Next Article in Journal
13C CP MAS NMR and DFT Studies of 6-Chromanyl Ethereal Derivatives
Next Article in Special Issue
Neutrophil Immunomodulatory Activity of (−)-Borneol, a Major Component of Essential Oils Extracted from Grindelia squarrosa
Previous Article in Journal
Maleimide-Functionalized Liposomes: Prolonged Retention and Enhanced Efficacy of Doxorubicin in Breast Cancer with Low Systemic Toxicity
Previous Article in Special Issue
Antitumor Effect of Glandora rosmarinifolia (Boraginaceae) Essential Oil through Inhibition of the Activity of the Topo II Enzyme in Acute Myeloid Leukemia
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Preliminary Study on Phytochemical Constituents and Biological Activities of Essential Oil from Myriactis nepalensis Less.

1
Sdu-Anu Joint Science College, Shandong University, Weihai 264209, China
2
Marine College, Shandong University, Weihai 264209, China
*
Author to whom correspondence should be addressed.
Molecules 2022, 27(14), 4631; https://doi.org/10.3390/molecules27144631
Submission received: 28 June 2022 / Revised: 16 July 2022 / Accepted: 18 July 2022 / Published: 20 July 2022
(This article belongs to the Special Issue Chemical Composition and Bioactivities of Essential Oils)

Abstract

:
In response to the need for novel therapeutic strategies to combat the development of microbial resistance, plant essential oils may represent a promising alternative source. This study set out to characterize the chemical composition and assess the antibacterial potential of Myriactis nepalensis Less. essential oil (MNEO). Essential oil isolated from M. nepalensis by hydrodistillation was analyzed using a GC–MS technique. The antibacterial properties of MNEO alone and combined with antibiotics (chloramphenicol and streptomycin) were tested via the disc diffusion, microbroth dilution, and checkerboard methods. MNEO was represented by oxygenated sesquiterpenes (60.3%) and sesquiterpene hydrocarbons (28.6%), with caryophyllene oxide, spathulenol, humulene epoxide II, β-elemene, neointermedeol, and β-caryophyllene as the main compounds. MNEO exhibited a strong antibacterial effect against Gram-positive bacteria, with MIC and MBC values of 0.039 mg/mL and 0.039–0.156 mg/mL, respectively, and synergistic effects were observed in both combinations with chloramphenicol and streptomycin. Furthermore, the antibiofilm and cytotoxic activities of MNEO were also evaluated. The crystal violet assay was used for quantification of Staphylococcus aureus biofilm formation, and an MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide) assay was conducted to determine cell viability. The results revealed MNEO could dose-dependently inhibit Staphylococcus aureus biofilm formation and possessed potential cytotoxic on both normal and cancer cells (IC50 values from 13.13 ± 1.90 to 35.22 ± 8.36 μg/mL). Overall, the results indicate that MNEO may have promising applications in the field of bacterial infections.

1. Introduction

As resistant pathogens develop and spread, antibiotic resistance poses a major threat to the effective treatment of a growing number of infections caused by bacteria [1]. The establishment of resistance among bacterial infections is acknowledged as a major global public health problem [2,3]. Therefore, it is imperative to search for novel antimicrobial agents and alternative therapeutic strategies. The study of plant-derived natural products plays an extremely essential part in the development of new therapeutic agents [4]. Antimicrobial compounds that come from plants have been studied closely in the last few years [5].
Under growth conditions, plants produce a large number of secondary metabolites to adapt to biotic and abiotic stresses [6,7]; these metabolites are now well documented to exhibit broad biological functions and play important roles in the plant defense system against pathogenic assaults and environmental factors [8,9,10,11]. Among the plants’ secondary metabolites, essential oils could be good candidates for new antimicrobial agents, as researchers have demonstrated that individual essential oils and their separated components exhibit antibacterial properties against a broad spectrum of microorganisms [12,13]. Essential oils are among the most prevalent naturally occurring antibacterial substances, and they are frequently employed as additives, preservatives, and decontaminants [14]. In addition, combination therapy employing both essential oils and classic antimicrobial agents has proven effective in preventing the development of resistant strains [15]. When such interactions result in synergistic effects, they can also be employed to improve therapy efficiency. Another benefit of such combinations is that lower doses are employed, which reduces side effects and treatment costs [16].
Myriactis nepalensis Less. (Compositae) is a perennial herb belonging to the genus Myriactis (comprising approximately 10 species) and distributed throughout Southwestern and Southern China, growing at altitudes between 1250 and 3400 m. The plant is 15–100 cm high with a short and procumbent rhizome, erect stem, and simple leaf, with a capitulum type of inflorescence. The flowering and fruiting period is from April to November [17]. To the best of our knowledge, no prior studies have investigated the chemical composition and biological activities of the plant Myriactis nepalensis Less. Therefore, the present study aimed to characterize the essential oil composition and evaluate the antibacterial properties of MNEO alone and combined with traditional antibiotics, as well as its antibiofilm and cytotoxic activities.

2. Results

2.1. Chemical Composition of MNEO

The isolation yield for MNEO was 0.68% (w/w based on air-dried plant material). The phytochemical composition determined by GC-MS is presented in Table 1. Altogether, 78 components, accounting for 96.4% of the total composition of the MNEO, were identified (Table 1). Sesquiterpene compounds dominated the chemical composition of MNEO (88.9%), with oxygenated and hydrocarbon components accounting for 60.3% and 28.6%, respectively. The main components were found to be caryophyllene oxide (10.2%), spathulenol (7.3%), and humulene epoxide II (7.2%), followed by β-elemene (6.1%), neointermedeol (4.5%), and β-caryophyllene (4.1%).

2.2. Antibacterial Activity of MNEO

The antibacterial properties of MNEO against ATCC strains were evaluated using the disc agar diffusion method to determine the diameter of inhibition zones (DIZ) and the microtiter broth dilution method to determine the MIC and MBC. Based on the susceptibility testing results (Table 2), the MNEO demonstrated a variable level of inhibitory effect against all bacteria tested. With DIZs ranging from 8.3 ± 0.9 mm (in Escherichia coli) to 16.7 ± 1.7 mm (in Bacillus subtilis), the disk diffusion assay demonstrated that MNEO exhibited broad-spectrum antimicrobial action. The MNEO showed low activity on the Gram-negative bacteria Pseudomonas aeruginosa (MIC = 0.625 mg/mL, MBC = 2.500 mg/mL) and E. coli (MIC = 1.250 mg/mL); however, the MBC for E. coli was not determined when the concentration of MNEO reached the maximum tested. Conversely, M. nepalensis EO presented significant bacteriostatic effects against Gram-positive bacteria (MIC values of 0.039–0.078 mg/mL and MBC values of 0.039–0.156 mg/mL). In addition, based on the MBC/MIC ratios, a bactericidal effect was confirmed for the M. nepalensis oil against B. subtilis, S. aureus, and P. larvae (ratios ≤ 4) [18].

2.3. Combined Effect of MNEO with Chloramphenicol and Streptomycin

Combination therapy employing both essential oils and classic antibiotics has proven effective in preventing the development of resistant strains and improving therapy efficiency [15,16]. Therefore, the combined effect of MNEO with conventional antibiotics (chloramphenicol and streptomycin) against four pathogens was investigated by using a checkerboard microtiter assay [19]. The fractional inhibitor concentration index (FICI) values were calculated to determine the interaction of MNEO with chloramphenicol and streptomycin, and these are reported in Table 3 and Table 4, respectively. The FICI values in association with chloramphenicol and streptomycin were, respectively, in the range of 0.13–0.56 and 0.12–0.50 against all tested bacterial strains. The mixtures of essential oil and chloramphenicol exhibited either synergism (FICI ≤ 0.5) or an additional synergistic effect (0.5 < FICI ≤ 1). The combined application of MNEO and streptomycin showed synergistic interaction against all the tested bacterial strains (FICIs of 0.12–0.50). The decrease in antibiotic MIC values in the presence of essential oil ranged from 4- to 16-fold. This suggested that the antibiotic activity of chloramphenicol and streptomycin was enhanced when combined with MNEO.

2.4. Antibiofilm Activity of MNEO

The microtiter-plate technique was conducted to assess S. aureus biofilm formation in the presence of MNEO, and a crystal violet (CV) staining assay based on a previously reported methodology was used to quantify biofilm biomass [20]. As shown in Figure 1, S. aureus demonstrated a statistically significant reduction in biofilm development when exposed to M. nepalensis essential oil. When M. nepalensis essential oil at concentrations of 1/16 MIC to 4 MIC was used, 7–88% of S. aureus biofilm formation was inhibited.

2.5. Cytotoxicity of MNEO

To study the possible cytotoxic activity of MNEO, in vitro metabolic analysis using an MTT colorimetric assay on four human cancer cell lines (colorectal carcinoma HCT-116 cells, hepatocellular carcinoma HepG2 cells, lung adenocarcinoma A-549 cells, and breast cancer MCF7 cells), and one normal cell line (HL-7702 human liver cells) was performed. The cytotoxic activity of MNEO is shown in Table 5 as IC50 values. The IC50 values obtained from MNEO were 19.53 ± 2.84 μg/mL, 13.13 ± 1.90 μg/mL, 19.19 ± 3.08 μg/mL, and 35.22 ± 8.36 μg/mL for HepG2, MCF-7, A-549, and HCT-116 cells, respectively. For HL-7702, the IC50 of MNEO was determined as 19.14 ± 0.63 μg/mL.

3. Discussion

As one of the main sources of phytochemical active ingredients, essential oils have been the object of increased interest from researchers, especially regarding their phytochemical profiles and biological activities. Essential oils comprise a range of compounds from diverse classes, including phenolics, terpenoids, aldehydes, ketones, ethers, and epoxides [21,22,23]. These complex mixtures contribute to the broad bioactivity of essential oils. In the present study, the major fraction of MNEO was represented by oxygenated sesquiterpenes (60.3%) and sesquiterpene hydrocarbons (28.6%), with caryophyllene oxide (10.2%), spathulenol (7.3%), humulene epoxide II (7.2%), β-elemene (6.1%), neointermedeol (4.5%), and β-caryophyllene (4.1%) as the main compounds. Among them, β-caryophyllene oxide, β-spathulenol, β-elemene, and β-caryophyllene are the most studied. β-Caryophyllene oxide is frequently found to co-occur with natural bicyclic sesquiterpene β-caryophyllene in many plant essential oils as its metabolite. Several reports have shown that both β-caryophyllene oxide and β-caryophyllene possess significant anti-cancer properties, inhibit the growth and proliferation of numerous cancer cell lines [24,25,26,27,28], and are able to enhance the antiproliferative effect of classical anticancer agents, such as 5-fluorouracil, oxaliplatin, paclitaxel, and doxorubicin [29,30,31,32]. In addition, both of these compounds were found to exhibit anti-inflammatory [33,34], analgesic [26,35], antifungal [36,37], and antibacterial activities [28,38,39].
Spathulenol, a tricyclic sesquiterpenoid with an aromadendrane carbon skeleton, due to its broad spectrum of biological properties, has also been extensively studied in recent years [40], showing antioxidant, anti-inflammatory, antiproliferative, and antimycobacterial activities [41]; insecticidal efficacy [40]; and immunomodulatory response effects [42]. In addition, spathulenol was also demonstrated to be a significantly effective repellent against Aedes aegypti and Anopheles stephensi [43]. Furthermore, a recent in vivo study showed the acute and persistent antiedematogenic, antihyperalgesic, and anxiolytic activities of spathulenol [44].
Another important compound in M. nepalensis essential oil is β-elemene, a noncytotoxic broad-spectrum antitumor drug utilized in Chinese traditional medicine (TCM) for more than 20 years to treat various cancer types, including lung, gastric, cervical, breast, liver, and bladder cancers, etc., including tyrosine kinase inhibitor (TKI)-resistant non-small-cell lung cancer [45,46,47,48,49,50,51,52,53]. Apart from its anticancer activity, β-elemene also possesses antioxidative and anti-inflammatory activities [54,55].
The results obtained from the antibacterial assays demonstrated the promising potential growth-inhibiting effect of MNEO against the tested gram-positive bacteria strains. The tested gram-negative stains were less sensitive to MNEO than tested gram-positive strains, which may be due to the structural differences in their outer membranes. The gram-negative outer membrane is surrounded by an outer layer composed of lipopolysaccharide, which provides an effective barrier of permeability to prevent the diffusion of hydrophobic essential oils [56,57]. The activity of MNEO could be contributed to by the presence of the main components, such as caryophyllene oxide, spathulenol, β-caryophyllene, (Z)-α-santalol, and α-humulene, which exhibit moderate or strong antimicrobial activity [28,39,41,58,59,60,61,62,63,64,65]. However, the possible synergistic effect of all components in the essential oil should also be considered.
Combination therapy using natural and antibacterial agents has been reported as an effective strategy to combat the development of bacterial resistance. Thus, we evaluated the combined effect of MNEO and two conventional antibiotics, namely, chloramphenicol and streptomycin. The most interesting result was obtained for P. aeruginosa, for which the MIC value of chloramphenicol was found to decrease from 15.60 to 0.98 μg/mL (FICI = 0.13), and that of streptomycin decreased from 1.95 to 0.12 μg/mL (FICI = 0.12). The most prevalent nosocomial pathogen, P. aeruginosa, is intrinsically resistant to numerous drug classes and has the ability to acquire resistance to all available treatment options [66]. Reduced cell permeability, efflux pumps, modifications to the target enzymes, and antibiotic inactivation are some of the primary mechanisms of drug resistance development [67,68]. Moreover, a promising result was also obtained against S. aureus (FICI = 0.12)—a 16-fold reduction in streptomycin’s MIC was observed when used with MNEO. The considerable synergistic interaction observed between chloramphenicol and M. nepalensis essential oil against the gram-negative P. aeruginosa and E. coli is also worthy of note. However, the combination of M. nepalensis essential oil and chloramphenicol showed only additive interaction when tested against S. aureus, as FICI = 0.56 for this combination; it should be considered that the additive manner is similar to a synergistic one, since lower dosages of drugs produce desirable results with additive effects [69].
As far as the microtiter plate biofilm assay results are concerned, interestingly, M. nepalensis essential oil revealed a significant effect in inhibiting S. aureus biofilm formation at the tested concentrations. Biofilm refers to multicellular surface-attached communities of bacteria embedded in a self-produced extracellular matrix consisting of polysaccharides, protein, and DNA [70]. It is widely acknowledged that bacteria are shielded from antibiotics and the hostile environment of the host by living in biofilms. There is proof that planktonic cells are 1000 times more susceptible to conventional medications than cells in biofilms on biotic or abiotic surfaces. [71,72,73,74,75]. Biofilm is difficult to eliminate once formed, systemic infections are more likely to occur, and the bacteria also become more resistant to treatment with traditional antibiotics [76,77,78]. Among bacteria, staphylococci, especially S. aureus, are the major causes of biofilm-associated infections [79,80]. S. aureus can colonize and develop biofilms in a variety of host environments [79,80,81]. Bacterial cells in biofilms may evade the host immunological response and tolerate much higher concentrations of antimicrobials compared with planktonic bacteria, making biofilm-related infections particularly difficult to eradicate [82,83,84].
It is worth noting that the biofilm responses to M. nepalensis essential oil were demonstrated to be concentration-dependent; even at sub-MICs, the essential oil exhibits inhibitory efficacy on biofilm formation. On the contrary, several studies have demonstrated that biofilm formation can still occur in the presence of sub-inhibitory levels of several conventional antibiotic agents [85,86,87,88,89,90]. This phenomenon was not observed with M. nepalensis essential oil, which emphasizes the fact that the essential oil has good therapeutic potential against biofilms.
Given the presence of β-elemene, caryophyllene oxide, β-caryophyllene, and β-spathulenol and their related cytotoxic effects, we also analyzed the metabolic activity of M. nepalensis essential oil on four cancer cell lines, as well as its cytotoxicity on a non-cancerous cell line, HL-7702. As expected, the M. nepalensis essential oil possessed significant cytotoxic activity on HepG2, MCF-7, and A-549, with IC50 values less than 20 μg/mL, and moderate activity on HCT-116. Unfortunately, the essential oil showed a cytotoxic effect on HL-7702 cells, with IC50 values of 19.14 ± 0.63 μg/mL. The cytotoxic activity of M. nepalensis essential oil could be attributed to the above-mentioned main compounds, as their cytotoxic effects on numerous cell lines have been widely reported [24,25,26,27,28,41,45,46,47,48,49,50,51,52,53,91].

4. Materials and Methods

4.1. Plant Material

The plant Myriactis nepalensis was collected in August 2020 from Badong County in Hubei Province, China. Plant material was identified by Professor Hong Zhao (Shandong University, Weihai, China). Samples (voucher specimen NO.020017) are stored in the herbarium of the Department of Biological Sciences, Shandong University, Weihai, China.

4.2. Extraction of Essential Oils

The aerial parts of M. nepalensis (300 g) were submitted to hydrodistillation using a Clevenger-type apparatus for 3.5 h to extract the essential oil, then dried with anhydrous Na2SO4 and kept at 4 °C until use.

4.3. Identification of Oil Components

The obtained essential oil was characterized through GC/MS (Agilent Technology 6890/5975C, St. Clara, CA, USA), and the relative percentage amounts of compounds in MNEO were assessed by GC/FID. An HP-5MS capillary column (30 m × 0.25 mm × 0.25 μm, Agilent, St. Clara, CA, USA) was used for the separation. Helium was used as the carrier gas with a flow rate of 1.3 mL/min. A 0.2 μL sample of essential oil was injected with split ratio 50:1, and the temperature was programmed from 60 °C (1 min) to 240 °C (12 min) at a rate of 8 °C/min. MS parameters: EI mode at 70 eV; mass range 50 to 550 amu. The identification of compounds of M. nepalensis oil was carried out by comparing their retention indices relative to C7-C30 n-alkanes and mass spectra with those reported in the literature and by comparing their mass spectra with the NIST and Wiley libraries [92,93,94].

4.4. Antibacterial Susceptibility Test

The strains studied are five reference strains of microorganisms from American Type Culture Collection (ATCC) and are representative of gram-positive and -negative strains—namely Staphylococcus aureus (ATCC 6538), Bacillus subtilis (ATCC 6633), and Paenibacillus larvae (ATCC 9545) representative of the gram-positive; and Escherichia coli (ATCC 25922) and Pseudomonas aeruginosa (ATCC 27853) with characteristics of gram-negative bacteria. The bacterial strains were cultured overnight at 37 °C on Mueller–Hinton (MH) medium before each experimental procedure. Before conducting susceptibility assays, the bacterial suspension was standardized versus 0.5 McFarland turbidimetrically and diluted with MH broth medium to reach the required concentration for each procedure.
A disc agar diffusion assay was applied to antibacterial susceptibility tests [95]. Briefly, filter paper discs were soaked with 20 µL of essential oil (10 mg/mL) or chloramphenicol positive control (1 mg/mL), and placed in triplicate on the plates in which the bacteria were inoculated. The plates were then kept at 37 °C for 24 h in an incubator. The results were determined by measuring the diameter of the zone of growth inhibition (DIZ, in millimeters) and are presented as the means of three measurements.

4.5. MIC and MBC Determination by a Microdilution Broth Method

The minimum inhibitory concentrations (MICs) of the essential oil and antibiotics were assessed according to the recommendations of CLSI, using the microdilution broth method in 96-well plates [96]. Stock solutions of the essential oil and antibiotics were prepared with dimethyl sulfoxide (DMSO). In 96-well microtiter plates, 100 µL of essential oil or antibiotic was serially two-fold diluted in Mueller–Hinton broth, and an equal volume of approximately 106 CFU× mL−1 bacterial suspension was added in each well. A mixture of culture medium and bacterial suspension acted as a growth control. After 24 h of incubation, to stain the bacteria, 1% 2,3,5-triphenyl tetrazolium chloride aqueous solution was added to each well (20 µL per well). The plates were incubated at 37 °C for 30 min, and the MIC values were visually determined as the minimal concentrations that did not produce a red color.
Samples (100 μL) from the MIC experiment wells with no color change were placed on MH agar plates and incubated for 18–24 h at 37 °C. The minimum bactericidal concentration (MBC) was defined as the lowest concentration at which no bacterial growth was observed.

4.6. In Vitro Synergistic Antibacterial Activity by the Checkerboard Method

To evaluate the combined action of MNEO and antibiotics (chloramphenicol and streptomycin), the microbroth checkerboard method was conducted to determine the fractional inhibitory concentration (FIC) index (FICI) [19]. The concentrations tested for each essential oil and antibiotic were selected from 4 × MIC to 1/32 × MIC. A 50 μL volume of essential oil was added at decreasing concentrations into the columns of the 96-well plates, and 50 μL of antibiotic was similarly distributed among the rows. Volumes of 100 μL of bacterial suspensions (106 CFU/mL) were then added to each well. The plates were incubated at 37 °C for 24 h. The FIC values were obtained following the same method for obtaining the MIC values, detailed above. The interaction of the association between the MNEO and antibiotics was evaluated by determining the FICI using the following equation:
FICI = MIC   of   EO   in   combination MIC   of   EO   alone + MIC   of   antibiotic   in   combination MIC   of   antibiotic   alone
The results were considered synergistic if FICI ≤ 0.5 and additive when 0.5 < FICI < 1 [97].

4.7. Biofilm Formation Inhibition

The potential of essential oil to inhibit biofilm formation by S. aureus (ATCC 6538) was assessed using the microtiter-plate technique [20,98]. The plates were assembled in a process similar to the MIC test. A suspension of approximately 106 CFU/mL S. aureus and different concentrations of essential oil (1/4 × MIC to 4 × MIC) were added to each well (100 μL per well) of a 96-well microtiter plate, with 3 parallel wells at each concentration, and the plate was incubated at 37 °C for 24 h. The nontreated bacterial suspension was used as the negative control. After biofilm formation, the supernatant was discarded and the wells were washed thrice with PBS (phosphate-buffered saline) to remove the planktonic and weakly attached cells. The remaining attached biofilms were fixed with 200 μL of methanol per well for 15 min. After discarding the supernatant, each well in the plate was stained with crystal violet (2%) for 10 min and washed with distilled water until the water was colorless. The residual CV, which represented the quantity of biofilm, was dissolved in 95% ethanol (200 μL). Finally, an ELISA microtiter plate reader was utilized to determine the optical density (OD) of each well at 570 nm.

4.8. Cytotoxic Activity Evaluation

Four human cancer cell lines (colorectal carcinoma HCT-116 cells, hepatocellular carcinoma HepG2 cells, lung adenocarcinoma A-549 cells, and breast cancer MCF7 cells) and one normal human cell line (HL-7702 human liver cells) were obtained from the Shanghai Institute for Biological Sciences (SIBS, Shanghai, China) and were developed in DMEM and RPMI 1640 (for HL-7702 cells) culture medium supplemented with 2 mM glutamine, 10% fetal bovine serum, 100 units/mL of streptomycin, and 100 units/mL of penicillin, cultured at 37 °C in 5% CO2. Cells were detached from the monolayer using 0.25% trypsin for 5 min once cells had grown to near confluence.
The metabolic activity of the cells was detected by an MTT-based assay [99]. Briefly, cells were seeded into 96-well microtiter plates (5 × 104 cells/well) and incubated for 24 h for cell adherens. The essential oil was first dissolved in DMSO (final concentration was 0.1%) and then diluted in culture medium. Subsequently, the cells were treated with essential oil or positive control (doxorubicin) for 48 h. Then, 20 μL of MTT (5 mg/mL) solution was added to all wells for 4 h to form formazan, which was then dissolved in 150 μL of DMSO. Absorbances were read at 570 nm. Cell viability in response to treatment was calculated as a percentage of control cells treated with DMSO at the final concentration 0.1%. The results are expressed as 50% inhibitory concentration of cell growth (IC50) values.

4.9. Statistical Analysis

All the experiments were carried out in triplicate and the data were presented as the mean ± SD. Data were statistically analyzed using GraphPad Prism 8.0 software (GraphPad Software, Inc., San Diego, CA, USA). One-way analysis of variance (ANOVA) was used to analyze the differences among the treatments, and the level of significance was 0.05.

5. Conclusions

The present study investigated the components of M. nepalensis essential oil, as well as the potential antibacterial activities of the essential oil, for the first time. The results show that M. nepalensis essential oil is rich in caryophyllene oxides (10.2%), spathulenol (7.3%), and humulene epoxide II (7.2%), followed by β-elemene (6.1%), neointermedeol (4.5%), and β-caryophyllene (4.1%). The essential oil of M. nepalensis demonstrated antibacterial activities against representative Gram-positive and -negative strains. Furthermore, the ability of M. nepalensis essential oil combined with traditional antibiotics to increase the sensitivity of the tested strains to chloramphenicol and streptomycin was investigated. Inhibitory activity was observed against S. aureus biofilm formation. Additionally, cytotoxic potential of M. nepalensis essential oil was observed on both normal and cancer cells. Considering all the results obtained, M. nepalensis essential oil could be a promising alternative for the treatment of various pathogenic bacterial strains; however, further detailed investigations in vitro and in vivo on the biological effects of this essential oil are required to elucidate the mechanism of action and evaluate safety.

Author Contributions

Conceptualization, X.X.; methodology, X.X., J.F. and Y.G.; formal analysis, X.X., Y.G. and J.F.; investigation, X.X. and J.F.; resources, X.X. and Y.G.; data curation, X.X. and J.F.; writing—original draft preparation, X.X. and J.F.; writing—review and editing, X.X. and J.F.; visualization, X.X. and J.F.; supervision, X.X.; project administration, X.X.; funding acquisition, X.X. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The data are contained in this article.

Acknowledgments

The authors are thankful to Xia Li for technical support.

Conflicts of Interest

The authors declare no conflict of interest.

Sample Availability

Samples of the essential oils are available from the authors.

References

  1. Levy, S.B.; Marshall, B. Antibacterial resistance worldwide: Causes, challenges and responses. Nat. Med. 2004, 10, 122–129. [Google Scholar] [CrossRef] [PubMed]
  2. Mandal, S.; Pal, N.K.; Chowdhury, I.H.; Debmandal, M. Antibacterial activity of ciprofloxacin and trimethoprim, alone and in combination, against Vibrio cholerae O1 Biotype El Tor serotype Ogawa isolates. Pol. J. Microbiol. 2009, 58, 57–60. [Google Scholar] [PubMed]
  3. Breijyeh, Z.; Jubeh, B.; Karaman, R. Resistance of gram-negative bacteria to current antibacterial agents and approaches to resolve it. Molecules 2020, 25, 1340. [Google Scholar] [CrossRef] [Green Version]
  4. Newman, D.J.; Cragg, G.M. Natural products as sources of new drugs from 1981 to 2014. J. Nat. Prod. 2016, 79, 629–661. [Google Scholar] [CrossRef] [Green Version]
  5. Ibrahim, H.; Elgindi, M.; Ibrahim, R.; El-Hosari, D. Antibacterial activities of triterpenoidal compounds isolated from Calothamnus quadrifidus leaves. BMC Complement. Altern. Med. 2019, 19, 102. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Yang, L.; Wen, K.S.; Ruan, X.; Zhao, Y.X.; Wei, F.; Wang, Q. Response of plant secondary metabolites to environmental factors. Molecules 2018, 23, 762. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Kessler, A.; Kalske, A. Plant secondary metabolite diversity and species interactions. Annu. Rev. Ecol. Evol. Syst. 2018, 49, 115–138. [Google Scholar] [CrossRef]
  8. Erb, M.; Kliebenstein, D.J. Plant secondary metabolites as defenses, regulators, and primary metabolites: The blurred functional trichotomy. Plant Physiol. 2020, 184, 39–52. [Google Scholar] [CrossRef]
  9. Piasecka, A.; Jedrzejczak-Rey, N.; Bednarek, P. Secondary metabolites in plant innate immunity: Conserved function of divergent chemicals. New Phytol. 2015, 206, 948–964. [Google Scholar] [CrossRef]
  10. Isah, T. Stress and defense responses in plant secondary metabolites production. Biol. Res. 2019, 52, 39. [Google Scholar] [CrossRef] [Green Version]
  11. Kesselmeier, J.; Staudt, M. Biogenic volatile organic compounds (VOC): An overview on emission, physiology and ecology. J. Atmos. Chem. 1999, 33, 23–88. [Google Scholar] [CrossRef]
  12. Wińska, K.; Mączka, W.; Łyczko, J.; Grabarczyk, M.; Czubaszek, A.; Szumny, A. Essential oils as antimicrobial agents—Myth or real alternative? Molecules 2019, 24, 2130. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. El-Tarabily, K.A.; El-Saadony, M.T.; Alagawany, M.; Arif, M.; Batiha, G.E.; Khafaga, A.F.; Elwan, H.A.; Elnesr, S.S.; Abd El-Hack, M.E. Using essential oils to overcome bacterial biofilm formation and their antimicrobial resistance. Saudi J. Biol. Sci. 2021, 28, 5145–5156. [Google Scholar] [CrossRef] [PubMed]
  14. Burt, S. Essential oils: Their antibacterial properties and potential applications in foods—A review. Int. J. Food Microbiol. 2004, 94, 223–253. [Google Scholar] [PubMed]
  15. Cho, T.J.; Park, S.M.; Yu, H.; Seo, G.H.; Kim, H.W.; Kim, S.A.; Rhee, M.S. Recent advances in the application of antibacterial complexes using essential oils. Molecules 2020, 25, 1752. [Google Scholar] [CrossRef]
  16. Visan, D.C.; Oprea, E.; Radulescu, V.; Voiculescu, I.; Biris, I.A.; Cotar, A.I.; Saviuc, C.; Chifiriuc, M.C.; Marinas, I.C. Original contributions to the chemical composition, microbicidal, virulence-arresting and antibiotic-enhancing activity of essential oils from four coniferous species. Pharmaceuticals 2021, 14, 1159. [Google Scholar] [CrossRef]
  17. Lin, R.; Chen, Y.L. Flora of China; Science Press: Beijing, China, 1985; Volume 74, pp. 87–88. [Google Scholar]
  18. Levison, M.E. Pharmacodynamics of antimicrobial drugs. Infect. Dis. Clin. N. Am. 2004, 18, 451–465. [Google Scholar] [CrossRef]
  19. Moody, J. Synergism testing: Broth microdilution checkerboard and broth macrodilution method. In Clinical Microbiology Procedures Handbook, 1st ed.; American Society for Microbiology: Washington, DC, USA, 2004; Volume 18, pp. 1–28. [Google Scholar]
  20. Stepanović, S.; Vuković, D.; Dakić, I.; Savić, B.; Švabić-Vlahović, M. A modified microtiter-plate test for quantification of staphylococcal biofilm formation. J. Microbiol. Methods 2000, 40, 175–179. [Google Scholar] [CrossRef]
  21. Wani, A.R.; Yadav, K.; Khursheed, A.; Rather, M.A. An updated and comprehensive review of the antiviral potential of essential oils and their chemical constituents with special focus on their mechanism of action against various influenza and coronaviruses. Microb. Pathog. 2021, 152, 104620. [Google Scholar] [CrossRef]
  22. Dagli, N.; Dagli, R.; Mahmoud, R.S.; Baroudi, K. Essential oils, their therapeutic properties, and implication in dentistry: A review. J. Int. Soc. Prev. Community Dent. 2015, 5, 335–340. [Google Scholar] [CrossRef] [Green Version]
  23. Di Sotto, A.; Mancinelli, R.; Gullì, M.; Eufemi, M.; Mammola, C.L.; Mazzanti, G.; Di Giacomo, S. Chemopreventive potential of caryophyllane sesquiterpenes: An overview of preliminary evidence. Cancers 2020, 12, 3034. [Google Scholar] [CrossRef] [PubMed]
  24. Jun, N.J.; Mosaddik, A.; Moon, J.Y.; Ki-Chang, J.; Dong-Sun, L.; Ahn, K.S.; Cho, S.K. Cytotoxic activity of β-caryophyllene oxide isolated from jeju guava (Psidium cattleianum sabine) leaf. Rec. Nat. Prod. 2011, 5, 242–246. [Google Scholar]
  25. Park, K.R.; Nam, D.; Yun, H.M.; Lee, S.G.; Jang, H.J.; Sethi, G.; Cho, S.K.; Ahn, K.S. β-Caryophyllene oxide inhibits growth and induces apoptosis through the suppression of PI3K/AKT/mTOR/S6K1 pathways and ROS-mediated MAPKs activation. Cancer Lett. 2011, 312, 178–188. [Google Scholar] [CrossRef] [PubMed]
  26. Fidyt, K.; Fiedorowicz, A.; Strządała, L.; Szumny, A. β-caryophyllene and β-caryophyllene oxide—Natural compounds of anticancer and analgesic properties. Cancer Med. 2016, 5, 3007–3017. [Google Scholar] [CrossRef]
  27. Kim, C.; Cho, S.K.; Kim, K.D.; Nam, D.; Chung, W.S.; Jang, H.J.; Lee, S.G.; Shim, B.S.; Sethi, G.; Ahn, K.S. β-Caryophyllene oxide potentiates TNFα-induced apoptosis and inhibits invasion through down-modulation of NF-κB-regulated gene products. Apoptosis 2014, 19, 708–718. [Google Scholar] [CrossRef] [PubMed]
  28. Dahham, S.S.; Tabana, Y.M.; Iqbal, M.A.; Ahamed, M.B.; Ezzat, M.O.; Majid, A.S.; Majid, A.M. The anticancer, antioxidant and antimicrobial properties of the sesquiterpene β-caryophyllene from the essential oil of Aquilaria crassna. Molecules 2015, 20, 11808–11829. [Google Scholar] [CrossRef] [PubMed]
  29. Ambrož, M.; Boušová, I.; Skarka, A.; Hanušová, V.; Králová, V.; Matoušková, P.; Szotáková, B.; Skálová, L. The influence of sesquiterpenes from Myrica rubra on the antiproliferative and pro-oxidative effects of doxorubicin and its accumulation in cancer cells. Molecules 2015, 20, 15343–15358. [Google Scholar] [CrossRef] [Green Version]
  30. Ambrož, M.; Šmatová, M.; Šadibolová, M.; Pospíšilová, E.; Hadravská, P.; Kašparová, M.; Hanušová Skarková, V.; Králová, V.; Skálová, L. Sesquiterpenes α-humulene and β-caryophyllene oxide enhance the efficacy of 5-fluorouracil and oxaliplatin in colon cancer cells. Acta Pharm. 2019, 69, 121–128. [Google Scholar] [CrossRef] [Green Version]
  31. Hanušová, V.; Caltová, K.; Svobodová, H.; Ambrož, M.; Skarka, A.; Murínová, N.; Králová, V.; Tomšík, P.; Skálová, L. The effects of β-caryophyllene oxide and trans-nerolidol on the efficacy of doxorubicin in breast cancer cells and breast tumor-bearing mice. Biomed. Pharmacother. 2017, 95, 828–836. [Google Scholar] [CrossRef]
  32. Legault, J.; Pichette, A. Potentiating effect of β-caryophyllene on anticancer activity of α-humulene, isocaryophyllene and paclitaxel. J. Pharm. Pharmacol. 2007, 59, 1643–1647. [Google Scholar] [CrossRef]
  33. Tung, Y.T.; Chua, M.T.; Wang, S.Y.; Chang, S.T. Anti-inflammation activities of essential oil and its constituents from indigenous cinnamon (Cinnamomum osmophloeum) twigs. Bioresour. Technol. 2008, 99, 3908–3913. [Google Scholar] [CrossRef] [PubMed]
  34. Medeiros, R.; Passos, G.; Vitor, C.; Koepp, J.; Mazzuco, T.; Pianowski, L.; Campos, M.; Calixto, J. Effect of two active compounds obtained from the essential oil of Cordia verbenacea on the acute inflammatory responses elicited by LPS in the rat paw. Br. J. Pharmacol. 2007, 151, 618–627. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Gertsch, J.; Leonti, M.; Raduner, S.; Racz, I.; Chen, J.Z.; Xie, X.Q.; Altmann, K.H.; Karsak, M.; Zimmer, A. β-caryophyllene is a dietary cannabinoid. Proc. Natl. Acad. Sci. USA 2008, 105, 9099–9104. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Yang, D.; Michel, L.; Chaumont, J.P.; Millet-Clerc, J. Use of caryophyllene oxide as an antifungal agent in an in vitro experimental model of onychomycosis. Mycopathologia 1999, 148, 79–82. [Google Scholar] [CrossRef] [PubMed]
  37. Hilgers, F.; Habash, S.S.; Loeschcke, A.; Ackermann, Y.S.; Neumann, S.; Heck, A.; Klaus, O.; Hage-Hülsmann, J.; Grundler, F.M.; Jaeger, K.E. Heterologous production of β-caryophyllene and evaluation of its activity against plant pathogenic fungi. Microorganisms 2021, 9, 168. [Google Scholar] [CrossRef] [PubMed]
  38. Souza, A.B.; Martins, C.H.; Souza, M.G.; Furtado, N.A.; Heleno, V.C.; de Sousa, J.P.; Rocha, E.M.; Bastos, J.K.; Cunha, W.R.; Veneziani, R.C. Antimicrobial activity of terpenoids from Copaifera langsdorffii Desf. against cariogenic bacteria. Phytother. Res. 2011, 25, 215–220. [Google Scholar] [CrossRef]
  39. Moo, C.L.; Yang, S.K.; Osman, M.A.; Yuswan, M.H.; Loh, J.Y.; Lim, W.M.; Lim, S.H.E.; Lai, K.S. Antibacterial activity and mode of action of β-caryophyllene on Bacillus cereus. Pol. J. Microbiol. 2020, 69, 49–54. [Google Scholar] [CrossRef] [Green Version]
  40. Benelli, G.; Pavela, R.; Drenaggi, E.; Desneux, N.; Maggi, F. Phytol, (E)-nerolidol and spathulenol from Stevia rebaudiana leaf essential oil as effective and eco-friendly botanical insecticides against Metopolophium dirhodum. Ind. Crops Prod. 2020, 155, 112844. [Google Scholar] [CrossRef]
  41. Do Nascimento, K.F.; Moreira, F.M.F.; Santos, J.A.; Kassuya, C.A.L.; Croda, J.H.R.; Cardoso, C.A.L.; do Carmo Vieira, M.; Ruiz, A.L.T.G.; Foglio, M.A.; de Carvalho, J.E. Antioxidant, anti-inflammatory, antiproliferative and antimycobacterial activities of the essential oil of Psidium guineense Sw. and spathulenol. J. Ethnopharmacol. 2018, 210, 351–358. [Google Scholar] [CrossRef]
  42. Ziaei, A.; Ramezani, M.; Wright, L.; Paetz, C.; Schneider, B.; Amirghofran, Z. Identification of spathulenol in Salvia mirzayanii and the immunomodulatory effects. Phytother. Res. 2011, 25, 557–562. [Google Scholar] [CrossRef]
  43. Cantrell, C.L.; Klun, J.A.; Bryson, C.T.; Kobaisy, M.; Duke, S.O. Isolation and identification of mosquito bite deterrent terpenoids from leaves of American (Callicarpa americana) and Japanese (Callicarpa japonica) beautyberry. J. Agric. Food Chem. 2005, 53, 5948–5953. [Google Scholar] [CrossRef] [PubMed]
  44. Dos Santos, E.; Radai, J.A.S.; do Nascimento, K.F.; Formagio, A.S.N.; de Matos Balsalobre, N.; Ziff, E.B.; Castelon Konkiewitz, E.; Kassuya, C.A.L. Contribution of spathulenol to the anti-nociceptive effects of Psidium guineense. Nutr. Neurosci. 2022, 25, 812–822. [Google Scholar] [CrossRef] [PubMed]
  45. Zhai, B.; Zhang, N.; Han, X.; Li, Q.; Zhang, M.; Chen, X.; Li, G.; Zhang, R.; Chen, P.; Wang, W. Molecular targets of β-elemene, a herbal extract used in traditional Chinese medicine, and its potential role in cancer therapy: A review. Biomed. Pharmacother. 2019, 114, 108812. [Google Scholar] [CrossRef]
  46. Wang, B.; Peng, X.X.; Sun, R.; Li, J.; Zhan, X.R.; Wu, L.J.; Wang, S.L.; Xie, T. Systematic review of β-elemene injection as adjunctive treatment for lung cancer. Chin. J. Integr. Med. 2012, 18, 813–823. [Google Scholar] [CrossRef]
  47. Zhai, B.; Zeng, Y.; Zeng, Z.; Zhang, N.; Li, C.; Zeng, Y.; You, Y.; Wang, S.; Chen, X.; Sui, X. Drug delivery systems for elemene, its main active ingredient β-elemene, and its derivatives in cancer therapy. Int. J. Nanomed. 2018, 13, 6279–6296. [Google Scholar] [CrossRef] [Green Version]
  48. Luo, H.; Vong, C.T.; Chen, H.; Gao, Y.; Lyu, P.; Qiu, L.; Zhao, M.; Liu, Q.; Cheng, Z.; Zou, J. Naturally occurring anti-cancer compounds: Shining from Chinese herbal medicine. Chin. Med. 2019, 14, 48. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  49. Pan, Y.; Wang, W.; Huang, S.; Ni, W.; Wei, Z.; Cao, Y.; Yu, S.; Jia, Q.; Wu, Y.; Chai, C. Beta-elemene inhibits breast cancer metastasis through blocking pyruvate kinase M2 dimerization and nuclear translocation. J. Cell. Mol. Med. 2019, 23, 6846–6858. [Google Scholar] [CrossRef]
  50. Wang, J.; Xu, C.; Chen, Y.; Shao, L.; Li, T.; Fan, X.; Yu, L.; Zhang, R.; Chen, B.; Chen, H. β-elemene enhances the antitumor activity of erlotinib by inducing apoptosis through AMPK and MAPK pathways in TKI-resistant H1975 lung cancer cells. J. Cancer 2021, 12, 2285–2294. [Google Scholar] [CrossRef]
  51. Liu, Y.; Jiang, Z.Y.; Zhou, Y.L.; Qiu, H.H.; Wang, G.; Luo, Y.; Liu, J.B.; Liu, X.W.; Bu, W.Q.; Song, J. β-elemene regulates endoplasmic reticulum stress to induce the apoptosis of NSCLC cells through PERK/IRE1α/ATF6 pathway. Biomed. Pharmacother. 2017, 93, 490–497. [Google Scholar] [CrossRef]
  52. Zhang, X.; Zhang, Y.; Li, Y. β-elemene decreases cell invasion by upregulating E-cadherin expression in MCF-7 human breast cancer cells. Oncol. Rep. 2013, 30, 745–750. [Google Scholar] [CrossRef] [Green Version]
  53. Lin, L.; Li, L.; Chen, X.; Zeng, B.; Lin, T. Preliminary evaluation of the potential role of β-elemene in reversing erlotinib-resistant human NSCLC A549/ER cells. Oncol. Lett. 2018, 16, 3380–3388. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Liu, M.; Chen, X.; Ma, J.; Hassan, W.; Wu, H.; Ling, J.; Shang, J. β-Elemene attenuates atherosclerosis in apolipoprotein E-deficient mice via restoring NO levels and alleviating oxidative stress. Biomed. Pharmacother. 2017, 95, 1789–1798. [Google Scholar] [CrossRef] [PubMed]
  55. Fang, Y.; Kang, Y.; Zou, H.; Cheng, X.; Xie, T.; Shi, L.; Zhang, H. β-elemene attenuates macrophage activation and proinflammatory factor production via crosstalk with Wnt/β-catenin signaling pathway. Fitoterapia 2018, 124, 92–102. [Google Scholar] [CrossRef] [PubMed]
  56. De Almeida, C.G.; Garbois, G.D.; Amaral, L.M.; Diniz, C.C.; Le Hyaric, M. Relationship between structure and antibacterial activity of lipophilic N-acyldiamines. Biomed. Pharmacother. 2010, 64, 287–290. [Google Scholar] [CrossRef]
  57. Wilson, A.; Ruiz, N. Transport of lipopolysaccharides and phospholipids to the outer membrane. Curr. Opin. Microbiol. 2021, 60, 51–57. [Google Scholar] [CrossRef]
  58. Schmidt, E.; Bail, S.; Friedl, S.M.; Jirovetz, L.; Buchbauer, G.; Wanner, J.; Denkova, Z.; Slavchev, A.; Stoyanova, A.; Geissler, M. Antimicrobial activities of single aroma compounds. Nat. Prod. Commun. 2010, 5, 1365–1368. [Google Scholar] [CrossRef] [Green Version]
  59. Woo, H.J.; Yang, J.Y.; Lee, M.H.; Kim, H.W.; Kwon, H.J.; Park, M.; Kim, S.K.; Park, S.Y.; Kim, S.H.; Kim, J.B. Inhibitory effects of β-caryophyllene on Helicobacter pylori infection in vitro and in vivo. Int. J. Mol. Sci. 2020, 21, 1008. [Google Scholar] [CrossRef] [Green Version]
  60. De Jesús Dzul-Beh, A.; García-Sosa, K.; Uc-Cachón, A.H.; Bórquez, J.; Loyola, L.A.; Barrios-García, H.B.; Peña-Rodríguez, L.M.; Molina-Salinas, G.M. In vitro growth inhibition and bactericidal activity of spathulenol against drug-resistant clinical isolates of Mycobacterium tuberculosis. Rev. Bras. Farmacogn. 2020, 29, 798–800. [Google Scholar] [CrossRef]
  61. Jang, H.I.; Rhee, K.J.; Eom, Y.B. Antibacterial and antibiofilm effects of α-humulene against Bacteroides fragilis. Can. J. Microbiol. 2020, 66, 389–399. [Google Scholar] [CrossRef]
  62. Pichette, A.; Larouche, P.L.; Lebrun, M.; Legault, J. Composition and antibacterial activity of Abies balsamea essential oil. Phytother. Res. 2006, 20, 371–373. [Google Scholar] [CrossRef]
  63. De Lacerda Leite, G.M.; de Oliveira Barbosa, M.; Lopes, M.J.P.; de Araújo Delmondes, G.; Bezerra, D.S.; Araújo, I.M.; de Alencar, C.D.C.; Coutinho, H.D.M.; Peixoto, L.R.; Barbosa-Filho, J.M. Pharmacological and toxicological activities of α-humulene and its isomers: A systematic review. Trends Food Sci. Technol. 2021, 115, 255–274. [Google Scholar] [CrossRef]
  64. Shafi, P.M.; Rosamma, M.K.; Jamil, K.; Reddy, P.S. Antibacterial activity of the essential oil from Aristolochia indica. Fitoterapia 2002, 73, 439–441. [Google Scholar] [CrossRef]
  65. Ochi, T.; Shibata, H.; Higuti, T.; Kodama, K.-H.; Kusumi, T.; Takaishi, Y. Anti-Helicobacter pylori compounds from Santalum album. J. Nat. Prod. 2005, 68, 819–824. [Google Scholar] [CrossRef] [PubMed]
  66. Rossolini, G.M.; Mantengoli, E. Treatment and control of severe infections caused by multiresistant Pseudomonas aeruginosa. Clin. Microbiol. Infect. 2005, 11, 17–32. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Lambert, R.J.; Joynson, J.; Forbes, B. The relationships and susceptibilities of some industrial, laboratory and clinical isolates of Pseudomonas aeruginosa to some antibiotics and biocides. J. Appl. Microbiol. 2001, 91, 972–984. [Google Scholar] [CrossRef] [Green Version]
  68. Matsuo, Y.; Eda, S.; Gotoh, N.; Yoshihara, E.; Nakae, T. MexZ-mediated regulation of mexXY multidrug efflux pump expression in Pseudomonas aeruginosa by binding on the mexZ-mexX intergenic DNA. FEMS Microbiol. Lett. 2004, 238, 23–28. [Google Scholar]
  69. Yang, S.K.; Yusoff, K.; Mai, C.W.; Lim, W.M.; Yap, W.S.; Lim, S.H.E.; Lai, K.S. Additivity vs. synergism: Investigation of the additive interaction of cinnamon bark oil and meropenem in combinatory therapy. Molecules 2017, 22, 1733. [Google Scholar] [CrossRef] [Green Version]
  70. Costerton, J.W.; Stewart, P.S.; Greenberg, E.P. Bacterial biofilms: A common cause of persistent infections. Science 1999, 284, 1318–1322. [Google Scholar] [CrossRef] [Green Version]
  71. Parsek, M.R.; Singh, P.K. Bacterial biofilms: An emerging link to disease pathogensis. Annu. Rev. Microbiol. 2003, 57, 677–701. [Google Scholar] [CrossRef]
  72. Verderosa, A.; Totsika, M.; Fairfull-Smith, K. Bacterial biofilm eradication agents: A current review. Front. Chem. 2019, 7, 824. [Google Scholar] [CrossRef] [Green Version]
  73. Høiby, N.; Bjarnsholt, T.; Givskov, M.; Molin, S.; Ciofu, O. Antibiotic resistance of bacterial biofilms. Int. J. Antimicrob. Agents 2010, 35, 322–332. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Kazemzadeh-Narbat, M.; Kindrachuk, J.; Duan, K.; Jenssen, H.; Hancock, R.E.; Wang, R. Antimicrobial peptides on calcium phosphate-coated titanium for the prevention of implant-associated infections. Biomaterials 2010, 31, 9519–9526. [Google Scholar] [CrossRef] [PubMed]
  75. Arciola, C.R.; Campoccia, D.; Montanaro, L. Implant infections: Adhesion, biofilm formation and immune evasion. Nat. Rev. Microbiol. 2018, 16, 397–409. [Google Scholar] [CrossRef]
  76. Bowler, P.G. Antibiotic resistance and biofilm tolerance: A combined threat in the treatment of chronic infections. J. Wound Care 2018, 27, 273–277. [Google Scholar] [CrossRef]
  77. Haaber, J.; Cohn, M.T.; Frees, D.; Andersen, T.J.; Ingmer, H. Planktonic aggregates of Staphylococcus aureus protect against common antibiotics. PLoS ONE 2012, 7, e41075. [Google Scholar] [CrossRef]
  78. Hall-Stoodley, L.; Stoodley, P. Evolving concepts in biofilm infections. Cell. Microbiol. 2009, 11, 1034–1043. [Google Scholar] [CrossRef]
  79. Otto, M. Staphylococcal biofilms. In Current Topics in Microbiology and Immunology; Romeo, T., Ed.; Springer: Berlin/Heidelberg, Germany, 2008; Volume 322, pp. 207–228. [Google Scholar]
  80. Bhattacharya, M.; Wozniak, D.J.; Stoodley, P.; Hall-Stoodley, L. Prevention and treatment of Staphylococcus aureus biofilms. Expert Rev. Anti-Infect. Ther. 2015, 13, 1499–1516. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  81. Brady, R.A.; Leid, J.G.; Calhoun, J.H.; Costerton, J.W.; Shirtliff, M.E. Osteomyelitis and the role of biofilms in chronic infection. FEMS Immunol. Med. Microbiol. 2008, 52, 13–22. [Google Scholar] [CrossRef] [Green Version]
  82. Del Pozo, J.L.; Patel, R. The challenge of treating biofilm-associated bacterial infections. Clin. Pharmacol. Ther. 2007, 82, 204–209. [Google Scholar] [CrossRef]
  83. Thurlow, L.R.; Hanke, M.L.; Fritz, T.; Angle, A.; Aldrich, A.; Williams, S.H.; Engebretsen, I.L.; Bayles, K.W.; Horswill, A.R.; Kielian, T. Staphylococcus aureus biofilms prevent macrophage phagocytosis and attenuate inflammation in vivo. J. Immunol. 2011, 186, 6585–6596. [Google Scholar] [CrossRef] [Green Version]
  84. Nadell, C.D.; Xavier, J.B.; Foster, K.R. The sociobiology of biofilms. FEMS Microbiol. Rev. 2009, 33, 206–224. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Ranieri, M.R.; Whitchurch, C.B.; Burrows, L.L. Mechanisms of biofilm stimulation by subinhibitory concentrations of antimicrobials. Curr. Opin. Microbiol. 2018, 45, 164–169. [Google Scholar] [CrossRef] [PubMed]
  86. Kaplan, J.B.; Izano, E.A.; Gopal, P.; Karwacki, M.T.; Kim, S.; Bose, J.L.; Bayles, K.W.; Horswill, A.R. Low levels of β-lactam antibiotics induce extracellular DNA release and biofilm formation in Staphylococcus aureus. mBio 2012, 3, e00198-12. [Google Scholar] [CrossRef] [Green Version]
  87. Kuehl, R.; Al-Bataineh, S.; Gordon, O.; Luginbuehl, R.; Otto, M.; Textor, M.; Landmann, R. Furanone at subinhibitory concentrations enhances staphylococcal biofilm formation by luxS repression. Antimicrob. Agents Chemother. 2009, 53, 4159–4166. [Google Scholar] [CrossRef] [Green Version]
  88. Weiser, J.; Henke, H.A.; Hector, N.; Both, A.; Christner, M.; Büttner, H.; Kaplan, J.B.; Rohde, H. Sub-inhibitory tigecycline concentrations induce extracellular matrix binding protein Embp dependent Staphylococcus epidermidis biofilm formation and immune evasion. Int. J. Med. Microbiol. 2016, 306, 471–478. [Google Scholar] [CrossRef] [PubMed]
  89. Jin, Y.; Guo, Y.; Zhan, Q.; Shang, Y.; Qu, D.; Yu, F. Subinhibitory concentrations of mupirocin stimulate Staphylococcus aureus biofilm formation by upregulating cidA. Antimicrob. Agents Chemother. 2020, 64, e01912–e01919. [Google Scholar] [CrossRef]
  90. Ma, H.; Bryers, J.D. Non-invasive determination of conjugative transfer of plasmids bearing antibiotic-resistance genes in biofilm-bound bacteria: Effects of substrate loading and antibiotic selection. Appl. Microbiol. Biotechnol. 2013, 97, 317–328. [Google Scholar] [CrossRef] [Green Version]
  91. Dai, Z.J.; Tang, W.; Lu, W.F.; Gao, J.; Kang, H.F.; Ma, X.B.; Min, W.L.; Wang, X.J.; Wu, W.Y. Antiproliferative and apoptotic effects of β-elemene on human hepatoma HepG2 cells. Cancer Cell Int. 2013, 13, 27. [Google Scholar] [CrossRef] [Green Version]
  92. Adams, R.P. Identification of Essential Oil Components by Gas Chromatography/Mass Spectrometry, 5th ed.; Texensis Publishing: Gruver, TX, USA, 2017. [Google Scholar]
  93. Babushok, V.I.; Linstrom, P.J.; Zenkevich, I.G. Retention indices for frequently reported compounds of plant essential oils. J. Phys. Chem. Ref. Data 2011, 40, 1–47. [Google Scholar] [CrossRef] [Green Version]
  94. Linstrom, P.J.; Mallard, W.G. NIST Chemistry WebBook, NIST Standard Reference Database Number 69. Available online: http://webbook.nist.gov/chemistry (accessed on 8 October 2021).
  95. Clinical and Laboratory Standards Institute. Document M02-A11. Performance Standards for Antimicrobial Disk Susceptibility Tests, 11th ed.; CLSI: Wayne, PA, USA, 2012. [Google Scholar]
  96. Clinical and Laboratory Standards Institute. Document M7-A7. Methods for Dilution Antimicrobial Susceptibility Tests for Bacteria That Grow Aerobically; Approved Standard, 7th ed.; CLSI: Wayne, PA, USA, 2006. [Google Scholar]
  97. Odds, F.C. Synergy, antagonism, and what the chequerboard puts between them. J. Antimicrob. Chemother. 2003, 52, 1. [Google Scholar] [CrossRef]
  98. Peng, L.; Xiong, Y.; Wang, M.; Han, M.; Cai, W.; Li, Z. Chemical composition of essential oil in Mosla chinensis Maxim cv. Jiangxiangru and its inhibitory effect on Staphylococcus aureus biofilm formation. Open Life Sci. 2018, 13, 1–10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  99. Mosmann, T. Rapid colorimetric assay for cellular growth and survival: Application to proliferation and cytotoxicity assays. J. Immunol. Methods 1983, 65, 55–63. [Google Scholar] [CrossRef]
Figure 1. The effect of different concentrations of essential oil from M. nepalensis against biofilm formation of Staphylococcus aureus. Differences were statistically significant in relation to the control for * p < 0.05; ** p < 0.01; *** p < 0.001; **** p < 0.0001.
Figure 1. The effect of different concentrations of essential oil from M. nepalensis against biofilm formation of Staphylococcus aureus. Differences were statistically significant in relation to the control for * p < 0.05; ** p < 0.01; *** p < 0.001; **** p < 0.0001.
Molecules 27 04631 g001
Table 1. Chemical composition of M. nepalensis essential oil.
Table 1. Chemical composition of M. nepalensis essential oil.
Peak No.CompoundRI aRI b% Area
1(E)-2-Octenal105010491.1
2Linalool109810950.6
3cis-Verbenol114111370.1
4α-Terpineol119011860.1
5cis-Myrtanol125212500.2
6(E)-2-Decenal125912600.1
7Bornyl acetate128512840.1
8Dihydroedulan129512930.2
9Menthyl acetate129712940.1
10Isomenthyl acetate130613040.2
11δ-Elemene133813350.1
127-epi-Silphiperfol-5-ene134613450.1
13α-Cubebene135013480.1
14(E)-2-Undecenal136113570.1
15Cyclosativene136913690.1
16α-Ylangene137313730.1
17α-Copaene137813740.3
18β-Bourbonene138713870.7
19β-Elemene139313896.1
20cis-α-Bergamotene141514110.8
21β-Caryophyllene142014174.1
22γ-Elemene143514341.0
23Aromadendrene144214390.1
24α-Himachalene144814490.1
25α-Humulene145514523.2
26Alloaromadendrene146014580.1
27γ-Muurolene147814781.5
28Valencene149114961.4
29Viridiflorene149314962.4
30α-Selinene149914982.0
31α-Muurolene150215000.8
32β-Bisabolene150815050.5
33γ-Cadinene151715130.7
34Cubebol151915140.1
35δ-Cadinene152515221.2
36cis-Sesquisabinene hydrate153815420.3
37Selina-3,7(11)-diene154315450.4
38α-Calacorene154615440.5
39Dehydronerolidol155715621.2
40(E)-Nerolidol156315610.7
41Mint oxide157215740.7
42Spathulenol157515777.3
43Caryophyllene oxide1585158210.2
44Salvial-4(14)-en-1-one159815941.8
45Widdrol160315990.7
46Humulene epoxide II160916087.2
471-epi-Cubenol162416271.0
48epi-α-Cadinol163716380.7
49Caryophylladienol II164116391.1
50Isospathulenol164316403.2
51α-Muurolol164516441.0
52α-Cadinol165316520.6
53Neointermedeol166016584.5
54Zizanol167216770.7
55(Z)-α-Santalol167516743.6
56Eudesma-4(15),7-dien-1β-ol169116872.2
57Aristol-1(10)-en-9-ol170817043.4
58β-Santalol171917151.5
59Vetiselinenol172617302.0
60γ-Costol174917450.2
61α-Vetivol175117560.7
62α-Cyperone175617580.3
63β-Acoradienol176017620.7
64β-Costol176817651.2
65α-Costol177617730.3
6614-hydroxy-α-muurolene178017790.6
67(E)-Isovalencenol178817930.3
68Neophytadiene183618400.7
69Hexahydrofarnesyl acetone184318470.7
70Rimuene190118960.2
71(5E,9E)-Farnesyl acetone190919130.2
72Carissone192719260.4
73Verrucarol194119390.1
74Methyl linolelaidate197619800.2
75(Z,E)-Geranyl linalool199619980.4
76Panaxynone202220182.0
77Thunbergol206920730.1
78Phytol210821140.1
Oxygenated monoterpenes 1.6
Sesquiterpene hydrocarbons 28.6
Oxygenated sesquiterpenes 60.3
Diterpenes hydrocarbons 0.7
Oxygenated diterpenes 0.6
Total identified 96.4
a Retention index calculated from n-alkanes (C7-C30) on an HP-5MS column; b Retention index data from the literature.
Table 2. Diameter of the inhibition zones (DIZ), minimum inhibitory concentrations (MIC) and minimum bactericidal concentration (MBC) of the essential oil of M. nepalensis (MNEO).
Table 2. Diameter of the inhibition zones (DIZ), minimum inhibitory concentrations (MIC) and minimum bactericidal concentration (MBC) of the essential oil of M. nepalensis (MNEO).
StrainDIZ (mm) ± SDMIC (mg/mL)MBC (mg/mL)
MNEOChlMNEOChlMNEOChl
Gram positive
B. subtilis ATCC 663314.0 ± 1.225.1 ± 1.00.0390.0020.1560.004
S. aureus ATCC 653816.7 ± 1.723.6 ± 1.30.0780.0020.0780.016
P. larvae ATCC 954513.4 ± 1.128.3 ± 1.70.0390.0010.0390.002
Gram negative
E. coli ATCC 259228.3 ± 0.925.9 ± 0.81.2500.002>2.5000.004
P. aeruginosa ATCC 2785310.5 ± 1.316.7 ± 0.40.6250.0312.5000.250
DIZ, diameter of the inhibition zones (mm) is given as the mean ± SD of triplicate experiments; positive control: Chl, chloramphenicol.
Table 3. Fractional inhibitory concentration index (FICI) values of the essential oil of M. nepalensis (MNEO) and chloramphenicol combinations.
Table 3. Fractional inhibitory concentration index (FICI) values of the essential oil of M. nepalensis (MNEO) and chloramphenicol combinations.
Microorganism MICa (μg/mL)MICc (μg/mL)FICI
Bacillus subtilisMNEO39.109.760.37 (S)
Chl3.900.48
Staphylococcus aureusMNEO78.134.880.56 (A)
Chl3.901.95
Escherichia coliMNEO1250.00312.500.31 (S)
Chl3.900.24
Pseudomonas aeruginosaMNEO625.0039.100.13 (S)
Chl15.600.98
MICa: MIC alone; MICc: MIC combined; Chl: chloramphenicol. S, synergy; A, additivity.
Table 4. Fractional inhibitory concentration index (FICI) values of the essential oil of M. nepalensis (MNEO) and streptomycin combinations.
Table 4. Fractional inhibitory concentration index (FICI) values of the essential oil of M. nepalensis (MNEO) and streptomycin combinations.
Microorganism MICa (μg/mL)MICc (μg/mL)FICI
Bacillus subtilisMNEO39.004.880.37 (S)
SM0.490.12
Staphylococcus aureusMNEO78.134.880.12 (S)
SM1.950.12
Escherichia coliMNEO1250.00312.500.50 (S)
SM3.900.98
Pseudomonas aeruginosaMNEO625.0039.000.12 (S)
SM1.950.12
MICa: MIC alone; MICc: MIC combined; SM: streptomycin. S, synergy.
Table 5. IC50 (μg/mL) ± SD values to the essential oil of M. nepalensis and positive control doxorubicin against cell lines.
Table 5. IC50 (μg/mL) ± SD values to the essential oil of M. nepalensis and positive control doxorubicin against cell lines.
Cell LineEssential OilDoxorubicin
HepG219.53 ± 2.840.46 ± 0.02
MCF-713.13 ± 1.900.70 ± 0.05
HL-770219.14 ± 0.630.60 ± 0.13
A-54919.19 ± 3.080.48 ± 0.01
HCT-11635.22 ± 8.360.57 ± 0.03
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Fu, J.; Gao, Y.; Xing, X. Preliminary Study on Phytochemical Constituents and Biological Activities of Essential Oil from Myriactis nepalensis Less. Molecules 2022, 27, 4631. https://doi.org/10.3390/molecules27144631

AMA Style

Fu J, Gao Y, Xing X. Preliminary Study on Phytochemical Constituents and Biological Activities of Essential Oil from Myriactis nepalensis Less. Molecules. 2022; 27(14):4631. https://doi.org/10.3390/molecules27144631

Chicago/Turabian Style

Fu, Jikai, Yang Gao, and Xiang Xing. 2022. "Preliminary Study on Phytochemical Constituents and Biological Activities of Essential Oil from Myriactis nepalensis Less." Molecules 27, no. 14: 4631. https://doi.org/10.3390/molecules27144631

Article Metrics

Back to TopTop