ArticlePDF Available

Increasing the Efficacy of Treatment of Staphylococcus aureus–Candida albicans Mixed Infections with Myrtenol

Authors:

Abstract and Figures

Infectious diseases caused by various nosocomial microorganisms affect worldwide both immunocompromised and relatively healthy persons. Bacteria and fungi have different tools to evade antimicrobials, such as hydrolysis damaging the drug, efflux systems, and the formation of biofilm that significantly complicates the treatment of the infection. Here, we show that myrtenol potentiates the antimicrobial and biofilm-preventing activity of conventional drugs against S. aureus and C. albicans mono- and dual-species cultures. In our study, the two optical isomers, (-)-myrtenol and (+)-myrtenol, have been tested as either antibacterials, antifungals, or enhancers of conventional drugs. (+)-Myrtenol demonstrated a synergistic effect with amikacin, fluconazole, and benzalkonium chloride on 64–81% of the clinical isolates of S. aureus and C. albicans, including MRSA and fluconazole-resistant fungi, while (-)-myrtenol increased the properties of amikacin and fluconazole to repress biofilm formation in half of the S. aureus and C. albicans isolates. Furthermore, myrtenol was able to potentiate benzalkonium chloride up to sixteen-fold against planktonic cells in an S. aureus–C. albicans mixed culture and repressed the adhesion of S. aureus. The mechanism of both (-)-myrtenol and (+)-myrtenol synergy with conventional drugs was apparently driven by membrane damage since the treatment with both terpenes led to a significant drop in membrane potential similar to the action of benzalkonium chloride. Thus, due to the low toxicity of myrtenol, it seems to be a promising agent to increase the efficiency of the treatment of infections caused by bacteria and be fungi of the genus Candida as well as mixed fungal–bacterial infections, including resistant strains.
Content may be subject to copyright.
Antibiotics 2022, 11, 1743. https://doi.org/10.3390/antibiotics11121743 www.mdpi.com/journal/antibiotics
Article
Increasing the Efficacy of Treatment of Staphylococcus
aureusCandida albicans Mixed Infections with Myrtenol
Ruba Y. Mahmoud 1, Elena Y. Trizna 1, Rand K. Sulaiman 1, Roman S. Pavelyev 1, Ilmir R. Gilfanov 1,2,
Svetlana A. Lisovskaya 3,4, Olga V. Ostolopovskaya 1,3, Larisa L. Frolova 5, Alexander V. Kutchin 5,
Galina B. Guseva 6, Elena V. Antina 6, Mikhail B. Berezin 6, Liliya E. Nikitina 1,3,†,* and Airat R. Kayumov 1,†,*
1 Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia
2 Varnishes and Paints Department, Kazan National Research Technological University, 420015 Kazan, Russia
3 Faculty of Medicine and Biology, Kazan State Medical University, 420012 Kazan, Russia
4 Scientific Research Institute of Epidemiology and Microbiology, 420015 Kazan, Russia
5 Institute of Chemistry, Federal Research Center “Komi Scientific Centre”, Ural Branch, Russian Academy
of Sciences, 167000 Syktyvkar, Russia
6 G.A. Krestov Institute of Solution Chemistry of Russian Academy of Sciences, 153045 Ivanovo, Russia
* Correspondence: nikitl@mail.ru (L.E.N.); kairatr@yandex.ru (A.R.K.); Tel.: +7 903 307-50-70 (L.E.N.);
Tel.: +7 904 665-19-08 (A.R.K.)
These authors contributed equally to this work.
Abstract: Infectious diseases caused by various nosocomial microorganisms affect worldwide both
immunocompromised and relatively healthy persons. Bacteria and fungi have different tools to
evade antimicrobials, such as hydrolysis damaging the drug, efflux systems, and the formation of
biofilm that significantly complicates the treatment of the infection. Here, we show that myrtenol
potentiates the antimicrobial and biofilm-preventing activity of conventional drugs against S. aureus
and C. albicans mono- and dual-species cultures. In our study, the two optical isomers, (-)-myrtenol
and (+)-myrtenol, have been tested as either antibacterials, antifungals, or enhancers of conventional
drugs. (+)-Myrtenol demonstrated a synergistic effect with amikacin, fluconazole, and ben-
zalkonium chloride on 64–81% of the clinical isolates of S. aureus and C. albicans, including MRSA
and fluconazole-resistant fungi, while (-)-myrtenol increased the properties of amikacin and flucon-
azole to repress biofilm formation in half of the S. aureus and C. albicans isolates. Furthermore, myr-
tenol was able to potentiate benzalkonium chloride up to sixteen-fold against planktonic cells in an
S. aureusC. albicans mixed culture and repressed the adhesion of S. aureus. The mechanism of both
(-)-myrtenol and (+)-myrtenol synergy with conventional drugs was apparently driven by mem-
brane damage since the treatment with both terpenes led to a significant drop in membrane poten-
tial similar to the action of benzalkonium chloride. Thus, due to the low toxicity of myrtenol, it
seems to be a promising agent to increase the efficiency of the treatment of infections caused by
bacteria and be fungi of the genus Candida as well as mixed fungal–bacterial infections, including
resistant strains.
Keywords: mixed infections; myrtenol; benzalkonium chloride; drug synergism; Staphylcoccus au-
reus; Candida albicans
1. Introduction
Infectious diseases caused by various nosocomial bacteria and fungi like Enterobac-
teriaceae (Klebsiella sp and Escherichia coli), Staphylococcus aureus, Candida albicans, Crypto-
coccus neoformans, and many others affect worldwide both immunocompromised and rel-
atively healthy persons [1]. In addition to the most vulnerable populations of patients,
such as neonatal, old, and AIDS-infected patients and persons with an intravenous cath-
eter, in the last three years, SARS-CoV2 led to an increased risk of mortality and a longer
Citation: Mahmoud, R.Y.; Trizna,
E.Y.; Sulaiman, R.K.; Pavelyev, R.S.;
Gilfanov, I.R.; Lisovskaya, S.A.;
Ostolopovskaya, O.V.; Frolova, L.L.;
Kutchin, A.V.; Guseva, G.B.; et al.
Increasing the Efficacy of Treatment
of Staphylococcus aureusCandida
albicans Mixed Infections with
Myrtenol. Antibiotics 2022, 11, 1743.
https://doi.org/10.3390/
antibiotics11121743
Academic Editors: Jorge H. Leitão,
Nitin Amdare and Joana R. Feliciano
Received: 17 November 2022
Accepted: 28 November 2022
Published: 2 December 2022
Publisher’s Note: MDPI stays neu-
tral with regard to jurisdictional
claims in published maps and institu-
tional affiliations.
Copyright: © 2022 by the authors. Li-
censee MDPI, Basel, Switzerland.
This article is an open access article
distributed under the terms and con-
ditions of the Creative Commons At-
tribution (CC BY) license (https://cre-
ativecommons.org/licenses/by/4.0/).
Antibiotics 2022, 11, 1743 2 of 17
course of ICU stays [2–4]. Antimicrobial therapy remains the only way to target patho-
genic microorganisms and save lives. Although conventional antimicrobial agents use
various strategies to repress the growth of pathogens, bacteria and fungi have different
tools to evade them, making the development and spread of antimicrobial resistance
(AMR) one of the factors that complicates the treatment of infectious diseases [5]. It has
been shown in the last decades that, in many cases, several pathogens rather than only
one are associated with disease [6]. These polymicrobial infections are often characterized
by more intense symptoms than any of the effects noticed by one microbe alone and in-
creased resistance to treatment [7,8]. S. aureus and C. albicans, an important, dangerous
twosome, have been shown to form a bacterial–fungal environment and were coisolated
from different infections, including periodontitis, cystic fibrosis, denture stomatitis, uri-
nary tract infections, burn wound infections, and infections of medical devices, such as
central venous catheters [9]. In bacterial–fungal coinfection, each counterpart has been re-
ported to contribute to resistance [10–12]. Moreover, 94% of S. aureus isolates are tolerant
to penicillin and its derivatives [13], and even cephalosporins and carbapenems often be-
come ineffective against this bacterium, leading to increased mortality of S. aureus-associ-
ated infections [14]. Some of the resistance mechanisms of S. aureus are limiting the drug
uptake, modifying the drug target, inactivating the drug, and active drug efflux [15]. C.
albicans also busts resistance to antifungals along the course of treatment [16] via trans-
forming between several morphological forms (blastospores, pseudohyphae, and hyphae)
[17], decreasing the permeability of drugs, and expressing efflux pumps or compromised
drug import [18].
In addition, biofilm formation plays an important role in S. aureus and C. albicans
protection. Biofilms are microbial communities (either mono- or polymicrobial) where the
cells are embedded into a matrix consisting of polysaccharides, proteins, and nucleotides
produced by the cells themselves [9,19,20]. The biofilm is formed in several stages, includ-
ing attachment to biotic or abiotic surfaces, maturation, and detachment (dispersal of ma-
ture biofilm) [21]. While in biofilm, microorganisms are characterized by a decreased sus-
ceptibility to antimicrobials due to the diffusional barrier for the latter as well as being
more virulent and capable to adhere to surfaces and form new biofilms [22]. Therefore,
the development of new approaches to increase the susceptibility of pathogenic microor-
ganisms to conventional antimicrobials could be promising in overcoming the AMR prob-
lem.
Various classes of compounds were reported to be able to potentiate the efficiency of
antimicrobials against planktonic- and biofilm-embedded bacteria and fungi: derivatives
of 5(H)furanone [23,24], various hydrolytic enzymes [25,26], and essential oils [27,28]. Ter-
penes, the active fraction of essential oils from plant extracts, make up the largest group
of secondary metabolites of plants (over 50,000 known substances) [29]. Monoterpenes
consist of two isoprene units and naturally occur in plants and essential oils [30] and are
introduced as key ingredients in the design and production of novel biologically active
compounds because of anti-inflammatory, antimicrobial, anticonvulsant, analgesic, anti-
viral, anticancer, antituberculosis, and antioxidant biological activities [31–33]. Addition-
ally, some researchers have described the ability of terpenes to inhibit the formation of S.
aureus biofilms as well as their antimicrobial and antifungal activity [34,35]. Myrtenol is a
monoterpene bicyclic derivative that has been well known for its antimicrobial activity
[36]. Myrtenol exhibited antibacterial activity against S. aureus and Acinetobacter baumannii
[37,38] and has repressed the growth of C. albicans, R. nigricans, A. fumigates, and F. solani
fungi species [36]. Several chemically synthesized myrtenol derivatives demonstrated sig-
nificant in vitro antifungal activity against Physalospora piricola with better or comparable
antifungal activity than those of positive controls (the commercial fungicides azoxystrobin
and chlorothalonil) [32]. In addition, the combination of myrtenol and antifungal agents
reduced the effective concentrations of the latter with synergistic and additive effects
[39,40]. The mechanism of myrtenol action is discussible. It has been suggested that myr-
tenol possibly damages the fungal membrane, affecting the change in the functional state
Antibiotics 2022, 11, 1743 3 of 17
of integrin-like proteins, which can lead to the disruption of morphogenesis of the fungal
cell [36].
Here, we show that myrtenol potentiates the antimicrobial and biofilm-preventing
activity of conventional drugs against S. aureus and C. albicans mono- and dual-species
cultures.
2. Results
2.1. Antibacterial and Antifungal Activity of Myrtenol
The antimicrobial activity of myrtenol was evaluated on S. aureus ATCC 29213 as
well as four methicillin-sensitive clinical isolates of S. aureus (MSSA), seven methicillin-
resistant isolates of S. aureus (MRSA), and 10 clinical isolates of C. albicans. (−)-myrtenol
and (+)-myrtenol exhibited low both antibacterial and antifungal activities (Tables 1 and
2). Worth noting, the minimum bactericidal concentration (MBC) either fit or exceeded
the minimum inhibiting concentration (MIC) two-fold, suggesting the bactericidal/fungi-
cidal property of terpenes. Furthermore, MRSA and MSSA were of similar susceptibility
to myrtenol, and the resistance to fluconazole did not affect the susceptibility of C. albicans
isolates to terpene.
Table 1. Minimum inhibitory concentration (MIC) and minimum bactericidal concentration (MBC)
values (expressed in µg/mL) of (−)-myrtenol, (+)-myrtenol, amikacin, and benzalkonium chloride
(BAC) against S. aureus isolates.
Strains (−)-Myrtenol (+)-Myrtenol Amikacin BAC
MIC MBC MIC MBC MIC MBC MIC MBC
S. aureus
ATCC 29213 (MSSA) 1024 1024 512 512 4 8 0.5 1
S. aureus
18 (MSSA) 1024 1024 1024 1024 16 32 0.25 1
S. aureus
25 (MSSA) 1024 1024 1024 1024 8 16 0.25 0.5
S. aureus
26 (MSSA) 1024 1024 512 512 16 16 0.5 1
S. aureus
27 (MSSA) 1024 1024 512 512 4 16 0.5 1
S. aureus
1053 (MRSA) 1024 1024 2048 2048 128 512 0.5 2
S. aureus
1065 (MRSA) 1024 1024 512 512 128 256 0.5 2
S. aureus
1130 (MRSA) 512 1024 512 512 256 512 0.5 2
S. aureus
1145 (MRSA) 1024 512 512 512 64 1024 1 1
S. aureus
1167 (MRSA) 2048 2048 512 512 256 512 0.5 2
S. aureus
1168 (MRSA) 2048 2048 512 512 8 8 0.5 1
S. aureus
1173 (MRSA) 512 512 256 256 256 256 0.5 1
Table 2. Minimum inhibitory concentration (MIC) and minimum fungicidal concentration (MFC)
(expressed in µg/mL) of (−)-myrtenol, (+)-myrtenol, fluconazole and benzalkonium chloride (BAC)
against C. albicans isolates.
Strains ()-Myrtenol (+)-Myrtenol Fluconazole BAC
MIC MFC MIC MFC MIC MFC MIC MFC
C. albicans
722 2048 2048 1024 1024 8 8 0.5 2
C. albicans
761 1024 1024 2048 2048 8 8 0.5 1
C. albicans
661FR 1024 1024 2048 2048 512 512 0.5 1
C. albicans
672 FR 1024 1024 2048 2048 512 512 0.5 0.5
C. albicans
688 FR 1024 1024 2048 2048 512 512 0.5 1
C. albicans
701 FR 2048 2048 2048 2048 512 512 1 1
C. albicans
703 FR 1024 1024 1024 1024 512 512 0.5 2
C. albicans
748 FR 1024 1024 2048 2048 512 512 1 2
C. albicans
762 FR 1024 1024 2048 2048 512 512 1 2
C. albicans
763 FR 2048 2048 2048 2048 512 512 0.5 1
2.2. Myrtenol Potentiates Both Antibacterial and Antifungal Agents
Antibiotics 2022, 11, 1743 4 of 17
The synergism of myrtenol with antimicrobials was assessed using the chequerboard
approach. For S. aureus, the concentrations of amikacin or benzalkonium chloride were in
the range of 0.06–4 × MIC, and myrtenol was added to concentrations of 0.125–1 × MIC.
After 24 h of incubation, the fractional inhibitory concentration index (FICI) was calcu-
lated for both the growth and biofilm repression assessed by crystal violet staining (Sup-
plementary Tables S1-S4). (−)-Myrtenol exhibited a synergistic effect with amikacin with
an FIC index in the range of 0.3–0.5 on 42% of clinical isolates of S. aureus; (+)-myrtenol
led to a four-fold decrease of the MIC of antibiotics against 75% of the studied isolates
regardless of their susceptibility to methicillin (MRSA or MSSA). On the other isolates, the
combined use of amikacin and either (−)- myrtenol or (+)-myrtenol led to an additive effect
(Table 3). On the biofilm-preventing activity, the synergistic effect of (−)-myrtenol and (+)-
myrtenol with amikacin was observed for 42% and 33% of isolates, respectively (Table 3).
In a combination of myrtenol with benzalkonium chloride, in most cases, the additive
effect was observed against the planktonic cells of S. aureus isolates. (−)-Myrtenol potenti-
ated the antiseptic only against one strain, and (+)-myrtenol demonstrated synergy with
benzalkonium chloride against four isolates (33%). Regarding biofilm prevention, (−)-
myrtenol significantly increased the efficiency of benzalkonium chloride against 50% of
isolates with an FICI ranging from 0.16–0.38 while (+)-myrtenol significantly increased the
effectiveness of the antiseptic in only 17% of isolates.
Table 3. FICI values of amikacin and benzalkonium chloride in combination with either (−)-myr-
tenol or (+)-myrtenol against various isolates of S. aureus.
Strains
Amikacin Benzalkonium Chloride
Growth Repression Biofilm Prevention Growth Repression Biofilm Prevention
(−)-Myr-
tenol
(+)-Myr-
tenol
(−)-Myr-
tenol
(+)-Myr-
tenol
(−)-Myr-
tenol
(+)-Myr-
tenol
(−)-Myr-
tenol
(+)-Myr-
tenol
S. aureus
ATCC (MSSA) 0.30 0.50 0.38 1.00 1.25 0.75 0.75 1.50
S. aureus
18 (MSSA) 0.50 0.31 1.12 0.75 0.75 0.5 0.63 0.75
S. aureus
25 (MSSA) 0.75 0.31 1.00 0.50 2.25 0.5 1.50 0.75
S. aureus
26 (MSSA) 0.75 0.75 0.75 1.25 0.75 0.75 0.75 0.28
S. aureus
27 (MSSA) 0.38 0.50 0.75 1.00 0.75 0.75 0.50 1.50
S. aureus
1053 (MRSA) 0.75 0.38 0.38 0.38 0.75 0.5 0.63 0.38
S. aureus
1065 (MRSA) 0.75 0.50 1.12 1.25 0.75 1.25 0.19 2.25
S. aureus
1130 (MRSA) 0.75 0.38 1.00 1.00 0.75 0.75 1.00 1.00
S. aureus
1145 (MRSA) 0.75 0.50 0.25 0.63 1.25 0.5 0.16 0.63
S. aureus
1167 (MRSA) 0.31 0.75 0.31 0.31 0.5 1.25 0.38 0.75
S. aureus
1168 (MRSA) 0.38 0.31 0.28 0.31 0.75 1.25 0.38 1.25
S. aureus
1173 (MRSA) 0.75 0.75 0.625 1.50 1.25 1.25 0.16 0.53
Fraction of strains with shown
synergy 42% 75% 42% 33% 8% 33% 50% 17%
Next, the synergistic effect of myrtenol with fluconazole and benzalkonium chloride
against C. albicans was evaluated. The fractional inhibitory concentration index (FICI) was
calculated for planktonic cell growth repression and biofilm formation prevention (Sup-
plementary Tables S5-S8). (+)-Myrtenol exhibited synergy with fluconazole in 64% of the
C. albicans isolates while (−)-myrtenol mainly led to an additive effect, and synergy was
only observed in 36% of isolates (Table 4). By contrast, when assessing the biofilm repres-
sion, (−)-myrtenol had an FICI less or equal to 0.5 for six out of 11 isolates while (+)-myr-
tenol was only for four out of 11 (Table 4). (-)-Myrtenol also demonstrated synergism with
benzalkonium chloride in relation to planktonic cells for five isolates, and the combined
use of (+)-myrtenol with antiseptic showed a clear synergistic effect in relation to nine
isolates. A similar result was obtained for C. albicans biofilm repression. Most of the iso-
lates (seven out of 11) were more sensitive to the combination of antiseptic with (+)-
Antibiotics 2022, 11, 1743 5 of 17
myrtenol while the use of (−)-myrtenol with benzalkonium chloride showed an FICI less
or equal to 0.5 for only four isolates (Table 4).
Table 4. FICI values of fluconazole and benzalkonium chloride in combination with either (−)-myr-
tenol or (+)-myrtenol against various isolates of C. albicans.
Strains
Fluconazole Benzalkonium Chloride
Growth Repression Biofilm Prevention Growth Repression Biofilm Prevention
(−)-Myr-
tenol
(+)-Myr-
tenol
()-Myr-
tenol
(+)-Myr-
tenol
()-Myr-
tenol
(+)-Myr-
tenol
(−)-Myr-
tenol
(+)-Myr-
tenol
C. albicans
722 1.25 1.25 0.37 0.40 0.50 0.50 1.25 0.31
C. albicans
761 1.25 1.25 0.37 0.50 0.75 0.50 0.38 0.50
C. albicans
661 FR 1.25 0.75 1.25 1.25 0.75 0.38 1.25 0.50
C. albicans
672 FR 0.27 0.50 0.28 0.50 0.75 0.50 0.38 0.75
C. albicans
688 FR 1.25 0.75 0.26 1.25 0.50 0.50 0.75 0.75
C. albicans
701 FR 0.28 0.27 0.75 0.75 0.75 0.75 0.50 0.38
C. albicans
703 FR 0.38 0.27 0.31 4.25 0.75 0.50 0.75 0.75
C. albicans
748 FR 1.25 0.27 0.26 1.25 0.75 0.50 0.75 0.50
C. albicans
762 FR 1.25 0.27 1.25 1.25 0.38 0.50 0.50 0.38
C. albicans
763 FR 0.27 0.50 0.75 0.30 0.50 1.25 0.75 0.50
Fraction of strains with shown
synergy 36% 64% 54% 36% 45% 81% 36% 72%
Thus, these data indicate that myrtenol reduces the effective concentrations of anti-
microbial and antifungal drugs, which, in turn, reduces both the general toxic effect on
the host organism and the risk of resistance development by bacterial and fungal cells.
2.3. Myrtenol Increases the Antimicrobial and Antifungal Activity of Benzalkonium Chloride
against an S. aureus and C. albicans Mixed Culture
Since the benzalkonium chloride demonstrated synergy with myrtenol against both
S. aureus and C. albicans, the effect of their combined use against the fungal–bacterial
mixed culture community was assessed. For this purpose, S. aureus and C. albicans were
cocultivated in a BM broth in a 24-well plate in the presence of benzalkonium chloride in
the concentration range of 0, 0.25, 0.5, 1, 4, or 8 µg/mL solely or in combination with (−)-
myrtenol and (+)-myrtenol at a concentration of 256 µg/mL. After a 24 h incubation, the
viability of planktonic bacterial and fungal cells was assessed by counting CFUs in a series
of ten-fold dilutions followed by plating on selective media for the differentiation of S.
aureus and C. albicans. The sole benzalkonium chloride led to a three-log decrease of viable
S. aureus and the death of C. albicans planktonic cells only at 8 µg/mL (Figure 1). In the
presence of either myrtenol, (−) or (+), a significant increase in the efficiency of the anti-
septic was observed, and the complete death of both C. albicans and S. aureus planktonic
cells was observed at a concentration of 0.5–1 µg/mL, suggesting an eight- to sixteen-fold
increase in the antiseptic’s efficiency by terpene. Worth noting, while the combination of
both (−)-myrtenol and (+)-myrtenol with benzalkonium chloride led to the prevention of
the adherence of S. aureus, although at 4 µg/mL of antiseptic, no significant effect on C.
albicans adherence was observed.
Antibiotics 2022, 11, 1743 6 of 17
Figure 1. Viability of S. aureus and C. albicans in mixed culture in presence of benzalkonium chloride
with concentrations 0, 0.25, 0.5, 1, 4, and 8 µg/mL separately and in combination with (−)-myrtenol
and (+)-myrtenol at a concentration of 256 µg/mL. The viability of bacterial and fungal cells was
assessed after 24 h growth in culture liquid and after 48 for adherent cells. The viable cells were
counted after a series of ten-fold dilutions followed by plating on selective media for differentiation
of S. aureus and C. albicans.
2.4. Myrtenol Damages the Cell Membrane of Bacterial and Fungal Cells
Since damage to the cell membrane has been proposed for various terpenes as the
mechanism of antimicrobial action [41], the effect of myrtenol on the membrane potential
of bacterial and fungal cells was investigated. Cells were preincubated with the fluores-
cent dye DioC2(3) which can be reduced on the membrane of intact cells; then the myr-
tenol was added until 0.5–2×MIC, and the fluorescence was recorded during 30 min of
incubation. As can be seen from Figure 2, in the presence of (−)-myrtenol and (+)-myrtenol,
the fluorescence intensity of S. aureus cells decreased compared to untreated cells in a
dose-dependent manner, confirming a drop in membrane potential, apparently, because
of its damage. A similar drop in fluorescence was observed in cells treated with ben-
zalkonium chloride, which also permeates the cell membrane, while no changes were de-
tected in ampicillin-treated cells. These data clearly suggest that both (−)-myrtenol and
(+)-myrtenol apparently damage the bacterial membrane, thus facilitating the penetration
of antimicrobials into the cell. Treatment of C. albicans cells with low concentrations of (−)-
myrtenol did not affect the fluorescence, similar to fluconazole, although the latter also
affects the integrity of the membrane via repression of the conversion of lanosterol to er-
gosterol (Figure 3). By contrast, (−)-myrtenol led to a significant decrease in fluorescence
comparable with the effect of benzalkonium chloride.
Antibiotics 2022, 11, 1743 7 of 17
Figure 2. Relative membrane potential of S. aureus cells expressed in fluorescence units of DioC2(3)
reduced on an intact cell membrane resulting in green fluorescence. Cells were grown for 18 h in LB
broth, washed with PBS, and resuspended until a final density of 106 CFU/mL in PBS supplemented
with DioC2(3) to a final concentration of 10 µM/mL. After a 30 min incubation at 25°C, compounds
of interest were added to the samples. Fluorescence detection was performed for 30 min with 5 min
intervals with excitation and emission wavelengths of 497 and 520 nm, respectively.
Figure 3. Relative membrane potential of C. albicans cells expressed in fluorescence units of DioC2(3)
reduced on an intact cell membrane resulting in green fluorescence. Cells were grown for 18 h in
Sabouraud broth, washed with PBS, and resuspended until a final density of 105 CFU/mL in PBS
supplemented with DioC2(3) to a final concentration of 10 µM/mL. After a 30 min incubation at
25°C, compounds of interest were added to the samples. Fluorescence detection was performed for
Antibiotics 2022, 11, 1743 8 of 17
30 min with 5 min intervals with excitation and emission wavelengths of 497 and 520 nm, respec-
tively.
To evaluate whether myrtenol binds to the membrane or diffuses into the cell, con-
focal laser scanning microscopy was performed to check the localization of terpenes in
bacterial cells. For this, S. aureus and C. albicans cells were incubated for 15 min in the
presence of myrtenol fused with a fluorophore (myrtenol-lum). Synthesis, physicochemi-
cal properties, and spectral data of BF2-ms-(4-((1″R)-6″,6″-dimethylbicyclo[3.1.1]hept-2″-
ene-2″)ylmethoxycarbonylpropyl)-3,3′,5,5′-tetramethyl-2,2′-dipyrromethene (mentioned
as “lum”) were described in detail in our previous paper [42]. To visualize the membranes
of bacteria and fungi, cells were additionally stained with CalcoFluor-White (CFW). Fig-
ures 4 and 5 show that myrtenol was evenly distributed in S. aureus cells (green fluoresce)
while the fluorophore was observed only over the cell surface, suggesting that myrtenol
diffuses through the membrane. A similar result was shown for C. albicans cells (Figures
4 and 5).
(a) (b)
Figure 4. Myrtenol-lum penetration into S. aureus cells. (a) The localization of either (−)-myrtenol or
(+)-myrtenol carrying the fluorophore BODIPY (Myrtenol-lum) assessed by confocal laser scanning
microscopy. The solely fluorophore (lum) and Calcofluor-White (CFW) membrane dyes served as
references. (b) Penetration of myrtenol-lum into S. aureus cells. Myrtenol-lum was added to S. aureus
cells, and the sole fluorophore (lum) was used as a control. After 4, 8, 16, 32, and 64 min of incuba-
tion, cells were harvested, washed with PBS, and residual fluorescence was measured. The half-time
of penetration (t½) was 24±1.3 min and 26±1.5 min for (−)-myrtenol and (+)-myrtenol, respectively,
while for the fluorophore solely (lum) was t½> 5000 min.
Antibiotics 2022, 11, 1743 9 of 17
(a) (b)
Figure 5. Myrtenol-lum penetration into C. albicans cells. (a) The localization of either (−)-myrtenol
or (+)-myrtenol carrying the fluorophore BODIPY (myrtenol-lum) assessed by confocal laser scan-
ning microscopy. The solely fluorophore (lum) and Calcofluor-White (CFW) membrane dyes served
as references. (b) Penetration of myrtenol-lum into C. albicans cells. Myrtenol-lum was added to C.
albicans cells, and the sole fluorophore (lum) was used as a control. After 4, 8, 16, 32, and 64 min of
incubation, cells were harvested, washed with PBS, and residual fluorescence was measured. The
half-time of penetration (t½) was 24±1.3 min and 18±1.2 min for (−)-myrtenol and (+)-myrtenol, re-
spectively, while for the fluorophore solely (lum) was t½> 5000 min.
In the next step, the rate of penetration of myrtenol into bacterial and fungal cells was
assessed. Myrtenol containing a fluorophore in its structure (Myrtenol-lum) was added to
S. aureus and C. albicans cells. The pure fluorophore (lum) itself was used as a control.
After 4, 8, 16, 32, and 64 min of incubation, cells were harvested, washed with PBS, and
the fluorescence in suspension was measured using a Tecan Infinite 200 Pro microplate
reader (Switzerland). Cells without any added compounds were considered point zero,
and the cell suspension with the fluorescent compound was considered 100%. As can be
seen from Figures 4 and 5, the half-time of maximal penetration (t½) of (+)-myrtenol-lum
was 26±1.5 min and 18±1.2 min for S. aureus and C. albicans, respectively. For (−)-myrtenol-
lum, the calculated t½ was 24 ± 1.3 min while the t½ of the sole fluorophore was t½> 5000
min in both bacterial and fungal cells, suggesting the interaction of myrtenol with the
membrane.
3. Discussion
The worldwide spread of pathogenic bacteria and micromycetes resistant or tolerant
to conventional antimicrobials drastically decreases the number of available options for
the treatment of infectious diseases and thus becomes a global challenge for healthcare
[43–46]. Furthermore, the coexistence of different microorganisms in mixed communities
leads to additional difficulties in treatment compared to monospecific infections [8,11,47].
Due to interbacterial and bacterial–fungal interactions in consortia, their counterparts
Antibiotics 2022, 11, 1743 10 of 17
change metabolism and morphology that consequently leads to altered susceptibility to
antimicrobials [7,48,49]. Therefore, the development of either novel universal antimicro-
bials or approaches to potentiate conventional ones could be tools to overcome the toler-
ance of microorganisms to antimicrobials.
Essential oils have been shown as both potential antimicrobials and enhancers of con-
ventional antimicrobials [27,28]. In particular, the bicyclic monoterpene myrtenol, a ter-
pene from the myrtenol tree, is able to repress the growth of bacteria [37,38] and fungi [36]
as well as reduce the effective concentrations of some antifungals [39,40]. In our study, the
two optical isomers of myrtenol, (−)-myrtenol and (+)-myrtenol, were tested as either an-
tibacterial, antifungal, or enhancers of conventional drugs. As can be seen from Tables 3
and 4, (+)-myrtenol demonstrated the synergistic effect with amikacin, fluconazole, and
benzalkonium chloride on most of the clinical isolates of S. aureus and C. albicans while
(−)-myrtenol exhibited synergy with conventional drugs only on a third of the isolates.
Thus, in the presence of myrtenol, the MICs of amikacin, fluconazole, and benzalkonium
chloride were reduced up to sixteen-fold (see Supplementary file), reaching medically rel-
evant concentrations. On the contrary, (−)-myrtenol more readily increased the property
of amikacin and fluconazole to repress biofilm formation by the S. aureus and C. albicans
isolates, respectively. The reason for such selectivity remains questionable since the half-
time penetration of both (−)-myrtenol and (+)-myrtenol into either S. aureus or C. albicans
was similar at 18–24 min (Figures 4 and 5). Additionally, the confocal microscopy of
treated cells revealed similar intracellular localization of (−)-myrtenol and (+)-myrtenol
fused to the fluorophore. However, in the membrane integrity assay, (+)-myrtenol led to
a faster drop in the membrane potential of treated C. albicans cells (Figure 3), which allows
for speculation about either the specificity of (+)-myrtenol to any molecular target or a
higher tropism to the membrane at least in fungal cells. The last assumption may be less
probable since no difference in the effect of either (−)-myrtenol or (+)-myrtenol on the S.
aureus membrane could be observed (Figure 2). Nevertheless, the mechanism of both (−)-
myrtenol and (+)-myrtenol synergy with conventional drugs is apparently driven by
membrane damage since the treatment with both terpenes led to a drop in membrane
potential similar to the action of benzalkonium chloride (Figures 2 and 3), the membrane-
permeating agent [50–52].
As has been reported in many works, S. aureus and C. albicans are opportunistic path-
ogens that live in the same niche and are capable of forming mixed-species consortia.
These consortia appear widely on various mucosa, including the mouth, vaginal tract, etc.
[10]. In this form, their resistance to antimicrobial and antifungal drugs increases signifi-
cantly [53,54]. Hence, we tested whether either (−)-myrtenol or (+)-myrtenol could poten-
tiate the antiseptic benzalkonium chloride against a mixed culture of S. aureus and C. albi-
cans. As can be seen from Figure 1, in this case, both isomers of myrtenol were able to
potentiate benzalkonium chloride up to sixteen-fold against planktonic cells, which al-
lows for reduction of the concentration of this toxic antiseptic for the treatment of various
mucosa with the same efficiency. On the other hand, the increase in antiseptic efficiency
decreases the risk of resistance development by pathogens [55]. Unfortunately, while the
combination of myrtenol with antiseptic could completely repress the adhesion of S. au-
reus, no effect of terpene on C. albicans adhesion repression by benzalkonium chloride
could be observed. This effect is probably due to the highly adaptive capabilities of the
fungal cells that make it possible to neutralize the negative effect of antimycotics at their
low concentrations.
Taken together, our data allow for the suggestion of myrtenol as a tool to increase the
susceptibility of pathogens to antimicrobials. While the terpene will apparently not be
effective against resistant strains, its combined use with antimicrobials could be helpful
when treating tolerant isolates. The lack of toxicity of terpenes [56,57] makes them a harm-
less and potential therapeutic agent to increase the efficiency of the treatment of bacterial
and fungal infections mediated by resistant strains. Thus, in much previous research, nei-
ther cytotoxicity nor acute toxicity on animals has been found for relatively high
Antibiotics 2022, 11, 1743 11 of 17
concentrations of myrtenol, up to 600 mg/L in vitro and 1.3 g per kg in vivo [58–60] It is
worth mentioning that a crucial benefit from using the described compounds is that their
resource is almost inexhaustible [61,62]. Thus, the knowledge of the clinical and economic
burden of antibiotic-resistant mixed infections, coupled with the benefits of the availabil-
ity of such compounds, will allow for optimal control and improved patient safety [63].
4. Materials and Methods
4.1. Chemistry
The (+)- or (-)-myrtenol were synthesized by the oxidation of (+)- or (-)-α-pinene with
tert-butyl hydroperoxide in the presence of catalytic amounts of SeO2 according to the
reported procedure [64]. The myrtenal formed during the reaction (content 70−75% by
Gas liquid chromatography) was isolated through a water-soluble sulfite derivative (al-
dehyde purity is 97−98%) with subsequent NaBH4 reduction of the aldehyde into myr-
tenol. A yield of 40–42% was observed. The spectral data and physical constants associ-
ated with the compounds obtained fit with the literature data. Synthesis, physicochemical
properties, and spectral data of BF2-ms-(4-((1″R)-6″,6″-dimethylbicyclo[3.1.1]hept-2″-ene-
2″)ylmethoxycarbonylpropyl)-3,3′,5,5′-tetramethyl-2,2′-dipyrromethene (mentioned as
“lum”) were described in detail in our previous paper [42]. A solution of ester 1 (0.128
mmol, 1 equiv) in isopropanol (5 mL) was stirred with 0.1 N KOH (2 mL) under argon
atmosphere at room temperature using thin layer chromatography (TLC) in a 1:10 methyl
tert-butyl ether (MTBE)−CCl4 system to monitor the reaction progress. After almost com-
plete transformation (1−2 h), the mixture was evaporated. Then, 20 mL of toluene and
diluted aqueous HCl were added to the mixture with intensive stirring. The organic layer
was separated and evaporated in vacuo. Then, 0.154 mmol (1.2 equiv) of (−)- or (+)-myr-
tenol and 0.128 mmol of DMAP in 20 mL of dichloromethane (DCM) were added. After
complete dissolution, 0.384 mmol of HATU was added to the mixture. The progress of the
reaction was monitored by TLC with a 1:10 MTBE−CCl4 system. After completion of the
reaction (about 5 h), the solvent was removed in vacuum and the product was purified by
silica gel column chromatography. A 1:19 MTBE−CCl4 mixture served as an eluent. A
yield of 59% was observed. The stock solutions of (−)-myrtenol and (+)-myrtenol were
prepared in pure DMSO at a concentration of 20 g/L. Working solutions were prepared in
a bacterial growth medium with a final concentration of DMSO of no more than 5%, which
is nontoxic for both bacterial and fungal strains. Amikacin (Sigma), benzalkonium chlo-
ride (Sigma), and fluconazole (Sigma) were used as reference antimicrobials.
4.2. Strains and Cultivation Conditions
A methicillin-sensitive Staphylococcus aureus ATCC 29213 as well as 10 clinical
MRSA isolates obtained from the Republican Clinical Hospital, Laboratory of Clinical Bac-
teriology in Kazan were used in this study (see Table 1). The bacterial strains were stored
in 50 % (V/V) glycerol stocks at -80 °C and freshly streaked on LB plates followed by their
overnight growth at 37 °C before use. Ten clinical isolates of Candida albicans (see Table
2 for resistance details) from the patients of Kazan Scientific Research Institute of Epide-
miology and Microbiology (Kazan, Russia) obtained during the year 2019 were used. Iso-
lates were identified as C. albicans by using AuxaColor 2 Colorimetric sugar-assimilation
yeast-identification kit (Bio-Rad) and confirmed on MALDI-TOF mass spectrometry
(Bruker Biotyper system, Bruker Daltonics, Germany). Fungal strains were stored as a 50%
glycerol stock at −80 °C and grown in the RPMI broth. The overnight cultures were used
to adjust an optical density to 0.5 McFarland (equivalent to 108 cells/mL) in growth me-
dium and used as a working suspension. To obtain a mature biofilm, fungal and bacterial
cells were seeded in TC-treated culture plates (at 106 cells/mL) and grown under static
conditions for 48 h at 37°C in BM-broth [65] supplemented with 1% glucose. Mannitol salt
agar and Sabouraud agar supplemented with ciprofloxacin (20 µg/mL) were used for the
Antibiotics 2022, 11, 1743 12 of 17
differential count of CFUs of S. aureus and C. albicans, respectively, in S. aureusC. albicans
mixed cultures.
4.3. Determination of the Minimum Inhibitory (MIC) and the Minimum Bactericidal/Fungicidal
Concentrations (MBC/MFC)
The minimum inhibitory concentration (MIC) was determined by serial microdilu-
tion in 96-well plates according to the EUCAST rules for antimicrobial susceptibility test-
ing [66]. The highest final concentration of each compound was 512 µg/mL. The next wells
contained two-fold decreasing concentrations of compound in the range of 0.5–1024
µg/mL. The wells were seeded with microbial culture to obtain density of 106 CFU/mL in
a volume of 200 µL per well. The plates were incubated under static conditions at 37°C
for 24 in case of bacterial culture and 48 h for yeast. The growth was assessed by measur-
ing the optical density at wavelength of 600 nm. The minimum inhibitory concentration
of the compound was defined as the concentration providing complete suppression of the
visible growth of cells. The minimum bactericidal/fungicidal concentration (MBC/MFC)
was determined by seeding 5 µL of culture fluid from wells with no visible growth in 3
mL of fresh nutrient broth. The MBC/MFC was considered the minimum concentration of
the studied compound, which ensures the complete absence of growth [67].
4.4. Determination of the Biofilm Prevention Concentration (BPC)
To determine the biofilm prevention concentration (BPC), bacterial and fungal cells
were grown in 96-well adhesive plates for 48 h under static conditions at 37 °C in BM
broth in wells of 200 µL with an initial density of 106 CFUs / ml in the presence of the test
substances. Next, staining with crystal violet was carried out as described in [68]. The
minimum biofilm inhibitory concentration was defined as the lowest concentration
providing no visible staining of the residual biofilm.
4.5. Analysis of the Antimicrobial Effect in the Combined Use of Antimicrobial Agents
(Chequerboard Approach)
The chequerboard approach was used to assess the possibility of increasing the ef-
fectiveness of other antimicrobial agents with myrtenol. The experimental methodology
was similar to the determination of the MIC in 96-well plates. Each plate contained serial
dilutions of a myrtenol derivative and various compounds in a chequerboard pattern, as
described previously [69]. One of the antimicrobial substances [A] was twice diluted hor-
izontally, and the other [B] vertically on a 96-well plate. The result was a combination of
77 concentrations of antimicrobial compounds [A] and [B]. The extreme lines and columns
contained only one of the considered substances to determine their MICs directly in the
experiment. The initial concentration of each studied antimicrobial agent was 4×MIC. The
final concentration of bacterial and fungal cells in the wells was 0.5×105 CFU/mL. The
plates were incubated at 37°C for 20 h. Then, the optical density OD600 was measured on
an Infinite 200 PRO plate spectrophotometer (Tecan, Switzerland). Each test was run in
triplicate and included a growth control without the addition of any antimicrobial agent.
The fractional inhibitory concentration index (FICI) for each double combination was cal-
culated as follows:
FICI =
[]  
 [] +[]  
[]
(1)
Interpretation of the obtained FICI values was carried out according to [70,71]; FICI
0.5 corresponded to synergy, 0.5 < IFIC 4 to an additive effect while IFIC > 4 corre-
sponded to antagonism.
4.6. Evaluation of Viability of Bacterial and Fungal Cells
To assess the viability of planktonic cells, samples from the upper layer of the culture
liquid were taken. Then the culture liquid was removed from the wells; wells were
Antibiotics 2022, 11, 1743 13 of 17
washed several times with a sterile NaCl solution (0.9%) to remove planktonic and de-
tached cells. The biofilms were mechanically destroyed, and cells were resuspended in a
sterile NaCl solution (0.9%). The viability of cells was evaluated by the drop plate assay
with minor modifications [72]. Serial ten-fold dilutions from each well were prepared, and
5 µL of suspension was dropped on Mannitol salt agar and Sabouraud agar with ciprof-
loxacin (20 µg/mL) to differentiate S. aureus and C. albicans cells, respectively. After 24 h
of incubation at 37°C, the number of colonies on the plates was counted; the values were
averaged and expressed as CFU/mL.
4.7. Membrane Potential Evaluation
Membrane potential was evaluated by the detection of 3,3’-diethyloxacarbocyanine
iodide (DioC2(3)) fluorescence as an indicator of the membrane potential level. Bacterial
or fungal cells were grown for 18 h in LB broth with stirring, then harvested and washed
with PBS. Cells were resuspended until a final density of 106 CFU/mL was reached in PBS
supplemented with DioC2(3) to a final concentration of 10 µM/mL. C. albicans cells were
resuspended until a final density of 105 CFU/mL. After a 30 min incubation at 25°C, com-
pounds were added to the samples. Fluorescence detection was performed for 30 min with
5 min intervals using carboxyfluorescein (FAM) wavelength detection (the excitation and
emission wavelengths were 497 and 520 nm, respectively).
4.8. Estimation of the Penetration Rate of Myrtenol into Bacterial and Fungal Cells
To assess the penetration rate of terpenoids into bacterial cells, (-)-myrtenol-lum and
(+)-myrtenol-lum, which contain a fluorophore (lum) in their structures, were used. Bac-
terial and fungal cells were grown overnight at 37 °C in LB culture medium with agitation,
then washed with BPS (pH = 7.4), and resuspended in a buffer to an optical density of 0.5
by McFarland. Either (-)-myrtenol-lum or (+)-myrtenol-lum was added to a final concen-
tration of 10 µg/mL and incubated at 25 °C in the dark. Pure fluorophore was used as a
control. After 4, 8, 16, 32, and 64 min of incubation, 150 µL of the suspension was taken;
cells were harvested by centrifugation, washed with PBS, and then resuspended in 150 µL
of the buffer. The fluorescence was measured using a Tecan Infinite 200 Pro microplate
reader (at an excitation and emission wavelength of 485 and 520 nm, respectively). The
time required to obtain half of the maximum fluorescence of stained cells (t 1/2) was cal-
culated by plotting log10 (time) as a function of percent fluorescence (taking into account
the fluorescence of unstained cells as 0% and the fluorescence of cell suspension in buffer
with test compound (10 µg/mL) as 100%) in GraphPad Prism 6. Additionally, the pene-
tration of either (−)-myrtenol-lum or (+)-myrtenol-lum in bacterial and fungal cells and
their localization there were assessed by confocal laser scanning microscopy on micro-
scope. (−)-Myrtenol-lum, (+)-myrtenol-lum, or pure fluorophore were added to the cells
at a concentration of 10 µg/mL. Cell membranes were additionally stained with calcofluor
dye (1 mg/mL). As a result, the membranes that were stained in blue (excitation emission)
and green fluorescence (excitation emission) indicated the localization of terpenes in the
cells.
4.9. Data Analysis
All experiments were performed in three biological replicates with three technical
replicates in each experiment. The data were analyzed and visualized using GraphPad
Prism version 6.00 for Windows (GraphPad Software, USA, www.graphpad.com). In each
experiment, a comparison with a negative control was performed using the nonparamet-
ric Kruskal–Wallis test of variance. Significant differences from control were considered
at p<0.05.
5. Conclusions
Antibiotics 2022, 11, 1743 14 of 17
Both (−)-myrtenol and (+)-myrtenol have weak antibacterial and antifungal activity
while demonstrating nonstrain-specific bactericidal and fungicidal effects and exhibiting
synergism with amikacin and benzalkonium chloride in relation to planktonic cells and
biofilms. The mechanism of these effects appears as a consequence of the membranotropic
property of the compound against bacterial and fungal cells. This may be considered as
further validation that these compounds contribute to an increase in the effectiveness of
various antimicrobial, antifungal, and antiseptic drugs, manifesting synergy with these
compounds. Moreover, our findings confirm that terpene derivatives increase the effec-
tiveness of benzalkonium chloride against microorganisms in the mixed community of S.
aureus and C. albicans. Thus, due to the low toxicity of terpenes, these compounds could
become promising agents in the treatment of infections caused by bacteria and by fungi
of the genus Candida as well as mixed fungal–bacterial infections, including resistant
strains.
Supplementary Materials: Table S1. MIC, FIC, and FICI values of amikacin in combination with
either (−)-myrtenol or (+)-myrtenol (expressed in µg/mL) against various isolates of S. aureus. Table
S2. Biofilm preventing concentrations (BPC), fractional biofilm preventing concentrations, and FICI
values of amikacin in combination with either (−)-myrtenol or (+)-myrtenol (expressed in µg/mL)
against various isolates of S. aureus. Table S3. MIC, FIC, and FICI values of benzalkonium chloride
(BAC) in combination with either (−)-myrtenol or (+)-myrtenol (expressed in µg/mL) against various
isolates of S. aureus. Table S4. Biofilm preventing concentrations (BPC), fractional biofilm preventing
concentrations, and FICI values of benzalkonium chloride (BAC) in combination with either (−)-
myrtenol or (+)myrtenol (expressed in µg/mL) against various isolates of S. aureus. Table S5. MIC,
FIC, and FICI values of fluconazole in combination with either (−)-myrtenol or (+)-myrtenol (ex-
pressed in µg/mL) against various isolates of C. albicans. Table S6. Biofilm preventing concentrations
(BPC), fractional biofilm preventing concentrations, and FICI values of fluconazole in combination
with either (−)-myrtenol or (+)-myrtenol (expressed in µg/mL) against various isolates of C. albicans.
Table S7. MIC, FIC, and FICI values of benzalkonium chloride (BAC) in combination with either (−)-
myrtenol or (+)-myrtenol (expressed in µg/mL) against various isolates of C. albicans. Table S8. Bio-
film preventing concentrations (BPC), fractional biofilm preventing concentrations, and FICI values
of benzalkonium chloride (BAC) in combination with either (−)-myrtenol or (+)-myrtenol (expressed
in µg/mL) against various isolates of C. albicans.
Author Contributions: Conceptualization, L.L.F., A.V.K., G.B.G., E.V.A., M.B.B., L.E.N. and A.R.K;
Data curation, A.R.K; Formal analysis, L.E.N.; Funding acquisition, A.R.K and E.V.A.; Investigation,
R.Y.M., E.Y.T., R.K.S., R.S.P., I.R.G., S.A.L., O.V.O., L.L.F., A.V.K., G.B.G., E.V.A. and M.B.B.; Meth-
odology, R.Y.M., E.Y.T., R.K.S., R.S.P., I.R.G., O.V.O., L.L.F., A.V.K., G.B.G., E.V.A. and M.B.B.; Pro-
ject administration, E.Y.T., L.E.N. and A.R.K; Resources, L.E.N. and A.R.K; Supervision, L.E.N. and
A.R.K; Validation, L.E.N. and A.R.K; Visualization, R.Y.M. and E.Y.T.; Writing—original draft,
R.Y.M., E.Y.T., R.K.S. and A.R.K; Writing—review and editing, R.Y.M., E.Y.T., R.K.S. and A.R.K. All
authors have read and agreed to the published version of the manuscript.
Funding: This research was funded by the Russian Science Foundation (grant N 20-64-47014 to
A.R.K.). The synthesis of BODIPY (BF2-ms-methoxycarbonylpropyl-3,3′,5,5′-tetramethyl-2,2′-dipyr-
romethene) was carried out in the ISC RAS under the Russian Science Foundation Project (grant N
20-63-47026 to E.V.A.).
Institutional Review Board Statement: Not applicable.
Informed Consent Statement: Not applicable.
Data Availability Statement: All data are included in the manuscript
Acknowledgments: This research was performed in frames of the Kazan Federal University Strate-
gic Academic Leadership Program (PRIORITY-2030).
Conflicts of Interest: The authors declare no conflict of interest.
References
1. Khan, H.A.; Baig, F.K.; Mehboob, R. Nosocomial infections: Epidemiology, prevention, control and surveillance. Asian Pac. J.
Trop. Biomed. 2017, 7, 478–482.
Antibiotics 2022, 11, 1743 15 of 17
2. Rangelova, V.; Kevorkyan, A.; Krasteva, M. Nosocomial infections in the neonatal intensive care unit. Arch. Balk Med. Union
2020, 55, 121–127.
3. Petrosillo, N.; Pagani, L.; Ippolito, G. Nosocomial infections in HIV-positive patients: An overview. Infection 2003, 31, 28–34.
4. Bardi, T.; Pintado, V.; Gomez-Rojo, M.; Escudero-Sanchez, R.; Azzam Lopez, A.; Diez-Remesal, Y.; Martinez Castro, N.; Ruiz-
Garbajosa, P.; Pestaña, D. Nosocomial infections associated to COVID-19 in the intensive care unit: Clinical characteristics and
outcome. Eur. J. Clin. Microbiol. Infect. Dis. 2021, 40, 495–502.
5. Abushaheen, M.A.; Fatani, A.J.; Alosaimi, M.; Mansy, W.; George, M.; Acharya, S.; Rathod, S.; Divakar, D.D.; Jhugroo, C.;
Vellappally, S. Antimicrobial resistance, mechanisms and its clinical significance. Disease-a-Month 2020, 66, 100971.
6. Brogden, K.A.; Guthmiller, J.M.; Taylor, C.E. Human polymicrobial infections. Lancet 2005, 365, 253–255.
7. Murray, J.L.; Connell, J.L.; Stacy, A.; Turner, K.H.; Whiteley, M. Mechanisms of synergy in polymicrobial infections. J. Microbiol.
2014, 52, 188–199.
8. Little, W.; Black, C.; Smith, A.C. Clinical implications of polymicrobial synergism effects on antimicrobial susceptibility.
Pathogens 2021, 10, 144.
9. Rodrigues, M.E.; Gomes, F.; Rodrigues, C.F. Candida spp./bacteria mixed biofilms. J. Fungi 2019, 6, 5.
10. Carolus, H.; Van Dyck, K.; Van Dijck, P. Candida albicans and Staphylococcus species: A threatening twosome. Front. Microbiol.
2019, 10, 2162.
11. Harriott, M.M.; Noverr, M.C. Candida albicans and Staphylococcus aureus form polymicrobial biofilms: Effects on
antimicrobial resistance. Antimicrob. Agents Chemother. 2009, 53, 3914–3922.
12. Harriott, M.M.; Noverr, M.C. Ability of Candida albicans mutants to induce Staphylococcus aureus vancomycin resistance
during polymicrobial biofilm formation. Antimicrob. Agents Chemother. 2010, 54, 3746–3755.
13. Algammal, A.M.; Hetta, H.F.; Elkelish, A.; Alkhalifah, D.H.H.; Hozzein, W.N.; Batiha, G.E.-S.; El Nahhas, N.; Mabrok, M.A.
Methicillin-Resistant Staphylococcus aureus (MRSA): One health perspective approach to the bacterium epidemiology,
virulence factors, antibiotic-resistance, and zoonotic impact. Infect. Drug Resist. 2020, 13, 3255.
14. Gajdács, M. The continuing threat of methicillin-resistant Staphylococcus aureus. Antibiotics 2019, 8, 52.
15. Reygaert, W.C. An overview of the antimicrobial resistance mechanisms of bacteria. AIMS Microbiol. 2018, 4, 482.
16. Costa-de-Oliveira, S.; Rodrigues, A.G. Candida albicans antifungal resistance and tolerance in bloodstream infections: The triad
yeast-host-antifungal. Microorganisms 2020, 8, 154.
17. Talapko, J.; Juzbašić, M.; Matijević, T.; Pustijanac, E.; Bekić, S.; Kotris, I.; Škrlec, I. Candida albicans—The virulence factors and
clinical manifestations of infection. J. Fungi 2021, 7, 79.
18. Prasad, R.; Nair, R.; Banerjee, A. Emerging mechanisms of drug resistance in Candida albicans. In Yeasts in Biotechnology and
Human Health; Springer: Berlin/Heidelberg, Germany, 2019; pp. 135–153.
19. Flemming, H.-C.; Wingender, J. The biofilm matrix. Nat. Rev. Microbiol. 2010, 8, 623–633.
20. Flemming, H.-C.; Wingender, J.; Szewzyk, U.; Steinberg, P.; Rice, S.A.; Kjelleberg, S. Biofilms: An emergent form of bacterial
life. Nat. Rev. Microbiol. 2016, 14, 563–575.
21. Wu, H.; Moser, C.; Wang, H.-Z.; Høiby, N.; Song, Z.-J. Strategies for combating bacterial biofilm infections. Int. J. Oral Sci. 2015,
7, 1–7.
22. Pereira, R.; dos Santos Fontenelle, R.O.; de Brito, E.H.S.; de Morais, S.M. Biofilm of Candida albicans: Formation, regulation and
resistance. J. Appl. Microbiol. 2021, 131, 11–22.
23. Kayumov, A.R.; Sharafutdinov, I.S.; Trizna, E.Y.; Bogachev, M.I. Antistaphylococcal activity of 2 (5H)-furanone derivatives. In
New and Future Developments in Microbial Biotechnology and Bioengineering: Microbial Biofilms; Elsevier: Amsterdam, The
Netherlands, 2020; pp. 77–89.
24. Sharafutdinov, I.S.; Ozhegov, G.D.; Sabirova, A.E.; Novikova, V.V.; Lisovskaya, S.A.; Khabibrakhmanova, A.M.; Kurbangalieva,
A.R.; Bogachev, M.I.; Kayumov, A.R. Increasing susceptibility of drug-resistant Candida albicans to fluconazole and terbinafine
by 2 (5 H)-furanone derivative. Molecules 2020, 25, 642.
25. Baidamshina, D.R.; Trizna, E.Y.; Holyavka, M.G.; Bogachev, M.I.; Artyukhov, V.G.; Akhatova, F.S.; Rozhina, E.V.; Fakhrullin,
R.F.; Kayumov, A.R. Targeting microbial biofilms using Ficin, a nonspecific plant protease. Sci. Rep. 2017, 7, 46068.
26. Baidamshina, D.R.; Koroleva, V.A.; Olshannikova, S.S.; Trizna, E.Y.; Bogachev, M.I.; Artyukhov, V.G.; Holyavka, M.G.;
Kayumov, A.R. Biochemical properties and anti-biofilm activity of chitosan-immobilized papain. Mar. Drugs 2021, 19, 197.
27. Sudarikov, D.V.; Gyrdymova, Y.V.; Borisov, A.V.; Lukiyanova, J.M.; Rumyantcev, R.V.; Shevchenko, O.G.; Baidamshina, D.R.;
Zakarova, N.D.; Kayumov, A.R.; Sinegubova, E.O. Synthesis and Biological Activity of Unsymmetrical Monoterpenylhetaryl
Disulfides. Molecules 2022, 27, 5101.
28. Szczepanski, S.; Lipski, A. Essential oils show specific inhibiting effects on bacterial biofilm formation. Food Control 2014, 36,
224–229.
29. Wojtunik-Kulesza, K.A.; Kasprzak, K.; Oniszczuk, T.; Oniszczuk, A. Natural monoterpenes: Much more than only a scent. Chem.
Biodivers. 2019, 16, e1900434.
30. Mohammed, A.E.; Abdul-Hameed, Z.H.; Alotaibi, M.O.; Bawakid, N.O.; Sobahi, T.R.; Abdel-Lateff, A.; Alarif, W.M. Chemical
diversity and bioactivities of monoterpene indole alkaloids (MIAs) from six Apocynaceae genera. Molecules 2021, 26, 488.
31. Soares-Castro, P.; Soares, F.; Santos, P.M. Current advances in the bacterial toolbox for the biotechnological production of
monoterpene-based aroma compounds. Molecules 2020, 26, 91.
Antibiotics 2022, 11, 1743 16 of 17
32. Zielińska-Błajet, M.; Feder-Kubis, J. Monoterpenes and their derivatives—Recent development in biological and medical
applications. Int. J. Mol. Sci. 2020, 21, 7078.
33. Elbe, H.; Yigitturk, G.; Cavusoglu, T.; Uyanikgil, Y.; Ozturk, F. Apoptotic effects of thymol, a novel monoterpene phenol, on
different types of cancer. Bratisl. Lek. Listy 2020, 121, 122–128.
34. Kifer, D.; Mužinić, V.; Klarić, M.Š. Antimicrobial potency of single and combined mupirocin and monoterpenes, thymol,
menthol and 1, 8-cineole against Staphylococcus aureus planktonic and biofilm growth. J. Antibiot. 2016, 69, 689–696.
35. Zacchino, S.A.; Butassi, E.; Cordisco, E.; Svetaz, L.A. Hybrid combinations containing natural products and antimicrobial drugs
that interfere with bacterial and fungal biofilms. Phytomedicine 2017, 37, 14–26.
36. Nikitina, L.E.; Lisovskaya, S.A.; Startseva, V.A.; Frolova, L.L.; Kutchin, A.V.; Shevchenko, O.G.; Ostolopovskaya, O.V.;
Pavelyev, R.S.; Khelkhal, M.A.; Gilfanov, I.R. Biological Activity of Bicyclic Monoterpene Alcohols. Bionanoscience 2021, 11, 970–
976.
37. Selvaraj, A.; Valliammai, A.; Sivasankar, C.; Suba, M.; Sakthivel, G.; Pandian, S.K. Antibiofilm and antivirulence efficacy of
myrtenol enhances the antibiotic susceptibility of Acinetobacter baumannii. Sci. Rep. 2020, 10, 21975.
38. Cordeiro, L.; Figueiredo, P.; Souza, H.; Sousa, A.; Andrade-Júnior, F.; Barbosa-Filho, J.; Lima, E. Antibacterial and antibiofilm
activity of myrtenol against Staphylococcus aureus. Pharmaceuticals 2020, 13, 133.
39. Maione, A.; La Pietra, A.; de Alteriis, E.; Mileo, A.; De Falco, M.; Guida, M.; Galdiero, E. Effect of Myrtenol and Its Synergistic
Interactions with Antimicrobial Drugs in the Inhibition of Single and Mixed Biofilms of Candida auris and Klebsiella
pneumoniae. Microorganisms 2022, 10, 1773.
40. Cavalcanti, B.B.; Neto, H.D.; da Silva-Rocha, W.P.; de Oliveira Lima, E.; Barbosa Filho, J.M.; de Castro, R.D.; Sampaio, F.C.;
Guerra, F.Q.S. Inhibitory Effect of (-)-myrtenol alone and in combination with antifungal agents on Candida spp. Res. Soc. Dev.
2021, 10, e35101522434–e35101522434.
41. Guimarães, A.C.; Meireles, L.M.; Lemos, M.F.; Guimarães, M.C.C.; Endringer, D.C.; Fronza, M.; Scherer, R. Antibacterial activity
of terpenes and terpenoids present in essential oils. Molecules 2019, 24, 2471.
42. Guseva, G.B.; Antina, E.V.; Berezin, M.B.; Pavelyev, R.S.; Kayumov, A.R.; Ostolopovskaya, O.V.; Gilfanov, I.R.; Frolova, L.L.;
Kutchin, A.V.; Akhverdiev, R.F. Design, spectral characteristics, and possibilities for practical application of BODIPY FL-labeled
monoterpenoid. ACS Appl. Bio Mater. 2021, 4, 6227–6235.
43. Tanwar, J.; Das, S.; Fatima, Z.; Hameed, S. Multidrug resistance: An emerging crisis. Interdiscip. Perspect. Infect. Dis. 2014, 2014,
541340.
44. Borgio, J.F.; Rasdan, A.S.; Sonbol, B.; Alhamid, G.; Almandil, N.B.; AbdulAzeez, S. Emerging Status of Multidrug-Resistant
Bacteria and Fungi in the Arabian Peninsula. Biology 2021, 10, 1144.
45. Kaye, K.S.; Kaye, D. Multidrug-resistant pathogens: Mechanisms of resistance and epidemiology. Curr. Infect. Dis. Rep. 2000, 2,
391–398.
46. Gulshan, K.; Moye-Rowley, W.S. Multidrug resistance in fungi. Eukaryot. Cell 2007, 6, 1933–1942.
47. Kim, H.-J.; Na, S.W.; Alodaini, H.A.; Al-Dosary, M.A.; Nandhakumari, P.; Dyona, L. Prevalence of multidrug-resistant bacteria
associated with polymicrobial infections. J. Infect. Public Health 2021, 14, 1864–1869.
48. Orazi, G.; O’Toole, G.A. “It takes a village”: Mechanisms underlying antimicrobial recalcitrance of polymicrobial biofilms. J.
Bacteriol. 2019, 202, e00530-19.
49. Kulshrestha, A.; Gupta, P. Polymicrobial interaction in biofilm: Mechanistic insights. Pathog. Dis. 2022, 80, ftac010.
50. Romanowski, E.G.; Mah, F.S.; Kowalski, R.P.; Yates, K.A.; Gordon, Y.J. Benzalkonium chloride enhances the antibacterial
efficacy of gatifloxacin in an experimental rabbit model of intrastromal keratitis. J. Ocul. Pharmacol. Ther. 2008, 24, 380–384.
51. Shadman, S.A.; Sadab, I.H.; Noor, M.S.; Khan, M.S. Development of a benzalkonium chloride based antibacterial paper for
health and food applications. ChemEngineering 2021, 5, 1.
52. Richards, R.M.E.; Mizrahi, L.M. Differences in antibacterial activity of benzalkonium chloride. J. Pharm. Sci. 1978, 67, 380–383.
53. Todd, O.A.; Peters, B.M. Candida albicans and Staphylococcus aureus pathogenicity and polymicrobial interactions: Lessons
beyond koch’s postulates. J. Fungi 2019, 5, 81.
54. Kong, E.F.; Tsui, C.; Kucharíková, S.; Andes, D.; Van Dijck, P.; Jabra-Rizk, M.A. Commensal protection of Staphylococcus aureus
against antimicrobials by Candida albicans biofilm matrix. MBio 2016, 7, e01365-16.
55. Tagkopoulos, I. Benzalkonium Chlorides: Uses, Regulatory Status, and Microbial Resistance. Appl. Environ. Microbiol. 2019, 85,
e00377-19.
56. Api, A.M.; Belsito, D.; Botelho, D.; Bruze, M.; Burton, G.A., Jr.; Buschmann, J.; Dagli, M.L.; Date, M.; Dekant, W.; Deodhar, C.
RIFM fragrance ingredient safety assessment, myrtenol, CAS Registry Number 515-00-4. Food Chem. Toxicol. 2019, 130, 110602.
57. Gomes, B.S.; Neto, B.P.S.; Lopes, E.M.; Cunha, F.V.M.; Araújo, A.R.; Wanderley, C.W.S.; Wong, D.V.T.; Júnior, R.C.P.L.; Ribeiro,
R.A.; Sousa, D.P. Anti-inflammatory effect of the monoterpene myrtenol is dependent on the direct modulation of neutrophil
migration and oxidative stress. Chem. Biol. Interact. 2017, 273, 73–81.
58. Huang, S.; Tan, Z.; Cai, J.; Wang, Z.; Tian, Y. Myrtenol improves brain damage and promotes angiogenesis in rats with cerebral
infarction by activating the ERK1/2 signalling pathway. Pharm. Biol. 2021, 59, 582–591.
59. Selvaraj, A.; Jayasree, T.; Valliammai, A.; Pandian, S.K. Myrtenol attenuates MRSA biofilm and virulence by suppressing sarA
expression dynamism. Front. Microbiol. 2019, 10, 2027.
60. Bhatia, S.P.; McGinty, D.; Letizia, C.S.; Api, A.M. Fragrance material review on myrtenol. Food Chem. Toxicol. 2008, 46, S237–
S240.
Antibiotics 2022, 11, 1743 17 of 17
61. Mahizan, N.A.; Yang, S.-K.; Moo, C.-L.; Song, A.A.-L.; Chong, C.-M.; Chong, C.-W.; Abushelaibi, A.; Lim, S.-H.E.; Lai, K.-S.
Terpene derivatives as a potential agent against antimicrobial resistance (AMR) pathogens. Molecules 2019, 24, 2631.
62. Noriega, P. Terpenes in Essential Oils: Bioactivity and Applications. In Terpenes and Terpenoids: Recent Advances; Books on
Demand: Norderstedt, Germany, 2020.
63. Naylor, N.R.; Atun, R.; Zhu, N.; Kulasabanathan, K.; Silva, S.; Chatterjee, A.; Knight, G.M.; Robotham, J.V. Estimating the
burden of antimicrobial resistance: A systematic literature review. Antimicrob. Resist. Infect. Control 2018, 7, 58.
64. Kiesgen de Richter, R.; Bonato, M.; Follet, M.; Kamenka, J.M. The (+)-and (-)-[2-(1, 3-dithianyl)] myrtanylborane. Solid and stable
monoalkylboranes for asymmetric hydroboration. J. Org. Chem. 1990, 55, 2855–2860.
65. Kayumov, A.R.; Khakimullina, E.N.; Sharafutdinov, I.S.; Trizna, E.Y.; Latypova, L.Z.; Thi Lien, H.; Margulis, A.B.; Bogachev,
M.I.; Kurbangalieva, A.R. Inhibition of biofilm formation in Bacillus subtilis by new halogenated furanones. J. Antibiot. 2015, 68,
297–301.
66. Leclercq, R.; Cantón, R.; Brown, D.F.J.; Giske, C.G.; Heisig, P.; MacGowan, A.P.; Mouton, J.W.; Nordmann, P.; Rodloff, A.C.;
Rossolini, G.M. EUCAST expert rules in antimicrobial susceptibility testing. Clin. Microbiol. Infect. 2013, 19, 141–160.
67. Testing, E.C. on A.S. European committee for antimicrobial susceptibility testing of the european society of clinical, M. &
infectious, D. EUCAST definitive document E. DEF 3.1, June 2000: Determination of minimum inhibitory concentrations (MICs)
of antibacterial agents by aga. Clin Microbiol Infect 2000, 6, 509–515.
68. Merritt, J.H.; Kadouri, D.E.; O’Toole, G.A. Growing and analyzing static biofilms. Curr. Protoc. Microbiol. 2011, 22, 1B-1.
69. Stein, C.; Makarewicz, O.; Forstner, C.; Weis, S.; Hagel, S.; Löffler, B.; Pletz, M.W. Should daptomycin–rifampin combinations
for MSSA/MRSA isolates be avoided because of antagonism? Infection 2016, 44, 499–504.
70. Odds, F.C. Synergy, antagonism, and what the chequerboard puts between them. J. Antimicrob. Chemother. 2003, 52, 1.
71. Den Hollander, J.G.; Mouton, J.W.; Verbrugh, H.A. Use of pharmacodynamic parameters to predict efficacy of combination
therapy by using fractional inhibitory concentration kinetics. Antimicrob. Agents Chemother. 1998, 42, 744–748.
72. Herigstad, B.; Hamilton, M.; Heersink, J. How to optimize the drop plate method for enumerating bacteria. J. Microbiol. Methods
2001, 44, 121–129.
... Among such approaches, the use of essential oils or crude phytoextracts is widely offered [11][12][13], where various terpenes and terpenoids are the active components. These compounds have been shown to be both enhancers of conventional drugs and solely antifungals, although with moderate activity [14][15][16][17][18][19][20][21]. Being of small size, these compounds and their conjugates with other pharmacophores could penetrate cells and affect them. ...
... Being of small size, these compounds and their conjugates with other pharmacophores could penetrate cells and affect them. Thus, the activity of fluconazole has been reported to be significantly increased by myrtenol [19] and a conjugate of borneol with 2(5H)furanone [18], suggesting these molecules as promising adjuvants. ...
... Since for various terpenes and their derivatives a synergy with antifungals has been reported previously [18,19], the ability of KS1 to reduce the MIC of antifungals (fluconazole and terbinafine) has been tested on five clinical isolates of C. albicans. The values of the FICI were determined as described in the materials and methods, and the median value from several wells was calculated. ...
Article
Background: In the last decade, the wide spread of the multidrug resistant yeast C. albicans has challenged the development of new approaches to treatment. Among various options, the use of natural and synthetic terpenes as antifungals or enhancers of conventional drugs has been offered in many works. Here we show the effect of the recently synthesized compound isobornane sulfide named KS1 on C. albicans and discuss its potential mechanism of action. Methods: To characterize the impact of the compound on the yeast minimum inhibitory concentration (MIC) determination on clinical isolates, quantitative PCR with reverse transcriptase (qRT-PCR) of efflux genes expression, cytotoxicity determination on eukaryotic cells, bright-field and fluorescent microscopy with KS1 conjugate with BODIPY fluorophore have been used. The in silico prediction of KS1 properties has been performed using the ADMET server, and molecular docking was used for the modeling of KS1 interaction with putative targets. Results: While exhibiting moderate antifungal activity as compared to fluconazole and terbinafine, KS1 has a synergy with both antifungals on resistant clinical isolates while not stimulating the expression of the CDR1 and MDR1 efflux genes. Moreover, KS1 represses hypha formation by C. albicans, reducing the number of germ tubes more than twice compared to the control. It readily penetrates the cell, as shown by confocal microscopy using the created KS1-BODIPY fluorophore conjugate, apparently thereby facilitating the penetration of antifungals into the cell. As judged by the ADMET server, KS1 falls into the category of drug-like compounds and neither inhibits the isoforms of cytochrome P450 nor manifests mutagenicity or carcinogenicity that fits with in vitro data. The molecular docking showed that KS1 has a high affinity for the transcription activator transcription activator (Tec1) protein of C. albicans, responsible for invasion and hypha formation, which fits with the in vitro data. Conclusions: These findings suggest KS1 as promising both a solely antifungal and an enhancer of conventional antimycotics blocking fungal virulence.
... Several infections are not due to a single pathogen alone. This is the case with infections caused by Staphylococcus aureus and Candida albicans, which include periodontitis, cystic fibrosis, denture stomatitis, urinary tract infections, burn wound infections, and infections associated with invasive medical devices such as venous catheters [14]. Mahmoud and co-authors [14] studied the antimicrobial activity of the monoterpene bicyclic derivative myrtenol in pure and mixed cultures of both organisms, as well as in combination with antibacterial, antifungal, and disinfectant compounds. ...
... This is the case with infections caused by Staphylococcus aureus and Candida albicans, which include periodontitis, cystic fibrosis, denture stomatitis, urinary tract infections, burn wound infections, and infections associated with invasive medical devices such as venous catheters [14]. Mahmoud and co-authors [14] studied the antimicrobial activity of the monoterpene bicyclic derivative myrtenol in pure and mixed cultures of both organisms, as well as in combination with antibacterial, antifungal, and disinfectant compounds. The results obtained led the authors to conclude that myrtenol potentiates the antimicrobial and anti-biofilm activity of the examined compounds against mono-and dual-species cultures of S. aureus and C. albicans [14]. ...
... Mahmoud and co-authors [14] studied the antimicrobial activity of the monoterpene bicyclic derivative myrtenol in pure and mixed cultures of both organisms, as well as in combination with antibacterial, antifungal, and disinfectant compounds. The results obtained led the authors to conclude that myrtenol potentiates the antimicrobial and anti-biofilm activity of the examined compounds against mono-and dual-species cultures of S. aureus and C. albicans [14]. ...
Article
Full-text available
The emergence of new pathogens, coupled with the reemergence of old pathogens and the steep worldwide increase in multiple resistances to available antimicrobials, poses major challenges to human health at the global scale [...]
... The combination of terpenes with known antimicrobials increases the activity of the latter [30][31][32]. The introduction of sulfur functional groups into the structure of biologically active terpenes often enhances the antibacterial and antifungal activity of the resulting thio-modified monoterpenoids compared to the original terpenes [21,29,[33][34][35][36]. Pinane and menthane sulfides containing a fragment of 2-mercaptoacetic acid methyl ester showed a wide range of antifungal activity against pathogenic strains of Candida albicans and a number of mycelial fungi [21,29]. ...
... The ability of monoterpenoids to inhibit the growth of diverse bacteria and fungi has been reported [21,[24][25][26][27][28][29]. The combination of terpenes with known antimicrobials increases the activity of the la er [30][31][32]. The introduction of sulfur functional groups into the structure of biologically active terpenes often enhances the antibacterial and antifungal activity of the resulting thio-modified monoterpenoids compared to the original terpenes [21,29,[33][34][35][36]. Pinane and menthane sulfides containing a fragment of 2-mercaptoacetic acid methyl ester showed a wide range of antifungal activity against pathogenic strains of Candida albicans and a number of mycelial fungi [21,29]. ...
Article
Full-text available
Monoterpene thiols are one of the classes of natural flavors that impart the smell of citrus fruits, grape must and wine, black currants, and guava and are used as flavoring agents in the food and perfume industries. Synthetic monoterpene thiols have found an application in asymmetric synthesis as chiral auxiliaries, derivatizing agents, and ligands for metal complex catalysis and organocatalysts. Since monoterpenes and monoterpenoids are a renewable source, there are emerging trends to use monoterpene thiols as monomers for producing new types of green polymers. Monoterpene thioderivatives are also known to possess antioxidant, anticoagulant, antifungal, and antibacterial activity. The current review covers methods for the synthesis of acyclic, mono-, and bicyclic monoterpene thiols, as well as some investigations related to their usage for the preparation of the compounds with antimicrobial properties.
... The acute anti-inflammatory antinociceptive, and antifungal effects of myrtenol have been reported. Moreover, the anxiolytic and anti-proliferative effects on human cancer cells have also been shown [7][8][9]. We assumed that the replacement of one of the bulky fragments of compounds 2, 3 with a fatty acid residue would increase the activity of the compounds obtained Fragments of myristic and lauric acids were chosen as agents with established antimicrobial properties [10]. ...
... The acute anti-inflammatory, antinociceptive, and antifungal effects of myrtenol have been reported. Moreover, the anxiolytic and anti-proliferative effects on human cancer cells have also been shown [7][8][9]. We assumed that the replacement of one of the bulky fragments of compounds 2, 3 with a fatty acid residue would increase the activity of the compounds obtained. ...
Article
Full-text available
The syntheses of the title compounds were performed using lauric and myristic acids. The compounds obtained were characterized using 1H-, 13C-NMR and 2D 1H-1H COSY, 1H-13C HSQC NMR, IR, and high-resolution mass spectrometry. Both compounds exhibited bactericidal activity on S. aureus comparable to that of a reference drug (miramistin). Compound 10, with lauric acid fragment, had a 16-fold higher activity on P. aeruginosa compared to compound 11, which in turn contains myristic acid fragment (with minimum inhibitory concentrations of 32 and 512 μg/mL, respectively). Compound 11 exhibited a pronounced activity against all types of fungi (higher than the activity of miramistin), while the activity of compound 10 was considerably lower. Thus, compound 11 can serve as a promising antimicrobial agent for the treatment of various fungal and staphylococcal infections, while compound 10 is of interest to treat P. aeruginosa-associated infections.
... (+)-Myrtenol 3 is a bicyclic monoterpene alcohol found in the essential oils of various plants, whose crude extracts show anti-inflammatory, antinociceptive and antifungal activities, have a pleasant odor, and are widely used in cosmetics [15][16][17][18][19][20]. Moreover, our findings confirmed that (+)-myrtenol is capable of increasing the efficiency of various antimicrobial, antifungal, and antiseptic drugs, exhibiting a pronounced synergy with these compounds [21]. The meso-substituted BODIPYs 1, 2 ( Figure 1)preferentially stain Gram-positive bacteria and can be proposed for the differential staining of Gram-positive and Gram-negative bacteria in mixed cultures [8]. ...
... (+)-Myrtenol 3 is a bicyclic monoterpene alcohol found in the essential oils of various plants, whose crude extracts show anti-inflammatory, antinociceptive and antifungal activities, have a pleasant odor, and are widely used in cosmetics [15][16][17][18][19][20]. Moreover, our findings confirmed that (+)-myrtenol is capable of increasing the efficiency of various antimicrobial, antifungal, and antiseptic drugs, exhibiting a pronounced synergy with these compounds [21]. ...
Article
Full-text available
The synthesis of new fluorescent probes, based on biocompatible luminophors and exhibiting various specificities, is intensively developed worldwide. Many luminophors contain a hydrophobic group that limits their application for cell staining under vital conditions. Herein, we report the synthesis of two BODIPY molecules—BF2-meso-(4-butan/pentanamido-N-(((1S,5R)-6,6-dimethylbicyclo [3.1.1]hept-2-en-2-yl)methyl)-N,N-dimethylpropan-1-aminium)-3,3′,5,5′-tetramethyl-2,2′-dipyrromethene bromides—designed as 10, 11 with a spacer of either four or three CH2 groups in length, respectively. These molecules present conjugates of BODIPY luminophors with (+)-myrtenol via a quaternary ammonium group. Both terpene-BODIPY conjugates demonstrated high fluorescence efficiency in various solvents such as OctOH, DMSO and water, and were characterized by their stability at pH 1.65–9.18. The fusion of the myrtenol, a monocyclic terpene, to the BODIPY fluorophore in the meso-substituent facilitated their penetration into the filamentous fungi Fusarium solani, while impairing the binding of the latter with S. aureus, K. pneumoniae and P. aeruginosa. The additional quaternary ammonium group between the myrtenol and fluorophore moieties restored the bacterial cell-staining while it did not affect the staining of fungi. Finally, the BODIPY conjugate 11 was able to stain both Gram-positive and Gram-negative bacteria by its interaction with their cell wall (or the membrane), as well as penetrating into filamentous fungi F. solani and staining their mitochondria.
... The use of carnosic acid isolated from Rosmarinus officinalis L. extract potentiated the effect of erythromycin against multidrug-resistant bacteria [45]. Myrtenol was able to increase the efficiency of various antimicrobials against S. aureus and C. albicans [46]. ...
Article
Full-text available
Plant extracts are in the focus of the pharmaceutical industry as potential antimicrobials for oral care due to their high antimicrobial activity coupled with low production costs and safety for eukaryotic cells. Here, we show that the extract from Hop (Humulus lupulus L.) exhibits antimicrobial activity against Staphylococcus aureus and Streptococci in both planktonic and biofilm-embedded forms. An extract was prepared by acetone extraction from hop infructescences, followed by purification and solubilization of the remaining fraction in ethanol. The effect of the extract on S. aureus (MSSA and MRSA) was comparable with the reference antibiotics (amikacin, ciprofloxacin, and ceftriaxone) and did not depend on the bacterial resistance to methicillin. The extract also demonstrated synergy with amikacin on six S. aureus clinical isolates, on four of six isolates with ciprofloxacin, and on three of six isolates with ceftriaxone. On various Streptococci, while demonstrating lower antimicrobial activity, an extract exhibited a considerable synergistic effect in combination with two of three of these antibiotics, decreasing their MIC up to 512-fold. Moreover, the extract was able to penetrate S. aureus and S. mutans biofilms, leading to almost complete bacterial death within them. The thin-layer chromatography and LC-MS of the extract revealed the presence of prenylated flavonoids (2′,4′,6′,4-tetrahydroxy-3′-geranylchalcone) and acylphloroglucides (cohumulone, colupulone, humulone, and lupulone), apparently responsible for the observed antimicrobial activity and ability to increase the efficiency of antibiotics. Taken together, these data suggest an extract from H. lupulus as a promising antimicrobial agent for use both as a solely antiseptic and to potentiate conventional antimicrobials.
Article
Staphylococcus aureus causes many diseases under pathological conditions. Due to the rapid development of antibiotic resistance, the search for alternative approaches to antimicrobial treatment of Staphylococci is challenging. Natural and synthetic terpenoids, due to their membranotropic properties, both exhibit antimicrobial properties and potentiate other antibiotics. The antimicrobial activity of 53 natural terpenes and their synthetic oxygen-containing derivatives, as well as their ability to potentiate the effect of conventional antimicrobials against S. aureus, has been assessed. It has been shown that (+)-3β,4β-carandiol and (−)-myrtenic acid increase the efficiency of amikacin, ceftriaxone, and miramistin up to 4-fold, thus suggesting them as promising agents for combination therapy with antimicrobials to reduce active concentrations of the latter.
Article
A series of new compounds containing monoterpenoid fragments, sulfide and carboxy groups was synthesized. The reaction of monoterpene thiols with bromoacetic acid and ethyl 2-bromo-2,2-difluoroacetate afforded new thiomonoterpene carboxylic acids in 80–96% and 56–80% yields, respectively. The synthesized compounds were studied for antimicrobial activity. Some structure–property interrelations were revealed, which depended on the monoterpene fragment structure and the presence of fluorine atoms in the carboxymethyl group.
Article
Full-text available
After the appearance of the green chemistry concept, which was introduced in the chemistry vocabulary in the early 1990s, its main statements have been continuously developed and modified. Currently, there are 10–12 cornerstones that should form the basis for an ideal chemical process. This review analyzes the accumulated experience and achievements towards the design of chemical products and processes that reduce or eliminate the use or generation of hazardous substances. The review presents the views of leading Russian scientists specializing in various fields of this subject, including homogeneous and heterogeneous catalysis, fine and basic organic synthesis, electrochemistry, polymer chemistry, chemistry based on bio-renewable feedstocks and chemistry of energetic compounds and materials. A new approach to the quantitative evaluation of the environmental friendliness of processes developed by Russian authors is described. The bibliography includes 1761.
Article
Colon cancer is a serious health problem across the globe with various dietary lifestyle modifications. It arises as an inflammation mediated crypts in the colon epithelial cells and undergoes uncontrolled cell division and proliferation. Bacterial enzymes contribute to a major outbreak in colon cancer development upon the release of toxic metabolites from the gut microflora. Pathogen associated molecular patterns and damage associated molecular patterns triggers the NLPR3 inflammasome pathways that releases pro-inflammatory cytokines to induce cancer of the colon. Contributing to this, specific chemokines and receptor complexes attribute to cellular proliferation and metastasis. Bacterial enzymes synergistically attack the colon mucosa and degenerate the cellular integrity causing lysosomal discharge. These factors further instigate the Tol like receptors (TLRs) and Nod like receptors (NLRs) to promote angiogenesis and supply nutrients for the cancer cells. Myrtenal, a monoterpene, is gaining more importance in recent times and it is being widely utilized against many diseases such as cancers, neurodegenerative diseases and diabetes. Based on the research data's, the reviews focus on the anticancer property of myrtenal by emphasizing its therapeutic properties which downregulate the inflammasome pathways and other signalling pathways. Combination therapy is gaining more importance as they can target every variant in the cellular stress condition. Clinical studies with compounds like myrtenal of the monoterpenes family is provided with positive results which might open an effective anticancer drug therapy. This review highlights myrtenal and its biological potency as a cost effective drug for prevention and treatment of colon cancer.
Article
Full-text available
The aim of this study was to examine the effects of (-)-myrtenol alone and combined with antifungal agents against Candida spp. The Minimum Inhibitory Concentration (MIC) and Minimum Fungicidal Concentration of (-)-myrtenol and fluconazole against C. albicans and C. parapsilosis strains was obtained using CLSI guidelines. Combination of (-)-myrtenol with antifungal drugs was determined by checkboard test. The (-) myrtenol showed MIC ranging from 256 to 512 µg/mL against both species assay. And the MFC was 512 µg/mL, demonstrated nature fungicidal (MFC/MIC < 4). In addition, combination of antifungal agents (amphotericin B and fluconazole) and (-) myrtenol showed synergistic and additive effects on strains assays. Based on these results, the present study demonstrates that (-) myrtenol showed strong fungicide activity against Candida spp. In addition, Combination of antifungal agents and (-) myrtenol reduces the effective concentrations of both the agents with synergistic to additive effects. Therefore, (-) myrtenol has potential to be developed into an antifungal agent.
Article
Full-text available
The increased incidence of mixed infections requires that the scientific community develop novel antimicrobial molecules. Essential oils and their bioactive pure compounds have been found to exhibit a wide range of remarkable biological activities and are attracting more and more attention. Therefore, the aim of this study was to evaluate myrtenol (MYR), one of the constituents commonly found in some essential oils, for its potential to inhibit biofilms alone and in combination with antimicrobial drugs against Candida auris/Klebsiella pneumoniae single and mixed biofilms. The antimicrobial activity of MYR was evaluated by determining bactericidal/fungicidal concentrations (MIC), and biofilm formation at sub-MICs was analyzed in a 96-well microtiter plate by crystal violet, XTT reduction assay, and CFU counts. The synergistic interaction between MYR and antimicrobial drugs was evaluated by the checkerboard method. The study found that MYR exhibited antimicrobial activity at high concentrations while showing efficient antibiofilm activity against single and dual biofilms. To understand the underlying mechanism by which MYR promotes single/mixed-species biofilm inhibition, we observed a significant downregulation in the expression of mrkA, FKS1, ERG11, and ALS5 genes, which are associated with bacterial motility, adhesion, and biofilm formation as well as increased ROS production, which can play an important role in the inhibition of biofilm formation. In addition, the checkerboard microdilution assay showed that MYR was strongly synergistic with both caspofungin (CAS) and meropenem (MEM) in inhibiting the growth of Candida auris/Klebsiella pneumoniae-mixed biofilms. Furthermore, the tested concentrations showed an absence of toxicity for both mammalian cells in the in vitro and in vivo Galleria mellonella models. Thus, MYR could be considered as a potential agent for the management of polymicrobial biofilms.
Article
Full-text available
New unsymmetrical monoterpenylhetaryl disulfides based on heterocyclic disulfides and monoterpene thiols were synthesized for the first time in 48–88% yields. Hydrolysis of disulfides with fragments of methyl esters of 2-mercaptonicotinic acid was carried out in 73–95% yields. The obtained compounds were evaluated for antioxidant, antibacterial, antifungal activity, cytotoxicity and mutagenicity.
Article
Full-text available
We aimed to identify the prevalence and emerging status of multidrug-resistant bacteria and fungi and their associated mortality in nine countries in the Arabian Peninsula. Original research articles and case studies regarding multidrug-resistant bacteria and fungi in the Arabian Peninsula, published during the last 10 years, were retrieved from PubMed and Scopus. A total of 382 studies were included as per the inclusion and exclusion criteria, as well as the PRISMA guidelines , from a thorough screening of 1705 articles, in order to analyse the emerging status and mortality. The emerging nature of >120 multidrug-resistant (MDR) bacteria and fungi in the Arabian Peninsula is a serious concern that requires continuous monitoring and immediate preventive measures. More than 50% (n = 453) of multidrug-resistant, microbe-associated mortality (n = 871) in the Arabian Peninsula was due to MDR Acinetobacter baumannii, Mycobacterium tuberculosis and Staphylococcus aureus infection. Overall, a 16.51% mortality was reported among MDR-infected patients in the Arabian Peninsula from the 382 articles of this registered systematic review. MDR A. baumannii (5600 isolates) prevailed in all the nine countries of the Arabian Peninsula and was one of the fastest emerging MDR bacteria with the highest mortality (n = 210). A total of 13087 Mycobac-terium tuberculosis isolates were reported in the region. Candida auris (580 strains) is the most prevalent among the MDR fungal pathogen in the Arabian Peninsula, having caused 54 mortalities. Active surveillance, constant monitoring, the development of a candidate vaccine, an early diagnosis of MDR infection, the elimination of multidrug resistance modulators and uninterrupted preventive measures with enhanced data sharing are mandatory to control MDR infection and associated diseases of the Arabian Peninsula. Accurate and rapid detection methods are needed to differentiate MDR strain from other strains of the species. This review summarises the logical relation, prevalence , emerging status and associated mortality of MDR microbes in the Arabian Peninsula.
Article
Full-text available
The present paper aims to study the biological properties of a series of bicyclic monoterpene alcohols. Firstly, we tested the obtained compounds for fungicidal activity against clinical and reference strains of microscopic fungi. Next, we determined the minimum inhibitory concentration of these compounds comparing to other drugs widely used in practical medicine (fluconazole, terbinafine). At this stage, we found that ( −)-myrtenol (47 MIC and 23.5 μg/ml) exhibits the most promising activity against filamentous and yeast fungi, respectively. Then, we have studied the membrane-protective and antioxidant activities of the obtained compounds and found out that ( −)-cis-verbenol and ( −)-myrtenol exhibit the highest activity on the model of erythrocytes oxidative hemolysis. Interestingly, among all the studied bicyclic monoterpene alcohols, the alcohols of the pinane series have been found to be the most promising. The obtained results from the present study suggest that ( −)-myrtenol would be a leading compound for further studies in terms of possible practical application.
Article
Full-text available
Context Cerebral ischaemia/reperfusion (I/R) injury has a high disability and fatality worldwide. Myrtenol has protective effects on myocardial I/R injury through antioxidant and anti-apoptotic effects. Objective This study investigated the effect of myrtenol on cerebral ischaemia/reperfusion (I/R) injury and the underlying mechanism. Materials and methods Cerebral I/R injury was induced in adult Sprague-Dawley rats by middle cerebral artery occlusion (MCAO) for 90 min. MCAO rats were treated with or without myrtenol (10, 30, or 50 mg/kg/day) or/and U0126 (10 μL) intraperitoneally for 7 days. Results In the present study, myrtenol had no toxicity at concentrations up to 1.3 g/kg. Myrtenol treatment improved neurological function of MCAO rats, with significantly (p < 0.05) improved neurological deficits (4.31 ± 1.29 vs. 0.00) and reduced brain edoema (78.95 ± 2.27% vs. 85.48 ± 1.24%). Myrtenol extenuated brain tissue injury and neuronal apoptosis, with increased Bcl-2 expression (0.48-fold) and decreased Bax expression (2.02-fold) and caspase-3 activity (1.36-fold). Myrtenol promoted angiogenesis in the brain tissues of MCAO rats, which was reflected by increased VEGF (0.86-fold) and FGF2 (0.51-fold). Myrtenol promoted the phosphorylation of MEK1/2 (0.80-fold) and ERK1/2 (0.97-fold) in MCAO rats. U0126, the inhibitor of ERK1/2 pathway, reversed the protective effects of myrtenol on brain tissue damage and angiogenesis in MCAO rats. Discussion and conclusions Myrtenol reduced brain damage and angiogenesis through activating the ERK1/2 signalling pathway, which may provide a novel alternative strategy for preventing cerebral I/R injury. Further in vitro work detailing its mechanism-of-action for improving ischaemic cerebral infarction is needed.
Article
Polymicrobial biofilm formation during multi-species infection is a serious threat growing worldwide. According to CDC, the microbial biofilm infection covers more than 65% of total infection. In many diseases, their natural habitat does not have one causative agent because most of the species exist in co-aggregation (such as CF, OM, Dental Caries) leading to polymicrobial biofilm. Polymicrobial biofilm is a big problem in bacterio-fungal and inter-species bacterial diseases developed during chronic illness and created a major health burden globally. This review focused on various aspects of polymicrobial biofilms such as why they are forming polymicrobial biofilm arrangements, the significance of studying these biofilms, the interaction between causative microbes. Also, we reviewed how these interactions and polymicrobial formations make the biofilm more recalcitrant towards the treatment. Understanding the mechanistic process behind these biofilm formations gives an insight into specific molecules, proteins responsible for their polymicrobial nature are likely to be very helpful in anti-microbial researches.
Article
Polymicrobial biofilm formation during multi-species infection is a serious threat growing worldwide. According to CDC, the microbial biofilm infection covers more than 65% of total infection. In many diseases, their natural habitat does not have one causative agent because most of the species exist in co-aggregation (such as CF, OM, Dental Caries) leading to polymicrobial biofilm. Polymicrobial biofilm is a big problem in bacterio-fungal and inter-species bacterial diseases developed during chronic illness and created a major health burden globally. This review focused on various aspects of polymicrobial biofilms such as why they are forming polymicrobial biofilm arrangements, the significance of studying these biofilms, the interaction between causative microbes. Also, we reviewed how these interactions and polymicrobial formations make the biofilm more recalcitrant towards the treatment. Understanding the mechanistic process behind these biofilm formations gives an insight into specific molecules, proteins responsible for their polymicrobial nature are likely to be very helpful in anti-microbial researches.
Article
Background Wounds remain the most important cause of postoperative mortality and morbidity and generate considerable additional social and healthcare costs. Most wounds are caused by various coliforms, Enterococcus fecalis, Proteus sp., and multidrug resistant Staphylococcus aureus. Wound is one of the leading cause of infections in the under developed and developing countries than developed nations. Methods A total of 43 samples associated with bacteremia and wound infection were collected. Biochemical characterization and culture characteristics of the drug resistant isolates were studied using MacConkey agar, blood agar and mannitol-salt agar. Antibiotic susceptibility analysis of the isolated strains was performed by disc diffusion method using various antibiotics. Prevalence of dug resistance among bacteria isolated from the wound was studied. The ability of Beta lactamase antibiotic producing bacterial strains were analyzed. Results A total of 168 bacterial strains were isolated showed high resistant towards ampicillin (89%), ciprofloxacin (90.8), cefepine (90.5), piperacillin (91.8), oxacillin (92.5), and imipenem (96.5). The isolated bacterial strains showed monobacterial as well as polybacterial growth on the surface of the wound. The isolated bacterial strains revealed 89% sensitivity against norfloxacin and 94.9 sensitivity against vancomycin. About 26% of bacterial strains degraded quinolones, whereas only 14% clinical isolates showed their ability to degrade aminoglycosides. A total of 27% bacteria degraded tetracycline and 51% of isolates degraded carbapenems compounds. Interestingly, E. faecalis was resistant against antibiotics such as, Oxacillin, Nalidic acid, Ofloxacin, Erythromycin, Norfloxacin, Ciprofloxacin, Ampicillin, Tetracycline, Cefepine, Amikacin, Cefurooxime, Vancomycin, Piperacillin, Imipenem and Gentamycin. Moreover, Proteus species was resistant against certain numbers of antibiotics namely, Ampicillin, Piperacillin, Oxacillin, Nalidic acid, Tetracycline, Erythromycin, Cefurooxime, Nitrofurantoin, Vancomycin and Imipenem Conclusions The isolated bacterial strains were resistant against various drugs including vancomycin. Staphylococci, and E. faecalisis strains showed resistance against various classes of antibiotics.