Skoči na glavni sadržaj

Izvorni znanstveni članak

https://doi.org/10.5599/admet.2684

Insight into antistaphylococcal effect of chlorinated 1-hydroxynaphthalene-2-carboxanilides

Lucia Vrablova orcid id orcid.org/0009-0008-4701-5265 ; Department of Analytical Chemistry, Faculty of Natural Sciences, Comenius University, Ilkovicova 6, 842 15 Bratislava, Slovakia
Tomas Gonec orcid id orcid.org/0000-0003-3712-8641 ; Department of Chemical Drugs, Faculty of Pharmacy, Masaryk University, Palackeho tr. 1946/1, 612 00 Brno, Czech Republic
Petra Majerova ; Institute of Neuroimmunology, Slovak Academy of Sciences, Dubravska cesta 9, 845 10 Bratislava, Slovakia
Andrej Kovac ; Institute of Neuroimmunology, Slovak Academy of Sciences, Dubravska cesta 9, 845 10 Bratislava, Slovakia
Dominika Kos ; Department of Molecular Pharmacy, Faculty of Pharmacy, Masaryk University, Palackeho tr. 1946/1, 612 00 Brno, Czech Republic
Peter Kollar orcid id orcid.org/0000-0003-2265-1528 ; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Masaryk University, Palackeho tr. 1946/1, 612 00 Brno, Czech Republic
Jiri Kos ; Department of Biochemistry, Faculty of Medicine, Masaryk University, Kamenice 5, Brno 625 00, Czech Republic
Alois Cizek ; Department of Infectious Diseases and Microbiology, Faculty of Veterinary Medicine, University of Veterinary Sciences Brno, Palackeho tr. 1946/1, 612 42 Brno, Czech Republic
Tereza Kauerova ; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Masaryk University, Palackeho tr. 1946/1, 612 00 Brno, Czech Republic
Josef Jampilek orcid id orcid.org/0000-0003-2003-9052 ; Department of Analytical Chemistry, Faculty of Natural Sciences, Comenius University, Ilkovicova 6, 842 15 Bratislava, Slovakia


Puni tekst: engleski pdf 1.032 Kb

preuzimanja: 98

citiraj

Preuzmi JATS datoteku


Sažetak

Abstract
Background and purpose
New compounds and innovative therapeutic approaches are trying to prevent antimicrobial resistance, which has become a global health challenge.
Experimental approach
This study includes a series of twelve mono-, di- and trichlorinated 1-hydroxynaphthalene-2-carboxanilides designed as multitarget agents. All compounds were evaluated for their antistaphylococcal activity. Furthermore, MTT assay and chemoproteomic analysis of selected compounds were performed. Cytotoxicity in human cells was also tested.
Key results
N-(3,5-Dichlorophenyl)-1-hydroxynaphthalene-2-carboxamide (10) demonstrated activity comparable to or higher than clinically used drugs, with minimum inhibitory concentrations (MICs) of 0.37 μM. The compound was equally effective against clinical isolates of methicillin-resistant S. aureus. On the other hand, compound 10 showed 96 % inhibition of S. aureus respiration only at a concentration of 16× MIC. Chemoproteomic analysis revealed that the effect of agent 10 on staphylococci resulted in the downregulation of four proteins. This compound expressed no in vitro cytotoxicity up to a concentration of 30 μM.
Conclusion
From the set of tested mono-, di- and trisubstituted derivatives, it is evident that the position of chlorine atoms is decisive for significant antistaphylococcal activity. Inhibition of energy metabolism does not appear to be one of the main mechanisms of action of compound 10; on the contrary, the antibacterial effect may likely be contributed by downregulation of proteins (especially ATP-dependent protease ATPase subunit HslU) involved in processes essential for bacterial survival and growth, such as protein, nucleotide/nucleic acid synthesis and efficient protein repair/degradation.

Ključne riječi

Lipophilicity; antistaphylococcal activity; cytotoxicity; MTT assay; chemoproteomic analysis

Hrčak ID:

330594

URI

https://hrcak.srce.hr/330594

Datum izdavanja:

26.3.2025.

Posjeta: 278 *




Introduction

Antimicrobial resistance (AMR) is a major global health challenge of the 21st century. Dangerous drug-resistant bacteria include third-generation cephalosporin-resistant Escherichia coli and methicillin-resistant Staphylococcus aureus, as well as Klebsiella pneumoniae, Pseudomonas aeruginosa, and vancomycin-resistant Enterococcus faecalis/Enterococcus faecium [1-5]. Studies published in the Lancet highlight a worrying trend in the burden of AMR among people over 70 years of age, which is critical given the rapidly ageing ‘Western’ global community. Given the high variability in the burden of AMR by location and age, it is important that interventions to prevent/reduce AMR, leading to a reduction in deaths, combine infection prevention, vaccination, minimization of inappropriate antibiotic use in agriculture and humans, and research into new antibiotics [5-7].

There are many innovative approaches to the discovery, design and development of new antimicrobial drugs [8-12]; one of them is the design of multitargeted agents, i.e. compounds capable of interfering with microorganisms at multiple sites and preventing them from developing resistance through multiple interventions, whether in serial or parallel action on metabolic pathways, reproduction or action on membrane systems [13-17].

Salicylamides are a typical example of such multitarget agents, which are characterized by a complex anti-invasive effect against parasites, microorganisms and cancer cells [16,18-20,21 and Refs. therein,22-26], they even have herbicidal effects [27,28]. Hydroxynaphthalenecarboxanilides, cyclic analogues of the parent salicylanilides prepared by our team, can also be considered multitarget compounds. 1-Hydroxynaphthalene--2-carboxanilides have been investigated primarily as potential anti-infective [29-31] and anti-cancer [32,33] agents. Although it is believed that these molecules may act as proton shuttle inhibitors, i.e. these compounds may destroy the cellular proton gradient [34,35], salicylamides' main mechanisms of action appear to be inhibition of many enzymes [19-27,28 and Refs. therein], including inhibition of the two-component regulatory system of bacteria [36,37].

A disadvantage of multitargeted agents is that it is difficult to precisely determine/identify their mechanisms of action. One option that could help uncover the mechanism of action of these compounds is the use of chemoproteomics, which has become a useful tool in modern drug discovery and preclinical research [38], providing critical insight into the interactions between bioactive compounds and protein targets within complex biological systems [39]. By identifying the proteins to which small molecules bind, chemoproteomics allows for mapping the mechanisms of action of potential therapeutics as well as identifying unintended off-target effects that could lead to toxicity [40]. The primary goal of chemoproteomics is to investigate how small bioactive molecules interact with proteins in their native cellular environment [41]. This is typically achieved using activity-based protein profiling (ABPP), a powerful chemoproteomics technique that uses small molecular probes to label and capture proteins in a functional state [42]. The resulting labeled proteins can then be analyzed using liquid chromatography-tandem mass spectrometry (LC-MS/MS), which provides a detailed proteomic profile of the cellular response to the compound [38,43].

Given the demonstrated improvement of anti-infective properties through chlorination [22,44-50], this study aims to elucidate the previously unreported antistaphylococcal activity of chlorinated 1-hydroxynaphthalene-2-carboxanilides. Specifically, the collection of susceptible S. aureus and its MRSA isolates were deliberately selected as they represent the most prevalent and clinically significant Gram-positive pathogens responsible for a spectrum of infections ranging from superficial skin lesions to severe conditions like pneumonia and sepsis [51]. To investigate the mechanism of action, not only the antistaphylococcal efficacy was evaluated, but also the impact of active derivatives on bacterial respiration (energy metabolism) and intracellular processes. The application of a contemporary chemoproteomic approach, including the comparative ABPP technique, supported the effort to determine the protein profile alterations induced in both reference S. aureus and MRSA strains following treatment with selected ring-substituted 1-hydroxynaphthalene-2-carboxanilides. This comprehensive study, including proteomic analysis, is expected to provide valuable insights into the molecular mechanisms underlying the antistaphylococcal effects of these novel chlorinated compounds.

Experimental

Synthesis

The synthetic pathway and characterization of compounds 1-4 were described by Gonec et al. [29], while compounds 5-13 were described in [52].

Lipophilicity determination by HPLC

An HPLC system Agilent 1200 equipped with a DAD detector (Agilent, Santa Clara, CA, USA) was used. A chromatographic column Symmetry® C18 5 μm, 4.6×250 mm, part No. WAT054275 (Waters Corp., Milford, MA, USA) was used. The HPLC separation process was monitored and evaluated with EZChrom Elite software ver. 3.3.2 [53] (Agilent). Isocratic elution by a mixture of MeOH p.a. (72 %) and H2O-HPLC Mili-Q grade (28 %) as a mobile phase was used for the determination of capacity factor k. Isocratic elution by a mixture of MeOH p.a. (72 %) and acetate-buffered saline (pH 7.4 and pH 6.5) (28 %) as a mobile phase was used for the determination of distribution coefficients expressed as D7.4 and D6.5. The total flow of the column was 1.0 mL/min, the injection volume was 20 μL, the column temperature was 40 °C, and the sample temperature was 10 °C. The detection wavelength of 210 nm was chosen. A KI methanolic solution was used for the determination of the dead times (tD). Retention times (tR) were measured in minutes. The capacity factors k were calculated according to the formula k = (tRtD)/tD, where tR is the retention time of the solute, and tD is the dead time obtained using an unretained analyte. The distribution coefficients DpH were calculated according to the formula DpH = (tRtD)/tD. Each experiment was repeated three times.

Antistaphylococcal screening

In vitro antistaphylococcal activity of the synthesized compounds was evaluated against three clinical isolates of methicillin-resistant S. aureus: clinical isolate of animal origin, MRSA 63718 (Department of Infectious Diseases and Microbiology, Faculty of Veterinary Medicine, University of Veterinary Sciences Brno, Czech Republic) [54], carrying the mecA gene [55]; and MRSA SA 630 and MRSA SA 3202 [54] (National Institute of Public Health, Prague, Czech Republic), both of human origin. Suspected colonies were confirmed by PCR; a 108bp fragment specific to S. aureus was detected [56]. All isolates were tested for the presence of the mecA gene encoding methicillin resistance [57]. These three clinical isolates were classified as vancomycin-susceptible (but with higher MIC of vancomycin equal to 2 μg/mL (VA2-MRSA) within the susceptible range for MRSA 63718) methicillin-resistant S. aureus (VS-MRSA) [54]. Vancomycin-susceptible methicillin-susceptible S. aureus (VS-MSSA) ATCC 29213, obtained from the American Type Culture Collection, was used as the reference and quality control strain. The minimum inhibitory concentrations (MICs) were evaluated by the microtitration broth method according to the CLSI [58,59] with some modifications. The compounds were dissolved in DMSO (Sigma, St. Louis, MO, USA) to get a concentration 10 μg/mL and diluted in a microtitration plate in a medium Cation Adjusted Mueller-Hinton (CaMH, Oxoid, Basingstoke, UK) to reach the final concentration of 256-0.0625 μg/mL. The plate was inoculated by the tested microorganisms. The final concentration of bacterial cells was 105 for bacteria. Ampicillin and ciprofloxacin (Sigma) were used as reference drugs. A drug-free control and a sterility control were included. The plates were incubated for 24 h at 37 °C. After static incubation in the darkness in an aerobic atmosphere, the MIC was visually evaluated as the lowest concentration of the tested compound, which completely inhibited the growth of the microorganism. The experiments were repeated three times.

MTT assay

Compounds were prepared as previously stated and diluted in CaMH broth for S. aureus to achieve the desired final concentrations. S. aureus bacterial suspension in sterile distilled water at 0.5 McFarland was diluted 1:3. Inocula were added to each well by multi-inoculator. Diluted mycobacteria in broth free from inhibiting compounds were used as the growth control. All compounds were prepared in duplicate. Plates were incubated at 37 °C for 24 h for S. aureus. After the incubation period, 10 % well volume of MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) reagent (Sigma) was mixed into each well and incubated at 37 °C for 1 h for S. aureus. Then 100 μL of 17 % sodium dodecyl sulphate in 40 % dimethylformamide was added to each well. The plates were read at 570 nm. The absorbance readings from the cells grown in the presence of the tested compounds were compared with uninhibited cell growth to determine the relative percent inhibition. The percent inhibition was determined through the MTT assay. The percent viability is calculated through the comparison of a measured value and that of the uninhibited control: Viability = (OD570E/OD570P)×100, where OD570E is the reading from the compound-exposed cells, while OD570P is the reading from the uninhibited cells (positive control). Cytotoxic potential is determined by a percent viability of <70 % [60,61].

Cytotoxicity assay

Cytotoxicity of the compounds was determined using an LDH assay kit (Roche, Mannheim, Germany) as described previously [31]. Human monocytic leukemia THP-1 cells (European Collection of Cell Cultures, Salisbury, UK) were exposed for 24 h at 37 °C to various compound concentrations ranging from 0.37 to 30 μM in RPMI 1640 medium. For LDH assays, cells were seeded into 96-well plates (5×104 cells/well in 100 μL of culture medium) in triplicate in serum-free RPMI 1640 medium and measurements were taken 24 h after the treatment with the compounds. The maximum concentration of DMSO (Sigma) in the assays never exceeded 0.1 %. Oxaliplatin and camptothecin (Sigma) were used as reference drugs. The median lethal dose values, LC50, were deduced through the production of a dose-response curve. All data from three independent experiments were evaluated using GraphPad Prism 5.00 software [62].

Proteomic study

This study utilized activity-based protein profiling (ABPP), specifically its comparative technique, to identify protein targets within the proteome of cell lysates. The experiments were conducted on the universally sensitive collection strain Staphylococcus aureus ATCC 29213 and MRSA SA 630 isolate [54]. Both strains were treated as described above. Prior to MS detection, proteins underwent tryptic digestion.

The bacterial pellet was homogenized in a lysis buffer containing 200 mM Tris, 150 mM NaCl, 1 mM ethylenediaminetetraacetic acid (EDTA), 1 mM sodium orthovanadate (Na3VO4), 20 mM sodium fluoride (NaF), 0.5 % Triton X-100 (pH 7.4) (Sigma), and a complete protease inhibitor cocktail (Roche). Protein precipitation was carried out overnight using 80 % ice-cold acetone (Sigma). After centrifugation, the pellets were solubilized in 8 M urea (Sigma), and protein concentrations were determined using the Bio-Rad protein assay (Bio-Rad Laboratories, Colbe, Germany). A total of 100 μg of protein was reduced with 10 mM dithiothreitol (Sigma) in 100 mM ammonium bicarbonate at 37 °C for one hour. Alkylation was performed with 15 mM iodoacetamide (Sigma) in 100 mM ammonium bicarbonate, protected from light for 30 min. Trypsin digestion (Promega, Madison, WI, USA) was performed at an enzyme-to-substrate ratio of 1:100, with overnight incubation at 37 °C.

Aliquots of purified complex peptide mixtures of 100 ng were separated using Acquity M-Class UHPLC (Waters). Samples were loaded onto the nanoEase Symmetry C18 trap column (25 mm length, 180 μm diameter, and 5 μm particle size). After 2 min of desalting/concentration by 1 % acetonitrile containing 0.1 % formic acid at a flow rate of 8 μL/min, peptides were introduced to the nanoEase HSS T3 C18 analytical column (100 mm length, 75 μm diameter, and 1.8 μm particle size). For the thorough separation, a 90 min gradient of 5 to 35 % acetonitrile with 0.1 % formic acid was applied at a flow rate of 300 nL/min. The samples were sprayed (3.1 kV capillary voltage) to the quadrupole time-of-flight mass spectrometer Synapt G2-Si (Waters) with an ion mobility option. Spectra were recorded in a data-independent manner in high-definition MSE mode. Ions with 50 to 2000 m/z were detected in both channels, with a 1 s spectral acquisition scan rate. Peak detection and processing were executed using Progenesis QI 4.0 software [63] (Waters). For peak picking, the following thresholds were applied: low-energy 320 counts and high-energy 40 counts. Precursors and fragment ions were coupled using correlations of chromatographic elution profiles in low/high-energy traces. Then, peak retention times were aligned across all chromatograms. Peak intensities were normalized to the median distribution of all ions, assuming that most signals were unaffected by the experimental conditions. Protein identification was performed in Progenesis QI 4.0 [63] utilizing the Staphylococcus aureus Uniprot database [64].

Results and discussion

Chemistry and computational ADME properties

The synthesis of the target anilides (seeScheme 1) was performed by click chemistry realized in a microwave reactor using phosphorus chloride in dry chlorobenzene from starting materials. 1-Hydroxynaphthalene-2-carboxylic acid and a suitable aniline as described in Gonec et al. [29] for 1-4 or Gonec et al. [52] for 5-13.

Scheme 1. Synthesis of ring-substituted 1-hydroxynaphthalene-2-carboxanilides 1-13. Reagents and conditions: (a) PCl3, chlorobenzene, microwave synthesis (500 W, 130 °C, 15 min).
ADMET-13-2684-g001.jpg

All the compounds are mentioned inTable 1, where their physicochemical properties are also listed as key parameters for their drug-likeness [65]. Their lipophilicity was measured experimentally (expressed as the logarithm of the capacity factor k and the logarithms of the distribution coefficients DpH at pH 6.5 and 7.4), and log P was predicted using the commercially available ACD/Percepta ver. 2012 program [66]. Lipinski's rule of five (Ro5) parameters [67,68] were also calculated using this software, and information about compounds related to the Veber rule was also added to clarify whether the compounds should have good oral bioavailability [68,69]. In addition to the parameters characterizing ADME,Table 1 includes the electronic σ(Ar) parameters of the entire substituted anilide ring, characterizing the ability to withdraw or donate electrons to the molecular system. The values of σ(Ar) are found in a wide range from 0.60 (unsubstituted derivative 1) to 1.56 (compound 11, R = 2,4,5-Cl).

Table 1. Structure of 1-hydroxynaphthalene-1-carboxanilides 1-13; experimentally determined log k, log D6.5, log D7.4 values and predicted lipophilicity (log P) values, their molecular weight (MW), number of H-bond donors (HBD), number of H-bond acceptors (HBA), number of rotatable bonds (RB), topological polar surface area (TPSA), and electronic σ(Ar) parameters of anilide ring of investigated compounds
ADMET-13-2684-t001.jpg
Comp.Rlog klog D6.5log D7.4log PaMWaHBDaHBAaRBaTPSAa, nm2σ(Ar)a
1H0.60840.51390.54874.52263.292320.490.60
22-Cl0.69000.58280.58365.02297.742320.491.05
33-Cl0.85560.73620.72275.25297.742320.490.85
44-Cl0.88800.76400.75015.24297.742320.490.75
52,3-Cl0.68380.43710.38705.76332.182320.491.22
62,4-Cl0.73870.51680.46805.78332.182320.491.12
72,5-Cl0.67170.39490.39385.82332.182320.491.22
82,6-Cl0.57290.45160.45945.52332.182320.491.33
93,4-Cl0.93040.90480.89095.99332.182320.491.19
103,5-Cl0.97040.95310.93066.01332.182320.491.11
112,4,5-Cl0.68940.53140.48646.31366.622320.491.56
122,4,6-Cl0.83980.72540.68256.15366.622320.491.48
133,4,5-Cl1.23871.09601.05616.28366.622320.491.46
Ro5---<5<500<5<10---
Veber rule-------<10<1.40-

[i] aACD/Percepta ver. 2012 (Advanced Chemistry Development. Inc., Toronto, ON, Canada, 2012) [66]; Ro5 = Lipinski’s rule of five.

Although this is a small series of compounds, it is a compact and coherent series, and interesting observations can be made due to the variability of the substitution positions in the anilide part of the molecule. The graphs inFigure 1 show the correlation between the experimental and calculated lipophilicity values. As can be seen from the values of the correlation coefficients r, which are in the interval 0.26-0.46 (n = 13), these are very poor agreements.

Figure 1. Comparison of experimentally determined values of log k (A), log D6.5 (B), and log D7.4 (C) with calculated log P (ACD/Percepta [66]) of carboxanilides 1-13
ADMET-13-2684-g002.jpg

The highest correlation was achieved for the comparison of log k versus log P, i.e. for measurements in water. At physiological pH 7.4 (i.e. slightly basic), the correlation is extremely low, indicating the probable role of the free phenol group in position 1 of the naphthalene scaffold and strong inter- and intramolecular interactions in the aqueous environment, which are imperceptible using the software but are essential for the behavior of this type of compounds in a biological system. Comparison of experimental log k versus log D values (Figure 2) yielded good correlations (r ranging from 0.91 to 0.99, n = 13).

Figure 2. Cross-correlations of experimentally determined values of log k versus log D6.5 (A), log k versus log D7.4 (B) and log D6.5 versus log D7.4 (C) of carboxanilides 1-13
ADMET-13-2684-g003.jpg

Figure 3 shows the order of compounds according to increasing log k values. The height of the bars indicates that log k values are the highest compared to log D values, with the lowest lipophilicity values being achieved when measured at pH 7.4.

Figure 3. Order of individual derivatives arranged according to increasing log k values
ADMET-13-2684-g004.jpg

However, measurements in all three types of media confirm that, surprisingly, the unsubstituted derivative 1 is not the least lipophilic compound of the discussed series, but N-(2,6-dichlorophenyl)-1-hydroxynaphthalene-2-carboxamide (8). The most lipophilic is N-(3,4,5-trichlorophenyl)-1-hydroxynaphthalene-2--carboxamide (13), with a large jump from all other derivatives. It is clear from the graph that substitution in ortho positions significantly reduces lipophilicity, while substitution mainly in meta positions increases it.

Regarding the calculated ADME parameters listed inTable 1, Lipinski's recommendation of molecular weight <500 is met by all compounds. On the contrary, their lipophilicity, which is higher than the recommended log P <5 for all molecules, is contradictory. All other parameters from Ro5 and the Veber rule are met by the discussed molecules. Therefore, it is possible to assume that the compounds should not have serious absorption problems after oral administration. Much more informative results could be obtained using in vitro or in vivo experiments, which are more time-consuming and expensive [70].

In vitro antistaphylococcal activities

All the investigated compounds were screened for their effect against S. aureus, as an important cause of various bacterial infections [51]. S. aureus ATCC 29213 was used as the reference and quality control strain, and in addition, clinical isolates of methicillin-resistant S. aureus (MRSA) 63718, SA 630, and SA 3202 [54] were tested. These three clinical MRSA isolates of human and veterinary origin carry the mecA gene, which causes their resistance [54-57]. Activities are expressed as the minimum inhibitory concentrations (MICs), seeTable 2.

Table 2. Antistaphylococcal activity (MIC) of carboxanilides 1-13 compared to ampicillin (APC) and ciprofloxacin (CPX), and cytotoxicity (LC50) in the human monocytic leukemia cell line (THP-1) compared to oxaliplatin (OXP) and camptothecin (CMT)
Comp.RMIC, μMLC50, μM THP-1
S. aureusMRSA 63718MRSA SA 630MRSA SA 3202
1H>972>972243121>20 [29]
22-Cl>859>859>859>859-
33-Cl>859>859429107>20 [29]
44-Cl>859>859429107>20 [29]
52,3-Cl770770770770-
62,4-Cl770770770770-
72,5-Cl770770770770-
82,6-Cl48.248.248.248.2>30
93,4-Cl77077096.3385-
103,5-Cl0.370.370.370.37>30
112,4,5-Cl698698698698-
122,4,6-Cl698698698698-
133,4,5-Cl698698698698-
APC-5.7245.845.845.80.8±0.1
CPX-0.7524.224.224.20.7±0.09
OXP-----1.7±0.6
CMP-----0.16±0.07

Only N-(3,5-dichlorophenyl)-1-hydroxynaphthalene-2-carboxamide (10) showed significant antistaphylococcal activity against the susceptible strain and resistant isolates. Partial activity was also observed for carboxamide 8. The other tested compounds were completely inactive. Therefore, no structure-activity relationships can be discussed from this set; it can only be stated that the introduction of the second chlorine substituent improves the biological activity compared to the unsubstituted derivative 1 and the monosubstituted compounds 2-4. On the contrary, the third chlorine atom (derivatives 11-13) again leads to a loss of activity.

Given that both active compounds have diametrically different lipophilicities (log k 0.9704 vs. 0.5729) and also distant electron-withdrawing values (σ(Ar) 1.11 vs. 1.33), it can be summarized that only the position of both chlorine substituents significantly affects biological activity, as discussed, e.g. in [45,47,71-73].

However, it should be noted that the antistaphylococcal activities of both compounds against all the tested strains are similar, so a specific activity against Staphylococcus spp. can be speculated. A similar type of compounds - 3-hydroxynaphthalene-2-carboxanilides - was evaluated for their combined effect with ciprofloxacin and oxacillin and synergism was found in combinations with ciprofloxacin and indifference in combinations with oxacillin. From these facts, it can be concluded that the compounds have their specific mechanism of action against S. aureus/MRSA without any interference with the mecA gene and without any interaction with the cell wall. On the other hand, due to the synergism with ciprofloxacin, a possible inhibition of efflux pumps could be hypothesized [74].

MTT assay

To contribute to the discovery of one of the possible mechanisms of action, a standard MTT assay was performed with active compound 10 in comparison with the drugs ampicillin and ciprofloxacin used as standards. The lowest multiples of MIC values that achieved inhibition of S. aureus ATCC 29213 viability greater than 70 % are shown inTable 3. The MTT assay can be used to assess cell growth by measuring respiration. The MTT-measured viability of bacterial cells is less than 70 % after exposure to the MIC values for each tested compound, which is considered a positive result of this assay. This low level of cell viability indicates inhibition of cell growth by inhibition of respiration [31,60,61]. Although the tested compound showed 96 % inhibition of respiration, this effect occurred only at 16× MIC, i.e. at a high concentration compared to the inhibitory effect on growth. Therefore, it can be assumed that the main mechanism of action is not inhibition of the respiratory chain.

Table 3. Lowest MIC values with at least 70 % inhibition of S. aureus ATCC 29213 viability (respiratory activity)
CompoundsConcentrationS. aureus viability inhibition, %
1016× MIC96.5
APC8× MIC90.0
CPX32× MIC92.8

Cytotoxicity

Preliminary in vitro cytotoxicity screening of selected compounds 8 and 10 was performed using the human monocytic leukemia cell line (THP-1). Cytotoxicity was expressed as LC50 value (lethal concentration for 50 % of the cell population), seeTable 2. Treatment with 30 μM did not lead to a significant lethal effect on THP-1 cells (e.g. the LC50 values of oxaliplatin and camptothecin were 1.7±0.6 and 0.16±0.07 μM, respectively). For comparison, the LC50 values of ampicillin and ciprofloxacin were 0.8±0.1 and 0.7±0.1 μM, respectively. Based on these observations, it can be concluded that both tested agents can be considered non-toxic compounds.

Previously, compounds 1, 3 and 4 were also tested for their potential cytotoxic activity in THP-1 cells. None of them induced significant lethal effects by any tested concentrations up to 20 μM [29]. Thus, compounds 1, 3 and 4 could also be considered non-toxic substances.

Chemoproteomic analysis

In order to gain a deeper understanding of what happens in the staphylococcal cell after exposure to these chlorinated derivatives, a primary chemoproteomic study was performed using the ABPP approach with LC-MS/MS detection. The discussed dichlorinated derivatives were tested against S. aureus ATCC 29213 strain and MRSA SA 630 isolate: compound 10 with high antibacterial activity and compound 9, a molecule with no antibacterial effect for comparison, as these xenobiotics cause cell stress but do not lead to bacterial cell death. In addition, the experiment was performed by culturing S. aureus/MRSA without the addition of the tested molecules (control sample, CN). In this way, the following information was obtained: (i) about the representation of proteins in native bacteria (CN); (ii) the change in the protein profile after exposure to an inactive substance (CN vs. 9); and (iii) changes in the protein profile induced by the active agent (CN vs. 9 vs. 10). This allows us to filter out misleading information that does not lead to a substantial effect on the viability of the studied staphylococci.

The results of the proteomic analysis are interpreted graphically as heatmaps and volcano plots, where differences in protein levels between samples are clearly visible. Both types of plots are created using the web platform MetaboAnalyst 6.0 [75], and brief information about the function of proteins and their assignment to specific biochemical pathways was obtained from the freely available Uniprot database [64]. From each protein set of approximately 100 proteins, the 50 proteins exhibiting the most significant changes were selected for the heatmaps. Each colored cell on the map corresponds to the value of the protein peak area in the data table, with samples in rows and proteins in columns.

The heatmap inFigure 4A shows the differences in protein expression between the control S. aureus sample (i.e. without treatment with any compound, CN) and S. aureus treated with antistaphylococcal inactive compound 9 and active agent 10. It is clear from the heatmap (Figure 4A) that inactive 9 caused changes in the proteome more significantly compared to active 10. The active agent specifically caused downregulation of the following proteins (their function is listed inTable 4): Probable tautomerase SAV1363 (P67526), ribonuclease J2 (Q5HPR6), ATP-dependent protease ATPase subunit HslU (Q5HPT8), adenylosuccinate synthetase (P65884). Arsenate reductase (P0A005) was reduced by both derivatives to a similar level. The heatmap inFigure 4B comparing the MRSA control sample with samples 9 and 10 shows smaller protein sets compared toFigure 4A. Compared to the control sample, agent 10 caused downregulation of only one protein – Uncharacterized oxidoreductase SAUSA300_2422 (Q2FE2), which was also more significantly downregulated by inactive compound 9. In addition, compound 9 caused a decrease in the expression of four other proteins (bottom of the heatmap): Large ribosomal subunit protein uL2 (A5IV31), Uncharacterized lipoprotein SAS2259 (Q6G6V2), Dihydroneopterin aldolase (Q5HRN9), and aminoacyltransferase FemB (Q5HPG5).

Figure 4. S. aureus heatmap comparing proteomic profiles in control sample (CN), sample with antibacterially inactive compound 9 and sample with antibacterially active agent 10 (A), and MRSA heatmap comparing protein expression in control sample (CN), sample with inactive compound 9 and sample with active agent 10 (B)
ADMET-13-2684-g005.jpg
Table 4. Downregulated proteins detected after treatment with active agent 10 and their functions in bacteria.
ProteinFunction
Probable tautomerase SAV1363 (P67526)Protein involved in processes mediating tautomerization, microbial metabolism in various environments, and degradation/detoxification of aromatic compounds (xenobiotics) [64,76].
Ribonuclease J2 (Q5HPR6)Metalloprotein (cofactor two Zn2+ ions) exhibiting 5'-3' exonuclease and endonuclease activity, respectively, which plays a role in rRNA maturation. It is also involved in mRNA synthesis and/or degradation. It affects bacterial viability through the regulation of gene expression at the RNA level [64].
ATP-dependent protease ATPase subunit HslU (Q5HPT8)ATPase subunit of the proteasome-like degradation complex with chaperone activity. It is involved in the vital function of ridding the cell of damaged, misfolded or abnormal proteins (e.g. due to stress induced by antibiotic treatment) as well as controlling the levels of regulatory proteins [64,77].
Adenylosuccinate synthetase (P65884)Key role in the de novo biosynthetic pathway of purine nucleotides essential for bacteria (basic cellular processes, DNA/RNA synthesis and energy metabolism). It catalyzes the first key step in the synthesis of adenosine monophosphate (AMP) from inosine monophosphate (IMP). It requires the presence of Mg2+ ions for its function [64].

The volcano plot of expression changes between samples shows several of the most significant differences between the control and samples 9 and 10. Inactive compound 9 caused downregulation of significantly more proteins (Figure 5A) compared to active agent 10 (Figure 5C). When comparing the scales on the x-axis, it is clear that the proteins inFigure 5A show several times greater changes in expression or inhibition, respectively, than those inFigure 5C. Most of the proteins that were affected by compound 9 are located in the region of the graph, showing high statistical significance, while the proteins affected by the effect of compound 10 are seen in the bottom parts of the graph. It is important to note that when compared to the heatmap (Figure 4A) showing the same compounds, four significant proteins were identified. The volcano plot shows that of these proteins, only the ATP-dependent protease ATPase subunit HslU (Q5HPT8) shows the most significant change. The others were downregulated but not significantly compared to the native state of the proteins in the control sample of S. aureus. The MRSA samples shown inFigure 5B (control vs. 9) andFigure 5D (control vs. 10) show a smaller difference in the extent of changes compared to the volcano plots inFigures 5A and 5C. At the same time, the MRSA proteome was significantly more affected by compound 9 than by agent 10. From the heatmap (Figure 4B) correlated with the given compounds, only the protein uncharacterized oxidoreductase SAUSA300_2422 (Q2FE2) was identified as downregulated, but its change was assessed as insufficiently significant (Figure 5D).

Figure 5. Volcano plot of fold changes (FC) in protein expression in S. aureus between control sample and compound 9 (A), between control and agent 10 (C), changes in expression in MRSA between control and compound 9 (B), between control and agent 10 (D)
ADMET-13-2684-g006.jpg

The proteins that were found to be downregulated after exposure to active agent 10 and their significance/function for bacterial viability are listed inTable 4.

If certain proteins are downregulated in bacteria even after treatment with an antibacterially inactive compound (high MIC value), as observed with compound 9 (compare 770 μM vs. 0.37 μM for agent 10), this suggests that decreased expression of these proteins does not significantly impact bacterial growth. While treatment with antibacterially inactive compounds induces stress [78-80], it also triggers the expression (upregulation) of alternative proteins that help maintain bacterial viability. Consequently, inactive compound 9 had a much greater impact on the bacterial proteome than active agent 10. In contrast, the active agent 10 induces fewer proteomic changes (downregulation) but selectively eliminates proteins essential for bacterial survival.

Conclusions

Twelve chlorinated 1-hydroxynaphthalene-2-carboxanilides were synthesized by microwave synthesis and tested for their antistaphylococcal activity against reference S. aureus ATCC 29213 and three clinical MRSA isolates carrying the mecA gene. Only one compound, N-(3,5-dichlorophenyl)-1-hydroxynaphthalene-2-carboxamide (10), was significantly active against all Staphylococcus sp. The MICs were the same against all strains (0.37 μM). From the set of tested mono-, di- and trisubstituted derivatives, it is evident that the position of the chlorine atoms is crucial for antistaphylococcal activity. This compound showed no lethal activity (LC50) against the human monocytic leukemia cell line (THP-1) up to a concentration of 30 μM. Compound 10 showed 96 % inhibition of S. aureus respiration at a concentration of 16× MIC. Thus, it has a significant effect, but at high concentrations, suggesting that interference with energy metabolism is not the primary mechanism of action. Proteomic analysis performed with antistaphylococcal effective agent 10 and inactive isomer 9 revealed that inactive derivative 9 alters the proteome of staphylococci more significantly than active compound 10. The effect of agent 10 on S. aureus resulted in the downregulation of four proteins: probable tautomerase SAV1363, ribonuclease J2, adenylosuccinate synthetase, and especially ATP-dependent protease ATPase subunit HslU, involved in essential processes for bacterial survival and growth, such as syntheses of proteins, nucleotides/nucleic acids, and efficient protein repair/degradation. It can be assumed that this downregulation probably contributes to the antibacterial effect. The effect of compound 10 on the proteome in a clinical MRSA isolate remains unclear despite several different experiments/measurements and will require further investigation.

Acknowledgements

This work was supported by the Slovak Research and Development Agency (projects APVV-22-0133, APVV-21-0321) and by International Centre for Genetic Engineering and Biotechnology (project CRP/24/017). Special thanks to Milos Makovsky (Department of Art and Education, Faculty of Education, J. E. Purkyne University in Usti nad Labem, Czech Republic), the author of the illustrations in the graphic abstract.

Notes

[2] Conflicts of interest Conflict of interest: The authors declare no conflicts of interest.

References

[1] 

WHO Bacterial Priority Pathogens List, 2024. https://www.who.int/news-room/fact-sheets/detail/antimicrobial-resistance (accessed on 10 February 2025).

[2] 

WHO Global Antimicrobial Resistance and Use Surveillance System (GLASS) Report: 2022. https://www.who.int/publications/i/item/9789240062702 (accessed on 10 February 2025).

[3] 

Assessing the health burden of infections with antibiotic-resistant bacteria in the EU/EEA, 2016-2020. European Centre for Disease Prevention and Control (ECDC): Stockholm, Sweden, 2022. https://www.ecdc.europa.eu/en/publications-data/health-burden-infections-antibiotic-resistant-bacteria-2016-2020 (accessed on 10 February 2025).

[4] 

Fongang H.; Mbaveng A.T.; Kuete V. Global burden of bacterial infections and drug resistance. in Advances in Botanical Research, Kuete V. (Ed.), Academic Press & Elsevier, Amsterdam, Netherlands, 2023, pp.1-20. https://doi.org/10.1016/bs.abr.2022.08.001 https://doi.org/10.1016/bs.abr.2022.08.001

[5] 

National Academies of Sciences, Engineering, and Medicine; Health and Medicine Division; Board on Population Health and Public Health Practice; Committee on the Long-Term Health and Economic Effects of Antimicrobial Resistance in the United States. in Combating Antimicrobial Resistance and Protecting the Miracle of Modern Medicine, Palmer G.H.; Buckley G.J. (Eds.), National Academies Press, Washington D.C., USA, 2022. https://nap.nationalacademies.org/catalog/26350/combating-antimicrobial-resistance-and-protecting-the-miracle-of-modern-medicine (accessed on 10 February 2025).

[6] 

GBD 2021 Antimicrobial Resistance Collaborators. Global burden of bacterial antimicrobial resistance 1990-2021: A systematic analysis with forecasts to 2050. Lancet 404 (2024) 1199-1226. https://doi.org/10.1016/s0140-6736(24)01867-1 https://doi.org/10.1016/s0140-6736(24)01867-1

[7] 

GBD 2019 Antimicrobial Resistance Collaborators. Global mortality associated with 33 bacterial pathogens in 2019: A systematic analysis for the Global Burden of Disease Study 2019. Lancet 400 (2022) 2221-2248. https://doi.org/10.1016/S0140-6736(22)02185-7 https://doi.org/10.1016/S0140-6736(22)02185-7

[8] 

Jampilek J. Design and discovery of new antibacterial agents: Advances, perspectives, challenges. Current Medicinal Chemistry 25 (2018) 4972-5006. https://doi.org/10.2174/0929867324666170918122633 https://doi.org/10.2174/0929867324666170918122633

[9] 

Miethke M.; Pieroni M.; Weber T.; Brönstrup M.; Hammann P.; Halby L.; Arimondo P.B.; Glaser P.; Aigle B.; Bode H.B.; Moreira R.; Li Y.; Luzhetskyy A.; Medema M.H.; Pernodet J.L.; Stadler M.; Tormo J.R.; Genilloud O.; Truman A.W.; Weissman K.J.; Takano E.; Sabatini S.; Stegmann E.; Brötz-Oesterhelt H.; Wohlleben W.; Seemann M.; Empting M.; Hirsch A.K.H.; Loretz B.; Lehr C.M.; Titz A.; Herrmann J.; Jaeger T.; Alt S.; Hesterkamp T.; Winterhalter M.; Schiefer A.; Pfarr K.; Hoerauf A.; Graz H.; Graz M.; Lindvall M.; Ramurthy S.; Karlén A.; van Dongen M.; Petkovic H.; Keller A.; Peyrane F.; Donadio S.; Fraisse L.; Piddock L.J.V.; Gilbert I.H.; Moser H.E.; Müller R. Towards the sustainable discovery and development of new antibiotics. Nature Reviews Chemistry 5 (2021) 726-749. https://doi.org/10.1038/s41570-021-00313-1 https://doi.org/10.1038/s41570-021-00313-1

[10] 

Ayon N.J. High-throughput screening of natural product and synthetic molecule libraries for antibacterial drug discovery. Metabolites 13 (2023) 625. https://doi.org/10.3390/metabo13050625 https://doi.org/10.3390/metabo13050625

[11] 

Boyd N.K.; Teng C.; Frei C.R. Brief overview of approaches and challenges in new antibiotic development: A focus on drug repurposing. Frontiers in Cellular and Infection Microbiology 11 (2021) 684515. https://doi.org/10.3389/fcimb.2021.684515 https://doi.org/10.3389/fcimb.2021.684515

[12] 

Mondal S.K.; Chakraborty S.; Manna S.; Mandal S.M. Antimicrobial nanoparticles: Current landscape and future challenges. RSC Pharmaceutics 1 (2024) 388-402. https://doi.org/10.1039/D4PM00032C https://doi.org/10.1039/D4PM00032C

[13] 

Gray D.A.; Wenzel M. Multitarget approaches against multiresistant superbugs. ACS Infectious Diseases 6 (2020) 1346-1365. https://doi.org/10.1021/acsinfecdis.0c00001 https://doi.org/10.1021/acsinfecdis.0c00001

[14] 

Suckling C.J.; Hunter I.S.; Scott F.J. Multitargeted anti-infective drugs: Resilience to resistance in the antimicrobial resistance era. Future Drug Discovery 4 (2022) FDD73. https://doi.org/10.4155/fdd-2022-0001 https://doi.org/10.4155/fdd-2022-0001

[15] 

Feng J.; Zheng Y.; Ma W.; Ihsan A.; Hao H.; Cheng G.; Wang X. Multitarget antibacterial drugs: An effective strategy to combat bacterial resistance. Pharmacology & Therapeutics 252 (2023) 108550. https://doi.org/10.1016/j.pharmthera.2023.108550 https://doi.org/10.1016/j.pharmthera.2023.108550

[16] 

Bremner J.B. An update review of approaches to multiple action-based antibacterials. Antibiotics 12 (2023) 865. https://doi.org/10.3390/antibiotics12050865 https://doi.org/10.3390/antibiotics12050865

[17] 

Stelitano G.; Sammartino J.C.; Chiarelli L.R. Multitargeting compounds: A promising strategy to overcome multi-drug resistant tuberculosis. Molecules 25 (2020) 1239. https://doi.org/10.3390/molecules25051239 https://doi.org/10.3390/molecules25051239

[18] 

Lagadinou M.; Onisor M.O.; Rigas A.; Musetescu D.V.; Gkentzi D.; Assimakopoulos S.F.; Panos G.; Marangos M. Antimicrobial properties on non-antibiotic drugs in the era of increased bacterial resistance. Antibiotics 9 (2020) 107. https://doi.org/10.3390/antibiotics9030107 https://doi.org/10.3390/antibiotics9030107

[19] 

Imramovsky A.; Pesko M.; Kralova K.; Vejsova M.; Stolarikova J.; Vinsova J.; Jampilek J. Investigating spectrum of biological activity of 4- and 5-chloro-2-hydroxy-N-[2-(arylamino)-1-alkyl-2-oxoethyl]benzamides. Molecules 16 (2011) 2414-2430. https://doi.org/10.3390/molecules16032414 https://doi.org/10.3390/molecules16032414

[20] 

Kushkevych I.; Kollar P.; Ferreira A.L.; Palma D.; Duarte A.; Lopes M.M.; Bartos M.; Pauk K.; Imramovsky A.; Jampilek J. Antimicrobial effect of salicylamide derivatives against intestinal sulfate-reducing bacteria. Journal of Applied Biomedicine 14 (2016) 125-130. https://doi.org/10.1016/j.jab.2016.01.005 https://doi.org/10.1016/j.jab.2016.01.005

[21] 

Borbala-Horvath L.; Kratky M.; Pflegr V.; Mahes E.; Gyulai G.; Kohut G.; Babiczky A.; Biri-Kovacs B.; Baranyai Z.; Vinsova J.; Bosze S. Host cell targeting of novel antimycobacterial 4-aminosalicylic acid derivatives with tuftsin carrier peptides. European Journal of Pharmaceutics and Biopharmaceutics 174 (2022) 111-130. https://doi.org/10.1016/j.ejpb.2022.03.009 https://doi.org/10.1016/j.ejpb.2022.03.009

[22] 

Paraskevopoulos G.; Monteiro S.; Vosatka R.; Kratky M.; Navratilova L.; Trejtnar F.; Stolarikova J.; Vinsova J. Novel salicylanilides from 4,5-dihalogenated salicylic acids: Synthesis, antimicrobial activity and cytotoxicity. Bioorganic and Medicinal Chemistry 25 (2017) 1524-1532. https://doi.org/10.1016/j.bmc.2017.01.016 https://doi.org/10.1016/j.bmc.2017.01.016

[23] 

Alhashimi M.; Mayhoub A.; Seleem M.N. Repurposing salicylamide for combating multidrug-resistant Neisseria gonorrhoeae. Antimicrobial Agents and Chemotherapy 63 (2019) e01225-19. https://doi.org/10.1128/aac.01225-19 https://doi.org/10.1128/aac.01225-19

[24] 

Almolhim H.; Elhassanny A.E.M.; Abutaleb N.S.; Abdelsattar A.S.; Seleem M.N.; Carlier P.R. Substituted salicylic acid analogs offer improved potency against multidrug-resistant Neisseria gonorrhoeae and good selectivity against commensal vaginal bacteria. Scientific Reports 13 (2023) 14468. https://doi.org/10.1038/s41598-023-41442-5 https://doi.org/10.1038/s41598-023-41442-5

[25] 

Yokoyama T.; Mizuguchi M.; Nabeshima Y.; Nakagawa Y.; Okada T.; Toyooka N.; Kusaka K. Rafoxanide, a salicylanilide anthelmintic, interacts with human plasma protein transthyretin. The FEBS Journal 290 (2023) 5158-5170. https://doi.org/10.1111/febs.16915 https://doi.org/10.1111/febs.16915

[26] 

Kauerova T.; Perez-Perez M.J.; Kollar P. Salicylanilides and their anti-cancer properties. International Journal of Molecular Sciences 24 (2023) 1728. https://doi.org/10.3390/ijms24021728 https://doi.org/10.3390/ijms24021728

[27] 

Otevrel J.; Mandelova Z.; Pesko M.; Guo J.; Kralova K.; Sersen F.; Vejsova M.; Kalinowski D.S.; Kovacevic Z.; Coffey A.; Csollei J.; Richardson D.R.; Jampilek J. Investigating the spectrum of biological activity of ring-substituted salicylanilides and carbamoylphenylcarbamates. Molecules 15 (2010) 8122-8142. https://doi.org/10.3390/molecules15118122 https://doi.org/10.3390/molecules15118122

[28] 

Imramovsky A.; Pesko M.; Ferriz J.M.; Kralova K.; Vinsova J.; Jampilek J. Photosynthesis-Inhibiting efficiency of 4-chloro-2-(chlorophenylcarbamoyl)phenyl alkylcarbamates. Bioorganic & Medicinal Chemistry Letters 21 (2011) 4564-4567. https://doi.org/10.1016/j.bmcl.2011.05.118 https://doi.org/10.1016/j.bmcl.2011.05.118

[29] 

Gonec T.; Kos J.; Zadrazilova I.; Pesko M.; Keltosova S.; Tengler J.; Bobal P.; Kollar P.; Cizek A.; Kralova K.; Jampilek J. Antimycobacterial and herbicidal activity of ring-substituted 1-hydroxynaphthalene-2-carboxanilides. Bioorganic and Medicinal Chemistry 21 (2013) 6531-6541. https://doi.org/10.1016/j.bmc.2013.08.030 https://doi.org/10.1016/j.bmc.2013.08.030

[30] 

Gonec T.; Pospisilova S.; Kauerova T.; Kos J.; Dohanosova J.; Oravec M.; Kollar P.; Coffey A.; Liptaj T.; Cizek A.; Jampilek J. N-Alkoxyphenylhydroxynaphthalenecarboxamides and their antimycobacterial activity. Molecules 21 (2016) 1068. https://doi.org/10.3390/molecules21081068 https://doi.org/10.3390/molecules21081068

[31] 

Michnova H.; Pospisilova S.; Gonec T.; Kapustikova I.; Kollar P.; Kozik V.; Musiol R.; Jendrzejewska I.; Vanco J.; Travnicek Z.; Cizek A.; Bak A.; Jampilek J. Bioactivity of methoxylated and methylated 1-hydroxynaphthalene-2-carboxanilides: comparative molecular surface analysis. Molecules 24 (2019) 2991. https://doi.org/10.3390/molecules24162991 https://doi.org/10.3390/molecules24162991

[32] 

Spaczynska E.; Mrozek-Wilczkiewicz A.; Malarz K.; Kos J.; Gonec T.; Oravec M.; Gawecki R.; Bak A.; Dohanosova J.; Kapustikova I.; Liptaj T.; Jampilek J.; Musiol R. Design and synthesis of anti-cancer 1-hydroxynaphthalene-2-carboxanilides with a p53 independent mechanism of action. Scientific Reports 9 (2019) 6387. https://doi.org/10.1038/s41598-019-42595-y https://doi.org/10.1038/s41598-019-42595-y

[33] 

Kauerova T.; Gonec T.; Jampilek J.; Hafner S.; Gaiser A.K.; Syrovets T.; Fedr R.; Soucek K.; Kollar P. Ring-substituted 1-hydroxynaphthalene-2-carboxanilides inhibit proliferation and trigger mitochondria-mediated apoptosis. International Journal of Molecular Sciences 21 (2020) 3416. https://doi.org/10.3390/ijms21103416 https://doi.org/10.3390/ijms21103416

[34] 

Terada H.; Goto S.; Yamamoto K.; Takeuchi I.; Hamada Y.; Miyake K. Structural requirements of salicylanilides for uncoupling activity in mitochondria: quantitative analysis of structure-uncoupling relationships. Biochimica et Biophysica Acta 936 (1988) 504-512. https://doi.org/10.1016/0005-2728(88)90027-8 https://doi.org/10.1016/0005-2728(88)90027-8

[35] 

Lee I.Y.; Gruber T.D.; Samuels A.; Yun M.; Nam B.; Kang M.; Crowley K.; Winterroth B.; Boshoff H.I.; Barry C.E. Structure-activity relationships of antitubercular salicylanilides consistent with disruption of the proton gradient via proton shuttling. Bioorganic and Medicinal Chemistry 21 (2013) 114-126. https://doi.org/10.1016/j.bmc.2012.10.056 https://doi.org/10.1016/j.bmc.2012.10.056

[36] 

Macielag M.J.; Demers J.P.; Fraga-Spano S.A.; Hlasta D.J.; Johnson S.G.; Kanojia R.M.; Russell R.K.; Sui Z.; Weidner-Wells M.A.; Werblood H.; Foleno B.D.; Goldschmidt R.M.; Loeloff M.J.; Webb G.C.; Barrett J.F. Substituted salicylanilides as inhibitors of two-component regulatory systems in bacteria. Journal of Medicinal Chemistry 41 (1998) 2939-2945. https://doi.org/10.1021/jm9803572 https://doi.org/10.1021/jm9803572

[37] 

Dasgupta N.; Kapur V.; Singh K.K.; Das T.K.; Sachdeva S.; Jyothisri K.; Tyagi J.S. Characterization of a two-component system, devR-devS, of Mycobacterium tuberculosis. Tubercle and Lung Disease 80 (2000) 141-159. https://doi.org/10.1054/tuld.2000.0240 https://doi.org/10.1054/tuld.2000.0240

[38] 

Moellering R.E.; Cravatt B.F. How chemoproteomics can enable drug discovery and development. Chemical Biology 19 (2012) 11-22. https://doi.org/10.1016/j.chembiol.2012.01.001 https://doi.org/10.1016/j.chembiol.2012.01.001

[39] 

Malarney K.P.; Chang P.V. Chemoproteomic approaches for unraveling prokaryotic biology. Israel Journal of Chemistry 63 (2023) e202200076. https://doi.org/10.1002/ijch.202200076 https://doi.org/10.1002/ijch.202200076

[40] 

Drewes G.; Knapp S. Chemoproteomics and chemical probes for target discovery. Trends in Biotechnology 36 (2018) 1275-1286. https://doi.org/10.1016/j.tibtech.2018.06.008 https://doi.org/10.1016/j.tibtech.2018.06.008

[41] 

Jones L.H.; Neubert H. Clinical chemoproteomics—Opportunities and obstacles. Science Translational Medicine 9 (2017) 7951. https://doi.org/10.1126/scitranslmed.aaf7951 https://doi.org/10.1126/scitranslmed.aaf7951

[42] 

Chen X.; Wong Y.K.; Wang J.; Zhang J.; Lee Y.; Shen H.; Lin Q.; Hua Z.C. Target identification with quantitative activity based protein profiling (ABPP). Proteomics 17 (2017) 1600212. https://doi.org/10.1002/pmic.201600212 https://doi.org/10.1002/pmic.201600212

[43] 

Wang S.; Tian Y.; Wang M.; Wang M.; Sun G.; Sun X. Advanced activity-based protein profiling application strategies for drug development. Frontiers in Pharmacology 9 (2018) 353. https://doi.org/10.3389/fphar.2018.00353 https://doi.org/10.3389/fphar.2018.00353

[44] 

Naumann K. Influence of chlorine substituents on biological activity of chemicals: A review. Pest Management Science 56 (2000) 3-21. https://doi.org/10.1002/(SICI)1526-4998(200001)56:1%3C3::AID-PS107%3E3.0.CO;2-P https://doi.org/10.1002/(SICI)1526-4998(200001)56:1%3C3::AID-PS107%3E3.0.CO;2-P

[45] 

Fang W.Y.; Ravindar L.; Rakesh K.P.; Manukumar H.M.; Shantharam C.S.; Alharbi N.S.; Qin H.L. Synthetic approaches and pharmaceutical applications of chloro-containing molecules for drug discovery: A critical review. European Journal of Medicinal Chemistry 173 (2019) 117-153. https://doi.org/10.1016/j.ejmech.2019.03.063 https://doi.org/10.1016/j.ejmech.2019.03.063

[46] 

Dolezal M.; Zitko J.; Osicka Z.; Kunes J.; Vejsova M.; Buchta V.; Dohnal J.; Jampilek J.; Kralova K. Synthesis, antimycobacterial, antifungal and photosynthesis-inhibiting activity of chlorinated N-phenylpyrazine-2-carboxamides. Molecules 15 (2010) 8567-8581. https://doi.org/10.3390/molecules15128567 https://doi.org/10.3390/molecules15128567

[47] 

Faleye O.S.; Boya B.R.; Lee J.H.; Choi I.; Lee J. Halogenated antimicrobial agents to combat drug-resistant pathogens. Pharmacological Reviews 76 (2023) 90-141. https://doi.org/10.1124/pharmrev.123.000863 https://doi.org/10.1124/pharmrev.123.000863

[48] 

Huber R.; Marcourt L.; Heritier M.; Luscher A.; Guebey L.; Schnee S.; Michellod E.; Guerrier S.; Wolfender J.L.; Scapozza L.; Kohler T.; Gindro K.; Queiroz E.F. Generation of potent antibacterial compounds through enzymatic and chemical modifications of the trans-δ-viniferin scaffold. Scientific Reports 13 (2023) 15986. https://doi.org/10.1038/s41598-023-43000-5 https://doi.org/10.1038/s41598-023-43000-5

[49] 

Krawczyk-Lebek A.; Zarowska B.; Janeczko T.; Kostrzewa-Suslow E. Antimicrobial activity of chalcones with a chlorine atom and their glycosides. International Journal of Molecular Sciences 25 (2024) 9718. https://doi.org/10.3390/ijms25179718 https://doi.org/10.3390/ijms25179718

[50] 

Perz M.; Szymanowska D.; Janeczko T.; Kostrzewa-Suslow E. Antimicrobial properties of flavonoid derivatives with bromine, chlorine, and nitro group obtained by chemical synthesis and biotransformation studies. International Journal of Molecular Sciences 25 (2024) 5540. https://doi.org/10.3390/ijms25105540 https://doi.org/10.3390/ijms25105540

[51] 

Taylor T.A.; Unakal C.G. Staphylococcus aureus Infection; StatPearls Publishing, Treasure Island, FL, USA, 2024. https://www.ncbi.nlm.nih.gov/books/NBK441868 (accessed on 10 February 2025).

[52] 

Gonec T.; Kos J.; Pesko M.; Dohanosova J.; Oravec M.; Liptaj T.; Kralova K.; Jampilek J. Halogenated 1-hydroxynaphthalene-2-carboxanilides affecting photosynthetic electron transport in photosystem II. Molecules 22 (2017) 1709. https://doi.org/10.3390/molecules22101709 https://doi.org/10.3390/molecules22101709

[53] 

EZChrom Elite software ver. 3.3.2. Agilent, Santa Clara, CA, USA. https://ezchrom-elite.software.informer.com/3.3/

[54] 

Zadrazilova I.; Pospisilova S.; Pauk K.; Imramovsky A.; Vinsova J.; Cizek A.; Jampilek J. In vitro bactericidal activity of 4- and 5-chloro-2-hydroxy-N-[1-oxo-1-(phenylamino)alkan-2-yl]benzamides against MRSA. BioMed Research International 2015 (2015) 349534. https://doi.org/10.1155/2015/349534 https://doi.org/10.1155/2015/349534

[55] 

Nubel U.; Dordel J.; Kurt K.; Strommenger B.; Westh H.; Shukla S.K.; Zemlickova H.; Leblois R.; Wirth T.; Jombart T.; Balloux F.; Witte W. A timescale for evolution, population expansion, and spatial spread of an emerging clone of methicillin-resistant Staphylococcus aureus. PLOS Pathogens 6 (2010) e1000855. https://doi.org/10.1371/journal.ppat.1000855 https://doi.org/10.1371/journal.ppat.1000855

[56] 

Bosgelmez-Tinaz G.; Ulusoy S.; Aridogan B.; Coskun-Ari F. Evaluation of different methods to detect oxacillin resistance in Staphylococcus aureus and their clinical laboratory utility. European Journal of Clinical Microbiology & Infectious Diseases 25 (2006) 410-412. https://doi.org/10.1007/s10096-006-0153-8 https://doi.org/10.1007/s10096-006-0153-8

[57] 

Martineau F.; Picard F.J.; Roy P.H.; Ouellette M.; Bergeron M.G. Species-specific and ubiquitous-DNA-based assays for rapid identification of Staphylococcus aureus. Journal of Clinical Microbiology 36 (1998) 618-623. https://doi.org/10.1128/jcm.36.3.618-623.1998 https://doi.org/10.1128/jcm.36.3.618-623.1998

[58] 

Weinstein M.P.; Patel J.B. Methods for dilution antimicrobial susceptibility tests for bacteria that grow aerobically: M07-A11, 11th edition, Committee for Clinical Laboratory Standards, Wayne, PA, 2018. https://clsi.org/media/1928/m07ed11_sample.pdf

[59] 

Schwalbe R.; Steele-Moore L.; Goodwin A.C. Antimicrobial Susceptibility Testing Protocols, CRC Press, Boca Raton, FL, USA, 2007. https://doi.org/10.1201/9781420014495 https://doi.org/10.1201/9781420014495

[60] 

Measuring Cell Viability/Cytotoxicity. Dojindo EU GmbH, Munich, Germany. https://www.dojindo.eu.com/Protocol/Dojindo-Cell-Proliferation-Protocol.pdf

[61] 

Grela E.; Kozłowska J.; Grabowiecka A. Current methodology of MTT assay in bacteria—A review. Acta Histochemica 120 (2018) 303-311. https://doi.org/10.1016/j.acthis.2018.03.007 https://doi.org/10.1016/j.acthis.2018.03.007

[62] 

GraphPad Prism 5.00 software. GraphPadSoftware, San Diego, CA, USA. http://www.graphpad.com

[64] 

UniProt: The Universal Protein Knowledgebase in 2023. https://www.uniprot.org/

[65] 

Wu K.; Kwon S.H.; Zhou X.; Fuller C.; Wang X.; Vadgama J.; Wu Y. Overcoming challenges in small-molecule drug bioavailability: A review of key factors and approaches. International Journal of Molecular Sciences 25 (2024) 13121. https://doi.org/10.3390/ijms252313121 https://doi.org/10.3390/ijms252313121

[66] 

ACD/Percepta ver. 2012. Advanced Chemistry Development, Inc., Toronto ON, Canada, 2012. https://www.acdlabs.com/products/percepta-platform/

[67] 

Lipinski C.A.; Lombardo F.; Dominy B.W.; Feeney P.J. Experimental and computational approaches to estimate solubility and permeability in drug discovery and development settings. Advanced Drug Delivery Reviews 46 (2001) 3-26. https://doi.org/10.1016/S0169-409X(00)00129-0 https://doi.org/10.1016/S0169-409X(00)00129-0

[68] 

Kerns E.H.; Di L. Drug-Like Properties: Concepts. Structure Design and Methods: From ADME to Toxicity Optimization; Academic Press, San Diego, CA, USA, 2008. ISBN 978-0-12-369520-8

[69] 

Veber D.F.; Johnson S.R.; Cheng H.Y.; Smith B.R.; Ward K.W.; Kopple K.D. Molecular properties that influence the oral bioavailability of drug candidates. Journal of Medicinal Chemistry 45 (2002) 2615-2623. https://doi.org/10.1021/jm020017n https://doi.org/10.1021/jm020017n

[70] 

Chung T.D.Y.; Terry D.B.; Smith L.H. In vitro and in vivo assessment of ADME and PK properties during lead selection and lead optimization - guidelines, benchmarks and rules of thumb. in The Assay Guidance Manual, Markossian S. (Ed), National Institutes of Health - National Center for Advancing Translational Sciences, Rockville, MD, USA, 2015. https://www.ncbi.nlm.nih.gov/books/NBK326710/ (accessed on 08 February 2025).

[71] 

Hansch C. Bioisosterism. Intra-Science Chemistry Reports 8 (1974) 17-25.

[72] 

Korona-Glowniak I.; Nitek W.; Tejchman W.; Zeslawska E. Influence of chlorine and methyl substituents and their position on the antimicrobial activities and crystal structures of 4-methyl-1,6-diphenylpyrimidine-2(1H)-selenone derivatives. Acta Crystallographica Section C: Structural Chemistry 77 (2021) 649-658. https://doi.org/10.1107/s205322962100975x https://doi.org/10.1107/s205322962100975x

[73] 

Janowska S.; Stefanska J.; Khylyuk D.; Wujec M. The importance of substituent position for antibacterial activity in the group of thiosemicarbazide derivatives. Molecules 29 (2024) 1333. https://doi.org/10.3390/molecules29061333 https://doi.org/10.3390/molecules29061333

[74] 

Bak A.; Kos J.; Michnova H.; Gonec T.; Pospisilova S.; Kozik V.; Cizek A.; Smolinski A.; Jampilek J. Consensus-based pharmacophore mapping for new set of N-(disubstituted-phenyl)-3-hydroxyl-naphthalene-2-carboxamides. International Journal of Molecular Sciences 21 (2020), 6583. https://doi.org/10.3390/ijms21186583 https://doi.org/10.3390/ijms21186583

[76] 

Staphylococcus aureus subsp. aureus Mu50 (MRSA/VISA): SAV1363. DBGET Search, Kyoto University Bioinformatics Center. https://www.genome.jp/entry/sav:SAV1363

[77] 

Couvreur B.; Wattiez R.; Bollen A.; Falmagne P.l.; Le Ray D.; Dujardin J.C. Eubacterial HslV and HslU subunits homologs in primordial eukaryotes. Molecular Biology and Evolution 19 (2002) 2110-2117. https://doi.org/10.1093/oxfordjournals.molbev.a004036 https://doi.org/10.1093/oxfordjournals.molbev.a004036

[78] 

Zhang F.; Cheng W. The mechanism of bacterial resistance and potential bacteriostatic strategies. Antibiotics 11 (2022) 1215. https://doi.org/10.3390/antibiotics11091215 https://doi.org/10.3390/antibiotics11091215

[79] 

Avci F.G. Unraveling bacterial stress responses: implications for next-generation antimicrobial solutions. World J Microbiol Biotechnol 40 (2024) 285. https://doi.org/10.1007/s11274-024-04090-z https://doi.org/10.1007/s11274-024-04090-z

[80] 

Zavizion B.; Zhao Z.; Nittayajarn A.; Rieder R.J. Rapid microbiological testing: monitoring the development of bacterial stress. PLoS One 5 (2010) e13374. https://doi.org/10.1371/journal.pone.0013374 https://doi.org/10.1371/journal.pone.0013374


This display is generated from NISO JATS XML with jats-html.xsl. The XSLT engine is libxslt.