Open access peer-reviewed chapter

Non-microbial Natural Products That Inhibit Drug-Resistant Staphylococcus aureus

Written By

Jactty Chew, Suat-Cheng Peh and Teow Sin Yeang

Submitted: 10 October 2017 Reviewed: 29 January 2018 Published: 06 April 2018

DOI: 10.5772/intechopen.74588

From the Edited Volume

Staphylococcus Aureus

Edited by Hassan Hemeg, Hani Ozbak and Farhat Afrin

Chapter metrics overview

1,753 Chapter Downloads

View Full Metrics

Abstract

Drug resistance developed in human pathogenic bacteria is emerging and has become a global problem. Methicillin-resistant Staphylococcus aureus (MRSA) spreading in both hospital and community areas has posed a great impact to global public health. Current antibiotics used against these resistant strains are no longer efficacious and the search for new alternative is in urgent need. In the past decades, natural products have demonstrated multiple biological activities in biomedical areas including their antibacterial actions against various drug-resistant bacteria. More promisingly, some natural products could reverse the resistance of bacteria to the antibiotics, making the target bacteria susceptible to these drugs again. Numerous natural products have also exhibited potent synergism against the drug-resistant bacteria when used in combination with various types of antibiotics. Recently, several antibacterials derived from microbes have been developed and approved by Food and Drug Administration (FDA) for clinical use. In this chapter, we discuss the potential use of non-microbial natural products in controlling Staphylococcus aureus (S. aureus)‘s growth, and the underlying challenges in developing the natural products into clinical applications.

Keywords

  • natural products
  • Staphylococcus aureus
  • methicillin-resistant
  • antibacterial
  • MRSA

1. Introduction

Staphylococcus aureus (S. aureus) is a coagulase-positive Gram-positive cocci bacterium, commonly found on human skin and mucous membranes. Up to 30% of the world population is colonised by this bacterium [1]. Despite being part of the human normal microbiota, it is known to be a pathogen causing various levels of diseases ranging from mild skin infections such as boils and rashes, to life-threatening diseases such as persistent bacteraemia, sepsis, and pneumonia [2]. The pathogenicity of this bacterium is attributed to its vast arrays of virulence factors such as adhesins, production of enzymes and toxins, biofilm formation, and evasion of immunity strategies [3, 4, 5]. Apart from the known virulence factors, this opportunistic pathogen is best known for its formidable reputation due to its antibiotic-resistant phenotype. Methicillin-resistant S. aureus (MRSA) and vancomycin-resistant S. aureus (VRSA) are among the two recognised health threats to the humans. As of now, MRSA is listed as a ‘serious threat’ by Centres for Disease Control and Prevent (CDC) and ‘priority pathogen’ by the World Health Organisation (WHO), while VRSA is listed as ‘concerning threat’ by CDC.

Despite the rapid advancement of modern medicine, S. aureus infections remain highly prevalent in the human populations as transmission of these pathogens can occur through direct contact [6, 7]. Drug-resistant S. aureus particularly MRSA can be defined either as healthcare- or community- associated, based on the ‘48-hour rule’. In the former, MRSA infections develop after 48 hours from hospital admission while the later develops within 48 hours of admission. In this classification system, there are three categories of MRSA infections, namely (i) healthcare-associated, hospital-onset, (ii) healthcare-associated, community onset and (iii) community-associated MRSA infections. Essentially, healthcare-acquired MRSA infections (HA-SA or HA-MRSA) lead to bacteraemia, infective endocarditis, and prosthetic-associated infections while community-acquired MRSA infections (CA-MRSA) often lead to skin and soft tissues infection as well as community-acquired pneumonia in healthy individuals [2, 8, 9]. Compounding to the situation, MRSA has been reported to infect livestock including pigs, poultry, and cattle. Livestock-associated MRSA (LA-MRSA) can be transmitted to individuals handling these infected livestock [10]. In the early days, S. aureus has already been recognised as the main culprit causing hospital-acquired infections (HAIs) such as surgical site infections, bloodstream infections, and pneumonia [11]. The epidemiology of S. aureus shifted in the 2000s with the observation of MRSA infections dominating HAIs. In fact, MRSA strains account for up to 75% of all S. aureus infections in different part of the world [12, 13, 14, 15, 16, 17]. In the US, for instance, MRSA causes approximately 11,000 deaths annually [18].

Antibiotic-resistant S. aureus is known to be associated with higher morbidity and mortality rates as compared to antibiotic-susceptible strains [19, 20, 21]. In the last decade, studies show that MRSA alone causes more death in the US hospitals than of HIV/AIDS, viral hepatitis, and tuberculosis in combination [22, 23]. In addition to health burden, these antibiotic-resistant S. aureus also imposes economic burden in order to eliminate the associated infections [24, 25]. The bacterium develops resistance to nearly all antibiotics introduced to treat infections caused by the bacterium. In 2011, the Expert Panel of the Infectious Diseases Society of America (IDSA) presented an evidence-based guideline for the management of antibiotic-resistant S. aureus infections, including antibiotic choices in both adult and paediatric patients [26]. The key antibiotic choices are described below.

1.1. Vancomycin

Vancomycin, a glycopeptide, was first introduced in the 1960s and has been the most reliable therapeutic agent for MRSA infections, including bacteraemia and endocarditis [27]. This broad spectrum antibiotic is a cell wall synthesis inhibitor. It binds to the c-terminal of D-Ala-D-Ala residues of the peptides of the N-acetyl-glucosamine (NAG) and N-acetyl-muramic acid (NAM) murein subunits, preventing transpeptidases from forming the peptide bridge between peptidoglycan chain, leading to bacterial cell death. However, there is also a group of S. aureus resistant to vancomycin known as VRSA.

1.2. Daptomycin

This antibiotic is a promising alternative to vancomycin for infections caused by MRSA. This cyclic lipopeptide was approved for clinical use in the U.S in 2003 and Europe in 2006. Daptomycin targets only Gram-positive bacteria and is commonly used for complicated skin and skin-structure infections, bacteraemia, and right-sided endocarditis [27]. This antibiotic, however, is not recommended for the pneumonia caused by MRSA. Some studies indicated that daptomycin interacts with pulmonary surfactants present in the lung tissues, leading to the inhibition of daptomycin antibacterial activity [2, 28]. Daptomycin works by targeting the cytoplasmic membrane of bacteria in a calcium ion-dependent manner. In the presence of calcium ions, daptomycin aggregates and forms micellar structures. Daptomycin is then inserted into the membrane and binds strongly to phosphatidylglycerol headgroups leading to depolarisation and permeabilisation of the membrane. This then leads to cytoplasmic content leakage and cell death [29, 30]. The emergence of daptomycin-resistant S. aureus is relatively uncommon. However, increasing records of daptomycin-resistant S. aureus have been reported [31, 32, 33].

1.3. Linezolid

Linezolid, an oxazolidinone, was first approved by the FDA in 2000 for skin bacteraemia and pneumonia-origin S. aureus infections [1]. Linezolid is considered as a standard broad-spectrum intravenous therapies directed towards vancomycin- and teicoplanin-resistant Gram-positive pathogens, including MRSA. Linezolid inhibits protein synthesis by binding to the 23S subunit of the 50S ribosome. Linezolid-resistant S. aureus is relatively uncommon. Resistant strains have been previously reported in staphylococci involving mutations in the 23S rRNA and rRNA methyltransferase. These mutations prevent the binding of linezolid to the ribosome for interfering protein synthesis [34].

1.4. Ceftaroline

Ceftaroline is a fifth generation cephalosporin with a broad-spectrum bactericidal activity against both Gram-positive bacteria including MRSA and some Gram-negative bacteria. This antibiotic is used primarily for the treatment of acute bacterial skin and skin structure infections, and community-acquired bacterial pneumonia caused by S. aureus [35]. Ceftaroline has an enhanced affinity for penicillin binding protein 2a (PBP2a), thus is an ideal antibiotic choice for MRSA infections. This antibiotic is relatively new, and was approved for use in 2010 in the U.S, 2012 in Europe, and 2013 in Australia [36]. However, the emergence of ceftaroline resistance in different parts of the world with a demonstrated decrease of PBP2a binding affinity and heteroresistance, has been documented [37, 38, 39]. The associated mechanisms of resistance involve glutamic acid-to-lysine substitutions in the non-penicillin binding domain and the transpeptidase domain of the PBP2a [39, 40].

There is increasing evidence demonstrating that S. aureus is becoming resistant against all possible antibiotic choices used to treat the infections in the past. Hence, the search for and development of new antibacterials against drug-resistant S. aureus is of pivotal importance. Natural products represent an enormous reservoir of compounds that are diverse in structures and chemical properties. These compounds have been used as antibiotics, such as penicillin and streptomycin. The discovery and use of natural products as antibiotics led to the Golden Age of antibiotics in the 1950s to 1960s. In the past decades, many pharmaceutical companies moved away from natural products programmes partly due to a shift to both high-throughput screening and combinatorial synthesis that focus on small synthetic molecules [41, 42]. However, these approaches are proven to have limited successes [43]. In 2015, the Nobel Prize in Physiology or Medicine was awarded to William C. Campbell and Satoshi Omura, and Youyou Tu for their discovery of new anti-parasitic drugs of natural sources, Avermectin and Artemisinin, respectively. This marks the new milestone and brings optimism for natural product drug discovery. Antimicrobial properties of countless natural products have been tested on S. aureus and an earlier review summarises these research findings collected between 1995 and 2003 [44]. The purpose of this review is to provide an update on natural products that have been shown to demonstrate promising bactericidal effects against drug-resistant S. aureus, published in journal between 2014 and 2017. The resistance mechanisms of drug-resistant S. aureus will be discussed, followed by new anti- S. aureus agents collected from non-microbial natural products, their potential synergism with antibiotics, the molecular targets and mechanisms of these agents, and potential challenges in developing them into clinical trials.

Advertisement

2. Mechanism of S. aureus antimicrobial resistance

Generally, bacteria acquire resistance against antibiotics via different molecular mechanisms, including enzymatic inactivation of antibiotics, alteration of antibiotics target(s) leading to decreased affinity for the antibiotics, removing antibiotics via efflux pumps and changing membrane permeability [45, 46]. S. aureus is known to resist all the clinically approved antibiotics using various resistance mechanisms mentioned above. The detailed resistance mechanisms for important antibiotics, including penicillin, methicillin, and vancomycin are discussed below.

2.1. Penicillin resistance

Penicillin was first isolated from a soil fungus, Penicillium in the 1940s. This antibiotic was once thought to be a miracle drug as it could cure previously fatal infections. However, few years after its introduction, penicillin resistance including penicillin-resistant S. aureus was isolated from hospitals. Penicillin resistance of S. aureus is highly prevalent with up to 86% of clinical S. aureus isolates being resistant to the antibiotic in the US [47]. Meantime, far way in Australia, a similar observation was made as 80% of S. aureus isolates were resistant to penicillin [48]. Penicillin resistance in staphylococci is mediated by the production of enzyme penicillinase or beta-lactamase encoded by the blaZ gene. This enzyme inactivates the antibiotic by hydrolysis of the beta-lactam ring of the antibiotic [49]. Studies show that penicillinase genes can be present on either plasmid of the chromosome of S. aureus [50].

2.2. Methicillin resistance

Methicillin is a penicillinase-resistant beta-lactam. It was first introduced in 1950s and prescribed for S. aureus infection. The first MRSA was documented in 1961 in the UK while the first MRSA in the US was first reported in 1968. Since then, many MRSA clones spread to every corner of the globe. Methicillin resistance is usually encoded by mecA gene that is located in a mobile genetic element of S. aureus, known as the Staphylococcal Chromosomal Cassette mec (SCCmec). mecA is responsible for the synthesis of low-affinity PBP2a which leads to decreased methicillin binding. Methicillin resistance confers broad spectrum of activity generally to the entire beta-lactam class of antibiotics including penicillins and cephalosporins [51]. The origin of SCCmec is thought to be originated from coagulase-negative staphylococcal species as there is no homologues of mecA present in methicillin-susceptible staphylococci. In recent years, a novel mecA homologue, mecC has been identified in both livestock and human in European countries. Similar to mecA, mecC codes for PBP2a with reduced affinity for methicillin and oxacillin, making them MRSA [52, 53].

2.3. Vancomycin resistance

As mentioned earlier, vancomycin is a gold standard antibiotic choice for MRSA infections. However, the emergence of vancomycin-intermediate S. aureus (VISA) (with a MIC value in the range of 3–8 μg/mL) and VRSA (with a MIC value ≥16 μg/mL) result in the failure of vancomycin treatment for MRSA infection. This antibiotic was first released in 1958. However, reduced vancomycin susceptibility in S. aureus was reported in 1997 in Japan [54]. VISA is also spreading to different parts of the world [38]. By comparison, the burden of VISA is relatively higher than VRSA, as the former is commonly associated with persistent infection, treatment failure and poor clinical outcomes. The molecular resistance of VISA is less-defined as compared to VRSA. Typically, VISA features increased cell wall thickness, reduced cross-linking rate, an increase of free D-alanyl-D-alanine residues in the peptidoglycan layers which provides more vancomycin binding, leading to an increased consumption of vancomycin while VISA remains unharmed [27, 55, 56, 57]. It is suggested that VISA involves accumulation of mutations, or rather, adaptation mechanisms in coping with the challenge of vancomycin. VRSA acquires complete resistance to vancomycin by obtaining plasmid(s) from vancomycin-resistant Enterococcus spp. that harbours vanA operon encoded on transposon Tn1546. VRSA maintains the resistance by retaining the original plasmid or by integrating Tn1546 from the enterococcal plasmid into staphylococcal resident plasmid. The vanA operon facilitates the synthesis of D-Ala-D-lactate instead of D-Ala-D-Ala peptidoglycan precursors. In doing so, vancomycin fails to bind hence leading to resistance observed in VRSA [57].

Advertisement

3. Bactericidal properties of natural products against drug-resistant S. aureus

Standard antibiotics treatment against drug-resistant S. aureus has failed in the clinical setting due to several causes as abovementioned. Interestingly, these resistant clinical isolates can be killed by various naturally derived compounds and more promisingly, the antibiotic resistance exhibited by the bacteria can be reversed, and making them susceptible to the antibiotics again. In this section, we discuss the non-microbial natural products that showed bactericidal action against drug-resistant S. aureus and their potential to be used in combination with current antibiotics for its synergistic effects.

3.1. Potent natural products against drug-resistant S. aureus

Numerous natural products have shown potent antibacterial effects against S. aureus. Interestingly, these antibacterial actions are not limited to drug-sensitive wild-type S. aureus, but also extended to antibiotic-resistant S. aureus, including MRSA [58, 59], VISA [60], and VRSA [61]. Some of these natural compounds that showed promising bactericidal effects against drug-resistant S. aureus are summarised in Table 1. Due to the extensive repertoire of natural compounds against drug-resistant S. aureus, Table 1 shows only those that are extracted from Pub-Med indexed publications, from year 2014 to 2017. These research articles reported the minimal inhibitory concentration (MIC) of the natural products against the drug-resistant S. aureus mainly MRSA using Clinical & Laboratory Standards Institute (CLSI) standard broth microdilution assay. As shown in Table 1, the MICs mostly range from micro to milligramme per millilitre. The bactericidal non-microbial natural products are derived from various sources including, plants, insects, animals, and fungi. These natural compounds have been reported to target and act on multiple bacterial targets such as cell wall [62, 63], pyruvate kinase [64], cell division [65], DNA topoisomerase [66], and efflux pump [67, 68]. These pharmacological targets are further discussed in Section 4.

Extract/ compound Test strain MIC References
Curcumin MRSA 217 μg/mL [69]
Quinolone alkaloids MRSA 8–128 μg/mL [70]
Bee venom MRSA 0.085–0.11 μg/mL [71]
Magnolol and honokiol MDR MRSA, MRSA 8–16 ppm [72]
Quercus infectoria gall extracts MRCoNS, MRSA 80–630 μg/mL [73]
Kaempferia pandurata rhizome extracts MRCoNS, MRSA 4–16 ppm [74]
Mulinum spinosum extracts MRSA 500–1000 μg/mL [75]
Isothiocyanates from cruciferous plants MRSA 2.9–110 μg/mL [76]
Lichen MRSA 3.9–500 μg/mL [77]
Marinopyrrole A MRSA 0.19–0.78 μM [78]
6,6′-dihydroxythiobinupharidine MRSA 1–4 μg/mL [66]
Mature carpels of Manglietiastrum sinicum MRSA 0.016–0.14 μM [79]
Pentacyclic triterpenoids MRSA 2–512 μg/mL [80]
Psoralea corylifolia fruit constituents MRSA 8–32 μg/mL [81]
Thai longan honey MRSA 12.5% (v/v) [82]
Rubiaceae, Fabaceae, and Poaceae leaves extract MRSA 5.5–388.4 μg/mL [83]
Cinnamomum extracts MRSA 19.5 μg/mL [84]
Garcinia mangostana pericarp extracts MRSA 17–20 μg/mL [85]
MFM 501 MRSA 15.6–31.3 μg/mL [86]
3′-demethoxy-6-O-demethylisoguaiacin MRSA 12.5 μg/mL [87]
Essential oils from Schinus areira leaves and fruits MRSA 6–30 μg/mL [88]
Spiromastixones A-O MRSA 0.125–8 μg/mL [89]
Hypericum riparium leaves extract MRSA 6.54–18.5 μM [90]
Demethyltexasin MDR MRSA, MRSA 16–128 μg/mL [91]
Oleoresin MRSA 18.2–30 μg/mL [92]
Thymoquinone MRSA 8–16 μg/mL [93]
Phenanthrene fraction MRSA 8–64 μg/mL [94]
Rhamnus californica and Umbellularia californica extracts MRSA 3.3–6 mg/mL [95]
Juncus and Luzula species MRSA 9.75–156 μg/mL [96]
E23 marine compound MRSA 0.5–2 μg/mL [97]
Piper betle extracts MRSA 78–625 μg/mL [98]
Letharia vulpine extracts MRSA 31.25 μg/mL [65]
Verrucosispora MS100047 MRSA 3.125–12.5 μg/mL [99]
Micromonohalimanes B MRSA 40 μg/mL [100]
Pterospartum tridentatum extracts MRSA 78.1 μg/mL [101]
N-3 substituted thiazolidine-2,4-dione derivatives MRSA 6.25–12.5 μg/mL [102]
Sapotaceae extracts MRSA 45–97 μg/mL [103]
Thymus daenensis MRSA 25 mg/mL [104]
Compositae extracts MRSA 31.25 μg/mL [105]
Roemerine MRSA 32–64 μg/mL [106]
Couroupita guianensis extracts MRSA 62.5–156 μg/mL [107]
Cotinus coggygria leaf extracts MRSA 0.313–0.625 mg/mL [108]
Eremophila alternifolia extracts MRSA 10–20 μM [109]
Baicuru MRSA 39 μg/mL [110]
Thymus bovei essential oil MRSA 0.5 mg/mL [111]
Formicamycins MRSA 0.625–80 μM [112]
Rumex aquaticus extracts MRSA 192.3–463 μM [113]
Endophenzine G MRSA 2–128 μg/mL [114]
Greek oregano isolates MRSA 160–640 μg/mL [115]
Macrocyclic bis(bibenzyl)s MRSA 0.5–16 μg/mL [116]
Acylquinic acids MRSA 0.63–1.25 mg/mL [117]
Houttuynia cordata poultice extracts MRSA 0.11–1.76 mg/mL [118]
Dandelion root extracts MRSA 62.5–500 μg/mL [119]
Solanioic acid MRSA 1 μg/mL [120]
Emodin MRSA 32–64 μg/mL [62]
Rhizoma coptidis MRSA 1.2–2.84 mg/mL [121]
Macrocyclic bis(bibenzyl)s MRSA 0.5–32 μg/mL [122]

Table 1.

Antibacterial natural products against drug-resistant Staphylococcus aureus: An update from publication year 2014 to 2017.

MIC—minimal inhibitory concentration; MDR—multidrug-resistant; MRCoNS—methicillin-resistant coagulase negative Staphylococcus aureus; MRSA—methicillin-resistant Staphylococcus aureus.

Compound Combination drug Test strain References
Curcumin Oxacillin, ampicillin, ciprofloxacin, norfloxacin MRSA [127]
Curcumin Gentamicin, amikacin, ciprofloxacin MRSA [128]
Lippia origanoides extracts Neomycin, amikacin MRSA [129]
Grape pornace extracts Oxacillin, ampicillin, nalidixic acid, ciprofloxacin, norfloxacin, levofloxacin, tetracycline, chloramphenicol MRSA [130]
Artocarpin Ampicillin, norfloxacin, tetracycline MRSA [131]
Apigenin Ampicillin, ceftriaxone MRSA [123]
Morin Oxacillin MRSA [124]
Bioactive fraction from Duabanga grandiflora Ampicillin MRSA [132]
Bee venom Ampicillin, penicillin, gentamicin, vancomycin MRSA [71]
Diosmetin Erythromycin MRSA [64]
Brazilin Amikacin, etimicin, gentamicin, streptomycin MRSA [133]
Sophora moorcroftiana genistein Norfloxacin MRSA [134]
Sophora moorcroftiana diosmetin Norfloxacin, streptomycin, ciprofloxacin MRSA [134]
Medihoney Rifampicin MRSA [135]
Magnolol and Honokiol Oxacillin MRSA [63]
9EA-FC-B Ampicillin MRSA [136]
Oxyresveratrol Ciprofloxacin, gentamicin MRSA [137]
Zanthoxylum capense constituents Ciprofloxacin MRSA [138]
Poncirus trifoliate extract Oxacillin MRSA [139]
Juglans regia Oxacillin MRSA [140]
Glabridin Norfloxacin, oxacillin, vancomycin MDR MRSA [141]
Coumarins Chloramphenicol, gentamicin, fosfomycin, levofloxacin, minocycline, piperacillin/ tazobactam, teicoplanin, vancomycin MRSA [142]
Polycarpol Oxacillin, amoxicillin, vancomycin MRSA, VISA [143]
Linoleic and oleic acids Erythromycin MRSA [144]
Salvianolate Fosfomycin, erythromycin, piperacillin-tazobactam, clindamycin MRSA [145]
Phellinus baumii extracts Oxacillin, cefazolin, cefepime, penicillin MRSA [146]
Epigallocatechin gallate Oxacillin, tetracycline, ciprofloxacin MRSA [147]
Clerodane diterpene Norfloxacin MRSA [67]
Carnosic acid Gentamicin MRSA [148]
Ursolic acid Ampicillin, tetracycline MRSA [149]
Anadenanthera colubrine extracts Neomycin, amikacin MRSA [150]
Herbal extracts Oxacillin, gentamicin MRSA [151]
Essential oils Amoxicillin, tetracycline, piperacillin, ofloxacin, oxacillin MRSA [125]
Coumarin derivatives Tetracycline, norfloxacin MRSA [68]

Table 2.

Synergistic anti-Staphylococcus aureus effects of natural products and drugs.

MDR—multidrug-resistant; MRSA—methicillin-resistant Staphylococcus aureus; VISA—vancomycin-intermediate Staphylococcus aureus; VRSA—vancomycin-resistant Staphylococcus aureus.

3.2. Synergism of natural products and antibiotics

While serving as potent antibacterial agents alone, several studies have been carried out to investigate the potential of natural products to be used in combination with current antibiotics. This is particularly important against drug-resistant S. aureus which have shown resistance against several antibiotics. Natural compounds have been shown to reverse the antibiotic resistance. For instances, Akilandeswari and coworkers demonstrated that apigenin (AP) reversed the bacterial resistance of MRSA when used in combination with ampicillin and ceftriaxone [123]. The resulting MIC for ampicillin was shifted from 800 to 107 μg/mL, and the MIC for ceftriaxone was shifted from 58 to 2.6 μg/mL. Similarly, Mun and colleagues also showed that a plant-derived flavonol, morin reversed the oxacillin- and ampicillin-treated MRSA [124]. Essential oils derived from Pituranthos chloranthus, Teucrium ramosissimum and Pistacia lentiscus also reduced the resistance of MRSA to various antibiotics in Penicillins’ group such as amoxicillin, piperacillin, and oxacillin [125].

Cumulative studies highlight the role of natural compounds in decreasing the reliance on antibiotics in bacterial treatment particularly in MRSA’s management, hence preventing the emergence of antibiotic resistance. In addition, production of antibacterial agents from natural products might be more cost-effective than antibiotics production. With advent of modern biotechnology, mass production of these antibacterial products is feasible. More importantly, the manufacturing process allows genetic modifications (e.g. to improve biological activity, solubility, stability, toxicity, production method, production cost and time, etc.) [41, 126]. Some of the challenges and limitation of using natural products as therapeutic modalities are discussed in Section 5.

Advertisement

4. Bacterial targets of S. aureus by natural products

In previous section, we discussed the anti-staphylococcal activities by various natural products alone and in combination with multiple types of antibiotics. As some of the mechanisms of antibiotic resistance have already been studied and reported, such as enzymes inactivation, antibiotics trapping, and efflux pumps, this information enables the anti-staphylococcal molecular targets of the natural products to be elucidated. These particular section summaries the molecular targets of natural products against drug resistant S. aureus such as bacterial cell wall and membrane, cell division protein FtsZ, pyruvate kinase, DNA topoisomerase, efflux pump proteins, and PBP2a. The reported pharmacological targets are depicted in Figure 1.

Figure 1.

Pharmacological targets of drug-resistant Staphylococcus aureus from reported bactericidal natural products.

4.1. Cell wall and membrane

Cell wall of S. aureus is a popular pharmacological target of various antibiotics such as penicillins, cephalosporins, vancomycin, bacitracin, and others [152]. These antibiotics interfere with the cell wall biosynthesis and leading to death of the bacteria. Among all, peptidoglycan is the major cell wall components and has been targeted by various drugs [153]. Other cell wall components including adhesins, teichoic acids, immunodominant antigens, and cell wall enzymes are also being targeted by multiple antibiotics [154]. Similarly, a variety of bactericidal natural compounds also act on bacterial cell wall. For instances, Cao and coworkers showed that a natural compound emodin that targets MRSA could damage the cell wall and compromise the intracellular components. The cellular morphology was altered after the treatment when observed under transmission electron microscopy (TEM) [62]. Kim and colleagues also showed that magnolol targeted cell wall components to exert its pharmacological effect. In a mechanistic study, it has been shown that magnolol inhibited mecI’s pathway [63]. In addition, magnolol also targets various resistant genes, such as mecA, femA, and femB in mRNA form. It has also been shown that Juglans regia (English walnut) targeted the bacterial cell wall and resulted in the anti-staphylococcal effects [140]. While showing synergism in combination with antibiotics, apigenin was shown to compromise the cell membrane followed by subsequent leakage of intracellular constituents. This finding was demonstrated using TEM which showed significant morphological change of bacterial cell wall, shape, and plasma membrane [123].

4.2. Efflux pump

The function of efflux pumps of bacteria is to eliminate metabolites or materials that are potentially toxic and stress-inducing to the cells including antimicrobial compounds [155]. Hence, the bacterial efflux pumps have been known to contribute significantly to antimicrobial resistance by extruding a large number of antibiotics or drugs. They are often known as multidrug resistance (MDR) efflux pumps [155]. For decades, MDR efflux system has served as an excellent antibacterial target. Numerous promising candidates have previously demonstrated their potencies in targeting efflux pumps as the major mechanism to killing the bacteria [156, 157]. For examples, Wang and colleagues showed that genistein killed the MRSA by inhibiting NorA efflux protein when used in combination with drugs [134]. Mechanistic studies have also shown that various bactericidal natural compounds such as coumarin derivatives [68], linoleic and oleic acids [144], clerodane diterpene [67], and Anadenanthera colubrina (Cebil/Vilca) [150] acted on MRSA’s efflux pump or proteins.

4.3. Penicillin-binding protein 2a (PBP2a)

PBP2a is encoded by mecA resistance gene and this gene can be acquired across different species for methicillin resistance [158]. Both PBP2a protein and mecA gene are emerging antimicrobial targets for therapeutics development [159, 160]. Various type of natural products targeting mecA gene or PBP2a have also been reported, these compounds include curcumin [161], tiliroside, pinoresinol, magnatriol B, and momorcharaside B [151], Acalypha wilkesiana (evergreen shrub) extract [136], and Poncirus trifoliata extract [139]. In combination with antibiotics, several natural compounds have also reduced the expression of PBP2a. For instances, Mun and colleagues showed that the combination of morin and oxacillin synergistically killed the MRSA depending on the PBP2a-mediated resistance mechanism [124]. Another study demonstrated that the combination of ampicillin and Duabanga grandiflora extract inhibited the PBP2a protein [132]. Hong and colleagues also showed that β-lactams and Phellinus baumii extracts synergistically killed the MRSA by targeting PBP2a [146].

4.4. Cell division protein FtsZ

FtsZ is a tubulin-like GTPase that recruit cell division proteins for new cell wall formation [162, 163]. Due to its pivotal role in cell division, it has been recognised as an important target for various antibacterial compounds or drugs including natural products. Liu and colleagues successfully developed several phenolic compounds targeting FtsZ of MRSA using a computer-aided simulation. These natural compounds showed potent bactericidal activities against MRSA [72]. It has also been shown that Letharia vulpina (lichen) extract possess antimicrobial activity by damaging cell membrane of MRSA, as well as disrupting cell division processes, possibly targeting FtsZ [65].

4.5. Other targets

Other bacterial proteins that are being targeted by natural products for antimicrobials discovery are pyruvate kinase (PK) and DNA topoisomerase IV. Pyruvate kinase serves as a catalyst to catalyse pyruvate and regulate carbohydrate metabolism [164] whereas DNA topoisomerase IV relaxes supercoiled DNA and performs decatenation events during DNA replication [165]. When used in combination with erythromycin, diosmetin drastically suppressed the MRSA PK activities in a dose-dependent manner. Chan and colleagues also speculated that the inhibition of PK could result in ATP deficiency and efflux pump malfunction [64]. Furthermore, Okamura and group demonstrated that a compound derived from Nuphar japonicum (water-lily) inhibited DNA topoisomerase IV of MRSA, but not DNA gyrase which is also carrying an important role in DNA replication [66].

Advertisement

5. Challenges and limitations

Despite great potentials shown by natural products of botanical origin, there is still a long way for them to be used for clinical application. Majority of these products function as supplements for their nutritional and immune-enhancing values, but none of these non-microbial derived natural products is FDA-approved, nor being used for treating bacterial infections. Several natural products antibacterials of microbial origins have been approved since 2010, including fidaxomicin, ceftaroline, dalbavancin, oritavancin, ceftolozane-tazobactam and ceftazidime-avibactam [166, 167]. Between 1980 and 2014, 59% of the total of 140 the FDA- approved antibacterials are originated from natural products or their derivatives, but none of them is originated from plants [166], despite the increasing evidences suggesting that plants may be promising antibacterials as discussed in this review. A few key challenges and limitations are highlighted and discussed in this section, including (a) design of antibacterial screening; (b) solubility and bioavailability of natural compounds; and (c) research directions towards clinical trials.

5.1. Design of antimicrobial screening

Antibacterial screening generally involves phenotypic screening relying on both Kirby-Bauer disc diffusion or broth micro-dilution methods. These methods are commonly used until today due to the cost-effective and ease of preparation nature [168]. In disc diffusion method, antibacterial activity of an extract or compound is determined based on the presence of inhibitory zone on agar plates seeded with susceptible bacteria while broth micro-dilution method examines the MIC of the antibacterial that inhibits bacterial growth [169]. Very often, these methods are used in the initial antibacterial screening of crude extracts, which may comprise up to hundreds of compounds. This complexity may jeopardise the identification of true antimicrobial effects, leading to false negative results, as some active components may be of low abundance nature [170]. The exclusion of extracts and compounds that have high MIC values following initial screening means giving up on potential novel antibacterials. To overcome this, if crude extract is used, pre-fractionate followed by antibacterial screening to identify the most potent fraction is recommended. These fractions with promising results can be further sub-fractionated until potent compounds are identified. The fractionation technique usually involves the use of HPLC coupled to mass spectrophotometry [171]. By doing this, it reduces the chances of losing potent antibacterial during the screening step

In addition to the use of disc diffusion and broth-dilution methods, various techniques are currently used in the antibacterial studies. One such technique is the time-kill assay (also known as time-kill curve). In this technique, following the broth-dilution method, the bactericidal effects of different concentrations of the antibacterial agents (usually covering the ½ x MIC, 1 x MIC and 2 x of the MIC) at different time points, e.g. 0, 4, 6, 8, 10, 12 and 24 h, are assayed, revealing a time-dependent or a concentration-dependent antibacterial effects of these antibacterials [172]. At the moment, there is a lack of such studies in most of the reviewed articles. As the time-kill assay is able to provide a wealth of information on the dynamic interaction between antibiotics and the microbial strains, specifically S. aureus in this context, the inclusion of time-kill assay will further verify the antibacterial activity observed in natural products.

5.2. Solubility and bioavailability of natural products

One of the main limitations of adopting natural products for clinical applications is its solubility and bioavailability [161, 173]. This is highly related to the chemical properties, in particular aqueous solubility of the natural compounds. For examples, curcumin which is a polyphenolic compound, is known to have poor solubility in water, and the main solvents used are usually DMSO, DMF or ethanol [161]. The water insolubility has significant impact on its antibacterial effect and the reported biological action is further reduced under the physiological conditions [161]. There have been several studies demonstrating the reduced antimicrobial effects of natural compounds in the presence of normal human serum. Marinopyrrole A, which has previously shown potent antibacterial action against MRSA, showed approximately 256-fold higher MIC when tested in the presence of 20% serum [72]. The reduced activity could be due to the non-specific serum protein bindings and protein degradation due to metabolic enzymes and complements that largely affect the bioavailability. It has also been reported that curcumin showed reduced antibacterial activities against S. aureus when tested in the presence of human plasma and whole blood [174]. Similarly, human serum albumin has significantly decreased the bactericidal properties of curcumin [174, 175].

Numerous methods have been developed to overcome the solubility and bioavailability issues. Natural products loaded into nanocarriers such as nanoparticles, microemulsions, micelles, etc. have improved the overall stability and bioavailability [176]. Incorporation of natural compounds such as resveratrol and thymol into liposomes has also increased the solubility and stability for their medicinal uses [173]. Furthermore, development of bioconjugates and nanoformulations also greatly improves the pharmacological action of natural products. This has been extensively reviewed for curcumin [161, 177].

5.3. Clinical trials

In the past decades, research organisations are de-prioritising natural products in their drug discovery programmes because of the costs associated with the development and licensure. For instance, between 1995 and 2001, Glaxo Smith Kline conducted 70 HTS campaigns, each worth approximately USD 1 million to identify only five potential antibacterial leads [178]. This early screening does not guarantee marketing and launching of these potential antibacterials as only approximately 30% of drugs, including natural products used as anti-infectives receive FDA approval [179]. Following initial in vitro testing, clinical (phase I to III) testing is required to ensure the efficacy and safety of new antibacterials on human subjects. The complexity of clinical trial adds another barrier to the development of new antibacterials [180, 181]. On one hand, pharmaceutical companies are faced with multiple regulatory bottlenecks such as increased stringency of trial design, increased demands regarding the design of phase III studies, and increased stringency of safety requirements for pre-licencing and post-licencing procedures of drugs [181]. On the other hand, bacteria are acquiring resistance at fast pace. It complicates clinical trials as these trials cannot be completed without a substantial number of the enrolled patients being infected with new, highly resistant strains. Clinical trials involving rare infectious diseases such as meningitis or endocarditis are most affected as these trials may take years and require multiple centres to complete [180]. Upon completion and success of clinical trials, pharmaceutical companies are required to file for approvals from the relevant agencies such as FDA in the US and European Medicines Agencies in the Europe. The entire process may take up to 15 years for the drug discovery to the launching stage [178]. The lack of interest and investment in antibacterial of natural sources reflects in the identification of only one such antibacterial agent, New Mexico Honey, as a decolonization agent for CA-MRSA abscess in the phase II clinical trial phase (ClinicalTrials.gov identifier number NCT00532324, accessed on the Dec 18, 2017).

In recognition of a lack of novel antibacterials in the clinical pipelines, FDA launched incentives such as Generating Antibiotics Incentives Now (GAIN) Act to foster the research and development of new antibacterial. For instance, granting five additional years of exclusivity to new antibacterials to the pharmaceutical companies, providing incentives for drugs used for treating serious and life-threatening infections, including S. aureus, and reducing new antibacterial drug application time to 6 months [182].

5.4. Future directions

The search for new antibacterial agent in natural products remains an exciting yet challenging task. Evidences show that regulatory agencies are working collaboratively with pharmaceutical companies in improving the development of new antibacterials from natural sources. The combined efforts are the key in shaping the development and marketing of potent antibacterials in the coming years. Scientists working in the field, however, may play a bigger role in the discovery of novel antibacterials by addressing technical shortcomings of the screening of natural products for novel antibacterials.

One such aspect for consideration is to expand the antibacterial screening to include anti-virulence screening such as quorum sensing systems, biofilm formation and pilus adhesins. The investigation of anti-virulence rationalises that because anti-virulence drugs do not kill bacterial cells and thus exerting less selective pressure for resistance. It is believed that the development of resistance is slower compared to bactericidal agents. Anti-virulence would constitute a valuable alternative to bactericidal agents [183, 184]. Anti-virulence of natural product such as anti-quorum sensing of goldenseal (Hydrastis canadensis L.) [185], anti-biofilm of dihydrocelastrol and dihydrocelastryl acetate present in many plants [186] in MRSA have been reported. This area of research is still lacking, in-depth investigation on anti-virulence potentials and solid evidence of slow resistance rate is still required.

Another challenging aspect of natural product not mentioned earlier is low bioavailability of natural products, creating inconsistent results between preclinical and clinical studies [187, 188, 189]. To overcome this challenge, scientists are exploring the incorporation of nanoparticles into a delivery system for natural products in order to increase therapeutic effects of natural products [190]. Preclinical successes of curcumin-nanoparticles in inhibiting in vitro growth of S. aureus [191] and MRSA and enhancing wound healing in in vivo murine wound model [192] have been documented thus far. This emerging field holds promises for natural products in treating bacterial infections. However, drug targeting using nanoparticles remains a challenge, toxicity and safety needs further in-depth evaluations.

Advertisement

6. Conclusions

Non-microbial natural products have shown promising bactericidal activities against drug-resistant S. aureus. The mechanisms of bacterial killings are under investigation and great efforts are being made to evaluate their antibacterial activities in clinical trials. This chapter provides an important update on the anti-staphylococcal activity of natural products against S. aureus and the underlying challenges are highlighted. These issues need to be addressed in order to transform the antibacterial natural products into clinically useful antibiotics in the future.

Advertisement

Acknowledgments

We thank Sunway Internal Research Grant (INT-2018-SHMS-SIHD-01 and INTS-2017-SST-DBS-02) from Sunway University and National Cancer Council Malaysia (MAKNA) Cancer Research Award (CRA) 2016 (EXT-SIDS-SIHD-MAKNA-2017-01) for partly supporting this work.

References

  1. 1. Salgado CD, Farr BM, Calfee DP. Community-acquired methicillin-resistant Staphylococcus aureus: A meta-analysis of prevalence and risk factors. Clinical Infectious Diseases. 2003;36(2):131-139
  2. 2. Tong SYC, Davis JS, Eichenberger E, Holland TL, Fowler VG. Staphylococcus aureus infections: Epidemiology, pathophysiology, clinical manifestations, and management. Clinical Microbiology Reviews. 2015;28(3):603-661
  3. 3. Foster TJ, Geoghegan JA, Ganesh VK, Höök M. Adhesion, invasion and evasion: The many functions of the surface proteins of Staphylococcus aureus. Nature Reviews Microbiology. 2014;12(1):49-62
  4. 4. Otto M. Staphylococcus aureus toxins. Current Opinion in Microbiology. 2014;17:32-37
  5. 5. Vandenesch F, Lina G, Henry T. Staphylococcus aureus hemolysins, bi-component leukocidins, and cytolytic peptides: A redundant arsenal of membrane-damaging virulence factors? Frontiers in Cellular and Infection Microbiology. 2012;2:12
  6. 6. O'Gara JP. Into the storm: Chasing the opportunistic pathogen Staphylococcus aureus from skin colonisation to life threatening infections. Environmental Microbiology. 2017;19(10):3823-3833
  7. 7. Rasigade JP, Dumitrescu O, Lina G. New epidemiology of Staphylococcus aureus infections. Clinical Microbiology and Infection. 2014;20(7):587-588
  8. 8. Naimi TS, LeDell KH, Como-Sabetti K, Borchardt SM, Boxrud DJ, Etienne J, et al. Comparison of community-and health care–associated methicillin-resistant Staphylococcus aureus infection. Journal of the American Medical Association. 2003;290(22):2976-2984
  9. 9. Kreisel K, Roghmann MC, Shardell M, Stine OC, Perencevich E, et al. Assessment of the 48-hour rule for identifying community-associated methicillin-resistant Staphylococcus aureus infection complicated by bacteremia. Infection Control and Hospital Epidemiology. 2010;31(6):657-659
  10. 10. Bosch T, Schouls LM. Livestock-associated MRSA: Innocent or serious health threat? Future Microbiology. 2015;10(4):445-447
  11. 11. Emori TG, Gaynes RP. An overview of nosocomial infections, including the role of the microbiology laboratory. Clinical Microbiology Reviews. 1993;6(4):428-442
  12. 12. Chen CJ, Huang YC. New epidemiology of Staphylococcus aureus infection in Asia. Clinical Microbiology and Infection. 2014;20(7):605-623
  13. 13. Fluit AC, Wielders CLC, Verhoet J, Schmitz FJ. Epidemiology and susceptibility of 3,051 Staphylococcus aureus isolates from 25 university hospitals participating in the European SENTRY study. Journal of Clinical Microbiology. 2001;39(10):3727-3732
  14. 14. Lai CC, Xiao Y, Ahmad N, Veeraragharan B, Thamlikitkul V, et al. High burden of antimicrobial drug resistance in Asia. Journal of Global Antimicrobial Resistance. 2014;2(3):141-147
  15. 15. Mendes RE, Flamm RK, Farrell DJ, Sader HS, Jones KN. Oritavancin in vitro Activity against the most Prevalent Antibiogram Resistance Patterns among Methicillin-Resistant Staphylococcus aureus, Including Multidrug-Resistant Strains from Patients in European Kospitals (2010-2013) in 25th European Congress of Clinical Microbiology and Infectious Diseases. Copenhagen: Denmark; 2015
  16. 16. Schaumburg F, Alabis S, Peters G, Becker K. New epidemiology of Staphylococcus aureus infection in Africa. Clinical Microbiology and Infection. 2014;20(7):589-596
  17. 17. Tokajian S. New epidemiology of Staphylococcus aureus infections in the Middle East. Clinical Microbiology and Infection. 2014;20(7):624-628
  18. 18. CDC. Antibiotic/antimicrobial resistance: biggest threats. 2016 April 14, 2017 [cited 2017 Dec 8, 2017]; Available from: https://www.cdc.gov/drugresistance/biggest_threats.html
  19. 19. Cosgrove SE, Sakoulas G, Perencevinch EN, Schwaber MJ, Karchmer AW, Carmeli Y. Comparison of mortality associated with methicillin-resistant and methicillin-susceptible Staphylococcus aureus bacteremia: A meta-analysis. Clinical Infectious Diseases. 2003;36(1):53-59
  20. 20. Fowler Jr VG, Boucher HW, Corey GR, Abrutyn E, Rupp ME, Levine DP. Daptomycin versus standard therapy for bacteremia and endocarditis caused by Staphylococcus aureus. The New England Journal of Medicine. 2006;355(7):653-665
  21. 21. Hanberger H, Walther S, Leone M, Barie PS, Rello J, Lipman J, et al. Increased mortality associated with meticillin-resistant Staphylococcus aureus (MRSA) infection in the intensive care unit: Results from the EPIC II study. International Journal of Antimicrobial Agents. 2011;38(4):331-335
  22. 22. Boucher HW, Corey GR. Epidemiology of methicillin-resistant Staphylococcus aureus. Clinical Infectious Diseases. 2008;46(Supplement_5):S344-S349
  23. 23. Klevens RM, Edwards JR, Tenover FC, McDonald LC, Haran T, Gaynes R, et al. Changes in the epidemiology of methicillin-resistant Staphylococcus aureus in intensive care units in US hospitals, 1992-2003. Clinical Infectious Diseases. 2006;42(3):389-391
  24. 24. Cameron JK, Paterson DL, Britton PN, Tong SYC, Hall L, Nimmo GR, et al. Co-MRSA Infections in Australia Cost $3.5 B Per Annum. In Australasian Society for Infectious Diseases Annual Scientific Meeting. Blue Mountains: N.S.W, Australia; 2017
  25. 25. Lee BY, Singh A, David MZ, Bartsch SM, Slayton RB, Huang SS, et al. The economic burden of community-associated methicillin-resistant Staphylococcus aureus (CA-MRSA). Clinical Microbiology and Infection. 2013;19(6):528-536
  26. 26. Liu C, Bayer A, Cosgrove SE, Daum RS, Fridkin SK, Gorwitz RS, et al. Clinical practice guidelines by the Infectious Diseases Society of America for the treatment of methicillin-resistant Staphylococcus aureus infections in adults and children. Clinical Infectious Diseases. 2011;52(3):e18-e55
  27. 27. Howden BP, Davies JK, Johnson PDR, Stinear TP, Grayson ML. Reduced vancomycin susceptibility in Staphylococcus aureus, including vancomycin-intermediate and heterogeneous vancomycin-intermediate strains: Resistance mechanisms, laboratory detection, and clinical implications. Clinical Microbiology Reviews. 2010;23(1):99-139
  28. 28. Purrello SG, Garau J, Giamarellos E, Mazzei T, Pea F, Soriano F, et al. Methicillin-resistant Staphylococcus aureus infections: A review of the currently available treatment options. Journal of Global Antimicrobial Resistance. 2016;7:178-186
  29. 29. Bayer AS, Schneider T, Sahl HG. Mechanisms of daptomycin resistance in Staphylococcus aureus: Role of the cell membrane and cell wall. Annals of the New York Academy of Sciences. 2013;1277(1):139-158
  30. 30. Taylor SD, Palmer M. The action mechanism of daptomycin. Bioorganic & Medicinal Chemistry. 2016;24(24):6253-6268
  31. 31. Cafiso V, Bertuccio T, Purrello S, Campanile F, Mammina C, Sartor A, et al. dltA overexpression: A strain-independent keystone of daptomycin resistance in methicillin-resistant Staphylococcus aureus. International Journal of Antimicrobial Agents. 2014;43(1):26-31
  32. 32. Cavalcante FS, Ferreira DC, Chamon RC, da Costa TM, Maia F, Barros EM, et al. Daptomycin and methicillin-resistant Staphylococcus aureus isolated from a catheter-related bloodstream infection: A case report. BMC Research Notes. 2014;7(1):759
  33. 33. Sader HS, Moet GJ, Farrell DJ, Jones RN. Antimicrobial susceptibility of daptomycin and comparator agents tested against methicillin-resistant Staphylococcus aureus and vancomycin-resistant enterococci: Trend analysis of a 6-year period in US medical centers (2005-2010). Diagnostic Microbiology and Infectious Disease. 2011;70(3):412-416
  34. 34. Flamm RK, Mendes RE, Hogan PA, Streit JM, Ross JE, Jones RN. Linezolid surveillance results for the United States (LEADER surveillance program 2014). Antimicrobial Agents and Chemotherapy. 2016;60(4):2273-2280
  35. 35. Cosimi RA, Beik N, Kubiak DW, Johnson JA. Ceftaroline for severe methicillin-resistant Staphylococcus aureus infections: A systematic review. Open Forum Infectious Diseases. 2017;4(2):ofx084
  36. 36. Abbott I, Jenney A, Jeremiah C, Mirčeta M, Kandiah J, Holt D, et al. Reduced in vitro activity of ceftaroline by Etest among clonal complex 239 methicillin-resistant Staphylococcus aureus clinical strains from Australia. Antimicrobial Agents and Chemotherapy. 2015;59(12):7837-7841
  37. 37. Biedenbach DJ, Alm RA, Lahiri SD, Reiszner E, Hoban DJ, Sahm DF, et al. In vitro activity of ceftaroline against Staphylococcus aureus isolated in 2012 from Asia-Pacific countries as part of the AWARE surveillance program. Antimicrobial Agents and Chemotherapy. 2016;60(1):343-347
  38. 38. Kelley WL, Jousselin A, Barras C, Lelong E, Renzoni A. Missense mutations in PBP2a affecting ceftaroline susceptibility detected in epidemic hospital-acquired methicillin-resistant Staphylococcus aureus clonotypes ST228 and ST247 in western Switzerland archived since 1998. Antimicrobial Agents and Chemotherapy. 2015;59(4):1922-1930
  39. 39. Lahiri SD, McLaughlin RE, Whiteaker JD, Ambler JE, Alm RA. Molecular characterization of MRSA isolates bracketing the current EUCAST ceftaroline-susceptible breakpoint for Staphylococcus aureus: The role of PBP2a in the activity of ceftaroline. The Journal of Antimicrobial Chemotherapy. 2015;70(9):2488-2498
  40. 40. Alm RA, McLaughlin RE, Kos VN, Sader HS, Iaconis JP, Lahiri SD. Analysis of Staphylococcus aureus clinical isolates with reduced susceptibility to ceftaroline: An epidemiological and structural perspective. The Journal of Antimicrobial Chemotherapy. 2014;69(8):2065-2075
  41. 41. Brown DG, Lister T, May-Dracka TL. New natural products as new leads for antibacterial drug discovery. Bioorganic & Medicinal Chemistry Letters. 2014;24(2):413-418
  42. 42. Pantosti A, Sanchini A, Monaco M. Mechanisms of antibiotic resistance in Staphylococcus aureus. Future Microbiology. 2007;2(3):323-334
  43. 43. Rossiter SE, Fletcher MH, Wuest WM. Natural products as platforms to overcome antibiotic resistance. Chemical Reviews. 2017;117(19):12415-12474
  44. 44. Gibbons S. Anti-staphylococcal plant natural products. Natural Product Reports. 2004;21(2):263-277
  45. 45. Blair JM, Webber MA, Baylay AJ, Ogbolu DO, Piddock LJ, et al. Molecular mechanisms of antibiotic resistance. Nature Reviews Microbiology. 2015;13(1):42-51
  46. 46. Munita JM, Arias CA. Mechanisms of antibiotic resistance. Microbiology Spectrum. 2016;4(2)
  47. 47. Richter SD, Doern GV, Heilmann K, Miner S, Tendolkar S, Riahi F, Diekema. Detection and prevalence of penicillin-susceptible Staphylococcus aureus in the United States in 2013. Journal of Clinical Microbiology. 2016;54(3):812-814
  48. 48. Coombs GW, Daley DA, Thin-Lee Y, Pearson JC, Robinson JO, Nimmo GR, et al. Australian group on antimicrobial resistance australian Staphylococcus aureus sepsis outcome programme annual report, 2014. Communicable Diseases Intelligence Quarterly Report. 2016;40(2):E244-E254
  49. 49. Lowy FD. Antimicrobial resistance: The example of Staphylococcus aureus. The Journal of Clinical Investigation. 2003;111(9):1265-1273
  50. 50. Livermore DM. Antibiotic resistance in staphylococci. International Journal of Antimicrobial Agents. 2000;16:3-10
  51. 51. Stapleton PD, Taylor PW. Methicillin resistance in Staphylococcus aureus: Mechanisms and modulation. Science Progress. 2002;85(1):57-72
  52. 52. Lindgren AK, Gustafsson E, Petersson A, Melander E. Methicillin-resistant Staphylococcus aureus with mecC: A description of 45 human cases in southern Sweden. European Journal of Clinical Microbiology & Infectious Diseases. 2016;35(6):971-975
  53. 53. Paterson GK, Harrison EM, Holmes MA. The emergence of mecC methicillin-resistant Staphylococcus aureus. Trends in Microbiology. 2014;22(1):42-47
  54. 54. Hiramatsu K, Hanaki H, Ino T, Yabuta K, Oguri T, Tenover F. Methicillin-resistant Staphylococcus aureus clinical strain with reduced vancomycin susceptibility. The Journal of Antimicrobial Chemotherapy. 1997;40(1):135-136
  55. 55. Gardete S, Tomasz A. Mechanisms of vancomycin resistance in Staphylococcus aureus. The Journal of Clinical Investigation. 2014;124(7):2836-2840
  56. 56. Holden MT, Feil EJ, Lindsay JA, Peacock SJ, Day NP, Enright MC, et al. Complete genomes of two clinical Staphylococcus aureus strains: Evidence for the rapid evolution of virulence and drug resistance. Proceedings of the National Academy of Sciences of the United States of America. 2004;101(26):9786-9791
  57. 57. McGuinness WA, Malachowa N, DeLeo FD. Focus: Infectious diseases: Vancomycin resistance in Staphylococcus aureus. The Yale Journal of Biology and Medicine. 2017;90(2):269-281
  58. 58. Kali A. Antibiotics and bioactive natural products in treatment of methicillin resistant Staphylococcus aureus: A brief review. Pharmacognosy Reviews. 2015;9(17):29-34
  59. 59. Fu L, Lu W, Zhou X. Phenolic compounds and in vitro antibacterial and antioxidant activities of three tropic fruits: Persimmon, guava, and sweetsop. Biomed Research International. 2016;2016:4287461
  60. 60. Hiramatsu K, Igarashi M, Morimoto Y, Baba T, Umekita M, Akamatsu Y. Curing bacteria of antibiotic resistance: Reverse antibiotics, a novel class of antibiotics in nature. International Journal of Antimicrobial Agents. 2012;39(6):478-485
  61. 61. Costa EM, Silva S, Veiga M, Vicente S, Tavaria FK, Pintado ME. Investigation of chitosan's antibacterial activity against vancomycin resistant microorganisms and their biofilms. Carbohydrate Polymers. 2017;174:369-376
  62. 62. Cao F, Peng W, Li X, Liu M, Li B, Qin R, et al. Emodin is identified as the active component of ether extracts from Rhizoma Polygoni Cuspidati, for anti-MRSA activity. Canadian Journal of Physiology and Pharmacology. 2015;93(6):485-493
  63. 63. Kim SY, Kim J, Jeong SI, Jahng KY, Yu KY. Antimicrobial effects and resistant regulation of Magnolol and Honokiol on methicillin-resistant Staphylococcus aureus. Biomed Research International. 2015;2015:283630
  64. 64. Chan BC, Ip M, Gong H, Lui SL, See RH, Jolivalt C, et al. Synergistic effects of diosmetin with erythromycin against ABC transporter over-expressed methicillin-resistant Staphylococcus aureus (MRSA) RN4220/pUL5054 and inhibition of MRSA pyruvate kinase. Phytomedicine. 2013;20(7):611-614
  65. 65. Shrestha G, Thompson A, Robison R, St Clair LL. Letharia vulpina, a vulpinic acid containing lichen, targets cell membrane and cell division processes in methicillin-resistant Staphylococcus aureus. Pharmaceutical Biology. 2016;54(3):413-418
  66. 66. Okamura S, Nishiyama E, Yamazaki T, Otsuka N, Taniguchi S, Ogawa W, et al. Action mechanism of 6, 6′-dihydroxythiobinupharidine from Nuphar japonicum, which showed anti-MRSA and anti-VRE activities. Biochimica et Biophysica Acta. 2015;1850(6):1245-1252
  67. 67. Gupta VK, Tiwari N, Gupta P, Verma S, Pal A, Srivastava SK, et al. A clerodane diterpene from Polyalthia longifolia as a modifying agent of the resistance of methicillin resistant Staphylococcus aureus. Phytomedicine. 2016;23(6):654-661
  68. 68. de Araújo RS, Barbosa-Filho JM, Scotti MT, Scotti L, da Cruz RM, Falcão-Silva Vdos S, et al. Modulation of drug resistance in Staphylococcus aureus with coumarin derivatives. Scientifica (Cairo). 2016;2016:6894758
  69. 69. Gunes H, Gulen D, Mutlu R, Gumus A, Tas T, Topkaya AE. Antibacterial effects of curcumin: An in vitro minimum inhibitory concentration study. Toxicology and Industrial Health. 2016;32(2):246-250
  70. 70. Pan X, Bligh SW, Smith E. Quinolone alkaloids from Fructus Euodiae show activity against methicillin-resistant Staphylococcus aureus. Phytotherapy Research. 2014;28(2):305-307
  71. 71. Han SM, Kim JM, Hong IP, Woo SO, Kim SG, Jang HR, et al. Antibacterial activity and antibiotic-enhancing effects of honeybee venom against methicillin-resistant Staphylococcus aureus. Molecules. 2016;21(1):79
  72. 72. Liu T, Pan Y, Lai R. New mechanism of magnolol and honokiol from Magnolia officinalis against Staphylococcus aureus. Natural Product Communications. 2014;9(9):1307-1309
  73. 73. Wan Nor Amilah WA, Masrah M, Hasmah A, Noor Izani NJ. In vitro antibacterial activity of Quercus infectoria gall extracts against multidrug resistant bacteria. Tropical Biomedicine. 2014;31(4):680-688
  74. 74. Sukandar EY, Sunderam N, Fidrianny I. Activity of Kaempferia pandurata (Roxb.) rhizome ethanol extract against MRSA, MRCNS, MSSA, Bacillus subtilis and Salmonella typhi. Pakistan Journal of Biological Sciences. 2014;17(1):49-55
  75. 75. Daniela E, Alejandra C, Pedro R, Claudia M, Lucía A, Carlos T, et al. Antibacterial activity of Mulinum spinosum extracts against slime-producing Staphylococcus aureus and methicillin-resistant Staphylococcus aureus isolated from nasal carriers. The Scientific World Journal. 2014;2014:342143
  76. 76. Dias C, Aires A, Saavedra MJ. Antimicrobial activity of isothiocyanates from cruciferous plants against methicillin-resistant Staphylococcus aureus (MRSA). International Journal of Molecular Sciences. 2014;15(11):19552-19561
  77. 77. Shrestha G, Raphael J, Leavitt SD, St Clair LL. In vitro evaluation of the antibacterial activity of extracts from 34 species of north American lichens. Pharmaceutical Biology. 2014;52(10):1262-1266
  78. 78. Liu Y, Haste NM, Thienphrapa W, Li J, Nizet V, Hensler M, et al. Marinopyrrole derivatives as potential antibiotic agents against methicillin-resistant Staphylococcus aureus (III). Marine Drugs. 2014;12(5):2458-2470
  79. 79. Ding JY, Yuan CM, Cao MM, Liu WW, Yu C, Zhang HY, et al. Antimicrobial constituents of the mature carpels of Manglietiastrum sinicum. Journal of Natural Products. 2014;77(8):1800-1805
  80. 80. Chung PY, Chung LY, Navaratnam P. Potential targets by pentacyclic triterpenoids from Callicarpa farinosa against methicillin-resistant and sensitive Staphylococcus aureus. Fitoterapia. 2014;94:48-54
  81. 81. Cui Y, Taniguchi S, Kuroda T, Hatano T. Constituents of Psoralea corylifolia fruits and their effects on methicillin-resistant Staphylococcus aureus. Molecules. 2015;20(7):12500-12511
  82. 82. Jantakee K, Tragoolpua Y. Activities of different types of Thai honey on pathogenic bacteria causing skin diseases, tyrosinase enzyme and generating free radicals. Biological Research. 2015;48:4
  83. 83. Sharifi-Rad M, Iriti M, Sharifi-Rad M, Gibbons S, Sharifi-Rad J. Anti-methicillin-resistant Staphylococcus aureus (MRSA) activity of Rubiaceae, Fabaceae and Poaceae plants: A search for new sources of useful alternative antibacterials against MRSA infections. Cellular and Molecular Biology. 2016;62(9):39-45
  84. 84. Buru AS, Pichika MR, Neela V, Mohandas K. In vitro antibacterial effects of Cinnamomum extracts on common bacteria found in wound infections with emphasis on methicillin-resistant Staphylococcus aureus. Journal of Ethnopharmacology. 2014;153(3):587-595
  85. 85. Tatiya-Aphiradee N, Chatuphonprasert W, Jarukamjorn K. In vivo antibacterial activity of Garcinia mangostana pericarp extract against methicillin-resistant Staphylococcus aureus in a mouse superficial skin infection model. Pharmaceutical Biology. 2016;54(11):2606-2615
  86. 86. Johari SA, Mohtar M, Mohammad SA, Sahdan R, Shaameri Z, Hamzah AS, et al. In vitro inhibitory and cytotoxic activity of MFM 501, a novel codonopsinine derivative, against methicillin-resistant Staphylococcus aureus clinical isolates. Biomed Research International. 2015;2015:823829
  87. 87. Favela-Hernández JM, Clemente-Soto AF, Balderas-Rentería I, Garza-González E, Camacho-Corona Mdel R. Potential mechanism of action of 3′-demethoxy-6-O-demethyl-isoguaiacin on methicillin resistant Staphylococcus aureus. Molecules. 2015;20(7):12450-12458
  88. 88. Celaya LS, Alabrudzińska MH, Molina AC, Viturro CI, Moreno S. The inhibition of methicillin-resistant Staphylococcus aureus by essential oils isolated from leaves and fruits of Schinus areira depending on their chemical compositions. Acta Biochimica Polonica. 2014;61(1):41-46
  89. 89. Niu S, Liu D, Hu X, Proksch P, Shao Z, Lin W. Spiromastixones A-O, antibacterial chlorodepsidones from a deep-sea-derived Spiromastix sp. fungus. Journal of Natural Products. 2014;77(4):1021-1030
  90. 90. Tala MF, Talontsi FM, Zeng GZ, Wabo HK, Tan NH, Spiteller M, et al. Antimicrobial and cytotoxic constituents from native Cameroonian medicinal plant Hypericum riparium. Fitoterapia. 2015;102:149-155
  91. 91. Hummelova J, Rondevaldova J, Balastikova A, Lapcik O, Kokoska L. The relationship between structure and in vitro antibacterial activity of selected isoflavones and their metabolites with special focus on antistaphylococcal effect of demethyltexasin. Letters in Applied Microbiology. 2015;60(3):242-247
  92. 92. Coté H, Boucher MA, Pichette A, Roger B, Legault J. New antibacterial hydrophobic assay reveals Abies balsamea oleoresin activity against Staphylococcus aureus and MRSA. Journal of Ethnopharmacology. 2016;194:684-689
  93. 93. Hariharan P, Paul-Satyaseela M, Gnanamani A. In vitro profiling of anti methicillin-resistant Staphylococcus aureus activity of thymoquinone against selected type and clinical strains. Letters of Applied Microbiology. 2016;62(3):283-289
  94. 94. Guo JJ, Dai BL, Chen NP, Jin LX, Jiang FS, Ding ZS, et al. The anti-Staphylococcus aureus activity of the phenanthrene fraction from fibrous roots of Bletilla striata. BMC Complementary and Alternative Medicine. 2016;16(1):491
  95. 95. Carranza MG, Sevigny MB, Banerjee D, Fox-Cubley L. Antibacterial activity of native California medicinal plant extracts isolated from Rhamnus californica and Umbellularia californica. Annals of Clinical Microbiology and Antimicrobials. 2015;14:29
  96. 96. Tóth B, Liktor-Busa E, Kúsz N, Szappanos Á, Mándi A, Kurtán T, et al. Phenanthrenes from Juncus inflexus with antimicrobial activity against methicillin-resistant Staphylococcus aureus. Journal of Natural Products. 2016;79(11):2814-2823
  97. 97. Igumnova EM, Mishchenko E, Haug T, Blencke HM, Sollid JUE, Fredheim EGA, et al. Synthesis and antimicrobial activity of small cationic amphipathic aminobenzamide marine natural product mimics and evaluation of relevance against clinical isolates including ESBL-CARBA producing multi-resistant bacteria. Bioorganic & Medicinal Chemistry. 2016;24(22):5884-5894
  98. 98. Valle DL Jr, Cabrera EC, Puzon JJ, Rivera WL. Antimicrobial activities of methanol, ethanol and supercritical CO2 extracts of Philippine Piper betle L. on clinical isolates of gram positive and gram negative bacteria with transferable multiple drug resistance. PLoS One. 2016;11(1):e0146349
  99. 99. Huang P, Xie F, Ren B, Wang Q, Wang J, Wang Q, et al. Anti-MRSA and anti-TB metabolites from marine-derived Verrucosispora sp. MS100047. Applied Microbiology and Biotechnology. 2016;100(17):7437-7447
  100. 100. Zhang Y, Adnani N, Braun DR, et al. Micromonohalimanes A and B: Antibacterial Halimane-type diterpenoids from a marine Micromonospora species. Journal of Natural Products. 2016;79(11):2968-2972
  101. 101. Aires A, Marrinhas E, Carvalho R, Dias C, Saavedra MJ. Phytochemical composition and antibacterial activity of hydroalcoholic extracts of pterospartum tridentatum and mentha pulegium against Staphylococcus aureus isolates. BioMed Research International. 2016;2016:5201879
  102. 102. Desai NC, Satodiya HM, Kotadiya GM, Vaghani HV. Synthesis and antibacterial and cytotoxic activities of new N-3 substituted thiazolidine-2,4-dione derivatives bearing the pyrazole moiety. Archiv der Pharmazie. 2014;347(7):523-532
  103. 103. Kipre BG, Guessennd NK, Koné MW, Gbonon V, Coulibaly JK, Dosso M. Antibacterial activity of the stem bark of Tieghemella Heckelii Pierre ex. A Chev against methicillin-resistant Staphylococcus aureus. BMC Complementary and Alternative Medicine. 2017;17(1):170
  104. 104. Saidi M, Sadeghifard N, Kazemian H, Sekawi Z, Badakhsh B, Friadian S, et al. Ex vivo evaluation of Thymus daenensis as an antioxidant and antibacterial medicinal herb. Drug Research. 2016;66(12):657-659
  105. 105. Njeru SN, Obonyo MA, Nyambati SO, Ngari SM. Antimicrobial and cytotoxicity properties of the crude extracts and fractions of Premna resinosa (Hochst.) Schauer (Compositae): Kenyan traditional medicinal plant. BMC Complementary and Alternative Medicine. 2015;15:295
  106. 106. Yin S, Rao G, Wang J, Luo L, He G, Wang C, et al. Roemerine improves the survival rate of septicemic BALB/c mice by increasing the cell membrane permeability of Staphylococcus aureus. PLoS One. 2015;10(11):e0143863
  107. 107. Costa DCM, Azevedo MMB, Silva DOE, Romanos MTV, Souto-Padrón TCBS, Alviano CS, et al. In vitro anti-MRSA activity of Couroupita guianensis extract and its component Tryptanthrin. Natural Product Research. 2017;31(17):2077-2080
  108. 108. Rendeková K, Fialová S, Jánošová L, Mučaji P, Slobodníková L. The activity of Cotinus coggygria Scop. Leaves on Staphylococcus aureus strains in planktonic and biofilm growth forms. Molecules. 2015;21(1):E50
  109. 109. Biva IJ, Ndi CP, Griesser HJ, Semple SJ. Antibacterial constituents of Eremophila alternifolia: An Australian aboriginal traditional medicinal plant. Journal of Ethnopharmacology. 2016;182:1-9
  110. 110. Blainski A, Gionco B, Oliveira AG, Andrade G, Scarminio IS, Silva DB, et al. Antibacterial activity of Limonium brasiliense (Baicuru) against multidrug-resistant bacteria using a statistical mixture design. Journal of Ethnopharmacology. 2017;198:313-323
  111. 111. Jaradat N, Adwan L, K'aibni S, Shraim N, Zaid AN. Chemical composition, anthelmintic, antibacterial and antioxidant effects of Thymus bovei essential oil. BMC Complementary and Alternative Medicine. 2016;16(1):418
  112. 112. Qin Z, Munnoch JT, Devine R, Holmes NA, Seipke RF, Wilkinson KA, et al. Formicamycins, antibacterial polyketides produced by Streptomyces formicae isolated from African Tetraponera plant-ants. Chemical Science. 2017;8(4):3218-3227
  113. 113. Orbán-Gyapai O, Liktor-Busa E, Kúsz N, Stefkó D, Urbán E, Hohmann J, et al. Antibacterial screening of Rumex species native to the Carpathian Basin and bioactivity-guided isolation of compounds from Rumex aquaticus. Fitoterapia. 2017;118:101-106
  114. 114. Udumula V, Endres JL, Harper CN, Jaramillo L, Zhong HA, Bayles KW, et al. Simple synthesis of endophenazine G and other phenazines and their evaluation as anti-methicillin-resistant Staphylococcus aureus agents. European Journal of Medicinal Chemistry. 2017;125:710-721
  115. 115. Stamenic M, Vulic J, Djilas S, Misic D, Tadic V, Petrovic S, et al. Free-radical scavenging activity and antibacterial impact of Greek oregano isolates obtained by SFE. Food Chemistry. 2014;165:307-315
  116. 116. Fujii K, Morita D, Onoda K, Kuroda T, Miyachi H. Minimum structural requirements for cell membrane leakage-mediated anti-MRSA activity of macrocyclic bis(bibenzyl)s. Bioorganic & Medicinal Chemistry Letters. 2016;26(9):2324-2327
  117. 117. Zheleva-Dimitrova D, Gevrenova R, Zaharieva MM, Najdenski H, Ruseva S, Lozanov V, et al. HPLC-UV and LC-MS analyses of acylquinic acids in Geigeria alata (DC) Oliv. & Hiern. and their contribution to antioxidant and antimicrobial capacity. Phytochemical Analysis. 2017;28(3):176-184
  118. 118. Sekita Y, Murakami K, Yumoto H, Mizuguchi H, Amoh T, Ogino S, et al. Anti-bacterial and anti-inflammatory effects of ethanol extract from Houttuynia cordata poultice. Bioscience, Biotechnology, and Biochemistry. 2016;80(6):1205-1213
  119. 119. Kenny O, Brunton NP, Walsh D, Hewage CM, McLoughlin P, Smyth TJ. Characterisation of antimicrobial extracts from dandelion root (Taraxacum officinale) using LC-SPE-NMR. Phytotherapy Research. 2015;29(4):526-532
  120. 120. Ratnaweera PB, Williams DE, Patrick BO, de Silva ED, Andersen RJ. Solanioic acid, an antibacterial degraded steroid produced in culture by the fungus Rhizoctonia solani isolated from tubers of the medicinal plant Cyperus rotundus. Organic Letters. 2015;17(9):2074-2077
  121. 121. Luo JY, Yan D, Yang MH. Study of the anti-MRSA activity of Rhizoma coptidis by chemical fingerprinting and broth microdilution methods. Chinese Journal of Natural Medicines. 2014;12(5):393-400
  122. 122. Onoda K, Sawada H, Morita D, Fujii K, Tokiwa H, Kuroda T, et al. Anti-MRSA activity of isoplagiochin-type macrocyclic bis(bibenzyl)s is mediated through cell membrane damage. Bioorganic & Medicinal Chemistry. 2015;23(13):3309-3316
  123. 123. Akilandeswari K, Ruckmani K. Synergistic antibacterial effect of apigenin with β-lactam antibiotics and modulation of bacterial resistance by a possible membrane effect against methicillin resistant Staphylococcus aureus. Cellular and Molecular Biology (Noisy-le-Grand, France). 2016;62(14):74-82
  124. 124. Mun SH, Lee YS, Han SH, Lee SW, Cha SW, Kim SB, et al. In vitro potential effect of morin in the combination with β-lactam antibiotics against methicillin-resistant Staphylococcus aureus. Foodborne Pathogens and Disease. 2015;12(6):545-550
  125. 125. Lahmar A, Bedoui A, Mokdad-Bzeouich I, Dhaouifi Z, Kalboussi Z, Cheraif I, et al. Reversal of resistance in bacteria underlies synergistic effect of essential oils with conventional antibiotics. Microbial Pathogenesis. 2017;106:50-59
  126. 126. Moloney MG. Natural products as a source for novel antibiotics. Trends in Pharmacological Sciences. 2016;37(8):689-701
  127. 127. Mun SH, Joung DK, Kim YS, Kang OH, Kim SB, Seo YS, et al. Synergistic antibacterial effect of curcumin against methicillin-resistant Staphylococcus aureus. Phytomedicine. 2013;20(8-9):714-718
  128. 128. Teow SY, Ali SA. Synergistic antibacterial activity of curcumin with antibiotics against Staphylococcus aureus. Pakistan Journal of Pharmaceutical Sciences. 2015;28(6):2109-2114
  129. 129. Medeiros Barreto H, Cerqueira Fontinele F, Pereira de Oliveira A, Arcanjo DD, Cavalcanti Dos Santos BH, de Abreu AP, et al. Phytochemical prospection and modulation of antibiotic activity in vitro by Lippia origanoides H.B.K. in methicillin resistant Staphylococcus aureus. Biomed Research International. 2014;2014:305610
  130. 130. 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(2):e0172273
  131. 131. Septama AW, Panichayupakaranant P. Synergistic effect of artocarpin on antibacterial activity of some antibiotics against methicillin-resistant Staphylococcus aureus, Pseudomonas aeruginosa, and Escherichia coli. Pharmaceutical Biology. 2016;54(4):686-691
  132. 132. Santiago C, Pang EL, Lim KH, Loh HS, Ting KN. Inhibition of penicillin-binding protein 2a (PBP2a) in methicillin resistant Staphylococcus aureus (MRSA) by combination of ampicillin and a bioactive fraction from Duabanga grandiflora. BMC Complementary and Alternative Medicine. 2015;15:178
  133. 133. Zuo GY, Han ZQ, Hao XY, Han J, Li ZS, Wang GC. Synergy of aminoglycoside antibiotics by 3-Benzylchroman derivatives from the Chinese drug Caesalpinia sappan against clinical methicillin-resistant Staphylococcus aureus (MRSA). Phytomedicine. 2014;21(7):936-941
  134. 134. Wang SY, Sun ZL, Liu T, Gibbons S, Zhang WJ, Qing M. Flavonoids from Sophora moorcroftiana and their synergistic antibacterial effects on MRSA. Phytotherapy Research. 2014;28(7):1071-1076
  135. 135. Müller P, Alber DG, Turnbull L, et al. Synergism between Medihoney and rifampicin against methicillin-resistant Staphylococcus aureus (MRSA). PLoS One. 2013;8(2):e57679
  136. 136. Santiago C, Pang EL, Lim KH, Loh HS, Ting KN. Reversal of ampicillin resistance in MRSA via inhibition of penicillin-binding protein 2a by Acalypha wilkesiana. Biomed Research International. 2014;2014:965348
  137. 137. Joung DK, Choi SH, Kang OH, Kim SB, Mun SH, Seo YS, et al. Synergistic effects of oxyresveratrol in conjunction with antibiotics against methicillin-resistant Staphylococcus aureus. Molecular Medicine Reports. 2015;12(1):663-667
  138. 138. Cabral V, Luo X, Junqueira E, Costa SS, Mulhovo S, A D, et al. Enhancing activity of antibiotics against Staphylococcus aureus: Zanthoxylum capense constituents and derivatives. Phytomedicine. 2015;22(4):469-476
  139. 139. Eom SH, Jung YJ, Lee DS, Yim MJ, Kim HS, Lee SH, et al. Studies on antimicrobial activity of Poncirus trifoliata ethyl extract fraction against methicillin-resistant Staphylococcus aureus and to elucidate its antibacterial mechanism. Journal of Environmental Biology. 2016;37(1):129-134
  140. 140. Farooqui A, Khan A, Borghetto I, Kazmi SU, Rubino S, Paglietti B. Synergistic antimicrobial activity of Camellia sinensis and Juglans regia against multidrug-resistant bacteria. PLoS One. 2015;10(2):e0118431
  141. 141. Singh V, Pal A, Darokar MP. A polyphenolic flavonoid glabridin: Oxidative stress response in multidrug-resistant Staphylococcus aureus. Free Radical Biology & Medicine. 2015;87:48-57
  142. 142. Zuo GY, Wang CJ, Han J, Li YQ, Wang GC. Synergism of coumarins from the Chinese drug Zanthoxylum nitidum with antibacterial agents against methicillin-resistant Staphylococcus aureus (MRSA). Phytomedicine. 2016;23(14):1814-1820
  143. 143. Pereira F, Madureira AM, Sancha S, Mulhovo S, Luo X, Duarte A, et al. Cleistochlamys kirkii chemical constituents: Antibacterialactivity and synergistic effects against resistant Staphylococcus aureus strains. Journal of Ethnopharmacology. 2016;178:180-187
  144. 144. Chan BC, Han XQ, Lui SL, Wong CW, Wang TB, Cheung DW, et al. Combating against methicillin-resistant Staphylococcus aureus - two fatty acids from Purslane (Portulaca oleracea L.) exhibit synergistic effects with erythromycin. The Journal of Pharmacy and Pharmacology. 2015;67(1):107-116
  145. 145. Liu QQ, Han J, Zuo GY, Wang GC, Tang HS. Potentiation activity of multiple antibacterial agents by Salvianolate from the Chinese medicine Danshen against methicillin-resistant Staphylococcus aureus (MRSA). Journal of Pharmacological Sciences. 2016;131(1):13-17
  146. 146. Hong SB, Rhee MH, Yun BS, Lim YH, Song HG, Shin KS. Synergistic anti-bacterial effects of Phellinus baumii ethyl acetate extracts and β-lactam antimicrobial agents against methicillin-resistant Staphylococcus aureus. Annals of Laboratory Medicine. 2016;36(2):111-116
  147. 147. Navrátilová A, Nešuta O, Vančatová I, Čížek A, Varela-M RE, López-Abán J, et al. C-Geranylated flavonoids from Paulownia tomentosa fruits with antimicrobial potential and synergistic activity with antibiotics. Pharmaceutical Biology. 2016;54(8):1398-1407
  148. 148. Vázquez NM, Fiorilli G, Cáceres Guido PA, Moreno S. Carnosic acid acts synergistically with gentamicin in killing methicillin-resistant Staphylococcus aureus clinical isolates. Phytomedicine. 2016;23(12):1337-1343
  149. 149. Wang CM, Chen HT, Wu ZY, Jhan YL, Shyu CL, Chou CH. Antibacterial and synergistic activity of pentacyclic triterpenoids isolated from Alstonia scholaris. Molecules. 2016;21(2):139
  150. 150. Barreto HM, Coelho KM, Ferreira JH, Dos Santos BH, de Abreu AP, Coutinho HD, et al. Enhancement of the antibiotic activity of aminoglycosides by extracts from Anadenanthera colubrine (Vell.) Brenan var. cebil against multi-drug resistant bacteria. Natural Product Research. 2016;30(11):1289-1292
  151. 151. Kuok CF, Hoi SO, Hoi CF, Chan CH, Fong IH, Ngok CK, et al. Synergistic antibacterial effects of herbal extracts and antibiotics on methicillin-resistant Staphylococcus aureus: A computational and experimental study. Experimental Biology and Medicine. 2017;242(7):731-743
  152. 152. Romaniuk JAH, Cegelski L. Bacterial cell wall composition and the influence of antibiotics by cell-wall and whole-cell NMR. Philosophical Transactions of the Royal Society, B: Biological Sciences. 2015;370(1679):20150024
  153. 153. Vollmer W, Blanot D, de Pedro MA. Peptidoglycan structure and architecture. FEMS Microbiology Reviews. 2008;32(2):149-167
  154. 154. Ohlsen K, Lorenz U. Novel targets for antibiotics in Staphylococcus aureus. Future Microbiology. 2007;2(6):655-666
  155. 155. Blanco P, Hernando-Amado S, Reales-Calderon JA, Corona F, Lira F, Alcalde-Rico M, et al. Bacterial multidrug efflux pumps: Much more than antibiotic resistance determinants. Microorganisms. 2016;4(1):14
  156. 156. Costa SS, Viveiros M, Amaral L, Couto I. Multidrug efflux pumps in Staphylococcus aureus: An update. The Open Microbiology Journal. 2013;7:59-71
  157. 157. Sun J, Deng Z, Yan A. Bacterial multidrug efflux pumps: Mechanisms, physiology and pharmacological exploitations. Biochemical and Biophysical Research Communications. 2014;453(2):254-267
  158. 158. Fuda C, Suvorov M, Vakulenko SB, Mobashery S. The basis for resistance to beta-lactam antibiotics by penicillin-binding protein 2a of methicillin-resistant Staphylococcus aureus. The Journal of Biological Chemistry. 2004;279(39):40802-40806
  159. 159. Klitgaard JK, Skov MN, Kallipolitis BH, Kolmos HJ. Reversal of methicillin resistance in Staphylococcus aureus by thioridazine. The Journal of Antimicrobial Chemotherapy. 2008;62(6):1215-1221
  160. 160. Meng J, He G, Wang H, Jia M, Ma X, Da F, et al. Reversion of antibiotic resistance by inhibiting mecA in clinical methicillin-resistant staphylococci by antisense phosphorothioate oligonucleotide. The Journal of Antibiotics. 2015;68(3):158-164
  161. 161. Teow SY, Liew K, Ali SA, Khoo ASB, Peh SC. Antibacterial action of curcumin against Staphylococcus aureus: A brief review. Journal of Tropical Medicine. 2016;2016:2853045
  162. 162. Matsui T, Yamane J, Mogi N, Yamaguchi H, Takemoto H, Yao M, et al. Structural reorganization of the bacterial cell-division protein FtsZ from Staphylococcus aureus. Acta Crystallographica, Section D: Biological Crystallography. 2012;68(Pt 9):1175-1188
  163. 163. Artola M, Ruíz-Avila LB, Ramírez-Aportela E, Martínez RF, Araujo-Bazán L, Vázquez-Villa H, et al. The structural assembly switch of cell division protein FtsZ probed with fluorescent allosteric inhibitors. Chemical Science. 2017;8(2):1525-1534
  164. 164. Zoraghi R, Worrall L, See RH, Strangman W, Popplewell WL, Gong H, et al. Methicillin-resistant Staphylococcus aureus (MRSA) pyruvate kinase as a target for bis-indole alkaloids with antibacterial activities. The Journal of Biological Chemistry. 2011;286(52):44716-44725
  165. 165. Drlica K, Hiasa H, Kerns R, Malik M, Mustaev A, Zhao X. Quinolones: Action and resistance updated. Current Topics in Medicinal Chemistry. 2009;9(11):981-998
  166. 166. Newman DJ, Cragg GM. Natural products as sources of new drugs from 1981 to 2014. Journal of Natural Products. 2016;79(3):629-661
  167. 167. Deak D, Outterson K, Powers JH, Kesselheim AD. Progress in the fight against multidrug-resistant bacteria? A review of US Food and Drug Administration–approved antibiotics, 2010-2015. Annals of Internal Medicine. 2016;165(5):363-372
  168. 168. Tan JBL, Lim YY. Critical analysis of current methods for assessing the in vitro antioxidant and antibacterial activity of plant extracts. Food Chemistry. 2015;172:814-822
  169. 169. Cos P, Vlietinck AJ, Berghe DV, Maes L. Anti-infective potential of natural products: How to develop a stronger in vitro ‘proof-of-concept’. Journal of Ethnopharmacology. 2006;106(3):290-302
  170. 170. Fallarero A, Hanski L, Vuorela P. How to translate a bioassay into a screening assay for natural products: General considerations and implementation of antimicrobial screens. Planta Medica. 2014;80(14):1182-1199
  171. 171. Spörri SA, Jan P, Cognard E, Ortelli D, Edder P. Comprehensive screening of veterinary drugs in honey by ultra-high-performance liquid chromatography coupled to mass spectrometry. Food Additives & Contaminants Part A. 2014;31(5):806-816
  172. 172. Balouiri M, Sadiki M, Ibnsouda SK. Methods for in vitro evaluating antimicrobial activity: A review. Journal of Pharmaceutical Analysis. 2016;6(2):71-79
  173. 173. Coimbra M, Isacchi B, van Bloois L, Torano JS, Ket A, Wu X, et al. Improving solubility and chemical stability of natural compounds for medicinal use by incorporation into liposomes. International Journal of Pharmaceutics. 2011;416(2):433-442
  174. 174. Teow SY, Ali SA. Altered antibacterial activity of Curcumin in the presence of serum albumin, plasma and whole blood. Pakistan Journal of Pharmaceutical Sciences. 2017;30(2):449-457
  175. 175. Teow SY, Ali SA. Impact of bovine and human serum albumin on Curcumin in vitro activity against Staphylococcus aureus. Pakistan Journal of Pharmaceutical Sciences. 2017;30(3):891-895
  176. 176. Bilia AR. Natural products loaded in nanocarriers: An opportunity to increase stability, oral bioavailability and bioefficacy. Journal of Nanomedicine & Nanotechnology. 2016;5(Suppl):7
  177. 177. Rahimi HR, Nedaeinia R, Sepehri Shamloo A, Nikdoust S, Kazemi Oskuee R. Novel delivery system for natural products: Nano-curcumin formulations. Avicenna Journal of Phytomedicine. 2016;6(4):383-398
  178. 178. Payne DJ, Gwynn MN, Holmes DJ, Pompliano DL. Drugs for bad bugs: Confronting the challenges of antibacterial discovery. Nature Reviews Drug Discovery. 2007;6(1):29-40
  179. 179. Patridge E, Gareiss P, Kinch MS, Hoyer D. An analysis of FDA-approved drugs: Natural products and their derivatives. Drug Discovery Today. 2016;21(2):204-207
  180. 180. Charles PG, Grayson ML. The dearth of new antibiotic development: Why we should be worried and what we can do about it. The Medical Journal of Australia. 2014;181:549-553
  181. 181. Gupta SK, Nayak RP. Dry antibiotic pipeline: Regulatory bottlenecks and regulatory reforms. The Journal of Pharmacy and Pharmacology. 2014;5(1):4-7
  182. 182. Food and Drug Administration (FDA). Safety and Innovation Act Antibiotic Incentives. Created by IDSA. 2012. Sep 7, [Last accessed on 2017 Dec 18]. Downloaded from http://www.idsociety.org/uploadedFiles/IDSA/Policy_and_Advocacy/Current_Topics_and_Issues/Antimicrobial_Resistance/10×20/Letters/To_Congress/IDSA%20Summary%20of%20Antibiotic%20Incentives%20in%20FDASIA.pdf
  183. 183. Baron C. Antivirulence drugs to target bacterial secretion systems. Current Opinion in Microbiology. 2010;13(1):100-105
  184. 184. Bhardwaj KA, Vinothkumar K, Rajpara N. Bacterial quorum sensing inhibitors: Attractive alternatives for control of infectious pathogens showing multiple drug resistance. Recent Patents on Anti-Infective Drug Discovery. 2013;8(1):68-83
  185. 185. Cech NB, Junio HA, Ackermann LW, Kavanaugh JS, Horswill AR. Quorum quenching and antimicrobial activity of goldenseal (Hydrastis canadensis) against methicillin-resistant Staphylococcus aureus (MRSA). Planta Medica. 2012;78(14):1556-1561
  186. 186. Woo SG, Lee SM, Lee SY, Lim KH, Ha EJ, Kim SH, Eom YB. The effectiveness of anti-biofilm and anti-virulence properties of dihydrocelastrol and dihydrocelastryl diacetate in fighting against methicillin-resistant Staphylococcus aureus. Archives of Microbiology. 2017;199(8):1-13
  187. 187. Bonifácio BV, da Silva PB, dos Santos Ramos MA, KMS N, Bauab TM, Chorilli M. Nanotechnology-based drug delivery systems and herbal medicines: A review. International Journal of Nanomedicine. 2014;9(1):1-15
  188. 188. Manach C, Williamson G, Morand C, Scalbert A, Rémésy C. Bioavailability and bioefficacy of polyphenols in humans. I. Review of 97 bioavailability studies. The American Journal of Clinical Nutrition. 2005;81(1):230S-242S
  189. 189. Thilakarathna SH, Rupasinghe H. Flavonoid bioavailability and attempts for bioavailability enhancement. Nutrients. 2013;5(9):3367-3387
  190. 190. Watkins R, Wu L, Zhang C, Davis RM, Xu B. Natural product-based nanomedicine: Recent advances and issues. International Journal of Nanomedicine. 2015;10:6055-6074
  191. 191. Basniwal R, Buttar HS, Jain V, Jain N. Curcumin nanoparticles: Preparation, characterization, and antimicrobial study. Journal of Agricultural and Food Chemistry. 2011;59(5):2056-2061
  192. 192. Krausz AE, Adler BL, Cabral V, Navati M, Doerner J, Charafeddine RA, et al. Curcumin-encapsulated nanoparticles as innovative antimicrobial and wound healing agent. Nanomedicine: Nanotechnology, Biology and Medicine. 2015;11(1):195-206

Written By

Jactty Chew, Suat-Cheng Peh and Teow Sin Yeang

Submitted: 10 October 2017 Reviewed: 29 January 2018 Published: 06 April 2018