You are on page 1of 11

RSC

Medicinal Chemistry
View Article Online
RESEARCH ARTICLE View Journal | View Issue

Sulfonium-based liposome-encapsulated
Published on 24 May 2021. Downloaded by Indian Institute of Science on 7/30/2023 7:45:51 AM.

Cite this: RSC Med. Chem., 2021, 12,


antibiotics deliver a synergistic antibacterial
1005 activity†
Anjali Patel,a Subhasis Dey,b Kamal Shokeen,c Tomasz M. Karpiński, d

Senthilkumar Sivaprakasam,c Sachin Kumar c and Debasis Manna *ab

The devastating antibacterial infections, coupled with their antibiotic resistance abilities, emphasize the
need for effective antibacterial therapeutics. In this prospect, liposomal delivery systems have been
employed in improving the efficacy of the antibacterial agents. The liposome-based antibiotics enhance
the therapeutic potential of the new or existing antibiotics and reduce their adverse effects. The current
study describes the development of sulfonium-based antibacterial lipids that demonstrate the delivery of
existing antibiotics. The presence of cationic sulfonium moieties and inherent membrane targeting abilities
of the lipids could help reduce the antibiotic resistance abilities of the bacteria and deliver the antibiotics to
remove the infectious pathogens electively. The transmission electron microscopic images and dynamic
light scattering analyses revealed the liposome formation abilities of the sulfonium-based amphiphilic
compounds in the aqueous medium. The effectiveness of the compounds was tested against the Gram-
negative and Gram-positive bacterial strains. The viability of the bacterial cells was remarkably reduced in
the presence of the compounds. The sulfonium-based compounds with pyridinium moiety and long
hydrocarbon chains showed the most potent antibacterial activities among the tested compounds.
Mechanistic studies revealed the membrane-targeted bactericidal activities of the compounds. The potent
compound also showed tetracycline and amoxicillin encapsulation and sustained release profiles in the
physiologically relevant medium. The tetracycline and amoxicillin-encapsulated lipid showed much higher
antibacterial activities than the free antibiotics at similar concentrations, emphasizing the usefulness of the
synergistic effect of sulfonium-based lipid and the antibiotics, signifying that the sulfonium lipid penetrated
Received 15th March 2021, the bacterial membrane and increased the cellular uptake of the antibiotics. The potent lipid also showed
Accepted 1st May 2021
therapeutic potential, as it is less toxic to mammalian cells (like HeLa and HaCaT cells) at concentrations
higher than their minimum inhibitory concentration values against S. aureus, E. coli, and MRSA. Hence, the
DOI: 10.1039/d1md00091h
sulfonium-based lipid exemplifies a promising framework for assimilating various warheads, and provides a
rsc.li/medchem potent antibacterial material.

Introduction outcomes due to bacterial infection and antibacterial drug


resistance.1–4 Over the years, bacteria such as Escherichia coli
Antimicrobial drug resistance has intimidated the success of (E. coli), Klebsiella pneumoniae, and Staphylococcus aureus (S.
preventing and treating infectious diseases, and other aureus) have developed multidrug resistance, which is
ailments such as cancer.1,2 Various medical procedures, such challenging to treat with common antibacterial drugs. The
as major surgery and organ implant, are also facing uncertain healthcare encumbrance related to the drug-resistant bacteria
is further impaired due to the ability of several bacterial
a
strains to become resistant towards the frontier antibiotics,
Indian Institute of Technology Guwahati, Centre for the Environment, Guwahati,
Assam, India. E-mail: dmanna@iitg.ac.in
such as vancomycin. Overexposure to drugs allows the
b
Biological Chemistry Laboratory, Indian Institute of Technology Guwahati, bacteria to develop drug resistance capability by modifying
Chemistry, Guwahati, Assam, India their genetic material. A plethora of bacteria also produce a
c
Indian Institute of Technology Guwahati, Biosciences and Bioengineering, sturdy biofilm to cover their colony and protect themselves
Guwahati, Assam, India
d
from unpropitious conditions.5,6 Thus, new or modified
Department of Medical Microbiology, Poznań University of Medical Sciences,
Wieniawskiego, Poznań, Poland
antibacterial drugs are highly demanding. Unexpectedly, the
† Electronic supplementary information (ESI) available. See DOI: 10.1039/ span is too large between the rate of development of new
d1md00091h medicines and the evolution of another drug-resistant

This journal is © The Royal Society of Chemistry 2021 RSC Med. Chem., 2021, 12, 1005–1015 | 1005
View Article Online

Research Article RSC Medicinal Chemistry

bacterial strain. Hence, exploring both new antibacterial quaternary ammonium and phosphonium-based compounds
agents and their delivery systems continues to be an and other cationic antimicrobial agents.15,16,18 Sulfonium-
imperative research emphasis, as it is crucial to fight against based compounds are naturally abundant in plants and
the emerging apprehension of drug-resistant pathogenic animals. Methyl sulfonium-containing compounds are
bacteria. A combinatorial strategy could also help to fight commonly used as a methyl transfer agent.19 However, the
against bacteria, as the synergistic effect is likely to antibacterial activities of these sulfonium-based compounds
demonstrate the superior bactericidal effect, reduce the host- are rarely studied, and practically used in comparison to that
specific toxicity, and operational killing. The therapeutic of ammonium and phosphonium-based compounds.
efficiency of this antibacterial strategy depends on the ability Currently, few sulfonium-based compounds, such as
Published on 24 May 2021. Downloaded by Indian Institute of Science on 7/30/2023 7:45:51 AM.

of the carrier to infringe the intrinsic resilience of the adenosylmethionine, bleomycin, and modified-echinomycin,
bacterial cells, and in re-establishing susceptibility to the are used in therapeutics. Recent studies revealed that the
antibacterial agent.7 sulfonium-linked vancomycin analog had enhanced
Antibacterial drugs primarily act on the bacterial cell wall, antibacterial activity against vancomycin-resistant bacteria
plasma membrane, replication machinery, metabolic activity, both under in vitro and in vivo conditions. The cationic
and protein translation machinery. Drugs with membrane- sulfonium moiety improved the interaction of vancomycin
targeted activity have an important role in the antibacterial with the cell membrane and disturbed the integrity of the
activity, and comparatively, they can better counteract the membrane of both Gram-negative and Gram-positive
bacterial resistance system. The membrane-targeted bacteria.18 In our previous study, we demonstrated that the
antibacterial drugs provide the extra benefit without sulfonium lipids encapsulate and deliver the anticancer drug,
developing resistance as they can act effectively towards the doxorubicin, to a mammalian cell.20 The self-assembly
dormant or spore form of the bacteria and biofilm-forming properties of those sulfonium lipids could be utilized to
strain because of the conserved membrane composition in deliver both aqueous soluble and insoluble antibiotics at the
all growth forms of the bacterial cell and distinct mechanism target site. The amphiphilicity and the presence of cationic
of action that implicates less specific membrane sulfonium moieties would also allow the sulfonium-based
permeabilization.8–10 Membrane targeting antibacterial lipids to fuse with the outer membrane of the bacterial cells
agents generally leads to bactericidal activity. Various reports and release the encapsulated antibiotics.21 Recently,
have already demonstrated that bactericidal drugs are more polymeric amphiphilic systems with inherent antibacterial
preferred over bacteriostatic drugs to treat meningitis, activity were reported.22 However, a small molecule-based
endocarditis, and several other bacteria-related diseases.11,12 biocompatible amphiphilic system can augment the
Membrane targeting antibacterial drugs target the bacterial therapeutic benefits.
cell membrane over mammalian cells because of the Herein, we report the synthesis and mechanism of the
differences in the lipid component and composition of their antibacterial activity of sulfonium-based compounds. The
architecture.13 role of the cationic charge and hydrocarbon chain length in
In this regard, small molecule-based membrane-active antimicrobial activity was investigated against both Gram-
synthetic amphiphilic compounds are considered as potential positive and Gram-negative strains of bacteria. The
antibacterial agents that hold the prospect of success for amphiphilic molecule with the pyridinium headgroup
combinatorial antimicrobial therapy. Amphiphilic showed the most potent antibacterial activity among the
compounds with a cationic moiety convey membrane- tested compounds against Gram-negative, Gram-positive, and
oriented activity because of the negatively charged even drug-resistant bacteria. The less-toxic sulfonium
lipopolysaccharide (LPS) and teichoic acid (TA) in Gram- compound could also prevent the formation of a biofilm. In
negative and Gram-positive bacteria, respectively.14–17 The addition, encapsulation of the commercial antibiotics and
hydrophobicity of an antimicrobial compound can easily antibacterial efficacies of the composite was investigated.
coordinate the bacterial membrane and is the panacea of The composite showed moderate loading of water-soluble
water-soluble antibiotic resistance because of the reduced antibiotics and a sustained release profile. Overall, our
porins in the bacterial outer membrane. Furthermore, studies propose that the electrostatic interaction of cationic
hydrophobic antimicrobial ointments are becoming the liposomes with the bacterial membrane allows its
choice of wound dressings to combat the severe skin disassembly and insertion of the amphiphilic compound to
infection caused by superbugs, like methicillin-resistant S. the bacterial membrane, resulting in the concomitant release
aureus (MRSA). However, the therapeutic prospective of of antibiotics at the target site to achieve synergistic
synthetic amphiphilic compounds depends on their host- antibacterial activities.
specific toxicity, suggesting the requirement of the judicious
design of antibacterial amphiphilic compounds so that both Results and discussion
carrier and antibiotics are accessible to the bacterial cells to
achieve the synergistic effect. Different research groups have Design and synthesis of compounds
demonstrated that the cationic sulfonium-based compounds The anticancer drug delivery and moderate antimicrobial
are potent antimicrobial agents, and less toxic than the activities of the sulfonium lipids motivated us to synthesize a

1006 | RSC Med. Chem., 2021, 12, 1005–1015 This journal is © The Royal Society of Chemistry 2021
View Article Online

RSC Medicinal Chemistry Research Article

new series of sulfonium-based compounds.20 The key features


in designing the compounds were the installation of a cationic
(pyridinium) or anionic (sulfonic acid) headgroup and variation
of the alkyl chain length, in addition to the sulfonium moieties
(Scheme 1). The variation in the alkyl chain length allowed us
to investigate the role of hydrophobicity and antimicrobial
activity. We envisage that the hydrophobicity of the dialkyl
chain lengths could enable the compounds to self-aggregate in
an aqueous medium, which could be useful in encapsulating
Published on 24 May 2021. Downloaded by Indian Institute of Science on 7/30/2023 7:45:51 AM.

the commercial antibiotics. The presence of both sulfonium


moieties and antibiotic encapsulation ability would generate a
composite antibacterial agent, which can increase the
bactericidal effect, reduce the antibiotic-related toxicity, and
enhance the efficacy of the antibiotics at lower doses (Fig. 1A).
Most of the conventional liposomes, micelles, or nanoparticle- Fig. 1 Cartoon diagram demonstrating the probable pathway for the
based delivery systems lack such a dual mode of action, bactericidal effect of the antibiotics-encapsulated liposomes of
suggesting that the combined antibacterial activity of these compound 7a (A). Representative TEM image of the soluble aggregates
sulfonium-based compounds can improve the killings of drug- generated from the 100% compound 7a (B). The scale bar for the TEM
image is 200 nm. Variation of the hydrodynamic diameter of the
resistant pathogenic bacteria.
soluble aggregates generated from the 100% compound 7a at different
The compounds were synthesized according to our pH values was measured by DLS analysis (C). The surface potential of
reported methods with minor modifications (Scheme 1).20 the soluble aggregates generated from the 100% compound 7a at
The compounds for the current study were synthesized using different pH values (D).
1,3,5-tris(bromomethyl)benzene. The mono-modification of
1,3,5-tris(bromomethyl)benzene with azide and pyridine
resulted in compounds 2 and 3, which were further modified a suspended aqueous solution. The presence of a short alkyl
with aliphatic thiols to provide compounds 4 and 5, chain length and sulfonic acid could be reasons for their
respectively. The azide–alkyne click reaction of compound 4 aqueous solubility. The field-emission transmission electron
with prop-2-yne-1-sulfonic acid produced compound 6. microscope (FE-TEM) analysis showed that compound 7a
Finally, the treatment of compounds 5 and 6 with methyl forms spherical aggregates in an aqueous medium (Fig. 1B).
iodide in the presence of AgBF4 provided the desired The dynamic light scattering (DLS) study showed that the size
products 7 and 8 with satisfactory yield. These compounds of the spherical aggregates varies between 270–340 nm at
were characterized by nuclear magnetic resonance (NMR) different pH values of the respective buffers at 25 °C
and high-resolution mass spectrometry (HRMS). (Fig. 1C and D, and S6†). The zeta-potential measurements
showed that the overall surface charge of the spherical
aggregates was positive, and the positive charge increases
Aggregation behavior in aqueous solution with the decrease in pH of the buffer, which could be due to
To investigate the behavior of these amphiphilic compounds the reduction in the solvation number in the acidic medium.
under an aqueous environment, the compounds were Hence, TEM, DLS, and zeta-potential measurements revealed
dissolved in phosphate-buffered saline (PBS).20 Compound that compound 7a formed stable spherical aggregates with
7b was completely soluble, but compounds 7a and 8 formed positive surface potential.

Antibacterial activities of the compounds


The sulfonium-based compounds showed antibacterial
activity against both Gram-positive and Gram-negative
bacteria.18,20 The minimum inhibitory concentrations (MIC)
of the compounds were calculated against Gram-negative
bacteria, such as E. coli MTCC 1687 and P. aeruginosa MTCC
2488, and Gram-positive bacteria, such as S. aureus MTCC 96
and methicillin-resistant S. aureus (MRSA), by micro broth
dilution method.23 The highly infectious E. coli strain resides
in the intestine and causes various diseases, including
pneumonia, diarrhea, and urinary tract infection (UTI). The
bacterial strains of S. aureus and Pseudomonas aeruginosa (P.
aeruginosa) are the frequent residents of bronchioles and
Scheme 1 Synthetic routes to the sulfonium-based compounds. alveoli of cystic fibrosis patients. The MIC values were

This journal is © The Royal Society of Chemistry 2021 RSC Med. Chem., 2021, 12, 1005–1015 | 1007
View Article Online

Research Article RSC Medicinal Chemistry

Table 1 Antibacterial activities of the synthesized compounds

Compound MIC (μM)/MBC (μM) HC50


n R E. coli P. aeruginosa S. aureus S. aureus (MRSA) (μM)
4 11 –N3 >100 >100 >100 >100 —
5a 11 >100 >100 >100 >100 —

5b 2 >100 >100 >100 >100 —

6 11 >100 >100 >100 >100 —


Published on 24 May 2021. Downloaded by Indian Institute of Science on 7/30/2023 7:45:51 AM.

7a 11 7.5 ± 1/10 ± 1 12 ± 0.5/24 ± 1 6.3 ± 1/12.5 ± 1 6.3 ± 1/12.5 ± 1.5 29

7b 2 >100 >100 >100 >100 —

8 11 12 ± 1/20 ± 1 25 ± 1/50 ± 1 10 ± 0.5/12.5 ± 1 25 ± 1/>100 35

Tetracycline 8.5 ± 1 14 ± 0.5 4.2 ± 0.5 >100


Amoxicillin 11 ± 1 4.3 ± 0.5 6.8 ± 1 >50

reckoned as the minimum concentration of the compounds, anionic lipids of the bacterial membrane compared to that of
which caused no visible growth, or the optical densities (OD) the sulfonic acid moiety. The cationic moieties showed better
of the compound-treated bacterial cultures were close to that antibacterial activity over neutral or anionic moieties.14–16
of the control (without any bacteria). Compounds 7a and 8 Similar antibacterial activities were reported when the
showed very good antibacterial activity against all four tested sulfonium moiety was appended to vancomycin.18 The
bacterial strains (Table 1). The MIC values of the pyridinium- sulfonium-containing vancomycin analogue showed
containing compound 7a were within 6.3–12 μM, whereas the improved antibacterial activity against vancomycin-resistant
sulfonic acid-containing compound 8 had slightly higher bacteria, suggesting the potential of the sulfonium moiety in
MIC values of 12–24 μM. It was encouraging to observe that antibacterial activities. The attachment of the sulfonium
the drug-resistant strain of S. aureus (MRSA) showed a similar moiety also alters the innate feature of vancomycin, leading
MIC value to that of its drug-sensitive strain. Further to its activity against Gram-negative bacteria.18 Interestingly,
antibacterial studies of compounds 7a and 8 were performed the antibacterial activity of these sulfonium lipids 7a and 8
to investigate the impetus of different structural moieties in are similar to other substances, especially to some widely
the compound, which provide the observed antimicrobial used antiseptics. The MIC values of those antiseptics against
property. However, compound 4 with azide but no sulfonium P. aeruginosa are the following: for octenidine
moieties showed no antibacterial activity, even at 100 μM.20 dihydrochloride, 3.9–31.3 μg mL−1 (6.25–50.1 μM); for
Compound 5a, with only pyridinium but no sulfonium chlorhexidine digluconate, 15.6–64.0 μg mL−1 (30.9–126.6
moieties, showed no antibacterial activity even at 100 μM, μM); and for polyhexamethylene biguanide, 2–31.3 μg
suggesting the role of the sulfonium moieties in the mL−1.24,25 Simultaneously, the activity of sulfonium
antibacterial activities of compound 7a. Compound 7b, with compounds 7a and 8 against P. aeruginosa is significantly
the same headgroup but with a short alkyl chain (ethyl higher than other very important antiseptics, like
group) length compared to that of compound 7a, showed no benzalkonium chloride (MICs, 32–512 μg ml−1), povidone-
antibacterial activity even at 100 μM, suggesting that the long iodine (MICs, 62.5–1024 μg ml−1), and triclosan (MICs > 512
hydrocarbon chain is as important as other moieties in the μg ml−1).24,25 Several sulfonium-based compounds showed
compound. Compound 5b with a short alkyl chain length better antimicrobial activities than some common cationic
also showed no antibacterial activity. The sulfonium and antimicrobials, including cetylpyridinium chloride and
pyridinium or sulfonic acid moieties could probably be benzalkonium chloride.26 The recently developed sulfonium
involved in electrostatic or hydrogen bond interactions with N-chloramines showed higher antibacterial activities than the
the phosphoryl and carboxyl groups of the bacterial formerly reported quaternary ammonium counterpart.27 The
membrane lipids, and elicit the insertion of the long polymeric salts of p-vinylbenzyl tetramethylenesulfonium
hydrocarbon chain lengths of compounds 7a and 8 to the tetrafluoroborate exhibited high antibacterial activity against
bacterial membrane.18 The higher antibacterial activity of Gram-positive bacteria.28
compound 7a over 8 could be due to much stronger The bactericidal activities of the compounds were tested
electrostatic interaction of the pyridinium moiety with the against the same bacterial strains.29 The minimum

1008 | RSC Med. Chem., 2021, 12, 1005–1015 This journal is © The Royal Society of Chemistry 2021
View Article Online

RSC Medicinal Chemistry Research Article

bactericidal concentrations (MBC) of the compound were


higher than the MIC values (bacteriostatic concentration),30
suggesting that a higher concentration of the compound is
required to demolish the bacterial membrane or metabolic
activity at a rate with which they might not be able to repair
it and die off (Table 1). Thus, these amphiphiles are
proficient in killing the bacterial cell at a low concentration,
making them good antimicrobial agents. The bactericidal
activity of the compounds was further confirmed by FESEM
Published on 24 May 2021. Downloaded by Indian Institute of Science on 7/30/2023 7:45:51 AM.

analysis (Fig. 2).20 A significant difference in morphology of


the bacterial cells was observed between the control and
compound-treated samples. The cocci shape of the S. aureus
and the rod shape of E. coli were disfigured or fragmented in
the drug-treated bacterial sample, which could be due to the
relentless disruption of the bacterial membrane. These
morphological changes of the bacterial cells suggest that
compound 7a may induce bacterial cell death.17,20
Additionally, the antibiofilm activity of the compound was
tested. The extra polymeric substance or biofilm is essentially
composed of extracellular DNA (eDNA), peptide or protein,
and intercellular polysaccharide with different ratios and
architecture, depending upon the bacterial strain. The
biofilm has been implicated in many infectious diseases,
including chronic sinusitis, atherosclerosis, chronic wounds,
endocarditis, and many more.31 The antibiofilm assay by the
crystal violet staining method showed that compound 7a
inhibits the formation of a biofilm by the S. aureus strain in
a dose-dependent manner (Fig. 3A).32 However, the Fig. 3 Compound 7a-mediated eradication of the S. aureus (MRSA)
biofilm was investigated by crystal violet staining assay (A and B). The
concentration of 7a required (24 μM) to abolish mature
diagram demonstrates the probable interaction pattern of compound
biofilm was 2–4-fold higher than the concentrations 7a with the negatively charged TA and LPS in the Gram-positive and
necessary to inhibit the formation of 90% biofilm (12 μM) Gram-negative bacteria, respectively (C).
(Fig. 3B and C). Thus, these amphiphiles have the capability
to degrade the mature biofilm. The presence of both
pyridinium and sulfonium (cationic) moieties could assist intercalating agent, PI can cross the damaged cell membrane,
the interaction of compound 7a with the TA and LPS of the but is impermeable to the intact and live cell. The PI uptake
Gram-positive and Gram-negative bacterial cells. This assay was performed at different concentrations and time
electrostatic interaction could drive the membrane-mediated intervals using S. aureus cells. The increase in the
antibacterial activities of compound 7a (Fig. 3C). The fluorescence intensity of PI revealed that the antibacterial
mechanism of the bactericidal activity was studied by activity of compound 7a is membrane-directed and time-
propidium iodide (PI) uptake assay.33,34 The fluorescent DNA dependent, suggesting that the number of dead cells
increases with time (Fig. 4A). Further mechanistic studies
were performed by membrane depolarization assay using the
DiSC3(5) dye.34 The fluorescence intensity of this dye gets
quenched on the negatively charged bacterial membrane.
However, it showed higher fluorescence intensity on the
disrupted membrane. The DiSC3(5) assay was tested on S.
aureus at various concentrations of compound 7a and
positive control (valinomycin). A significant disparity was
observed in the fluorescence intensity even at a very low
concentration range, increasing with the increase in
concentration. The increase in the fluorescence intensity of
the DiSC3(5) dye of the compound 7a-treated cells suggests
the disruption in the membrane integrity of S. aureus cells
Fig. 2 Representative FESEM images of untreated (A and C) and
(Fig. 4B). A fluorescence-based live and dead cell imaging
compound 7a-treated (B and D) bacterial cells. The scale bars for the study by staining the drug-treated cells with PI and 5(6)-
images are 1.0 μm (A, C, and D) and 200 nm (B). carboxyfluorescein diacetate N-succinimidyl ester (cFDA-SE)

This journal is © The Royal Society of Chemistry 2021 RSC Med. Chem., 2021, 12, 1005–1015 | 1009
View Article Online

Research Article RSC Medicinal Chemistry


Published on 24 May 2021. Downloaded by Indian Institute of Science on 7/30/2023 7:45:51 AM.

Fig. 5 Cartoon diagram demonstrating the antibiotic release pathways


from the liposomes of compound 7a (A). The antibiotics (tetracycline
and amoxicillin) release profile of compound 7a at pH 7.4 (B).
Concentration-dependent viabilities of the HeLa and HaCaT cells in
the presence of compounds 7a and 8 (C). Concentration-dependent
viabilities of the HaCaT cells in the presence of compounds 7a and
antibiotics-encapsulated compound 7a (D).

amoxicillin, respectively, were selected. Encapsulation and


Fig. 4 Fluorescence-based PI-uptake assay of compound 7a using S. release profiles of the antibiotics in compound 7a were
aureus (MRSA) cells (A). Compound 7a-treated membrane depolarization investigated by the fluorometric method (Fig. S7 and S8†).36
assay using DiSC3(5) dye on S. aureus (MRSA) cells (B). Representative Compound 7a showed the satisfactory loading ability for
fluorescence microscopic images of untreated and compound 7a-treated tetracycline (18%) and amoxicillin (28%) at pH 7.4,
bacterial cells (C). The scale bar for the images is 10 μm.
suggesting that these water-soluble antibiotics could be
entrapped within the hydrophilic core of the self-assembled
structure (Fig. 5A). Fluorescence measurements of these
was also done to investigate the bactericidal activity of the antibiotics showed sustained release profiles with 40% and
compound (Fig. 4C). The membrane-permeable 60% of tetracycline and amoxicillin, respectively, which were
nonfluorescent cFDA-SE dye only shows fluorescence due to released after 60 hours of incubation at 37 °C (Fig. 5B). These
the formation of carboxyfluorescein succinimidyl ester (CFSE) sustained antibiotics release profiles of compound 7a are
in the presence of esterase enzyme, which is an indicator of advantageous for their biological applications, including a
bacterial cell viability. During the measurements, the S. reduction in antibiotic-associated toxicity, augmentation of
aureus cells were stained with the cFDA-SE dye, and the activity of the antibiotics at lower doses, and others.
microscopic fluorescence images were recorded. The analysis The antibacterial activity of tetracycline and amoxicillin
revealed that the compound 7a-treated S. aureus cells lacked encapsulated liposomes of 7a (tetracycline@7a and
CFSE fluorescence, which demonstrated the inactivity of amoxicillin@7a) showed synergistic effects (Table S1†), and
bacterial metabolism due to superior membrane damage, the antibacterial activities were 4 to 6.5-folds lower than that
which resulted in cell death.

Table 2 Antibacterial activities of the commercial antibiotics and


Antibiotics encapsulation and antibacterial efficacy antibiotic-encapsulated sulfonium-based amphiphilic compound

The self-assembly property of compound 7a inspired us to MIC of 7a in


encapsulate the antibiotics and generate an antibacterial Bacterial MIC MIC antibiotic@7a
composite, where antibiotic and sulfonium-based Compound train (μM) (antibiotic@7a; μM) (μM)
compounds would show a potential synergistic effect. When 7a S. aureus 6.3 — —
encapsulated in liposomes, antibiotics and other drugs are E. coli 7.5 — —
Tetracycline S. aureus 4.2 0.7 1.0
more effective and protected from the adverse environment
E. coli 8.4 2.0 1.4
of the cell.35 To explore the antibiotic encapsulation, both Amoxicillin S. aureus 6.8 1.2 1.0
bacteriostatic and bactericidal antibiotics tetracycline and E. coli 10.9 1.7 1.3

1010 | RSC Med. Chem., 2021, 12, 1005–1015 This journal is © The Royal Society of Chemistry 2021
View Article Online

RSC Medicinal Chemistry Research Article

for free tetracycline and amoxicillin, suggesting the removed under reduced pressure, and CH2Cl2 (25 mL) was
accomplishment of dual activity of compound 7a and added. The salt was filtered off, and the filtrate was
antibiotics encapsulated within the liposome (Table 2). concentrated under reduced pressure. The crude product was
Therefore, the encapsulation of these antibiotics increases purified through silica gel column chromatography with a
the drug efficacy, and the controlled release of drugs could solvent gradient system using ethyl acetate and hexane (0–
provide the long-term effect and minimize the necessity of 1.5% EtOAc in hexanes) to obtain the pure product 2 as a
multiple dosing. clear yellow oil (200 mg; 45% yield). The compound was
An antimicrobial agent can be used for further bio- characterized by 1H and 13C NMR, and the results were in
application only when it is biocompatible and non-toxic to accordance with the literature report.37
Published on 24 May 2021. Downloaded by Indian Institute of Science on 7/30/2023 7:45:51 AM.

human cells. As our drug targets the bacterial membrane, it Synthesis of 1-(3,5-bis(bromomethyl)benzyl)pyridin-1-ium
should be specific to that only. Although the bacterial (3). To a stirring solution of 1,3,5-tris(bromomethyl)benzene
membrane differs from the mammalian cell membrane and (1, 1.40 mmol) in acetone solvent, pyridine (1.3 mmol) was
red blood cell membrane architecture, the drug should be added. The resulting mixture was stirred for 12 hours at
tested against mammalian cells because of the few room temperature. A white precipitate was observed, which
similarities in the membrane nature. In this regard, the was filtered and washed with acetone to afford the desired
cytocompatibility of the synthesized potent amphiphiles and product 3 as a white solid (350 mg, 70%). The compound was
antibiotic-loaded amphiphiles were verified against HeLa characterized by 1H and 13C NMR, and the results were in
(human cervical cancer) and HaCaT (human keratinocyte; accordance with the literature report.38
normal cells) cells by assessing the NADH-dependent cellular Synthesis of ((5-(azidomethyl)-1,3-phenylene)
oxidoreductase enzyme activities with the MTT dye (3-(4,5- bis(methylene))bis(dodecylsulfane) (4). To a stirring solution
dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) of 1-(azidomethyl)-3,5-bis(bromomethyl)benzene (2, 0.31
(Fig. 5C). The HaCaT cell line showed no toxicity up to 100 mmol) in CH3CN/H2O (3 : 1 in volume) was slowly added
μM against compound 8. The calculated IC50 value of (dropwise) a previously stirring solution of dodecanthiol and
compound 7a was 27 μM, while the IC50 values of sodium bicarbonate in CH3CN/H2O (3 : 1 in volume) at room
tetracycline@7a and amoxicillin@7a were 8 and 29.41 μM, temperature. The resulting reaction mixture was stirred for
respectively (Fig. 5D). Furthermore, the toxicity of the 36 hours. After that, the solvent was removed under reduced
synthesized amphiphiles was tested against the membrane of pressure. Then, the reaction mixture was diluted with cold
red blood cells, which is very much elastic, and the only water and ethyl acetate. The organic layer was extracted with
structural component it has is the plasma membrane that EtOAc (3 × 50 mL) and washed with brine, and dried over
provides mechanical support to the cell. To ensure the anhydrous Na2SO4. The organic solvent was removed under
compatibility of the sulfonium-based amphiphiles against reduced pressure. The crude reaction mixture was purified
red blood cells, the haemolytic activity was performed. through silica gel column chromatography with a solvent
Disruption of the membrane of the red blood cells causes gradient system using ethyl acetate and hexane (0–10% EtOAc
heme leakage from the cell that can be measured by the in hexane) to afford compound 4 as a colorless gummy solid.
spectrophotometric method (Fig. S9†). Amphiphiles at Characterization of the compound: colourless gummy liquid
various concentration ranges were incubated with 5% (yield – 87%) 1H NMR (600 MHz, CDCl3) δppm 7.23–7.22 (m,
haematocrit at room temperature. The extent of heme release 1H), 7.14 (s, 1H), 4.31 (s, 2H), 3.69–3.66 (m, 4H), 2.42–2.37
was measured by UV-vis spectroscopy at 410 nm after (m, 4H), 1.57–1.50 (m, 4H), 1.35–1.24 (m, 36H), 0.88 (t, 3H);
centrifugation. The result was compared with negative (no 13
C NMR (151 MHz, CDCl3) δppm 139.7, 135.8, 129.3, 127.2,
compound) and positive controls (0.1% Triton X-100), and it 54.5, 36.0, 31.9, 31.4, 29.7, 29.6, 29.6, 29.5, 29.4, 29.3, 29.2,
was found that at the MIC and MBC value, the compound 28.9, 22.7, 14.1; HRMS (ESI) calcd. for C33H63N4S2 [M +
has very less or no haemolytic activity (Table 1 and Fig. S9†). NH4]+: 579.4494, found: 579.4463.
The calculated therapeutic index (TI; IC50/MIC) of compound Synthesis of 1-(3,5-bis((dodecylthio)methyl)benzyl)pyridin-
7a and the composites were within 3.6–29.4 for both E. coli 1-ium (5a). To a stirring solution of 1-(3,5-bis(bromomethyl)
and S. aureus bacterial cells (Table S2†). benzyl)pyridin-1-ium (3, 0.14 mmol) in CH3CN/H2O (3 : 1 in
volume) was slowly added (dropwise) a previously stirring
Experimental solution of dodecanthiol (0.30 mmol) and sodium
bicarbonate (0.30 mmol) in CH3CN/H2O (3 : 1 in volume) at
I. Synthesis and characterization of the compounds room temperature. The resulting reaction mixture was stirred
Synthesis of 1-(azidomethyl)-3,5-bis(bromomethyl)benzene for 36 hours. After that, the solvent was removed under
(2). To a stirring solution of 1,3,5-tris(bromomethyl)benzene reduced pressure. Then, the reaction mixture was diluted
(1) (1.40 mmol) in dry DMF solvent, sodium azide (1.2 mmol) with cold water and ethyl acetate. The organic layer was
was added portion-wise, and the resulting mixture was stirred extracted with EtOAc (3 × 50 mL) and washed with brine, and
for 3 hours at room temperature. The progress of the reaction dried over anhydrous Na2SO4. The organic solvent was
was monitored by thin-layer chromatography (TLC). After the removed under reduced pressure. The crude reaction mixture
maximum consumption of compound 1, the solvent was was purified through silica gel column chromatography with

This journal is © The Royal Society of Chemistry 2021 RSC Med. Chem., 2021, 12, 1005–1015 | 1011
View Article Online

Research Article RSC Medicinal Chemistry

a solvent gradient system using ethyl acetate and hexane to afford compound 7b as a brown gummy liquid.
(0.1–10% EtOAc in hexanes) to afford compound 5a as a Characterization of the compound: dark brown gummy liquid
colorless gummy liquid. Characterization of the compound: (yield – 71%) 1H NMR (600 MHz, DMSO-d6) δppm 9.15–9.08
colorless gummy liquid (yield – 81%) 1H NMR (600 MHz, (m, 2H), 8.69–8.61 (m, 1H), 8.23–8.21 (m, 2H), 7.95 (s, 1H),
CDCl3 + DMSO-d6) 9.38–9.37 (m, 2H), 8.38–8.34 (m, H), 7.99– 7.58 (s, 2H), 5.93 (s, 2H), 4.71–4.62 (m, 4H), 2.89 (m, 2H),
7.95 (m, 1H), 7.33 (s, 1H), 7.27 (s, 1H), 7.20 (s, 1H), 6.17 (s, 2.77 (s, 6H), 2.73 (s, 2H), 1.33 (t, 3H); 13C NMR (151 MHz,
2H), 2.68 (s, 1H), 2.37–2.30 (m, 4H), 1.51–1.43 (m, 4H), 1.25– DMSO-d6) δppm 162.8, 146.8, 145.5, 136.6, 133.8, 132.2, 130.9,
1.18 (m, 38H), 0.81 (t, 3H); 13C NMR (151 MHz, CDCl3) δppm 128.9, 66.1, 62.9, 59.3, 43.4, 35.5, 31.2, 21.2, 11.25, 8.84; ES-
145.2, 145.0, 140.9, 132.9, 130.9, 128.3, 128.2, 64.3, 41.0, 36.0, MS (ESI+) m/z: [(M + 3BF4−] 889.525.
Published on 24 May 2021. Downloaded by Indian Institute of Science on 7/30/2023 7:45:51 AM.

32.1, 31.9, 29.6, 29.5, 29.3, 29.2, 28.9, 22.6, 14.0; HRMS (ESI) Synthesis of ((5-((4-(sulfomethyl)-1H-1,2,3-triazol-1-yl)
calcd. for C38H64NS2 [M]+: 598.4480, found: 598.4487. methyl)-1,3-phenylene)bis(methylene))bis(dodecyl- (methyl)
Synthesis of ((5-(pyridin-1-ium-1-ylmethyl)-1,3-phenylene) sulfonium) (8). To a stirring solution of ((5-(azidomethyl)-1,3-
bis(methylene))bis(dodecyl(methyl)sulfonium) (7a). To a phenylene)bis(methylene))bis-(dodecylsulfane) (4, 0.09 mmol)
stirring solution of 1-(3,5-bis((dodecylthio)methyl)benzyl) in DMF (3 mL) was added prop-2-yne-1-sulfonic acid (0.09
pyridin-1-ium (5a, 0.08 mmol) in dry DCM solvent was added mmol) and the mixture was stirred for 10 minutes. After that,
AgBF4 (0.08 mmol) and methyl iodide (0.2 mmol), and the sodium ascorbate (0.003 mmol) and CuI (0.002 mmol) were
resulting mixture was stirred for 5 hours at room added to the reaction mixture, and the solution was allowed
temperature. The progress of the reaction was monitored by to stir for 24 hours at room temperature. The reaction was
TLC. After consumption of compound 5a, the solvent was monitored by TLC. After that, the unused solvent was
removed under reduced pressure and the resulting mixture removed under reduced pressure and diluted with ethyl
was filtered off using a pad of celite to remove silver salt. acetate. The organic layer was separated, washed with brine,
Then, the crude mixture was washed with ether and CH3CN and dried over anhydrous Na2SO4. The solid crude product
to afford compound 7a as a brown gummy liquid. was washed with distilled hexane, the desired product 6 was
Characterization of the compound: brown gummy liquid almost pure, and the reaction mixture was used for the next
(yield – 77%); 1H NMR (600 MHz, DMSO-d6) δppm 9.14–9.13 reaction without further purification. To a stirring solution of
(m, 2H), 8.70–8.67 (m, 1H), 8.24–8.21 (m, 2H), 7.63 (s, 2H), compound 6 (0.58 mmol) in dry DCM, AgBF4 (0.058 mmol)
7.57 (s, 1H), 5.93 (s, 1H), 4.80–4.63 (m, 4H), 2.79 (s, 6H), was added, followed by methyl iodide (0.15 mmol). The
1.76–1.63 (m, 4H), 1.34–1.18 (m, 39H), 0.86 (t, 6H); 13C NMR resulting mixture was stirred for 5 hours at room
(151 MHz, DMSO-d6) δppm 146.8, 145.5, 136.6, 132.4, 130.9, temperature. TLC monitored the progress of the reaction.
128.9, 62.9, 46.1, 44.1, 40.91, 31.7, 29.5, 29.4, 29.2, 29.1, 28.8, After maximum consumption of compound 6, the solvent
28.2, 23.7, 22.5, 21.9, 14.4; ES-MS (ESI+) m/z: [(M + 3BF4−] was removed under reduced pressure, and the resulting
609.4756. mixture was filtered off using a pad of celite to remove silver
Synthesis of ((5-(pyridin-1-ium-1-ylmethyl)-1,3-phenylene) salt. Then, the crude mixture was washed with ether and
bis(methylene))bis(ethyl(methyl)sulfonium) (7b). To a stirring CH3CN to afford compound 8 as a brown gummy solid.
solution of 1-(3,5-bis(bromomethyl)benzyl)pyridin-1-ium (3, Characterization of the compound: dark brown gummy solid
0.28 mmol) in CH3CN/H2O (3 : 1 in volume) was slowly added (yield – 68%) - 1H NMR (600 MHz, CDCl3) δppm 9.82 (s, 1H),
(dropwise) a previously stirring solution of ethanethiol (0.60 8.02 (s, 2H), 7.95 (s, 1H), 4.71–4.62 (m, 2H), 3.78–3.76 (m,
mmol) and sodium bicarbonate (0.60 mmol) in CH3CN/H2O 2H), 3.34–3.17 (m, 4H), 2.81 (s, 6H), 2.35–2.28 (m, 1H), 1.87–
(3 : 1 in volume) at room temperature. The resulting reaction 1.78 (m, 4H), 1.44–1.40 (m, 4H), 1.29–1.23 (m, 41H), 0.86 (t,
mixture was stirred for 36 hours. After that, the solvent was 6H); 13C NMR (151 MHz, CDCl3) δppm 138.3, 133.0, 130.1,
removed under reduced pressure. Then, the reaction mixture 129.0, 128.0, 125.3, 68.1, 47.2, 44.6, 41.4, 31.9, 29.7, 29.7,
was diluted with cold water and ethyl acetate. The organic 29.6, 29.4, 29.0, 28.4, 25.6, 24.1, 22.7, 21.3, 14.1; ES-MS (ESI+)
layer was extracted with EtOAc (3 × 50 mL) and washed with m/z: [(M/2 + 2BF4− + Na+] 552.0522.
brine, and dried over anhydrous Na2SO4. After that, the
solvent was removed under reduced pressure to afford a
white solid, which was used for the next reaction without II. Zeta-potential and DLS measurement
further purification. Then, to the stirring solution of the The surface potential and hydrodynamic diameter of the
above white solid 1-(3,5-bis((ethylthio)methyl)benzyl)pyridin- compounds in the aqueous environment was measured by
1-ium (0.06 mmol) (5b) in dry DCM was added AgBF4 (0.06 zeta potential and dynamic light scattering (DLS) using a Zeta
mmol) and methyl iodide (0.16 mmol), and the resulting sizer ZS90 (Malvern, Westborough, MA) instrument at 25 °C.
mixture was stirred for 5 hours at room temperature. TLC The stock solution of the compound was prepared in
monitored the progress of the reaction. After total chloroform, and multi-layered vesicles were prepared by
consumption of the starting compound, the solvent was rotary evaporation method with the help of a vacuum pump.
removed under reduced pressure, and the resulting mixture To this dried compound, phosphate-buffered saline (PBS; 137
was filtered off using a pad of celite to remove silver salt. mM NaCl, 2.7 mM KCl, 8 mM Na2HPO4, and 2 mM KH2PO4
Then, the crude mixture was washed with ether and CH3CN at pH 7.4) buffer was added and kept for hydration overnight

1012 | RSC Med. Chem., 2021, 12, 1005–1015 This journal is © The Royal Society of Chemistry 2021
View Article Online

RSC Medicinal Chemistry Research Article

at 50 °C. To obtain the unilamellar vesicles, a sample vial encapsulation of the antibiotics. The unloaded antibiotic was
underwent vortexing and intermittent sonication. Disposable removed by centrifugation. The antibiotics loading efficacy
zeta cells (DTS1061) and a 3 mL fluorescence cuvette were was measured by a UV-vis spectrophotometer. The
used for the surface potential and DLS measurement. encapsulated liposome was treated with 5% Triton X-100 to
break the liposome, and the released antibiotic was
III. Transmission electron microscopic measurements monitored by its absorbance. The absorption of the antibiotic
The transmission electron microscope (TEM) was used to alone was also recorded. The concentration of the loaded
investigate the morphology of the aqueous soluble aggregates antibiotic was calculated by Lambert–Beer's law, A = ε·Cab·l,
of the lipids. The lipid solution was prepared by the method where Cab is the released antibiotic concentration from the
Published on 24 May 2021. Downloaded by Indian Institute of Science on 7/30/2023 7:45:51 AM.

mentioned above in PBS, pH 7.4. The freshly prepared lipid liposome. For this calculation, the calibration curve was
solution (without extrusion) was diluted to half of its original obtained to calculate the extinction coefficient (ε) by the
concentration using 1 mL PBS, pH 7.4. Then, 10 μL of lipid absorption of the antibiotics at different concentrations. The
solution was taken and placed onto a carbon-coated copper drug release profile was observed at different time intervals
grid and allowed to absorb for 1 minute. The grid was then up to 60 hours by fluorescence spectrofluorometer (for
carefully blotted with filter paper, and only a trace amount of tetracycline, the λex and λem values were 390 and 550 nm,
the solution in the middle of the grid was kept. After that, respectively; for amoxicillin, the λex and λem values were 390
the grid was allowed to dry for 10 minutes at 30 °C. Finally, and 550 nm, respectively36). All of the measurements were
5–10 μL of 2% uranyl acetate solution (in water) was added to performed in PBS. For 100% release of the antibiotic, the
the grid, and allowed to dry for another 1 min. The excess liposome suspension was treated with Triton X-100, followed
uranyl acetate solution was wicked off, and the grid was dried by sonication and fluorescence measurement. The same
overnight at 30 °C. The images were collected using a JEOL experimental protocol measured the antimicrobial activity of
JEM 2100 transmission electron microscope (operated at a the antibiotic encapsulated liposome.
maximum accelerating voltage of 200 kV).
VI. Antibiofilm activity study
IV. Antimicrobial activity and bactericidal activity
The antibiofilm activity study was performed by the crystal
The antimicrobial activities of the compounds were tested by violet staining method. S. aureus MTCC 96 is a well-known
using a broth dilution assay in 96-well plates. E. coli MTCC non-motile, sessile strain and forms a biofilm on the bottom
1687 and P. aeruginosa MTCC 2488 were cultured in Luria of the wells. The bacterial cells were grown and treated with
Bertani (LB), and S. aureus MTCC 96 and methicillin-resistant compounds, as mentioned in an earlier section. After the
S. aureus (MRSA) cells were cultured in Brain Heart Infusion overnight incubation, the planktonic cells were pipetted out,
(BHI) broth media until the mid-logarithmic phase at 37 °C and the wells were rinsed with PBS. The wells were air-dried
in a shaker incubator and harvested by centrifugation. Then, under laminar airflow. Then, the crystal violet solution (1%,
the cells were washed with PBS buffer and suspended in the v/v) was added to each well, and the plate was incubated for
same buffer. In the presence of varying concentrations of the 20 minutes. After incubation, the crystal violet solution was
compounds, bacterial cells were incubated at 106 CFU mL−1 taken out, and the wells were dried. Then, ethanol (95%) was
at 37 °C. After 14–16 hours of incubation, the optical density added to each well, and the absorbance was taken at 590 nm
(OD) was measured at 600 nm by using a Tecan infinite using a 96 micro-titer plate reader (Infinite M200, TECAN,
M200 Plate reader. For the bactericidal activity test, the Switzerland).
solution with no visible growth was transferred into a fresh
well containing only media. The plate was incubated for 24
hours at 37 °C, and then the OD was measured. To get the VII. Propidium iodide uptake assay
closest minimum inhibitory concentration (MIC) value, this The stock solution (1.5 mM) of propidium iodide (PI) was
antimicrobial experiment was repeated at a variable prepared in sterile Milli-Q water and stored at 4 °C. The S.
concentration range, and was repeated thrice at the reported aureus MTCC 96 cells were cultured overnight and collected
MIC value procedure. by centrifugation, and resuspended in the same media. Cells
were again diluted in media at 106 CFU mL−1. After the
V. Antibiotic loading and release profile study addition of the compound, the cells were incubated at 37 °C
The stock solutions of tetracycline and amoxicillin antibiotics at 180 rpm. The sample (1 mL) was collected at the 1st, 2nd,
were prepared in PBS. The compound was dissolved in and 3rd hour. The culture was centrifuged and washed with
chloroform and dried by the rotary evaporation method to PBS, and the cells were incubated with PI at 30 μM
form a multilayer film. To this dried vial containing a dried concentration for half an hour. Cells were again centrifuged,
layer of the compound, the aqueous solution of antibiotic washed, and resuspended with PBS, and the fluorescence
was added and incubated at 37 °C in a water bath for measurement was recorded at an excitation wavelength of
hydration. The liposomes underwent 7–8 freeze–thaw cycles, 535 and emission wavelength of 617 in a spectrofluorometer
followed by vigorous vortexing for 2 hours for superior (HORIBA, flioroMax-4).

This journal is © The Royal Society of Chemistry 2021 RSC Med. Chem., 2021, 12, 1005–1015 | 1013
View Article Online

Research Article RSC Medicinal Chemistry

VIII. Fluorescence imaging assay (DMSO) in each well. The absorbance was recorded at 570
The PI and cFDA-SE dyes were used to stain the dying and nm to measure the cell viability by using a plate reader.
viable bacterial cells. The S. aureus cells were cultured and
harvested, as mentioned in an earlier section. Cells were XII. Haemolytic assay
treated with the compound, and only buffer was added to the To test the toxicity of the compound against the blood cells,
cell culture as a control. After 5 h of incubation, the cells haemolysis screening was performed according to the
were collected by centrifugation and washed with buffer, and reported protocol. The haemolytic assay was performed with
diluted up to 106 CFU mL−1. The cells were separately stained erythrocytes extracted from fresh human blood. Blood was
with PI at a concentration of 30 μM and cFDA-SE at a centrifuged at 1500 rpm for 10 minutes, the supernatant was
Published on 24 May 2021. Downloaded by Indian Institute of Science on 7/30/2023 7:45:51 AM.

concentration of 10 μM. After 30 min of incubation, 10 μL of discarded, and cells were washed with PBS. The 5%
the stained sample was taken on a thin glass slide and haematocrit was prepared in the same media. Serially diluted
observed under the fluorescence microscope. sulfonium compounds were added in a vial up to 240 μL,
including a negative control with only buffer and positive
control with 0.5% Triton X-100 and 60 μL of the 5%
IX. Morphological study
haematocrit was added to each vial, and incubated for 1 hour
The morphological assessment was done by field emission at 37 °C. After incubation, the vials were centrifuged at 1500
scanning electron microscope (FESEM). The S. aureus MTCC rpm for 10 minutes, and 50 μL of the supernatant was added
96 and E. coli MTCC 1687 cells were cultured and harvested to fresh 96-well plates. The absorbance was recorded at 410
by centrifugation at 5000 rpm for 5 minutes. Cells were nm. The% haemolysis was calculated by comparing the
washed and treated with the compound, and the control was absorbance of the sample with the positive control and the
taken as the bacterial culture without compound treatment. absorbance of the negative control. All experiments were
After 5 hours of incubation, cells were collected by performed in accordance with the Guidelines of the
centrifugation, washed with Milli-Q water, drop-casted on an Institutional Human Ethics Committee (IHEC), and the
aluminum foil studded glass grid, and air-dried under experiments were approved by the ethics committee at Indian
laminar airflow. The drop-casted sample was again mounted Institute of Technology Guwahati. Informed consents were
on the FESEM metal grid sandwiched by carbon tape before obtained from the human participants of this study.
the FESEM analysis sample was double-coated by gold.
Conclusions
X. Membrane depolarization study
Bacterial infections and antibiotic resistance are rapidly
For this study, S. aureus and MTCC 96 cells were grown as becoming one of the most daunting healthcare burdens of
mentioned in the above section until the mid-log phase and our day, which immediately need effective therapeutic
harvested, followed by resuspension in HEPES buffer (10 mM strategies. The current study demonstrates that the modular
HEPES and 50 mM glucose). To this, DiSC35 was added at a synthesis of membrane-active sulfonium-based lipids can
final concentration of 0.4 μM, and cells were incubated for 1 effectively form liposomes, and encapsulate the commercially
h. After this, KCl was added to the cell suspension at a available antibiotics to have a synergistic effect against
concentration of 100 mM. After 10 min incubation, variable bacterial strains responsible for respiratory infections like
concentrations (0, 2, and 4 μM) of the compound and 30 μM cystic fibrosis. Liposomal aggregates are the rising armour to
valinomycin as a positive control were added to the cell strive against such respiratory syndromes. The fascinating
suspension, and the fluorescence spectra were recorded at properties of the antibiotic-encapsulated liposomes were the
excitation and emission wavelengths of 620 nm and 650 nm, prudent assimilation of two weapons, which permitted
respectively. harnessing their concomitant activity. The membrane-
directed properties of the lipids make them less prone to
XI. Cytocompatibility test drug resistance, and the most potent lipid also augments the
activity of commercial antibiotics. We envisage that this
To test the viability of human cells in the presence of the inherent antimicrobial drug carrier can be proven as the
antimicrobial drugs, HeLa cells (1 × 104 cells/well) were panacea towards the path of antibiotic stewardship.
seeded into 96-well plates in 100 μL of DMEM per well and
left overnight for attachment. The cells were then treated
with variable concentrations of the compound, and only
Conflicts of interest
buffer as the control. After incubation for 24 h, 10 μL MTT There are no conflicts to declare.
reagent (5 mg mL−1) was added to each well (containing cells
in DMEM media with compounds), and then incubated for Acknowledgements
another 3 h at 37 °C in a CO2 incubator. Later, the MTT
reagent was removed from the wells, and formazan crystals We are thankful to the Department of Biotechnology [BT/
were dissolved by adding 100 μL of dimethyl sulfoxide PR41335/NNT/28/1780/2021 and NECBH/2019-20/160] for

1014 | RSC Med. Chem., 2021, 12, 1005–1015 This journal is © The Royal Society of Chemistry 2021
View Article Online

RSC Medicinal Chemistry Research Article

financial support. We are also thankful to the Department of 19 R. G. Carden, K. J. Sommers, C. L. Schrank, A. J. Leitgeb,
Chemistry, Department of Bioscience and Bioengineering, J. A. Feliciano, W. M. Wuest and K. P. C. Minbiole,
Central Instruments Facility and Centre for the Environment, ChemMedChem, 2020, 15, 1974–1984.
IIT Guwahati for instrument facilities. 20 S. Dey, A. Patel, K. Raina, N. Pradhan, O. Biswas, R. P.
Thummer and D. Manna, Chem. Commun., 2020, 56,
Notes and references 1661–1664.
21 D. Y. Wang, H. C. van der Mei, Y. J. Ren, H. J. Busscher and
1 J. A. Fishman, Am. J. Transplant., 2017, 17, 856–879. L. Q. Shi, Front. Chem., 2020, 7, 872.
2 C. Gudiol and J. Carratala, Expert Rev. Anti-Infect. Ther., 22 A. Kumar, C. Boyer, L. Nebhani and E. H. H. Wong, Sci. Rep.,
Published on 24 May 2021. Downloaded by Indian Institute of Science on 7/30/2023 7:45:51 AM.

2014, 12, 1003–1016. 2018, 8, 7965.


3 L. E. Papanicolas, D. L. Gordon, S. L. Wesselingh and G. B. 23 M. Balouiri, M. Sadiki and S. K. Ibnsouda, J. Pharm. Anal.,
Rogers, Trends Microbiol., 2018, 26, 393–400. 2016, 6, 71–79.
4 T. Wang, E. B. Ahmed, L. Chen, J. Xu, J. Tao, C. R. Wang, 24 T. Koburger, N.-O. Hübner, M. Braun, J. Siebert and A.
M. L. Alegre and A. S. Chong, Am. J. Transplant., 2010, 10, Kramer, J. Antimicrob. Chemother., 2010, 65, 1712–1719.
1524–1533. 25 T. M. Karpiński, Eur. J. Biol. Res., 2019, 3, 135–140.
5 K. Thitiananpakorn, Y. Aiba, X. E. Tan, S. Watanabe, K. Kiga, 26 M. Hirayama, Biocontrol Sci., 2011, 16, 23–31.
Y. Sato’o, T. Boonsiri, F. Y. Li, T. Sasahara, Y. Taki, A. H. 27 L. Li, D. Jia, H. Wang, C. Chang, J. Yana and Z. K. Zhao, New
Azam, Y. C. Zhang and L. Z. Cui, Sci. Rep., 2020, 10, 16107. J. Chem., 2020, 44, 303–307.
6 Y. L. Guo, G. H. Song, M. L. Sun, J. Wang and Y. Wang, 28 A. Kanazawa, T. Ikeda and T. Endo, Polym. Chem., 1993, 31,
Front. Cell. Infect. Microbiol., 2020, 10, 107. 2873–2876.
7 Y. C. Yeh, T. H. Huang, S. C. Yang, C. C. Chen and J. Y. 29 D. S. Uppu, P. Akkapeddi, G. B. Manjunath, V. Yarlagadda, J.
Fang, Front. Chem., 2020, 8, 286. Hoque and J. Haldar, Chem. Commun., 2013, 49, 9389–9391.
8 M. P. Mingeot-Leclercq and J. L. Decout, MedChemComm, 30 C. Vijayakumar and C. E. Wolf-Hall, Food Microbiol.,
2016, 7, 586–611. 2002, 19, 383–388.
9 C. Dias and A. P. Rauter, Future Med. Chem., 2019, 11, 211–228. 31 G. Zhao, M. L. Usui, S. I. Lippman, G. A. James, P. S. Stewart,
10 J. G. Hurdle, A. J. O'Neill, I. Chopra and R. E. Lee, Nat. Rev. P. Fleckman and J. E. Olerud, Adv. Wound Care, 2013, 2,
Microbiol., 2011, 9, 62–75. 389–399.
11 R. W. Finberg, R. C. Moellering, F. P. Tally, W. A. Craig, G. A. 32 R. Funari, N. Bhalla, K. Y. Chu, B. Soderstrom and A. Q.
Pankey, E. P. Dellinger, M. A. West, M. Joshi, P. K. Linden, Shen, ACS Sens., 2018, 3, 1499–1509.
K. V. Rolston, J. C. Rotschafer and M. J. Rybak, Clin. Infect. 33 M. Yasir, D. Dutta and M. D. P. Willcox, Sci. Rep., 2019, 9,
Dis., 2004, 39, 1314–1320. 7063.
12 G. A. Pankey and L. D. Sabath, Clin. Infect. Dis., 2004, 38, 34 R. Kannan, P. Prabakaran, R. Basu, C. Pindi, S. Senapati, V.
864–870. Muthuvijayan and E. Prasad, ACS Appl. Bio Mater., 2019, 8,
13 R. M. Epand, C. Walker, R. F. Epand and N. A. Magarvey, 3212–3224.
Biochim. Biophys. Acta, 2016, 1858, 980–987. 35 A. G. Gomez and Z. Hosseinidoust, ACS Infect. Dis., 2020, 6,
14 P. Gilbert and L. E. Moore, J. Appl. Microbiol., 2005, 99, 896–908.
703–715. 36 L. H. Ma, L. Chen and S. P. Huang, Spectrosc. Spect. Anal.,
15 L. E. Moore, R. G. Ledder, P. Gilbert and A. J. McBain, Appl. 1999, 19, 230–232.
Environ. Microbiol., 2008, 74, 4825–4834. 37 P. R. Werkhoven, M. Elwakiel, T. J. Meuleman, H. C. Q. van
16 L. E. Papanicolas, D. L. Gordon, S. L. Wesselingh and G. B. Ufford, J. A. W. Kruijtzer and R. M. J. Liskamp, Org. Biomol.
Rogers, Trends Microbiol., 2018, 26, 393–400. Chem., 2016, 14, 701–710.
17 P. Dey, S. Mukherjee, G. Das and A. Ramesh, J. Mater. Chem. 38 J. N. Marafino, T. M. Gallagher, J. Barragan, B. L. Volkers,
B, 2018, 6, 2116–2125. J. E. LaDow, K. Bonifer, G. Fitzgerald, J. L. Floyd, K.
18 D. Guan, F. Chen, Y. Qiu, B. Jiang, L. Gong, L. Lan and W. McKenna, N. T. Minahan, B. Walsh, K. Seifert and K. L.
Huang, Angew. Chem., Int. Ed., 2019, 58, 6678–6682. Caran, Bioorg. Med. Chem., 2015, 23, 3566–3573.

This journal is © The Royal Society of Chemistry 2021 RSC Med. Chem., 2021, 12, 1005–1015 | 1015

You might also like