Abstract
Antibiotic resistance has become a serious global concern, and the discovery of antimicrobial herbal constituents may provide valuable solutions to overcome the problem. In this study, the effects of therapies combining antibiotics and four medicinal herbs on methicillin-resistant Staphylococcus aureus (MRSA) were investigated. Specifically, the synergistic effects of Magnolia officinalis, Verbena officinalis, Momordica charantia, and Daphne genkwa in combination with oxacillin or gentamicin against methicillin-resistant (ATCC43300) and methicillin-susceptible (ATCC25923) S. aureus were examined. In vitro susceptibility and synergistic testing were performed to measure the minimum inhibitory concentration and fractional inhibitory concentration (FIC) index of the antibiotics and medicinal herbs against MRSA and methicillin-susceptible S. aureus. To identify the active constituents in producing these synergistic effects, in silico molecular docking was used to investigate the binding affinities of 139 constituents of the four herbs to the two common MRSA inhibitory targets, penicillin binding proteins 2a (PBP2a) and 4 (PBP4). The physicochemical and absorption, distribution, metabolism, and excretion properties and drug safety profiles of these compounds were also analyzed. D. genkwa extract potentiated the antibacterial effects of oxacillin against MRSA, as indicated by an FIC index value of 0.375. M. officinalis and V. officinalis produced partial synergistic effects when combined with oxacillin, whereas M. charantia was found to have no beneficial effects in inhibiting MRSA. Overall, tiliroside, pinoresinol, magnatriol B, and momorcharaside B were predicted to be PBP2a or PBP4 inhibitors with good drug-like properties. This study identifies compounds that deserve further investigation with the aim of developing therapeutic agents to modulate the effect of antibiotics on MRSA.
Impact statement
Antibiotic resistant is a well-known threat to global health and methicillin-resistant Staphylococcus aureus is one of the most significant ones. These resistant bacteria kill thousands of people every year and therefore a new effective antimicrobial treatment is necessary. This study identified the herbs and their associated bioactive ingredients that can potential the effects of current antibiotics. These herbs have long history of human usage in China and have well-defined monograph in the Chinese Pharmacopeia. These indicate their relatively high clinical safety and may have a quicker drug development process than that of a new novel antibiotic. Based on the results of this study, the authors will perform further in vitro and animal studies, aiming to accumulate significant data for the application of clinical trial.
Keywords: Antibiotic resistance, methicillin-resistant Staphylococcus aureus, synergistic effects, penicillin binding proteins, traditional Chinese medicines, molecular docking
Introduction
Methicillin-resistant Staphylococcus aureus (MRSA) is one of the most common causes of multidrug resistance infections with significant morbidity and mortality.1 In 2011, 80,461 invasive MRSA infections were recorded in the US,2 causing substantial economic and logistical problems.3 The rates of MRSA infections have been stabilized in certain developed countries, but those in many developing regions continue to increase rapidly.4 More importantly, MRSA infections previously occurred exclusively in hospitals, but in the past decade, the number of community-acquired infections has significantly increased.4 Presently, most of the MRSA infections that occur in communities affect the skin, whereas life-threatening infections such as bacteremia, endocarditis, and pneumonia still occur in medical facilities.5 No one knows whether these less severe community-acquired MRSA infections will ultimately prove as life threatening as those occurring in hospitals, but it is safe to assume that hospital infection prevention is no longer the only consideration in reducing MRSA rates, and that more effective treatments are urgently needed.
Over the past few decades, several antibiotics, including vancomycin and daptomycin, have been considered drugs of choice for treating and preventing severe MRSA infections.6 However, their effectiveness is declining.5 Much evidence has shown that the increased resistance of vancomycin has led to poor therapeutic outcomes and increased patient mortality.7 More importantly, another strong resistance strain of S. aureus, called heterogeneous vancomycin-resistant S. aureus (hVRSA) has been detected globally. hVRSA is closely related to MRSA and clinical study has demonstrated that hVRSA phenotype was detected in 50% of clinical MRSA in certain countries.8 In addition, vancomycin dosing that achieves an appropriate trough serum level (15–20 mg/L) for serious infection is difficult and has become associated with significant increased risk of toxicity, mainly nephrotoxicity.9 Daptomycin is generally considered to be clinically effective, with less risk of nephrotoxicity than vancomycin in treating MRSA-based endocarditis and bacteremia infections.10 However, an increasing number of cases of daptomycin treatment failure due to resistance have been reported,11,12 and the typical minimum inhibitory concentration (MIC) has been raised from 0.5 to 4–8 mg/L in the past few decades.13 Thus, scientists are always seeking new ways to counter the effects of MRSA antibiotic resistance.
The most common ways to overcome antibiotic resistance are to develop new drugs or via combined treatment therapy.14 The latter method is generally considered to be less complicated because much of the necessary scientific data may already exist, especially when the drugs being combined are already marketed for human use. Examples of recent studies include the combination of clarithromycin and daptomycin15 and rifampicin and daptomycin.16 Both studies revealed potentiated antimicrobial efficacy against different strains of S. aureus. However, combining antibiotics may also make their adverse effects more pronounced.17
Apart from the combined use of antibiotics, the synergistic effects of combining phytopharmaceuticals and antibiotics have been demonstrated in various studies,18,19 particularly in relation to traditional Chinese medicines (TCMs),20 which has flourished for thousands of years in clinical practice. Many TCMs have been documented as effective in treating various infectious diseases. Thus, we performed in vitro studies of four traditional Chinese medicinal plants to investigate their anti-MRSA activity – alone and in combination with oxacillin or gentamicin. The four plants studied – Daphne genkwa, Magnolia officinalis, Verbena officinalis, and Momordica charantia – are commonly used in China as anti-infection treatments (Additional file 1: Table S1).
Hundreds of studies have verified that active compounds present in medicinal plants can be used as therapeutic agents, and that in silico methods are effective approaches to screening out these lead molecules for further investigation.21 In this study, 139 constituents of the four anti-infection medicinal plants were identified using the TCM Database@Taiwan.22 Molecular docking was used to investigate their binding affinities to the two common MRSA inhibitory targets, Penicillin binding proteins 2a (PBP2a) and 4 (PBP4).23,24 This helped us identify the anti-MRSA compounds and may explain any inhibitory or synergistic effects reported from the in vitro synergy testing. The physicochemical and absorption, distribution, metabolism, and excretion (ADME) properties and drug safety of these compounds were also analyzed to estimate their drug-likeness and reduce the failure rate among further drug discovery processes.25 Hence, this study uses in vitro and in silico methods to investigate the anti-MRSA properties of four medicinal herbs and their 139 constituents (Table S1).
Methods
Reagents and consumables for in vitro studies
Mueller–Hinton broth and agar were purchased from Hope Biol-Technology Company (Qingdao, China). Trypticase soy agar (TSA), brain heart infusion broth (BHIB), 30 µg cefoxitin filter paper discs, and strains of MRSA (ATCC43300) and methicillin-susceptible S. aureus (MSSA, ATCC25923) were commercially purchased from Compass Biotechnology Company (Shenyang, China). Extracts of M. officinalis and V. officinalis were from Ruikang Biological Company (Shanxi, China). Extracts of M. charantia and D. genkwa were from Hengdetang Biological Company (Zhengzhou, China) and Runxue Biological Company (Xi’an, China), respectively. These suppliers characterized all the extracts with HPLC fingerprints. The oxacillin and gentamicin were from Yuanye Bio-Technology Company (Shanghai, China).
Strains of ATCC43300 (methicillin resistant) and ATCC25923 (methicillin susceptible) were grown on TSA and BHIB, respectively, at 37℃ for 24 h. Both strains were then stored at −80℃ for future experiments. A Vitek 2 microbial identification system (bioMérieux, Marcy l’Etoile, France) was used to identify the S. aureus strain. The Kirby–Bauer disk diffusion method with 30 µg cefoxitin filter paper discs was used to investigate the drug resistance of both strains.
Susceptibility testing
The MIC is the lowest concentration of the control antibiotics and herbal extracts that will inhibit the visible growth of the ATCC43300 and ATCC25923 after 24 h of incubation at 37℃. MIC is one of the most common measurements of antibacterial activity against MRSA and MSSA.26 Here, tube dilation methods were used to determine the MIC of oxacillin and gentamicin against both MRSA and MSSA. The MICs of the extracts of M. officinalis, V. officinalis, M. charantia, and D. genkwa were determined by plate dilation methods with concentrations ranging from 0.00625 to 0.4 g/ml for both MRSA and MSSA strains. The gentamicin was tested with concentrations ranging from 1 to 32 mg/L for both strains, and the oxacillin was tested from 0.25 to 8 mg/L for MSSA and 8 to 256 mg/L for MRSA. These experiments were performed according to the guidelines from the Clinical and Laboratory Standards Institute.27
Synergistic testing
The fractional inhibitory concentration (FIC) index was used to quantify the synergistic interactions between the plant extracts and antibiotics against both MRSA and MSSA. The antimicrobial assays were performed using the checkerboard method28 with oxacillin or gentamicin in combination with the extracts of M. officinalis, V. officinalis, M. charantia, and D. genkwa. Both the MRSA and MSSA cultures were grown in the presence of one of the four herbal extracts with the following concentrations: 1/8 × MIC, 1/4 × MIC, 1/2 × MIC, 1 × MIC, 2 × MIC, and 4 × MIC in combination with either oxacillin or gentamicin, with concentrations ranging from 1/8 × MIC to 4 × MIC. These experiments were conducted in the same manner as for the MIC determination in the susceptibility testing.
The FIC index was calculated with the following formulas
where FIC index values of less than 0.5 indicated synergy, 0.5–0.75 indicated partial synergy, 0.76–1 indicated an additive effect, and >2 indicated antagonism.28,29
In silico molecular docking studies
Molecular docking has been successfully used in many studies to investigate the binding potential between proteins and ligands. It is popular for screening active compounds from medicinal plants for inhibition against pharmacological receptors.30,31 Here, the crystal structures of two common MRSA inhibitory targets, PBP2a and PBP4, were obtained from the Protein Data Bank (PDB) with PDB codes 4CJN32 and 3HUM,33 respectively. The constituents of M. officinalis, V. officinalis, M. charantia, and D. genkwa were downloaded from the TCM Database@Taiwan (http://tcm.cmu.edu.tw/), which covers more than 20,000 pure compounds isolated from 453 TCMs ingredients.22 The downloaded structures were built by ChemBioOffice 2008 (CambridgeSoft, Cambridge, MA) and energy minimized in a MM2 force field. Here, 139 constituents were identified and downloaded for in silico docking to both the PBP2a and PBP4 crystal structures.
The Surflex-Dock module of the SYBYL-X 1.3 molecular modeling suite was used to perform the molecular docking simulations. The accuracy of Surflex-Dock in scoring protein–ligand interactions has been successfully validated for large and diverse sets of target protein structures, demonstrating a high screening enrichment performance.34–36 In this study, the 139 constituents in the crystal structures of 4CJN and 3HUM were docked into the PBP2a and PBP4 independently. Apart from the 139 constituents, the binding scores of the original ligands bound to each of the PBP2a and PBP4 crystal structures were also calculated (Figure 1). These ligands were confirmed as PBP2a and PBP4 inhibitors from experimental results downloaded from the PDB, and their binding scores of the two ligands were compared with those of the top-ranked herbal constituents. The structures of all the molecules were minimized using the standard tripos force field37 before the docking simulations. The docking parameters were set to the default in Surflex-Dock; for instance, the protomol threshold and bloat were set at 0.5 and 0.0, respectively. The protomols were generated by removing the ligands of the co-crystallized compounds and defined by the residues surrounding the active site. This method gives a fixed degree of coverage against the residues proximal to the specified ligands or explicit residues. The water molecules in the PBP2a and PBP4 crystal structures were retained for docking because water molecules may have cross-linkages between proteins and ligands that help to stabilize the protein–ligand complex.38 More importantly, the presence of water molecules may increase docking accuracy.39 All of the atoms were allowed to move during the simulations because flexible docking is generally more accurate than rigid docking.40 Finally, the resulting binding modes of the top-scoring constituents were analyzed using SYBYL-X 1.3 and PyMOL. The docking procedures and calculations were the same as in the following evaluation of docking methods, which would ensure that the results were consistently reliable.
Figure 1.
Two-dimensional structures of the two ligands bound to the crystal structures of PBP2a (a) and PBP4 (b). PBP2a: penicillin binding protein 2a; PBP4: penicillin binding protein 4
Evaluation of docking methods
Surflex-Dock has been successfully validated on large and diverse sets of target proteins and has produced valuable results in investigating PBP inhibition.41 We further evaluated its specific performance in simulating PBP2a–ligand interactions using receiver operating characteristic (ROC) analysis. Docking was performed on the X-ray crystallography structure of PBP2a (PDB: 4CJN) and the 11,421 ligands from the Zinc In Man database,42 plus eight experimentally approved PBP2a inhibitors with IC50 less than 100 µM.43 These eight approved inhibitors served as the “true” hits in this validation. Added together, a ligand library of 11,429 was formed. ROC analysis has previously been used to evaluate the accuracy of docking algorithms in various protein–ligand systems.36,44 For PBP4, only five experimentally proved inhibitors were identified from our literature search,45 which covered various databases including Medline, Embase, Cochrane Library, and the China National Knowledge Infrastructure database. These five inhibitors are in the same series and have very similar structures. Thus, using these compounds exclusively would not provide acceptable statistical power to evaluate the docking accuracy for this particular protein.
Prediction of drug-like properties
The structures of the 139 herbal constituents downloaded from the TCM Database@Taiwan database were applied to ACD/Percepta 14.0 software46 for drug-like properties evaluation. The drug profiler module in ACD/Percepta uses high-quality predictive methods to screen compounds with three types of profiler parameters46: physicochemical, ADME, and drug safety profiling. The physicochemical profiler evaluates an array of physicochemical properties including log P, molecular weight, H-bond donors, H-bond receptors, rotatable bond, solubility, Lipinski’s Rule-of-5, and predefined lead-like categories. Based on these properties, each constituent is classified as having favorable or unfavorable drug-like physicochemical properties.47 The ADME profiler predicts P-glycoprotein specificity, passive permeability across Caco-2 cell monolayers, and human intestinal absorption (HIA), all of which are important factors that affect oral bioavailability.48 The drug safety profiler includes the prediction of Cytochrome P450 (CYP) regioselectivity (CYP3A4, CYP2D6, CYP2C9, CYP2C19, and CYP1A2), probability of producing positive Ames (mutagenicity prediction), and human ether-a-go-go-related gene (hERG) (cardiotoxicity prediction).49,50 The accuracy and sensitivity of all the above methods were successfully evaluated by the supplier, ACD/Labs. All the data sheets are available on the company website (http://www.acdlabs.com).
Results
In this study, in vitro susceptibility and synergistic testing were conducted to investigate the inhibitory effects of M. officinalis, V. officinalis, M. charantia, and D. genkwa on MRSA. In silico work was also performed to predict the active ingredients that are responsible for the inhibitory effects.
In vitro susceptibility and synergistic studies
Our in vitro susceptibility testing found that the extracts of all four TCMs had antibacterial effects on both MRSA and MSSA strains, with V. officinalis performed the best (Table 1). The MIC of V. officinalis was lower than that of gentamicin for MRSA (Table 1), which indicated the strong inhibitory effect of V. officinalis on MRSA. In terms of the synergistic testing, the extract of D. genkwa obtained an FIC index value of 0.375 (Table 2), which demonstrated a potentiated antibacterial effect of oxacillin against MRSA. The extract of D. genkwa partially potentiated the effect of gentamicin, as indicated by an FIC index value of 0.750 (Table 2). The extracts of M. officinalis and V. officinalis demonstrated partial synergistic effects in combination with the antibiotics (Table 2).
Table 1.
Minimum inhibitory concentration (MIC) of test samples for MRSA ATCC43300 and MSSA ATCC25923
| MIC (mg/L) |
||
|---|---|---|
| Test samples | MRSA ATCC43300 | MSSA ATCC25923 |
| Oxacillin | 128 | 1 |
| Gentamicin | 16,000 | 32 |
| D. genkwa extract | 35,000 | 60,000 |
| M. officinalis extract | 35,000 | 40,000 |
| V. officinalis extract | 6250 | 6250 |
| M. charantia extract | 70,000 | 90,000 |
Table 2.
Fractional inhibitory concentration (FIC) index of test samples for MRSA ATCC43300 in combination with oxacillin or gentamicin
| Oxacillin |
Gentamicin |
|||
|---|---|---|---|---|
| Test samples | FIC index | Outcome | FIC index | Outcome |
| D. genkwa extract | 0.375 | Synergy | 0.750 | Partial synergy |
| M. officinalis extract | 0.625 | Partial synergy | 0.750 | Partial synergy |
| V. officinalis extract | 0.625 | Partial synergy | 1.125 | No effect |
| M. charantia extract | >2 | Antagonism | 1.200 | No effect |
In silico docking simulations
The docking accuracy on PBP2a was successfully validated using an ROC analysis (Figure 2), where a calculated area under the curve (AUC) value indicated the high predictive power. An AUC value of 0.5 indicates a random result with no predictive power and 1.0 indicates a perfect prediction. In general, a model with an AUC value of 0.7 or above is considered reliable.36,44 Here, the obtained AUC value was 0.72, showing that the accuracy of our docking methods was reliable.
Figure 2.
Receiver operating characteristic (ROC) curves of the docking results for PBP2a and the ligands from the ZIM database. The green reference line indicates random results, with an AUC value of 0.50. An AUC value of 1.0 indicates a perfect prediction and one of 0.7 or above is considered reliable. AUC: area under the curve; PBP2a: penicillin binding protein 2a. (A color version of this figure is available in the online journal.)
The docking results of PBP2a revealed that 25 of the 139 constituents had advanced scores than the crystallographic derived PBP2a inhibitor, quinazolinone (PDB: 4CJN).32 The first ranked constituent was tiliroside, which obtained a Surflex score of 10.5 (Table 3). This score is superior to that of the experimentally proved inhibitor, quinazolinone, which obtained a score of 7.6. The binding mode analysis revealed that the tiliroside interacted with the residues Tyr105, Gly106, Ile144, Asn146, Lys273, Asp275, Asp295, and Tyr297 in the PBP2a binding pocket. In contrast, quinazolinone demonstrated significantly fewer interactions in the same binding pocket (Figure 3). Tiliroside also achieved a high score in binding to the PBP4 (Table 4), as indicated by a score of 12.3. The binding mode analysis showed that tiliroside formed interactions with the residues Glu114, Ser116, Ser139, Phe241, Gly261, and Ser263. These interactions appeared to be stronger than those between cefotaxime and PBP4 (Figure 4). Luteolin 7-diglucuronide of V. officinalis and yuanhuapine of D. genkwa achieved the second and third highest score in the PBP2a docking simulation (Table 3). Other highly ranked compounds against PBP2a were (-)-pinoresinol, neo-chlorogenic acid, momorcharaside B, 3,4-dihydroverbenalin, magnatriol B, and daucosterol (Table 3).
Table 3.
Surflex scores and structural properties of the top 10 ranked constituents against PBP2a
| Constituents | Botanical source | Surflex score | |
|---|---|---|---|
| Tiliroside |
|
D. genkwa | 10.5 |
| Luteolin-7-diglucuronide |
|
V. officinalis | 10.1 |
| Yuanhuapine |
|
D. genkwa | 9.3 |
| (-)-pinoresinol |
|
D. genkwa | 9.0 |
| Neo-chlorogenic acid |
|
V. officinalis | 8.9 |
| Momorcharaside B |
|
M. charantia | 8.8 |
| 3,4-dihydroverbenalin |
|
V. officinalis | 8.7 |
| Magnatriol B |
|
M. officinalis | 8.7 |
| Daucosterol |
|
V. officinalis | 8.7 |
| 5beta-hydroxyresiniferonol-6alpha, 7alpha-epoxy-12beta-acetoxy-9, 13,14-ortho-2E-decenoate |
|
D. genkwa | 8.4 |
PBP2a: penicillin binding protein 2a.
Figure 3.
(a) Crystallographic binding mode of quinazolinone in the PBP2a active site. (b) Surflex-Dock predicted binding mode of tiliroside in the PBP2a active site. All distances are measured in Angstroms (Å). PBP2a: penicillin binding protein 2a. (A color version of this figure is available in the online journal.)
Table 4.
Surflex scores and structures of the top 10 ranked constituents against PBP4
| Constituents | Botanical source | Surflex score | |
|---|---|---|---|
| Apigenin-7-O-beta-d-glucopyranoside |
|
D. genkwa | 12.5 |
| Tiliroside |
|
D. genkwa | 12.3 |
| Genkwanine E |
|
D. genkwa | 12.1 |
| Jionoside D |
|
V. officinalis | 11.5 |
| Genkwanine F |
|
D. genkwa | 11.2 |
| Luteolin-7-O-beta-d-galactopyranoside |
|
V. officinalis | 11.0 |
| Daucosterol |
|
V. officinalis | 10.9 |
| 3-caffeoylquinic acid |
|
V. officinalis | 10.7 |
| Yuanhuagine |
|
D. genkwa | 10.5 |
| Yuanhuajine |
|
D. genkwa | 10.4 |
Figure 4.
(a) Crystallographic binding mode of cefotaxime in the PBP4 active site. (b) Surflex-Dock predicted binding mode of tiliroside in the PBP4 active site. All distances are measured in Angstroms (Å). PBP4: penicillin binding protein 4. (A color version of this figure is available in the online journal.)
The docking results of PBP4 and herbal constituents showed that 40 compounds have advanced binding score than the experimentally proven PBP4 inhibitor, cefotaxime. The binding scores of these constituents ranged from 12.5 to 8.8, whereas cefotaxime obtained a score of 8.7. The top-ranked constituent was apigenin,7-O-beta-d-glucopyranoside (Table 4), which is a constituent of D. genkwa.
Discussion
In vitro studies
The results of the in vitro susceptibility testing revealed that the MICs of oxacillin and gentamicin for MRSA were 128 and 500 times than that of MSSA (Table 1). This clearly showed a high level of antibiotic resistance to MRSA but interestingly, the MIC values of all four TCMs for MRSA were lower or equal to that of MSSA. This indicated that the TCMs produced stronger inhibitory effects on MRSA than on MSSA, which led to a theory that the inhibitory mechanisms are different between the antibiotics and TCMs. Future research on this mechanism could lead to a novel pathway for designing anti-MRSA agents. For the synergistic testing, the extract of D. genkwa potentiated the antibacterial effects of oxacillin against MRSA (Table 2); this synergistic effect indicated that the efficacy of D. genkwa in combination with oxacillin was greater than the sum of their individual use. It also suggested that the extract of D. genkwa somehow enhanced the oxacillin pharmaceutical actions, inhibiting the synthesis of a peptidoglycan layer of bacterial cell walls by binding to PBPs. The extract of D. genkwa partially potentiated the effect of gentamicin, which is an aminoglycoside antibiotic, has a different pharmacological mechanism than oxacillin. Gentamicin irreversibly binds to the 30S subunit of the bacterial ribosome and disrupts protein synthesis. There are different opinions about the mechanism of aminoglycoside resistance on MRSA, and the widely accepted hypotheses are ribosomal mutations, active efflux, and aminoglycoside modifying enzymes.51 Thus, further investigations of these synergistic effects caused by D. genkwa on oxacillin and gentamicin should focus on areas relating to these resistance mechanisms.
The extracts of M. officinalis and V. officinalis demonstrated partial synergistic effects in combination with the antibiotics (Table 2), indicating that the active constituents of these plants must somehow overcome the resistance properties of MRSA. The lower synergistic effects of M. officinalis and V. officinalis than D. genkwa may be due to the lower efficiency and/or concentrations of these constituents. To further investigate, we performed in silico studies on PBP2a and PBP4.
In silico studies
Molecular docking analyses were performed to predict the binding affinities among the 139 herbal constituents of the four herbs against PBP2a and PBP4. Drug-like physicochemical properties, ADME, and drug safety were also predicted to increase the chance of success in developing a drug treatment for clinical use.
PBP2a
The first ranked constituent against PBP2a was tiliroside, which obtained a higher binding score than that of the experimentally proved PBP2a inhibitor, quinazolinone (Table 3). This indicates that the binding of tiliroside to PBP2a will from a stable complex and the defined binding site of PBP2a is an appropriate target of tiliroside. Expressly, this high affinity molecule is being predicted as a PBP2a inhibitor. Tiliroside is a glycosidic flavonoid and a kaempferol derivative and its antibacterial properties have been controversially documented in various studies. For example, tiliroside was found to be a significantly active compound against Candida albicans (MIC value = 1 µg/mL), Escherichia coli (2 µg/mL), and Bacillus subtilis (8 µg/mL).52 However, the findings of Zhang et al.53 suggested that tiliroside had no relevant antibacterial activity on E. coli, B. subtilis, or C. albicans. The reasons for these diverse results are still uncertain. In terms of the inhibition of S. aureus, tiliroside has not been found to have substantial antibacterial activity, but it was shown to somehow modulate the activities of other conventional antibiotics against the resistant strain of S. aureus. It increased the activity of norfloxacin 16 times, ciprofloxacin 16 times, lomefloxacin 4 times, and ofloxacin by 2 times.54 Thus, tiliroside could be a constituent of D. genkwa capable of producing a synergistic effect with oxacillin against MRSA in this study. The potential mechanism could be related to the binding of PBP2a or PBP4, as indicated by the high binding scores calculated in this study. In terms of ADME and drug safety, no significant information was found in the literature. Here, the in silico methods predicted that tiliroside would have an optimal log P value and good HIA properties (Tables 5 and 6). It was also predicted to be a non-penetrant of the central nervous system (CNS) and a non-inhibitor of cardiac hERG channels (Table 7), which indicate a low risk of CNS adverse effects and hERG-related cardiotoxicity when used clinically. However, the plasma protein binding (PPB) value indicated that a high percentage (94%) of administrated tiliroside will bind with blood plasma protein (Table 6). This means that only 6% of tiliroside may reach the target receptor and produce pharmaceutical effects. This high PPB value may reduce the efficiency and delay the excretion of tilrioside.55 Moreover, a high PPB value indicates a high potential for producing drug–drug interactions, especially with other highly protein-bound drugs. In general, drugs with less than an 80–85% PPB value have appeared to be of slight clinical importance.55 Nevertheless, there are still many highly protein-bound drugs with >95% PPB being used, especially for severe illness and serious infections. Classic examples are dicloxacillin and flucloxacillin.56
Table 5.
Physicochemical drug-like properties of the top-ranked herbal constituents of PBP2a and PBP4
| log P | MW | H-bond donors | H-bond acceptors | Rotatable bonds | Rings | Lipinski | Lead like | Aqueous solubility | |
|---|---|---|---|---|---|---|---|---|---|
| Tiliroside | 2.26 | 594.5 | 7 | 13 | 8 | 5 | 3 | 3 | 0.03 |
| LD | 0.44 | 476.4 | 6 | 12 | 5 | 4 | 2 | 3 | 2.68 |
| Yuanhuapine | 4.32 | 542.6 | 3 | 10 | 5 | 7 | 1 | 3 | 2.42 |
| (-)-pinoresinol | 1.90 | 358.4 | 2 | 4 | 4 | 4 | 0 | 0 | 0.06 |
| Neo-chlorogenic acid | −0.33 | 354.3 | 6 | 9 | 5 | 2 | 1 | 1 | 1000 |
| Momorcharaside B | 2.64 | 654.9 | 8 | 10 | 8 | 5 | 2 | 3 | 0.002 |
| 3,4-dihydroverbenalin | −2.53 | 390.4 | 5 | 10 | 5 | 3 | 1 | 2 | 1000 |
| Magnatriol B | 2.59 | 242.3 | 3 | 3 | 3 | 2 | 0 | 0 | 0.32 |
| Daucosterol | 7.18 | 576.9 | 4 | 6 | 9 | 5 | 2 | 2 | 0.0001 |
| 5HAD | 7.13 | 588.7 | 3 | 10 | 11 | 6 | 2 | 3 | 0.003 |
| AG | 0.18 | 432.4 | 6 | 10 | 4 | 4 | 1 | 2 | 1.38 |
| Genkwanine E | 4.59 | 638.7 | 6 | 10 | 8 | 6 | 2 | 4 | 0.05 |
| Jionoside D | 0.75 | 624.6 | 9 | 15 | 11 | 4 | 3 | 3 | 15.7 |
| Genkwanine F | 4.68 | 640.8 | 6 | 10 | 9 | 6 | 2 | 4 | 0.04 |
| LG | 0.13 | 448.4 | 7 | 11 | 4 | 4 | 2 | 2 | 1.86 |
| 3-caffeoylquinic acid | −2.53 | 390.4 | 5 | 10 | 5 | 3 | 1 | 2 | 1000 |
| Yuanhuagine | 5.78 | 584.7 | 3 | 10 | 9 | 6 | 2 | 3 | 0.07 |
| Yuanhuajine | 7.34 | 660.8 | 3 | 10 | 11 | 7 | 2 | 3 | 0.02 |
5HAD: 5beta-hydroxyresiniferonol-6alpha,7alpha-epoxy-12beta-acetoxy-9,13,14-ortho-2E-decenoate; LD: luteolin-7-diglucuronide; LG: luteolin-7-O-beta-d-galactopyranoside; PBP2a: penicillin binding protein 2a; PBP4: penicillin binding protein 4; 3AG: apigenin-7-O-beta-d-glucopyranoside.
og P = octanol–water partition coefficient under standard conditions at 25℃ (optimal value: −1.00 to 4.20); MW: molecular weight (values for optimal drug-like properties are <=460.00); optimal numbers of H-bond donors, H-bond acceptors, rotatable bonds and rings are <=5, <=10, <=10, and <4, respectively; optimal numbers of violations for both Lipinski and lead like are <1; aqueous solubility was calculated at pH 6.4 and in mg/mL (<=0.01 indicates highly insoluble, 0.01–0.1 is insoluble, >0.1 is soluble).
Table 6.
ADME drug-like properties of the top 10 ranked herbal constituents of PBP2a and PBP4
| Caco-2 | PPB (%) | CNS | HIA (%) | |
|---|---|---|---|---|
| Tiliroside | 0.3 × 10−6 | 94 | −5.78 | 85 |
| LD | 0.1 × 10−6 | 84 | −5.98 | 23 |
| Yuanhuapine | 1.5 × 10−4 | 8 | −1.64 | 100 |
| (-)-pinoresinol | 1.2 × 10−4 | 81 | −2.70 | 100 |
| Neo-chlorogenic acid | 0.0 × 10−6 | 76 | −6.25 | 1 |
| Momorcharaside B | 2.0 × 10−6 | 93 | −4.37 | 96 |
| 3,4-dihydroverbenalin | 0.0 × 10−6 | 51 | −6.98 | 1 |
| Magnatriol B | 130 × 10−6 | 78 | −2.24 | 100 |
| Daucosterol | 0.4 × 10−6 | 99 | −4.38 | 100 |
| 5HAD | 1.0 × 10−6 | 13 | −2.73 | 100 |
| AG | 0.0 × 10−6 | 74 | −5.68 | 18 |
| Genkwanine E | 1.6 × 10−5 | 55 | −2.44 | 100 |
| Jionoside D | 0.1 × 10−6 | 53 | −5.22 | 9 |
| Genkwanine F | 1.4 × 10−5 | 47 | −2.36 | 100 |
| LG | 0.0 × 10−6 | 64 | −5.89 | 7 |
| 3-caffeoylquinic acid | 0.0 × 10−6 | 51 | −6.98 | 1 |
| Yuanhuagine | 2.3 × 10−5 | 4 | −1.82 | 100 |
| Yuanhuajine | 0.7 × 10−6 | 17 | −2.92 | 100 |
5HAD: 5beta-hydroxyresiniferonol-6alpha,7alpha-epoxy-12beta-acetoxy-9,13,14-ortho-2E-decenoate; LD: luteolin-7-diglucuronide; LG: luteolin-7-O-beta-d-galactopyranoside; PBP2a: penicillin binding protein 2a; PBP4: penicillin binding protein 4; 3AG: apigenin-7-O-beta-d-glucopyranoside;.
Caco-2 predicts passive intestinal permeability and was measured in cm/s (<=1.00 indicates poorly permeable, 1.00–7.00 is moderately permeable, >7.00 is highly permeable); plasma protein binding estimates the overall percentage bound in blood plasma protein; CNS values of <=−3.50 indicates non-penetrant to central nervous system, −3.50 to −3.00 is weak penetrant, and >−3.00 is penetrant; HIA: human intestinal absorption (<=30% indicates poorly absorbed, 30–70% is moderately absorbed, >70% is highly absorbed).
Table 7.
Drug safety profiles of the top 10 ranked herbal constituents of PBP2a and PBP4
| Ames | hERG | CYP1A2 | CYP2C9 | CYP2C19 | CYP2D6 | CYP4A4 | |
|---|---|---|---|---|---|---|---|
| Tiliroside | 0.64 | 0.25 | NI | U | U | NI | U |
| LD | 0.50 | 0.34 | NI | NI | NI | NI | U |
| Yuanhuapine | 0.36 | 0.44 | NI | U | U | NI | U |
| (-)-pinoresinol | 0.41 | 0.42 | U | U | U | U | U |
| Neo-chlorogenic acid | 0.22 | 0.27 | NI | NI | NI | NI | NI |
| Momorcharaside B | 0.27 | 0.33 | NI | NI | NI | NI | NI |
| 3,4-dihydroverbenalin | 0.38 | 0.29 | NI | NI | NI | NI | NI |
| Magnatriol B | 0.30 | 0.38 | U | U | U | NI | U |
| Daucosterol | 0.23 | 0.38 | NI | NI | NI | NI | NI |
| 5HAD | 0.37 | 0.42 | NI | U | U | NI | U |
| AG | 0.70 | 0.30 | NI | NI | NI | NI | NI |
| Genkwanine E | 0.40 | 0.40 | NI | NI | U | NI | U |
| Jionoside D | 0.44 | 0.28 | NI | NI | NI | NI | NI |
| Genkwanine F | 0.41 | 0.40 | NI | NI | U | NI | U |
| LG | 0.52 | 0.30 | NI | NI | NI | NI | NI |
| 3-caffeoylquinic acid | 0.38 | 0.29 | NI | NI | NI | NI | NI |
| Yuanhuagine | 0.52 | 0.39 | NI | U | U | U | U |
| Yuanhuajine | 0.37 | 0.43 | NI | U | U | U | U |
CYP: Cytochrome P450; 5HAD: 5beta-hydroxyresiniferonol-6alpha,7alpha-epoxy-12beta-acetoxy-9,13,14-ortho-2E-decenoate; I: inhibitor; LD: luteolin-7-diglucuronide; LG: luteolin-7-O-beta-d-galactopyranoside; 3AG: apigenin-7-O-beta-d-glucopyranoside; NI: non-inhibitor; PBP2a: penicillin binding protein 2a; PBP4: penicillin binding protein 4; U: undefined.
Ames test predicts mutagenic potential (<=0.33 indicates non-mutagenic, 0.33–0.67 is undefined, >0.67 is mutagenic); hERG estimates cardiotoxicity of compounds related to drug interactions with the hERG channel (<=0.33 indicates non-inhibitor, 0.33–0.67 is undefined, >0.67 is inhibitor).
Luteolin 7-diglucuronide of V. officinalis achieved the second highest score in the PBP2a docking simulation (Table 3). Its antibacterial activity was uncertain in our literature search. However, its structurally related compounds, luteolin and a series of its derivatives are well-documented antibacterial agents against various species, including S. aureus.57 Luteolin was also characterized as a selective inhibitor to the growth of S. aureus, including MRSA and MSSA, as indicated by an MIC value of 62.5–125 µg/mL.58 It has also been found to have synergetic effects on MRSA in combination with several quinolones and aminoglycosides antibiotics.59 Thus, as a derivative of luteolin, luteolin 7-diglucuronide may also have anti-MRSA effects. It may have produced the partial synergistic effects of V. officinalis observed in this study. Luteolin 7-diglucuronide achieved an optimal log P value (Table 5), which indicated an optimal balance of lipophilicity and hydrophilicity for oral absorption. However, as a more accurate parameter, this compound appeared to have poor HIA properties (Table 6),60 suggesting that luteolin 7-diglucuronide may only be administered by injection or using costly formulation strategies to improve its bioavailability, such as self-emulsifying systems.61 In terms of causing CNS adverse reactions, luteolin 7-diglucuronide is a proven moderate GABAA inhibitor (IC50 40 μM).62 GABAA inhibitors produce stimulant effects and are mainly located in the CNS.63 This in silico work predicted that luteolin 7-diglucuronide would be a CNS non-penetrant (Table 6), making the CNS effect minimal. Luteolin 7-diglucuronide was also predicted to be a non-inhibitor of four of the most significant Cytochrome P450s (CYPs) (Table 7), indicating a low likelihood of causing drug–drug and drug–food interactions. Thus, further experimental investigations of this compound, especially on the inhibition of PBP2a, would be truly worthwhile.
The third ranked compound against PBP2a was yuanhuapine (Table 3), which had no significant MRSA-related data in our literature search. Its structurally related compound, yuanhuacine, has been identified as a DNA topoisomerase I inhibitor based on an animal study.64 This type of inhibitor has also been suggested for use in treating cancer and antibacterial activity.65 Its effect on MRSA is unknown. In terms of ADME and drug safety, yuanhuapine is a very lipophilic compound (log P = 4.32) with 100% HIA (Tables 5 and 6). Its binding ability to most CYP and hERG is undefined, as indicated by a borderline score between inhibitor and non-inhibitor. Thus, the probability of obtaining a correct prediction is low.
Other highly ranked compounds against PBP2a were (-)-pinoresinol, neo-chlorogenic acid, momorcharaside B, 3,4-dihydroverbenalin, magnatriol B, and daucosterol (Table 3). All of them had no significant MRSA-related data in our literature search. Regarding ADME, (-)-pinoresinol and magnatriol B had excellent physicochemical drug-like properties and were predicted as having high HIA properties (Tables 5 and 6). However, they were both CNS penetrant and undefined in most CYP profiling (Tables 6 and 7). Neo-chlorogenic acid and 3,4-dihydroverbenalin were highly hydrophilic compounds, both with poor absorption (<1% HIA) (Tables 5 and 6). Daucosterol was also highly ranked in the docking simulations for PBP4, suggesting that it may also bind tightly to PBP4. However, it is highly lipophilic (log P = 7.18) with 99% of PPB (Tables 5 and 6). The log P value of momorcharaside B was optimal (2.64) and predicted to have 96% HIA (Tables 5 and 6). It was a non-inhibitor to all five common CYP and hERG (Tables 6 and 7). It was also non-mutagenic, as indicated by a low Ames value (0.27) (Table 7). Further in vitro investigation on momorcharaside B against PBP2a would be worthwhile.
PBP4
The docking simulations of PBP4 and 139 herbal constituents suggested that 40 of the latter may bind to PBP4 in a more stable mode than the experimentally proven PBP4 inhibitor, cefotaxime.66 The top-ranked constituent was apigenin,7-O-beta-d-glucopyranoside (Table 4), a constituent of D. genkwa. According to our literature search, the effect of this compound on MRSA is unknown. However, based on its structurally related compounds, apigenin was found to be selectively toxic to S. aureus, including the MRSA and MSSA strains (MIC 3.9–15.6 µg/mL).58 Thus, apigenin,7-O-beta-d-glucopyranoside could be another active constituent of D. genkwa, which produced a synergistic anti-MRSA effect in combination with oxacillin in this study. Our docking simulations suggested that its mechanism is PBP4 inhibition. In terms of ADME, apigenin,7-O-beta-d-glucopyranoside was predicted to be poorly absorbed (HIA = 18%) (Table 6), such that a specific formulation would be necessary to improve its oral absorption. It was also predicted as a non-inhibitor for all five CYPs and hERG. However, it appeared to be mutagenic (Table 7).
In our literature search, we found no MRSA-related articles on the other top-ranked constituents, including genkwanine E, jionoside D, genkwanine F, luteolin-7-O-beta-d-galactopyranoside, 3-caffeoylquinic acid, and yuanhuagine. None of these compounds had excellent ADME or drug-safety properties. Genkwanine E, genkwanine F, jionoside D, and yuanhuagine were predicted to have poor physiochemical drug-like properties but high HIA (Tables 5 and 6). Luteolin-7-O-beta-d-galactopyranoside was poorly absorbed and mutagenic. 3-caffeoylquinic acid had acceptable physiochemical drug-like properties and a good drug-safety profile, but poor absorption. Again, absorption of this constituent can be modified by drug formulation technologies.
Conclusions
In vitro susceptibility testing revealed that the extracts of the four TCMs – M. officinalis, V. officinalis, M. charantia, and D. genkwa – had anti-MRSA and anti-MSSA effects, with V. officinalis exhibiting advanced performance in comparison to gentamicin. All four TCMs produced stronger inhibitory effects on MRSA than on MSSA. The synergistic testing results demonstrated the potentiated antibacterial effects, when combined with antibiotics, on MRSA of M. officinalis, V. officinalis, and D. genkwa. To detect which active constituents were responsible for producing those effects, validated in silico simulations were performed on 139 constituents of the four herbs. The results indicated that some of the constituents had the ability to binding more strongly to PBP2a and PBP4 than their experimentally proven inhibitors, indicating their potential to inhibit MRSA. However, most of the constituents had poor physicochemical drug-like properties, ADME, and drug safety profiles. Tiliroside achieved excellent scores for binding to both PBP2a and PBP4, further supporting current scientific evidence of its synergistic inhibition effects on MRSA when combined with antibiotics. This study revealed that tiliroside’s mechanism could be the inhibition of PBP2a and PBP4. Pinoresinol, magnatriol B, and momorcharaside B bind to both PBP2a and PBP4 in a stable mode and are thus potentially strong inhibitors with acceptable drug-like properties and drug safety profiles. Further investigation and optimization of these compounds could assist in developing therapeutic agents to modulate the effect of antibiotics on MRSA for use as adjuvant therapy.
Supplementary Material
Acknowledgements
The financial support of the Macao Polytechnic Institute Research Fund (Project no: RP/ESS-04/2016) is gratefully acknowledged. The authors thank Kosca Leong for technical assistance.
Authors’ contributions
All authors participated in the design, interpretation of the studies and analysis of the data, and review of the manuscript; SOH, CFH, CHC, IHF, CKN, and PF conducted the experiments, LRM provide technical advices and lead the studies, and PF and CFK wrote the manuscript.
Declaration of Conflicting Interests
The author(s) declared no potential conflicts of interest with respect to the research, authorship, and/or publication of this article.
Supplemental Material
Supplementary material for this paper can be found at http://journals.sagepub.com/doi/suppl/10.1177/1535370216689828.
References
- 1.Huang SS, Hinrichsen VL, Datta R, Spurchise L, Miroshnik I, Nelson K, Platt R. Methicillin-resistant Staphylococcus aureus infection and hospitalization in high-risk patients in the year following detection. PLoS One 2011; 6: e24340–e24340. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Malani PN. National burden of invasive methicillin-resistant Staphylococcus aureus infection. JAMA 2014; 311: 1438–9. [DOI] [PubMed] [Google Scholar]
- 3.Lee BY, Singh A, David MZ, Bartsch SM, Slayton RB, Huang SS, Zimmer SM, Potter MA, Macal CM, Lauderdale DS, Miller LG, Daum RS. The economic burden of community-associated methicillin-resistant Staphylococcus aureus (CA-MRSA). Clin Microbiol Infect 2013; 19: 528–36. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Stefani S, Chung DR, Lindsay JA, Friedrich AW, Kearns AM, Westh H, Mackenzie FM. Meticillin-resistant Staphylococcus aureus (MRSA): global epidemiology and harmonisation of typing methods. Int J Antimicrob Agents 2012; 39: 273–82. [DOI] [PubMed] [Google Scholar]
- 5.Edwards B, Andini R, Esposito S, Grossi P, Lew D, Mazzei T, Novelli A, Soriano A, Gould IM. Treatment options for methicillin-resistant Staphylococcus aureus (MRSA) infection: where are we now? J Glob Antimicrob Resist 2014; 2: 133–40. [DOI] [PubMed] [Google Scholar]
- 6.Holland TL, Arnold C, Fowler VG. Clinical management of Staphylococcus aureus bacteremia: a review. JAMA 2014; 312: 1330–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.van Hal SJ, Fowler VG. Is it time to replace vancomycin in the treatment of methicillin-resistant Staphylococcus aureus infections? Clin Infect Dis 2013; 56: 1779–88. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Horne KC, Howden BP, Grabsch EA, Graham M, Ward PD, Xie S, Mayall BC, Johnson PDR, Grayson ML. Prospective comparison of the clinical impact of heterogeneous vancomycin-intermediate methicillin-resistant Staphylococcus aureus (MRSA) and vancomycin-susceptible MRSA. Antimicrob Agents Chemother 2009; 53: 3447–452. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Wong-Beringer A, Joo J, Tse E, Beringer P. Vancomycin-associated nephrotoxicity: a critical appraisal of risk with high-dose therapy. Int J Antimicrob Agents 2011; 37: 95–101. [DOI] [PubMed] [Google Scholar]
- 10.Fowler VG, Boucher HW, Corey GR, Abrutyn E, Karchmer AW, Rupp ME, Levine DP, Chambers HF, Tally FP, Vigliani GA, Cabell CH, Link AS, DeMeyer I, Filler SG, Zervos M, Cook P, Parsonnet J, Bernstein JM, Price CS, Forrest GN, Fatkenheuer G, Gareca M, Rehm SJ, Brodt HR, Tice A, Cosgrove SE. S aureus Endocarditis and Bacteremia Study Group. Daptomycin versus standard therapy for bacteremia and endocarditis caused by Staphylococcus aureus. N Engl J Med 2006; 355: 653–65. [DOI] [PubMed] [Google Scholar]
- 11.Mangili A, Bica I, Snydman DR, Hamer DH. Daptomycin-resistant methicillin-resistant Staphylococcus aureus bacteremia. Clin Infect Dis 2005; 40: 1058–60. [DOI] [PubMed] [Google Scholar]
- 12.Jacobson LM, Milstone AM, Zenilman J, Carroll KC, Arav-Boger R. Daptomycin therapy failure in an adolescent with methicillin-resistant Staphylococcus aureus bacteremia. Pediatr Infect Dis J 2009; 28: 445–7. [DOI] [PubMed] [Google Scholar]
- 13.Gould IM, David MZ, Esposito S, Garau J, Lina G, Mazzei T, Peters G. New insights into methicillin-resistant Staphylococcus aureus (MRSA) pathogenesis treatment and resistance. Int J Antimicrob Agents 2012; 39: 96–104. [DOI] [PubMed] [Google Scholar]
- 14.Nakazawa H, Kikuchi Y, Honda T, Isago T, Nozaki M. Enhancement of antimicrobial effects of various antibiotics against methicillin-resistant Staphylococcus aureus (MRSA) by combination with fosfomycin. J Infect Chemother 2003; 9: 304–9. [DOI] [PubMed] [Google Scholar]
- 15.Fujimura S, Sato T, Hayakawa S, Kawamura M, Furukawa E, Watanabe A. Antimicrobial efficacy of combined clarithromycin plus daptomycin against biofilms-formed methicillin-resistant Staphylococcus aureus on titanium medical devices. J Infect Chemother 2015; 21: 756–9. [DOI] [PubMed] [Google Scholar]
- 16.El Haj C, Murillo O, Ribera A, Vivas M, Garcia-Somoza D, Tubau F, Cabellos C, Cabo J, Ariza J. Daptomycin combinations as alternative therapies in experimental foreign-body infection caused by methicillin-susceptible Staphylococcus aureus. Int J Antimicrob Agents 2015; 46: 189–95. [DOI] [PubMed] [Google Scholar]
- 17.Garnacho-Montero J, Amaya-Villar R, Gutierrez-Pizarraya A, Espejo-Gutierrez de Tena E, Artero-Gonzalez ML, Corcia-Palomo Y, Bautista-Paloma J. Clinical efficacy and safety of the combination of colistin plus vancomycin for the treatment of severe infections caused by carbapenem-resistant Acinetobacter baumannii. Chemotherapy 2013; 59: 225–31. [DOI] [PubMed] [Google Scholar]
- 18.An J, Zuo GY, Hao XY, Wang GC, Li ZS. Antibacterial and synergy of a flavanonol rhamnoside with antibiotics against clinical isolates of methicillin-resistant Staphylococcus aureus (MRSA). Phytomedicine 2011; 18: 990–3. [DOI] [PubMed] [Google Scholar]
- 19.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: 936–41. [DOI] [PubMed] [Google Scholar]
- 20.Zuo GY, Zhang XJ, Yang CX, Han J, Wang GC, Bian ZQ. Evaluation of traditional Chinese medicinal plants for anti-MRSA activity with reference to the treatment record of infectious diseases. Molecules 2012; 17: 2955–67. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Barlow DJ, Buriani A, Ehrman T, Bosisio E, Eberini I, Hylands PJ. In-silico studies in Chinese herbal medicines’ research: evaluation of in-silico methodologies and phytochemical data sources and a review of research to date. J Ethnopharmacol 2012; 140: 526–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Chen CY. TCM Database@Taiwan: the world’s largest traditional Chinese medicine database for drug screening in silico. PLoS One 2011; 6: e15939–e15939. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.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 Complement Altern Med 2015; 15: 178–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Memmi G, Filipe SR, Pinho MG, Fu Z, Cheung A. Staphylococcus aureus PBP4 is essential for beta-lactam resistance in community-acquired methicillin-resistant strains. Antimicrob Agents Chemother 2008; 52: 3955–66. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Beck B, Geppert T. Industrial applications of in silico ADMET. J Mol Model 2014; 20: 2322–36. [DOI] [PubMed] [Google Scholar]
- 26.Cos P, Vlietinck AJ, Berghe DV, Maes L. Anti-infective potential of natural products: how to develop a stronger in vitro ‘proof-of-concept’. J Ethnopharmacol 2006; 106: 290–302. [DOI] [PubMed] [Google Scholar]
- 27.Clinical and Laboratory Standards Institute. Performance standards for antimicrobial susceptibility testing, twenty-fourth informational supplement . M100 2014; 34: 128–31. [Google Scholar]
- 28.White RL, Burgess DS, Manduru M, Bosso JA. Comparison of three different in vitro methods of detecting synergy: time-kill checkerboard and E test. Antimicrob Agents Chemother 1996; 40: 1914–18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.AL-Ani I, Zimmermann S, Reichling J, Wink M. Pharmacological synergism of bee venom and melittin with antibiotics and plant secondary metabolites against multi-drug resistant microbial pathogens. Phytomedicine 2015; 22: 245–55. [DOI] [PubMed] [Google Scholar]
- 30.Meng XY, Zhang HX, Mezei M, Cui M. Molecular docking: a powerful approach for structure-based drug discovery. Curr Comput Aided Drug Des 2011; 7: 146–57. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Fong P, Tong HH, Chao CM. In silico prediction of tyrosinase and adenylyl cyclase inhibitors from natural compounds. Nat Prod Commun 2014; 9: 189–94. [PubMed] [Google Scholar]
- 32.Bouley R, Kumarasiri M, Peng Z, Otero LH, Song W, Suckow MA, Schroeder VA, Wolter WR, Lastochkin E, Antunes NT, Pi H, Vakulenko S, Hermoso JA, Chang M, Mobashery S. Discovery of antibiotic (E)-3-(3-carboxyphenyl)-2-(4-cyanostyryl)quinazolin-4(3H)-one. J Am Chem Soc 2015; 137: 1738–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Navratna V, Nadig S, Sood V, Prasad K, Arakere G, Gopal B. Molecular basis for the role of Staphylococcus aureus penicillin binding protein 4 in antimicrobial resistance. J Bacteriol 2010; 192: 134–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Spitzer R, Jain AN. Surflex-Dock: docking benchmarks and real-world application. J Comput Aided Mol Des 2012; 26: 687–99. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Cleves AE, Jain AN. Knowledge-guided docking: accurate prospective prediction of bound configurations of novel ligands using Surflex-Dock. J Comput Aided Mol Des 2015; 29: 485–509. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Fong P, Tong HH, Ng KH, Lao CK, Chong CI, Chao CM. In silico prediction of prostaglandin D2 synthase inhibitors from herbal constituents for the treatment of hair loss. J Ethnopharmacol 2015; 175: 470–80. [DOI] [PubMed] [Google Scholar]
- 37.Clark M, Cramer RD, Van Opdenbosch N. Validation of the general purpose tripos 5.2 force field. J Comput Chem 1989; 10: 982–1012. [Google Scholar]
- 38.Thilagavathi R, Mancera RL. Ligand-protein cross-docking with water molecules. J Chem Inf Model 2010; 50: 415–21. [DOI] [PubMed] [Google Scholar]
- 39.Lemmon G, Meiler J. Towards ligand docking including explicit interface water molecules. PLoS One 2013; 8: e67536–e67536. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Lexa KW, Carlson HA. Protein flexibility in docking and surface mapping. Q Rev Biophys 2012; 45: 301–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Kumar KM, Anbarasu A, Ramaiah S. Molecular docking and molecular dynamics studies on beta-lactamases and penicillin binding proteins. Mol Biosyst 2014; 10: 891–900. [DOI] [PubMed] [Google Scholar]
- 42.Irwin JJ, Sterling T, Mysinger MM, Bolstad ES, Coleman RG. ZINC: a free tool to discover chemistry for biology. J Chem Inf Model 2012; 52: 1757–68. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Turk S, Verlaine O, Gerards T, Zivec M, Humljan J, Sosic I, Amoroso A, Zervosen A, Luxen A, Joris B, Gobec S. New noncovalent inhibitors of penicillin-binding proteins from penicillin-resistant bacteria. PLoS One 2011; 6: e19418–e19418. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Wang Q, Park J, Devkota AK, Cho EJ, Dalby KN, Ren P. Identification and validation of novel PERK inhibitors. J Chem Inf Model 2014; 54: 1467–75. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Richter HG, Angehrn P, Hubschwerlen C, Kania M, Page MG, Specklin JL, Winkler FK. Design synthesis and evaluation of 2 beta-alkenyl penam sulfone acids as inhibitors of beta-lactamases. J Med Chem 1996; 39: 3712–22. [DOI] [PubMed] [Google Scholar]
- 46.ACD/Percepta version 14.0. Advanced Chemistry Development Inc, Toronto, ON, Canada. www.acdlabs.com. 2015.
- 47.Leeson P. Drug discovery: chemical beauty contest. Nature 2012; 481: 455–6. [DOI] [PubMed] [Google Scholar]
- 48.Wang J, Hou T. Advances in computationally modeling human oral bioavailability. Adv Drug Deliv Rev 2015; 86: 11–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Xu C, Cheng F, Chen L, Du Z, Li W, Liu G, Lee P.W, Tang Y. In silico prediction of chemical Ames mutagenicity. J Chem Inf Model 2012; 52: 2840–47. [DOI] [PubMed] [Google Scholar]
- 50.Jing Y, Easter A, Peters D, Kim N, Enyedy IJ. In silico prediction of hERG inhibition. Future Med Chem 2015; 7: 571–86. [DOI] [PubMed] [Google Scholar]
- 51.Livermore DM. Antibiotic resistance in staphylococci. Int J Antimicrob Agents 2000; 16: S3–10. [DOI] [PubMed] [Google Scholar]
- 52.Ozcelik B, Orhan I, Toker G. Antiviral and antimicrobial assessment of some selected flavonoids. Z Naturforsch C 2006; 61: 632–8. [DOI] [PubMed] [Google Scholar]
- 53.Zhang Q, Fan D, Xiong B, Kong L, Zhu X. Isolation of new flavan-3-ol and lignan glucoside from Loropetalum chinense and their antimicrobial activities. Fitoterapia 2013; 90: 228–32. [DOI] [PubMed] [Google Scholar]
- 54.Falcão-Silva VS, Silva DA, Souza V, Siqueira-Junior JP. Modulation of drug resistance in Staphylococcus aureus by a kaempferol glycoside from Herissantia tiubae (malvaceae). Phytother Res 2009; 23: 1367–70. [DOI] [PubMed] [Google Scholar]
- 55.Scheife RT. Protein binding: what does it mean? DICP 1989; 23: S27–31. [DOI] [PubMed] [Google Scholar]
- 56.Roder BL, Frimodt-Moller N, Espersen F, Rasmussen SN. Dicloxacillin and flucloxacillin: pharmacokinetics protein binding and serum bactericidal titers in healthy subjects after oral administration. Infection 1995; 23: 107–12. [DOI] [PubMed] [Google Scholar]
- 57.Lv PC, Li HQ, Xue JY, Shi L, Zhu HL. Synthesis and biological evaluation of novel luteolin derivatives as antibacterial agents. Eur J Med Chem 2009; 44: 908–14. [DOI] [PubMed] [Google Scholar]
- 58.Sato Y, Suzaki S, Nishikawa T, Kihara M, Shibata H, Higuti T. Phytochemical flavones isolated from Scutellaria barbata and antibacterial activity against methicillin-resistant Staphylococcus aureus. J Ethnopharmacol 2000; 72: 483–88. [DOI] [PubMed] [Google Scholar]
- 59.Zhang X, Zuo G, Wang T, Zhang Y, Wang G, Luo X. In vitro study on synergetic anti-MRSA effects of luteolin combined with antibacterial drugs. Prog Pharm Sci 2012; 4: 173–9. [Google Scholar]
- 60.Reynolds DP, Lanevskij K, Japertas P, Didziapetris R, Petrauskas A. Ionization-specific analysis of human intestinal absorption. J Pharm Sci 2009; 98: 4039–54. [DOI] [PubMed] [Google Scholar]
- 61.Gupta S, Kesarla R, Omri A. Formulation strategies to improve the bioavailability of poorly absorbed drugs with special emphasis on self-emulsifying systems. ISRN Pharm 2013; 2013: 848043–848043. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Hennebelle T, Sahpaz S, Gressier B, Joseph H, Bailleul F. Antioxidant and neurosedative properties of polyphenols and iridoids from Lippia alba. Phytother Res 2008; 22: 256–8. [DOI] [PubMed] [Google Scholar]
- 63.Mohler H, Schoch P, Richards JG, Haring P, Takacs B. Structure and location of a GABA-A receptor complex in the central nervous system. J Recept Res 1987; 7: 617–28. [DOI] [PubMed] [Google Scholar]
- 64.Zhang SX, Yang PW, Zhang DC, Dong WQ, Zhang FH, Sun YM. Pharmacokinetics tissue distribution and metabolism of novel DNA topoisomerase I inhibitor yuanhuacine in rabbit. Xenobiotica 2009; 39: 273–81. [DOI] [PubMed] [Google Scholar]
- 65.Mitscher LA. Bacterial topoisomerase inhibitors: quinolone and pyridone antibacterial agents. Chem Rev 2005; 105: 559–92. [DOI] [PubMed] [Google Scholar]
- 66.Navratna V, Nadig S, Sood V, Prasad K, Arakere G, Gopal B. Molecular basis for the role of Staphylococcus aureus penicillin binding protein 4 in antimicrobial resistance. J Bacteriol 2010; 192: 134–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.




