Next Article in Journal
A Systematic Review on Microbial Profiling Techniques in Goat Milk: Implications for Probiotics and Shelf-Life
Next Article in Special Issue
Extracellular Vesicles Released by Bovine Alphaherpesvirus 1-Infected A549 Cells May Limit Subsequent Infections of the Progeny Virus
Previous Article in Journal
Human Papillomavirus and Other Relevant Issues in Cervical Cancer Pathogenesis
Previous Article in Special Issue
Effects of Sodium Alginate Infusion on Intramammary Immunity Against Subclinical Mastitis in Dairy Cows
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Plant Metabolites as Potential Agents That Potentiate or Block Resistance Mechanisms Involving β-Lactamases and Efflux Pumps

by
Muhammad Jawad Zai
1,2,
Ian Edwin Cock
1,2,* and
Matthew James Cheesman
3,*
1
Centre for Planetary Health and Food Security, Griffith University, Brisbane, QLD 4111, Australia
2
School of Environment and Science, Griffith University, Brisbane, QLD 4111, Australia
3
School of Pharmacy and Medical Sciences, Griffith University, Southport, QLD 4222, Australia
*
Authors to whom correspondence should be addressed.
Int. J. Mol. Sci. 2025, 26(12), 5550; https://doi.org/10.3390/ijms26125550
Submission received: 15 May 2025 / Revised: 4 June 2025 / Accepted: 9 June 2025 / Published: 10 June 2025
(This article belongs to the Special Issue Microbial Infections and Novel Biological Molecules for Treatment)

Abstract

The dramatic increase in antimicrobial resistance (AMR) in recent decades has created an urgent need to develop new antimicrobial agents and compounds that can modify and/or block bacterial resistance mechanisms. An understanding of these resistance mechanisms and how to overcome them would substantially assist in the development of new antibiotic chemotherapies. Bacteria may develop AMR through multiple differing mechanisms, including modification of the antibiotic target site, limitation of antibiotic uptake, active efflux of the antibiotic, and via direct modification and inactivation of the antibiotic. Of these, efflux pumps and the production of β-lactamases are the most common resistance mechanisms that render antibiotics inactive. The development of resistance-modifying agents (particularly those targeting efflux pumps and β-lactamase enzymes) is an important consideration to counteract the spread of AMR. This strategy may repurpose existing antibiotics by blocking bacterial resistance mechanisms, thereby increasing the efficacy of the antibiotic compounds. This review focuses on known phytochemicals that possess efflux pump inhibitory and/or β-lactamase inhibitory activities. The interaction of phytochemicals possessing efflux pumps and/or β-lactamase inhibitory activities in combination with clinical antibiotics is also discussed. Additionally, the challenges associated with further development of these phytochemicals as potentiating agents is discussed to highlight their therapeutic potential, and to guide future research.

1. Introduction

Bacterial resistance to clinical antibiotics (AMR) poses a global challenge, contributing significantly to high mortality and morbidity [1]. The increased incidence of multidrug resistance (MDR) in both Gram-negative and Gram-positive bacteria has led to infections that are challenging (or impossible) to treat with current antibacterial therapies [2]. The rise in AMR has been driven by multiple factors, including the increased use of antimicrobials in both humans and animals, and inappropriate prescribing of these therapies (e.g., to treat viral infections). Excessive and improper antibiotic use in humans promotes the emergence of resistant microorganisms [3]. The Centres for Disease Control and Prevention (CDC) reported that over 2 million people in the United States (U.S.) fall ill each year due to antimicrobial-resistant infections, leading to more than 23,000 deaths annually [4]. The cost associated with these resistant infections is estimated to be from USD7000 to over USD29,000 per patient [5]. Indeed, methicillin-resistant Staphylococcus aureus (MRSA) infections cost over USD18,000 each to treat effectively in the U.S., nearly EUR9000 in Germany, and more than 100,000 Swiss francs per case in Switzerland [6,7,8].
Bacterial antibiotic resistance has arisen from multiple origins, with various adaptive mechanisms identified that contribute to resistant behaviour in bacterial populations [9]. Efflux pumps are perhaps the quickest and most effective resistance mechanism bacteria can develop to neutralise antibiotics. Indeed, MDR efflux pumps are a major factor in the reduced susceptibility to antimicrobials seen in multiple clinical pathogenic bacteria isolates [10]. To date, six families of bacterial drug efflux pumps have been identified that contribute to efflux pathways [11]. The ATP binding cassette (ABC) family of efflux pumps directly uses ATP as an energy source to power antimicrobial movement out of the cell [12]. The other five groups, which include the multidrug and toxin extrusion family, major facilitator family, small multidrug resistance family, proteobacterial antimicrobial compound efflux family, and the resistance nodulation cell division superfamily function as secondary active transporters, with efflux driven by electrochemical energy from transmembrane ion gradients [13,14].
Antibiotics of the β-lactam class are among the most prescribed antibiotics for treating microbial infections. They are valued for their safety, reliable (and often broad-spectrum) bactericidal properties, and their clinical effectiveness. However, β-lactam antibiotic effectiveness is increasingly compromised by the global spread of genes encoding β-lactamase enzymes with extensive hydrolytic abilities, particularly in MDR Gram-negative pathogens. β-Lactamase enzymes are categorised into four classes based on their sequence identities [15]. Whilst only a few β-lactamase enzymes had been documented by the early 1970s, their numbers have since grown rapidly, with new enzymes capable of hydrolysing carbapenems now increasingly identified in clinical isolates [16]. A notable example is the class A β-lactamase KPC-2 which, within a few years, has become one of the most concerning β-lactamases spreading globally [17]. Class A β-lactamases may be plasmid-encoded (GES, KPC, FRI-1), chromosomally encoded (NmcA, SME, BIC-1, SFC-1, FPH-1, PenA, SHV-38), or both (IMI). Plasmid encoded enzymes are frequently linked to mobile genetic elements that facilitate their transfer. These enzymes exhibit shared structural characteristics and operate through a common mechanism of action. They variably hydrolyse cephalosporins, penicillins, carbapenems, monobactams, and are inhibited by tazobactam and clavulanate [17].
In recent years, there has been growing interest in natural products for the development of new antibiotic chemotherapies, as numerous studies have shown that natural product compounds exhibit significant antimicrobial effects, which are often distinct from those of existing clinical antibiotics [18,19]. Notably, phytochemicals that exert antimicrobial activity via mechanism(s) that differ from those of presently available antibiotics may be of substantial clinical value for treating antibiotic-resistant infections [20,21]. Many phytochemicals have demonstrated effective antibiotic resistance reversal activity, primarily by inhibiting drug efflux pumps or modifying enzymes [22].

2. Medicinal Plant Secondary Metabolites as Antimicrobial Agents

Plants derive their antimicrobial properties from their secondary metabolites. Plant secondary metabolites constitute over 8000 known phenolic compounds, of which there are more than 12,000 known alkaloids and 25,000 terpenoids, many of which possess strong antibacterial activity [23]. Of these, phenolic compounds are the most frequently reported antibacterial agents [24]. Phenolic compounds can contain either multiple phenol units, in which case they are classified as polyphenolic, or they may contain a single substituted phenolic ring. Phenols can be further classified into phenolic acids, coumarins, stilbenes, flavonoids, lignans, and tannins (Figure 1). In particular, terpenes and terpenoids are widely recognised for their antimicrobial activities [25]. Another noteworthy class of plant-derived antimicrobials are alkaloids. These compounds form a diverse and structurally varied group, serving as foundational scaffolds for critical antibacterial drugs including quinolones and metronidazole [26]. The antibacterial activity of selected plant secondary metabolites is summarised in Table 1.

3. Bacterial Efflux Pump Mechanisms

Antibiotic efflux is one of the most prevalent resistance mechanisms among various pathogenic bacteria [46]. Efflux pumps play a role in maintaining the internal environment of bacteria by expelling quorum-sensing molecules, toxic substances, bacterial virulence factors, and biofilm-forming molecules [47]. Efflux pumps can be selective for a single substrate, or capable of transporting a variety of structurally diverse compounds, including multiple classes of antibiotics [48]. Efflux pumps are classified as primary or secondary pumps, based on the energy source they use to expel substrates. Primary efflux pumps use the energy derived from ATP hydrolysis to transport substrates across the membrane. In contrast, secondary efflux pumps rely on energy from electrochemical gradients, such as the proton motive force, created by protons or ions. To date, six main families of bacterial efflux pumps have been identified, and include the major facilitator superfamily, the ATP binding cassette superfamily, the resistance nodulation cell division family, the multidrug and toxic compound extrusion family, the proteobacterial antimicrobial compound efflux and the small multidrug resistance family (Figure 2) [12]. Efflux pumps are crucial for bacterial survival in various stress environments, making them a promising target for developing new inhibitors to restore the effectiveness of existing antibiotics.

4. Mechanism of β-Lactamases

The extensive use of β-lactam antibiotics has driven the emergence and spread of resistance. Enzyme-mediated β-lactam resistance is due to the action of the β-lactamase enzymes, which are produced by both Gram-negative and Gram-positive bacteria. The enzyme hydrolyses the β-lactam amide bond (Figure 3) [49]. β-Lactamases are categorised into the four classes A, B, C, and D and this is based on primary sequence homology and variations in their hydrolytic mechanisms [50]. Class A, C, and D β-lactamases are serine-based enzymes that hydrolyse the β-lactam ring through a serine-bound acyl intermediate in the active site. In contrast, class B β-lactamases, known as metallo-β-lactamases, contain one or two zinc ions in their active site, which are essential for their enzymatic function [51]. Over 400 distinct types of β-lactamases have been identified. Among the Gram-negative bacteria, including Enterobacteriaceae spp., Haemophilus influenza, P. aeruginosa, and Neisseria gonorrhoeae, the most common plasmid-mediated class A β-lactamases are TEM and a closely related enzyme, TEM-2. These enzymes are capable of hydrolysing multiple penicillins, and some cephalosporins [52]. A related, but less prevalent class A β-lactamases called SHV have been identified primarily in Enterobacteriaceae spp. and in P. aeruginosa. These enzymes can hydrolyse aztreonam and third generation cephalosporins, including ceftazidime and cefotaxime [52].

5. Phytochemicals as Efflux Pump and β-Lactamase Inhibitors

Herbal remedies and phytochemical-based therapies have gained substantial recent attention as targets for the discovery of efflux pumps and β-lactamase inhibitors due to their structural diversity and multiple modes of action. Berberine (Figure 4a) was first discovered by Herberger and Buchner in 1830 and is the most extensively researched protoberberine alkaloid found in nature, with a long history of use [53]. The primary plants containing berberine include Berberis vulgaris L., and Scutellaria baicalensis Georgi., which have been traditionally used as folk medicines in India, China, Iran, and various other countries [54]. Berberine’s pharmacological activity has been associated with nearly all types of bodily disorders, including blood disease and cancer [55,56], cardiovascular diseases [57], the central nervous system [58] and immune diseases [59]. Research on medicinal plant extracts has revealed several phytochemicals that exert antimicrobial activity by inhibiting efflux pumps and β-lactamases in Gram-positive and Gram-negative bacteria (Table 2). By blocking these resistance mechanisms, such phytochemicals enable antibiotics to reach sufficient concentrations within bacterial cells to achieve bactericidal effects.

6. Plant Metabolite Structure Activity Relationship

Plant metabolites are known for their wide range of biological activities, including antimicrobial effects and their extensive structural diversity makes them promising candidates for drug discovery [90]. The most common classification of phytochemicals is based on their chemical structures, including groups such as alkaloids, limonoids, phenolics, saponins, secoiridoids, terpenoids and others. Alkaloids are typically defined as nitrogen-containing basic metabolites with heterocyclic structures, although this definition does not always clearly distinguish them from other nitrogenous compounds. They are classified in various ways, often based on their biogenic origins or the characteristics of their carbon skeleton. Among phytochemicals, alkaloids exhibit particularly high structural diversity. They are generally categorised according to their core carbon skeletons [91]. Limonoids are distinctive secondary metabolites characterised by a tetranortriterpenoid backbone that includes a furan ring. They are commonly isolated from Citrus and Meliaceae plants and have been shown to exhibit antibacterial activities [92]. Several simple phenols and phenolic acids also exhibit antibacterial activities against a broad range of pathogens. Structure-activity relationship studies suggest that phenols with varying alkyl chain lengths and hydroxyl groups may play a crucial role in determining antimicrobial activity [93]. Studies have reported that the antimicrobial activity of phenolic acid derivatives increases with the length of the alkyl chain [94]. The antimicrobial toxicity of phenolic compounds is also influenced by the presence of hydroxyl groups and the oxidation state of the phenol moiety. An increase in the length of hydrophobic alkyl chains can disrupt the fluidity of microbial cell membranes. Phenolic acids may integrate into the membrane structure, with their polar hydroxyl groups forming hydrogen bonds in the aqueous phase, while their nonpolar alkyl chains align with the lipid bilayer through dispersion forces [93]. Therefore, when the hydrophilic interactions outweigh the hydrophobic ones, the antimicrobial activity tends to diminish. Additionally, the number and position of substituents on the benzene ring, along with the length of the saturated side chain, influence the bactericidal effects of phenolic acids. However, these effects vary between Gram-positive and Gram-negative bacteria [95].

7. Synergistic Interactions Between Efflux Pump and β-Lactamase Inhibitor Phytochemicals and Conventional Antibiotics

Synergistic interactions refer to a phenomenon where the combined therapeutic potential of two or more compounds is greater than the sum of their individual effects. Combinations of phytochemicals possessing efflux pump and/or β-lactamase inhibitor activity with conventional antibiotics (or antibiotic natural compounds) can reduce the minimum inhibitory concentration (MIC) of the antibiotic components and thus sensitise MDR bacteria to these antibiotics [96]. In synergistic interactions, one molecule can enhance or support the primary action of another compound, resulting in improved therapeutic efficacy. Such synergistic interaction occurs when antibiotics are paired with compounds capable of partially or entirely inhibiting bacterial resistance mechanisms. These combinations enhance the effectiveness of antibiotics by reducing bacterial defences, allowing for a more robust therapeutic response. For example, combining clavulanic acid with penicillin (or other β-lactam antibiotics) effectively blocks penicillinase resistance [97]. Table 3 summarises known combinations of phytochemicals and antibiotics that minimise antimicrobial resistance by targeting efflux pumps and β-lactamase enzymes.

8. Plant Metabolites Efflux Pump Inhibitor Mechanisms

Efflux pump inhibitors aim to restore bacterial sensitivity to antibiotics by preventing the bacteria from expelling the drug through their intramembrane efflux pump systems [119]. Efflux pump inhibitors work through various methods, including blocking of ATP synthesis to inhibit antibiotic efflux, repressing the gene responsible for encoding efflux pumps and dissipation of the proton gradient essential for the pump activity. Other mechanisms include disrupting the assembly of efflux systems and obstructing the membrane proteins involved in efflux (Figure 5).
Different studies have examined baicalin for its ability to inhibit the MsrA efflux pump in ARSS (azithromycin-resistant Staphylococcus saprophyticus), a strain that expresses the MsrA efflux gene and is resistant to azithromycin [120]. MsrA is an ATP-dependent efflux pump, suggesting that baicalin’s inhibitory effects on the ARSS efflux pump are more closely associated with the ATP-dependent processes in bacteria [121]. Baicalin may disrupt ATP production, potentially inhibiting the function of the MsrA efflux pump and enhancing its efflux pump inhibitory activity. Studies have also shown capsaicin (Figure 4f) is an effective efflux pump inhibitor activity against S. aureus strains that express NorA efflux pump [70]. Capsaicin is a known P-glycoprotein (P-gp) inhibitor, an active drug transporter belonging to the ATP-binding cassette transporter family. P-gp is involved in the efflux of chemotherapeutic agents from the cells and is highly expressed in the apical membrane of various pharmacologically significant epithelial barriers, such as the hepatocytes, intestinal epithelium and renal tubular cells indicating its critical role in drug absorption, distribution and elimination [122,123].
Cumin, and its key active component cuminaldehyde, inhibit the activity of the S. aureus LmrS multidrug efflux pump [76]. Host cells lacking the LmrS efflux pump exhibited a reduced response to treatment with cumin extract. Studies have also noted significantly higher ethidium bromide (a common substrate for efflux pump assays) accumulation in E. coli KAM32 host cells expressing the LmrS multidrug efflux pump when treated with cumin extract compared to KAM32 host cells lacking the LmrS efflux pump [76]. These efflux and accumulation studies suggest that cumin extract specifically targets the LmrS multidrug efflux pump. Cumin and cuminaldehyde also suppress the growth of host cells lacking LmrS, albeit at relatively higher concentrations [76]. This observation suggests that these compounds may disrupt the proton motive force generated during respiration and metabolism. Cumin appears to exert a multifaceted effect, inhibiting the LmrS efflux pump at low concentrations and compromising the integrity of the cell membrane at higher concentrations.
The plant alkaloid reserpine directly binds specific RND and MFS efflux pumps in Gram-negative bacteria through competitive inhibition [119]. In Gram-positive bacteria, reserpine interacts directly with specific amino acid residues within the efflux pump transporter proteins, such as the Bmr efflux pump in B. subtilis [119]. This interaction affects the pump ability to expel antimicrobial agents, thereby reducing resistance and potentially making the bacteria more susceptible to treatment with the antibiotic. Eugenol, an active component of clove oil and trans-cinnamaldehyde extracted from cinnamon bark inhibit efflux pumps in A. baumannii at a genetic level by downregulating gene adeA and adeB in the RND efflux family [124].
Catechin gallates, including epicatechin gallate and epigallocatechin gallate (Figure 4i), are plant-derived flavonoid-tannin hybrids that act as mild inhibitors of the NorA efflux pump, with epicatechin gallate showing slightly greater potency [119]. It has been suggested that these molecules interact with two distinct binding sites on the NorA efflux transporter, each with varying affinities. At low concentrations, catechins bind to high-affinity sites, increasing efflux of the NorA substrate [119]. Epigallocatechin gallate also acts on TetK efflux pumps in Staphylococci spp. and Campylobacter spp. to increase their susceptibility to tetracycline, erythromycin and ciprofloxacin, although the exact mechanism was not determined [119]. In K. pneumoniae, epigallocatechin has been shown to restore susceptibility to tetracycline due to a change in the capsule structure of K. pneumoniae and the inhibition of efflux pumps from the RND family (AcrAB) [125]. Epigallocatechin gallate also inhibits the growth of B. subtilis by targeting cell surface proteins including glucose phosphotransferase system transporter protein, oligopeptide ABC transporter binding lipoprotein, penicillin-binding protein 5 and phosphate ABC transporter substrate-binding protein [126].
Resveratrol, a phytochemical sourced from the seeds and skins of red grape, is another efflux pump inhibitor [127]. Resveratrol has been shown to enhance the susceptibility of clinical A. baumannii isolates resistant to chlorhexidine [127]. Whilst resveratrol itself does not inhibit the growth of A. baumannii, it targets the expression of AdeB protein, a component of AdeABC efflux pump system. The expression of AdeB is significantly reduced when resveratrol is combined with chlorhexidine, thereby substantially increasing antibiotic sensitivity [127].

9. Plant Metabolites β-Lactamase Inhibitor Mechanism

β-Lactam antibiotics are among the most used antimicrobial agents in hospital environments due to their broad spectrum of activity, high efficacy and safety [128]. The four primary classes of β-lactam antibiotics include cephalosporins, penicillins, monobactams and carbapenems, all of which feature a four-membered cyclic amide (azetidinone), commonly known as β-lactam rings. All β-lactam antibiotics share a common mode of bacterial killing, via the inhibition of transpeptidase enzymes that are essential for the final step in bacterial cell wall synthesis. Alexander Fleming observed that the β-lactam antibiotic penicillin failed to inhibit the growth of certain bacteria within the colityphoid group [129]. Further studies revealed an enzyme in E. coli (initially termed as penicillinase) inactivated penicillin by breaking its β-lactam ring, producing penicilloic acid, thereby rendering it ineffective [130]. One strategy to counteract bacterial resistance to β-lactam antibiotics involves the targeting and inhibition of the β-lactamase enzyme (Figure 6). Clavulanic acid, a naturally occurring compound derived from Streptomyces clavuligerus, has been identified as a powerful inhibitor of staphylococcal and TEM-1 penicillinase and is perhaps the most widely known example of a β-lactamase inhibitor [131]. Mechanistically, clavulanic acid is a high-efficacy irreversible inactivator of TEM-1, creating an irreversibly inactivated enzyme complex [132].
The phytochemical 2-methoxy chrysophanol (Figure 4c) derived from Clutia myricoides Jaub. & Spach possesses an anti-ESBL activity against K. pneumoniae. To investigate the mechanism of action of 2-methoxy chrysophanol, molecular docking was conducted with CTX-M-27 ESBL [61]. 2-Methoxy chrysophanol formed two hydrogen bonds with the side chains of Ser70 and Thr235 through its two phenolic hydroxyl groups. Although it did not form a hydrogen bond with the catalytic residue Ser237, its carbonyl group compensated by forming a hydrogen bond with the nitrogen atom of the backbone amide of this residue. Notably, 2-methoxy chrysophanol also established two critical hydrogen bonds via its methoxy group with the side chains of Asn132 and Asn104. These interactions are particularly significant as they contribute to the compound’s activity against ESBL enzymes [61].
Studies have reported the β-lactamase inhibitory effects of 1,4-naphthalenedione (Figure 4b), a compound isolated from the plant Holoptelea integrifolia (Roxb.) Planch [60]. Molecular docking studies showed that 1,4-naphthalenedione binds to the active site of β-lactamase and one of its carbonyl oxygens hydrogen bonds with residues Lys 73 and Ser 70. The ligand is surrounded by several hydrophobic residues, suggesting that van der Waals interactions may serve as the primary stabilising factor of the complex [60].
Epigallocatechin, a phytochemical present in green tea, also is an effective inhibitor of β-lactamase enzymes. It binds directly to peptidoglycan in the bacterial cell membrane and inhibits penicillinase activity in a dose-dependent manner, achieving 50% inhibition at a concentration of 10 µg/mL [133]. The synergistic interaction between quercetin and amoxicillin has also been shown to be very effective in inhibiting peptidoglycan synthesis in the bacterial cell membrane, enhancing cell membrane permeability, suppressing β-lactamase activity, reducing the presence of fatty acids within bacterial cells and increasing protein amide I (alpha-helix and beta-sheet) and amide II levels [134]. These findings suggest that quercetin, both individually and in combination with amoxicillin, may alter the fatty acid composition of various membrane amphiphiles, resulting in cytoplasmic membrane damage and increased membrane permeability. Additionally, this combination may interact with β-lactamase, transpeptidase and other proteins, potentially forming complexes that lead to the stacking and accumulation of proteins within the bacterial cells.

10. Challenges and Future Perspectives

Some phytochemicals offer natural antibacterial properties and are generally safer than synthetic alternatives. Additionally, other compounds have antibiotic-potentiating activity by blocking bacterial resistance pathways. Herbal material containing bioactive phytochemicals is often readily available and locally accessible, easy to administer, cost-effective and provides significant therapeutic benefits, making treatments more affordable and practical for many communities [135]. The current proportion of antibacterial agents derived from medicinal plants does not represent their full potential for future antimicrobial therapies [136]. Isolating individual phytochemicals with targeted antibacterial activity can be a lengthy process and may require the use of large quantities of plant material and extraction solvents that can be toxic or environmentally damaging. Synergistic interactions within complex mixtures of compounds pose specific challenges, as current technology is not yet fully equipped to analyse multiple compounds working together across various biological targets [137]. The concept of synergistic effects is not standardised due to differences between the studies, and there is no uniform use of the term across studies. The interactions between phytochemicals can be favourable, such as synergy, or harmful, antagonistic. Synergistic interactions among medicinal plant extracts, as well as between their compounds and antibiotics, should be further investigated to uncover the underlying mechanisms driving their antimicrobial activity. This research could help identify multiple pathways that can be targeted for enhanced therapeutic outcomes. Phytochemicals are a largely untapped reservoir of bioactive compounds, with only a limited number of them having been explored for the discovery of new compounds that can modify AMR and serve as an effective therapeutic tool. Whilst all of the combinational therapies summarised herein highlight the potential to overcome bacterial resistance mechanisms, it is noteworthy that the bacteria may develop resistance against the potentiating compound, and further work is required to address this.
Future in vitro studies should assess the clinical relevance of these compounds and validate their in vivo efficacy through established correlations with in vitro efficacy results. Further studies should identify efflux pump and β-lactamase inhibitors by performing further studies including combinational, metabolomics and in silico methods. The structures of these compounds should also be modified to optimise their pharmacodynamics and pharmacokinetics, alongside conducting structure-activity analysis to enhance their safety and efficacy. Studies should also focus on investigating synergistic interactions within phytochemicals and antibiotics, to elucidate mechanisms underlying their antibacterial activity. This could reveal multiple molecular pathways to target, broadening the therapeutic options available to counter AMR. The interactions between phytochemicals and antibiotics can be either beneficial such as synergistic, or unsafe, as in antagonistic. Thus, additional studies are essential, particularly in vivo and toxicity studies, for these products to be validated and recognised as antimicrobial agents

11. Conclusions

The increasing incidence of antibiotic-resistant infections poses a challenge to effective bacterial infection treatment and control. Developing new and effective treatments against antibiotic-resistant pathogens has become a global priority, as these alternatives are essential to address the growing challenge of AMR. Whilst there is a relatively good amount of data reporting synthetic inhibitors, plant-based compounds are relatively ignored. Phytochemicals have the potential as a promising alternative to conventional antibiotics for combating infections caused by antibiotic-resistant pathogens. There are multiple other mechanisms that render the antibiotics ineffective. In this review, we focus on only two resistance mechanisms: efflux pumps and β-lactamases. These are arguably the most important because β-lactams are widely used, and efflux pumps are a common resistant mechanism. Some phytochemicals can inhibit key antibiotic resistance mechanisms, including efflux pumps and β-lactamase activity, which are necessary for pathogen survival and resistance. Although there are numerous examples of phytochemicals effectively combating antibiotic-resistant infections, the success in translating these findings into commercial applications remains limited and requires substantial improvement. There is a critical need to accelerate research, clinical trials and regulatory approval processes to facilitate the application of phytochemicals in combating MDR infections.

Author Contributions

Conceptualization, M.J.Z. and I.E.C.; data curation, M.J.Z.; original draft preparation, M.J.Z.; writing—review and editing, supervision, M.J.C. and I.E.C. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

The authors are grateful to Griffith University, Australia for providing M.J.Z with a PhD scholarship to support this work.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Akova, M. Epidemiology of antimicrobial resistance in bloodstream infections. Virulence 2016, 7, 252–266. [Google Scholar] [CrossRef] [PubMed]
  2. Frieri, M.; Kumar, K.; Boutin, A. Antibiotic resistance. J. Infect. Public Health 2017, 10, 369–378. [Google Scholar] [CrossRef] [PubMed]
  3. Goossens, H. Antibiotic consumption and link to resistance. Clin. Microbiol. Infect. 2009, 15, 12–15. [Google Scholar] [CrossRef] [PubMed]
  4. Solomon, S.L.; Oliver, K.B. Antibiotic resistance threats in the United States: Stepping back from the brink. Am. Fam. Physician 2014, 89, 938–941. [Google Scholar]
  5. Maragakis, L.L.; Perencevich, E.N.; Cosgrove, S.E. Clinical and economic burden of antimicrobial resistance. Expert Rev. Anti-Infect. Ther. 2008, 6, 751–763. [Google Scholar] [CrossRef]
  6. Filice, G.A.; Nyman, J.A.; Lexau, C.; Lees, C.H.; Bockstedt, L.A.; Como-Sabetti, K.; Lesher, L.J.; Lynfield, R. Excess costs and utilization associated with methicillin resistance for patients with Staphylococcus aureus infection. Infect. Control. Hosp. Epidemiol. 2010, 31, 365–373. [Google Scholar] [CrossRef]
  7. Hübner, C.; Hübner, N.O.; Hopert, K.; Maletzki, S.; Flessa, S. Analysis of MRSA-attributed costs of hospitalized patients in Germany. Eur. J. Clin. Microbiol. 2014, 33, 1817–1822. [Google Scholar] [CrossRef]
  8. Macedo-Vinas, M.; De Angelis, G.; Rohner, P.; Safran, E.; Stewardson, A.; Fankhauser, C.; Schrenzel, J.; Pittet, D.; Harbarth, S. Burden of methicillin-resistant Staphylococcus aureus infections at a Swiss University hospital: Excess length of stay and costs. J. Hosp. Infect. 2013, 84, 132–137. [Google Scholar] [CrossRef]
  9. Alcalde-Rico, M.; Hernando-Amado, S.; Blanco, P.; Martinez, J.L. Multidrug efflux pumps at the crossroad between antibiotic resistance and bacterial virulence. Front. Microbiol. 2016, 7, 1483. [Google Scholar] [CrossRef]
  10. Grkovic, S.; Brown, M.H.; Skurray, R.A. Regulation of bacterial drug export systems. Microbiol. Mol. Biol. Rev. 2002, 66, 671–701. [Google Scholar] [CrossRef]
  11. Avakh, A.; Grant, G.D.; Cheesman, M.J.; Kalkundri, T.; Hall, S. The art of war with Pseudomonas aeruginosa: Targeting mex efflux pumps directly to strategically enhance antipseudomonal drug efficacy. Antibiotics 2023, 12, 1304. [Google Scholar] [CrossRef] [PubMed]
  12. Du, D.J.; Wang-Kan, X.; Neuberger, A.; van Veen, H.W.; Pos, K.M.; Piddock, L.J.V.; Luisi, B.F. Multidrug efflux pumps: Structure, function and regulation. Nat. Rev. Microbiol. 2018, 16, 577. [Google Scholar] [CrossRef]
  13. Hassan, K.A.; Jackson, S.M.; Penesyan, A.; Patching, S.G.; Tetu, S.G.; Eijkelkamp, B.A.; Brown, M.H.; Henderson, P.J.F.; Paulsen, I.T. Transcriptomic and biochemical analyses identify a family of chlorhexidine efflux proteins. Proc. Natl. Acad. Sci. USA 2013, 110, 20254–20259. [Google Scholar] [CrossRef]
  14. Hassan, K.A.; Liu, Q.; Henderson, P.J.F.; Paulsena, I.T. Homologs of the Acinetobacter baumannii AceI transporter represent a new family of bacterial multidrug efflux systems. mBio 2015, 6, e01982-14. [Google Scholar] [CrossRef] [PubMed]
  15. Bonomo, R.A. β-Lactamases: A focus on current challenges. Cold Spring Harb. Perspect. Med. 2017, 7, a025239. [Google Scholar] [CrossRef] [PubMed]
  16. Nordmann, P.; Naas, T.; Poirel, L. Global spread of carbapenemase-producing Enterobacteriaceae. Emerg. Infect. Dis. 2011, 17, 1791–1798. [Google Scholar] [CrossRef]
  17. Naas, T.; Dortet, L.; Iorga, B.I. Structural and functional aspects of Class A carbapenemases. Curr. Drug Targets 2016, 17, 1006–1028. [Google Scholar] [CrossRef]
  18. Savoia, D. Plant-derived antimicrobial compounds: Alternatives to antibiotics. Future Microbiol. 2012, 7, 979–990. [Google Scholar] [CrossRef]
  19. González-Lamothe, R.; Mitchell, G.; Gattuso, M.; Diarra, M.S.; Malouin, F.; Bouarab, K. Plant antimicrobial agents and their effects on plant and human pathogens. Int. J. Mol. Sci. 2009, 10, 3400–3419. [Google Scholar] [CrossRef]
  20. Keita, K.; Darkoh, C.; Okafor, F. Secondary plant metabolites as potent drug candidates against antimicrobial-resistant pathogens. SN Appl. Sci. 2022, 4, 209. [Google Scholar] [CrossRef]
  21. Li, J.M.; Feng, S.S.; Liu, X.; Jia, X.; Qiao, F.L.; Guo, J.L.; Deng, S.S. Effects of traditional Chinese medicine and its active ingredients on drug-resistant bacteria. Front. Pharmacol. 2022, 13, 837907. [Google Scholar] [CrossRef] [PubMed]
  22. Tiwari, P.; Khare, T.; Shriram, V.; Bae, H.; Kumar, V. Plant synthetic biology for producing potent phyto-antimicrobials to combat antimicrobial resistance. Biotechnol. Adv. 2021, 48, 107729. [Google Scholar] [CrossRef]
  23. Radulovic, N.S.; Blagojevic, P.D.; Stojanovic-Radic, Z.Z.; Stojanovic, N.M. Antimicrobial plant metabolites: Structural diversity and mechanism of action. Curr. Med. Chem. 2013, 20, 932–952. [Google Scholar] [PubMed]
  24. 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] [PubMed]
  25. Masyita, A.; Sari, R.M.; Astuti, A.D.; Yasir, B.; Rumata, N.R.; Bin Emran, T.; Nainu, F.; Simal-Gandara, J. Terpenes and terpenoids as main bioactive compounds of essential oils, their roles in human health and potential application as natural food preservatives. Food Chem. X 2022, 13, 100217. [Google Scholar] [CrossRef]
  26. Cushnie, T.P.; Cushnie, B.; Lamb, A.J. Alkaloids: An overview of their antibacterial, antibiotic-enhancing and antivirulence activities. Int. J. Antimicrob. Agents 2014, 44, 377–386. [Google Scholar] [CrossRef]
  27. Dib, M.E.; Allali, H.; Bendiabdellah, A.; Meliani, N.; Tabti, B. Antimicrobial activity phytochemical screening of Arbutus unedo L. J. Saudi Chem. Soc. 2013, 17, 381–385. [Google Scholar] [CrossRef]
  28. Tan, K.K.; Khoo, T.J.; Rajagopal, M.; Wiart, C. Antibacterial alkaloids from Artabotrys crassifolius Hook.f. & Thomson. Nat. Prod. Res. 2015, 29, 2346–2349. [Google Scholar]
  29. Sardessai, Y.; Desai, R.R.; Joshi, A.B.; Bhobe, M.P. Screening for antimicrobial activity of the stem bark of Bauhinia purpurea Linn. Glob. J. Pharmacol. 2013, 7, 288–293. [Google Scholar]
  30. Azimi, G.; Hakakian, A.; Ghanadian, M.; Joumaa, A.; Alamian, S. Bioassay-directed isolation of quaternary benzylisoquinolines from Berberis integerrima with bactericidal activity against Brucella abortus. Res. Pharm. Sci. 2018, 13, 149–158. [Google Scholar]
  31. Mandi, F.; Morgan, J.B.; Liu, W.L.; Agarwal, A.K.; Jekabsons, M.B.; Liu, Y.; Zhou, Y.D.; Nagle, D.G. Sampangine (a Copyrine Alkaloid) Exerts Biological Activities through Cellular Redox Cycling of Its Quinone and Semiquinone Intermediates. J. Nat. Prod. 2015, 78, 3018–3023. [Google Scholar]
  32. Farha, M.A.; El-Halfawy, O.M.; Gale, R.T.; MacNair, C.R.; Carfrae, L.A.; Zhang, X.; Jentsch, N.G.; Magolan, J.; Brown, E.D. Uncovering the hidden antibiotic potential of cannabis. ACS Infect. Dis. 2020, 6, 338–346. [Google Scholar] [CrossRef]
  33. Subhadradevi, V.; Asokkumar, K.; Umamaheswari, M.; Sivashanmugam, A.; Ushanandhini, J.; Jagannath, P. Antimicrobial activity of leaves and flowers of Cassia auriculata Linn. Bangladesh J. Sci. Ind. Res. 2011, 46, 513–518. [Google Scholar] [CrossRef]
  34. Oloyede, G.K.; Onocha, P.A.; Soyinka, J.; Oguntokun, O.; Thonda, E. Phytochemical screening, antimicrobial and antioxidant activities of four Nigerian medicinal plants. Ann. Biol. Res. 2010, 1, 114–120. [Google Scholar]
  35. Kumar, R.S.; Balasubramanian, P.; Govindaraj, P.; Krishnaveni, T. Preliminary studies on phytochemicals and antimicrobial activity of solvent extracts of Coriandrum sativum L. roots (Coriander). J. Pharmacogn. Phytochem. 2014, 2, 74–78. [Google Scholar]
  36. Shen, F.G.; Tang, X.D.; Wang, Y.; Yang, Z.Q.; Shi, X.C.; Wang, C.; Zhang, Q.L.; An, Y.A.; Cheng, W.; Jin, K.Q.; et al. Phenotype and expression profile analysis of Staphylococcus aureus biofilms and planktonic cells in response to licochalcone A. Appl. Microbiol. Biotechnol. 2015, 99, 359–373. [Google Scholar] [CrossRef]
  37. Pájaro-Gonzalez, Y.; Oliveros-Diaz, A.F.; Cabrera-Barraza, J.; Fernández-Daza, E.; Reyes, N.; Montes-Guevara, O.A.; Caro-Fuentes, D.; Franco-Ospina, L.; Quiñones-Fletcher, W.; Quave, C.L.; et al. Mammea B/BA isolated from the seeds of Mammea americana L. (Calophyllaceae) is a potent inhibitor of methicillin-resistant Staphylococcus aureus. Front. Pharmacol. 2022, 13, 826404. [Google Scholar] [CrossRef]
  38. Mada, S.; Garba, A.; Mohammed, H.a.A.; Muhammad, A.; Olagunju, A.; Muhammad, A. Antimicrobial activity and phytochemical screening of aqueous and ethanol extracts of Momordica charantia L. leaves. J. Med. Plants Res. 2013, 7, 579–586. [Google Scholar]
  39. Singh, A.R.; Bajaj, V.K.; Sekhawat, P.S.; Singh, K. Phytochemical estimation and antimicrobial activity of aqueous and methanolic extract of Ocimum sanctum L. J. Nat. Prod. Plant Resour. 2013, 3, 51–58. [Google Scholar]
  40. Dhale, D.; Mogle, U. Phytochemical screening and antibacterial activity of Phyllanthus emblica (L.). Sci. Res. Rep. 2011, 1, 138–142. [Google Scholar]
  41. Pandey, A. Antibacterial properties of Psidium guajava leaves, fruits and stems against various pathogens. Int. J. Pharm. Res. Dev. 2011, 3, 15–24. [Google Scholar]
  42. Gosset-Erard, C.; Zhao, M.J.; Lordel-Madeleine, S.; Ennahar, S. Identification of punicalagin as the bioactive compound behind the antimicrobial activity of pomegranate (Punica granatum L.) peels. Food Chem. 2021, 352, 129396. [Google Scholar] [CrossRef]
  43. Zhao, L.Y.; Liu, H.X.; Wang, L.; Xu, Z.F.; Tan, H.B.; Qiu, S.X. Rhodomyrtosone B, a membrane-targeting anti-MRSA natural acylgphloroglucinol from Rhodomyrtus tomentosa. J. Ethnopharmacol. 2019, 228, 50–57. [Google Scholar] [CrossRef]
  44. Hamoud, R.; Reichling, J.; Wink, M. Synergistic antibacterial activity of the combination of the alkaloid sanguinarine with EDTA and the antibiotic streptomycin against multidrug resistant bacteria. J. Pharm. Pharmacol. 2015, 67, 264–273. [Google Scholar] [CrossRef] [PubMed]
  45. Gehrke, I.T.S.; Neto, A.T.; Pedroso, M.; Mostardeiro, C.P.; Da Cruz, I.B.M.; Silva, U.F.; Ilha, V.; Dalcol, I.I.; Morel, A.F. Antimicrobial activity of Schinus lentiscifolius (Anacardiaceae). J. Ethnopharmacol. 2013, 148, 486–491. [Google Scholar] [CrossRef]
  46. Webber, M.A.; Piddock, L.J.V. The importance of efflux pumps in bacterial antibiotic resistance. J. Antimicrob. Chemother. 2003, 51, 9–11. [Google Scholar] [CrossRef] [PubMed]
  47. Piddock, L.J.V. Clinically relevant chromosomally encoded multidrug resistance efflux pumps in bacteria. Clin. Microbiol. Rev. 2006, 19, 382–402. [Google Scholar] [CrossRef]
  48. Van Bambeke, F.; Balzi, E.; Tulkens, P.M. Antibiotic efflux pumps—Commentary. Biochem. Pharmacol. 2000, 60, 457–470. [Google Scholar] [CrossRef]
  49. Bush, K. Past and present perspectives on beta-lactamases. Antimicrob. Agents Chemother. 2018, 62. [Google Scholar] [CrossRef]
  50. Tehrani, K.H.M.E.; Martin, N.I. β-lactam/β-lactamase inhibitor combinations: An update. Medchemcomm 2018, 9, 1439–1456. [Google Scholar] [CrossRef]
  51. Tooke, C.L.; Hinchliffe, P.; Bragginton, E.C.; Colenso, C.K.; Hirvonen, V.H.A.; Takebayashi, Y.; Spencer, J. β-Lactamases and β-Lactamase Inhibitors in the 21st Century. J. Mol. Biol. 2019, 431, 3472–3500. [Google Scholar] [CrossRef] [PubMed]
  52. Varela, M.F.; Stephen, J.; Lekshmi, M.; Ojha, M.; Wenzel, N.; Sanford, L.M.; Hernandez, A.J.; Parvathi, A.; Kumar, S.H. Bacterial resistance to antimicrobial agents. Antibiotics 2021, 10, 593. [Google Scholar] [CrossRef] [PubMed]
  53. Mazzini, S.; Bellucci, M.C.; Mondelli, R. Mode of binding of the cytotoxic alkaloid berberine with the double helix oligonucleotide D(AAGAATTCTT). Bioorg. Med. Chem. 2003, 11, 505–514. [Google Scholar] [CrossRef]
  54. Imanshahidi, M.; Hosseinzadeh, H. Pharmacological and therapeutic effects of Berberis vulgaris and its active constituent, berberine. Phytother. Res. 2008, 22, 999–1012. [Google Scholar] [CrossRef]
  55. Huang, C.G.; Chu, Z.L.; Wei, S.J.; Jiang, H.; Jiao, B.H. Effect of berberine on arachidonic acid metabolism in rabbit platelets and endothelial cells. Thromb. Res. 2002, 106, 223–227. [Google Scholar] [CrossRef] [PubMed]
  56. Ruan, H.; Zhan, Y.Y.; Hou, J.; Xu, B.; Chen, B.; Tian, Y.; Wu, D.; Zhao, Y.; Zhang, Y.; Chen, X.; et al. Berberine binds RXRα to suppress β-catenin signaling in colon cancer cells. Oncogene 2017, 36, 6906–6918. [Google Scholar] [CrossRef]
  57. Ko, W.H.; Yao, X.Q.; Lau, C.W.; Law, W.I.; Chen, Z.Y.; Kwok, W.; Ho, K.; Huang, Y. Vasorelaxant and antiproliferative effects of berberine. Eur. J. Pharmacol. 2000, 399, 187–196. [Google Scholar] [CrossRef]
  58. Liu, Y.M.; Niu, L.; Wang, L.L.; Bai, L.; Fang, X.Y.; Li, Y.C.; Yi, L.T. Berberine attenuates depressive-like behaviors by suppressing neuro-inflammation in stressed mice. Brain Res. Bull. 2017, 134, 220–227. [Google Scholar] [CrossRef]
  59. Kim, T.S.; Kang, B.Y.; Cho, D.H.; Kim, S.H. Induction of interleukin-12 production in mouse macrophages by berberine, a benzodioxoloquinolizine alkaloid, deviates CD4+T cells from a Th2 to a Th1 response. Immunology 2003, 109, 407–414. [Google Scholar] [CrossRef]
  60. Vinod, N.V.; Shijina, R.; Dileep, K.V.; Sadasivan, C. Inhibition of beta-lactamase by 1,4-Naphthalenedione from the Plant Holoptelea integrifolia. Appl. Biochem. Biotechnol. 2010, 160, 1752–1759. [Google Scholar] [CrossRef]
  61. Elfaky, M.A.; El-Halawany, A.M.; Koshak, A.E.; Alshali, K.Z.; El-Araby, M.E.; Khayat, M.T.; Abdallah, H.M. Bioassay guided isolation and docking studies of a potential β-lactamase inhibitor from Clutia myricoides. Molecules 2020, 25, 2566. [Google Scholar] [CrossRef]
  62. Zhou, X.; Jia, F.; Liu, X.; Yang, J.; Zhang, Y.; Wang, Y. In vitro synergistic interaction of 5-O-methylglovanon and ampicillin against ampicillin resistant Staphylococcus aureus and Staphylococcus epidermidis isolates. Arch. Pharm. Res. 2011, 34, 1751–1757. [Google Scholar] [CrossRef] [PubMed]
  63. Gibbons, S.; Oluwatuyi, M.; Kaatz, G.W. A novel inhibitor of multidrug efflux pumps in Staphylococcus aureus. J. Antimicrob. Chemoth. 2003, 51, 13–17. [Google Scholar] [CrossRef] [PubMed]
  64. Konaté, K.; Mavoungou, J.F.; Lepengué, A.N.; Aworet-Samseny, R.R.R.; Hilou, A.; Souza, A.; Dicko, M.H.; M’Batchi, B. Antibacterial activity against β-lactamase producing methicillin and ampicillin-resistants Staphylococcus aureus: Fractional Inhibitory Concentration Index (FICI) determination. Ann. Clin. Microb. Antimicrob. 2012, 11, 18. [Google Scholar] [CrossRef]
  65. Suga, T.; Ishii, T.; Iwatsuki, M.; Yamamoto, T.; Nonaka, K.; Masuma, R.; Matsui, H.; Hanaki, H.; Omura, S.; Shiomi, K. Aranorosin circumvents arbekacin-resistance in MRSA by inhibiting the bifunctional enzyme AAC(6′)/APH(2″). J. Antibiot. 2012, 65, 527–529. [Google Scholar] [CrossRef]
  66. Chan, B.C.; Ip, M.; Lau, C.B.; Lui, S.L.; Jolivalt, C.; Ganem-Elbaz, C.; Litaudon, M.; Reiner, N.E.; Gong, H.; See, R.H.; et al. Synergistic effects of baicalein with ciprofloxacin against NorA over-expressed methicillin-resistant Staphylococcus aureus (MRSA) and inhibition of MRSA pyruvate kinase. J. Ethnopharmacol. 2011, 137, 767–773. [Google Scholar] [CrossRef]
  67. Qian, M.Y.; Tang, S.S.; Wu, C.M.; Wang, Y.; He, T.; Chen, T.T.; Xiao, X.L. Synergy between baicalein and penicillins against penicillinase-producing Staphylococcus aureus. Int. J. Med. Microbiol. 2015, 305, 501–504. [Google Scholar] [CrossRef] [PubMed]
  68. Aghayan, S.S.; Kalalian Mogadam, H.; Fazli, M.; Darban-Sarokhalil, D.; Khoramrooz, S.S.; Jabalameli, F.; Yaslianifard, S.; Mirzaii, M. The effects of berberine and palmatine on efflux pumps inhibition with different gene patterns in Pseudomonas aeruginosa isolated from burn infections. Avicenna J. Med. Biotechnol. 2017, 9, 2–7. [Google Scholar]
  69. Fiamegos, Y.C.; Kastritis, P.L.; Exarchou, V.; Han, H.; Bonvin, A.M.J.J.; Vervoort, J.; Lewis, K.; Hamblin, M.R.; Tegos, G.P. Antimicrobial and efflux pump inhibitory activity of caffeoylquinic acids from Artemisia absinthium against Gram-positive pathogenic bacteria. PLoS ONE 2011, 6, e18127. [Google Scholar] [CrossRef]
  70. Kalia, N.P.; Mahajan, P.; Mehra, R.; Nargotra, A.; Sharma, J.P.; Koul, S.; Khan, I.A. Capsaicin, a novel inhibitor of the NorA efflux pump, reduces the intracellular invasion of Staphylococcus aureus. J. Antimicrob. Chemother. 2012, 67, 2401–2408. [Google Scholar] [CrossRef]
  71. Dwivedi, G.R.; Tyagi, R.; Sanchita; Tripathi, S.; Pati, S.; Srivastava, S.K.; Darokar, M.P.; Sharma, A. Antibiotics potentiating potential of catharanthine against superbug Pseudomonas aeruginosa. J. Biomol. Struct. Dyn. 2018, 36, 4270–4284. [Google Scholar] [CrossRef]
  72. Siriyong, T.; Srimanote, P.; Chusri, S.; Yingyongnarongkul, B.E.; Suaisom, C.; Tipmanee, V.; Voravuthikunchai, S.P. Conessine as a novel inhibitor of multidrug efflux pump systems in Pseudomonas aeruginosa. BMC Complement. Altern. Med. 2017, 17, 405. [Google Scholar] [CrossRef] [PubMed]
  73. Roy, S.K.; Kumari, N.; Pahwa, S.; Agrahari, U.C.; Bhutani, K.K.; Jachak, S.M.; Nandanwar, H. NorA efflux pump inhibitory activity of coumarins from Mesua ferrea L. Fitoterapia 2013, 90, 140–150. Fitoterapia 2013, 90, 140–150. [Google Scholar] [CrossRef] [PubMed]
  74. Stermitz, F.R.; Scriven, L.N.; Tegos, G.; Lewis, K. Two flavonols from Artemisa annua which potentiate the activity of berberine and norfloxacin against a resistant strain of Staphylococcus aureus. Planta Medica 2002, 68, 1140–1141. [Google Scholar] [CrossRef]
  75. Ramalhete, C.; Spengler, G.; Martins, A.; Martins, M.; Viveiros, M.; Mulhovo, S.; Ferreira, M.-J.U.; Amaral, L. Inhibition of efflux pumps in methicillin-resistant Staphylococcus aureus and Enterococcus faecalis resistant strains by triterpenoids from Momordica balsamina. Int. J. Antimicrob. Agents 2011, 37, 70–74. [Google Scholar] [CrossRef] [PubMed]
  76. Kakarla, P.; Floyd, J.; Mukherjee, M.; Devireddy, A.R.; Inupakutika, M.A.; Ranweera, I.; Ranjana, K.C.; Shrestha, U.; Cheeti, U.R.; Willmon, T.M.; et al. Inhibition of the multidrug efflux pump LmrS from Staphylococcus aureus by cumin spice Cuminum cyminum. Arch. Microbiol. 2017, 199, 465–474. [Google Scholar] [CrossRef]
  77. Zhao, W.H.; Hu, Z.Q.; Okubo, S.; Hara, Y.; Shimamura, T. Mechanism of synergy between epigallocatechin gallate and β-lactams against methicillin-resistant Staphylococcus aureus. Antimicrob. Agents Chemother. 2001, 45, 1737–1742. [Google Scholar] [CrossRef]
  78. Smith, E.C.J.; Williamson, E.M.; Wareham, N.; Kaatz, G.W.; Gibbons, S. Antibacterials and modulators of bacterial resistance from the immature cones of Chamaecyparis lawsoniana. Phytochemistry 2007, 68, 210–217. [Google Scholar] [CrossRef]
  79. Bag, A.; Chattopadhyay, R.R. Efflux-pump inhibitory activity of a gallotannin from Terminalia chebula fruit against multidrug-resistant uropathogenic Escherichia coli. Nat. Prod. Res. 2014, 28, 1280–1283. [Google Scholar] [CrossRef]
  80. Ponnusamy, K.; Ramasamy, M.; Savarimuthu, I.; Paulraj, M.G. Indirubin potentiates ciprofloxacin activity in the NorA efflux pump of Staphylococcus aureus. Scand. J. Infect. Dis. 2010, 42, 500–505. [Google Scholar] [CrossRef]
  81. Holler, J.G.; Christensen, S.B.; Slotved, H.C.; Rasmussen, H.B.; Gúzman, A.; Olsen, C.E.; Petersen, B.; Molgaard, P. Novel inhibitory activity of the NorA efflux pump by a kaempferol rhamnoside isolated from Persea lingue Nees. J. Antimicrob. Chemother. 2012, 67, 1138–1144. [Google Scholar] [CrossRef] [PubMed]
  82. Fakudze, N.T.; Sarbadhikary, P.; George, B.P.; Abrahamse, H. Ethnomedicinal Uses, phytochemistry, and anticancer potentials of African medicinal fruits: A Comprehensive Review. Pharmaceuticals 2023, 16, 1117. [Google Scholar] [CrossRef] [PubMed]
  83. Joshi, P.; Singh, S.; Wani, A.; Sharma, S.; Jain, S.K.; Singh, B.; Gupta, B.D.; Satti, N.K.; Koul, S.; Khan, I.A.; et al. Osthol and curcumin as inhibitors of human Pgp and multidrug efflux pumps of Staphylococcus aureus: Reversing the resistance against frontline antibacterial drugs. MedChemComm 2014, 5, 1540–1547. [Google Scholar] [CrossRef]
  84. Negi, N.; Prakash, P.; Gupta, M.L.; Mohapatra, T.M. Possible role of curcumin as an efflux pump inhibitor in multi drug resistant clinical isolates of Pseudomonas aeruginosa. J. Clin. Diagn. Res. 2014, 8, DC04–DC07. [Google Scholar] [CrossRef] [PubMed]
  85. Cabral, V.; Luo, X.; Junqueira, E.; Costa, S.S.; Mulhovo, S.; Duarte, A.; Couto, I.; Viveiros, M.; Ferreira, M.J. Enhancing activity of antibiotics against Staphylococcus aureus: Zanthoxylum capense constituents and derivatives. Phytomedicine 2015, 22, 469–476. [Google Scholar] [CrossRef]
  86. Mahamoud, A.; Chevalier, J.; Alibert-Franco, S.; Kern, W.V.; Pagès, J.M. Antibiotic efflux pumps in Gram-negative bacteria: The inhibitor response strategy. J. Antimicrob. Chemother. 2007, 59, 1223–1229. [Google Scholar] [CrossRef]
  87. Coates, N.J.; Gilpin, M.L.; Gwynn, M.N.; Lewis, D.E.; Milner, P.H.; Spear, S.R.; Tyler, J.W. SB-202742, a novel beta-lactamase inhibitor isolated from Spondias mombin. J. Nat. Prod. 1994, 57, 654–657. [Google Scholar] [CrossRef]
  88. Wang, D.; Xie, K.; Zou, D.; Meng, M.; Xie, M. Inhibitory effects of silybin on the efflux pump of methicillin-resistant Staphylococcus aureus. Mol. Med. Rep. 2018, 18, 827–833. [Google Scholar]
  89. Shiota, S.; Shimizu, M.; Sugiyama, J.i.; Morita, Y.; Mizushima, T.; Tsuchiya, T. Mechanisms of action of corilagin and tellimagrandin I that remarkably potentiate the activity of β-lactams against methicillin-resistant Staphylococcus aureus. Microbiol. Immunol. 2004, 48, 67–73. [Google Scholar] [CrossRef]
  90. Koche, D.; Shirsat, R.; Kawale, M. An overview of major classes of phytochemicals: Their types and role in disease prevention. Hislopia J. 2016, 9, 1–11. [Google Scholar]
  91. Hegnauer, R. Biochemistry, Distribution and Taxonomic Relevance of Higher-Plant Alkaloids. Phytochemistry 1988, 27, 2423–2427. [Google Scholar] [CrossRef]
  92. Atawodi, S.E.; Atawodi, J.C. Azadirachta indica (neem): A plant of multiple biological and pharmacological activities. Phytochem. Rev. 2009, 8, 601–620. [Google Scholar] [CrossRef]
  93. Kubo, I.; Muroi, H.; Kubo, A. Structural Functions of Antimicrobial Long-Chain Alcohols and Phenols. Bioorg. Med. Chem. 1995, 3, 873–880. [Google Scholar] [CrossRef]
  94. Merkl, R.; Hrádková, I.; Filip, V.; Smidrkal, J. Antimicrobial and Antioxidant Properties of Phenolic Acids Alkyl Esters. Czech J. Food Sci. 2010, 28, 275–279. [Google Scholar] [CrossRef]
  95. Cueva, C.; Moreno-Arribas, M.V.; Martín-Alvarez, P.J.; Bills, G.; Vicente, M.F.; Basilio, A.; Rivas, C.L.; Requena, T.; Rodríguez, J.M.; Bartolomé, B. Antimicrobial activity of phenolic acids against commensal, probiotic and pathogenic bacteria. Res. Microbiol. 2010, 161, 372–382. [Google Scholar] [CrossRef]
  96. Stapleton, P.D.; Shah, S.; Anderson, J.C.; Hara, Y.; Hamilton-Miller, J.M.T.; Taylor, P.W. Modulation of β-lactam resistance in Staphylococcus aureus by catechins and gallates. Int. J. Antimicrob. Agents 2004, 23, 462–467. [Google Scholar] [CrossRef]
  97. Wagner, H.; Ulrich-Merzenich, G. Synergy research: Approaching a new generation of phytopharmaceuticals. Phytomedicine 2009, 16, 97–110. [Google Scholar] [CrossRef] [PubMed]
  98. Marquez, B.; Neuville, L.; Moreau, N.J.; Genet, J.P.; dos Santos, A.F.; de Andrade, M.C.C.; Sant’Ana, A.E.G. Multidrug resistance reversal agent from Jatropha elliptica. Phytochemistry 2005, 66, 1804–1811. [Google Scholar] [CrossRef]
  99. Obiang-Obounou, B.W.; Jang, Y.P. Enriching modern pharmacotherapy through synergy assessment for the combination of natural products and synthetic drugs. Arch. Pharm. Res. 2011, 34, 1579–1581. [Google Scholar] [CrossRef]
  100. Freitas, P.R.; de Araújo, A.C.J.; Barbosa, C.R.; Muniz, D.F.; Tintino, S.R.; Ribeiro, J.; Siqueira, J.P.; Filho, J.M.B.; de Sousa, G.R.; Coutinho, H.D.M. Inhibition of efflux pumps by monoterpene (α-Pinene) and impact on Staphylococcus aureus resistance to tetracycline and erythromycin. Curr. Drug Metab. 2021, 22, 123–126. [Google Scholar] [CrossRef]
  101. Liu, I.X.; Durham, D.G.; Richards, R.M.E. Baicalin synergy with β-lactam antibiotics against methicillin-resistant Staphylococcus aureus and other β-lactam-resistant strains of S. aureus. J. Pharm. Pharmacol. 2000, 52, 361–366. [Google Scholar] [CrossRef] [PubMed]
  102. 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] [PubMed]
  103. Magnini, R.D.; Pedinielli, F.; Vergalli, J.; Ouedraogo, N.; Remy, S.; Hilou, A.; Brunel, J.M.; Pagés, J.M.; Davin-Regli, A. Acacia senegal Budmunchiamines as a potential adjuvant for rejuvenating phenicol activities towards Escherichia coli-resistant strains. Int. J. Mol. Sci. 2023, 24, 8790. [Google Scholar] [CrossRef]
  104. Oluwatuyi, M.; Kaatz, G.W.; Gibbons, S. Antibacterial and resistance modifying activity of Rosmarinus officinalis. Phytochemistry 2004, 65, 3249–3254. [Google Scholar] [CrossRef]
  105. Magi, G.; Marini, E.; Facinelli, B. Antimicrobial activity of essential oils and carvacrol, and synergy of carvacrol and erythromycin, against clinical, erythromycin-resistant Group A Streptococci. Front. Microbiol. 2015, 6, 165. [Google Scholar] [CrossRef] [PubMed]
  106. Dwivedi, G.R.; Maurya, A.; Yadav, D.K.; Singh, V.; Khan, F.; Gupta, M.K.; Singh, M.; Darokar, M.P.; Srivastava, S.K. Synergy of clavine alkaloid ‘chanoclavine’ with tetracycline against multi-drug-resistant E. coli. J. Biomol. Struct. Dyn. 2019, 37, 1307–1325. [Google Scholar] [CrossRef]
  107. Zhou, Y.L.; Lv, X.H.; Chen, M.S.; Guo, Y.; Ding, R.; Liu, B.; Deng, X.M.; Wang, J.F. Characterization of corosolic acid as a KPC-2 inhibitor that increases the susceptibility of KPC-2-positive bacteria to carbapenems. Front. Pharmacol. 2020, 11, 1047. [Google Scholar] [CrossRef]
  108. Sundaramoorthy, N.S.; Sivasubramanian, A.; Nagarajan, S. Simultaneous inhibition of MarR by salicylate and efflux pumps by curcumin sensitizes colistin resistant clinical isolates of Enterobacteriaceae. Microb. Pathog. 2020, 148, 104445. [Google Scholar] [CrossRef]
  109. Roccaro, A.S.; Blanco, A.R.; Giuliano, F.; Rusciano, D.; Enea, V. Epigallocatechin-gallate enhances the activity of tetracycline in staphylococci by inhibiting its efflux from bacterial cells. Antimicrob. Agents Chemother. 2004, 48, 1968–1973. [Google Scholar] [CrossRef]
  110. Hu, Z.Q.; Zhao, W.H.; Hara, Y.; Shimamura, T. Epigallocatechin gallate synergy with ampicillin/sulbactam against 28 clinical isolates of methicillin-resistant Staphylococcus aureus. J. Antimicrob. Chemother. 2001, 48, 361–364. [Google Scholar] [CrossRef]
  111. Sanhueza, L.; Melo, R.; Montero, R.; Maisey, K.; Mendoza, L.; Wilkens, M. Synergistic interactions between phenolic compounds identified in grape pomace extract with antibiotics of different classes against Staphylococcus aureus and Escherichia coli. PLoS ONE 2017, 12, e0172273. [Google Scholar] [CrossRef] [PubMed]
  112. Zhou, Y.L.; Guo, Y.; Wen, Z.M.; Ci, X.X.; Xia, L.N.; Wang, Y.L.; Deng, X.M.; Wang, J.F. Isoalantolactone enhances the antimicrobial activity of penicillin G against Staphylococcus aureus by inactivating β-lactamase during protein translation. Pathogens 2020, 9, 161. [Google Scholar] [CrossRef]
  113. Gibbons, S.; Oluwatuyi, M.; Veitch, N.C.; Gray, A.I. Bacterial resistance modifying agents from Lycopus europaeus. Phytochemistry 2003, 62, 83–87. [Google Scholar] [CrossRef]
  114. Gallique, M.; Wei, K.; Maisuria, V.B.; Okshevsky, M.; McKay, G.; Nguyen, D.; Tufenkji, N. Cranberry-derived proanthocyanidins potentiate β-lactam antibiotics against resistant bacteria. Appl. Environ. Microb. 2021, 87, e00127-21. [Google Scholar] [CrossRef]
  115. Pal, A.; Tripathi, A. Quercetin inhibits carbapenemase and efflux pump activities among carbapenem-resistant Gram-negative bacteria. APMIS 2020, 128, 251–259. [Google Scholar] [CrossRef]
  116. Yi, K.F.; Liu, S.B.; Liu, P.Y.; Luo, X.W.; Zhao, J.F.; Yan, F.B.; Pan, Y.S.; Liu, J.H.; Zhai, Y.J.; Hu, G.Z. Synergistic antibacterial activity of tetrandrine combined with colistin against MCR-mediated colistin-resistant Salmonella. Biomed. Pharmacother. 2022, 149, 112873. [Google Scholar] [CrossRef] [PubMed]
  117. Kilic, I.; Ayar, Y.; Ceylan, I.; Kaya, P.K.; Caliskan, G. Nephrotoxicity caused by colistin use in ICU: A single centre experience. BMC Nephrol. 2023, 24, 302. [Google Scholar] [CrossRef]
  118. Barbosa, C.R.D.; Scherf, J.R.; de Freitas, T.S.; de Menezes, I.R.A.; Pereira, R.L.S.; dos Santos, J.F.S.; de Jesus, S.S.P.; Lopes, T.P.; Silveira, Z.D.; Oliveira-Tintino, C.D.D.; et al. Effect of Carvacrol and Thymol on NorA efflux pump inhibition in multidrug-resistant (MDR) Staphylococcus strains. J. Bioenerg. Biomembr. 2021, 53, 489–498. [Google Scholar] [CrossRef] [PubMed]
  119. Sharma, A.; Gupta, V.K.; Pathania, R. Efflux pump inhibitors for bacterial pathogens: From bench to bedside. Indian J. Med. Res. 2019, 149, 129–145. [Google Scholar] [CrossRef]
  120. Wang, J.L.; Jiao, H.H.; Meng, J.W.; Qiao, M.Y.; Du, H.X.; He, M.; Ming, K.; Liu, J.G.; Wang, D.Y.; Wu, Y. Baicalin inhibits biofilm formation and the quorum-sensing system by regulating the MsrA drug efflux pump in Staphylococcus saprophyticus. Front. Microbiol. 2019, 10, 2800. [Google Scholar] [CrossRef]
  121. Hu, Z.Y.; Zhou, Z.G.; Hu, Y.H.; Wu, J.H.; Li, Y.M.; Huang, W.L. HZ08 reverse p-glycoprotein mediated multidrug resistance in vitro and in vivo. PLoS ONE 2015, 10, e0116886. [Google Scholar] [CrossRef] [PubMed]
  122. Gottesman, M.M.; Pastan, I. Biochemistry of multidrug-resistance mediated by the multidrug transporter. Annu. Rev. Biochem. 1993, 62, 385–427. [Google Scholar] [CrossRef] [PubMed]
  123. Yu, D.K. The contribution of P-glycoprotein to pharmacokinetic drug-drug interactions. J. Clin. Pharmacol. 1999, 39, 1203–1211. [Google Scholar] [CrossRef]
  124. Park, S.; Lee, K.M.; Yoo, Y.S.; Yoo, J.S.; Yoo, J.I.; Kim, H.S.; Lee, Y.S.; Chung, G.T. Alterations of gyrA, gyrB, and parC and activity of efflux pump in fluoroquinolone-resistant Acinetobacter baumannii. Osong Public Health Res. Perspect. 2011, 2, 164–170. [Google Scholar] [CrossRef] [PubMed]
  125. Buchmann, D.; Schultze, N.; Borchardt, J.; Bottcher, I.; Schaufler, K.; Guenther, S. Synergistic antimicrobial activities of epigallocatechin gallate, myricetin, daidzein, gallic acid, epicatechin, 3-hydroxy-6-methoxyflavone and genistein combined with antibiotics against ESKAPE pathogens. J. Appl. Microbiol. 2022, 132, 949–963. [Google Scholar] [CrossRef]
  126. Nakayama, M.; Shimatani, K.; Ozawa, T.; Shigemune, N.; Tomiyama, D.; Yui, K.; Katsuki, M.; Ikeda, K.; Nonaka, A.; Miyamoto, T. Mechanism for the antibacterial action of epigallocatechin gallate (EGCg) on Bacillus subtilis. Biosci. Biotechnol. Biochem. 2015, 79, 845–854. [Google Scholar] [CrossRef]
  127. Singkham-In, U.; Higgins, P.G.; Wannigama, D.L.; Hongsing, P.; Chatsuwan, T. Rescued chlorhexidine activity by resveratrol against carbapenem-resistant Acinetobacter baumannii via down-regulation of AdeB efflux pump. PLoS ONE 2020, 15, e0243082. [Google Scholar] [CrossRef]
  128. Bush, K.; Bradford, P.A. Beta-lactams and beta-lactamase inhibitors: An Overview. Cold Spring Harb. Perspect. Med. 2016, 6, a025247. [Google Scholar] [CrossRef]
  129. Fleming, A. On the antibacterial action of cultures of a penicillium, with special reference to their use in the isolation of B. influenzae. Br. J. Exp. Pathol. 1929, 10, 226–236. [Google Scholar] [CrossRef]
  130. Abraham, E.P.; Chain, E. An enzyme from bacteria able to destroy penicillin. Nature 1940, 146, 837. [Google Scholar] [CrossRef]
  131. Reading, C.; Cole, M. Clavulanic acid: A Beta-lactamase-inhibiting beta-lactam from Streptomyces-Clavuligerus. Antimicrob. Agents Chemother. 1977, 11, 852–857. [Google Scholar] [CrossRef] [PubMed]
  132. Fisher, J.; Charnas, R.L.; Knowles, J.R. Kinetic studies on inactivation of Escherichia coli RTEM beta-lactamase by clavulanic acid. Biochemistry 1978, 17, 2180–2184. [Google Scholar] [CrossRef] [PubMed]
  133. Zhao, W.H.; Hu, Z.Q.; Hara, Y.; Shimamura, T. Inhibition of penicillinase by epigallocatechin gallate resulting in restoration of antibacterial activity of penicillin against penicillinase-producing Staphylococcus aureus. Antimicrob. Agents Chemother. 2002, 46, 2266. [Google Scholar] [CrossRef] [PubMed]
  134. Siriwong, S.; Teethaisong, Y.; Thumanu, K.; Dunkhunthod, B.; Eumkeb, G. The synergy and mode of action of quercetin plus amoxicillin against amoxicillin-resistant Staphylococcus epidermidis. BMC Pharmacol. Toxicol. 2016, 17, 39. [Google Scholar] [CrossRef]
  135. Ghosh, A.; Das, B.K.; Roy, A.; Mandal, B.; Chandra, G. Antibacterial activity of some medicinal plant extracts. J. Nat. Med. 2008, 62, 259–262. [Google Scholar] [CrossRef]
  136. Deutch, C.E. Limited effectiveness of over-the-counter plant preparations used for the treatment of urinary tract infections as inhibitors of the urease activity from Staphylococcus saprophyticus. J. Appl. Microbiol. 2017, 122, 1380–1388. [Google Scholar] [CrossRef]
  137. 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]
Figure 1. Base structures of (a) phenolic acid, (b) coumarins, (c) flavonoids, (d) lignans, (e) stilbenes, (f) tannins and (g) terpenoids.
Figure 1. Base structures of (a) phenolic acid, (b) coumarins, (c) flavonoids, (d) lignans, (e) stilbenes, (f) tannins and (g) terpenoids.
Ijms 26 05550 g001
Figure 2. Efflux pump families.
Figure 2. Efflux pump families.
Ijms 26 05550 g002
Figure 3. Inactivation of penicillin by beta lactamase enzymes. The red line indicates the peptide bond that is hydrolysed by the enzyme.
Figure 3. Inactivation of penicillin by beta lactamase enzymes. The red line indicates the peptide bond that is hydrolysed by the enzyme.
Ijms 26 05550 g003
Figure 4. (a) Berberine, (b) 1, 4-naphthalenedione, (c) 2-methoxy chrysophanol (d) aranorosin, (e) baicalein, (f) capsaicin, (g) catharanthine, (h) conessine, (i) epigallocatechin gallate, (j) ferruginol, (k) indirubin, (l) kaempferol rhamnoside, (m) oleanolic acid, (n) osthol, (o) reserpine and (p) silybin.
Figure 4. (a) Berberine, (b) 1, 4-naphthalenedione, (c) 2-methoxy chrysophanol (d) aranorosin, (e) baicalein, (f) capsaicin, (g) catharanthine, (h) conessine, (i) epigallocatechin gallate, (j) ferruginol, (k) indirubin, (l) kaempferol rhamnoside, (m) oleanolic acid, (n) osthol, (o) reserpine and (p) silybin.
Ijms 26 05550 g004
Figure 5. Efflux pump inhibitory mechanisms (indicated by the red crosses), including proton motive force disruption, blockage of ATP synthesis, altering the integrity of the membrane permeability, down-regulating gene expression and targeting the functional assembly of the efflux pump. The black rectangles indicate altered membrane permeability.
Figure 5. Efflux pump inhibitory mechanisms (indicated by the red crosses), including proton motive force disruption, blockage of ATP synthesis, altering the integrity of the membrane permeability, down-regulating gene expression and targeting the functional assembly of the efflux pump. The black rectangles indicate altered membrane permeability.
Ijms 26 05550 g005
Figure 6. β-lactamase inhibitor mechanism of action.
Figure 6. β-lactamase inhibitor mechanism of action.
Ijms 26 05550 g006
Table 1. Antimicrobial activity of plant secondary metabolites.
Table 1. Antimicrobial activity of plant secondary metabolites.
Plant NameSecondary MetabolitesAntimicrobial Activity AgainstReferences
Arbutus unedo L.Anthocyanins, flavonoids, Quinines,
tannins
S. aureus, E. coli, P. aeruginosa[27]
Artabotrys crassifolius Hook.f. & ThomsonLiridineB. cereus[28]
Bauhinia purpurea Linn.Alkaloids, fatty acids, phytol esters
Steroids, triterpenoids,
A. niger, B. subtilis, Claviceps purpurea, E. coli, Klebsiella, P. aeruginosa, S. aureus, S. typhimurium[29]
Berberis vulgaris L., Berberis petiolaris Kunth., Berberis aristate DC., Berberis integerrima Bonge.BerberineBrucella abortus[30]
Cananga odorata (Lam.) Hook. f. & Thomson.SampangineM. intracellulare[31]
Cannabis sativa L.CannabinoidsMRSA, E. faecalis, S. pneumoniae[32]
Cassia auriculatalinn Linn.Alkaloids, flavonoids, glycosides,
phenols terpenoids, proteins, saponins,
tannins,
B. subtilis, C. albicans, Aspergillus niger, E. coli, S. aureus[33]
Cnidoscolus acontifolius (Mill) I.M.Johnst, Newbouldia laevis (P.Beauv.) Seem.ex Bureau, Adansonia digitata L., Alchornea laxiflorab (Benth.) Pax & K. Hoffm.,Alkaloids, anthraquinones, reducing sugars flavonoids, phenols, resins, Saponins, steroids tannins, terpenoids, carbohydrates, cardioactive glycosidesE. coli, S. aureus, B. subtilis, P. aeruginosa[34]
Gloriosa superba Linn.Terpenoids, tannin, steroids, glycosides, alkaloidsB. cereus, B. subtilis, E. coli, K. pneumonia, P. aeruginosa, P. vulgaris, S. aureus, S. faecalis, S. typhimurium, S. cremoris[35]
Glycyrrhiza inflata Batalin.Licochalcone AS. aureus[36]
Mammea americana L.Mammea B/BAMRSA, S. aureus[37]
Momordica charantia L.Alkaloids, flavonoids, glycosides,
Saponins, steroids, tannins
B. subtilis, E. coli, P. aeruginosa,
S. aureus
[38]
Ocimum sanctum L.Alkaloids, Steroidal compounds,
tannins
E. coli, S. aureus, P. mirabilis[39]
Phyllanthus emblica L.Alkaloids, fats,
glyceroids, carbohydrates, phenolics, lignin, oil, tannins, flavonoids,
saponins, terpenoids
E. coli, S. aureus, P. aeruginosa, B. subtilis[40]
Psidium guajava L.Alkaloid, polyphenols, saponins,
tannins, terpenoids,
S. aureus, E. coli, P. aeruginosa
[41]
Punica granatum L.PunicalaginS. aureus, P. aeruginosa, Enterococcus mundtii[42]
Rhodomyrtus tomentosa (Ait.) Hassk.Rhodomyrtosone BVancomycin-resistant E. faecium, MRSA[43]
Sanguinaria canadensis L.SanguinarineB. subtilis, S. epidermidis, S. aureus, MRSA[44]
Schinus lentiscifolius Marchand.Moronic acidS. pyogenes, S. aureus, B. subtilis[45]
Table 2. Phytochemicals possessing efflux pump and β-lactamase inhibition activity.
Table 2. Phytochemicals possessing efflux pump and β-lactamase inhibition activity.
PhytochemicalsPlant NameEfflux Pump Inhibitory Activity Againstβ-Lactamase Inhibitory Activity AgainstReferences
1,4-Naphthalenedione (Figure 4b)Holoptelea integrifolia (Roxb.) Planch.Not reportedS. aureus[60]
2-Methoxy chrysophanol (Figure 4c)Clutia myricoides Jaub. & SpachNot reportedK. pneumoniae[61]
5-O-MethylglovanonGlycosmis plantsNot reportedS. epidermidis (MIC = 25–50 µg/mL) and ampicillin-resistant S. aureus (MIC = 12.50–50 µg/mL)[62]
Abietane diterpenesRosmarinus officinalis L.S. aureus (MIC = 16–64 µg/mL)Not reported[63]
Alkaloid compoundsCienfuegosia digitata Cav.Not reportedAmpicillin and methicillin-resistant S. aureus (ZOI = 10–16 mm)[64]
Aranorosin (Figure 4d)Gymnascella aurantiaca. PeckNot reportedMRSA (ZOI = 10 mm)[65]
Baicalein (Figure 4e)Scutellaria baicalensis GeorgiMRSA (MIC = 64–256 µg/mL)S. aureus (MIC = 128 µg/mL)[66,67]
Berberine (Figure 4a)Berberis spp.P. aeruginosa (MIC = 125–250 µg/mL)Not reported[68]
Caffeoylquinic acidsArtemisia absinthium L.E. faecalis, S. aureus (MIC = 32–>256 µg/mL)Not reported[69]
Capsaicin (Figure 4f)Capsicum spp.S. aureus (MIC ≥ 100 µg/mL)Not reported[70]
Catharanthine (Figure 4g)Catharanthus roseus (L.) G.DonP. aeruginosa (MIC = 400 µg/mL)Not reported[71]
Conessine (Figure 4h)Holarrhena antidysenterica (L.) Wall.ex A. DC.P. aeruginosa (MIC = 40 µg/mL)Not reported[72]
CoumarinsMesua ferrea L.S. aureus (MIC = 3.12–100 µg/mL)Not reported[73]
Crysoplenol, CrysoplenetinArtemissia annua L.S. aureus (MIC = 250–500 µg/mL)Not reported[74]
Cucurbitane-type triterpenoidsMomordica balsamina L. (Cucurbitaceae)E. faecalis (MIC = 100–>200 µg/mL), MRSA (MIC = 25–50 µg/mL)Not reported[75]
CuminCuminum cyminum L.S. aureus (MIC = 5–25 µg/mL)Not reported[76]
Epigallocatechin gallate (Figure 4i)Camellia sinensis (L.) KuntzeNot reportedMRSA (MIC ≤ 100 µg/mL)[77]
Ferruginol (Figure 4j)Chamaecyparis lawsoniana (A.Murray bis) Parl.S. aureus (MIC = 4–128 µg/mL)Not reported[78]
Gallotannin glucopyranoseTerminalia chebula RetziusE. coli (MIC = 12.1–97.5 µg/mL)Not reported[79]
Indirubin (Figure 4k)Wrightia tinctoria (Roxb.) R. Br.S. epidermidis (MIC = 25 µg/mL), S. aureus (MIC = 12.5 µg/mL)Not reported[80]
Kaempferol rhamnoside (Figure 4l)Persea lingue (Ruiz & Pav.) NeesS. aureus (IC50 = 2 µM)Not reported[81]
Oleanolic acid (Figure 4m)Carpobrotus edulis (L.) L.BolusMRSA (MIC = 25–50 µg/mL)Not reported[82]
Osthol (Figure 4n)Cnidii monnieri (L.)S. aureus (MIC = 25 µg/mL), P. aeruginosa (MIC = 128 µg/mL)Not reported[83,84]
Phenylpropanoid ailanthoidiolZanthoxylum capense (Thunb.) Harv.S. aureus (MIC = 50–100 µg/mL)Not reported[85]
Reserpine (Figure 4o)Rauwolfia vomitoria Afzel., Rauwolfia serpentina (L.) Benth. ex KurzE. coli (ZOI = 15.5–16.5 mm)Not reported[86]
SB-202742 (Anacardic acid derivatives)Spondias mombin L.Not reportedE. coli (IC50 = 10.1 µg/mL), P. aeruginosa (IC50 = 40.5 µg/mL), P. mirabilis (IC50 = 111.3 µg/mL)[87]
Silybin (Figure 4p)Silybum marianum (L.) GaertnMRSA (ZOI = 29 mm)Not reported[88]
Tellimagrandin IRosa canina L.Not reportedMRSA (MIC = 150 µg/mL)[89]
Table 3. Synergistic interactions of phytochemicals in combination with clinical antibiotics.
Table 3. Synergistic interactions of phytochemicals in combination with clinical antibiotics.
PhytochemicalsCombination with AntibioticBacterial TargetInhibitory Activity AgainstReferences
2,6-dimethyl-4-phenyl-pyridine-3,5-dicarboxylic acid diethyl esterCiprofloxacinS. aureusEfflux pump (MIC = 3–8 µg/mL)[98]
5-O-MethylglovanonAmpicillinS. aureus, S. epidermidisβ-lactamase[99]
α-pineneErythromycin, TetracyclineS. aureusEfflux pump (MIC = 128 µg/mL)[100]
BaicalinCefotaxime, methicillin, benzylpenicillin, amoxicillin, ampicillinMRSAβ-lactamase (MIC = 4–25 µg/mL)[101]
Berberine5ʹ- MethoxyhydnocarpinS. aureusEfflux pump (MIC = 32 µg/mL)[102]
BudmunchiaminesChloramphenicolE. coliEfflux pump (MIC = 8–64 µg/mL)[103]
Carnosol, carnosic acidErythromycinS. aureusEfflux pump (MIC = 32–256 µg/mL)[104]
CarvacrolErythromycinErythromycin-resistant Group A StreptococciEfflux pump (MIC = 8–64 µg/mL)[105]
ChanoclavineTetracyclineMDE E. coliEfflux pump (MIC = 0.78–100 µg/mL)[106]
Corosolic acidCarbapenemsE. coliβ-lactamase (MIC = 0.08–1.5 µg/mL)[107]
Curcumin, salicylateColistinCarbapenem resistant EnterobacteriaceaeEfflux pump (MIC = 0.01–4 µg/mL)[108]
Epigallocatechin-gallateTetracyclineStaphylococcal isolatesEfflux pump (MIC = 0.06–32 µg/mL)[109]
Epigallocatechin-gallateSulbactam/ampicillinMRSAβ-Lactamase (MIC = 4 µg/mL)[110]
Gallic acid, quercetin, luteolin, protocatechuic acidAmphenicol, tetracycline, fluoroquinolone, quinolone, β lactamsE. coli, S. aureusβ-Lactamase (MIC = 0.02–47 µg/mL) for E. coli and (MIC = 0.02–7.8 µg/mL) for S. aureus[111]
IsoalantolactonePenicillin GS. aureusβ-Lactamase (MIC = 0.008–16 µg/mL)[112]
Methyl-1alpha-acetoxy-7alpha 14alpha-dihydroxy-8,15-isopimaradien-18-oate, Methyl-1alpha,14alpha-diacetoxy-7alpha-hydroxy-8,15-isopimaradien-18-oateErythromycin, tetracyclineS. aureusEfflux pump (MIC = 32–256 µg/mL)[113]
ProanthocyanidinsAmpicillin, meropenem, cefotaximeE. coli, Staphylococci strains, Klebsiellaβ-Lactamase (MIC = 37.5–2400 µg/mL)[114]
QuercetinMeropenemCarbapenem resistant K. pneumoniae, E. coli, P. aeruginosa, A. baumanniiEfflux pump[115]
Tellimagrandin I, corilagin, ellagic acid, gallic acidOxacillinMRSAβ-Lactamase (MIC = 0.25–256 µg/mL)[89]
TetrandrineColistin (Toxicity has been reported)Colistin-resistant SalmonellaEfflux pump (MIC = 0.004–4 µg/mL)[116,117]
Thymol, CarvacrolNorfloxacinS. aureusEfflux pump (MIC = 32 µg/mL)[118]
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

Zai, M.J.; Cock, I.E.; Cheesman, M.J. Plant Metabolites as Potential Agents That Potentiate or Block Resistance Mechanisms Involving β-Lactamases and Efflux Pumps. Int. J. Mol. Sci. 2025, 26, 5550. https://doi.org/10.3390/ijms26125550

AMA Style

Zai MJ, Cock IE, Cheesman MJ. Plant Metabolites as Potential Agents That Potentiate or Block Resistance Mechanisms Involving β-Lactamases and Efflux Pumps. International Journal of Molecular Sciences. 2025; 26(12):5550. https://doi.org/10.3390/ijms26125550

Chicago/Turabian Style

Zai, Muhammad Jawad, Ian Edwin Cock, and Matthew James Cheesman. 2025. "Plant Metabolites as Potential Agents That Potentiate or Block Resistance Mechanisms Involving β-Lactamases and Efflux Pumps" International Journal of Molecular Sciences 26, no. 12: 5550. https://doi.org/10.3390/ijms26125550

APA Style

Zai, M. J., Cock, I. E., & Cheesman, M. J. (2025). Plant Metabolites as Potential Agents That Potentiate or Block Resistance Mechanisms Involving β-Lactamases and Efflux Pumps. International Journal of Molecular Sciences, 26(12), 5550. https://doi.org/10.3390/ijms26125550

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