Skip to main content
PLOS One logoLink to PLOS One
. 2025 Jun 3;20(6):e0318135. doi: 10.1371/journal.pone.0318135

Antibacterial, antibiofilm and anti-quorum sensing activities of 1,2,3,5-tetrazine derivatives linked to a benzothiazole moiety

Jean Paul Dzoyem 1,2,*, Joseph Tsemeugne 3, Boniface Pone Kamdem 4,5,*, Rostand Foyou Meupiap 3, Boris Arnaud Kuate 3, Pierre Mkounga 3, Fabrice Fekam Boyom 4,5, Lyndy Joy McGaw 2
Editor: M Alejandro Dinamarca6
PMCID: PMC12133013  PMID: 40460187

Abstract

A series of known tetrazine derivatives, containing benzothiazole scaffold, were prepared during the coupling reactions of selected diazotized 2-aminobenzo[d]thiazole derivatives with p-acetaminophen. The as-prepared compounds were characterized based on NMR and MS spectrometry. The antibacterial and anti-biofilm activities of the synthesized compounds were evaluated by microdilution method, whereas the anti-quorum sensing effect was carried out using assay for the inhibition of violacein formation. As a result, compounds 4a, 4b and 4c revealed minimum inhibitory concentrations and minimum bactericidal concentrations ranging from 8 to 128 µg/mL and from 32 to 256 µg/mL, respectively. Compounds 4a (52–86.5%), 4b (57.7–79.4%) and 4c (59.9–80.3%) prevented biofilm formation in all the four bacteria tested with percentages of inhibition more than 50%. The concentrations of 4a, 4b and 4c that inhibited 50% of violacein production were found to be 62.71, 28.56 and 107.45 µg/mL, respectively, thus attesting that these compounds possess anti-quorum sensing activity. Noteworthy, our previous investigation attested that these compounds are non-cytotoxic on the human mammalian cells Vero. This novel contribution demonstrates the antibacterial, antibiofilm and anti-quorum sensing activities of tetrazine-based benzothiazoles, which might be prospected as scaffolds for the discovery of efficient antibiotics with decreased risk of microbial drug resistance.

Introduction

Resistance to currently available antibiotics has become a grave menace to the treatment of infectious diseases [1,2]. According to the 2019’s World Health Organization (WHO) report, antibacterial resistance was accountable for 1.27 million deaths worldwide [3,4]. The main mechanisms of this notorious phenomenon include enzymatic alteration and efflux pump systemic elimination of antimicrobials, structural modification of bacterial target proteins, changes in bacterial membrane permeability, etc. In the meantime, many pathogenic bacteria can form a dense biofilm [5,6], thus making bacteria highly resistant to antibiotics [7]. In fact, the contribution of biofilms in antimicrobial resistance is highly complex and may significantly drive resistance [810]. By controlling the formation of biofilms and drug efflux pumps, the quorum sensing system plays a crucial role in developing bacterial drug-resistant pathways [11]. Growing evidence has demonstrated the link between quorum sensing and biofilm development [1214]. As a matter of fact, a quorum sensing is a density-dependent cell-signalling mechanism by which bacteria crosstalk to each other [14,15]. Because of its connection to bacterial pathogenicity, virulence and biofilm formation, quorum sensing has gained more research attention in the last decade [16,17]. Thus, targeting quorum sensing and biofilm formation would be a prominent approach to unravel antibiotic resistance in pathogenic bacteria [18,19].

Modern research has substantially identified antibacterial compounds with anti-quorum sensing and anti-biofilm properties [6,2024], even though almost no such chemotype is reported to have succeeded the last clinical trial stage in humans.

Thus, the development of effective chemotypes that inhibit bacterial growth while preventing biofilm formation and attenuating quorum sensing-dependent virulence factors is of paramount importance.

Benzothiazole is a heterocyclic and bicyclic pharmacophore that contains benzene fused with 1,3-thiazole skeleton [25,26]. A number of scientists have established the potential of benzothiazole and its derivatives as antimicrobial hit compounds [2729].

On the other hand, modern research on tetrazine derivatives in relation to their antibacterial activity has been reported by many researchers [30,31].

The incorporation of 1,2,3,5-tetrazine derivatives into the benzothiazole structure introduces new chemical functionalities that can potentially disrupt bacterial biofilms and interfere with quorum sensing mechanisms. This dual approach aims not only to inhibit bacterial growth but also to prevent biofilm formation and attenuate virulence factors, thereby offering a comprehensive strategy to combat bacterial infections.

In our previous research investigation, we demonstrated the cytotoxic effects of 1,2,3,5-tetrazine tethered benzothiazole derivatives against a number of cancer cells, including A549, Hela and MCF-7 cells. However; these compounds were non cytotoxic vis-à-vis the human mammalian cells Vero.

In our continuing effort to search for effective antibacterial hit compounds that might aid in drug discovery campaigns, this study sought to investigate the inhibitory effects of certain 1,2,3,5-tetrazine tethered benzothiazole derivatives on the growth of selected bacteria. Moreover, anti-biofilm and anti-quorum sensing activities of these compounds are also investigated.

Materials and methods

Chemistry

General.

The reagents of analytical grade were purchased from commercial sources and used without any further purification. 1HNMR spectra were measured with a 400 MHz spectrometer NMR Bruker Advance 400 at room temperature in DMSO-d6 with tetramethylsilane as the internal reference. 13C-NMR spectra were recorded in DMSO-d6 with a 100 MHz spectrometer NMR Bruker Advance 400. UV-visible absorption spectra were recorded on Beckman U-640 Spectrophotometer, using samples’ solutions of concentration 5 × 10−5 mol.L−1. Infrared spectra were taken in KBr on a Perkin Elmer FT-IR 2000 spectrophotometer. Masss spectra were measured with a Waters Xevo TQD tandem quadrupole mass spectrometry system running in MS scan mode, 1 minute of acquired spectra were combined and centroided. Melting points were obtained with a Buchii melting point apparatus and are uncorrected. The Thin Layer Chromatography (T.L.C.) was carried out on Eastman Chromatogram Silica Gel Sheets (13181; 6060) with fluorescent indicators. A mixture of hexane and ethyl acetate (4:6) was used as the eluent and iodine was used for the visualization of the chromatograms.

Preparation of diazonium salt solution.

As per a previously reported protocol [32], dried sodium nitrite (0.69 g, 10 mmol) was slowly added over a period of 30 minutes to concentrated sulphuric acid (10 mL) with occasional stirring. The solution was cooled to 0–5 °C. Compound 1 was dissolved in DMSO (10 mL) and cooled to 0–5 °C. The nitrosyl sulphuric acid solution was added to the solution of 1 and the temperature was maintained between 0–5 °C. The clear diazonium salt solution thus obtained consisting of the in situ-formed intermediate 2, was used immediately in the coupling reactions.

General procedure for the preparation of the coupling products (4).

Acetaminophen (3) (1.51 g, 10 mmol) or 2-amino-6-nitrobenzothiazole (1b) (1.952 g, 10 mmol) was dissolved in DMSO (10 mL) and then cooled in an ice-bath at 0–5 °C. A prepared diazonium solution of 2 was added drop wise over 1 hour, and then 15 mL of sodium acetate solution (10%) was added to the mixture. The pH of the mixtures was in the range 9–11. The solid precipitate was collected on a filter and crystallised from methanol to give the title compound.

N-(3-((5,6-dimethylbenzo[d]thiazol-2-yl)diazenyl)-4-hydroxyphenyl)acetamide (4a) .

Compound 4a was obtained in 58% yield as red powder; m.p. 118–120 °C; [Litt: 119–121 °C, [32]; 1H-NMR (DMSO-d6, 400MHz): δ 10.67 (s, 1H, O-H), 9.94 (s, 1H, N-H), 8.10 (d, 1H, J = 4.0 Hz, H-2’), 7.89 (s, 1H, H-4), 7.84 (s, 1H, H-7), 7.60 (dd, 1H, J = 4.0 and 8.0 Hz, H-6’), 7.06 (d, 1H, J = 8.0 Hz H-5’), 2.36 (s, 6H, 2CH3), 2.01 (s, 3H, COCH3); 13C-NMR (DMSO-d6,100 MHz): δ 174.6 (CO), 168.1 (C-2), 153.6 (C-3a), 131.3 (C-4a), 124.2 (C-4), 137.4 (C-5), 132.1 (C-6), 122.5 (C-7), 138.5 (C-1’), 108.1 (C-2’), 136.1 (C-3’), 151.1 (C-4’), 118.8 (C-5’), 128.1 (C-6’), 23.8 (CH3CO), 19.9 (CH3), 19.6 (CH3); UV-Vis λmax (DMSO) (Log ε): 274 (5.00), 327 (4.39), 364 (4.49), 452 (4.01) nm; IR (KBr) υmax: 3248 (O-H and N-H), 1659 (C = O), 1604−1557 (C = C), 1483−1450 (N = N), 1274 (C-S), 1239 (C-S), 861−510 (Ar def C = N str thiazole) cm-1. (ESI+ ) m/z (%) 394 (8), 389 (10), 375 (13), 372 (7), 316 (65), 304 (48), 283 (19), 202 (14), 192 (21), 150 (70); Anal. Calcd. for C17H22N4O5S: C, 59.98; H, 4.74; N, 16.46; S, 9.42. Found: C, 59.63; H, 4.80; N, 16.41; S, 9.40. Rf = 0.62.

4-((5-acetamido-2-hydroxyphenyl)diazenyl)-3-(2-mercapto-4,5-dimethylphenyl)-7,8-dimethylbenzo[4,5]thiazolo[2,3-d][1,2,3,5]tetrazine-3,5-diium sulfate (4a’).

Compound 4a’ was obtained in 34% yield as brown powder; m.p. 318–319 °C; [Litt. 318–320 °C [32]; 1H-NMR (DMSO-d6, 400 MHz): δ 11.35 (s, 1H, OH), 10.67 (s, 1H, NH), 8.37 (s, 1H, H-9”’), 8.13 (d, 1H, J = 2.8 Hz, H-6’), 7.93 (s, 1H, H-6”’), 7.77 (s, 1H, H-6”), 7.69 (s, 1H, H-3”), 7.63 (dd, 1H, J = 8.8 and 2.8 Hz, H-4’), 7.09 (d, 1H, J = 8.8 Hz, H-3’), 2.50, 2.39, 2.37, 1.23 (s, 12H, CH3), 2.04 (s, 3H, COCH3); 13C-NMR (DMSO-d6, 100 MHz): δ 198.6 (C-4), 174.5 (C = O), 168.1 (C-6), 153.5 (C-1”), 151.0 (C-2’), 138.4 (C-5”’), 137.4 (C-8”’ and C-5’), 136.0 (C-5”), 132.1 (C-7”’), 131.3 (C-4”’ and C-2”), 128.1 (C-1’), 124.2 (C-4”), 122.4 (C-3”), 122.1 (C-9”’), 121.6 (C-6”’), 118.8 (C-6”), 118.3 (C-4’), 108.0 (C-3’), 105.4 (C-6’), 23.8 (COCH3), 23.7, 19.86, 19.7, 19.6 (Ph-CH3); UV-Vis λmax (MeOH) (Log ε): 227 (4.06), 257 (4.12), 272 (4.26), 290 (4.09), 295 (4.08), 302 (4.12), 325 (4.19), 348 (4.18), 355 (4.19), 399 (4.23), 445 (4.25), 486 (4.22) nm; IR (KBr) νmax: 3887−3282 (O-H and N-H), 2920 (ArC-H), 2324 (S-H), 1664−1655 (C = O), 1533 (C = C), 1490−1449 (N = N), 1370 (δtetrazine ring), 1269 (C-S), 1240 (C-O), 889 (δtetrazine ring) cm-1; ms: (ESI+ ) m/z (%) 699 (8), 673 (9), 643 (11), 659 (75), 601 (10), 599 (58), 581 (22), 485 (41), 410 (34), 409 (74), 316 (100), 166 (47); Anal. Calcd. for C26H33N7O10S3: C, 44.63; H, 4.75; N, 14.01; S, 13.74. Found: C, 44.59; H, 4.80; N, 14.05; S, 13.71. Rf = 0.30.

N-4-Hydroxy-2,3-bis[3-(3-Nitro-benzenethiol-5)-yl-7-nitro-9-thia-1,2-diaza-3,4a-diazonia-fluorene-4)-yl-diazenyl]-5,6-bis[(6-Nitro-benzothiazol-2)-yl-diazenyl]-phenyl-acetamide disulfate (4b).

Compound 4b was obtained in 67% yield as brown powder; m.p. 169–172 °C; [Litt. 171–173 °C [32]]; 1H-NMR (DMSO-d6, 400 MHz): δ 8.65 (d, 1H, J = 2.4 Hz, H-8v), 8.58 (d, 1H, J = 2.8 Hz, H-4’), 8.43 (dd, 1H, J = 8.8 and 2.8 Hz, H-5’), 8.43 (dd, 1H, J = 9.2 and 2.4 Hz, H-6v), 8.38 (dd, 1H, J = 8.8 and 2.4 Hz, H-4iv), 8.38 (dd, 1H, J = 9.2 and 2.0 Hz, H-6vii), 8.31 (d, 1H, J = 9.2 Hz, H-5v), 8.20 (dd, 1H, J = 6.4 and 2.0 Hz, H-4viii), 8.19 (d, 1H, J = 2.4 Hz, H-2iv), 8.18 (d, 1H, J = 6.4 Hz, H-2viii), 8.10 (dd, 1H, J = 8.8 and 2.4 Hz, H-5”), 7.85 (d, 1H, J = 8.8 Hz, H-4”), 7.68 (d, 1H, J = 2.0 Hz, H-5viii), 7.42 (d, 1H, J = 9.2 Hz, H-8vii), 7.35 (d, 1H, J = 2.0 Hz, H-5vii), 7.28 (d, 1H, J = 8.8 Hz, H-7’), 7.13 (d, 1H, J = 2.4 Hz, H-7”), 7.05 (d, 1H, J = 8.8 Hz, H-5iv), 3.17 (s, 2H, SH), 2.05 (s, 3H, COCH3); 13C-NMR (DMSO-d6, 100 MHz): δ 180.4 (CO), 171.7 (C-4”’), 170.5 (C-4vi), 169.8 (C-2’), 168.3 (C-2”), 168.0 (C-4), 158.2 (C-3vii), 155.8 (C-3a’), 155.3 (C-3a”), 155.1 (C-3iv), 152.5 (C-7v), 145.3 (C-6iv), 143.7 (C-6vii), 143.2 (C-7viii), 142.4 (C-6’), 141.9 (C-6”), 140.7 (C-9a”’ and 9avi), 138.6 (C-1vii), 135.6 (C-7a”), 134.4 (C-7a’), 132.2 (C-5av and 5aviii), 131.7 (C-1iv), 131.4 (C-1), 129.9 (C-8aviii), 125.0 (C-8av), 124.5 (C-6viii), 124.4 (C-6v), 122.7 (C-5iv), 122.3 (C-5vii), 122.1 (C-5v), 121.9 (C-5viii), 121.8 (C-8viii), 121.8 (C-4iv), 121.5 (C-8v), 120.4 (C-4vii), 119.8 (C-4’), 119.1 (C-4”), 119.0 (C-2), 119.0 (C-2vii), 118.6 (C-6), 118.4 (C-5’), 118.2 (C-5”), 117.6 (C-2iv), 116.8 (C-7”), 116.7 (C-7’), 111.4 (C-5), 107.1 (C-3), 23.8 (COCH3); UV-Vis λmax (MeOH) (Log ε): 272 (4.67), 348 (4.87), 437 (4.25), 483 (4.25), 555 (3.75) nm; IR (KBr) νmax: 3285 (O-H and N-H), 3097 (ArC-H), 1654 (C = O), 1599 (C = N), 1512 (C = C), 1442 (N = N), 1269 (C-S), 1234 (C-O), 910−502 (Ar def C = N str thiazole) cm-1; ms: (ESI+) m/z (%) 1052 (4), 996 (3), 882 (3), 713 (3), 694 (3), 659 (3), 637 (7), 599 (13), 409 (16), 317 (26), 316 (81); Anal. Calcd. for C50H33N25O26S8: C, 36.26; H, 2.01; N, 21.14; S, 15.48. Found: C, 36.24; H, 1.98; N, 21.17; S, 15.43. Rf = 0.53.

3,11-dinitrobenzo[4,5]thiazolo[3,2-c]benzo[4,5]thiazolo[3,2-e][1,2,3,5]tetrazine-8,14-diium sulfate (4c).

Compound 4c was obtained in 41% yield as orange powder; m.p. 244–246 °C; [Litt. 245–247 °C [32]]; 1H-NMR (DMSO-d6, 600 MHz): δ 8.95 (d, 1H, J = 1.8 Hz, H-7), 8.69 (d, 1H, J = 2.4 Hz, H-7’), 8.29 (s, 2H, NH), 8.25 (dd, 1H, J= 2.8 and 8.8 Hz, H-5), 8.11 (dd, 1H, J = 2.4 and 8.8 Hz, H-5’), 7.90 (d, 1H, J = 8.8 Hz, H-4), 7.42 (d, 1H, J= 8.8 Hz, H-4’); 13C-NMR (DMSO-d6, 150 MHz): δ 153.2 (C-2), 155.0 (C-3a), 121.0 (C-4), 122.5 (C-5 and C-5’), 144.0 (C-6), 119.3 (C-7), 132.6 (C-7a), 172.3 (C-2’), 158.7 (C-3a’), 117.3 (C-4’), 141.2 (C-6’), 118.3 (C-7’), 131.9 (C-7a’); UV-Vis (MeOH) λmax (log ε): 260 (4.54), 285 (4.48), 352 (4.66), 393 (4.62), 421 (4.63), 450 (4.64), 472 (4.62); IR (KBr) υmax/cm-1: 3097 (CAr-H), 1556 (C = N), 1444 (N = N), 1336 (CAr-NO2), 1120 (C-S), 1514 (C = C); ms: (ESI+) m/z (%) 590 (5), 568 (3), 562 (10), 558 (15), 554 (10), 550 (17), 514 (35), 450 (76), 378 (10), 294 (13), 248 (34). Anal. Calcd. for: C14H18N6O14S3: C, 28.48; H, 3.07; N, 14.23; S, 16.29. Found: C, 28.50; H, 3.1; N, 14.25; S, 16.32. Rf = 0.45.

1,2-bis(6-nitrobenzothiazol-2-yl)diazene-1,2-diium sulfate (4c’).

Compound 4c’ was obtained in 28% yield as red powder; m.p. 243–245 °C; [Litt. 243–245 °C [32]; 1H-NMR (DMSO-d6, 600 MHz): δ 8.69 (d, 2H, J= 2.4 Hz, H-7 and H-7’), 8.24 (2H, s, NH), 8.10 (dd, 2H, J= 2.4 and 8.8 Hz, H-5 and H-5’), 7.42 (d, 1H, J= 8.8 Hz, H-4 and H-4’); 13C-NMR (DMSO-d6, 150 MHz): δ 172.3 (C-2 and C-2’), 159.1 (C-3a and C-3a’), 117.3 (C-4 and C-4’), 122.5 (C-5 and C-5’), 141.2 (C-6 and C-6’), 118.2 (C-7 and C-7’), 132.1 (C-7a and C-7a’). UV-Vis (MeOH) λmax (log ε): 269.7 (4.53), 279.6 (4.30), 351.3 (5.17); IR (KBr) υmax/cm-1: 3508 (N-H), 3068 (CAr-H), 1644 (C = N), 1568 (C = C), 1486 (N = N), 1282 (CAr-NO2), 1120 (C-S); ms: (ESI+) m/z (%) 484 (5), 452 (100), 456 (5), 466 (8), 438 (10), 428 (17). Anal. Calcd. for C14H8N6O8S3: C, 34.71; H, 1.66; N, 17.35; S, 19.85 Found: C, 34.73; H, 1.70; N, 17.33; S, 19.88. Rf = 0.48.

Antimicrobial susceptibility and antibiofilm assays

Microbial strains.

Strains of Klebsiella aerogenes ATCC 130148, Acinetobacter baumannii ATCC BAA-1605, Enterococcus faecium ATCC700221, Staphylococcus epidermidis ATCC35984 and Chromobacterium violaceum ATCC12472 from the American Type Culture Collection (ATCC) were used. They were maintained in Muller Hinton agar (MHA) at 37°C, while Chromobacterium violaceum strain was maintained in Luria–Bertani (LB) agar at 25°C.

Determination of the minimum inhibitory concentration (MIC) and the minimum bactericidal concentration (MBC).

MIC and MBC values were determined by the broth microdilution method using Muller Hinton broth (MHB). Stock solutions of compounds and reference antibacterial gentamicin were prepared in 100% dimethyl sulfoxide (DMSO; Sigma), and twofold serial dilutions were prepared in media in amounts of 100 μL per well in a 96-well plate. Then, 100 μL of a bacterial suspension was added to each well of the plate except those of the sterility control, resulting in a final inoculum of 1.5 × 106 CFU/mL. The final concentration of samples ranged 0.125 to 256 μg/mL. The final concentration of DMSO was lower than 2.5% and does not affect the bacterial growth. The medium without the agents was used as a growth control, and the blank control contained only the medium. Gentamicin (final concentrations’ range: 0.031–64 μg/mL) was used as a positive control. The microtitre plates were incubated at 37°C for 24 h. The assay was repeated three times in triplicate. The MIC of the samples was detected following the addition (40 µL) of 0.2 mg/mL p-iodonitrotetrazolium chloride and incubation at 37°C for 30 minutes. Viable microorganisms reduced the yellow dye to a pink color. MIC was defined as the lowest sample concentration that prevented this change and exhibited complete inhibition of bacterial growth.

The MBC was determined by adding 50 μL of the suspensions from the wells, which did not show any growth after incubation during MIC assays, to 150 μL of fresh MHB. These suspensions were incubated at 37°C for 48 h. The MBC was determined as the lowest concentration of sample that inhibited bacterial viability.

Determination of the minimum biofilm inhibitory concentration (MBIC50) and the minimum biofilm eradicating concentration (MBEC50).

In a preliminary experiment, all the test compounds were evaluated at their MIC concentration, and those with more than 50% biofilm inhibition/eradication were selected for a dose‒response assay to determine the MBIC50 and MBEC50 values. This test was carried out by the broth microdilution method as previously described [33]. Briefly, 100 μL of MHB supplemented with 2% glucose containing the samples was introduced into the first wells followed by a serial twofold dilution. Subsequently, 100 μL of bacterial suspension was added to all wells except those of the sterility control, resulting in a final inoculum of 1.5 × 106 CFU/mL, followed by incubation at 37°C for 24 h. After incubation, the plate was washed three times with phosphate-buffered saline (PBS; pH 7.2) to remove non-adherent bacteria cells. Wells containing MHB without bacteria served as the negative control. The remaining bacterial cells that attached to the well surface were considered as true biofilm. Then, the plates were stained with 0.1% crystal violet solution for 20 min at room temperature. After staining, the plates were washed three times with PBS. Then, the plates were air-dried and destained with 150 μL of 95% ethanol (v/v) for 30 min. Finally, the optical density was measured at 590 nm using a microplate reader (BioTek Epoch Microplate Spectrophotometer). Untreated wells and wells containing broth only were used as positive and blank controls, respectively, and the percentage of biofilm inhibition was calculated by using the following formula:

% inhibition = 100  [(ODsample  ODblank )/(ODcontrol  ODblank ) × 100]\newsavebox\eqbox

The MBEC50 was determined under the same conditions as the MBIC50, with the only difference being that the biofilm was allowed to form for 24 h before treatment with the samples.

Median MBIC50 and MBEC50 values were defined as the concentration inhibiting 50% of biofilm formation and preformed biofilm, respectively. This was calculated by plotting the percentage of inhibition or eradication versus the concentrations using GraphPad Prism software. Samples were tested in triplicate, and experiments were repeated three times.

Anti-quorum sensing assay

Inhibition of violacein production.

The inhibition of violacein production was performed according to a previously described method [34] and miniaturized in a 48-well microplate. This was achieved by transferring 1000 μL of test compounds’ solutions into the first well of a 48-well microplate, followed by a serial twofold dilution in LB broth. Then, 500 μL of C. violaceum inoculum, standardized at 3x106 CFU/mL, was added to all wells except those of the sterility control to obtain a final concentration of 4–256 μg/mL. Vanillin, at final concentrations ranging from 0.5 to 1024 μg/mL, was used as a reference compound [35]. Plates were properly sealed with parafilm and incubated in an orbital shaker (140 rpm) at 30°C for 24 h. The MIC was defined as the minimum concentration inhibiting visible bacterial growth and therefore preventing the production of purple pigmentation. The minimum quorum sensing inhibitory concentration (MQSIC) was defined as the lowest compound’s concentration allowing bacterial growth (shown by turbidity) without the visible production of purple pigmentation.

Quantification of violacein.

The inhibitory effect of selected compounds (4a, 4b and 4c) on violacein production was further quantified using a spectrophotometric method. After collection of MIC and MQSIC data, the plates were centrifuged at 4000 rpm for 20 min, and the supernatant was discarded. Then, the bacterial pellet was resuspended in 1 mL of DMSO, and the plates were further left in an orbital shaker for 10–15 min. Then, 200 µL of the supernatant was transferred into a 96-well microplate, and the optical density was measured at 595 nm. The percentage of violacein inhibition was calculated using the following formula:

% violacein inhibition = 100  [(ODsample  ODblank )/(ODcontrol  ODblamk ) × 100].

The IC50 values of the test compounds were defined as the concentrations inhibiting 50% of violacein production. These values were calculated by plotting the percentages of violacein production versus the concentrations using GraphPad Prism software. Samples were tested in triplicate, and each experiment was repeated three times.

Kinetics of bacterial growth and biofilm formation at sub-MIC concentrations

The effect of sub-MIC concentrations on bacterial growth and biofilm was evaluated by performing the kinetics of bacterial growth and biofilm formation at MIC, 1/2 xMIC, 1/4 xMIC, 1/8 xMIC, 1/16 xMIC and 1/32xMIC. Then, the average of optical density values obtained at 570 nm, were plotted against the concentrations.

Statistical analysis

The data are presented as the mean ± standard deviation (SD) of three independent experiments. Statistical differences between the IC50 values inhibiting the violacein production of samples and the reference compound (vanillin) were assessed by two-way ANOVA followed by Sidak’s multiple comparisons test in GraphPad Prism software.

Results

Chemistry

The synthesis of 1,2,3,5-tetrazine and azo dyes 4 is shown in Fig 1. All synthesized compounds were synthesised according to previous experimental procedure [32]. The yields, the melting points and all the spectroscopic data for these compounds described in the present study are in full agreement with those originally reported [32].

Fig 1. Reactions’ sequences to compounds 4.

Fig 1

Assays for the inhibition of bacteria

Table 1 summarizes the minimum inhibitory concentrations (MICs) and minimum bactericidal concentrations (MBCs) of 1,2,3,5-tetrazine derivatives upon screening against Klebsiella aerogenes, Acinetobacter baumannii, Enterococcus faecium and Staphylococcus epidermidis. The incubation of test compounds with the four pathogens afforded MIC and MBC values ranging from 8 to 256 µg/mL and from 64 to 256 µg/mL, respectively (Table 1). Against Klebsiella aerogenes, Acinetobacter baumannii, and Enterococcus faecium, the most promising compound viz. 4b showed bactericidal trend as evidenced by the ratios MBC/MIC (128/64, 64/16 and 32/8= 4), which is more than 2. Also, compound 4a (MBC/MIC: 128/16 = 8) and 4c (MBC/MIC: 64/16 = 4) were found to be bactericidal when incubated with Staphylococcus epidermidis. Gentamicin, the standard antibiotic agent showed antibacterial activity against the tested bacteria with MIC values ranging from 0.25 to 2 µg/mL.

Table 1. The minimum inhibitory concentrations (µg/mL) and the minimum bactericidal concentrations (µg/mL) of synthesized 1,2,3,5-tetrazine derivatives.

Samples Bacterial strains
Ka Ab Ef Se
MIC MBC MIC MBC MIC MBC MIC MBC
1a 256 256 256
1b 256 256 128 256 128 256
4a 128 128 64 128 32 64 16 128
4a’ 256 128 256 128 256
4b 64 128 16 64 8 32 32 64
4c 128 256 32 128 32 128 16 64
4c’ 256 128 256
Gentamicin 2 4 1 2 0.5 2 0.25 1

– = > 256 µg/mL Ka: Klebsiella aerogenes, Ab: Acinetobacter baumannii, Ef: Enterococcus faecium, Se: Staphylococcus epidermidis.

Anti-biofilm activity

The anti-biofilm effect of 1,2,3,5-tetrazine derivatives was evaluated on biofilms formed by Klebsiella aerogenes, Acinetobacter baumannii, Enterococcus faecium, and Staphylococcus epidermidis. The percentage of biofilm formation and eradication are summarized in Table 2. The degree of anti-biofilm activity of the test compounds was classified as highly and poorly actives for percentages of inhibition of >50%, and 0 < % < 50, respectively. Any compound with inhibition percentage of antibiofilm formation of 0 was considered inactive [36]. Compounds 4a (52–86.5%), 4b (57.7–79.4%) and 4c (59.9–80.3%) prevented biofilm formation in all the bacteria tested with percentages of inhibition >50%.

Table 2. Percentages of inhibition of biofilm formation and eradication of synthesized 1,2,3,5-tetrazine derivatives.

Samples Biofilm formation inhibition (%) Biofilm eradication (%)
Ka Ab Ef Se Ka Ab Ef Se
1a 4.8 ± 0.5 8.5 ± 1.2 18.9 ± 0.6 15.7 ± 1.7 −6.4 ± 1.3 3.5 ± 0.2 3.5 ± 0.1 12.5 ± 1.5
1b 13.7 ± 0.9 15.7 ± 1.4 25.3 ± 0.4 35.2 ± 2.7 5.6 ± 0.6 5.0 ± 1.2 5.4 ± 0.3 24.3 ± 2.7
4a 68.5 ± 5.2 86.5 ± 5.2 56.6 ± 3.4 52.0 ± 3.5 52.4 ± 4.6 75.4 ± 4.7 34.5 ± 2.4 42.0 ± 4.3
4a’ 28.9 ± 3.4 4.3 ± 0.4 37.4 ± 3.8 28.1 ± 1.6 13.5 ± 1.8 4.4 ± 1.0 0.09 ± 0.0 20.1 ± 2.6
4b 65.8 ± 6.9 79.4 ± 5.8 57.7 ± 2.4 70.3 ± 4.6 55.9 ± 3.6 66.3 ± 5.2 45.8 ± 2.4 61.0 ± 4.2
4c 65.8 ± 5.0 80.3 ± 8.7 59.9 ± 4.1 63.0 ± 4.2 59.0 ± 4.8 65.3 ± 5.7 48.0 ± 3.5 53.0 ± 5.8
4c’ 32.0 ± 2.5 22.5 ± 2.6 9.0 ± 0.2 21.6 ± 1.8 15.9 ± 1.4 10.0 ± 2.1 −5.4 ± 0.7 19.0 ± 1.2
Gentamicin 95.7 ± 6.2 99.1 ± 7.9 98.0 ± 8.0 99.4 ± 5.1 84.7 ± 6.5 75.2 ± 6.4 72.2 ± 8.5 86.2 ± 8.5

Ka Klebsiella aerogenes, Ab: Acinetobacter baumannii Ef: Enterococcus faecium, Se: Staphylococcus epidermidis.

Similarly, compounds 4b (55.9–61.0%) and 4c (53.0–65.3%) eradicated biofilms formed by K. aerogenes, A. baumannii and S. epidermidis with percentages of eradication of >50%. Moreover, compound 4a eradicated biofilms formed by K. aerogenes and A. baumannii with percentages of inhibition of 52.4 and 75.4%, respectively. Gentamicin, the antibiotic that was used as a positive control showed percentages of inhibition for biofilm formation and eradication ranging from 95.7 and 99.4% and from 72.2 and 86.2%, respectively (Table 2).

Table 3 summarizes the minimum biofilm inhibitory concentrations (MBIC50s) and minimum biofilm eradicating concentrations (MBEC50s) of compounds (4a, 4b and 4c) that exhibited more than 50% inhibition upon anti-biofilm formation and eradication assays. The minimum biofilm inhibitory concentrations (MBIC50s) ranged from 5.29 to 54.91 µg/mL, 7.79 to 45.23 µg/mL, and 8.56 to 87.35 µg/mL for compounds 4a, 4b and 4c, respectively (Table 3). Moreover, compounds 4a, 4b and 4c displayed minimum biofilm eradication concentrations (MBIC50s) of 98.62 and 60.98 µg/mL, 54.21 and 15.70 µg/mL, as well as 105.08 and 29.55 µg/mL, respectively, when tested against Klebsiella aerogenes and Acinetobacter baumannii (Table 3). All three compounds showed minimal growth inhibition at sub-MIC concentrations (≤64 μg/mL), with optical density (OD) curves closely paralleling the untreated control. Notably, 4b demonstrated the most pronounced separation between biofilm inhibition and growth curves, maintaining >80% biofilm reduction while showing <10% growth impact at 32 μg/mL (Supporting data S1 Fig). Similarly, the three compounds showed concentration-dependent suppression of violacein production, with 4b exhibiting the steepest dose-response curve, achieving 50% inhibition at as low as 28.56 μg/mL concentration while permitting 90% of Chromobacterium violaceum growth (Supporting data S2 Fig).

Table 3. MBIC50 and MBEC50 values (µg/mL) of synthesized 1,2,3,5-tetrazine derivatives against bacteria pathogen strains.

Samples MBIC50 (µg/mL)
Ka Ab Ef Se
4a 54.91 ± 5.3 44.34 ± 4.1 31.87 ± 2.8 5.29 ± 1.1
4b 45.23 ± 4.7 6.54 ± 1.0 7.79 ± 1.5 19.22 ± 4.8
4c 87.35 ± 7.7 18.33 ± 2.5 30.51 ± 3.1 8.56 ± 1.3
Gentamicin 1.22 ± 0.1 2.28 ± 0.4 1.03 ± 0.2 0.11 ± 0.0
MBEC50 (µg/mL)
4a 98.62 ± 8.5 60.98 ± 5.4 nd nd
4b 54.21 ± 4.9 15.70 ± 1.9 nd 25.44 ± 2.2
4c 105.08 ± 9.4 29.55 ± 3.1 nd 10.45 ± 2.1
Gentamicin 1.92 ± 0.1 0.95 ± 0.0 1.75 ± 0.2 0.18 ± 0.0

- = > 1024 µg/mL, Ka: Klebsiella aerogenes, Ab: Acinetobacter baumannii Ef: Enterococcus faecium, Se: Staphylococcus epidermidis.

Assay for the inhibition of quorum sensing

Inhibition of violacein formation.

An indicator strain of bacteria, i.e., Chromobacterium violaceum 12472 was used to test the inhibitory potential of the 1,2,3,5-tetrazine derivatives (4a, 4b and 4c) on the production of violacein, a water soluble pigment that result from the disruption of quorum-sensing signals or inhibition of cell growth in a number of gram negative bacteria [37,38]. Herein, the inhibitory effects of compounds 4a, 4b and 4c vis-à-vis violacein formation was evaluated at concentrations ranging from 0 to 256 µg/mL and the results are illustrated in Fig 2. As much as 256 µg/mL of compounds 4a, 4b and 4c completely inhibited violacein production as evidenced by the trends of the curves that almost overlapped with the x axis at 256 µg/mL concentration. 50% inhibition of violacein formation was observed at approximately 32, 64 and 110 µg/mL concentrations for compounds 4a, 4b and 4c, respectively, vs vanillin (50% inhibition of violacein formation at 64 µg/mL). These results demonstrate that compounds 4a, 4b and 4c possess anti-quorum sensing activity.

Fig 2. Percentage of violacein production in Chromobacterium violaceum by the most active synthesized 1,2,3,5-tetrazine derivatives (4a, 4b and 4c).

Fig 2

Determination of the minimum inhibitory concentration (MIC) and minimum quorum sensing inhibitory concentration (MQSIC).

Table 4 summarizes the minimum inhibitory concentration (MIC) and minimum quorum sensing inhibitory concentration (MQSIC) of compounds 4a, 4b and 4c, the most promising antibacterial tetrazine derivatives. Compounds 4a, 4b and 4c revealed MIC and MQSIC values ranging from 256 to 512 µg/mL and from 64 and 256 µg/mL, respectively (Table 4). The concentrations of 4a, 4b and 4c that inhibited 50% of violacein production were found to be 62.71, 28.56 and 107.45 µg/mL, respectively, vs vanillin (IC50: 56.75 µg/mL).

Table 4. Minimum quorum sensing inhibitory concentrations (MQSICs), minimum inhibitory concentrations (MICs), and concentrations inhibiting 50% of violacein production (IC50s) of compounds 4a, 4b and 4c.
Samples MIC MQSIC IC50
4a 512 256 62.71 ± 5.11
4b 256 64 28.56 ± 1.24***
4c 256 128 107.45 ± 8.56***
Vanillin 256 128 56.75 ± 4.32

Statistical analysis was performed with Sidak’s multiple comparisons test using two-way ANOVA; ***p < 0.001.

Discussion

Combating antimicrobial resistance (AMR) has been declared as a priority concern by the World Health Organization (WHO) ever since antibacterial AMR was directly responsible for 1.27 million global deaths in 2019 [4]. Adequate research and development on quality diagnosis, effective treatment of bacterial infections, and innovation are among the strategies that can slow down and eliminate bacterial drug resistance. To contribute toward the identification of effective treatments against drug resistant bacterial infections, numerous scientists have investigated the inhibitory effects of natural and synthetic compounds against different pathogens. This observation is exemplified by the number of recently published review articles on the antibacterial activity of heterocyclic compounds, including benzothiazole derivatives [3941], tetrazine compounds [42,43], among others. On the other hand, the implication of biofilm formation [6,44,45] and quorum sensing system [4648] in developing bacterial drug-resistance is prominent [49,50]. Thus, the search for effective antibacterial treatments with potential inhibition of biofilm formation and quorum sensing activity is valuable and might significantly contribute to antimicrobial drug discovery against multi-resistant bacteria. In point of fact, a number of antibacterial chemotypes were reported to exhibit anti-quorum sensing [51,52] and anti-biofilm activity [53,54]. However, the identification of antibacterial pharmacophores with novel features is still needed since none of these promising hit compounds is indicated to have entered the clinical trial phase, perhaps the reason being their unfavourable pharmacokinetic characteristics, their safety limits, and so on. As the benzothiazole and tetrazine rings hold promise as antibacterial potential candidates and that previously prepared derivatives from compounds bearing both the pharmacophores revealed non cytotoxicity against normal and cancer cells, the present study evaluated the antibacterial efficacy of 1,2,3,5-tetrazine amalgamated benzothiazole derivatives.

As a result, the synthesized 1,2,3,5-tetrazine derivatives showed MIC and MBC values ranging from 8 to 256 µg/mL and from 64 to 256 µg/mL, respectively, thus highlighting the antibacterial activity of these compounds. Compound 4b was found to be the most potent, followed by compound 4c and 4a. Noteworthy, compound 4b harbours more benzothiazole rings than its counterparts 4a and 4c, thus justifying the high inhibition of selected bacteria by compound 4b.

Similarly, the compound 4a contain a number of nitro groups, which might have aided in the observed antibacterial activity. By contrast, compound 4c bears only two nitro groups, whereas compound 4a do not contain any nitro moiety. The chemical and physical properties of the nitro group (−NO2) including its size, electron-withdrawing ability, polarity, ability to form hydrogen bonds and redox properties contribute to its key role in the action of many drugs, especially antimicrobial agents [55]. Accumulated evidence has shown the significant role of the nitro group in the inhibition of several bacteria [5658].

The anti-biofilm and anti-quorum sensing effects of the most promising derivatives were also evaluated. As a result, compounds 4a, 4b and 4c, inhibited biofilm formation with minimum biofilm inhibitory concentrations (MBIC50s) ranging from 5.29 to 54.91 µg/mL, 7.79 to 45.23 µg/mL, and 8.56 to 87.35 µg/mL for compounds 4a, 4b and 4c, respectively. Moreover, compounds 4a, 4b and 4c displayed minimum biofilm eradication concentrations (MBIC50s) of 98.62 and 60.98 µg/mL, 54.21 and 15.70 µg/mL, as well as 105.08 and 29.55 µg/mL, respectively, when tested against Klebsiella aerogenes and Acinetobacter baumannii. As shown in the supporting information (S1 and S2 Fig), the sub-inhibitory concentrations of compounds 4a, 4b and 4c did not significantly affect the bacterial growth, biofilm formation and biological synthesis of violacein over the tested time period, thus justifying the selective inhibitory effects of the test compounds on these specific bacterial processes rather than a general decrease in the bacterial cell viability. These results are consistent with previously reported data on the anti-quorum sensing effects of benzothiazole derivatives containing an isopropanolamine moiety [51], pyrazole-based benzothiazoles [59], etc. Moreover, series of 2-azidobenzothiazoles [51] and benzothiazole‒urea hybrids [60] were reported to exhibit anti-biofilm activity. On the other hand, tetrazine groups are reputed for their antibacterial potential [61,62]. In general, the antibacterial activity of benzothiazole derivatives has been attributed to binding onto enzymes that are important for essential processes in the bacterial cells, such as cell-wall synthesis, cell division, and DNA replication [63,64]. Inhibition of violacein formation has been used as an approach to determine anti-quorum sensing activity [65]. The antibacterial, anti-biofilm, and anti-quorum sensing (QS) activities of the synthesized 1,2,3,5-tetrazine-benzothiazole hybrids (4a-4c) can be attributed to their unique structural features and potential interactions with bacterial targets. The potent bactericidal effects of 4b (MIC: 8–64 µg/mL) likely stem from the benzothiazole moieties. These heterocycles may inhibit DNA gyrase or topoisomerase IV, which is a critical enzyme for bacterial DNA replication [63]. The additional benzothiazole rings in 4b could improve target binding affinity compared to 4a and 4c. At sub-MIC concentrations, 4a-4c selectively inhibited biofilm formation and violacein production without affecting bacterial growth. These results suggest the disruption of extracellular polymeric substances (EPS), since the azo and tetrazine groups are well known to interfere with EPS synthesis [66]. Growing evidence has shown that QS interference by violacein suppression might result from the inhibition of the CviR/I system in C. violaceum [65,67]. The planar structure of compound 4b may competitively bind LuxR-type receptors, thus blocking the signal transduction [51].

Conclusions

This work has unveiled the antibacterial, anti-quorum sensing and anti-biofilm formation of selected 1,2,3,5-tetrazine-benzothiazole hybrids, which can be prospected as potential pharmacophores for the discovery of effective antibacterial agents. Future studies could dissect the mechanisms of action of the antibacterial compounds using transcriptional analysis such as RT-qPCR of QS genes, or genetic knockouts to confirm target specificity. Nevertheless, our findings contribute to the rationale for developing sub-MIC therapies targeting virulence, particularly in biofilm-associated infections where conventional antibiotics have failed due to several factors, including the physical barrier of the biofilm matrix, slow bacterial growth rates, and the presence of persistent cells within the biofilm.

Supporting information

S1 Fig. Effect of selected compounds at sub-inhibitory concentrations on biofilm and bacterial growth inhibition.

A: effect of compound 4a, B: effect of compound 4b, C: effect of compound 4c, MIC: minimum inhibitory concentration.

(DOCX)

pone.0318135.s001.docx (128.1KB, docx)
S2 Fig. Effect of selected compounds 4a, 4b and4c onChromobacterium violaceum growth at sub-inhibitory concentrations.

MIC: minimum inhibitory concentration.

(DOCX)

pone.0318135.s002.docx (32.4KB, docx)

Data Availability

All relevant data are within the manuscript and its Supporting Information files.

Funding Statement

The author(s) received no specific funding for this work.

References

  • 1.O’Neill L, Manzanilla EG, Ekhlas D, Leonard FC. Antimicrobial Resistance in Commensal Escherichia coli of the Porcine Gastrointestinal Tract. Antibiotics (Basel). 2023;12(11):1616. doi: 10.3390/antibiotics12111616 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Oliveira M, Antunes W, Mota S, Madureira-Carvalho Á, Dinis-Oliveira RJ, Dias da Silva D. An Overview of the Recent Advances in Antimicrobial Resistance. Microorganisms. 2024;12(9):1920. doi: 10.3390/microorganisms12091920 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Antimicrobial Resistance Collaborators. Global burden of bacterial antimicrobial resistance in 2019: a systematic analysis. Lancet. 2022;399(10325):629–55. doi: 10.1016/S0140-6736(21)02724-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.World Health Organization (WHO). The Fact Sheets. Antimicrobial Resistance. 2024. [cited 09th October 2024] Available from: https://www.who.int/news-room/fact-sheets/detail/antimicrobial-resistance [Google Scholar]
  • 5.Rajput A, Thakur A, Sharma S, Kumar M. aBiofilm: a resource of anti-biofilm agents and their potential implications in targeting antibiotic drug resistance. Nucleic Acids Res. 2018;46(D1):D894–900. doi: 10.1093/nar/gkx1157 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Sharma S, Mohler J, Mahajan SD, Schwartz SA, Bruggemann L, Aalinkeel R. Microbial Biofilm: A Review on Formation, Infection, Antibiotic Resistance, Control Measures, and Innovative Treatment. Microorganisms. 2023;11(6):1614. doi: 10.3390/microorganisms11061614 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Juszczuk-Kubiak E. Molecular Aspects of the Functioning of Pathogenic Bacteria Biofilm Based on Quorum Sensing (QS) Signal-Response System and Innovative Non-Antibiotic Strategies for Their Elimination. Int J Mol Sci. 2024;25(5):2655. doi: 10.3390/ijms25052655 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Singh S, Singh SK, Chowdhury I, Singh R. Understanding the Mechanism of Bacterial Biofilms Resistance to Antimicrobial Agents. Open Microbiol J. 2017;11:53–62. doi: 10.2174/1874285801711010053 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Abebe GM. The Role of Bacterial Biofilm in Antibiotic Resistance and Food Contamination. Int J Microbiol. 2020;2020:1705814. doi: 10.1155/2020/1705814 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Shree P, Singh CK, Sodhi KK, Surya JN, Singh DK. Biofilms: Understanding the structure and contribution towards bacterial resistance in antibiotics. Medicine in Microecology. 2023;16:100084. doi: 10.1016/j.medmic.2023.100084 [DOI] [Google Scholar]
  • 11.Zhao X, Yu Z, Ding T. Quorum-Sensing Regulation of Antimicrobial Resistance in Bacteria. Microorganisms. 2020;8(3):425. doi: 10.3390/microorganisms8030425 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Kirisits MJ, Parsek MR. Does Pseudomonas aeruginosa use intercellular signalling to build biofilm communities?. Cell Microbiol. 2006;8(12):1841–9. doi: 10.1111/j.1462-5822.2006.00817.x [DOI] [PubMed] [Google Scholar]
  • 13.Spoering AL, Gilmore MS. Quorum sensing and DNA release in bacterial biofilms. Curr Opin Microbiol. 2006;9(2):133–7. doi: 10.1016/j.mib.2006.02.004 [DOI] [PubMed] [Google Scholar]
  • 14.Hooshangi S, Bentley WE. From unicellular properties to multicellular behavior: bacteria quorum sensing circuitry and applications. Curr Opin Biotechnol. 2008;19(6):550–5. doi: 10.1016/j.copbio.2008.10.007 [DOI] [PubMed] [Google Scholar]
  • 15.Wu L, Luo Y. Bacterial Quorum-Sensing Systems and Their Role in Intestinal Bacteria-Host Crosstalk. Front Microbiol. 2021;12:611413. doi: 10.3389/fmicb.2021.611413 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Zhang A, Chu W-H. Anti-Quorum Sensing Activity of Forsythia suspense on Chromobacterium violaceum and Pseudomonas aeruginosa. Pharmacogn Mag. 2017;13(50):321–5. doi: 10.4103/0973-1296.204547 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Sadik A, Viswaswar JP, Rajamoney A, Rekha A, Raj DM, Prakashan D, et al. Mitigation of quorum sensing mediated virulence factors of Pseudomonas aeruginosa: the role of Meldrum’s acid activated furan. Front Microbiol. 2024;14:1272240. doi: 10.3389/fmicb.2023.1272240 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Sionov RV, Steinberg D. Targeting the Holy Triangle of Quorum Sensing, Biofilm Formation, and Antibiotic Resistance in Pathogenic Bacteria. Microorganisms. 2022;10(6):1239. doi: 10.3390/microorganisms10061239 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Grooters KE, Ku JC, Richter DM, Krinock MJ, Minor A, Li P, et al. Strategies for combating antibiotic resistance in bacterial biofilms. Front Cell Infect Microbiol. 2024;14:1352273. doi: 10.3389/fcimb.2024.1352273 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Kalia VC. Quorum sensing inhibitors: an overview. Biotechnol Adv. 2013;31(2):224–45. doi: 10.1016/j.biotechadv.2012.10.004 [DOI] [PubMed] [Google Scholar]
  • 21.Kalia VC, Patel SKS, Lee J-K. Bacterial biofilm inhibitors: An overview. Ecotoxicol Environ Saf. 2023;264:115389. doi: 10.1016/j.ecoenv.2023.115389 [DOI] [PubMed] [Google Scholar]
  • 22.Asfour HZ. Anti-Quorum Sensing Natural Compounds. J Microsc Ultrastruct. 2018;6(1):1–10. doi: 10.4103/JMAU.JMAU_10_18 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Bouyahya A, Chamkhi I, Balahbib A, Rebezov M, Shariati MA, Wilairatana P, et al. Mechanisms, Anti-Quorum-Sensing Actions, and Clinical Trials of Medicinal Plant Bioactive Compounds against Bacteria: A Comprehensive Review. Molecules. 2022;27(5):1484. doi: 10.3390/molecules27051484 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Iaconis A, De Plano LM, Caccamo A, Franco D, Conoci S. Anti-Biofilm Strategies: A Focused Review on Innovative Approaches. Microorganisms. 2024;12(4):639. doi: 10.3390/microorganisms12040639 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Booysen IN, Ismail MB, Akerman MP. N-[(E)-Thio-phen-2-yl-methyl-idene]-1,3-benzothia-zol-2-amine. Acta Crystallogr Sect E Struct Rep Online. 2012;68(Pt 8):o2489. doi: 10.1107/S1600536812030498 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Sumit, Kumar A, Mishra AK. Advancement in Pharmacological Activities of Benzothiazole and its Derivatives: An Up to Date Review. Mini Rev Med Chem. 2021;21(3):314–35. doi: 10.2174/1389557520666200820133252 [DOI] [PubMed] [Google Scholar]
  • 27.Zhilitskaya LV, Shainyan BA, Yarosh NO. Modern Approaches to the Synthesis and Transformations of Practically Valuable Benzothiazole Derivatives. Molecules. 2021;26(8):2190. doi: 10.3390/molecules26082190 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Catalano A, Rosato A, Salvagno L, Iacopetta D, Ceramella J, Fracchiolla G, et al. Benzothiazole-Containing Analogues of Triclocarban with Potent Antibacterial Activity. Antibiotics (Basel). 2021;10(7):803. doi: 10.3390/antibiotics10070803 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Yadav RK, Kumar R, Singh H, Mazumdar A, Chauhan B, et al. Recent Insights on Synthetic Methods and Pharmacological Potential in Relation with Structure of Benzothiazoles. Med Chem. 2023;19(4):325–60. doi: 10.2174/1573406418666220820110551 [DOI] [PubMed] [Google Scholar]
  • 30.El-Reedy AAM, Soliman NK. Synthesis, biological activity and molecular modeling study of novel 1,2,4-triazolo[4,3-b][1,2,4,5]tetrazines and 1,2,4-triazolo[4,3-b][1,2,4]triazines. Sci Rep. 2020;10(1):6137. doi: 10.1038/s41598-020-62977-x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Ishmetova RI, Ignatenko NK, Gerasimova NA, Belyaev DV, Butorin II, Konovalova OA. 3,6-disubstituted derivatives of 1,2,4,5-tetrazine with pyridinyl amidine moieties and condensed systems on their basis: synthesis, docking, and antibacterial activity. Russ Chem Bull. 2024;73:1686–97. [Google Scholar]
  • 32.Tsemeugne J, Bah YA, Dzoyem JP, Ndefongang JN, Famuyide IM, McGaw LJ, et al. Synthesis and anticancer activity evaluation of some new 1,2,3,5-tetrazine derivatives attached to benzothiazole moiety. Arkivoc. 2022; part ix:73–89. [Google Scholar]
  • 33.Bisso BN, Makuété AL, Tsopmene JU, Dzoyem JP. Biofilm Formation and Phospholipase and Proteinase Production in Cryptococcus neoformans Clinical Isolates and Susceptibility towards Some Bioactive Natural Products. ScientificWorldJournal. 2023;2023:6080489. doi: 10.1155/2023/6080489 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Ahmad A, Viljoen AM, Chenia HY. The impact of plant volatiles on bacterial quorum sensing. Lett Appl Microbiol. 2015;60(1):8–19. doi: 10.1111/lam.12343 [DOI] [PubMed] [Google Scholar]
  • 35.Choo JH, Rukayadi Y, Hwang J-K. Inhibition of bacterial quorum sensing by vanilla extract. Lett Appl Microbiol. 2006;42(6):637–41. doi: 10.1111/j.1472-765X.2006.01928.x [DOI] [PubMed] [Google Scholar]
  • 36.Sandasi M, Leonard C, Van Vuuren S, Viljoen A. Peppermint (Mentha piperita) inhibits microbial biofilms in vitro. S Afr J Bot. 2011;77(1):80–5. [Google Scholar]
  • 37.Kocak G, Tamfu A, Bütün V, Ceylan O. Synthesis of quaternary piperazine methacrylate homopolymers and their antibioflm and anti-quorum sensing efects on pathogenic bacteria. J Appl Polym Sci. 2021;138(21):50466. [Google Scholar]
  • 38.Tamfu AN, Kucukaydin S, Ceylan O, Sarac N, Duru EM. Phenolic composition, enzyme inhibitory and anti-quorum sensing activities of cinnamon (Cinnamomum zeylanicum blume) and basil (Ocimum basilicum linn). Chem Afr. 2021;4(4):759–67. [Google Scholar]
  • 39.Franchini C, Muraglia M, Corbo F, Florio MA, Di Mola A, Rosato A, et al. Synthesis and biological evaluation of 2-mercapto-1,3-benzothiazole derivatives with potential antimicrobial activity. Arch Pharm (Weinheim). 2009;342(10):605–13. doi: 10.1002/ardp.200900092 [DOI] [PubMed] [Google Scholar]
  • 40.Ali R, Siddiqui N. Biological Aspects of Emerging Benzothiazoles: A Short Review. Journal of Chemistry. 2013;2013(1). doi: 10.1155/2013/345198 [DOI] [Google Scholar]
  • 41.Kashyap P, Verma S, Gupta P, Narang R, Lal S, Devgun M. Recent insights into antibacterial potential of benzothiazole derivatives. Med Chem Res. 2023;:1–31. doi: 10.1007/s00044-023-03077-z [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Neamah IJ, Baqer FM, Hassan BA. Review nomenclature systems of tetrazine and its pharmacology application. Int J Pharm Sci Res. 2022;7:1–6. [Google Scholar]
  • 43.Abdulridha MM, Hassan BA, Neamah IJ. Review theoretical study of synthesis and pharmaceutical study of tetrazine derivatives. Ann Rom Soc Cell Biol. 2022;26(01):1657–69. [Google Scholar]
  • 44.Rather MA, Gupta K, Mandal M. Microbial biofilm: formation, architecture, antibiotic resistance, and control strategies. Braz J Microbiol. 2021;52(4):1701–18. doi: 10.1007/s42770-021-00624-x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Mirghani R, Saba T, Khaliq H, Mitchell J, Do L, Chambi L, et al. Biofilms: Formation, drug resistance and alternatives to conventional approaches. AIMS Microbiol. 2022;8(3):239–77. doi: 10.3934/microbiol.2022019 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Gupta DS, Kumar MS. The implications of quorum sensing inhibition in bacterial antibiotic resistance- with a special focus on aquaculture. J Microbiol Methods. 2022;203:106602. doi: 10.1016/j.mimet.2022.106602 [DOI] [PubMed] [Google Scholar]
  • 47.Patel R, Soni M, Soyantar B, Shivangi S, Sutariya S, Saraf M, et al. A clash of quorum sensing vs quorum sensing inhibitors: an overview and risk of resistance. Arch Microbiol. 2023;205(4):107. doi: 10.1007/s00203-023-03442-x [DOI] [PubMed] [Google Scholar]
  • 48.Rodríguez-Urretavizcaya B, Vilaplana L, Marco M-P. Strategies for quorum sensing inhibition as a tool for controlling Pseudomonas aeruginosa infections. Int J Antimicrob Agents. 2024;64(5):107323. doi: 10.1016/j.ijantimicag.2024.107323 [DOI] [PubMed] [Google Scholar]
  • 49.Vashistha A, Sharma N, Nanaji Y, Kumar D, Singh G, Barnwal RP, et al. Quorum sensing inhibitors as Therapeutics: Bacterial biofilm inhibition. Bioorg Chem. 2023;136:106551. doi: 10.1016/j.bioorg.2023.106551 [DOI] [PubMed] [Google Scholar]
  • 50.Hemmati J, Nazari M, Abolhasani FS, Ahmadi A, Asghari B. In vitro investigation of relationship between quorum-sensing system genes, biofilm forming ability, and drug resistance in clinical isolates of Pseudomonas aeruginosa. BMC Microbiol. 2024;24(1):99. doi: 10.1186/s12866-024-03249-w [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Chu P-L, Feng Y-M, Long Z-Q, Xiao W-L, Ji J, Zhou X, et al. Novel Benzothiazole Derivatives as Potential Anti-Quorum Sensing Agents for Managing Plant Bacterial Diseases: Synthesis, Antibacterial Activity Assessment, and SAR Study. J Agric Food Chem. 2023;71(17):6525–40. doi: 10.1021/acs.jafc.2c07810 [DOI] [PubMed] [Google Scholar]
  • 52.Talla RM, Tamfu AN, Wakeu BNK, Ceylan O, Mbazoa CD, Kapche GDWF, et al. Evaluation of anti-quorum sensing and antibiofilm effects of secondary metabolites from Gambeya lacourtiana (De Wild) Aubr. & Pellegr against selected pathogens. BMC Complement Med Ther. 2023;23(1):300. doi: 10.1186/s12906-023-04115-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Stojković D, Petrović J, Carević T, Soković M, Liaras K. Synthetic and Semisynthetic Compounds as Antibacterials Targeting Virulence Traits in Resistant Strains: A Narrative Updated Review. Antibiotics (Basel). 2023;12(6):963. doi: 10.3390/antibiotics12060963 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Adeyemo RO, Famuyide IM, Dzoyem JP, Lyndy Joy M. Anti-Biofilm, Antibacterial, and Anti-Quorum Sensing Activities of Selected South African Plants Traditionally Used to Treat Diarrhoea. Evid Based Complement Alternat Med. 2022;2022:1307801. doi: 10.1155/2022/1307801 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Rice AM, Long Y, King SB. Nitroaromatic Antibiotics as Nitrogen Oxide Sources. Biomolecules. 2021;11(2):267. doi: 10.3390/biom11020267 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Lobana TS, Indoria S, Sood H, Arora DS, Kaur M, Jasinski JP. Synthesis of (3-nitro-2-oxo-benzaldehyde thiosemicarbazonato)-zinc(II) complexes: the position of nitro group in phenyl ring alters antimicrobial activity against K. pneumoniae 1, S. typhimurium 2, MRSA and C. albicans. Dalton Trans. 2021;50(20):6823–33. doi: 10.1039/d1dt00657f [DOI] [PubMed] [Google Scholar]
  • 57.Noriega S, Cardoso-Ortiz J, López-Luna A, Cuevas-Flores MDR, Flores De La Torre JA. The Diverse Biological Activity of Recently Synthesized Nitro Compounds. Pharmaceuticals (Basel). 2022;15(6):717. doi: 10.3390/ph15060717 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Ayoup MS, Rabee AR, Abdel-Hamid H, Harras MF, El Menofy NG, Ismail MMF. Exploration of Nitroaromatic Antibiotics via Sanger’s Reagent: Synthesis, In Silico, and Antimicrobial Evaluation. ACS Omega. 2022;7(6):5254–63. doi: 10.1021/acsomega.1c06383 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Gabr MT, El-Gohary NS, El-Bendary ER, El-Kerdawy MM, Ni N, Shaaban MI. Synthesis, antimicrobial, antiquorum-sensing and cytotoxic activities of new series of benzothiazole derivatives. Chin Chem Lett. 2015;26(12):1522–8. [Google Scholar]
  • 60.Zha L, Xie Y, Wu C, Lei M, Lu X, Tang W, et al. Novel benzothiazole‒urea hybrids: Design, synthesis and biological activity as potent anti-bacterial agents against MRSA. Eur J Med Chem. 2022;236:114333. doi: 10.1016/j.ejmech.2022.114333 [DOI] [PubMed] [Google Scholar]
  • 61.Shawali AS, Tawfik NM. Novel facile synthesis of imidazo[1,2-b]-[1,2,4,5]tetrazines with potential antimicrobial activity. Arch Pharmacal Res. 2009; 32: 975–82. [DOI] [PubMed] [Google Scholar]
  • 62.Al-Omair MA, Sayed AR, Youssef MM. Synthesis of novel triazoles, tetrazine, thiadiazoles and their biological activities. Molecules. 2015;20(2):2591–610. doi: 10.3390/molecules20022591 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Gjorgjieva M, Tomašič T, Kikelj D, Mašič LP. Benzothiazole-based Compounds in Antibacterial Drug Discovery. Curr Med Chem. 2018;25(38):5218–36. doi: 10.2174/0929867324666171009103327 [DOI] [PubMed] [Google Scholar]
  • 64.Haroun M. Review on the Developments of Benzothiazole-containing Antimicrobial Agents. Curr Top Med Chem. 2022;22(32):2630–59. doi: 10.2174/1568026623666221207161752 [DOI] [PubMed] [Google Scholar]
  • 65.Dimitrova PD, Ivanova V, Trendafilova A, Paunova-Krasteva T. Anti-Biofilm and Anti-Quorum-Sensing Activity of Inula Extracts: A Strategy for Modulating Chromobacterium violaceum Virulence Factors. Pharmaceuticals (Basel). 2024;17(5):573. doi: 10.3390/ph17050573 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Roy R, Tiwari M, Donelli G, Tiwari V. Strategies for combating bacterial biofilms: A focus on anti-biofilm agents and their mechanisms of action. Virulence. 2018;9(1):522–54. doi: 10.1080/21505594.2017.1313372 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Khan MA, Shahid M, Celik I, Khan HM, Shahzad A, Husain FM, et al. Attenuation of quorum sensing regulated virulence functions and biofilm of pathogenic bacteria by medicinal plant Artemisia annua and its phytoconstituent 1, 8-cineole. Microsc Res Tech. 2024;87(1):133–48. doi: 10.1002/jemt.24418 [DOI] [PubMed] [Google Scholar]

Decision Letter 0

M Alejandro Dinamarca

28 Mar 2025

PONE-D-25-01821Antibacterial, antibiofilm and anti-quorum sensing activities of 1,2,3,5-tetrazine derivatives linked to a benzothiazole moietyPLOS ONE

Dear Dr. Pone Kamdem,

Thank you for submitting your manuscript to PLOS ONE. After careful consideration, we feel that it has merit but does not fully meet PLOS ONE’s publication criteria as it currently stands. Therefore, we invite you to submit a revised version of the manuscript that addresses the points raised during the review process.

The submitted manuscript has scientific merit and it is positively valued to work with chemically synthesized compounds by the authors' own team in the context of bacterial quorum sensing inhibition. However, in light of the review, the manuscript must correct and clarify aspects related to the methodology, specifically the relationship between MICs, MIBCs, and the biomass necessary to perform QS and form biofilms. Additionally, the authors should delve into the chemical and molecular mechanisms that explain the observed results. This should be done by proposing chemical or molecular biology assays that can elucidate the pathways through which the derived triazines exert their action.

Please submit your revised manuscript by april 25th. If you will need more time than this to complete your revisions, please reply to this message or contact the journal office at plosone@plos.org . When you're ready to submit your revision, log on to https://www.editorialmanager.com/pone/ and select the 'Submissions Needing Revision' folder to locate your manuscript file.

Please include the following items when submitting your revised manuscript:

  • A rebuttal letter that responds to each point raised by the academic editor and reviewer(s). You should upload this letter as a separate file labeled 'Response to Reviewers'.

  • A marked-up copy of your manuscript that highlights changes made to the original version. You should upload this as a separate file labeled 'Revised Manuscript with Track Changes'.

  • An unmarked version of your revised paper without tracked changes. You should upload this as a separate file labeled 'Manuscript'.

If you would like to make changes to your financial disclosure, please include your updated statement in your cover letter. Guidelines for resubmitting your figure files are available below the reviewer comments at the end of this letter.

If applicable, we recommend that you deposit your laboratory protocols in protocols.io to enhance the reproducibility of your results. Protocols.io assigns your protocol its own identifier (DOI) so that it can be cited independently in the future. For instructions see: https://journals.plos.org/plosone/s/submission-guidelines#loc-laboratory-protocols . Additionally, PLOS ONE offers an option for publishing peer-reviewed Lab Protocol articles, which describe protocols hosted on protocols.io. Read more information on sharing protocols at https://plos.org/protocols?utm_medium=editorial-email&utm_source=authorletters&utm_campaign=protocols .

We look forward to receiving your revised manuscript.

Kind regards,

M. Alejandro Dinamarca, Dr.

Academic Editor

PLOS ONE

Journal Requirements:

When submitting your revision, we need you to address these additional requirements.

1. Please ensure that your manuscript meets PLOS ONE's style requirements, including those for file naming. The PLOS ONE style templates can be found at 

https://journals.plos.org/plosone/s/file?id=wjVg/PLOSOne_formatting_sample_main_body.pdf and 

https://journals.plos.org/plosone/s/file?id=ba62/PLOSOne_formatting_sample_title_authors_affiliations.pdf

Additional Editor Comments:

The article titled Antibacterial, antibiofilm and anti-quorum sensing activities of 1,2,3,5-tetrazine derivatives linked to a benzothiazole moiety, is a work that evaluates a series of organic compounds derived from chemical synthesis conducted by the group of authors. Specifically, the authors evaluate their synthesis products in two related bacterial behavior systems, which are quorum sensing communication and biofilm formation. The evaluations are conducted on a series of bacterial pathogens or pathogenicity models such as: Klebsiella aerogenes ATCC 130148, Acinetobacter baumannii ATCC BAA-1605, Enterococcus faecium ATCC700221, Staphylococcus epidermidis. Additionally, the ability to inhibit the quorum sensing mechanism is evaluated using the production of violacein in Chromobacterium violaceum ATCC12472 as a model. To methodologically relate their work, the authors propose an experimental design based on determining the respective MICs and MIBCs of each compound in concentrations of µg/ml. The results of the work are expressed in percentage values through known formulas.

The results are consistent with the experimental design; however, they present a methodological question that must be clarified by the authors. Specifically, if bacteria need a specific concentration to perform QS communication, how does this relate to the concentrations related to MICs and MIBCs used for each evaluated triazine? The same is considered for the formation or eradication of biofilms. This must be adequately clarified in the methodology section and in the results, with a special mention in the discussion section.

Finally, although the article is coherent and it is positively valued to work with compounds derived from synthesis, it is required to delve deeper methodologically or in the discussion regarding the molecular and chemical mechanisms that may be involved in the observed results.

The manuscript must be presented in the format required by PLOS.

[Note: HTML markup is below. Please do not edit.]

Reviewers' comments:

Reviewer's Responses to Questions

Comments to the Author

1. Is the manuscript technically sound, and do the data support the conclusions?

The manuscript must describe a technically sound piece of scientific research with data that supports the conclusions. Experiments must have been conducted rigorously, with appropriate controls, replication, and sample sizes. The conclusions must be drawn appropriately based on the data presented.

Reviewer #1: Partly

**********

2. Has the statistical analysis been performed appropriately and rigorously?

Reviewer #1: N/A

**********

3. Have the authors made all data underlying the findings in their manuscript fully available?

The PLOS Data policy requires authors to make all data underlying the findings described in their manuscript fully available without restriction, with rare exception (please refer to the Data Availability Statement in the manuscript PDF file). The data should be provided as part of the manuscript or its supporting information, or deposited to a public repository. For example, in addition to summary statistics, the data points behind means, medians and variance measures should be available. If there are restrictions on publicly sharing data—e.g. participant privacy or use of data from a third party—those must be specified.

Reviewer #1: Yes

**********

4. Is the manuscript presented in an intelligible fashion and written in standard English?

PLOS ONE does not copyedit accepted manuscripts, so the language in submitted articles must be clear, correct, and unambiguous. Any typographical or grammatical errors should be corrected at revision, so please note any specific errors here.

Reviewer #1: Yes

**********

5. Review Comments to the Author

Please use the space provided to explain your answers to the questions above. You may also include additional comments for the author, including concerns about dual publication, research ethics, or publication ethics. (Please upload your review as an attachment if it exceeds 20,000 characters)

Reviewer #1: The manuscript addresses a relevant topic and evaluates new potential antimicrobial and antivirulence compounds. While the evaluated compounds show potential against relevant pathogens, there are some aspects of the microbiological assays that are not included and without which a solid conclusion can not be generated.

Major comments

There are some major methodological issues regarding the evaluation of biofilm inhibition and violacein production. While authors test different concentrations of the selected compounds below MIC and MBC, they do not take into consideration the potential effect of subinhibitory concentrations of these on bacterial growth. Therefore, it is not possible to determine if the lower biofilm formation and violacein production is due to the selective effect of the compounds on these cellular processes or to a reduction of bacterial cells. Normalization by total cell number (CFU or Optical density at 600 nm) or growth curves at the selected concentrations of the compounds should be included to support the conclusions.

Minor comments:

Introduction, L5: change "The main mechanisms of ..... includes" for "include" (plural)

Introduction, L7: "degradation" may be replace bt a more accurate concept as "structural modification"

Methods, 2.2.1: Please indicate why C. violaceum was cultured at 30 ºC instead of 26 ºC (its optimal growth temperature)

Methods 2.2.3: Delete ")" after gentamicin.

Please specify gentamicin concentrations used as control condition.

"The MBC was determined as the ...that completely inhibited the growth of bacteria". Because authors are evaluating bactericidal effect, the more accurate concept should be "that inhibited bacterial viability".

Methods, 2.3.1. Please specify vanillin concentration

Results, 3.1.2: Authors should include ATCC numbers for all strains (or none).

Results, 3.4.1, Figure 2. According to the figure and its Y-axis, lower concentrations of the selected compouds produced higher ihnibition of violacein production. Please check this information and plot´s axis names.

Discussion "By contrast, compound 4c bears only two nitro groups, whereas

compound 4c do not contain any nitro moiety". Authors mention compound 4c twice, please check.

Conclusion. Authors do not expose the conclusion of their research. Instead, in this section, they summarize the relevance and of their results. Please, include the conclusions.

**********

6. PLOS authors have the option to publish the peer review history of their article (what does this mean? ). If published, this will include your full peer review and any attached files.

If you choose “no”, your identity will remain anonymous but your review may still be made public.

Do you want your identity to be public for this peer review? For information about this choice, including consent withdrawal, please see our Privacy Policy .

Reviewer #1: No

**********

[NOTE: If reviewer comments were submitted as an attachment file, they will be attached to this email and accessible via the submission site. Please log into your account, locate the manuscript record, and check for the action link "View Attachments". If this link does not appear, there are no attachment files.]

While revising your submission, please upload your figure files to the Preflight Analysis and Conversion Engine (PACE) digital diagnostic tool, https://pacev2.apexcovantage.com/ . PACE helps ensure that figures meet PLOS requirements. To use PACE, you must first register as a user. Registration is free. Then, login and navigate to the UPLOAD tab, where you will find detailed instructions on how to use the tool. If you encounter any issues or have any questions when using PACE, please email PLOS at figures@plos.org . Please note that Supporting Information files do not need this step.

PLoS One. 2025 Jun 3;20(6):e0318135. doi: 10.1371/journal.pone.0318135.r003

Author response to Decision Letter 0


25 Apr 2025

Manuscript number: [PONE-D-25-01821] - [EMID:c96f77a474010caa]

Title: Antibacterial, antibiofilm and anti-quorum sensing activities of 1,2,3,5-tetrazine derivatives linked to a benzothiazole moiety

Answers to Reviewers and Editor Comments

Additional Editor Comments:

The article titled Antibacterial, antibiofilm and anti-quorum sensing activities of 1,2,3,5-tetrazine derivatives linked to a benzothiazole moiety, is a work that evaluates a series of organic compounds derived from chemical synthesis conducted by the group of authors. Specifically, the authors evaluate their synthesis products in two related bacterial behavior systems, which are quorum sensing communication and biofilm formation. The evaluations are conducted on a series of bacterial pathogens or pathogenicity models such as: Klebsiella aerogenes ATCC 130148, Acinetobacter baumannii ATCC BAA-1605, Enterococcus faecium ATCC700221, Staphylococcus epidermidis. Additionally, the ability to inhibit the quorum sensing mechanism is evaluated using the production of violacein in Chromobacterium violaceum ATCC12472 as a model. To methodologically relate their work, the authors propose an experimental design based on determining the respective MICs and MIBCs of each compound in concentrations of µg/ml. The results of the work are expressed in percentage values through known formulas.

The results are consistent with the experimental design; however, they present a methodological question that must be clarified by the authors. Specifically, if bacteria need a specific concentration to perform QS communication, how does this relate to the concentrations related to MICs and MIBCs used for each evaluated triazine? The same is considered for the formation or eradication of biofilms. This must be adequately clarified in the methodology section and in the results, with a special mention in the discussion section.

Finally, although the article is coherent and it is positively valued to work with compounds derived from synthesis, it is required to delve deeper methodologically or in the discussion regarding the molecular and chemical mechanisms that may be involved in the observed results.

Response: We sincerely appreciate the Editor’s thoughtful comments and suggestions for improving our manuscript. We have now clarified in the methodology section that the sub-inhibitory concentrations tested (1/2x, 1/4x, 1/8x, 1/16x, and 1/32x MIC) were specifically selected to evaluate bacterial growth and biofilm formation while avoiding growth effects. We have also expanded the discussion to include potential molecular targets of the tested compounds.

The manuscript must be presented in the format required by PLOS.

Response: We have now presented the manuscript in the format required by PLOS. The changes have been colored red with yellow stripes across the manuscript.

Reviewers' comments:

Comments to the Author

Reviewer #1:

Major comments

There are some major methodological issues regarding the evaluation of biofilm inhibition and violacein production. While authors test different concentrations of the selected compounds below MIC and MBC, they do not take into consideration the potential effect of subinhibitory concentrations of these on bacterial growth. Therefore, it is not possible to determine if the lower biofilm formation and violacein production is due to the selective effect of the compounds on these cellular processes or to a reduction of bacterial cells. Normalization by total cell number (CFU or Optical density at 600 nm) or growth curves at the selected concentrations of the compounds should be included to support the conclusions.

Response: We appreciate the reviewer’s insightful comment regarding the potential effect of subinhibitory concentrations of the tested compounds on bacterial growth. To clarify this point, we provided data of the kinetics of bacterial growth and biofilm formation in response to subinhibitory concentrations of the tested compounds, alongside our biofilm and violacein production experiments. We have now explicitly discussed this point in the revised manuscript to ensure clarity for readers. The changes have been colored red with yellow stripes in the main text. Thank you for raising this important consideration.

Minor comments:

Introduction, L5: change "The main mechanisms of ..... includes" for "include" (plural)

Introduction, L7: "degradation" may be replace bt a more accurate concept as "structural modification"

Response: The appropriate correction has been made.

Methods, 2.2.1: Please indicate why C. violaceum was cultured at 30 ºC instead of 26 ºC (its optimal growth temperature).

Response: The mistake has been corrected.

Methods 2.2.3: Delete ")" after gentamicin.

Please specify gentamicin concentrations used as control condition.

Response: The gentamicin range of concentrations used has been provided.

"The MBC was determined as the ...that completely inhibited the growth of bacteria". Because authors are evaluating bactericidal effect, the more accurate concept should be "that inhibited bacterial viability".

Response: The appropriate correction has been made.

Methods, 2.3.1. Please specify vanillin concentration

Response: The appropriate correction has been made.

Results, 3.1.2: Authors should include ATCC numbers for all strains (or none).

Response: ATCC numbers are now provided only once, in the methodology section.

Results, 3.4.1, Figure 2. According to the figure and its Y-axis, lower concentrations of the selected compouds produced higher ihnibition of violacein production. Please check this information and plot´s axis names.

Response: We appreciate the reviewer’s comments and suggestions for improving our manuscript. The mistake has been corrected. Y-axis of Figure 2 is the % of violacein production, where higher concentrations of the selected compounds produced lower violacein production.

Discussion "By contrast, compound 4c bears only two nitro groups, whereas compound 4c do not contain any nitro moiety". Authors mention compound 4c twice, please check.

Response: We agree with the reviewer’s inquisitiveness. The change has now been amended as suggested.

Conclusion. Authors do not expose the conclusion of their research. Instead, in this section, they summarize the relevance and of their results. Please, include the conclusions.

Response: The conclusion section has now been re-written as suggested.

We request the Editorial office for re-evaluation and kind consideration.

Attachment

Submitted filename: Response to the reviewers comments of 24th April.docx

pone.0318135.s004.docx (20.1KB, docx)

Decision Letter 1

M Alejandro Dinamarca

6 May 2025

Antibacterial, antibiofilm and anti-quorum sensing activities of 1,2,3,5-tetrazine derivatives linked to a benzothiazole moiety

PONE-D-25-01821R1

Dear 

Dr. Boniface Pone Kamdem,

We’re pleased to inform you that your manuscript has been judged scientifically suitable for publication and will be formally accepted for publication once it meets all outstanding technical requirements.

Within one week, you’ll receive an e-mail detailing the required amendments. When these have been addressed, you’ll receive a formal acceptance letter and your manuscript will be scheduled for publication.

An invoice will be generated when your article is formally accepted. Please note, if your institution has a publishing partnership with PLOS and your article meets the relevant criteria, all or part of your publication costs will be covered. Please make sure your user information is up-to-date by logging into Editorial Manager at Editorial Manager®  and clicking the ‘Update My Information' link at the top of the page. If you have any questions relating to publication charges, please contact our Author Billing department directly at authorbilling@plos.org.

If your institution or institutions have a press office, please notify them about your upcoming paper to help maximize its impact. If they’ll be preparing press materials, please inform our press team as soon as possible -- no later than 48 hours after receiving the formal acceptance. Your manuscript will remain under strict press embargo until 2 pm Eastern Time on the date of publication. For more information, please contact onepress@plos.org.

Kind regards,

M. Alejandro Dinamarca, Dr.

Academic Editor

PLOS ONE

Additional Editor Comments (optional):

Reviewers' comments:

Acceptance letter

M Alejandro Dinamarca

PONE-D-25-01821R1

PLOS ONE

Dear Dr. Pone Kamdem,

I'm pleased to inform you that your manuscript has been deemed suitable for publication in PLOS ONE. Congratulations! Your manuscript is now being handed over to our production team.

At this stage, our production department will prepare your paper for publication. This includes ensuring the following:

* All references, tables, and figures are properly cited

* All relevant supporting information is included in the manuscript submission,

* There are no issues that prevent the paper from being properly typeset

You will receive further instructions from the production team, including instructions on how to review your proof when it is ready. Please keep in mind that we are working through a large volume of accepted articles, so please give us a few days to review your paper and let you know the next and final steps.

Lastly, if your institution or institutions have a press office, please let them know about your upcoming paper now to help maximize its impact. If they'll be preparing press materials, please inform our press team within the next 48 hours. Your manuscript will remain under strict press embargo until 2 pm Eastern Time on the date of publication. For more information, please contact onepress@plos.org.

If we can help with anything else, please email us at customercare@plos.org.

Thank you for submitting your work to PLOS ONE and supporting open access.

Kind regards,

PLOS ONE Editorial Office Staff

on behalf of

Mr M. Alejandro Dinamarca

Academic Editor

PLOS ONE

Associated Data

    This section collects any data citations, data availability statements, or supplementary materials included in this article.

    Supplementary Materials

    S1 Fig. Effect of selected compounds at sub-inhibitory concentrations on biofilm and bacterial growth inhibition.

    A: effect of compound 4a, B: effect of compound 4b, C: effect of compound 4c, MIC: minimum inhibitory concentration.

    (DOCX)

    pone.0318135.s001.docx (128.1KB, docx)
    S2 Fig. Effect of selected compounds 4a, 4b and4c onChromobacterium violaceum growth at sub-inhibitory concentrations.

    MIC: minimum inhibitory concentration.

    (DOCX)

    pone.0318135.s002.docx (32.4KB, docx)
    Attachment

    Submitted filename: Response to the reviewers comments of 24th April.docx

    pone.0318135.s004.docx (20.1KB, docx)

    Data Availability Statement

    All relevant data are within the manuscript and its Supporting Information files.


    Articles from PLOS One are provided here courtesy of PLOS

    RESOURCES