Next Article in Journal
Nanocrystalline TiO2 Composite Films for the Photodegradation of Formaldehyde and Oxytetracycline under Visible Light Irradiation
Previous Article in Journal
The Complete Chloroplast Genome Sequences of Fritillaria ussuriensis Maxim. and Fritillaria cirrhosa D. Don, and Comparative Analysis with Other Fritillaria Species
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Synthesis and Bioactivity Evaluation of N-Arylsulfonylindole Analogs Bearing a Rhodanine Moiety as Antibacterial Agents

Basic Medical and Pharmacy College, Jinggangshan University, Ji’an 343009, Jiangxi, China
*
Author to whom correspondence should be addressed.
Molecules 2017, 22(6), 970; https://doi.org/10.3390/molecules22060970
Submission received: 28 April 2017 / Revised: 6 June 2017 / Accepted: 9 June 2017 / Published: 14 June 2017

Abstract

:
Due to the rapidly growing bacterial resistance to antibiotics and the scarcity of novel agents under development, bacterial infections are still a pressing global problem, making new types of antibacterial agents, which are effective both alone and in combination with traditional antibiotics, urgently needed. In this paper, seven series of N-arylsulfonylindole analogs 511 bearing rhodanine moieties were synthesized, characterized, and evaluated for antibacterial activity. According to the in vitro antimicrobial results, half of the synthesized compounds showed potent inhibition against four Gram-positive bacteria, with MIC values in the range of 0.5–8 µg/mL. For multidrug-resistant strains, compounds 6a and 6c were the most potent, with MIC values of 0.5 µg/mL, having comparable activity to gatifloxacin, moxiflocaxin and norfloxacin and being 128-fold more potent than oxacillin (MIC = 64 µg/mL) and 64-fold more active than penicillin (MIC = 32 µg/mL) against Staphylococcus aureus ATCC 43300.

1. Introduction

Bacterial drug resistance has emerged and become increasingly serious on a global scale, both in developed and developing countries [1]. Drug-resistant bacteria, such as methicillin-resistant Staphylococcus aureus (MRSA), multi-drug resistant Escherichia coli, and multi-drug resistant Pseudomonas aeruginosa, cause lethal diseases and cause great difficulties in the treatment of nosocomial infections [2,3,4,5], which severely threaten global public health while resulting in very large economic costs [6]. Currently the development of new antimicrobial drugs cannot keep pace with the development of bacterial drug resistance, and the number of new antibiotics approved for marketing per year is declining continuously. Taking advantage of pathogens virulence is an alternative strategy to inhibit antibiotic resistance development, and some studies have focused on anti-virulence agents against Gram-positive pathogens and Gram-negative pathogens [7,8]. However, this strategy has some weaknesses that limiting its development, such as the lack of a good way to assessing the efficacy and the inapplicability in immunocompromised patients. Over the last decade, a few new antibiotics (e.g., linezolid, ceftolozane, telavancin, ceftaroline fosamil, Xifaxan®, and daptomycin) were approved and dozens of antibiotics are currently in Phase 2 or Phase 3 clinical trials [9]. However, these drugs cannot address the entire spectrum of bacteria resistance [10], therefore, there is an urgent need to develop new antimicrobial agents, especially those with a new drug target or with the ability to overcome drug resistance.
Indole, an intercellular signaling molecule, regulates various aspects of bacterial physiology, including spore formation, plasmid stability, resistance to drugs, biofilm formation, and virulence. The amino acid tryptophan is an indole derivative and the precursor of the neurotransmitter serotonin. Up to now, indoles have displayed important physiological functions and potent pharmacological activities, including anti-inflammatory and antioxidant [11], antineoplastic [12,13,14], antimicrobial [15,16], antiviral [14,17,18], and anti-HIV activity [19,20]. N-Arylsulfonylindoles, as a kind of indole derivative, have received a great deal of attention in the field of chemical drug research, behaving as 5-HT6 receptor antagonists [21], anti-AIDS drugs [22], and antifungal agents [23]. However, the antibacterial activity of N-arylsulfonylindoles has not yet been reported to our knowledge.
Previously, we reported a series of rhodanine derivatives (compounds I, Scheme 1) which showed good inhibitory activity against Gram-positive bacteria (including multidrug-resistant clinical isolates) [24,25,26,27,28,29], which suggested that the rhodanine moiety was an important fragment for antimicrobial activity. Fragment-based drug discovery and skeleton migration strategy is a rational technique for molecular modification and drug design when some active fragments are obtained. Based upon these observations, and as part of our ongoing program aiming at the discovery and development of bioactive molecules, in this work, seven series of N-arylsulfonyl-3-substituted indoles 511 (Scheme 1) were designed using compound I as the lead compound. The target compounds were prepared by combining the rhodanine groups with N-arylsulfonylindoles, and their anti-bacterial activities were screened.
Chen et al. [30] have reported the synthesis and antibacterial activity evaluation of chalcone derivatives containing a rhodanine-3-acetic acid moiety, and found that the antibacterial activity of compounds with halogen and methyl substituents on the phenyl group was obviously higher than that of compounds with other substituents. In view of these findings, in this paper, only halogen and methyl substituents were chosen on the indole and phenyl sulfonyl moieties.

2. Results and Discussion

2.1. Chemistry

The target compounds were synthesized as outlined in Scheme 2. A series of 3-substituted-5-((1-(phenylsulfonyl)-1H-indol-3-yl)methylene)-2-thioxothiazolidin-4-ones were synthesized using 1H-indole-3-carbaldehydes as the starting material. Firstly, benzenesulfonyl chlorides 1a, 1b were reacted with 1H-indole-3-carbaldehydes 2a2c in the presence of anhydrous potassium carbonate (K2CO3) at 40 °C to give 1-(phenylsulfonyl)-1H-indole-3-carbaldehydes 3af, which were directly used in the next step without purification. Next compounds 3af were subjected to a Knoevenagel condensation reaction with appropriate N-substituted rhodanines to provide seven new series of target compounds 511. The structures of the products were well characterized by 1H-NMR, 13C-NMR, and high-resolution mass spectrometry.

2.2. Antimicrobial Activity

All of the target compounds 511 were evaluated for their in vitro anti-bacterial activity using a serial dilution method to obtain the minimum inhibitory concentration (MIC) against five Gram-positive strains (S. aureus (CMCC(B) 26003 and CMCC 25923, Streptococcus pyogenes CMCC 32067, Enterococcus faecalis CMCC 29212, and Bacillus subtilis CMCC 63501), four Gram-negative strains (E. coli (CMCC 25922 and CMCC 44568) and P. aeruginosa (CMCC 27853 and CMCC 10104))m as well as two methicillin-resistant clinical isolates (S. aureus ATCC 43300 and ATCC 33591). Gatifloxacin, moxifloxacin, norfloxacin, oxacillin, and penicillin were used as positive control drugs.
Preliminarily, compounds 511 were screened for their activity against five Gram-positive strains and four Gram-negative strains. Initial screening results described as MIC values are presented in Table 1. The results illustrate that the inhibition of the seven series of derivatives against Gram-positive strains (effective against four bacteria) is in general superior to that of Gram-negative strains (effective against one bacterium). For Gram-positive strains, more than half of the tested compounds showed potent inhibition activity against S. aureus (CMCC(B) 26003 and CMCC 25923), with MIC values in the range of 0.5–4 µg/mL. Half of the target compounds exhibited moderate activity against E. faecalis CMCC 29212 (MIC = 4–32 µg/mL) and S. pyogenes CMCC 32067 (MIC = 2–128 µg/mL). Conversely, five positive control agents did not exhibit inhibition activity for the two strains (MICs > 128 µg/mL). All compounds, however, had no effect on B. subtilis CMCC 63501 at 128 µg/mL. For Gram-negative strains, only a few of compounds showed moderate activity against P. aeruginosa CMCC 10104 with MICs of 4–64 µg/mL, while showing no inhibitory activity against three other strains at 128 µg/mL. The study found that, among the compounds synthesized, compound 8b was the most active compound against two S. aureus with MIC values of 1 or 0.5 µg/mL, along with MIC = 2 or 4 µg/mL against S. pyogenes CMCC 32067 and E. faecalis CMCC 29212, respectively.
In the following trials, five compounds (6a, 6c, 8a–c) were chosen to evaluate their inhibitory activity against two clinical isolates of multidrug-resistant Gram-positive bacterial strains (S. aureus ATCC 43300 and S. aureus ATCC 33591), whose MICs against S. aureus (CMCC(B) 26003 or CMCC 25923 are less than 1 µg/mL. The results are listed in Table 2. The data illustrated that five compounds had excellent inhibitory activities against the two multidrug-resistant strains, with MICs of 0.5, 1, or 2 µg/mL. Among them, compounds 6a and 6c were the most potent, with MIC values of 0.5 µg/mL, having comparable activity to gatifloxacin, moxiflocaxin and norfloxacin, while being 128-fold more potent than oxacillin (MIC = 64 µg/mL) and 64-fold more active than penicillin (MIC = 32 µg/mL) against S. aureus ATCC 43300.

2.3. Cytotoxicity

The cytotoxic properties of compounds 6a, 8b, and 8c were also investigated on HEK 293T cells using the CCK-8 method and the results are shown in Table 3. Compounds 6a, 8b, and 8c, with IC50 values of 36.90, 54.09, and 32.28 µg/mL, respectively, were not cytotoxic at concentrations in the range of 0.5–16 µg/mL. The comparison between the MIC and IC50 values of the tested compounds suggests that compounds 6a, 8b, and 8c exhibit in vitro antibacterial activity at non-cytotoxic concentrations.

2.4. The Structure-Activity Relationships (SARs) Analysis

Based on the present data of the synthesized compounds, simple SARs could be proposed. For the derivatives bearing weak electron-donating substituents (R1 = -CH3), it seems that there is no obvious impact on the antibacterial activity in comparison with non-substituted compounds (R1 = H). In series 6, for example, the MIC values against CMCC 26003 of compound 6c (R1 = -CH3) and compound 6a (R1 = H) were both 2 µg/mL, and the same result was discovered in the series 7. As evidenced from Table 1, this could lead to the conclusion that a general inhibitory activity order of seven series of target compounds was series 6, 8 > series 7, 10 > series 9 > series 5, 11, but the differences are not remarkable. Upon comparison of series 7 and 8, it can be found that the activity of the R-configuration compounds appears to be slightly better than that of S-configuration compounds.

3. Materials and Methods

3.1. Instruments and Reagents

All of the reagents and solvents were purchased from Aladdin (Shanghai, China) or Sinopharm Chemical Reagent Co. Ltd. (Shanghai, China), and were used as received. Melting points were determined in open capillary tubes and are uncorrected. Reaction courses were monitored by thin-layer chromatography on silica gel-precoated F254 plates (Merck, Darmstadt, Germany). Developed plates were examined with UV lamps (254 nm). Nuclear magnetic resonance spectroscopy was performed on an AV-300 spectrometer (Bruker, Zurich, Switzerland) operating at 300 MHz for 1H and 75 MHz for 13C and using DMSO-d6 as solvent and tetramethylsilane as the internal standard. Electrospray Ionization Mass Spectrometry (ESI-MS) experiments were performed on an IT-TOF mass spectrometer (Shimadzu, Tokyo, Japan) in negative ion mode. Specific optical rotation was measured on a Digital automatic polariscope JASCO P-1020 (JASCO, Tokyo, Japan).

3.2. Synthesis Method and Spectral Data

3.2.1. General Procedure for the Preparation of Compounds 3a3b

To a dry dichloromethane solution (10 mL) of the appropriate 1H-indole-3-carbaldehydes (2 mmol), anhydrous potassium carbonate (6 mmol) and benzenesulfonyl chlorides (4 mmol) in dry dichloromethane (20 mL) were added, and the mixture was stirred for 12 h at 40 °C. After the completion of the reaction, excess solvent was removed under reduced pressure to obtain a yellow crude solid of 3a3b which was directly used in the next step without purification.

3.2.2. General Procedure for the Preparation of Compounds 3c3f

To a solution of the appropriate 1H-indole-3-carbaldehydes (1 mmol) in dry dichloromethane (30 mL), sodium hydroxide (1.75 mmol), benzyltriethylammonium chloride (TEBA, 0.1 mmol), and benzenesulfonyl chlorides (1.2 mmol) were added and stirred for 12 h at room temperature. After the completion of the reaction, 15 mL water was added into the mixture. Then the mixture was extracted with dichloromethane (30 mL × 3). The combined organic layers were dried over anhydrous MgSO4 before being concentrated in vacuo. The crude products 3c3f obtained were directly used in the next step without purification.

3.2.3. General Procedure for the Preparation of Compounds 511

A mixture of 3 (1 mmol), corresponding rhodanine (1 mmol), 10 drops glacial acetic acid and 10 drops piperidine in ethanol (20 mL) was refluxed for 16 h. After cooling, the solvent was evaporated in vacuo, followed by the purification of the resulting residue by silica gel column chromatography (dichloromethane/methanol = 100/1 or 150/1) to obtain a yellow solid 511.

3.2.4. Spectral Data

2-(5-((5-Bromo-1-(phenylsulfonyl)-1H-indol-3-yl)methylene)-4-oxo-2-thioxothiazolidin-3-yl)acetic acid (5a). Yellow solid; yield 45%; m.p. 242–246 °C. 1H-NMR: δ 4.64 (s, 2H, NCH2), 7.65–8.38 (m, 10H, Ar-H, CH=C), 12.80 (br.s, 1H, COOH). 13C-NMR: δ 192.57, 167.35, 166.27, 136.47, 135.98, 133.12, 131.05, 130.66, 129.50, 129.00, 127.79, 127.75, 125.95, 123.79, 117.92, 116.80, 115.59, 53.18. ESI-HRMS calcd. for C20H12BrN2O5S3 ([M − H]): 534.9097; found: 534.9118.
2-(5-((6-Chloro-1-(phenylsulfonyl)-1H-indol-3-yl)methylene)-4-oxo-2-thioxothiazolidin-3-yl)acetic acid (5b). Yellow solid; yield 47%; m.p. 206–208 °C. 1H-NMR: δ 4.40 (s, 2H, NCH2), 7.44–8.24 (m, 10H, Ar-H, CH=C), 9.24 (br.s, 1H, COOH). 13C-NMR: δ 192.54, 167.13, 166.62, 136.53, 136.02, 134.67, 131.51, 130.75, 128.35, 127.99, 127.76, 125.39, 124.67, 122.56, 121.59, 117.33, 113.31, 43.98. ESI-HRMS calcd. for C20H12ClN2O5S3 ([M − H]): 490.9602; found: 490.9615.
2-(5-((5-Bromo-1-tosyl-1H-indol-3-yl)methylene)-4-oxo-2-thioxothiazolidin-3-yl)acetic acid (5c). Yellow solid; yield 44%; m.p. 226 °C. 1H-NMR: δ 2.33 (s, 3H, CH3), 4.40 (s, 2H, NCH2), 7.37–8.40 (m, 9H, Ar-H, CH=C), 9.31 (br.s, 1H, COOH). 13C-NMR: δ 194.93, 178.62, 176.66, 147.06, 146.25, 133.97, 133.13, 132.73, 132.63, 131.08, 130.76, 128.74, 127.75, 127.53, 117.81, 116.94, 115.62, 43.85, 22.70. ESI-HRMS calcd. for C21H14BrN2O5S3 ([M − H]): 548.9254; found: 548.9272.
2-(5-((6-Chloro-1-tosyl-1H-indol-3-yl)methylene)-4-oxo-2-thioxothiazolidin-3-yl)acetic acid (5d). Yellow solid; yield 57%; m.p. 205 °C. 1H-NMR: δ 2.35 (s, 3H, CH3), 4.41 (s, 2H, NCH2), 7.42–8.11 (m, 9H, Ar-H, CH=C), 9.28 (br.s, 1H, COOH). 13C-NMR: δ 192.52, 167.45, 166.62, 147.13, 134.63, 133.57, 131.44, 131.16, 128.36, 127.96, 127.79, 125.29, 124.48, 122.46, 121.63, 117.15, 113.30, 43.89, 22.73. ESI-HRMS calcd. for C21H14ClN2O5S3 ([M − H]): 504.9759; found: 504.9774.
(R)-2-(5-((1-(Phenylsulfonyl)-1H-indol-3-yl)methylene)-4-oxo-2-thioxothiazolidin-3-yl)-4-methylpentanoic acid (6a). Yellow solid; yield 51%; m.p. 271–276 °C. [ α ] D 20 : +35 (c = 0.20, DMF). 1H-NMR: δ 0.89 (d, 3H, J = 6.4 Hz, CHCH3), 0.94 (d, 3H, J = 6.4 Hz, CHCH3), 1.35–1.45 (m, 1H, CHCH3), 1.99–2.07 (m, 1H, CH-Ha), 2.20–2.28 (m, 1H, CH-Hb), 5.61 (br.s, 1H, NCH), 7.38–8.20 (m, 11H, Ar-H, CH=C), 13.31 (br.s, 1H, COOH). 13C-NMR: δ 193.05, 169.83, 166.48, 136.66, 135.80, 134.29, 130.56, 129.03, 128.46, 127.72, 126.88, 125.03, 123.82, 122.02, 120.82, 117.21, 113.72, 56.47, 36.88, 25.29, 23.36, 22.37. ESI-HRMS calcd. for C24H21N2O5S3([M − H]): 513.0618; found: 513.0629.
(R)-2-(5-((6-Chloro-1-(phenylsulfonyl)-1H-indol-3-yl)methylene)-4-oxo-2-thioxothiazolidin-3-yl)-4-methyl-pentanoic acid (6b). Yellow solid; yield 53%; m.p. 180–181 °C. [ α ] D 20 : +62.5 (c = 0.20, DMF). 1H-NMR: δ 0.86 (d, 3H, J = 6.6 Hz, CHCH3), 0.91 (d, 3H, J = 6.6 Hz, CHCH3), 1.33–1.38 (m, 1H, CHCH3), 2.00–2.07 (m, 1H, CH-Ha), 2.24–2.31 (m, 1H, CH-Hb), 5.50 (br.s, 1H, NCH), 7.20–8.22 (m, 10H, Ar-H, CH=C), 9.30 (br.s, 1H, COOH). 13C-NMR: δ 193.22, 170.01, 166.56, 136.51, 136.00, 134.63, 131.53, 130.70, 129.78, 128.10, 127.76, 125.94, 125.38, 122.47, 120.63, 117.16, 113.29, 56.50, 37.23, 25.57, 23.49, 22.68. ESI-HRMS calcd. for C24H20ClN2O5S3 ([M − H]): 547.0228; found: 547.0244.
(R)-2-(5-((1-Tosyl-1H-indol-3-yl)methylene)-4-oxo-2-thioxothiazolidin-3-yl)-4-methylpentanoic acid (6c). Yellow solid; yield 66%; m.p. 200 °C. [ α ] D 20 +45 (c = 0.10, DMF). 1H-NMR: δ 0.88 (d, 3H, J = 6.4 Hz, CHCH3), 0.93 (d, 3H, J = 6.5 Hz, CHCH3), 1.28–1.33 (m, 1H, CHCH3), 1.97–2.07 (m, 1H, CH-Ha), 2.20–2.27 (m, 1H, CH-Hb), 2.33 (s, 3H, Ph-CH3), 5.61 (br.s, 1H, NCH), 7.37–7.51 (m, 4H, Ar-H), 7.98–8.07 (m, 6H, Ar-H, CH=C), 13.04 (br.s, 1H, COOH). 13C-NMR: δ 193.01, 171.68, 169.83, 157.49, 146.85, 134.27, 133.73, 130.97, 129.02, 128.48, 127.76, 126.80, 124.95, 123.87, 120.75, 117.05, 113.74, 56.46, 36.88, 25.82, 23.36, 22.37, 21.54. ESI-HRMS calcd. for C25H23N2O5S3 ([M − H]): 527.0775; found: 527.0790.
(R)-2-(5-((5-Bromo-1-tosyl-1H-indol-3-yl)methylene)-4-oxo-2-thioxothiazolidin-3-yl)-4-methylpentanoic acid (6d). Yellow solid; yield 61%; m.p. 256–260 °C. [ α ] D 20 : +57.5 (c = 0.08, DMF). 1H-NMR: δ 0.89 (d, 3H, J = 6.3 Hz, CHCH3), 0.94 (d, 3H, J = 6.3 Hz, CHCH3), 1.45–1.53 (m, 1H, CHCH3), 1.99–2.08 (m, 1H, CH-Ha), 2.20–2.26 (m, 1H, CH-Hb), 2.34 (s, 3H, Ph-CH3), 5.62 (br.s, 1H, NCH), 7.43 (d, 2H, J = 7.9 Hz, Ph-H), 7.61 (d, 1H, J = 8.7 Hz, Ph-H), 7.93–8.07 (m, 5H, Ar-H), 8.37 (s, 1H, CH=C), 13.23 (br.s, 1H, COOH). 13C-NMR: δ 193.01, 171.68, 169.83, 157.49, 146.85, 134.27, 133.73, 130.97, 129.02, 128.48, 127.76, 126.80, 124.95, 123.87, 120.75, 117.05, 113.74, 56.46, 36.88, 25.82, 23.36, 22.37, 21.54. ESI-HRMS calcd. for C25H22BrN2O5S3 ([M − H]): 604.9880; found: 604.9899.
(R)-2-(5-((6-Chloro-1-tosyl-1H-indol-3-yl)methylene)-4-oxo-2-thioxothiazolidin-3-yl)-4-methylpentanoic acid (6e). Yellow solid; yield 63%; m.p. 247–249 °C. [ α ] D 20 : +31.5 (c = 0.20, DMF). 1H-NMR: δ 0.88 (d, 3H, J = 6.6 Hz, CHCH3), 0.94 (d, 3H, J = 6.6 Hz, CHCH3), 1.46–1.54 (m, 1H, CHCH3), 1.97–2.07 (m, 1H, CH-Ha), 2.21–2.28 (m, 1H, CH-Hb), 2.34 (s, 3H, Ph-CH3), 5.60 (br.s, 1H, NCH), 7.45 (d, 3H, J = 8.3 Hz, Ar-H), 7.97–8.11 (m, 6H, Ar-H, CH=C), 13.04 (br.s, 1H, COOH). 13C-NMR: δ 193.01, 169.80, 166.44, 147.16, 134.61, 133.54, 131.50, 131.13, 129.00, 127.88, 127.84, 125.34, 123.48, 122.51, 122.42, 116.93, 113.30, 56.46, 36.86, 25.28, 23.36, 22.36, 21.58. ESI-HRMS calcd. for C25H22ClN2O5S3 ([M − H]): 561.0385; found: 561.0406.
(S)-2-(4-Oxo-5-((1-(phenylsulfonyl)-1H-indol-3-yl)methylene)-2-thioxothiazolidin-3-yl)-3-phenylpropanoic acid (7a). Yellow solid; yield 58%; m.p. 156–157 °C. [ α ] D 20 : –150 (c = 0.12, DMF). 1H-NMR: δ 3.56 (d, 2H, J = 7.6 Hz, CHCH2), 5.76 (br.s, 1H, NCH), 7.15–8.19 (m, 16H, Ar-H, CH=C), 8.85 (br.s, 1H, COOH). 13C-NMR: δ 192.67, 169.05, 166.55, 138.07, 136.66, 135.78, 134.23, 130.57, 129.32, 129.03, 128.73, 128.20, 127.74, 126.93, 126.85, 124.99, 122.82, 122.17, 120.77, 117.11, 113.70, 44.05, 22.70. ESI-HRMS calcd. for C27H19N2O5S3 ([M − H]): 547.0462; found: 547.0491.
(S)-2-(5-((6-Chloro-1-(phenylsulfonyl)-1H-indol-3-yl)methylene)-4-oxo-2-thioxothiazolidin-3-yl)-3-phenyl-propanoic acid (7b). Yellow solid; yield 54%; m.p. 220–224 °C. [ α ] D 20 : –144.5 (c = 0.40, DMF). 1H-NMR: δ 3.54 (br.s, 2H, CHCH2), 5.91 (br.s, 1H, NCH), 7.19–8.24 (m, 15H, Ar-H, CH=C), 13.26 (br.s, 1H, COOH). 13C-NMR: δ 192.36, 169.17, 166.37, 137.05, 136.49, 136.03, 134.56, 131.54, 130.70, 129.83, 129.45, 128.93, 128.78, 128.47, 127.85, 127.20, 125.39, 123.23, 122.52, 116.87, 113.25, 44.85, 25.77. ESI-HRMS calcd. for C27H18ClN2O5S3 ([M − H]): 581.0072; found: 581.0094.
(S)-2-(5-((1-Tosyl-1H-indol-3-yl)methylene)-4-oxo-2-thioxothiazolidin-3-yl)-3-phenylpropanoic acid (7c). Yellow solid; m.p. 164–166 °C, yield 55%. [ α ] D 20 : –174.25 (c = 0.40, DMF). 1H-NMR: δ 2.32 (s, 3H, Ph-CH3), 3.56 (d, 2H, J = 7.0 Hz, CHCH2), 5.71 (br.s, 1H, NCH), 7.14–7.50 (m, 9H, Ar-H), 7.90–8.05 (m, 6H, Ar-H, CH=C), 9.10 (br.s, 1H, COOH). 13C-NMR: δ 192.81, 169.00, 166.64, 146.83, 138.35, 134.21, 133.72, 130.98, 129.27, 129.04, 128.70, 128.13, 127.77, 126.84, 126.77, 124.90, 122.62, 122.16, 120.71, 116.99, 113.70, 43.94, 22.70, 21.55. ESI-HRMS calcd. for C28H21N2O5S3 ([M − H]): 561.0618; found: 561.06392.
(S)-2-(5-((5-Bromo-1-tosyl-1H-indol-3-yl)methylene)-4-oxo-2-thioxothiazolidin-3-yl)-3-phenylpropanoic acid (7d). Yellow solid; yield 59%; m.p.217–221 °C. [ α ] D 20 : –216.25 (c = 0.08, DMF). 1H-NMR: δ 2.34 (s, 3H, Ph-CH3), 3.56 (d, 2H, J = 6.8 Hz, CHCH2), 5.65 (br.s, 1H, NCH), 7.15–7.63 (m, 9H, Ar-H), 7.86–8.05 (m, 4H, Ar-H), 8.35 (s, 1H, CH=C). 13C-NMR: δ 191.51, 173.04, 170.18, 148.15, 146.96, 142.46, 133.52, 133.24, 133.06, 131.08, 131.04, 129.33, 129.23, 128.68, 127.92, 127.80, 126.93, 126.82, 121.1, 117.9, 113.5, 44.13, 22.72, 22.17. ESI-HRMS calcd. for C28H20BrN2O5S3 ([M − H]): 638.9723; found: 638.9745.
(R)-2-(4-Oxo-5-((1-(phenylsulfonyl)-1H-indol-3-yl)methylene)-2-thioxothiazolidin-3-yl)-3-phenylpropanoic acid (8a). Yellow solid; yield 65%; m.p. 198–199 °C. [ α ] D 20 : +255 (c = 0.02, DMF). 1H-NMR: δ 3.57 (d, 2H, J = 6.8 Hz, CHCH2), 5.63 (br.s, 1H, NCH), 7.13–8.17 (m, 16H, Ar-H, CH=C), 9.26 (br.s, 1H, COOH). 13C-NMR: δ 192.92, 169.15, 166.77, 139.01, 136.66, 135.76, 134.23, 130.56, 129.17, 129.07, 128.66, 127.93, 127.71, 126.82, 126.66, 124.97, 122.65, 121.98, 120.73, 117.21, 113.69, 43.89, 22.71. ESI-HRMS calcd. for C27H19N2O5S3 ([M − H]): 547.0462; found: 547.0487.
(R)-2-(5-((6-Chloro-1-(phenylsulfonyl)-1H-indol-3-yl)methylene)-4-oxo-2-thioxothiazolidin-3-yl)-3-phenyl-propanoic acid (8b). Yellow solid; yield 61%; m.p. 264–265 °C. [ α ] D 20 : +170.36 (c = 0.28, DMF). 1H-NMR: δ 3.54 (d, 2H, J = 6.9 CHCH2), 5.91 (br.s, 1H, NCH), 7.20–8.23 (m, 15H, Ar-H, CH=C), 12.77 (br.s, 1H, COOH). 13C-NMR: δ 192.35, 169.15, 166.35, 137.00, 136.48, 136.04, 134.57, 131.55, 130.71, 129.45, 128.96, 128.79, 127.85, 127.21, 125.39, 123.29, 122.54, 122.25, 116.86, 113.26, 100.00, 58.76, 29.48. ESI-HRMS calcd. for C27H18ClN2O5S3 ([M − H]): 581.0072; found: 581.0090.
(R)-2-(5-((1-Tosyl-1H-indol-3-yl)methylene)-4-oxo-2-thioxothiazolidin-3-yl)-3-phenylpropanoic acid (8c). Yellow solid; yield 57%; m.p. 176–181 °C. [ α ] D 20 : +178.25 (c = 0.40, DMF). 1H-NMR: δ 2.32 (s, 3H, Ph-CH3), 3.57 (d, 2H, J = 7.1 Hz, CHCH2), 5.71 (br.s, 1H, NCH), 7.14–7.50 (m, 9H, Ar-H), 7.90–8.05 (m, 6H, Ar-H, CH=C), 9.16 (br.s, 1H, COOH). 13C-NMR: δ 191.80, 169.17, 166.65, 146.82, 138.37, 134.21, 133.73, 130.97, 129.26, 129.04, 128.69, 128.12, 127.76, 126.83, 126.76, 124.89, 122.58, 122.17, 120.70, 116.99, 113.70, 43.90, 22.70, 21.54. ESI-HRMS calcd. for C28H21N2O5S3 ([M − H]): 561.0618; found: 561.0643.
(R)-2-(5-((5-Bromo-1-tosyl-1H-indol-3-yl)methylene)-4-oxo-2-thioxothiazolidin-3-yl)-3-phenylpropanoic acid (8d). Yellow solid; yield 62%; m.p. 192–193 °C. [ α ] D 20 : +159.13 (c = 0.80, DMF). 1H-NMR: δ 2.34 (s, 3H, Ph-CH3), 3.56 (d, 2H, J = 6.5 Hz, CHCH2), 5.71 (br.s, 1H, NCH), 7.13–7.20 (m, 5H, Ar-H), 7.43 (d, 2H, J = 8.3 Hz, Ar-H), 7.61 (dd, 1H, J1 = 8.9 Hz, J2 = 1.8 Hz, Ar-H), 7.91–8.06 (m, 5H, Ar-H), 8.37 (s, 1H, CH=C). 13C-NMR: δ 192.71, 169.11, 166.51, 147.05, 138.38, 138.27, 133.52, 133.05, 131.04, 129.39, 129.27, 129.01, 128.70, 127.80, 126.86, 123.67, 122.62, 122.51, 117.80, 116.53, 115.57, 43.94, 22.71, 21.56. ESI-HRMS calcd. for C28H20BrN2O5S3 ([M − H]): 638.9723; found: 638.9740.
(R)-2-(5-((6-Chloro-1-tosyl-1H-indol-3-yl)methylene)-4-oxo-2-thioxothiazolidin-3-yl)-3-phenylpropanoic acid (8e). Yellow solid; yield 55%; m.p. 277–280 °C. [ α ] D 20 : +182.5 (c = 0.80, DMF). 1H-NMR: δ 2.34 (s, 3H, Ph-CH3), 3.56 (d, 2H, J = 7.0 Hz, CHCH2), 5.84 (br.s, 1H, NCH), 7.18–7.45 (m, 8H, Ar-H), 7.92–8.10 (m, 6H, Ar-H). 13C-NMR: δ 192.32, 169.23, 166.43, 147.14, 137.43, 134.54, 133.53, 131.46, 131.12, 129.38, 128.88, 128.76, 127.87, 127.09, 125.30, 122.97, 122.49, 122.34, 116.76, 113.26, 110.60, 33.77, 29.48, 21.58. ESI-HRMS calcd. for C28H20ClN2O5S3 ([M − H]): 595.0228; found: 595.0245.
2-(4-Oxo-5-((1-(phenylsulfonyl)-1H-indol-3-yl)methylene)-2-thioxothiazolidin-3-yl)3-methylbutanoic acid (9). Yellow solid; yield 46%; m.p. 255–257 °C. 1H-NMR: δ 0.78 (d, 3H, J = 5.8 Hz, CHCH3), 1.22 (d, 3H, J = 4.6 Hz, CHCH3), 2.77 (d, 1H, CH(CH3)2), 5.21 (d, 1H, J = 7.5 Hz, NCH), 7.41–8.20 (m, 11H, Ar-H, CH=C), 13.15 (br.s, 1H, COOH). 13C-NMR: δ 193.02, 169.09, 166.53, 136.67, 135.81, 134.29, 130.57, 129.03, 128.57, 127.76, 126.90, 125.04, 124.16, 121.71, 120.83, 117.17, 113.73, 62.69, 27.65, 22.15, 19.41. ESI-HRMS calcd. for C23H19N2O5S3 ([M − H]): 499.0462; found: 499.0479.
(S)-2-(4-Oxo-5-((1-(phenylsulfonyl)-1H-indol-3-yl)methylene)-2-thioxothiazolidin-3-yl)-3-methylpentanoic acid (10). Yellow solid; yield 42%; m.p. 256–260 °C. [ α ] D 20 : –32.5 (c = 0.4, CHCl3). 1H-NMR: δ 0.82 (t, 3H, J = 7.2 Hz, CH2CH3), 0.94–1.01 (m, 1H, CH-Ha), 1.18 (d, 3H, J = 6.5 Hz, CHCH3), 1.47–1.53 (m, 1H, CH-Hb), 2.54–2.59 (m, 1H, CHCH3), 5.25 (d, 1H, J = 7.4 Hz, NCH), 7.39–8.20 (m, 11H, Ar-H, CH=C), 13.18 (br.s, 1H, COOH). 13C-NMR: δ 193.00, 169.09, 166.57, 136.65, 135.81, 134.27, 130.57, 129.02, 128.58, 127.75, 126.89, 125.03, 124.19, 121.66, 120.84, 117.17, 113.71, 62.15, 33.53, 25.37, 18.05, 11.34. ESI-HRMS calcd. for C24H21N2O5S3 ([M − H]): 513.0629; found: 513.0618.
(S)-3-(4-Hydroxyphenyl)-2-(4-oxo-5-((1-(phenylsulfonyl)-1H-indol-3-yl)methylene)-2-thioxothiazolidin-3-yl)propanoic acid (11). Yellow solid; yield 47%; m.p. 167–181 °C. [ α ] D 20 : –150 (c = 0.2, DMF). 1H-NMR: δ 3.46 (br.s, 2H, CHCH2), 5.69 (br.s, 1H, NCH), 6.59 (d, 2H, J = 8.1 Hz, Ar-H), 6.94 (d, 2H, J = 8.1 Hz, Ar-H), 7.38–8.19 (m, 11H, Ar-H, CH=C), 9.13 (s, 1H, OH). 13C-NMR: δ 192.67, 170.93, 166.57, 156.25, 136.66, 135.77, 134.22, 130.70, 130.56, 130.18, 129.05, 128.69, 128.16, 127.95, 127.75, 126.83, 124.98, 120.75, 117.10, 115.57, 113.69, 44.84, 25.78. ESI-HRMS calcd. for C27H19N2O6S3 ([M − H]): 563.0411; found: 563.0430.

3.3. Evaluation of Anti-Bacterial Activity In Vitro

The anti-bacterial activity in vitro against S. aureus (CMCC(B) 26003 and CMCC 25923, S. pyogenes CMCC 32067, E. faecalis CMCC 29212, B. subtilis CMCC 63501; E. coli CMCC 25922 and CMCC 44568, P. aeruginosa CMCC 27853 and CMCC 10104, as well as two methicillin-resistant clinical isolates (S. aureus ATCC 43300 and ATCC 33591) was evaluated using a two-fold serial dilution technique [31], and the final concentrations of compounds obtained were in the range of 0.5–128 μg/mL. Test bacteria were grown to mid-log phase in Mueller-Hinton broth (MHB) or Tryptone Soya Broth (TSB) and diluted 1000-fold in the same medium. The bacteria of 105 CFU/mL were inoculated into MHB or TSB and dispensed at 0.2 mL/well in a 96-well microtiter plate. As positive controls, gatifloxacin, moxifloxacin, norfloxacin, oxacillin, and penicillin were used. Test compounds were prepared in DMSO, the final concentration of which did not exceed 0.05%. The MIC was defined as the concentration of a test compound that completely inhibited bacteria growth during 24 h incubation at 37 °C. Bacteria growth was determined by measuring the absorption at 630 nm using a microtiter enzyme-linked immunosorbent assay (ELISA) reader. All experiments were carried out three times.

3.4. Evaluation of Cytotoxicity In Vitro

HEK 293T cells were used to test the cytotoxicity of the new compounds. HEK 293T cells were grown in Dulbecco modified Eagle medium supplemented with fetal bovine serum (10%), and antibiotics (penicillin-streptomycin mixture (100 U/mL)). Cells at 80% to 90% confluence were split by trypsin (0.25% in PBS; pH 7.4), and the medium was changed at 24 h intervals. The cells were cultured at 37 °C in a 5% CO2 incubator. The cells were grown to three passages, and approximately 1 × 104 cells were seeded into each well of a 96-well plate and allowed to incubate to allow attachment of the cells to the substrate. After 24 h, the medium was replaced with DMEM supplemented with 10% FBS containing various concentrations (4, 8, 16, 32, 64, 128 μg/mL) of test compounds and incubated for 48 h. Each concentration set three wells in parallel. Then 20 µL of CCK-8 solution was added to each well. After incubation for 3 h, the optical density was measured at 450 nm using a microtiter ELISA reader. The IC50 values were defined as the concentrations inhibiting 50% of cell growth.

4. Conclusions

In summary, seven new series of N-arylsulfonylindoles 511 bearing rhodanine moieties were designed, synthesized, and evaluated for their antibacterial activities. In accordance to the results of antibacterial tests in vitro, some of the compounds showed good antibacterial activities against Staphylococcus aureus, including multidrug-resistant strains. Among them compounds 6a and 6c showed the most potent levels of activity (MIC = 0.5 µg/mL) against selected MRSA strains. These results illustrate that N-arylsulfonylindole analogs bearing rhodanine moieties are promising leads to develop novel antimicrobial agents against many infections caused by Gram-positive strains, especially Staphylococcus aureus, incluing MRSA. Future studies will focus on the mechanism of action of these compounds.

Acknowledgments

This work was supported by the National Science Foundation of China (No. 81560561) and Natural Science Foundation of Jiangxi Province, China (No. 20161BAB215207).

Author Contributions

Xian-Qing Deng designed experiments and conducted the spectrum analysis of target compounds; Wen-Hui Xu, Shao-Feng Xiong synthesized all the presented compounds; Ming-Xia Song, Song-Hui Li, Jiao-Yang Peng, and Ting-Ting Guo conducted the antimicrobial activity determination; Ming-Xia Song wrote the article, and made the discussion and conclusions.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Livermore, D.M. Has the era of untreatable infections arrived? J. Antimicrob. Chemother. 2009, 64 (Suppl. 1), i29–i36. [Google Scholar] [CrossRef] [PubMed]
  2. Bi, Y.; Liu, X.X.; Zhang, H.Y.; Yang, X.; Liu, Z.Y.; Lu, J.; Lewis, P.J.; Wang, C.Z.; Xu, J.Y.; Meng, Q.G.; et al. Synthesis and Antibacterial Evaluation of Novel 3-Substituted Ocotillol-Type Derivatives as Leads. Molecules 2017, 22, 590. [Google Scholar] [CrossRef] [PubMed]
  3. Carrel, M.; Perencevich, E.N.; David, M.Z. USA300 Methicillin-Resistant Staphylococcus aureus, United States, 2000–2013. Emerg. Infect. Dis. 2015, 21, 1973–1980. [Google Scholar] [CrossRef] [PubMed]
  4. Hvistendahl, M. Public health. China takes aim at rampant antibiotic resistance. Science 2012, 336, 795. [Google Scholar] [CrossRef] [PubMed]
  5. Yezli, S.; Li, H. Antibiotic resistance amongst healthcare-associated pathogens in China. Int. J. Antimicrob. Agents 2012, 40, 389–397. [Google Scholar] [CrossRef] [PubMed]
  6. Azeredo da Silveira, S.; Perez, A. Liposomes as novel anti-infectives targeting bacterial virulence factors? Expert. Rev. Anti-Infect. Ther. 2015, 13, 531–533. [Google Scholar] [CrossRef] [PubMed]
  7. Cascioferro, S.; Cusimano, M.G.; Schillaci, D. Antiadhesion agents against Gram-positive pathogens. Future Microbiol. 2014, 9, 1209–1220. [Google Scholar] [CrossRef] [PubMed]
  8. Tay, S.B.; Yew, W.S. Development of quorum-based anti-virulence therapeutics targeting Gram negative bacterial pathogens. Int. J. Mol. Sci. 2013, 14, 16570–16599. [Google Scholar] [CrossRef] [PubMed]
  9. Rademacher, J.; Welte, T. New antibiotics - standstill or progress. Med. Klin. Intensivmed. Notfmed. 2017, 112, 206–213. [Google Scholar] [CrossRef] [PubMed]
  10. Boucher, H.W.; Talbot, G.H.; Benjamin, D.K.; Bradley, J.; Guidos, R.J.; Jones, R.N.; Murray, B.E.; Bonomo, R.A.; Gilbert, D. Infectious Diseases Society of America. 10 x '20 Progress--development of new drugs active against gram-negative bacilli: an update from the Infectious Diseases Society of America. Clin. Infect. Dis. 2013, 56, 1685–1694. [Google Scholar] [CrossRef] [PubMed]
  11. Bhale, P.S.; Chavan, H.V.; Dongare, S.B.; Shringare, S.N.; Mule, Y.B.; Nagane, S.S.; Bandgar, B.P. Synthesis of extended conjugated indolyl chalcones as potent anti-breast cancer, anti-inflammatory and antioxidant agents. Bioorg. Med. Chem. Lett. 2017, 27, 1502–1507. [Google Scholar] [CrossRef] [PubMed]
  12. Diao, P.C.; Li, Q.; Hu, M.J.; Ma, Y.F.; You, W.W.; Hong, K.H.; Zhao, P.L. Synthesis and biological evaluation of novel indole-pyrimidine hybrids bearing morpholine and thiomorpholine moieties. Eur. J. Med. Chem. 2017, 13, 110–118. [Google Scholar] [CrossRef] [PubMed]
  13. Manuel-Manresa, P.; Korrodi-Gregório, L.; Hernando, E.; Villanueva, A.; Martínez-García, D.; Rodilla, A.M.; Ramos, R.; Fardilha, M.; Moya, J.; Quesada, R.; et al. Novel indole-based tambjamine-analogues induce apoptotic lung cancer cell death through p38 mitogen-activated protein kinase activation. Mol. Cancer. Ther. 2017. (In press) [Google Scholar] [CrossRef] [PubMed]
  14. Barbosa, V.A.; Baréa, P.; Mazia, R.S.; Ueda-Nakamura, T.; Costa, W.F.; Foglio, M.A.; Goes Ruiz, A.L.; Carvalho, J.E.; Vendramini-Costa, D.B.; Nakamura, C.V.; et al. Synthesis and evaluation of novel hybrids β-carboline-4-thiazolidinones as potential antitumor and antiviral agents. Eur. J. Med. Chem. 2016, 124, 1093–1104. [Google Scholar] [CrossRef] [PubMed]
  15. Kumar, V.P.; Renjitha, J.; Salfeena, C.F.; Ashitha, K.T.; Keri, R.S.; Varughese, S.; Sasidhar, B.S. Antibacterial and anti-tubercular evaluation of Dihydronaphthalenone-indole hybrid analogues. Chem. Biol. Drug. Des. 2017. (In press) [Google Scholar] [CrossRef]
  16. Patil, S.A.; Patil, S.A.; Patil, R. Medicinal applications of (benz)imidazole- and indole-based macrocycles. Chem. Biol. Drug. Des. 2017, 89, 639–649. [Google Scholar] [CrossRef] [PubMed]
  17. Tan, M.C.; Wong, W.Y.; Ng, W.L.; Yeo, K.S.; Mohidin, T.B.; Lim, Y.Y.; Lafta, F.; Mohd Ali, H.; Ea, C.K. Identification of 5-Methoxy-2-(Diformylmethylidene)-3,3-Dimethylindole as an Anti-Influenza A Virus Agent. PLoS ONE 2017, 12, e0170352. [Google Scholar] [CrossRef] [PubMed]
  18. Scuotto, M.; Abdelnabi, R.; Collarile, S.; Schiraldi, C.; Delang, L.; Massa, A.; Ferla, S.; Brancale, A.; Leyssen, P.; Neyts, J.; et al. Discovery of novel multi-target indole-based derivatives as potent and selective inhibitors of chikungunya virus replication. Bioorg. Med. Chem. 2017, 25, 327–337. [Google Scholar] [CrossRef] [PubMed]
  19. Che, Z.; Tian, Y.; Hu, Z.; Chen, Y.; Liu, S.; Chen, G. Synthesis and in vitro anti-HIV-1 activity of a series of N-arylsulfonyl-3-propionylindoles. Z. Naturforsch. C 2016, 71, 105–109. [Google Scholar] [CrossRef] [PubMed]
  20. Silvestri, R.; De Martino, G.; La Regina, G.; Artico, M.; Massa, S.; Vargiu, L.; Mura, M.; Loi, A.G.; Marceddu, T.; La Colla, P. Novel indolyl aryl sulfones active against HIV-1 carrying NNRTI resistance mutations: Synthesis and SAR studies. J. Med. Chem. 2003, 46, 2482–2493. [Google Scholar] [CrossRef] [PubMed]
  21. Vera, G.; Lagos, C.F.; Almendras, S.; Hebel, D.; Flores, F.; Valle-Corvalán, G.; Pessoa-Mahana, C.D.; Mella-Raipán, J.; Montecinos, R.; Recabarren-Gajardo, G. Extended N-Arylsulfonylindoles as 5-HT6 Receptor Antagonists: Design, Synthesis & amp; Biological Evaluation. Molecules 2016, 21, 1070. [Google Scholar]
  22. Fan, L.L.; Liu, W.Q.; Xu, H.; Yang, L.M.; Lv, M.; Zheng, Y.T. Anti human immunodeficiency virus-1 (HIV-1) agents 3. synthesis and in vitro anti-HIV-1 activity of some N-arylsulfonylindoles. Chem. Pharm. Bull. (Tokyo) 2009, 57, 797–800. [Google Scholar] [CrossRef] [PubMed]
  23. Xu, H.; Wang, Y.Y. Antifungal agents. Part 5: Synthesis and antifungal activities of aminoguanidine derivatives of N-arylsulfonyl-3-acylindoles. Bioorg. Med. Chem. Lett. 2010, 20, 7274–7277. [Google Scholar] [CrossRef] [PubMed]
  24. Song, M.X.; Deng, X.Q.; Li, Y.R.; Zheng, C.J.; Hong, L.; Piao, H.R. Synthesis And biological evaluation of (E)-1-(substituted)-3-phenylprop-2-en-1-ones bearing rhodanines as potent antimicrobial agents. J. Enzym. Inhib. Med. Chem. 2014, 29, 647–653. [Google Scholar] [CrossRef] [PubMed]
  25. Song, M.X.; Zheng, C.J.; Deng, X.Q.; Wei, Z.Y.; Piao, H.R. The Synthesis and Anti-Bacterial Activities of N-carboxymethyl Rhodanines. Med. Chem. 2014, 4, 441–448. [Google Scholar]
  26. Song, M.X.; Zheng, C.J.; Sun, L.P.; Wu, Y.; Hong, L.; Li, Y.J.; Liu, Y.; Wei, Z.Y.; Jin, M.J.; Piao, H.R. Synthesis and antibacterial evaluation of rhodanine-based 5-aryloxy pyrazoles against selected methicillin resistant and quinolone-resistant Staphylococcus aureus (MRSA and QRSA). Eur. J. Med. Chem. 2013, 60, 376–385. [Google Scholar] [CrossRef] [PubMed]
  27. Song, M.X.; Zheng, C.J.; Deng, X.Q.; Wang, Q.; Hou, S.P.; Liu, T.T.; Xing, X.L.; Piao, H.R. Synthesis and bioactivity evaluation of rhodanine derivatives as potential anti-bacterial agents. Eur. J. Med. Chem. 2012, 54, 403–412. [Google Scholar] [CrossRef] [PubMed]
  28. Zheng, C.J.; Song, M.X.; Sun, L.P.; Wu, Y.; Hong, L.; Piao, H.R. Synthesis and biological evaluation of 5-aryloxypyrazole derivatives bearing a rhodanine-3-aromatic acid as potential antimicrobial agents. Bioorg. Med. Chem. Lett. 2012, 22, 7024–7028. [Google Scholar] [CrossRef] [PubMed]
  29. Jin, X.; Zheng, C.J.; Song, M.X.; Wu, Y.; Sun, L.P.; Li, Y.J.; Yu, L.J.; Piao, H.R. Synthesis and antimicrobial evaluation of l-phenylalanine-derived C5-substituted rhodanine and chalcone derivatives containing thiobarbituric acid or 2-thioxo-4-thiazolidinone. Eur. J. Med. Chem. 2012, 56, 203–209. [Google Scholar] [CrossRef] [PubMed]
  30. Chen, Z.H.; Zheng, C.J.; Sun, L.P.; Piao, H.R. Synthesis of new chalcone derivatives containing a rhodanine-3-acetic acid moiety with potential anti-bacterial activity. Eur. J. Med. Chem. 2010, 45, 5739–5743. [Google Scholar] [CrossRef] [PubMed]
  31. Clinical and Laboratory Standards Institute. Methods for Dilution Antimicrobial Susceptibility Tests for Bacteria that Grow Aerobically, Approved Standard M7-A6; Clinical and Laboratory Standards Institute: Wayne, PA, USA, 2003. [Google Scholar]
Sample Availability: Samples of the compounds are available from the authors for a short period of time.
Scheme 1. The design of the target compounds 511.
Scheme 1. The design of the target compounds 511.
Molecules 22 00970 sch001
Scheme 2. The synthesis route of compounds 511.
Scheme 2. The synthesis route of compounds 511.
Molecules 22 00970 sch002
Table 1. Inhibitory activity (MIC, μg/mL) of compounds 511 against Gram-positive and Gram-negative bacteria.
Table 1. Inhibitory activity (MIC, μg/mL) of compounds 511 against Gram-positive and Gram-negative bacteria.
Molecules 22 00970 i023
Compd.R1-R2-R3-Gram-Positive StrainsGram-Negative Strains
26003 a25923 b32067 c29212 d63501 e25922 f44568 g27853 h10104 i
5aH5-BrCH2COOH88>12832>128>128>128>12832
5bH6-ClCH2COOH>128>128>128>128>128>128>128>128>128
5cCH35-BrCH2COOH816>128>128>128>128>128>128>128
5dCH36-ClCH2COOH44>12832>128>128>128>12832
6aHHMolecules 22 00970 i00121168>128>128>128>1288
6bH6-ClMolecules 22 00970 i00242>12816>128>128>128>12816
6cCH3HMolecules 22 00970 i00321>1284>128>128>128>1284
6dCH35-BrMolecules 22 00970 i00422>1284>128>128>128>1284
6eCH36-ClMolecules 22 00970 i00564>128>128>128>128>128>128>128>128
7aHHMolecules 22 00970 i006221284>128>128>128>1284
7bH6-ClMolecules 22 00970 i0073216>1288>128>128>128>1288
7cCH3HMolecules 22 00970 i008216>128>128>128>128>128>128>128
7dCH35-BrMolecules 22 00970 i00922>12816>128>128>128>12816
8aHHMolecules 22 00970 i01021416>128>128>128>12816
8bH6-ClMolecules 22 00970 i01110.524>128>128>128>1284
8cCH3HMolecules 22 00970 i01211128>128>128>128>128>128>128
8dCH35-BrMolecules 22 00970 i01322>12864>128>128>128>12864
8eCH36-ClMolecules 22 00970 i01422>12864>128>128>128>12864
9HHMolecules 22 00970 i01544>12832>128>128>128>12832
10HHMolecules 22 00970 i016221616>128>128>128>12816
11HHMolecules 22 00970 i017>1288>12832>128>128>128>12832
Gatifloxacin0.1250.125>128>128120.1250.1252
Moxifloxacin0.1250.125>128>128120.1250.1252
Norfloxacin0.1250.125>128>128120.1250.1252
Oxacillin0.1250.125>128>128128>128128>128>128
penicillin0.1250.125>128>128128128128>128>128
a S. aureus CMCC(B) 26003; b S. aureus CMCC 25923; c S. pyogenes CMCC 32067; d E. faecalis CMCC 29212; e B. subtilis CMCC 63501; f E. coli CMCC 25922; g E. coli CMCC 44568; h P. aeruginosa CMCC 27853; I P. aeruginosa CMCC 10104.
Table 2. Inhibitory activity (MIC, µg/mL) of compounds 6a, 6c and 8ac against clinical isolates of multidrug-resistant strains.
Table 2. Inhibitory activity (MIC, µg/mL) of compounds 6a, 6c and 8ac against clinical isolates of multidrug-resistant strains.
CompoundR1-R2-R3-Multidrug-Resistant Gram-Positive Strains
43300 a33591 b
6aHHMolecules 22 00970 i0180.51
6cCH3HMolecules 22 00970 i0190.51
8aHHMolecules 22 00970 i02012
8bH6-ClMolecules 22 00970 i02122
8cCH3HMolecules 22 00970 i02212
Gatifloxacin0.50.25
Moxifloxacin0.50.25
Norfloxacin0.50.25
Oxacillin648
Penicillin32>32
a S. aureus ATCC 43300; b S. aureus ATCC 33591.
Table 3. Cytotoxic activity of compounds 6a, 8b and 8c on the HEK 293T cells.
Table 3. Cytotoxic activity of compounds 6a, 8b and 8c on the HEK 293T cells.
CompoundIC50 (µg/mL) a
6a36.90
8b54.09
8c32.28
a IC50 is the concentration required to inhibit 50% of cell growth.

Share and Cite

MDPI and ACS Style

Song, M.-X.; Li, S.-H.; Peng, J.-Y.; Guo, T.-T.; Xu, W.-H.; Xiong, S.-F.; Deng, X.-Q. Synthesis and Bioactivity Evaluation of N-Arylsulfonylindole Analogs Bearing a Rhodanine Moiety as Antibacterial Agents. Molecules 2017, 22, 970. https://doi.org/10.3390/molecules22060970

AMA Style

Song M-X, Li S-H, Peng J-Y, Guo T-T, Xu W-H, Xiong S-F, Deng X-Q. Synthesis and Bioactivity Evaluation of N-Arylsulfonylindole Analogs Bearing a Rhodanine Moiety as Antibacterial Agents. Molecules. 2017; 22(6):970. https://doi.org/10.3390/molecules22060970

Chicago/Turabian Style

Song, Ming-Xia, Song-Hui Li, Jiao-Yang Peng, Ting-Ting Guo, Wen-Hui Xu, Shao-Feng Xiong, and Xian-Qing Deng. 2017. "Synthesis and Bioactivity Evaluation of N-Arylsulfonylindole Analogs Bearing a Rhodanine Moiety as Antibacterial Agents" Molecules 22, no. 6: 970. https://doi.org/10.3390/molecules22060970

Article Metrics

Back to TopTop