 10.1016/bs.enz.2015.05.001 [PubMed] [CrossRef] [Google Scholar]
  • Shen X. F., Ren L. B., Teng Y., Zheng S., Yang X. L., Guo X. J., et al. [2014]. Luteolin decreases the attachment, invasion and cytotoxicity of UPEC in bladder epithelial cells and inhibits UPEC biofilm formation. Food Chem. Toxicol. 72, 204–211. 10.1016/j.fct.2014.07.019 [PubMed] [CrossRef] [Google Scholar]
  • Shields M. [2017]. Chemotherapeutics. Chemotherapeutics, 295–313. 10.1016/b978-0-12-802104-0.00014-7 [CrossRef] [Google Scholar]
  • Shin J. E., Kim J. M., Bae E. A., Hyun Y. J., Kim D. H. [2005]. In Vitro Inhibitory Effect of Flavonoids on Growth, Infection and Vacuolation of Helicobacter pylori. Planta Med. 71, 197–201. 10.1055/s-2005-837816 [PubMed] [CrossRef] [Google Scholar]
  • Shivashankara A. R., Venkatesh S., Bhat H. P., Palatty P. L., Baliga M. S. [2015]. Can Phytochemicals be Effective in Preventing Ethanol-Induced Hepatotoxicity in the Geriatric Population? An Evidence-Based Revisit. Foods Diet. Suppl. Prev. Treat. Dis. Older Adults, 163–170. 10.1016/B978-0-12-418680-4.00017-8 [CrossRef] [Google Scholar]
  • Shukla R., Pandey V., Vadnere G. P., Lodhi S. [2019]. Role of Flavonoids in Management of Inflammatory Disorders. Bioact. Food as Diet. Interv. Arthritis Relat. Inflamm. Dis., 293–322. –322. 10.1016/b978-0-12-813820-5.00018-0 [CrossRef] [Google Scholar]
  • Sinsinwar S., Vadivel V. [2020]. Catechin isolated from cashew nut shell exhibits antibacterial activity against clinical isolates of MRSA through ROS-mediated oxidative stress. Appl. Microbiol. Biotechnol. 104, 8279–8297. 10.1007/s00253-020-10853-z [PubMed] [CrossRef] [Google Scholar]
  • Sivaranjani M., Gowrishankar S., Kamaladevi A., Pandian S. K., Balamurugan K., Ravi A. V. [2016]. Morin inhibits biofilm production and reduces the virulence of Listeria monocytogenes - An In Vitro and In Vivo approach. Int. J. Food Microbiol. 237, 73–82. 10.1016/j.ijfoodmicro.2016.08.021 [PubMed] [CrossRef] [Google Scholar]
  • Sivasothy Y., Sulaiman S. F., Ooi K. L., Ibrahim H., Awang K. [2013]. Antioxidant and antibacterial activities of flavonoids and curcuminoids from Zingiber spectabile Griff. Food control. 30, 714–720. 10.1016/j.foodcont.2012.09.012 [CrossRef] [Google Scholar]
  • Smyth E. C., Nilsson M., Grabsch H. I., van Grieken N. C., Lordick F. [2020]. Gastric cancer. Lancet 396, 635–648. 10.1016/S0140-6736[20]31288-5 [PubMed] [CrossRef] [Google Scholar]
  • Spósito L., Oda F. B., Vieira J. H., Carvalho F. A., dos Santos Ramos M. A., de Castro R. C., et al. [2019]. In Vitro and In Vivo anti-Helicobacter pylori activity of Casearia sylvestris leaf derivatives. J. Ethnopharmacol. 233, 1–12. 10.1016/j.jep.2018.12.032 [PubMed] [CrossRef] [Google Scholar]
  • Srinivasan S., Vinothkumar V., Murali R. [2019]. Antidiabetic Efficacy of Citrus Fruits With Special Allusion to Flavone Glycosides. Bioact. Food as Diet. Interv. Diabetes, 335–346. 10.1016/b978-0-12-813822-9.00022-9 [CrossRef] [Google Scholar]
  • Sun Y., Sun F., Feng W., Qiu X., Liu Y., Yang B., et al. [2017]. Hyperoside inhibits biofilm formation of Pseudomonas aeruginosa. Exp. Ther. Med. 14, 1647–1652. 10.3892/etm.2017.4641[PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Sung H., Ferlay J., Siegel R. L., Laversanne M., Soerjomataram I., Jemal A., et al. [2021]. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. Ca. Cancer J. Clin. 71, 209–249. 10.3322/caac.21660 [PubMed] [CrossRef] [Google Scholar]
  • Tatsimo S. J., Tamokou Jde D., Havyarimana L., Csupor D., Forgo P., Hohmann J., et al. [2012]. Antimicrobial and antioxidant activity of kaempferol rhamnoside derivatives from Bryophyllum pinnatum. BMC Res. Notes 5, 1–6. 10.1186/1756-0500-5-158[PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Thrift A. P., El-Serag H. B. [2020]. Burden of Gastric Cancer. Clin. Gastroenterol. Hepatol. 18, 534–542. 10.1016/j.cgh.2019.07.045[PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Tian C., Chang Y., Zhang Z., Wang H., Xiao S., Cui C., et al. [2019]. Extraction technology, component analysis, antioxidant, antibacterial, analgesic and anti-inflammatory activities of flavonoids fraction from Tribulus terrestris L. leaves. Heliyon 5, e02234. 10.1016/j.heliyon.2019.e02234[PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Tombola F., Campello S., De Luca L., Ruggiero P., Del Giudice G., Papini E., et al. [2003]. Plant polyphenols inhibit VacA, a toxin secreted by the gastric pathogen Helicobacter pylori. FEBS Lett. 543, 184–189. 10.1016/S0014-5793[03]00443-5 [PubMed] [CrossRef] [Google Scholar]
  • Tran Trung H., Truong Thi Huynh H., Nguyen Thi Thuy L., Nguyen Van Minh H., Thi Nguyen M. N., Luong Thi M. N. [202020080–20089]. 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 5, 20080–20089. 10.1021/acsomega.0c01640[PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Trojan-Rodrigues M., Alves T. L., Soares G. L., Ritter M. R. [2012]. Plants used as antidiabetics in popular medicine in Rio Grande Do Sul, southern Brazil. J. Ethnopharmacol. 139, 155–163. 10.1016/j.jep.2011.10.034 [PubMed] [CrossRef] [Google Scholar]
  • Tsukamoto T., Tatematsu M. [2014]. Role of Helicobacter pylori in gastric neoplasia. Curr. Infect. Dis. Rep. 16, 402–406. 10.1007/s11908-014-0402-4 [PubMed] [CrossRef] [Google Scholar]
  • Tungmunnithum D., Thongboonyou A., Pholboon A., Yangsabai A. [2018]. Flavonoids and Other Phenolic Compounds from Medicinal Plants for Pharmaceutical and Medical Aspects: An Overview. Med. [Basel] 5, 93. 10.3390/medicines5030093[PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Ustün O., Ozçelik B., Akyön Y., Abbasoglu U., Yesilada E. [2006]. Flavonoids with anti-Helicobacter pylori activity from Cistus laurifolius leaves. J. Ethnopharmacol. 108, 457–461. 10.1016/j.jep.2006.06.001 [PubMed] [CrossRef] [Google Scholar]
  • Van Der Watt E., Pretorius J. C. [2001]. Purification and identification of active antibacterial components in Carpobrotus edulis L. J. Ethnopharmacol. 76, 87–91. 10.1016/S0378-8741[01]00197-0 [PubMed] [CrossRef] [Google Scholar]
  • Vaquero M. J. R., Alberto M. R., de Nadra M. C. M. [2007]. Antibacterial effect of phenolic compounds from different wines. Food control. 18, 93–101. 10.1016/j.foodcont.2005.08.010 [CrossRef] [Google Scholar]
  • Waglechner N., Wright G. D. [2017]. Antibiotic resistance: it's bad, but why isn't it worse? BMC Biol. 15, 84–88. 10.1186/s12915-017-0423-1[PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Wang F., Meng W., Wang B., Qiao L. [2014]. Helicobacter pylori-induced gastric inflammation and gastric cancer. Cancer Lett. 345, 196–202. 10.1016/j.canlet.2013.08.016 [PubMed] [CrossRef] [Google Scholar]
  • Wang S., Alseekh S., Fernie A. R., Luo J. [2019]. The Structure and Function of Major Plant Metabolite Modifications. Mol. Plant 12, 899–919. 10.1016/j.molp.2019.06.001 [PubMed] [CrossRef] [Google Scholar]
  • Wang Y., Zhang J. Y., Song X. N., Zhang Z. Y., Li J. F., Li S. [2018]. Anti-ulcer and anti-Helicobacter pylori potentials of the ethyl acetate fraction of Physalis alkekengi L. var. franchetii [Solanaceae] in rodent. J. Ethnopharmacol. 211, 197–206. 10.1016/j.jep.2017.09.004 [PubMed] [CrossRef] [Google Scholar]
  • Wei R., Ma Q., Zhong G., He J., Sang Z. [2020]. Isolation and characterization of flavonoid derivatives with anti-prostate cancer and hepatoprotective activities from the flowers of Hosta plantaginea [Lam.] Aschers. J. Ethnopharmacol. 253, 112685. 10.1016/j.jep.2020.112685 [PubMed] [CrossRef] [Google Scholar]
  • Wu B.-L., Wu Z.-W., Yang F., Shen X.-F., Wang L., Chen B., et al. [2019]. Flavonoids from the seeds of Oroxylum indicum and their anti-inflammatory and cytotoxic activities. Phytochem. Lett. 32, 66–69. 10.1016/j.phytol.2019.05.003 [CrossRef] [Google Scholar]
  • Wu D., Kong Y., Han C., Chen J., Hu L., Jiang H., et al. [2008]. d-Alanine:d-alanine ligase as a new target for the flavonoids quercetin and apigenin. Int. J. Antimicrob. Agents 32, 421–426. 10.1016/j.ijantimicag.2008.06.010 [PubMed] [CrossRef] [Google Scholar]
  • Wu T., Zang X., He M., Pan S., Xu X. [2013]. Structure-activity relationship of flavonoids on their anti-Escherichia coli activity and inhibition of DNA gyrase. J. Agric. Food Chem. 61, 8185–8190. 10.1021/jf402222v [PubMed] [CrossRef] [Google Scholar]
  • Xie Y., Yang W., Tang F., Chen X., Ren L. [2014]. Antibacterial Activities of Flavonoids: Structure-Activity Relationship and Mechanism. Curr. Med. Chem. 22, 132–149. 10.2174/0929867321666140916113443 [PubMed] [CrossRef] [Google Scholar]
  • Yamaoka Y. [2010]. Mechanisms of disease: Helicobacter pylori virulence factors. Nat. Rev. Gastroenterol. Hepatol. 7, 629–641. 10.1038/nrgastro.2010.154[PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Yamaoka Y. [2012]. Pathogenesis ofHelicobacter pylori-Related Gastroduodenal Diseases from Molecular Epidemiological Studies. Gastroenterology Res. Pract. 2012, 1–9. 10.1155/2012/371503[PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Yeon M. J., Lee M. H., Kim D. H., Yang J. Y., Woo H. J., Kwon H. J., et al. [2019]. Anti-inflammatory effects of Kaempferol on Helicobacter pylori-induced inflammation. Biosci. Biotechnol. Biochem. 83, 166–173. 10.1080/09168451.2018.1528140 [PubMed] [CrossRef] [Google Scholar]
  • Yin X., Gyles C. L., Gong J. [2012]. Grapefruit juice and its constituents augment the effect of low pH on inhibition of survival and adherence to intestinal epithelial cells of Salmonella enterica serovar Typhimurium PT193. Int. J. Food Microbiol. 158, 232–238. 10.1016/j.ijfoodmicro.2012.07.022 [PubMed] [CrossRef] [Google Scholar]
  • Zengin G., Menghini L., Di Sotto A., Mancinelli F., Carradori S., Cesa S., et al. [2018]. Chromatographic analyses, In Vitro biological activities, and cytotoxicity of cannabis sativa l. Essential oil: A multidisciplinary study. Molecules 23. 10.3390/molecules23123266[PMC free article] [PubMed] [CrossRef] [Google Scholar] Zhang L., Kong Y., Wu D., Zhang H., Wu J., Chen J., et al. [2008]. Three flavonoids targeting the beta-hydroxyacyl-acyl carrier protein dehydratase from Helicobacter pylori: crystal structure characterization with enzymatic inhibition assay

Chủ Đề