Next Article in Journal
Discovery of the Role of Tick Salivary Glands in Enhancement of Virus Transmission—Beginning of an Exciting Story
Previous Article in Journal
Hepatitis E Virus Infection in Patients with Chronic Inflammatory Bowel Disease Treated with Immunosuppressive Therapy
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Plant-Derived Products with Therapeutic Potential against Gastrointestinal Bacteria

by
Fatimah I. Qassadi
1,2,
Zheying Zhu
1 and
Tanya M. Monaghan
3,4,*
1
School of Pharmacy, University of Nottingham, Nottingham NG7 2RD, UK
2
Department of Pharmacognosy, College of Pharmacy, Prince Sattam bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
3
NIHR Nottingham Biomedical Research Centre, University of Nottingham, Nottingham NG7 2RD, UK
4
Nottingham Digestive Diseases Centre, School of Medicine, University of Nottingham, Nottingham NG7 2RD, UK
*
Author to whom correspondence should be addressed.
Pathogens 2023, 12(2), 333; https://doi.org/10.3390/pathogens12020333
Submission received: 23 January 2023 / Revised: 12 February 2023 / Accepted: 14 February 2023 / Published: 15 February 2023
(This article belongs to the Section Bacterial Pathogens)

Abstract

:
The rising burden of antimicrobial resistance and increasing infectious disease outbreaks, including the recent COVID-19 pandemic, has led to a growing demand for the development of natural products as a valuable source of leading medicinal compounds. There is a wide variety of active constituents found in plants, making them an excellent source of antimicrobial agents with therapeutic potential as alternatives or potentiators of antibiotics. The structural diversity of phytochemicals enables them to act through a variety of mechanisms, targeting multiple biochemical pathways, in contrast to traditional antimicrobials. Moreover, the bioactivity of the herbal extracts can be explained by various metabolites working in synergism, where hundreds to thousands of metabolites make up the extract. Although a vast amount of literature is available regarding the use of these herbal extracts against bacterial and viral infections, critical assessments of their quality are lacking. This review aims to explore the efficacy and antimicrobial effects of herbal extracts against clinically relevant gastrointestinal infections including pathogenic Escherichia coli, toxigenic Clostridioides difficile, Campylobacter and Salmonella species. The review will discuss research gaps and propose future approaches to the translational development of plant-derived products for drug discovery purposes for the treatment and prevention of gastrointestinal infectious diseases.

1. Introduction

Globally, gastrointestinal (GI) infections are the second most prevalent infectious diseases after respiratory infections +. Bacterial gastroenteritis and diarrhoea are commonly caused by specific pathogenic strains of Escherichia coli, Campylobacter, Enterococcus faecalis, Salmonella spp., Shigella, and Clostridioides difficile [1,2,3]. Contaminated food and water and inadequate hygiene are key factors in the transmission of GI infections [4]. The most common GI symptom is diarrhoea, which is usually self-limiting and typically resolves within a few days for most healthy individuals. If diarrhoeal disease progresses, young children and older adults with co-morbidities may succumb to excessive dehydration, malnutrition, bacteremia, and other complications, which may result in death [5]. The World Health Organization (WHO) has reported more than 1.7 billion diarrhoeal disease cases each year. In addition, diarrhoeal diseases were responsible for the deaths of 370,000 children under five years of age in 2019 [4]. Identifying the aetiology of infectious gastroenteritis and making appropriate treatment decisions are crucial for patient management.
The search for new antimicrobial agents has intensified in recent years due to bacterial resistance to current antibiotics, with natural products playing an integral role [6]. Additionally, the lengthy testing process for new drugs before they are approved for commercialization adds to the call for alternative therapeutic approaches. [7]. Several methods have been proposed to investigate the antibacterial properties of natural products. The ancient world has used plants as medicaments for thousands of years, especially in Asian regions like India, China, and Japan, and in some African nations [8]. Among tribal people, these plants are mostly used as folkloric remedies mainly due to their ease of availability and relatively low prices [9,10]. According to a study examining the use of complementary and alternative medicine (CAM) in patients with GI disorders, nearly half were using herbal treatment, acupuncture, or mind/body treatments [11]. Several plant-derived products with various chemical structures have been discovered based on traditional knowledge of their medicinal uses [12]. The structural diversity of phytochemicals enables them to act through a variety of mechanisms (targeting multiple biochemical pathways), in contrast to traditional antibiotics [13,14]. Despite a significant increase in publications on this topic, the mechanisms of action of herbal extracts have remained elusive. In this review, we will discuss the preclinical and clinical evidence for the use of herbal-derived products as antibacterial agents for the treatment of clinically relevant infectious diseases associated with the GI tract (GIT).

2. Proposed Mechanisms of Action of Plant-Derived Products

Phytochemicals show promise in supporting and maintaining GI health (including the management of diarrhoea, gastritis, and gastric ulcers), and combating bacterial infections. There are thousands of structurally different compounds produced by plants (polyphenols, terpenoids, phenolic acids, essential oils, lectins, polypeptides, and alkaloids), each possessing different biological properties, which provide the scientific basis for the use of herbs in traditional medicine in many ancient communities [15]. Most of these compounds are secondary metabolites, which are heterogeneous from both a biosynthetic and structural perspective. The presence/absence of some specific functional groups increases the diversity of compounds belonging to the same biosynthetic class [16].
A great example of the strong relationship between the molecular structure of a compound and its possible biological properties can be found in the p-menthane type monoterpenoids Figure 1. The differences between these molecules, even if they appear insignificant at first glance, can be very important when considering the biological properties of these compounds, even if some are only mutually isomeric, differ in opposition to double bonds, and others have different functionalities/hetero atoms [12]. As an example, carvacrol, a compound isomer to thymol, was found to have antimicrobial effects primarily via cytoplasmic membranes, where the effect of carvacrol is enhanced by its hydroxyl group, which acts as a transmembrane carrier of monovalent cations, disrupting the membrane potential [17,18], while thymol, another aromatic p-menthane type monoterpene phenol, is thought to interact with both inner and outer membranes of the cell by integrating in the polar headgroup region of the lipid bilayer, increasing its permeability [19].
Ternanthranin, an alkaloid found in Choisya ternata Kunth. essential oil (Mexican orange blossom), is not only structurally similar to aspirin (acetylsalicylic acid), but also pharmacologically similar (N-methyl derivative of anthranilic acid) (Figure 2) [20]. Interestingly, anthranilic acid derivatives are considered to be important drug discovery pharmacophores [21] Many of these compounds are currently in use, including mefenamic acid and meclofenamates, which have analgesic, anti-inflammatory, and antipyretic properties [21,22].
In general, antibacterial actions of action of plant-derived products include inhibition of bacterial cell wall synthesis, cell membrane destruction, inhibition of bacterial protein synthesis, DNA replication, and metabolic pathways, making them excellent candidates for future drug development [6,15,23,24]. Additionally, they can inhibit mechanisms involved in bacterial resistance to antibiotics, including overexpression of efflux pumps, structural modification of porins, and antibiotic-target modification, which may lead to new approaches to overcoming antibiotic resistance [6,15]. Some plant-derived compounds with antibacterial properties against clinically relevant GI bacteria and their mechanism of actions are illustrated in Table 1.
Enterohemorrhagic Escherichia coli (EHEC), specifically, E. coli O157:H7, is one of the most important serotypes among the different E. coli pathogenic groups, which include enterotoxigenic (ETEC), enteropathogenic (EPEC), enterohaemorrhagic (EHEC), enteroinvasive (EIEC), diffusely adherent (DAEC), and enteroaggregative E. coli (EAEC) [45]. It is estimated that these serotypes cause 73,000 illnesses annually and 60 deaths in the United States alone [46]. Plant-derived products used for treating these pathogens possess antibacterial activities through inhibition of toxic production and toxin-mediated cellular damage on host cells [33].
Ginger (Zingiber officinale Roscoe) is one of the most used spices. Zingerone (vanillylacetone) is reported to be the active constituent responsible for the antidiarrhoeal efficacy of ginger [27]. Zingerone reduced heat-labile enterotoxin (LT)-induced diarrhoea in ETEC through blocking the binding to GM1 ganglioside receptors, thus preventing accumulation of diarrhoeal fluids in vivo [27]. The FDA recognizes ginger as a safe herb, and German monographs report no known side effects or drug interactions with ginger [27]. Similarly, extracts of Chinese medicinal herbs, Rhus chinensis Mill (Chinese sumac or nutgall tree), and Indian medicinal plant, Berberis aristata DC (Indian barberry or tree turmeric), were effective in preventing ETEC heat-labile enterotoxins from binding to their surfaces [47]. Gallic acid, the major component of Galla chinensis, was effective for treating LT-induced diarrhoea by causing inactivation of ETEC enterotoxins [47]. Galla chinensis is gall caused by the Chinese aphid (family Pemphigidae) on the Rhus leaves of the family Anacardiaceous (mainly, Rhus chinensis Mill) [48]. Furthermore, large amounts of tannins present in Galla chinensis inhibit intestinal bacterial growth in vitro including Clostridium perfringens, Clostridium paraputrificum, and E. coli without affecting the growth of the intestinal probiotics (bifidobacterial and lactobacilli) [49]. It was reported that tannins extract from Galla chinensis was one of the active compounds to resist E. coli and protect mice from infection by Enterotoxigenic E. coli [50].
Camellia sinensis tea leaves contain galloylated tea catechins, which have been observed to act at the level of the bacterial cell membrane level [51,52] inducing perturbations of the ordered structure of bilayers of phosphatidylcholine and phosphatidylethanolamine found in bacterial cell walls [51]. Epigallocatechin-3-gallate (EGCG), a major polyphenolic compound extracted from green tea leaves, was the most effective catechin causing inhibition of the growth of E. coli cells by damaging the lipid bilayer membrane. These results suggest a strong effect of catechins from green tea on bacterial phospholipid synthesis [53]. Furthermore, a decrease in Vero toxins (VTs) production in pathogenic EHEC has been observed following treatment with catechin and epigallocatechin in green tea. At concentrations of 0.05 mg/mL, these compounds markedly inhibit the release of extracellular VT into culture supernatant fluid [28].
Eugenol, the major component of clove (Syzygium aromaticum (L.) Merr. & L.M. Perry.), inhibits the production of two verotoxins (VT1 and VT2) in EHEC by modulating the transcription of stx 1 and stx 2, respectively [33]. Stx1 and Stx2 gene expression levels were similarly reduced with sub-inhibitory concentrations of Curtisia dentata extract, trans-cinnamaldehyde (major component of cinnamon) and carvacrol (obtained from oregano oil) [33,54]. These studies suggest that plant-derived compounds affect not only toxin-receptor binding, but also toxin genes and host receptor expression, reducing toxin-mediated pathology.
Proanthocyanidins show antibacterial activity against E. coli, the leading cause of bacteria-mediated gastrointestinal infections, by reducing bacterial adhesion to the gastrointestinal mucosa [55]. These phenolic compounds are produced in relatively high concentrations in Vaccinium macrocarpon (cranberry) and show promising activity against bacterial virulence toxins and biofilm formation [25,56]. Although the activity of phenolics is generally weak and nonspecific, they target Gram-positive, rather than Gram-negative bacteria [29,57].
An alkaloid, berberine, derived from the rhizomes of Berberis vulgaris, exhibits antibacterial properties against a wide range of pathogenic bacteria either alone or in combination with antibiotics [58,59]. Berberine was effective in targeting proteins responsible for upholding the structure of cells and for cell division. This led Domadia, et al. [60] to examine the effect of berberine on filamenting temperature sensitive mutant Z (FtsZ) activity, a bacterial cytokinesis protein essential for bacterial cell division. In silico modelling suggested that the hydrophobic residues in the GTP binding region may facilitate berberine binding to FtsZ, which makes it effective towards Gram-negative bacteria [61]. Scanning electron microscopy revealed that berberine altered bacterial morphology and ruptured cells, causing leakage of intracellular substances and the release of more K+ and Ca+, which inhibited protein synthesis [62]. Furthermore, this compound could decrease the activity of the Na+/K+-ATPase in cell membranes. Due to these effects, berberine prevents bacterial proteins from being expressed by destroying the cell membrane structures, ultimately resulting in the death of bacteria [60,62]. Interestingly, berberine, an efflux pump substrate, was shown to reduce the resistance of pathogenic strains to conventional antibiotics when it is co-administered with different efflux pump inhibitors [63]. Through inhibition of the MexXY-OprM efflux pump system, berberine exhibits synergistic effects with carbapenem in re-sensitizing imipenem-resistant Pseudomonas aeruginosa [64].
The mechanism by which berberine treats diarrhoea caused by V. cholerae and E. coli has been extensively investigated. Berberine directly inhibited V. cholerae and E. coli enterotoxins in vitro [32]. The inhibition of E.coli biofilm by berberine was associated with the suppression of quorum-sensing genes such as pfS, hflX, and fliA [65]. In addition, berberine significantly inhibited the secretion of E. coli heat-stable enterotoxins in a mouse model [32]. Even though the mechanism of action of the drug is not yet fully known, these data support its clinical value in treating acute diarrhoea [32]. Berberine significantly decreased the expression levels of the ATP-binding protein of the lipoprotein release system (LolD) and the binding protein of the phospholipid ABC transporter (MlaB) [66]. The absence of LolD decreases lipoprotein transport to the outer membrane of bacteria [67]. Additionally, MlaB contributes to maintaining outer membrane lipid asymmetry through the transport of aberrantly localized phospholipids between the outer and inner membranes, while Mla plays an important role in transporter assembly and function [68]. In Gram-negative bacteria, lipid asymmetry is crucial to the outer membrane’s function as a selectively permeable barrier. In the absence of LloD and MlaB expression, cell membrane stability is weakened [66] Berberine can significantly inhibit bacterial cell wall synthesis in E.coli by downregulating the capsule synthesis promoter (RcsF) and Undecaprenyl-diphosphatase (YbjG) [66]. Both are involved in bacterial cell wall synthesis, and the latter is necessary to synthesize undecaprenyl phosphate, a C55 lipid carrier for cell wall synthesis [69,70]. Consequently, incomplete cell wall synthesis reduced the antibiotic resistance of E. coli [32]. Even though the mechanism of action of the drug is not yet fully known, these data support its clinical value in treating acute diarrhoea [32]. Berberine significantly decreased the expression levels of the ATP-binding protein of the lipoprotein release system (LolD) and the binding protein of the phospholipid ABC transporter (MlaB) [66]. The absence of LolD decreases lipoprotein transport to the outer membrane of bacteria [67]. Additionally, MlaB contributes to maintain outer membrane lipid asymmetry through the transport of aberrantly localized phospholipids between the outer and inner membranes, while Mla plays an important role in transporter assembly and function [68]. In Gram-negative bacteria, lipid asymmetry is crucial to the function of the outer membrane as a selectively permeable barrier. In the absence of LloD and MlaB expression, cell membrane stability is weakened. [66] Berberine can significantly inhibit bacterial cell wall synthesis in E.coli by downregulating the capsule synthesis promoter (RcsF) and Undecaprenyl-diphosphatase (YbjG) [66]. Both are involved in bacterial cell wall synthesis, and the latter is necessary to synthesize undecaprenyl phosphate, a C55 lipid carrier for cell wall synthesis [69,70]. Consequently, incomplete cell wall synthesis reduced the antibiotic resistance of E. coli [66].
Aromatic essential oil constituents such as carvacrol (2-methyl-5-(1-methylethyl)-phenol) and cinnamaldehyde (3-phenyl-2-propenal) possess broad antimicrobial activity that extends to food spoilage and pathogenic microorganisms, including drug-resistant and biofilm-forming microorganisms [33,34]. The antibacterial activity of these compounds is due to the considerable changes in the structural and functional properties of cytoplasmic membranes [19,71]. An in vitro study found that carvacrol and cinnamaldehyde reduce production of C. difficile toxins A (TcdA) and toxin B (TcdB) and toxicity while maintaining normal gut flora growth [34]. These compounds were found to down-regulate toxin production genes. Several toxin repressor genes were found to be positively modulated by the compounds as well, potentially through global pleiotropic repressor CodY [34,72]. As a result of carvacrol supplementation, diarrhoea incidence in mice was significantly reduced and C. difficile-induced clinical symptoms were mitigated, causing a favourable shift in gut microbiota composition without negatively affecting gut microbiome diversity [73]. Based on these findings, carvacrol may be a potential anti-C. difficile agent, but further clinical studies are needed.
Several studies have shown that allicin in garlic juice (Allium sativum L.) inhibits C. difficile toxin production (TcdA) along with the genes encoding TcdR, an RNA polymerase sigma factor needed for maximal expression of TcdA (4.7 mg/mL) [74,75]. The antibacterial activity of garlic may also be attributed to other compounds such as saponins and flavonoids. A study found that the ginger component zingerone protected cells against C. difficile toxins TcdA and TcdB by blocking either the toxin-binding site on toxin molecules or the host cell receptors (0.3 mg/mL) [75]. Further investigation of biologically active components showed that zingerone was the active constituent responsible for the anti-diarrhoeal effect of ginger [27]. In an earlier study, Doughari et al. investigated the effect of onion bulb extract and its bioactive components on inhibition of toxigenic C. difficile toxins TcdA and TcdB in vitro. Fresh onion bulb extract (12.5% v/v) significantly decreased toxin production and activity. The primary mechanism of action of this extract was the inhibition of C. difficile sporulation at different stages [57]. In a mouse model of C. difficile infection (CDI), berberine (100 mg/kg/day) combined with vancomycin was more effective than vancomycin alone. Treatment with berberine prevents CDI relapses and reduces mortality significantly [76].
A novel lead compound, chromomycin A2/NP-00387, with potent anticlostridial activity in vitro and in vivo, conferred 100% protection to infected mice against clinical manifestations of CDI at a concentration of 0.03 μg/mL [77]. The compound belongs to the aureolic acid family of bacterial natural products, and exhibits time-kill kinetics similar to fidaxomicin [77]. Furthermore, chromomycin A2 has been shown to be non-toxic to Caco-2 cells and to be as effective as vancomycin in protecting mice against C. difficile [78]. It will be necessary to conduct further trials to evaluate its efficacy in ameliorating CDI symptoms, including validating its mechanism of action and determining its efficacy in a recurrent infection model.
Scutellaria baicalensis Georgi. (Chinese skullcap) roots contain baicalin (5,6,7-trihydroxyflavone), a major flavone glycoside identified in the Chinese Pharmacopoeia as a medicinal herb [37]. Researchers have observed that baicalin was capable of significantly inhibiting H. pylori in the murine stomach [35]. Previously, researchers have identified that baicalin improved renal function and significantly reduced Shiga-Like Toxin 2 lethality in mice. Additionally, structural and biophysical studies have shown that baicalin binds directly to Stx to inactivate the toxin and promote toxin oligomerization [36]. Moreover, the same research group challenged mice with Enterohemorrhagic E. coli O157:H7 and showed that baicalin reduced lethality in mice, but its exact mechanism of action remains unclear [38].
There are only a few reports available in which researchers have assessed essential oils [39,79,80] and fruits extract [81] against C. difficile. A recent study examined the effect of essential oil compounds on mixed human faecal microbiota. As a result of the study, thymol and geraniol at a concentration of around 100ppm may be effective in suppressing pathogens in the small intestine without damaging beneficial commensal bacteria of the colon [39]. Several of the fatty acids found in virgin coconut oil have been tested for their antimicrobial properties against C. difficile in vitro [79]. Lauric acid (C12) significantly inhibited the growth of the bacteria. At 2 mg/mL of lauric acid exposure, transmission electron microscopy clearly demonstrated that both the cell membrane and cytoplasm were disrupted. Furthermore, live/dead staining of bacterial cells following treatment with selected fatty acids confirmed the physiological changes in bacterial cell membrane integrity [79].
It has become increasingly difficult to manage campylobacteriosis because of antibiotic-resistant strains of Campylobacter. Campylobacter has been reported to possess a wide variety of cytotoxic activities [82], but only the cytolethal distending toxin (CDT) has been clearly defined. There is evidence that certain strains of Campylobacter jejuni that cause dysentery-like illness are more cytotoxic than other Campylobacter species [83]. Although some plants have been evaluated for their ability to inhibit Campylobacter jejuni and Campylobacter coli, little is known about their effect on virulence factors of these bacteria [84]. A preliminary analysis showed 21 of the 28 plant extracts inhibited Campylobacter growth, 11 of which showed a high level of inhibition (inhibition zone of 2 to 4 cm) [85]. The four plants with the best anti-Campylobacter activity were Acacia farnesiana, Artemisia ludoviciana, Opuntia ficus-indica, and Cynara scolymus. It has also been reported that Japanese green tea extracts show anti-Campylobacter jejuni activity [86]. A critical step in Campylobacter pathogenesis is adhesion to intestinal mucosa [82,87]). In a study by Lengsfeld, et al. [88], okra extracts inhibited the adhesion of C. jejuni, with a strong antiadhesive effect in vitro, but not in vivo. Similarly, extracts of A. farnesiana, A. ludoviciana, C. scolmymus, and O. ficus-indica significantly reduced Campylobacter adhesion to Vero cells [85]. This inhibitory activity was caused by flavonoids and polyphenols. It’s thought these compounds work as antimicrobials because they form a complex with extracellular and soluble proteins, which are then able to bind to bacterial cell walls [89]. Furthermore, evidence indicates that lipophilic flavonoids can disrupt microbial membranes [12,15].

3. Preclinical and Clinical Studies of Herbal Products in the Management of GI Infectious Diarrhoeal Disorders

The global healthcare system is heavily burdened by GIT disorders. Globally, diarrhoeal-inducing GIT disorders are one of the leading causes of death [90]. Even though pharmaceutical drugs are effective in treating most GIT ailments, a growing trend has been observed in the use of traditional treatments (e.g., through herbal remedies) [91].
Due to the absence of significant pre-clinical and clinical research on the effectiveness of herbal medicine, it is important to gather whatever scientific data to support their use in treating different pathologies. A large proportion of herbal medicines are used in developing countries [11]. There are many biologically active compounds in the natural products derived from medicinal plants. Globally, there are approximately 500,000 species of plants, but only 1% have been examined for novel bioactive compounds, and only a small fraction has been tested in randomized controlled trials as illustrated in Table 2 [92,93].
In India, raw baobab fruit (Adansonia digitata) is eaten to treat diarrhoea and dysentery. According to a clinical study conducted in Senegal comparing Adansonia fruit with oral rehydration therapy, the two treatments did not differ significantly in terms of diarrhoea duration and weight gain [94]. It is possible that the fruit’s astringent properties explain its medicinal properties, although high levels of tartaric acid can cause gastrointestinal irritation if consumed in large quantities [95].
Guava is well-known for treating diarrhoea in tropical countries through ingesting teas, decoctions, or macerations [96]. Guava leaves, Psidium guajava (myrtaceae), originate from Central and South America where this tree forms part of the pantropical region. Additionally, the plant can be used to treat coughs, diabetes, dysentery, fevers, leukorrhea, rheumatism, toothaches, and wounds [97]. The use of decoctions of dried leaves and aqueous extracts of leaves to treat diarrhoea has been supported by independent experiments in mice. A mixture of flavonoids including quercitrin, kaempferol, avicularin, and guaijaverin is believed to be the active components [98]. Guava has been evaluated using various in vivo models of infectious diarrhoea. An ethanol extract of P. guajava was shown to clear infection faster after 19 days in mice infected with Citrobacter rodentium [99], and mice infected with V. cholerae had intestinal ameliorative effects after a 4-h infection when treated with 250 mg/kg of ethanol extracts of P. guajava leaves [100]. A randomized double-blind clinical trial was conducted to evaluate the efficacy of the phytodrug QG5®, which contains P. guajava leaves with high flavonoid concentrations, on patients with infectious gastroenteritis. The duration of abdominal pain was significantly reduced by QG5® in these patients without any serious side effects [101]. The literature lacks sufficient information on the toxicological effects of P. guajava extracts. According to one study, oral administration of 5000 mg/kg body weight of methanolic extract of P. guajava bark for 28 days did not cause toxicity in Wistar rats [102], although in histopathological analyses, repeated doses exceeding 1000 mg/kg b.w. resulted in minor liver inflammation in females treated at high doses, and a sensitivity to the extract was observed in the males [102]. Early clinical trials showed that berberine tannate combined with sulfadimidine and neomycin were effective in treating acute infective diarrhoea in children [103,104]. Among the bacteria isolated from the stools of paediatric patients, E. coli was most prevalent. Clinical trials conducted on 100 gastroenteritis patients showed that berberine tannate was more effective than conventional antibiotics in treating diarrhoea [105]. When berberine tannate was administered to children with acute diarrhoea, their recovery was faster than with standard antibiotic therapy [106]. Many diarrhoea-causing bacteria, such as E. coli, Shigella spp., and Salmonella spp. were isolated from the patients [106].
In a randomized controlled trial, berberine sulphate reduced mean stool volume in 165 adult patients with enterotoxigenic E. coli and V. cholerae diarrhoea during three consecutive 8-h periods after treatment. There have been no reported side effects associated with berberine [107]. There are probably several mechanisms underlying berberine’s anti-diarrhoeal activity, including the inhibition of intestinal secretion caused by microbial enterotoxins [32], inhibition of gastrointestinal motility [108,109], enhanced water and Na+ absorption [110], and its direct antimicrobial properties. It is still noteworthy that Minimum Inhibitory Concentrations (MICs) of berberine for bacteria associated with diarrhoea, such as Bacillus cereus, E. coli, and V. cholerae, present at relatively high levels (12.5–469 μg/mL) [31,111].
In a study of 50 patients with CDI and non-C. difficile antibiotic-associated diarrhoea, berberine cure rates were 84 and 92 percent, respectively [112]. A meta-analysis has concluded that traditional Chinese medicine such as berberine, alone or combined with Western medicine, is superior to Western medicine alone in treating antibiotic-associated diarrhoea [113]. The inhibition of C. difficile toxin genes was reported by Shu et al. [114] to be a potential pharmacological mechanism of traditional Chinese medicine for the treatment of CDI. Studies have shown that berberine may induce apoptosis and adversely affect liver function in vivo and in vitro [115]. Aside from gastric troubles, berberine toxicity also causes hepato- and haematotoxicity, haemorrhagic inflammatory consequences, and immune cell damage [115,116]. The literature reveals, however, that berberine’s efficacy data are far more abundant than its toxicity data [64,117,118,119]
As part of a parallel, double-blind, randomized trial of 133 European tourists in West Africa and 112 American students in Mexico, bismuth subsalicylate significantly shortened travellers’ diarrhoea duration at both study sites [120]. Bismuth subsalicylate was found to be effective and well-tolerated in an open-label, two-day study in 94 adult students with acute diarrhoea [121]. The use of bismuth subsalicylate led to the clearance of pathogenic E. coli in all cases and was well tolerated with no reported adverse effects. In a placebo-controlled randomized trial conducted with 252 children, 150 mg of bismuth subsalicylate per kilogram of body weight per day decreased the duration of acute diarrhoea. C. jejuni, enteropathogenic E. coli, Salmonella spp., Shigella spp., and Vibrio spp. were found in stool samples collected at the beginning of the study [122]. There was an in vitro inhibitory effect of bismuth subsalicylate against C. difficile, enterotoxigenic E. coli, S. enterica subsp. enterica serovar Typhimurium, and Shigella sonnei. Bismuth subsalicyclate was found to have MICs of 2, 8, 4–8, and 2–8 μg/mL, respectively [123]. Despite its therapeutic efficacy, Chowdhury and his team observed a significant increase in weight gain in patients treated with bismuth subsalicylate compared with placebo, without major toxicity [124] The use of over-the-counter bismuth subsalicylate for long periods of time has been reported to cause acute toxicity manifested as nephrotoxicity [125].
Several dietary supplements, herbal medicines, and over-the-counter pharmaceuticals are available on the international market that support and maintain GI health (including diarrhoea, gastritis, and gastric ulcers) [126]. Various forms are available, including capsules, liquids, tablets, and tinctures. In addition, plant extracts and derivatives containing antibacterial properties are available commercially on the international market [127]. There are various food and herbal supplements available in Europe containing extracts from roots and rhizomes of Coptis spp., Berberis spp., and Hydrastis canadensis as capsules, tablets, and tinctures (e.g., Huang Lian tincture (Coptis chinensis extract), Napiers, Edinburgh, UK; Berberine HCL 97% Capsules, British supplements, Milton Keynes, UK; and Hydrastis Canadensis capsules, Alfa herbal, Barcelona, Spain). In addition to supporting digestive function, these supplements are also used to treat gastrointestinal disorders, including diarrhoea [59,110]. Most clinical data on their anti-diarrhoeal effects are based on berberine, an isoquinoline alkaloid (especially in its hydrochloride, sulphate, and tannate forms) [58,104]. In Asia, berberine hydrochloride tablets are sold for the treatment of intestinal infectious diseases and diarrhoea (Northeastern Pharmaceutical Group, Shenyang, China). Furthermore, Bismuth subsalicylate (the Procter & Gamble Company, Cincinnati, OH, USA) is available in the form of liquids or chewable/swallowable tablets for the treatment of stomach and GI discomfort, including diarrhoea, indigestion, heartburn, and nausea, under the brand name Pepto-Bismol [122,123]. In Latin America, QG5 is sold to relieve inflammation, spasms, and abdominal pain, containing 166 milligrams of Psidium guajava leaves dry extract, which contains flavonoids like quercetin [101].
Table 2. Herbal-derived products used for treating GI infections in clinical trials.
Table 2. Herbal-derived products used for treating GI infections in clinical trials.
DiseaseTreatmentNo. of PatientsStudy DesignDosage/DurationOutcomes Reference
Diarrhoea-predominant irritable bowel syndrome
(i)
Ayurvedic herbal compound: Murraya koenigii (curry), Punica granatum (pomegranate), and Curcuma longa (turmeric)
(ii)
Placebo
22Double-blind, cross-over randomized clinical trial Twice daily/4 weeks followed by a one week wash out periodNo significant improvement in the symptoms of diarrhoea-pre-dominant irritable bowel syndrome compared to placebo.[128]
Diarrhoea-predominant irritable bowel syndrome
(i)
Traditional Chinese medicine containing 11 herbs including Punica granatum
(ii)
Placebo
119Double-blind, randomized clinical trialTwice daily/8 weeksNo significant difference was observed in symptom and Quality of Life (QoL) scores between two groups.[129]
Diarrhoea
(i)
Adansonia digitata fruit
(ii)
Oral rehydration therapy
161Prospective clinical trialDaily with a follow-up after 4 to 48 h The two treatments did not differ significantly in terms of diarrhoea duration and weight gain[130]
Acute infective diarrhoea
(i)
Berberine tannate
(ii)
Sulfadimidine and neomycin
55Randomized clinical trial Initial dose 40 mg
followed by 20 mg
every 4 h for 5 days
Faster recovery using berberine compared to standard antibiotic therapy with a clinical cure 72%[103]
Antibiotic-associated
diarrhoea (AAD)
(i)
Berberine
63Prospective studyNRCure rates 84 and 92% for C.
difficile and non-C.
difficile AAD, respectively
[112]
Acute gastroenteritis
and dysentery
(i)
Berberine and its combinations with, chloramphenicol, streptomycin, sulphaimidine and iodochlorhydroxyqinoline
(ii)
Antibiotics only
129Clinical trial 300 mg/day for 7 days Combinations of berberine with antibiotics were more effective than berberine and antibiotics alone[131]
Acute nonspecific
diarrhoea
(i)
Bismuth subsalicylate
(ii)
loperamide hydrochloride
94An open-label parallel comparison study
bismuth subsalicylate
4900 mg/day for 2 days Loperamide was significantly effective for diarrhoea treatment than bismuth subsalicylate[121]
Enterotoxigenic E. coli and V. cholerae diarrhoea
(i)
Berberine sulphate
(ii)
Berberine sulphate plus tetracyclic (for Cholera patients)
(iii)
Tetracycline alone
165Randomized clinical trial 400 mg or 1200 mg berberine sulphate in a single oral doseReduced mean stool volume during three consecutive 8-hr periods after treatment [107]
Infectious gastroenteritis
(i)
Phytodrug (QG-5®) developed from guava leaves, standardized in its content of quercetin
(ii)
Control
100Randomized clinical trial 500 mg every 8 h for 3 daysDecreased the duration of abdominal pain with no significant changes in the consistency and frequency of liquid stools compared with the control group.[101]

4. OMICS Applications for Studying Biological Effects of Herbal Products against GI Bacteria

Currently, omics approaches, which encompass genomics, transcriptomics, proteomics, and metabolomics, are becoming important in identifying and characterizing crucial gene–protein–metabolite networks, discovering new drug metabolites, identifying complete genomes, transcriptomes, and proteomes of medicinal plants, evaluating human cell responses to drugs and whole ethnobotanical plants for medicinal use, and facilitating large-scale production of plant-derived medicines [132,133]. A research strategy that is based on the study of complex interactions between active constituents utilizing “omics” technologies (transcriptomics, proteomics, and metabolomics) and systems biology will be imperative to developing effective herbal-based treatments for infectious diseases.

4.1. Metabolomics in Antimicrobial Medicinal Plant-Based Drug Discovery

The discovery of plant biosynthetic pathways and gene clusters has increased dramatically over the past few decades, spurred by technological advances in sequencing and mass spectrometry technologies [134]. Due to the functional diversity and structural complexity of biosynthetic pathways in plants, the expansion of biosynthetic pathway discovery in plants lags behind that of bacteria and fungi [135]. A wide range of plant biosynthetic pathways have been explored using metabolomics-based approaches, such as chromatography coupled with mass spectrometry. A variety of analytical and computational metabolomics approaches being developed in recent decades for profiling specialized metabolites and analysing biosynthetic pathways, plant-based natural product discovery remains challenging despite significant technological advances in instrumentation, software, and databases [136]. This is mainly because of two reasons. The first is the diversity of functional and chemical properties of specialized metabolites in plants. Secondly, the specialized metabolites produced by a plant are part of its total metabolome. Since classical reductive approaches (experimentation and targeted approaches) to characterizing metabolites are time-consuming and laborious, untargeted metabolomics using mass spectrometry (MS) can be used to profile specialized metabolites in crude extracts and identify unprecedented numbers of metabolic classes [134]. Identification of metabolites from a variety of sources, including leaves, roots, soils, and volatiles, has led to the identification of biosynthesis pathways by identifying key shifts in the metabolite profile. While full metabolite scans with chromatography and mass spectrometry (MS1 mode) are (more accurately) quantified, they are unreliable in metabolite annotation, since several compounds can have the same mass but have different molecular formulas or may have the same molecular formula but differ in their chemical structure [133,137]. To address this challenge, metabolites are further fragmented using tandem MS mode (also known as MS2 or MS/MS or MSn when deeper fragmentation levels are included). With the aid of a variety of software and tools that have been developed for mining and annotating MS2 fragmentation data, metabolites can be structurally annotated and readily identified [137]. Aside from developing new classes of antibiotics with direct growth inhibition effects against bacteria, future research should also focus on finding indirect antimicrobials that are more effective and have better safety profiles. To understand the mechanisms of action at a molecular level, it is crucial to investigate the metabolome of pathogens after treatment.
Metabolomics experiments offer a more efficient route to drug discovery than the classical approach in natural products research. In essence, metabolomics analyses all metabolites present in a living system qualitatively and quantitatively [136]. Interestingly, this concept can be extended to the study of the relationship between whole metabolomes of natural products and their biological effects [134]. Different studies suggest that various enzymes, metabolic pathways, cell membrane or cell wall transport proteins and chaperones may also serve as potent targets for berberine, a natural product isolated from medicinal plants [66,138,139,140,141]. In a metabolomics study which combined principal component analysis, the antibacterial activity of traditional Chinese medicine Aquilegia oxysepala extract on Staphylococcus aureus and its main chemical components (genkwanin, apigenin, maguoflorine, and berberine), which are used for treating GI infections, were examined in comparison with nine antibiotics with known mechanisms of action [142]. As shown by this study, A. oxysepala targets are similar to those of protein synthesis inhibitors including lincolmensin, erythromycin, chloromycetin, streptomycin, and acheomycin, and its bioactive component maguoflorine accounted for most of the antibacterial activity. In this study, berberine showed similar effects to rifampicin and norfloxacin, both of which target nucleic acids [142]. These metabolomics strategies, which compare phytochemicals with known antibiotics, may be somewhat limited since they do not have the ability to define a new mechanism of action, but are viable and unexplored options for tagging new bioactive compounds with known mechanisms.

4.2. Transcriptomics and Functional Analysis of Proteins in Medicinal Plants

Transcriptomics is an effective method for assembling genomic data from a variety of non-model therapeutic plants that lack a reference genome. Transcriptomic studies assist in understanding the molecular mechanisms involved in the formation of secondary metabolites and in probing pharmaceutically significant mechanisms [143]. The transcriptomic analysis of Catharanthus roseus (L.) G. Don (Madagascar Periwinkle) revealed diverse iridoid-based monoterpene indole alkaloids [144]. It was determined from these transcriptomic data that a new iridoid synthase was responsible for converting 10-oxogeranial into iridoid scaffold [145] and that cytochrome P450 hydroxylation genes are involved in the biosynthesis of monoterpenoid indole alkaloids [146]. There are a number of non-glycosidic iridoids that possess strong antibacterial properties, including mussaenin A, gardendiol, isoboonein, and rehmaglutin D, which have a similar activity to chloramphenicol against E. coli [147].
A wide range of functions can be achieved with proteomics in medicinal plant research [148]. Using proteomics, we can visualize how proteins are structured, function, and modified, as well as how they interact with each other in vitro and in vivo [143]. Proteomics is a multi-functional field that can help predict the protein targets of plant-based bioactive compounds. It can also be used to understand how traditional Chinese medicine (TCM) acts on bacterial cells and how protein–drug interactions occur at the molecular level [149].
The proteomic analysis of Salmonella enterica revealed that energy metabolism-related proteins and flagellar proteins were up-regulated with berberine treatment, indicating a possible antibacterial action at the molecular level [150]. Although berberine is a cationic alkaloid, its water solubility is poor, which explains its reduced intestinal absorption. In animal models, it was observed that berberine’s bioavailability was much lower than 1%, since the amount that is absorbed by the gut is excreted back into the colon by P-glycoprotein [127].
These studies show how OMICS technologies have potential for identifying the mechanisms of action of bioactive compounds, as well as combating drug-resistant bacteria. Furthermore, these studies suggest that bacterial cell death cannot be explained solely by the direct interaction between bioactive compounds and their cellular targets [151,152], with several reports indicating that bacterial metabolic states can affect antibiotic effectiveness [153,154].

5. Safety Implications Regarding the Use of Plant-Derived Antimicrobials

The appeal of herbal medicine is multifactorial. It is widely believed by the general public that herbal medicines are safe to consume and are not associated with any significant side effects. Since these treatments are “natural”, they are thought to be safe as an alternative to conventional medicine. However, several side effects have been reportedly associated with herbal products used in the treatment of GI disorders. These are listed in Table 3. Further, herbal products are rarely subject to the same rigorous regulations as applied to other non-herbal medicinal products [155]. In the European Union (EU), herbal products are either categorized as food supplements or medicines; those which are “Classified as Medicine” are regulated under EU medicinal law Directive 2004/24/EC. The European Medicines Agency (EMA) handles the safety, efficacy, and premarket authorisation of these products. However, in the United States, the Food and Drug Administration (FDA) considers herbal products as dietary supplements and as such, they do not need to be registered with the FDA. The regulatory landscape in other countries has been summarised elsewhere [126].

6. Future Perspectives

Many previous studies concerning plant-derived antibacterial agents failed to standardize their methods to achieve reproducible and comparable results. The effectiveness of a plant’s antimicrobial properties is generally demonstrated through large-scale, well-designed clinical trials. It is therefore essential that clinical trials follow the guidelines of the U.S. FDA and the EMA when evaluating medicines indicated for treating bacterial infections [180,181]. Plant-derived products should also be evaluated in vitro for their antimicrobial effectiveness according to the methods recommended by the Clinical and Laboratory Standards Institute (CLSI) or by the European Committee on Antimicrobial Susceptibility Testing (EUCAST) [182].
As bacteria become increasingly resistant to commonly used antibiotics, new classes of plant-derived antimicrobials to treat infections caused by pathogens, such as C. difficile, Enterobacter, E. coli, K. pneumonia, and Pseudomonas, are of intense interest. The potential synergy or potentiation of phytochemicals with other antibacterial agents has already been demonstrated in previous studies [14,63,183]. In this sense, the use of plant constituents in the development of resistance-modifying drugs, such as agents affecting membrane permeability or modified antimicrobial targets, as well as agents affecting bacterial enzymes, inactivating antibiotics or efflux pumps, is becoming an area of active research [184].
As part of their defence against microbial infections, plants use a variety of chemical strategies to reduce the selective pressure for developing antibiotic resistance. The activity of a compound can be significantly increased or decreased depending on its synergistic and antagonistic effects with other compounds found in plants. The complex chemical composition of plant extracts may also produce pharmacological effects, as is the case for traditional herbal medicines, whose efficacy is based on their complex composition, due to their synergistic or antagonistic interactions with a wide spectrum of phytochemicals [63,185]. Developing more effective plant-derived antibacterial agents seems to be possible by developing chemically standardized complex phytotherapeutic preparations with a balanced ratio of antimicrobial, synergistic, and solubility-enhancing constituents. Nevertheless, there are specific problems associated with the use of complex phytotherapeutics as well as those associated with single-ingredient plant drugs, such as a lack of standardization, safety, and effectiveness validation, quality control, and the lack of patent rights over herbal medicines [186] In order for herbal products to succeed, clear intellectual property rights and standardized chemical and quality control standards must be established.

7. Conclusions

This review summarizes data on the clinical efficacy and antimicrobial properties of antibacterial agents of plant origin currently used to prevent and treat GI infections. A paradigm shift has occurred from focusing on conventional antibiotic therapy to alternative approaches due to new resistant bacterial strains. There is a wide variety of active constituents found in plants, making them an excellent source of antimicrobial agents with therapeutic potential as alternatives or potentiators of antibiotics. The identification of new and valuable antibacterial compounds from plants and the testing of their antibacterial properties are crucial for this purpose. Several factors may contribute to the effectiveness of herbal-derived compounds for fighting bacterial infections, two of which may be pivotal. First, from a phytochemical point of view, these compounds belong to various classes of plant secondary metabolites such as isoquinoline alkaloids, monoterpenoids, phenolic acids, and tannins, which may provide alternative antibacterial mechanisms to conventional antibiotics. Second, it is extremely likely that the utilization of unique traditional knowledge of herbal medicine will result in biocompatible, cost-effective, and promising solutions, ultimately leading to the discovery of new antibacterial agents. Although new plant-derived antibacterial compounds are increasingly being studied, few are still in clinical trials. However, most of them show antibacterial activity in vitro and in vivo. Most of these products act as direct growth inhibitors; however, certain products have antimicrobial properties due to their antitoxin properties (e.g., green tea extract). Inhibitory agents of biofilm formation may also be of interest. Plant-derived products may be used as biofilm inhibitors and synergistic agents in the future. It may be more fruitful to investigate combinations of plant compounds with conventional antibiotics. There may be a reason for this, since phytomedicines require complex combinational effects between their bioactive compounds to synergize their activities. Innovative approaches using high-throughput technologies (omics) and systems biology are crucial for effective proof-of-concept research and development of new types of plant-derived products that could be effective against antimicrobial resistance.

Author Contributions

Conceptualization, T.M.M. and F.I.Q.; Writing—Original draft presentation, F.I.Q.; Writing—Review and editing, T.M.M. and Z.Z.; Visualization, F.I.Q.; Supervision, T.M.M. All authors have read and agreed to the published version of the manuscript.

Funding

F.I.Q. is funded by the Saudi Ministry of Higher Education as part of her PhD project.

Data Availability Statement

Not applicable.

Acknowledgments

We wish to thank James Thomas for assisting with copyediting.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Thielman, N.M.; Guerrant, R.L. Acute infectious diarrhea. N. Engl. J. Med. 2004, 350, 38–47. [Google Scholar] [CrossRef]
  2. Casburn-Jones, A.C.; Farthing, M.J. Management of infectious diarrhoea. Gut 2004, 53, 296–305. [Google Scholar] [CrossRef] [Green Version]
  3. Zboromyrska, Y.; Vila, J. Advanced PCR-based molecular diagnosis of gastrointestinal infections: Challenges and opportunities. Expert Rev. Mol. Diagn. 2016, 16, 631–640. [Google Scholar] [CrossRef]
  4. WHO. Diarrhoeal Disease. Key Facts. Available online: https://www.who.int/news-room/fact-sheets/detail/diarrhoeal-disease (accessed on 5 December 2022).
  5. Fischer Walker, C.L.; Sack, D.; Black, R.E. Etiology of diarrhea in older children, adolescents and adults: A systematic review. PLoS Negl. Trop. Dis. 2010, 4, e768. [Google Scholar] [CrossRef] [Green Version]
  6. Khameneh, B.; Diab, R.; Ghazvini, K.; Fazly Bazzaz, B.S. Breakthroughs in bacterial resistance mechanisms and the potential ways to combat them. Microb. Pathog. 2016, 95, 32–42. [Google Scholar] [CrossRef]
  7. Spellberg, B.; Powers, J.H.; Brass, E.P.; Miller, L.G.; Edwards, J.E., Jr. Trends in Antimicrobial Drug Development: Implications for the Future. Clin. Infect. Dis. 2004, 38, 1279–1286. [Google Scholar] [CrossRef] [Green Version]
  8. Pan, S.Y.; Litscher, G.; Gao, S.H.; Zhou, S.F.; Yu, Z.L.; Chen, H.Q.; Zhang, S.F.; Tang, M.K.; Sun, J.N.; Ko, K.M. Historical perspective of traditional indigenous medical practices: The current renaissance and conservation of herbal resources. Evid. Based Complement. Altern. Med. 2014, 2014, 525340. [Google Scholar] [CrossRef] [Green Version]
  9. Pandey, M.M.; Rastogi, S.; Rawat, A.K.S. Indian Traditional Ayurvedic System of Medicine and Nutritional Supplementation. Evid.-Based Complement. Altern. Med. 2013, 2013, 376327. [Google Scholar] [CrossRef] [Green Version]
  10. Ezekwesili-Ofili Josephine, O.; Okaka Antoinette Nwamaka, C. Herbal Medicine in African Traditional Medicine. In Herbal. Medicine; Philip, F.B., Ed.; IntechOpen: Rijeka, Croatia, 2019; p. 191. [Google Scholar]
  11. Tillisch, K. Complementary and alternative medicine for functional gastrointestinal disorders. Gut 2006, 55, 593–596. [Google Scholar] [CrossRef] [Green Version]
  12. Radulović, N.S.; Blagojević, P.D.; Stojanović-Radić, Z.Z.; Stojanović, N.M. Antimicrobial plant metabolites: Structural diversity and mechanism of action. Curr. Med. Chem. 2013, 20, 932–952. [Google Scholar] [CrossRef]
  13. AlSheikh, H.M.A.; Sultan, I.; Kumar, V.; Rather, I.A.; Al-Sheikh, H.; Tasleem Jan, A.; Haq, Q.M.R. Plant-Based Phytochemicals as Possible Alternative to Antibiotics in Combating Bacterial Drug Resistance. Antibiotics 2020, 9, 480. [Google Scholar] [CrossRef]
  14. Cheesman, M.J.; Ilanko, A.; Blonk, B.; Cock, I.E. Developing New Antimicrobial Therapies: Are Synergistic Combinations of Plant Extracts/Compounds with Conventional Antibiotics the Solution? Pharm. Rev. 2017, 11, 57–72. [Google Scholar] [CrossRef] [Green Version]
  15. Porras, G.; Chassagne, F.; Lyles, J.T.; Marquez, L.; Dettweiler, M.; Salam, A.M.; Samarakoon, T.; Shabih, S.; Farrokhi, D.R.; Quave, C.L. Ethnobotany and the Role of Plant Natural Products in Antibiotic Drug Discovery. Chem. Rev. 2021, 121, 3495–3560. [Google Scholar] [CrossRef]
  16. Vaou, N.; Stavropoulou, E.; Voidarou, C.; Tsigalou, C.; Bezirtzoglou, E. Towards Advances in Medicinal Plant Antimicrobial Activity: A Review Study on Challenges and Future Perspectives. Microorganisms 2021, 9, 2041. [Google Scholar] [CrossRef]
  17. Ultee, A.; Bennik, M.H.; Moezelaar, R. The phenolic hydroxyl group of carvacrol is essential for action against the food-borne pathogen Bacillus cereus. Appl. Env. Microbiol. 2002, 68, 1561–1568. [Google Scholar] [CrossRef] [Green Version]
  18. Ben Arfa, A.; Combes, S.; Preziosi-Belloy, L.; Gontard, N.; Chalier, P. Antimicrobial activity of carvacrol related to its chemical structure. Lett. Appl. Microbiol. 2006, 43, 149–154. [Google Scholar] [CrossRef]
  19. Lambert, R.J.; Skandamis, P.N.; Coote, P.J.; Nychas, G.J. A study of the minimum inhibitory concentration and mode of action of oregano essential oil, thymol and carvacrol. J. Appl. Microbiol. 2001, 91, 453–462. [Google Scholar] [CrossRef] [Green Version]
  20. Radulović, N.S.; Miltojević, A.B.; McDermott, M.; Waldren, S.; Parnell, J.A.; Pinheiro, M.M.G.; Fernandes, P.D.; de Sousa Menezes, F. Identification of a new antinociceptive alkaloid isopropyl N-methylanthranilate from the essential oil of Choisya ternata Kunth. J. Ethnopharmacol. 2011, 135, 610–619. [Google Scholar] [CrossRef]
  21. Jahan, H.; Choudhary, M.I.; Atta, A.; Khan, K.M.; Ur-Rahman, A. Anthranilic Acid Derivatives: Novel Inhibitors of Protein Glycation and the Associated Oxidative Stress in the Hepatocytes. Med. Chem. 2018, 14, 516–523. [Google Scholar] [CrossRef]
  22. Asif, M. Study of Anthranylic Acid Derivatives: Mefenamic Acid and Its Various Analogues. Am. J. Med. Stud. 2014, 2, 24–30. [Google Scholar] [CrossRef]
  23. Álvarez-Martínez, F.J.; Barrajón-Catalán, E.; Herranz-López, M.; Micol, V. Antibacterial plant compounds, extracts and essential oils: An updated review on their effects and putative mechanisms of action. Phytomedicine 2021, 90, 153626. [Google Scholar] [CrossRef]
  24. Gorlenko, C.L.; Kiselev, H.Y.; Budanova, E.V.; Zamyatnin, A.A.; Ikryannikova, L.N. Plant Secondary Metabolites in the Battle of Drugs and Drug-Resistant Bacteria: New Heroes or Worse Clones of Antibiotics? Antibiotics 2020, 9, 170. [Google Scholar] [CrossRef] [Green Version]
  25. Ranfaing, J.; Dunyach-Remy, C.; Lavigne, J.-P.; Sotto, A. Propolis potentiates the effect of cranberry (Vaccinium macrocarpon) in reducing the motility and the biofilm formation of uropathogenic Escherichia coli. PLoS ONE 2018, 13, e0202609. [Google Scholar] [CrossRef]
  26. Shane, A.L.; Mody, R.K.; Crump, J.A.; Tarr, P.I.; Steiner, T.S.; Kotloff, K.; Langley, J.M.; Wanke, C.; Warren, C.A.; Cheng, A.C.; et al. 2017 Infectious Diseases Society of America Clinical Practice Guidelines for the Diagnosis and Management of Infectious Diarrhea. Clin. Infect. Dis. 2017, 65, e45–e80. [Google Scholar] [CrossRef] [Green Version]
  27. Chen, J.C.; Huang, L.J.; Wu, S.L.; Kuo, S.C.; Ho, T.Y.; Hsiang, C.Y. Ginger and its bioactive component inhibit enterotoxigenic Escherichia coli heat-labile enterotoxin-induced diarrhea in mice. J. Agric. Food. Chem. 2007, 55, 8390–8397. [Google Scholar] [CrossRef]
  28. Sugita-Konishi, Y.; Hara-Kudo, Y.; Amano, F.; Okubo, T.; Aoi, N.; Iwaki, M.; Kumagai, S. Epigallocatechin gallate and gallocatechin gallate in green tea catechins inhibit extracellular release of Vero toxin from enterohemorrhagic Escherichia coli O157:H7. Biochim. Biophys. Acta. 1999, 1472, 42–50. [Google Scholar] [CrossRef]
  29. Garg, S.S.; Gupta, J.; Sharma, S.; Sahu, D. An insight into the therapeutic applications of coumarin compounds and their mechanisms of action. Eur. J. Pharm. Sci. 2020, 152, 105424. [Google Scholar] [CrossRef]
  30. Dadi, P.K.; Ahmad, M.; Ahmad, Z. Inhibition of ATPase activity of Escherichia coli ATP synthase by polyphenols. Int. J. Biol. Macromol. 2009, 45, 72–79. [Google Scholar] [CrossRef]
  31. Amin, A.H.; Subbaiah, T.V.; Abbasi, K.M. Berberine sulfate: Antimicrobial activity, bioassay, and mode of action. Can. J. Microbiol. 1969, 15, 1067–1076. [Google Scholar] [CrossRef]
  32. Sack, R.B.; Froehlich, J.L. Berberine inhibits intestinal secretory response of Vibrio cholerae and Escherichia coli enterotoxins. Infect. Immun. 1982, 35, 471–475. [Google Scholar] [CrossRef] [Green Version]
  33. Baskaran, S.A.; Kollanoor-Johny, A.; Nair, M.S.; Venkitanarayanan, K. Efficacy of Plant-Derived Antimicrobials in Controlling Enterohemorrhagic Escherichia coli Virulence In Vitro. J. Food. Prot. 2016, 79, 1965–1970. [Google Scholar] [CrossRef]
  34. Mooyottu, S.; Kollanoor-Johny, A.; Flock, G.; Bouillaut, L.; Upadhyay, A.; Sonenshein, A.L.; Venkitanarayanan, K. Carvacrol and trans-cinnamaldehyde reduce Clostridium difficile toxin production and cytotoxicity in vitro. Int. J. Mol. Sci. 2014, 15, 4415–4430. [Google Scholar] [CrossRef] [Green Version]
  35. Chen, J.; Zhang, R.; Wang, J.; Yu, P.; Liu, Q.; Zeng, D.; Song, H.; Kuang, Z. Protective effects of baicalin on LPS-induced injury in intestinal epithelial cells and intercellular tight junctions. Can. J. Physiol. Pharmacol. 2015, 93, 233–237. [Google Scholar] [CrossRef]
  36. Dong, J.; Zhang, Y.; Chen, Y.; Niu, X.; Zhang, Y.; Yang, C.; Wang, Q.; Li, X.; Deng, X. Baicalin inhibits the lethality of Shiga-like toxin 2 in mice. Antimicrob. Agents Chemother. 2015, 59, 7054–7060. [Google Scholar] [CrossRef] [Green Version]
  37. Liu, I.X.; Durham, D.G.; Richards, R.M. Baicalin synergy with beta-lactam antibiotics against methicillin-resistant Staphylococcus aureus and other beta-lactam-resistant strains of S. aureus. J. Pharm. Pharm. 2000, 52, 361–366. [Google Scholar] [CrossRef]
  38. Zhang, Y.; Qi, Z.; Liu, Y.; He, W.; Yang, C.; Wang, Q.; Dong, J.; Deng, X. Baicalin Protects Mice from Lethal Infection by Enterohemorrhagic Escherichia coli. Front. Microbiol. 2017, 8, 395. [Google Scholar] [CrossRef] [Green Version]
  39. Thapa, D.; Louis, P.; Losa, R.; Zweifel, B.; Wallace, R.J. Essential oils have different effects on human pathogenic and commensal bacteria in mixed faecal fermentations compared with pure cultures. Microbiology 2015, 161, 441–449. [Google Scholar] [CrossRef]
  40. Khan, U.A.; Rahman, H.; Qasim, M.; Hussain, A.; Azizllah, A.; Murad, W.; Khan, Z.; Anees, M.; Adnan, M. Alkanna tinctoria leaves extracts: A prospective remedy against multidrug resistant human pathogenic bacteria. BMC. Complement. Altern. Med. 2015, 15, 127. [Google Scholar] [CrossRef] [Green Version]
  41. Zhao, Q.; Yu, Y. Antibacterial activity of matrine and oxymatrine on pathogenic bacteria causing cow endometritis in vitro. Prog. Vet. Med. 2017, 38, 65–68. [Google Scholar] [CrossRef]
  42. Nowicki, D.; Maciąg-Dorszyńska, M.; Bogucka, K.; Szalewska-Pałasz, A.; Herman-Antosiewicz, A. Various modes of action of dietary phytochemicals, sulforaphane and phenethyl isothiocyanate, on pathogenic bacteria. Sci. Rep. 2019, 9, 13677. [Google Scholar] [CrossRef] [Green Version]
  43. Gupta, S.; Yadava, J.N.S.; Tandon, J.S. Antisecretory (Antidiarrhoeal) Activity of Indian Medicinal Plants against Escherichia Coli Enterotoxin-Induced Secretion in Rabbit and Guinea Pig Ileal Loop Models. Int. J. Pharmacogn. 1993, 31, 198–204. [Google Scholar] [CrossRef]
  44. Pereira da Cruz, R.; Sampaio de Freitas, T.; Socorro Costa, M.D.; Lucas Dos Santos, A.T.; Ferreira Campina, F.; Pereira, R.L.S.; Bezerra, J.W.A.; Quintans-Júnior, L.J.; De Souza Araújo, A.A.; Júnior, J.P.S.; et al. Effect of α-Bisabolol and Its β-Cyclodextrin Complex as TetK and NorA Efflux Pump Inhibitors in Staphylococcus aureus Strains. Antibiot 2020, 9, 28. [Google Scholar] [CrossRef] [Green Version]
  45. Kaper, J.B.; Nataro, J.P.; Mobley, H.L. Pathogenic Escherichia coli. Nat. Rev. Microbiol. 2004, 2, 123–140. [Google Scholar] [CrossRef]
  46. Rangel, J.M.; Sparling, P.H.; Crowe, C.; Griffin, P.M.; Swerdlow, D.L. Epidemiology of Escherichia coli O157: H7 outbreaks, united states, 1982–2002. Emerg. Infect. Dis. 2005, 11, 603. [Google Scholar] [CrossRef]
  47. Chen, J.-C.; Ho, T.-Y.; Chang, Y.-S.; Wu, S.-L.; Hsiang, C.-Y. Anti-diarrheal effect of Galla Chinensis on the Escherichia coli heat-labile enterotoxin and ganglioside interaction. J. Ethnopharmacol. 2006, 103, 385–391. [Google Scholar] [CrossRef]
  48. Tian, F.; Li, B.; Ji, B.; Yang, J.; Zhang, G.; Chen, Y.; Luo, Y. Antioxidant and antimicrobial activities of consecutive extracts from Galla chinensis: The polarity affects the bioactivities. Food. Chem. 2009, 113, 173–179. [Google Scholar] [CrossRef]
  49. Ahn, Y.J.; Lee, C.O.; Kweon, J.H.; Ahn, J.W.; Park, J.H. Growth-inhibitory effects of Galla Rhois-derived tannins on intestinal bacteria. J. Appl. Microbiol. 1998, 84, 439–443. [Google Scholar] [CrossRef]
  50. Song, X.; Yang, Y.; Li, J.; He, M.; Zou, Y.; Jia, R.; Li, L.; Hang, J.; Cui, M.; Bai, L.; et al. Tannins extract from Galla Chinensis can protect mice from infection by Enterotoxigenic Escherichia coli O101. BMC. Complement. Med. Ther. 2021, 21, 84. [Google Scholar] [CrossRef]
  51. Caturla, N.; Vera-Samper, E.; Villalaín, J.; Mateo, C.R.; Micol, V. The relationship between the antioxidant and the antibacterial properties of galloylated catechins and the structure of phospholipid model membranes. Free. Radic. Biol. Med. 2003, 34, 648–662. [Google Scholar] [CrossRef]
  52. Friedman, M. Overview of antibacterial, antitoxin, antiviral, and antifungal activities of tea flavonoids and teas. Mol. Nutr. Food. Res. 2007, 51, 116–134. [Google Scholar] [CrossRef]
  53. Zeller, W.E. Activity, purification, and analysis of condensed tannins: Current state of affairs and future endeavors. Crop. Sci. 2019, 59, 886–904. [Google Scholar] [CrossRef]
  54. Doughari, J.H.; Ndakidemi, P.A.; Human, I.S.; Benade, S. Antioxidant, antimicrobial and antiverotoxic potentials of extracts of Curtisia dentata. J. Ethnopharmacol. 2012, 141, 1041–1050. [Google Scholar] [CrossRef]
  55. Khameneh, B.; Eskin, N.A.M.; Iranshahy, M.; Fazly Bazzaz, B.S. Phytochemicals: A Promising Weapon in the Arsenal against Antibiotic-Resistant Bacteria. Antibiotics 2021, 10, 1044. [Google Scholar] [CrossRef]
  56. Di Martino, P.; Agniel, R.; David, K.; Templer, C.; Gaillard, J.L.; Denys, P.; Botto, H. Reduction of Escherichia coli adherence to uroepithelial bladder cells after consumption of cranberry juice: A double-blind randomized placebo-controlled cross-over trial. World J. Urolog. 2006, 24, 21–27. [Google Scholar] [CrossRef]
  57. Enioutina, E.Y.; Teng, L.; Fateeva, T.V.; Brown, J.C.S.; Job, K.M.; Bortnikova, V.V.; Krepkova, L.V.; Gubarev, M.I.; Sherwin, C.M.T. Phytotherapy as an alternative to conventional antimicrobials: Combating microbial resistance. Expert. Rev. Clin. Pharmacol. 2017, 10, 1203–1214. [Google Scholar] [CrossRef]
  58. Petronio Petronio, G.; Cutuli, M.A.; Magnifico, I.; Venditti, N.; Pietrangelo, L.; Vergalito, F.; Pane, A.; Scapagnini, G.; Di Marco, R. In Vitro and In Vivo Biological Activity of Berberine Chloride against Uropathogenic, E. coli Strains Using Galleria mellonella as a Host Model. Molecules 2020, 25, 5010. [Google Scholar] [CrossRef]
  59. Xu, C.; Wang, F.; Huang, F.; Yang, M.; He, D.; Deng, L. Targeting effect of berberine on type I fimbriae of Salmonella Typhimurium and its effective inhibition of biofilm. Appl. Microbiol. Biotechnol. 2021, 105, 1563–1573. [Google Scholar] [CrossRef]
  60. Domadia, P.N.; Bhunia, A.; Sivaraman, J.; Swarup, S.; Dasgupta, D. Berberine targets assembly of Escherichia coli cell division protein FtsZ. Biochemistry 2008, 47, 3225–3234. [Google Scholar] [CrossRef]
  61. Boberek, J.M.; Stach, J.; Good, L. Genetic evidence for inhibition of bacterial division protein FtsZ by berberine. PLoS ONE 2010, 5, e13745. [Google Scholar] [CrossRef]
  62. Jin, J.L.; Hua, G.Q.; Meng, Z.; Gao, P.J. J. Antibacterial Mechanisms of Berberine and Reasons for Little Resistance of Bacteria. Chin. Herb. Med. 2010, 3, 27–35. [Google Scholar] [CrossRef]
  63. Stermitz, F.R.; Lorenz, P.; Tawara, J.N.; Zenewicz, L.A.; Lewis, K. Synergy in a medicinal plant: Antimicrobial action of berberine potentiated by 5′-methoxyhydnocarpin, a multidrug pump inhibitor. Proc. Natl. Acad. Sci. USA 2000, 97, 1433–1437. [Google Scholar] [CrossRef] [Green Version]
  64. Su, F.; Wang, J. Berberine inhibits the MexXY-OprM efflux pump to reverse imipenem resistance in a clinical carbapenem-resistant Pseudomonas aeruginosa isolate in a planktonic state. Exp. Med. 2018, 15, 467–472. [Google Scholar] [CrossRef] [Green Version]
  65. Sun, T.; Xiaodong, L.; Hong, J.; Liu, C.; Zhang, X.-L.; Zheng, J.-P.; Xu, Y.-J.; Ou, Z.-Y.; Zheng, J.-L.; Yu, D.-J. Inhibitory Effect of Two Traditional Chinese Medicine Monomers, Berberine and Matrine, on the Quorum Sensing System of Antimicrobial-Resistant Escherichia coli. Front. Microbiol. 2019, 10, 2584. [Google Scholar] [CrossRef]
  66. Shilei, Z.; Ze, J.; Xianghe, Z.; Chunguang, W.; Tie, Z. The Effect of Berberine on the Transcriptome and Proteome of E. coli. bioRxiv 2018, 318733. [Google Scholar] [CrossRef] [Green Version]
  67. Lorenz, C.; Dougherty, T.J.; Lory, S. Transcriptional Responses of Escherichia coli to a Small-Molecule Inhibitor of LolCDE, an Essential Component of the Lipoprotein Transport Pathway. J. Bacteriol. 2016, 198, 3162–3175. [Google Scholar] [CrossRef] [Green Version]
  68. Thong, S.; Ercan, B.; Torta, F.; Fong, Z.Y.; Wong, H.Y.A.; Wenk, M.R.; Chng, S.-S. Defining key roles for auxiliary proteins in an ABC transporter that maintains bacterial outer membrane lipid asymmetry. eLife 2016, 5, e19042. [Google Scholar] [CrossRef]
  69. Majdalani, N.; Heck, M.; Stout, V.; Gottesman, S. Role of RcsF in signaling to the Rcs phosphorelay pathway in Escherichia coli. J. Bacteriol. 2005, 187, 6770–6778. [Google Scholar] [CrossRef] [Green Version]
  70. Ghachi, M.E.; Derbise, A.; Bouhss, A.; Mengin-Lecreulx, D. Identification of Multiple Genes Encoding Membrane Proteins with Undecaprenyl Pyrophosphate Phosphatase (UppP) Activity in Escherichia coli*. J. Biol. Chem. 2005, 280, 18689–18695. [Google Scholar] [CrossRef] [Green Version]
  71. Gill, A.O.; Holley, R.A. Mechanisms of bactericidal action of cinnamaldehyde against Listeria monocytogenes and of eugenol against L. monocytogenes and Lactobacillus sakei. Appl. Env. Microbiol. 2004, 70, 5750–5755. [Google Scholar] [CrossRef] [Green Version]
  72. Dineen, S.S.; McBride, S.M.; Sonenshein, A.L. Integration of metabolism and virulence by Clostridium difficile CodY. J. Bacteriol. 2010, 192, 5350–5362. [Google Scholar] [CrossRef] [Green Version]
  73. Mooyottu, S.; Flock, G.; Upadhyay, A.; Upadhyaya, I.; Maas, K.; Venkitanarayanan, K. Protective Effect of Carvacrol against Gut Dysbiosis and Clostridium difficile Associated Disease in a Mouse Model. Front. Microbiol. 2017, 8, 625. [Google Scholar] [CrossRef] [Green Version]
  74. Roshan, N.; Riley, T.V.; Hammer, K.A. Antimicrobial activity of natural products against Clostridium difficile in vitro. J. Appl. Microbiol. 2017, 123, 92–103. [Google Scholar] [CrossRef] [Green Version]
  75. Roshan, N.; Riley, T.V.; Knight, D.R.; Hammer, K.A. Effect of natural products on the production and activity of Clostridium difficile toxins in vitro. Sci. Rep. 2018, 8, 15735. [Google Scholar] [CrossRef] [Green Version]
  76. Lv, Z.; Peng, G.; Liu, W.; Xu, H.; Su, J. Berberine blocks the relapse of Clostridium difficile infection in C57BL/6 mice after standard vancomycin treatment. Antimicrob. Agents. Chemother. 2015, 59, 3726–3735. [Google Scholar] [CrossRef] [Green Version]
  77. Pal, R.; Seleem, M.N. Discovery of a novel natural product inhibitor of Clostridioides difficile with potent activity in vitro and in vivo. PLoS ONE 2022, 17, e0267859. [Google Scholar] [CrossRef]
  78. Pal, R.; Dai, M.; Seleem, M.N. High-throughput screening identifies a novel natural product-inspired scaffold capable of inhibiting Clostridioides difficile in vitro. Sci. Rep. 2021, 11, 1–7. [Google Scholar] [CrossRef]
  79. Shilling, M.; Matt, L.; Rubin, E.; Visitacion, M.P.; Haller, N.A.; Grey, S.F.; Woolverton, C.J. Antimicrobial effects of virgin coconut oil and its medium-chain fatty acids on Clostridium difficile. J. Med. Food. 2013, 16, 1079–1085. [Google Scholar] [CrossRef] [Green Version]
  80. Fraternale, D.; Flamini, G.; Ricci, D. Essential oil composition and antimicrobial activity of Angelica archangelica L.(Apiaceae) roots. J. Med. Food. 2014, 17, 1043–1047. [Google Scholar] [CrossRef]
  81. Finegold, S.M.; Summanen, P.H.; Corbett, K.; Downes, J.; Henning, S.M.; Li, Z. Pomegranate extract exhibits in vitro activity against Clostridium difficile. Nutrition 2014, 30, 1210–1212. [Google Scholar] [CrossRef]
  82. Nadeau, É.; Messier, S.; Quessy, S. Comparison of Campylobacter isolates from poultry and humans: Association between in vitro virulence properties, biotypes, and pulsed-field gel electrophoresis clusters. Appl. Environ. Microbiol. 2003, 69, 6316–6320. [Google Scholar] [CrossRef] [Green Version]
  83. Mahajan, S.; Rodgers, F.G. Isolation, characterization, and host-cell-binding properties of a cytotoxin from Campylobacter jejuni. J. Clin. Microbiol. 1990, 28, 1314–1320. [Google Scholar] [CrossRef] [Green Version]
  84. Valtierra-Rodríguez, D.; Heredia, N.L.; García, S.; Sánchez, E. Reduction of Campylobacter jejuni and Campylobacter coli in poultry skin by fruit extracts. J. Food. Prot. 2010, 73, 477–482. [Google Scholar] [CrossRef]
  85. Castillo, S.L.; Heredia, N.; Contreras, J.F.; García, S. Extracts of Edible and Medicinal Plants in Inhibition of Growth, Adherence, and Cytotoxin Production of Campylobacter jejuni and Campylobacter coli. J. Food. Sci. 2011, 76, M421–M426. [Google Scholar] [CrossRef]
  86. Toda, M.; Okubo, S.; Ohnishi, R.; Shimamura, T. Antibacterial and bactericidal activities of Japanese green tea. Nihon. Saikingaku. Zasshi. Jpn. J. Bacteriol. 1989, 44, 669–672. [Google Scholar] [CrossRef]
  87. Zhu, J.; Hua, X.; Hou, J.; Zhao, W. The virulence determinants of Campylobacter jejuni and its ability to colonize hosts. Rev. Med. Microbiol. 2008, 19, 13–18. [Google Scholar] [CrossRef]
  88. Lengsfeld, C.; Faller, G.; Hensel, A. Okra polysaccharides inhibit adhesion of Campylobacter jejuni to mucosa isolated from poultry in vitro but not in vivo. Anim. Feed. Sci. Technol. 2007, 135, 113–125. [Google Scholar] [CrossRef]
  89. Samy, R.; Gopalakrishnakone, P. Review: Therapeutic potential of plants asanti-microbials for drug discovery. J. Evid. -Based Compl. Altern. Med. 2008, 10, 1093. [Google Scholar] [CrossRef] [Green Version]
  90. Stickel, F.; Schuppan, D. Herbal medicine in the treatment of liver diseases. Dig. Liver. Dis. 2007, 39, 293–304. [Google Scholar] [CrossRef]
  91. CDC. Diarrhea: Common Illness, Global Killer. Available online: https://stacks.cdc.gov/view/cdc/13557 (accessed on 11 December 2022).
  92. Groombridge, B.; Jenkins, M. World Atlas of Biodiversity: Earth’s Living Resources in the 21st Century; University of California Press: Berkeley, CA, USA, 2002; p. 340. ISBN 0-520-23668-8. [Google Scholar] [CrossRef] [Green Version]
  93. WHO. WHO Guidelines on Safety Monitoring of Herbal Medicines in Pharmacovigilance Systems; World Health Organization: Geneva, Switzerland, 2004.
  94. Silva, M.; Bittner, M.; Mueller, M.S.; Mechler, E. Medicinal Plants in Tropical Countries, Traditional Use—Experience—Facts; Georg Thieme Verlag: Stuttgart, Germany, 2005; ISBN 1-58890-253-6. [Google Scholar] [CrossRef]
  95. Palombo, E.A. Phytochemicals from traditional medicinal plants used in the treatment of diarrhoea: Modes of action and effects on intestinal function. Phytother. Res. 2006, 20, 717–724. [Google Scholar] [CrossRef]
  96. Hirudkar, J.R.; Parmar, K.M.; Prasad, R.S.; Sinha, S.K.; Lomte, A.D.; Itankar, P.R.; Prasad, S.K. The antidiarrhoeal evaluation of Psidium guajava L. against enteropathogenic Escherichia coli induced infectious diarrhoea. J. Ethnopharmacol. 2020, 251, 112561. [Google Scholar] [CrossRef]
  97. Gutiérrez, R.M.P.; Mitchell, S.; Solis, R.V. Psidium guajava: A review of its traditional uses, phytochemistry and pharmacology. J. Ethnopharmacol. 2008, 117, 1–27. [Google Scholar] [CrossRef]
  98. Galvez, J.; Zarzuelo, A.; Crespo, M.E.; Lorente, M.D.; Ocete, M.A.; Jiménez, J. Antidiarrhoeic activity of Euphorbia hirta extract and isolation of an active flavonoid constituent. Planta. Med. 1993, 59, 333–336. [Google Scholar] [CrossRef]
  99. Gupta, P.; Birdi, T. Psidium guajava leaf extract prevents intestinal colonization of Citrobacter rodentium in the mouse model. J. Ayurveda Integr. Med. 2015, 6, 50. [Google Scholar] [CrossRef] [Green Version]
  100. Shittu, O.B.; Ajayi, O.L.; Bankole, S.O.; Popoola, T.O. Intestinal ameliorative effects of traditional Ogi-tutu, Vernonia amygdalina and Psidium guajava in mice infected with Vibrio cholera. Afr. Health. Sci. 2016, 16, 620–628. [Google Scholar] [CrossRef] [Green Version]
  101. Lozoya, X.; Reyes-Morales, H.; Chávez-Soto, M.A.; del Carmen Martínez-García, M.a.; Soto-González, Y.; Doubova, S.V. Intestinal anti-spasmodic effect of a phytodrug of Psidium guajava folia in the treatment of acute diarrheic disease. J. Ethnopharmacol. 2002, 83, 19–24. [Google Scholar] [CrossRef]
  102. Manekeng, H.T.; Mbaveng, A.T.; Ntyam Mendo, S.A.; Agokeng, A.D.; Kuete, V. Evaluation of Acute and Subacute Toxicities of Psidium guajava Methanolic Bark Extract: A Botanical with In Vitro Antiproliferative Potential. Evid. Based Complement. Altern. Med. 2019, 2019, 8306986. [Google Scholar] [CrossRef] [Green Version]
  103. Chauhan, R.K.; Jain, A.M.; Dube, M.K.; Bhandari, B. A combination of sulfadimidine, neomycin and berberine in the treatment of infectious diarrhoea. Indian. J. Pediatr. 1969, 36, 242–244. [Google Scholar] [CrossRef]
  104. Chauhan, R.K.S.; Jain, A.M.; Bhandari, B. Berberine in the treatment of childhood diarrhoea. Indian. J. Pediatr. 1970, 37, 577–579. [Google Scholar] [CrossRef]
  105. Sharda, D.C. Berberine in the treatment of diarrhoea of infancy and childhood. J. Indian. Med. Assoc. 1970, 54, 22–24. [Google Scholar] [CrossRef]
  106. Sharma, R.; Joshi, C.K.; Goyal, R.K. Berberine tannate in acute diarrhoea. Indian. Pediatr. 1970, 7, 496–501. [Google Scholar]
  107. Rabbani, G.H.; Butler, T.; Knight, J.; Sanyal, S.C.; Alam, K. Randomized controlled trial of berberine sulfate therapy for diarrhea due to enterotoxigenic Escherichia coli and Vibrio cholerae. J. Infect. Dis. 1987, 155, 979–984. [Google Scholar] [CrossRef]
  108. Yamamoto, K.; Takase, H.; Abe, K.; Saito, Y.; Suzuki, A. [Pharmacological studies on antidiarrheal effects of a preparation containing berberine and geranii herba]. Nihon Yakurigaku Zasshi 1993, 101, 169–175. [Google Scholar] [CrossRef]
  109. Tsai, C.S.; Ochillo, R.F. Pharmacological effects of berberine on the longitudinal muscle of the guinea-pig isolated ileum. Arch. Int. Pharm. 1991, 310, 116–131. [Google Scholar]
  110. Zhang, Y.; Wang, X.; Sha, S.; Liang, S.; Zhao, L.; Liu, L.; Chai, N.; Wang, H.; Wu, K. Berberine increases the expression of NHE3 and AQP4 in sennosideA-induced diarrhoea model. Fitoterapia 2012, 83, 1014–1022. [Google Scholar] [CrossRef]
  111. Cernáková, M.; Kostálová, D. Antimicrobial activity of berberine--A constituent of Mahonia aquifolium. Folia. Microbiol. 2002, 47, 375–378. [Google Scholar] [CrossRef]
  112. Zhou, F.F.; Wu, S.; Klena, J.D.; Huang, H.H. Clinical characteristics of Clostridium difficile infection in hospitalized patients with antibiotic-associated diarrhea in a university hospital in China. Eur. J. Clin. Microbiol. Infect. Dis. 2014, 33, 1773–1779. [Google Scholar] [CrossRef] [Green Version]
  113. He, C.; Gao, P. Meta-analysis of antibiotic-associated diarrhea treated by traditional Chinese medicine. Chin. J. TCM. WM. Crit. Care 2010, 17, 69–72. [Google Scholar] [CrossRef]
  114. Shu, Q.; Peng, S.; Feng, Y.; Zhang, Z.; Xie, Y.; Zhou, S. The influences of Chinese herbal compounds on in vitro expression of toxic genes tcd A/tcd B of Clostridium difficile. Chin. J. Microecol. 2013, 25, 373–380. [Google Scholar]
  115. Singh, N.; Sharma, B. Toxicological Effects of Berberine and Sanguinarine. Front. Mol. Biosci. 2018, 5, 21. [Google Scholar] [CrossRef] [Green Version]
  116. Rad, S.Z.K.; Rameshrad, M.; Hosseinzadeh, H. Toxicology effects of Berberis vulgaris (barberry) and its active constituent, berberine: A review. Iran. J. Basic. Med. Sci. 2017, 20, 516–529. [Google Scholar] [CrossRef]
  117. Ai, X.; Yu, P.; Peng, L.; Luo, L.; Liu, J.; Li, S.; Lai, X.; Luan, F.; Meng, X. Berberine: A Review of its Pharmacokinetics Properties and Therapeutic Potentials in Diverse Vascular Diseases. Front. Pharmacol. 2021, 12, 3104. [Google Scholar] [CrossRef]
  118. Baska, A.; Leis, K.; Gałązka, P. Berberine in the Treatment of Diabetes Mellitus: A Review. Endocr. Metab. Immune. Disord. Drug Targets. 2021, 21, 1379–1386. [Google Scholar] [CrossRef]
  119. Ekor, M. The growing use of herbal medicines: Issues relating to adverse reactions and challenges in monitoring safety. Front. Pharmacol. 2014, 4, 177. [Google Scholar] [CrossRef] [Green Version]
  120. Steffen, R.; Mathewson, J.J.; Ericsson, C.D.; DuPont, H.L.; Helminger, A.; Balm, T.K.; Wolff, K.; Witassek, F. Travelers’ diarrhea in West Africa and Mexico: Fecal transport systems and liquid bismuth subsalicylate for self-therapy. J. Infect. Dis. 1988, 157, 1008–1013. [Google Scholar] [CrossRef] [Green Version]
  121. DuPont, H.L.; Flores Sanchez, J.; Ericsson, C.D.; Mendiola Gomez, J.; DuPont, M.W.; Cruz Luna, A.; Mathewson, J.J. Comparative efficacy of loperamide hydrochloride and bismuth subsalicylate in the management of acute diarrhea. Am. J. Med. 1990, 88, 15s–19s. [Google Scholar] [CrossRef]
  122. Figueroa-Quintanilla, D.; Salazar-Lindo, E.; Sack, R.B.; León-Barúa, R.; Sarabia-Arce, S.; Campos-Sánchez, M.; Eyzaguirre-Maccan, E. A controlled trial of bismuth subsalicylate in infants with acute watery diarrheal disease. N. Engl. J. Med. 1993, 328, 1653–1658. [Google Scholar] [CrossRef]
  123. Pitz, A.M.; Park, G.W.; Lee, D.; Boissy, Y.L.; Vinjé, J. Antimicrobial activity of bismuth subsalicylate on Clostridium difficile, Escherichia coli O157:H7, norovirus, and other common enteric pathogens. Gut Microbes. 2015, 6, 93–100. [Google Scholar] [CrossRef] [Green Version]
  124. Chowdhury, H.R.; Yunus, M.; Zaman, K.; Rahman, A.; Faruque, S.M.; Lescano, A.G.; Sack, R.B. The efficacy of bismuth subsalicylate in the treatment of acute diarrhoea and the prevention of persistent diarrhoea. Acta. Paediatr. 2001, 90, 605–610. [Google Scholar] [CrossRef]
  125. Halani, S.; Wu, P.E. Salicylate toxicity from chronic bismuth subsalicylate use. BMJ. Case Rep. 2020, 13, e236929. [Google Scholar] [CrossRef]
  126. Thakkar, S.; Anklam, E.; Xu, A.; Ulberth, F.; Li, J.; Li, B.; Hugas, M.; Sarma, N.; Crerar, S.; Swift, S.; et al. Regulatory landscape of dietary supplements and herbal medicines from a global perspective. Regul. Toxicol. Pharmacol. 2020, 114, 104647. [Google Scholar] [CrossRef]
  127. Alibi, S.; Crespo, D.; Navas, J. Plant-Derivatives Small Molecules with Antibacterial Activity. Antibiotics 2021, 10, 231. [Google Scholar] [CrossRef]
  128. Lauche, R.; Kumar, S.; Hallmann, J.; Lüdtke, R.; Rampp, T.; Dobos, G.; Langhorst, J. Efficacy and safety of Ayurvedic herbs in diarrhoea-predominant irritable bowel syndrome: A randomised controlled crossover trial. Complement. Ther. Med. 2016, 26, 171–177. [Google Scholar] [CrossRef]
  129. Leung, W.K.; Wu, J.C.Y.; Liang, S.M.; Chan, L.S.; Chan, F.K.L.; Xie, H.; Fung, S.S.L.; Hui, A.J.; Wong, V.W.S.; Che, C.-T.; et al. Treatment of Diarrhea-Predominant Irritable Bowel Syndrome with Traditional Chinese Herbal Medicine: A Randomized Placebo-Controlled Trial. Off. J. Am. Coll. Gastroenterol. ACG. 2006, 101, 1574–1580. [Google Scholar] [CrossRef]
  130. Tal-Dia, A.; Toure, K.; Sarr, O.; Sarr, M.; Cisse, M.F.; Garnier, P.; Wone, I. A baobab solution for the prevention and treatment of acute dehydration in infantile diarrhea. Dakar. Med. 1997, 42, 68–73. [Google Scholar]
  131. Khin Maung, U.; Myo, K.; Nyunt Nyunt, W.; Aye, K.; Tin, U. Clinical trial of berberine in acute watery diarrhoea. Br. Med. J. Clin. Res. Ed. 1985, 291, 1601–1605. [Google Scholar] [CrossRef] [Green Version]
  132. Chakraborty, P. Herbal genomics as tools for dissecting new metabolic pathways of unexplored medicinal plants and drug discovery. Biochim. Open. 2018, 6, 9–16. [Google Scholar] [CrossRef]
  133. Alseekh, S.; Aharoni, A.; Brotman, Y.; Contrepois, K.; D’Auria, J.; Ewald, J.J.; Ewald, J.C..; Fraser, P.D.; Giavalisco, P.; Hall, R.D.; et al. Mass spectrometry-based metabolomics: A guide for annotation, quantification and best reporting practices. Nat. Methods. 2021, 18, 747–756. [Google Scholar] [CrossRef]
  134. Wang, M.; Lamers, R.J.A.; Korthout, H.A.; van Nesselrooij, J.H.; Witkamp, R.F.; van der Heijden, R.; Voshol, P.J.; Havekes, L.M.; Verpoorte, R.; van der Greef, J. Metabolomics in the context of systems biology: Bridging traditional Chinese medicine and molecular pharmacology. Phytother. Res. Int. J. Devoted. Pharmacol. Toxicol. Eval. Nat. Prod. Deriv. 2005, 19, 173–182. [Google Scholar] [CrossRef]
  135. Medema, M.H.; Osbourn, A. Computational genomic identification and functional reconstitution of plant natural product biosynthetic pathways. Nat. Prod. Rep. 2016, 33, 951–962. [Google Scholar] [CrossRef] [Green Version]
  136. Goodacre, R.; Vaidyanathan, S.; Dunn, W.B.; Harrigan, G.G.; Kell, D.B. Metabolomics by numbers: Acquiring and understanding global metabolite data. TRENDS Biotechnol. 2004, 22, 245–252. [Google Scholar] [CrossRef]
  137. Zhou, Z.; Luo, M.; Zhang, H.; Yin, Y.; Cai, Y.; Zhu, Z.-J. Metabolite annotation from knowns to unknowns through knowledge-guided multi-layer metabolic networking. Nat. Commun. 2022, 13, 6656. [Google Scholar] [CrossRef]
  138. Yi, Z.-B.; Yu, Y.; Liang, Y.-Z.; Zeng, B. Evaluation of the antimicrobial mode of berberine by LC/ESI-MS combined with principal component analysis. J. Pharm. Biomed. Anal. 2007, 44, 301–304. [Google Scholar] [CrossRef]
  139. Budeyri Gokgoz, N.; Avci, F.G.; Yoneten, K.K.; Alaybeyoglu, B.; Ozkirimli, E.; Sayar, N.A.; Kazan, D.; Sariyar Akbulut, B. Response of Escherichia coli to prolonged berberine exposure. Microb. Drug. Resist. 2017, 23, 531–544. [Google Scholar] [CrossRef]
  140. Karaosmanoglu, K.; Sayar, N.A.; Kurnaz, I.A.; Akbulut, B.S. Assessment of berberine as a multi-target antimicrobial: A multi-omics study for drug discovery and repositioning. Omics. A. J. Integr. Biol. 2014, 18, 42–53. [Google Scholar] [CrossRef]
  141. Ozbalci, Ç.; Unsal, Ç.; Kazan, D.; Sariyar-Akbulut, B. Proteomic response of Escherichia coli to the alkaloid extract of Papaver polychaetum. Ann. Microbiol. 2010, 60, 709–717. [Google Scholar] [CrossRef]
  142. Yu, Y.; Yi, Z.B.; Liang, Y.Z. Validate antibacterial mode and find main bioactive components of traditional Chinese medicine Aquilegia oxysepala. Bioorg. Med. Chem. Lett. 2007, 17, 1855–1859. [Google Scholar] [CrossRef]
  143. Pandita, D.; Pandita, A.; Wani, S.H.; Abdelmohsen, S.A.M.; Alyousef, H.A.; Abdelbacki, A.M.M.; Al-Yafrasi, M.A.; Al-Mana, F.A.; Elansary, H.O. Crosstalk of Multi-Omics Platforms with Plants of Therapeutic Importance. Cells 2021, 10, 1296. [Google Scholar] [CrossRef]
  144. Dinda, B.; Chowdhury, D.R.; Mohanta, B.C. Naturally Occurring Iridoids, Secoiridoids and Their Bioactivity. An Updated Review, Part 3. Chem. Pharm. Bull. 2009, 57, 765–796. [Google Scholar] [CrossRef] [Green Version]
  145. Geu-Flores, F.; Sherden, N.H.; Courdavault, V.; Burlat, V.; Glenn, W.S.; Wu, C.; Nims, E.; Cui, Y.; O’Connor, S.E. An alternative route to cyclic terpenes by reductive cyclization in iridoid biosynthesis. Nature 2012, 492, 138–142. [Google Scholar] [CrossRef]
  146. Giddings, L.-A.; Liscombe, D.K.; Hamilton, J.P.; Childs, K.L.; DellaPenna, D.; Buell, C.R.; O’Connor, S.E. A stereoselective hydroxylation step of alkaloid biosynthesis by a unique cytochrome P450 in Catharanthus roseus. J. Biol. Chem. 2011, 286, 16751–16757. [Google Scholar] [CrossRef] [Green Version]
  147. Tundis, R.; Loizzo, M.R.; Menichini, F.; Statti, G.A.; Menichini, F. Biological and pharmacological activities of iridoids: Recent developments. Mini. Rev. Med. Chem. 2008, 8, 399–420. [Google Scholar] [CrossRef]
  148. Lao, Y.; Wang, X.; Xu, N.; Zhang, H.; Xu, H. Application of proteomics to determine the mechanism of action of traditional Chinese medicine remedies. J. Ethnopharmacol. 2014, 155, 1–8. [Google Scholar] [CrossRef] [Green Version]
  149. Wang, Y.; Yu, R.-Y.; He, Q.-Y. Proteomic analysis of anticancer TCMs targeted at mitochondria. Evid.-Based Complement. Altern. Med. 2015, 2015, 539260. [Google Scholar] [CrossRef] [Green Version]
  150. Wang, L.; Tian, R. Proteomic profiling of salmonella under berberine stress. IOP. Conf. Ser. Mater. Sci. Eng. 2020, 768, 052054. [Google Scholar] [CrossRef]
  151. Dwyer, D.J.; Belenky, P.A.; Yang, J.H.; MacDonald, I.C.; Martell, J.D.; Takahashi, N.; Chan, C.T.; Lobritz, M.A.; Braff, D.; Schwarz, E.G.; et al. Antibiotics induce redox-related physiological alterations as part of their lethality. Proc. Natl. Acad. Sci. USA 2014, 111, E2100–E2109. [Google Scholar] [CrossRef] [Green Version]
  152. Dwyer, D.J.; Camacho, D.M.; Kohanski, M.A.; Callura, J.M.; Collins, J.J. Antibiotic-induced bacterial cell death exhibits physiological and biochemical hallmarks of apoptosis. Mol. Cell. 2012, 46, 561–572. [Google Scholar] [CrossRef] [Green Version]
  153. Cohen, N.R.; Lobritz, M.A.; Collins, J.J. Microbial persistence and the road to drug resistance. Cell. Host. Microbe. 2013, 13, 632–642. [Google Scholar] [CrossRef] [Green Version]
  154. Van Acker, H.; Van Dijck, P.; Coenye, T. Molecular mechanisms of antimicrobial tolerance and resistance in bacterial and fungal biofilms. Trends. Microbiol. 2014, 22, 326–333. [Google Scholar] [CrossRef]
  155. Bent, S. Herbal medicine in the United States: Review of efficacy, safety, and regulation: Grand rounds at University of California, San Francisco Medical Center. J. Gen. Intern. Med. 2008, 23, 854–859. [Google Scholar] [CrossRef] [Green Version]
  156. Tangjitman, K.; Wongsawad, C.; Kamwong, K.; Sukkho, T.; Trisonthi, C. Ethnomedicinal plants used for digestive system disorders by the Karen of northern Thailand. J. Ethnobiol. Ethnomed. 2015, 11, 27. [Google Scholar] [CrossRef] [Green Version]
  157. Yakubu, M.T.; Nurudeen, Q.O.; Salimon, S.S.; Yakubu, M.O.; Jimoh, R.O.; Nafiu, M.O.; Akanji, M.A.; Oladiji, A.T.; Williams, F.E. Antidiarrhoeal Activity of Musa paradisiaca Sap in Wistar Rats. Evid. Based. Complement. Altern. Med. 2015, 2015, 683726. [Google Scholar] [CrossRef] [Green Version]
  158. Ugbogu, E.A.; Ude, V.C.; Elekwa, I.; Arunsi, U.O.; Uche-Ikonne, C.; Nwakanma, C. Toxicological profile of the aqueous-fermented extract of Musa paradisiaca in rats. Avicenna. J. Phytomed. 2018, 8, 478–487. [Google Scholar]
  159. Hossain, F.; Mostofa, M.G.; Alam, A.K. Traditional uses and pharmacological activities of the genus leea and its phytochemicals: A review. Heliyon 2021, 7, e06222. [Google Scholar] [CrossRef]
  160. Mishra, G.; Khosa, R.; Singh, P.; Jha, K. Hepatoprotective activity of ethanolic extract of Leea indica (Burm. f.) Merr.(Leeaceae) stem bark against paracetamol induced liver toxicity in rats. Niger. J. Exp. Clin. Biosci. 2014, 2, 59. [Google Scholar] [CrossRef]
  161. Dandiya, P.; Cullumbine, H. Studies on Acorus calamus (III): Some pharmacological actions of the volatile oil. J. Pharmacol. Exp. Ther. 1959, 125, 353–359. [Google Scholar]
  162. Sharma, V.; Sharma, R.; Gautam, D.S.; Kuca, K.; Nepovimova, E.; Martins, N. Role of Vacha (Acorus calamus Linn.) in Neurological and Metabolic Disorders: Evidence from Ethnopharmacology, Phytochemistry, Pharmacology and Clinical Study. J. Clin. Med. 2020, 9, 1176. [Google Scholar] [CrossRef] [Green Version]
  163. Oli, A.N.; Obaji, M.; Enweani, I.B. Combinations of Alchornea cordifolia, Cassytha filiformis and Pterocarpus santalinoides in diarrhoegenic bacterial infections. BMC. Res. Notes 2019, 12, 649. [Google Scholar] [CrossRef]
  164. Libman, A.; Bouamanivong, S.; Southavong, B.; Sydara, K.; Soejarto, D. Medicinal plants: An important asset to health care in a region of Central Laos. J. Ethnopharmacol. 2006, 106, 303–311. [Google Scholar] [CrossRef]
  165. Nikkhah Bodagh, M.; Maleki, I.; Hekmatdoost, A. Ginger in gastrointestinal disorders: A systematic review of clinical trials. Food. Sci. Nutr. 2019, 7, 96–108. [Google Scholar] [CrossRef]
  166. Weidner, M.S.; Sigwart, K. Investigation of the teratogenic potential of a Zingiber officinale extract in the rat. Reprod. Toxicol. 2000, 15, 75–80. [Google Scholar] [CrossRef]
  167. Chivapat, S.; Chavalittumrong, P.; Attawish, A.; Bansiddhi, J.; Padungpat, S. Chronic toxicity of Thunbergia laurifolia Lindl. extract. J. Tradit. Thai. Altern. Med. 2009, 7, 17–25. [Google Scholar]
  168. Alshehri, M.M.; Quispe, C.; Herrera-Bravo, J.; Sharifi-Rad, J.; Tutuncu, S.; Aydar, E.F.; Topkaya, C.; Mertdinc, Z.; Ozcelik, B.; Aital, M.; et al. A Review of Recent Studies on the Antioxidant and Anti-Infectious Properties of Senna Plants. Oxid. Med. Cell. Longev. 2022, 2022, 6025900. [Google Scholar] [CrossRef]
  169. Nadal, S.R.; Calore, E.E.; Manzione, C.R.; Puga, F.R.; Perez, N.M. Effects of long-term administration of Senna occidentalis seeds in the large bowel of rats. Pathol. -Res. Pract. 2003, 199, 733–737. [Google Scholar] [CrossRef]
  170. Oladeji, O.S.; Adelowo, F.E.; Oluyori, A.P.; Bankole, D.T. Ethnobotanical Description and Biological Activities of Senna alata. Evid. Based Complement. Altern. Med. 2020, 2020, 2580259. [Google Scholar] [CrossRef] [Green Version]
  171. Hennebelle, T.; Weniger, B.; Joseph, H.; Sahpaz, S.; Bailleul, F. Senna alata. Fitoterapia 2009, 80, 385–393. [Google Scholar] [CrossRef]
  172. Ali, M.Z.; Mehmood, M.H.; Saleem, M.; Gilani, A.H. The use of Euphorbia hirta L. (Euphorbiaceae) in diarrhea and constipation involves calcium antagonism and cholinergic mechanisms. BMC. Complement. Med. 2020, 20, 14. [Google Scholar] [CrossRef]
  173. Adedapo, A.A.; Abatan, M.O.; Olorunsogo, O.O. Toxic effects of some plants in the genus Euphorbia on haematological and biochemical parameters of rats. Vet. Arh. 2004, 74, 53–62. [Google Scholar]
  174. Rujjanawate, C.; Kanjanapothi, D.; Amornlerdpison, D.; Pojanagaroon, S. Anti-gastric ulcer effect of Kaempferia parviflora. J. Ethnopharmacol. 2005, 102, 120–122. [Google Scholar] [CrossRef]
  175. Sudwan, P.; Saenphet, K.; Saenphet, S.; Suwansirikul, S. Effect of Kaempferia parviflora Wall. ex. Baker on sexual activity of male rats and its toxicity. Southeast. Asian. J. Trop. Med. Public. Health. 2006, 37 Suppl. 3, 210–215. [Google Scholar]
  176. Syiem, D.; Khup, P. Evaluation of Flemingia macrophylla L., a traditionally used plant of the north eastern region of India for hypoglycemic and anti-hyperglycemic effect on mice. Pharmacologyonline 2007, 2, 355–366. [Google Scholar]
  177. Palle, S.; Kanakalatha, A.; Kavitha, C.N. Gastroprotective and Antiulcer Effects of Celastrus paniculatus Seed Oil Against Several Gastric Ulcer Models in Rats. J. Diet. Suppl. 2018, 15, 373–385. [Google Scholar] [CrossRef]
  178. Borrelli, F.; Borbone, N.; Capasso, R.; Montesano, D.; De Marino, S.; Aviello, G.; Aprea, G.; Masone, S.; Izzo, A.A. Potent relaxant effect of a Celastrus paniculatus extract in the rat and human ileum. J. Ethnopharmacol. 2009, 122, 434–438. [Google Scholar] [CrossRef]
  179. Nalini, K.; Karanth, K.; Rao, A.; Aroor, A. Effects of Celastrus paniculatus on passive avoidance performance and biogenic amine turnover in albino rats. J. Ethnopharmacol. 1995, 47, 101–108. [Google Scholar] [CrossRef]
  180. FDA. Complementary and Alternative Medicine Products and their Regulation by the Food and Drug Administration. Available online: https://www.fda.gov/regulatory-information (accessed on 5 December 2022).
  181. EMA. The Committee on Herbal Medicinal Products (HMPC). Available online: https://www.ema.europa.eu/en/committees/committee-herbal-medicinal-products-hmpc (accessed on 10 December 2022).
  182. Gajic, I.; Kabic, J.; Kekic, D.; Jovicevic, M.; Milenkovic, M.; Mitic Culafic, D.; Trudic, A.; Ranin, L.; Opavski, N. Antimicrobial Susceptibility Testing: A Comprehensive Review of Currently Used Methods. Antibiotics 2022, 11, 427. [Google Scholar] [CrossRef]
  183. Netopilova, M.; Houdkova, M.; Rondevaldova, J.; Kmet, V.; Kokoska, L. Evaluation of in vitro growth-inhibitory effect of carvacrol and thymol combination against Staphylococcus aureus in liquid and vapour phase using new broth volatilization chequerboard method. Fitoterapia 2018, 129, 185–190. [Google Scholar] [CrossRef]
  184. Abreu, A.C.; McBain, A.J.; Simões, M. Plants as sources of new antimicrobials and resistance-modifying agents. Nat. Prod. Rep. 2012, 29, 1007–1021. [Google Scholar] [CrossRef]
  185. Hadacek, F. Secondary Metabolites as Plant Traits: Current Assessment and Future Perspectives. Crit. Rev. Plant. Sci. 2002, 21, 273–322. [Google Scholar] [CrossRef]
  186. Atanasov, A.G.; Waltenberger, B.; Pferschy-Wenzig, E.-M.; Linder, T.; Wawrosch, C.; Uhrin, P.; Temml, V.; Wang, L.; Schwaiger, S.; Heiss, E.H. Discovery and resupply of pharmacologically active plant-derived natural products: A review. Biotechnol. Adv. 2015, 33, 1582–1614. [Google Scholar]
Figure 1. Chemical structures of P-Menthane type monoterpenoids.
Figure 1. Chemical structures of P-Menthane type monoterpenoids.
Pathogens 12 00333 g001
Figure 2. Chemical structure of Ternanthranin (N-methyl derivative of anthranilic acid), an alkaloid constituent of Mexican orange blossom, has anti-inflammatory and antibacterial activities [20].
Figure 2. Chemical structure of Ternanthranin (N-methyl derivative of anthranilic acid), an alkaloid constituent of Mexican orange blossom, has anti-inflammatory and antibacterial activities [20].
Pathogens 12 00333 g002
Table 1. Mechanism of action of plant-derived substances (PDSs) against a broad range of GI microbes.
Table 1. Mechanism of action of plant-derived substances (PDSs) against a broad range of GI microbes.
PhytochemicalsChemical Structure PlantMode of ActionMicroorganism(s)
ProanthocyanidinPathogens 12 00333 i001Vaccinium macrocarpon L.Modifies biofilm formationEnterococcus faecalis, E. coli[25,26]
ZingeronePathogens 12 00333 i002Zingiber officinale Rosc.Reduce heat-labile enterotoxin (LT)-induced diarrhoea in ETEC through blocking the binding to GM1 ganglioside receptorsETEC[27]
EpigallocatechinPathogens 12 00333 i003Camellia sinensis L.Inhibit extracellular release of Vero toxin from enterohemorrhagic Escherichia coli O157:H7EHEC[28]
QuercetinPathogens 12 00333 i004Allium cepa L.Inhibition of ATPase activity,
elevates extracellular phosphatase and galactosidase.
E. coli[29,30]
BerberinePathogens 12 00333 i005Berberis vulgaris L.Targeting proteins responsible for upholding the structure of cells and for cell divisionE. coli, Salmonella spp., V. cholerae[31,32]
EugenolPathogens 12 00333 i006Syzygium aromaticum L. Inhibition of toxin production EHEC[33]
CinnamaldehydePathogens 12 00333 i007Cinnamomum verum J. PreslInhibition of toxin genes and host receptor expression, reducing toxin-mediated pathologyEHEC, C. difficile[33,34]
Baicalin (5,6,7-trihydroxyflavone)Pathogens 12 00333 i008Scutellaria baicalensis Georgi.Inhibition of toxin productionH. pylori[35,36,37,38]
Thymol Pathogens 12 00333 i009Thymus vulgaris L.Disturbance of the cell membrane and cytoplasm C. difficile[39]
GeraniolPathogens 12 00333 i010Monarda fistulosa L.Disturbance of the cell membrane and disruption of the cytoplasm. C. difficile[39]
PyrrolizidinePathogens 12 00333 i011Alkanna tinctoria L.Disturbance of the cell membrane and cytoplasmE. coli[40]
MartinePathogens 12 00333 i012Sophora flavescens Ait.Inhibiting the synthesis
of proteins
E. coli[41]
IsothiocyanatesPathogens 12 00333 i013Brassica oleracea var. botrytisActing on cell membranes and leakage of cellular
metabolites
Pathogenic E. coli strains[42]
AndrographolidePathogens 12 00333 i014Andrographis paniculata (Burm.f.) NeesAnti-secretory activity against enterotoxins (Heat Labile (LT) and Heat Stable (ST) forms)Pathogenic E. coli[43]
α-bisabololPathogens 12 00333 i015Matricaria chamomilla L.Inhibition of efflux pump E. coli[44]
EAEC Enteroaggregative Escherichia coli, EHEC Enterohemorrhagic Escherichia coli.
Table 3. Toxicological effects of selected plant species used for the treatment of different GI disorders.
Table 3. Toxicological effects of selected plant species used for the treatment of different GI disorders.
Plant Species Common Name Family GI Disorder(s)Toxicological Effects
Psidium guajava L.GuavaMyrtaceaeAcute diarrhoea [96,156]Minor liver inflammation in rats, LD50 = 1000 mg/kg [102]
Musa × paradisiaca L.PlantainMusaceaeInfectious diarrhoea [157]Significant changes in white blood cells, eosinophils, platelets, neutrophils, and monocytes count [158].
Leea indica (Burm.f.) Merr.Bandicoot berryVitaceaeDiarrhoea, dysentery [159]Liver toxicity in rats [160]
Acorus calamus L.Sweet flagAcoraceaeGI infections, diarrhoea, dysentery [161,162]Acute liver, spleen, and kidney toxicity and genotoxic effects in rats, LD50 = 221 g/kg [161]
Cassytha filiformis L.Love-vineLauraceaeDiarrhoeagenic bacterial infections [163]Acute haematological and biochemical toxicity (significant increase in alanine aminotransferase (ALT), aspartate aminotransferase (AST), and total and direct bilirubin in rats, LD50 = 625.8 g/kg [164])
Zingiber officinale RoscoeGingerZingiberaceaeGastric ulceration, flatulence, diarrhoea [27,165]Embryo toxic to pregnant rats [166]
Thunbergia laurifolia Lindl.Laurel clock vineAcanthaceaeGastric ulcer, diarrhoea [156]Decrease red blood cells in male rat [167]
Senna occidentalis (L.) LinkCoffee sennaFabaceaeConstipation, GI infections [168]Intestinal disturbance in long term use [169]
Senna alata (L.) Roxb.Candle BushFabaceaeConstipation, abdominal pain [168,170]Decrease haemoglobin and erythrocyte (RBC) count values in rats [171]
Euphorbia hartal L.Hairy SpurgeEuphorbiaceaeDiarrhoea, dysentery, constipation, intestinal parasites [172]Leucocytosis, dullness, anorexia, stairy haircoat and 20% mortality in rats [173]
Euphorbia heterophylla L.MilkweedEuphorbiaceaeIntestinal bacterial infections, diarrhoea [172,173]Leucopaenia in rats [173]
Kaempferia parviflora L.Thai ginsengZingiberaceaeFlatulence, gastric ulcer [156,174]Hepatotoxic to rats [175]
Flemingia macrophylla (Willd.) Kuntze ex Merr.Apa apaFabaceaeFlatulence, indigestive [156]Severe hypoglycaemia followed by death within 24 h after administration to normoglycemic mice [176]
Celastrus paniculatus Willd.Black oil plantCelastraceaeDiarrhoea, gastric ulcer, bowel spasms [177,178]Hyperactivity and loss of behavioural responsiveness (loss of righting reflex in rat) [179]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Qassadi, F.I.; Zhu, Z.; Monaghan, T.M. Plant-Derived Products with Therapeutic Potential against Gastrointestinal Bacteria. Pathogens 2023, 12, 333. https://doi.org/10.3390/pathogens12020333

AMA Style

Qassadi FI, Zhu Z, Monaghan TM. Plant-Derived Products with Therapeutic Potential against Gastrointestinal Bacteria. Pathogens. 2023; 12(2):333. https://doi.org/10.3390/pathogens12020333

Chicago/Turabian Style

Qassadi, Fatimah I., Zheying Zhu, and Tanya M. Monaghan. 2023. "Plant-Derived Products with Therapeutic Potential against Gastrointestinal Bacteria" Pathogens 12, no. 2: 333. https://doi.org/10.3390/pathogens12020333

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop