Next Article in Journal
Protective Role of Sphingomyelin in Eye Lens Cell Membrane Model against Oxidative Stress
Next Article in Special Issue
Eicosapentaenoic Acid Inhibits KRAS Mutant Pancreatic Cancer Cell Growth by Suppressing Hepassocin Expression and STAT3 Phosphorylation
Previous Article in Journal
Low Expression of miR-375 and miR-190b Differentiates Grade 3 Patients with Endometrial Cancer
Previous Article in Special Issue
The Influence of Physical Activity on the Bioactive Lipids Metabolism in Obesity-Induced Muscle Insulin Resistance
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

α-Ketoheterocycles Able to Inhibit the Generation of Prostaglandin E2 (PGE2) in Rat Mesangial Cells

by
Anastasia Psarra
1,†,
Maria A. Theodoropoulou
1,†,
Martin Erhardt
2,
Marina Mertiri
1,
Christiana Mantzourani
1,
Sofia Vasilakaki
1,
Victoria Magrioti
1,
Andrea Huwiler
2 and
George Kokotos
1,*
1
Department of Chemistry, National and Kapodistrian University of Athens, Panepistimiopolis, 15771 Athens, Greece
2
Institute of Pharmacology, University of Bern, CH-3010 Bern, Switzerland
*
Author to whom correspondence should be addressed.
These authors contributed equally.
Biomolecules 2021, 11(2), 275; https://doi.org/10.3390/biom11020275
Submission received: 17 December 2020 / Revised: 28 January 2021 / Accepted: 10 February 2021 / Published: 13 February 2021
(This article belongs to the Collection Bioactive Lipids in Inflammation, Diabetes and Cancer)

Abstract

:
Prostaglandin E2 (PGE2) is a key mediator of inflammation, and consequently huge efforts have been devoted to the development of novel agents able to regulate its formation. In this work, we present the synthesis of various α-ketoheterocycles and a study of their ability to inhibit the formation of PGE2 at a cellular level. A series of α-ketobenzothiazoles, α-ketobenzoxazoles, α-ketobenzimidazoles, and α-keto-1,2,4-oxadiazoles were synthesized and chemically characterized. Evaluation of their ability to suppress the generation of PGE2 in interleukin-1β plus forskolin-stimulated mesangial cells led to the identification of one α-ketobenzothiazole (GK181) and one α-ketobenzoxazole (GK491), which are able to suppress the PGE2 generation at a nanomolar level.

1. Introduction

Prostaglandins are a class of highly bioactive eicosanoids, which are generated from arachidonic acid by the subsequent action of various enzymes [1,2]. Among them, prostaglandin E2 (PGE2) is the most abundant in humans, playing physiological and pathological roles [3]. The biosynthesis of PGE2 begins when phospholipase A2 (PLA2) hydrolyzes membrane glycerophospholipids to release free fatty acids, including arachidonic acid (Figure 1) [4]. Then, arachidonic acid is converted to prostaglandin H2 (PGH2) by the enzymes cyclooxygenase-1 (COX-1) and cyclooxygenase-2 (COX-2) [1]. Finally, prostaglandin synthases, such as microsomal prostaglandin E synthase-1 (mPGES-1), catalyze the generation of PGE2 [5,6], which exerts its actions interacting with PGE2 receptors (Figure 1).
PGE2 is a key mediator of inflammation [7,8], and consequently huge efforts have been devoted to the discovery of agents able to inhibit its production [9]. The involvement of PGE2 in tumorigenesis and cancer is well described in recent review articles [10,11,12]. A wide variety of inhibitors targeting the various enzymes involved in the PGE2 biosynthesis have been developed both in pharmaceutical industry and in academia. Inhibitors of PLA2 targeting the release of arachidonic acid have been described, but none of them reached the market [13]. Numerous clinically validated COX-1 and COX-2 inhibitors are known. Non-steroidal anti-inflammatory drugs (NSAIDs) are non-selective COX inhibitors, while selective COX-2 inhibitors, such as celecoxib, overcome the gastrointestinal side effects of COX-1 inhibitors, however exhibiting potential cardiovascular toxicity [14]. Although mPGES-1 inhibitors have been proposed as safer alternatives to COX-2 inhibitors, lackingcardiovascular toxicity, further research is needed so that such inhibitors enter clinical practice [5,6].
Sometimes, although an inhibitor for one particular enzyme involved in PGE2 generation presents high potency in vitro, tremendous discrepancies can be observed when it is studied in cells. Thus, we have focused our attention on evaluating potential anti-inflammatory compounds in a cellular system consisting of renal mesangial cells. We have previously shown that inhibitors of secreted PLA2 exhibit interesting suppression of the production of PGE2 in mesangial cells [15], while small peptides were also found to inhibit the generation of PGE2 [16]. Inspired by the anti-inflammatory properties which α-keto-thiazoles 1 and 2 (Figure 2) and related compounds exhibit [17,18,19,20], we synthesized various α-ketoheterocycles and studied their ability to inhibit the generation of PGE2 at a cellular level. We present herein the synthesis of a number of α-ketobenzothiazoles and related heterocycles, and we demonstrate that two of them exhibit potent suppression of the generation of PGE2 in rat mesangial cells.

2. Materials and Methods

2.1. General Chemistry Methods

Chromatographic purification of products was accomplished using forced-flow chromatography on Merck® (Merck, Darmstadt, Germany) Kieselgel 60 F254 230–400 mesh. Thin-layer chromatography (TLC) was performed on aluminum-backed silica plates (0.2 mm, 60 F254). Visualization of the developed chromatogram was performed by fluorescence quenching using phosphomolybdic acid, ninhydrin, or potassium permagnate stains. Melting points were determined on a Buchi® 530 (Buchi, Flawil, Switzerland) spectrometer and were uncorrected. 1H and 13C NMR spectra were recorded on a Varian® Mercury (Varian, Palo Alto, CA, USA) (200 MHz and 50 MHz, respectively), a Bruker Avance Neo (400 MHz and 100 MHz, respectively) (Bruker, Faellanden, Switzerland), or a Bruker Avance (500 MHz and 125 MHz, respectively) (Bruker, Santa Barbara, CA, USA), and are internally referenced to residual solvent signals. Data for 1H NMR are reported as follows: chemical shift (δ ppm), multiplicity (s = singlet, d = doublet, t = triplet, m = multiplet, br s = broad signal), coupling constant, integration, and peak assignment. Data for 13C NMR are reported in terms of chemical shift (δ ppm). IR spectra were recorded with an OSTEC, IROS-05, FTIR spectrophotometer equipped with ATR diamond crystal (Simex Co., Ltd., Nizhny Novgorod, Russia). Mass spectra (ESI) were recorded on a Finnigan® Surveyor MSQ LC-MS spectrometer (Thermo, Darmstadt, Germany). High-resolution mass spectrometry (HRMS) spectra were recorded on a Bruker® Maxis Impact QTOF (Bruker Daltonics, Bremen, Germany) spectrometer. A microwave synthesizer, Discover (CEM, Charlotte, NC, USA), was used for the microwave synthesis. 1H NMR and 13C NMR spectra of the final products are shown in the Supplementary Materials.
Compounds 18, 19, 20a, 21a, and 22a were synthesized as previously described [21], and their analytical data were in accordance with literature.
General procedure for the synthesis of Weinreb amides 4af from carboxylic acids.
To a stirred solution of the carboxylic acid 3af (1 mmol) in dry CH2Cl2 (7 mL), 4-dimethylaminopyridine (DMAP) (1 mmol), N,O-dimethyl hydroxylamine hydrochloride (1 mmol), N-methylmorpholine (1 mmol), and N-(3-dimethylaminopropyl)-N’-ethyl carbodiimide hydrochloride (WSCI·HCl) (1 mmol) were added consecutively at room temperature. The reaction mixture was left stirring for 18 h. It was then washed with an aqueous solution of 10% citric acid (3 × 10 mL), brine (10 mL), an aqueous solution of 5% NaHCO3 (3 × 10 mL), and brine (10 mL). The organic layer was dried over Na2SO4 and concentrated under reduced pressure. The amide was purified by flash chromatography eluting with the appropriate mixture of EtOAc:petroleum ether (40–60 °C) to afford the desired product.
N-Methoxy-N-methyl-5-(naphthalen-2-yl) pentanamide (4a) [19]. Yield 75%; Colorless oil; 1H NMR (200 MHz, CDCl3): δ = 7.85–7.70 (m, 3H, 3 × ArH), 7.61 (s, 1H, ArH), 7.50–7.27 (m, 3H, 3 × ArH), 3.64 (s, 3H, OCH3), 3.16 (s, 3H, NCH3), 2.80 (t, J = 7.0 Hz, 2H, CH2), 2.45 (t, J = 7.0 Hz, 2H, CH2), 1.87–1.60 (m, 4H, 2 × CH2); 13C NMR (50 MHz, CDCl3): δ = 175.8, 139.9, 133.6, 131.9, 127.8, 127.6, 127.4, 127.3, 126.3, 125.8, 125.0, 61.2, 35.9, 31.7, 31.1, 24.4; MS (ESI) m/z (%): 272 [(M+H)+, 100].
N-Methoxy-5-(4-methoxyphenyl)-N-methylpentanamide (4b). Yield 64%; Colorless oil; 1H NMR (200 MHz, CDCl3): δ = 7.10 (d, J = 8.0 Hz, 2H, 2 × ArH), 6.82 (d, J = 8.0 Hz, 2H, 2×ArH), 3.78 (s, 3H, OCH3), 3.66 (s, 3H, OCH3), 3.17 (s, 3H, NCH3), 2.59 (t, J = 6.0 Hz, 2H, CH2), 2.44 (t, J = 6.0 Hz, 2H, CH2), 1.73–1.61 (m, 4H, 2 × CH2); 13C NMR (50 MHz, CDCl3): δ = 176.4, 157.5, 134.3, 129.2, 113.6, 61.1, 55.1, 34.7, 31.6, 31.4, 24.2; MS (ESI) m/z (%): 252.2 [(M+H)+, 100].
N-Methoxy-N-methyl-2-(phenethylthio)acetamide (4c). Yield 83%; Colorless oil; 1H NMR (200 MHz, CDCl3): δ = 7.31−7.10 (m, 5H, 5 × ArH), 3.63 (s, 3H, OCH3), 3.31 (s, 2H, SCH2), 3.16−3.04 (m, 4H, SCH2,CH2), 2.87 (s, 3H, NCH3); 13C NMR (50 MHz, CDCl3): δ = 159.2, 156.7, 128.1, 128.0, 125.9, 61.1, 35.3, 33.3, 31.2; HRMS (ESI) [M+Na]+ m/z: 262.0872; (calculated for [C12H17NNaO2S]+ 262.0872).
N-Methoxy-N-methyl-2-(2-(naphthalen-2-yl)ethoxy)acetamide (4d). Yield 72%; Colorless oil; 1H NMR (200 MHz, CDCl3): δ = 7.83−7.35 (m, 7H, 7 × ArH), 4.24 (s, 2H, OCH2), 3.85 (t, J = 7.1 Hz, 2H, OCH2), 3.52 (s, 3H, OCH3), 3.17−3.03 (m, 5H, NCH3, CH2); 13C NMR (50 MHz, CDCl3): δ = 170.7, 135.9, 133.2, 131.8, 127.6, 127.3, 127.22, 127.17, 126.9, 125.6, 125.0, 72.2, 68.1, 61.0, 36.0, 31.9; HRMS (ESI) [M+Na]+ m/z: 296.1254; (calculated for [C16H19NNaO3]+ 296.1257).
3-([1,1′-Biphenyl]-4-yl)-N-methoxy-N-methylpropanamide (4e). Yield 88%; Colorless oil; 1H NMR (200 MHz, CDCl3): δ = 7.60−7.23 (m, 9H, 9 × ArH), 3.63 (s, 3H, OCH3), 3.19 (s, 3H, NCH3), 3.00 (t, J = 8.1 Hz, 2H, CH2), 2.81 (t, J = 8.2 Hz, 2H, CH2); 13C NMR (50 MHz, CDCl3): δ = 140.9, 140.4, 139.0, 128.8, 128.7, 127.1, 127.0, 126.9, 61.2, 33.7, 32.2, 30.2; HRMS (ESI) [M+Na]+ m/z: 292.1308; (calculated for [C17H19NNaO2]+ 292.1308).
N-Methoxy-N-methyl-3-(naphthalen-2-yl)propanamide (4f). Yield 93%; Colorless oil; 1H NMR (200 MHz, CDCl3): δ = 7.82−7.32 (m, 7H, 7 × ArH), 3.59 (s, 3H, OCH3), 3.23−3.07 (m, 5H, NCH3, CH2), 2.91−2.76 (m, 2H, CH2); 13C NMR (50 MHz, CDCl3): δ = 173.4, 138.7, 133.4, 131.9, 127.9, 127.5, 127.3, 127.1, 126.4, 125.8, 125.1, 61.1, 33.5, 32.0, 30.7; HRMS (ESI) [M+Na]+ m/z: 266.1151; (calculated for [C15H17NNaO2]+ 266.1151).
General procedure for the synthesis of Weinreb amides 4g,h from esters.
To a stirred solution of ester 5 or 6 (1 mmol) in dry tetrahydrofuran (2 mL) at −20 °C, N,O-dimethyl hydroxylamine hydrochloride (1.5 mmol) was added. Isopropyl magnesium chloride was then added dropwise over 15 min and the reaction mixture was left stirring for 35 min at −20 °C. The reaction mixture was quenched with a saturated solution of NH4Cl (5 mL) and the reaction mixture was extracted with diethyl ether (2 × 10 mL). The combined extracts were dried over Na2SO4 and concentrated under reduced pressure. Purification by flash chromatography eluting with the appropriate mixture of EtOAc:petroleum ether (40–60 °C) afforded the desired product.
N-Methoxy-N-methyl-2-(naphthalen-2-yloxy)acetamide (4g). Yield 41%; White solid; mp: 74–75 °C; 1H NMR (200 MHz, CDCl3): δ = 7.88–6.99 (m, 7H, 7 × ArH), 4.91 (s, 2H, CH2), 3.77 (s, 3H, OCH3), 3.25 (s, 3H, NCH3); 13C NMR (50 MHz, CDCl3): δ = 201.7, 156.2, 144.2, 134.4, 129.7, 127.7, 126.9, 126.5, 124.0, 118.9, 107.3, 65.7, 61.8, 32.5. HRMS (ESI) [M+Na]+ m/z: 268.0940; (calculated for [C14H15NNaO3]+ 268.0944).
N-Methoxy-2-(4-methoxyphenoxy)-N-methylacetamide (4h). Yield 52%; Colorless oil; 1H NMR (200 MHz, CDCl3): δ = 6.88–6.66 (m, 4H, 4 × ArH), 4.67 (s, 2H, CH2), 3.66 (s, 3H, OCH3), 3.64 (s, 3H, OCH3), 3.13 (s, 3H, NCH3); 13C NMR (50 MHz, CDCl3): δ = 169.3, 154.1, 152.3, 115.8, 114.4, 66.2, 61.4, 55.4, 32.1. HRMS (ESI) [M+Na]+ m/z: 248.0891; (calculated for [C11H15NNaO4]+ 248.0893.
General procedure for the synthesis of α-ketobenzothiazoles 8ah.
To a stirred solution of benzothiazole (3 mmol) in dry Et2O (20 mL) at −78 °C, under a dry argon atmosphere, a solution of n-BuLi (1.6 M in hexane, 3 mmol) was added dropwise over a period of 10 min. The resulting orange solution was stirred for 45 min. Then, a solution of the Weinreb amide (1 mmol) in dry Et2O (2 mL) was slowly added giving the mixture a dark brown color. After stirring for 30 min at −78 °C, the mixture was allowed to warm up to room temperature over a period of 2 h. Then, saturated aqueous ammonium chloride solution was added, and the reaction mixture was extracted with diethyl ether (2 × 10 mL). The combined extracts were washed with brine (10 mL) and then dried over Na2SO4 and concentrated under reduced pressure. Purification by flash chromatography eluting with the appropriate mixture of EtOAc:petroleum ether (40–60 °C) afforded the desired product.
1-(Benzo[d]thiazol-2-yl)-5-(naphthalen-2-yl)pentan-1-one (8a) [19]. Yield 72%; Yellow solid; 1H NMR (400 MHz, CDCl3): δ = 8.18 (d, J = 8.0 Hz, 1H, ArH), 7.97 (d, J = 8.0 Hz, 1H, ArH), 7.83−7.72 (m, 3H, 3 × ArH), 7.63 (s, 1H, ArH), 7.61−7.49 (m, 2H, 2 × ArH), 7.49−7.38 (m, 2H, 2 × ArH), 7.35 (dd, J1 = 8.4, J2 = 1.7 Hz, 1H, ArH), 3.34 (t, J = 6.9 Hz, 2H, CH2CO), 2.86 (t, J = 7.1 Hz, 2H, CH2Ar), 1.98–1.79 (m, 4H, 2 × CH2); 13C NMR (100 MHz, CDCl3): δ = 195.5, 166.7, 153.7, 139.8, 137.4, 133.8, 132.1, 128.0, 127.8, 127.7, 127.6, 127.5, 127.1, 126.6, 126.0, 125.5, 125.2, 122.6, 38.5, 36.0, 30.9, 23.8; IR: v ˜ = 3052, 1687, 1601, 1551, 1490 cm−1; HRMS (ESI) [M+Na]+ m/z: 368.1084; (calculated for [C22H19NNaOS]+ 368.1080).
1-(Benzo[d]thiazol-2-yl)-5-(4-methoxyphenyl)pentan-1-one (8b). Yield 52%; Orange solid; mp: 65–66 °C; 1H NMR (200 MHz, CDCl3): δ = 8.18 (d, J = 7.6 Hz, 1H, ArH), 7.96 (d, J = 7.2 Hz, 1H, ArH), 7.62−7.45 (m, 2H, 2 × ArH), 7.12 (d, J = 8.6 Hz, 2H, 2 × ArH), 6.83 (d, J = 8.6 Hz, 2H, 2 × ArH), 3.77 (s, 3H, OCH3), 3.30 (t, J = 7.1 Hz, 2H, CH2), 2.64 (t, J = 7.3 Hz, 2H, CH2), 1.95−1.64 (m, 4H, 2 × CH2); 13C NMR (50 MHz, CDCl3): δ = 195.4, 166.5, 157.7, 153.6, 137.3, 134.2, 129.3, 127.6, 127.0, 125.4, 122.5, 113.7, 55.2, 38.4, 34.8, 31.2, 23.6; HRMS (ESI) [M+Na]+ m/z: 348.1032; (calculated for [C19H19NNaO2S]+ 348.1029).
1-(Benzo[d]thiazol-2-yl)-2-(phenethylthio)ethan-1-one (8c). Yield 86%; Orange solid; mp: 63–65 °C; 1H NMR (500 MHz, CDCl3): δ = 8.24 (d, J = 8.0 Hz, 1H, ArH), 8.03 (d, J = 7.8 Hz, 1H, ArH), 7.63 (t, J = 7.3 Hz, 1H, ArH), 7.58 (t, J = 7.0 Hz, 1H, ArH), 7.39−7.33 (m, 2H, 2 × ArH), 7.32−7.26 (m, 3H, 3 × ArH), 4.14 (s, 2H, SCH2), 3.03−2.96 (m, 4H, SCH2, CH2); 13C NMR (125 MHz, CDCl3): δ = 188.9, 165.4, 153.5, 140.1, 137.7, 128.62, 128.56, 127.9, 127.1, 126.5, 125.6, 122.5, 36.2, 35.7, 33.9; IR: v ˜ = 3060, 1678, 1603, 1556 cm−1; HRMS (ESI) [M+Na]+ m/z: 336.0484; (calculated for [C17H15NNaOS2]+ 336.0487).
1-(Benzo[d]thiazol-2-yl)-2-(2-(naphthalen-2-yl)ethoxy)ethan-1-one (8d). Yield 20%; Orange solid; mp: 57–59 °C; 1H NMR (200 MHz, CDCl3): δ = 8.22−8.08 (m, 1H, ArH), 8.06−7.92 (m, 1H, ArH), 7.91−7.69 (m, 4H, 4 × ArH), 7.60−7.38 (m, 5H, 5 × ArH), 5.15 (s, 2H, OCH2), 3.99 (t, J = 7.2 Hz, 2H, OCH2), 3.21 (t, J = 7.2 Hz, 2H, CH2Ar); 13C NMR (50 MHz, CDCl3): δ = 191.2, 164.0, 153.5, 136.9, 136.0, 133.7, 132.3, 128.1, 128.0, 127.74, 127.66, 127.4, 127.3, 126.1, 125.5, 122.6, 73.7, 73.0, 36.5; IR: v ˜ = 3058, 1708, 1634, 1589 cm−1; HRMS (ESI) [M+Na]+ m/z: 370.0883; (calculated for [C21H17NNaO2S]+ 370.0872).
3-([1,1′-Biphenyl]-4-yl)-1-(benzo[d]thiazol-2-yl)propan-1-one (8e). Yield 91%; Yellowish solid; mp: 44–46 °C; 1H NMR (400 MHz, CDCl3): δ = 8.20 (d, J = 8.1 Hz, 1H, ArH), 7.97 (d, J = 7.9 Hz, 1H, ArH), 7.65–7.50 (m, 6H, 6 × ArH), 7.46 (t, J = 7.6 Hz, 2H, 2 × ArH), 7.42-7.33 (m, 3H, 3 × ArH), 3.69 (t, J = 7.6 Hz, 2H, CH2), 3.22 (t, J = 7.6 Hz, 2H, CH2); 13C NMR (100 MHz, CDCl3): δ = 194.4, 166.2, 153.6, 141.0, 139.8, 139.2, 137.3, 129.0, 128.8, 127.7, 127.3, 127.2, 127.1, 127.0, 125.5, 122.5, 40.2, 29.4; HRMS (ESI) [M+Na]+ m/z: 366.0934; (calculated for [C22H17NNaOS]+ 366.0923).
1-(Benzo[d]thiazol-2-yl)-3-(naphthalen-2-yl)propan-1-one (8f). Yield 91%; Yellowish solid of low melting point; 1H NMR (400 MHz, CDCl3): δ = 8.20 (d, J = 8.2 Hz, 1H, ArH), 7.96 (d, J = 8.0 Hz, 1H, ArH), 7.87−7.78 (m, 3H, 3 × ArH), 7.75 (s, 1H, ArH), 7.60−7.41 (m, 5H, 5 × ArH), 3.73 (t, J = 7.6 Hz, 2H, CH2), 3.32 (t, J = 7.6 Hz, 2H, CH2); 13C NMR (100 MHz, CDCl3): δ = 194.4, 166.2, 153.6, 138.2, 137.3, 133.7, 132.2, 128.2, 127.71, 127.68, 127.6, 127.3, 127.0, 126.7, 126.1, 125.5, 125.4, 122.5, 40.2, 29.9; HRMS (ESI) [M+Na]+ m/z: 340.0779; (calculated for [C20H15NNaOS]+ 340.0767).
1-(Benzo[d]thiazol-2-yl)-2-(naphthalen-2-yloxy)ethan-1-one (8g). Yield 22%; Pale yellow solid; mp: 144–145 °C; 1H NMR (400 MHz, CDCl3): δ = 8.28 (d, J = 8.1 Hz, 1H, ArH), 8.07 (d, J = 7.9 Hz, 1H, ArH), 7.86−7.79 (m, 2H, 2 × ArH), 7.76 (d, J = 8.2 Hz, 1H, ArH), 7.70−7.59 (m, 2H, 2 × ArH), 7.50−7.44 (m, 1H, ArH), 7.42−7.34 (m, 2H, 2 × ArH), 7.28 (d, J = 8.9 Hz, 1H, ArH), 5.81 (s, 2H, CH2); 13C NMR (100 MHz, CDCl3): δ = 188.7, 163.6, 155.9, 153.5, 137.1, 134.3, 129.8, 129.5, 128.1, 127.7, 127.4, 126.9, 126.5, 125.6, 124.1, 122.6, 118.7, 107.6, 70.3; IR: v ˜ = 3058, 1715, 1634, 1598, 1484 cm-1; HRMS (ESI) [M+Na]+ m/z: 342.0568; (calculated for [C19H13NNaO2S]+ 342.0559).
1-(Benzo[d]thiazol-2-yl)-2-(4-methoxyphenoxy)ethan-1-one (8h). Yield 27%; Pale yellow solid of low melting point; 1H NMR (400 MHz, CDCl3): δ = 8.20 (d, J = 8.0 Hz, 1H, ArH), 8.02 (d, J = 7.9 Hz, 1H, ArH), 7.67−7.53 (m, 2H, 2 × ArH), 6.99 (d, J = 8.4 Hz, 2H, 2 × ArH), 6.85 (d, J = 8.4 Hz, 2H, 2 × ArH), 5.61 (s, 2H, CH2), 3.77 (s, 3H, CH3); 13C NMR (100 MHz, CDCl3): δ = 189.5, 163.7, 154.8, 153.6, 152.3, 137.1, 128.2, 127.4, 125.6, 122.7, 116.4, 114.9, 71.5, 55.9; HRMS (ESI) [M+Na]+ m/z: 322.0514; (calculated for [C16H13NNaO3S]+ 322.0508).
tert-Butyl 2-(2-(naphthalen-2-yl)ethoxy)acetate (10). Alcohol 9 (1 mmol), tert-butyl bromoacetate (1.2 mmol), and Bu4NHSO4 (0.2 mmol) were diluted in toluene (1 mL) and in an aqueous solution of 50% NaOH (1 mL). The reaction mixture was stirred for 18 h. After completion of the reaction, the organic layer was collected and washed with brine (2 mL), and then dried over Na2SO4 and concentrated under reduced pressure. Purification by flash chromatography eluting with a mixture of EtOAc:petroleum ether (40–60 °C) 2:8 afforded the desired product. Yield 94%; Colorless oil; 1H NMR (200 MHz, CDCl3): δ = 7.90–7.31 (m, 7H, 7 × ArH), 4.01 (s, 2H, OCH2), 3.85 (t, J = 7.2 Hz, 2H, OCH2), 3.14 (t, J = 7.2 Hz, 2H, CH2Ar), 1.50 (s, 9H, 3 × CH3); 13C NMR (50 MHz, CDCl3): δ = 169.6, 136.0, 133.5, 132.1, 127.8, 127.5, 127.4, 127.1, 125.8, 125.2, 81.5, 72.3, 68.8, 36.3, 28.0. HRMS (ESI) [M+Na]+ m/z: 309.1458; (calculated for [C18H22NaO3]+ 309.1461).
2-(2-(Naphthalen-2-yl)ethoxy)acetic acid (3d). To a stirred solution of tert-butyl ester 10 (1 mmol) in dry CH2Cl2 (2 mL), under an inert argon atmosphere, trifluoroacetic acid (2 mL) was added and the reaction mixture was left stirring for 2 h. Toluene (2 mL) was then added and the solvents were evaporated under reduced pressure. The latter was repeated until complete removal of trifluoroacetic acid. The residue was diluted in H2O (5 mL) and diethyl ether (5 mL) and transferred to a separating funnel. An aqueous solution of 5% NaHCO3 (5 mL) was added, and the aqueous layer was acidified with an aqueous solution of 5% citric acid (5 mL) and then extracted with diethyl ether (3 × 5 mL). Concentration of the combined organic layers under reduced pressure afforded the desired product. Yield 81%; White solid; mp: 63–67 °C; 1H NMR (200 MHz, CD3OD): δ = 7.67−7.19 (m, 7H, 7 × ArH), 5.43 (br s, 1H, COOH), 4.02−3.88 (m, 2H, OCH2), 3.71−3.56 (m, 2H, OCH2), 2.97−2.85 (m, 2H, CH2Ar); 13C NMR (50 MHz, CD3OD): δ = 174.1, 137.3, 134.9, 133.5, 128.8, 128.4, 128.1, 126.8, 126.2, 73.1, 68.6, 36.9. HRMS (ESI) [M-H]- m/z: 229.0866; (calculated for [C14H13O3]- 229.0870).
1-(Benzo[d]thiazol-2-yl)-5-(naphthalen-2-yl)pentan-1-ol (12). To a stirred solution of benzothiazole (1.2 mmol) in dry diethyl ether (6.5 mL), at −78 °C, under an inert argon atmosphere, a solution of n-BuLi 1M (1.2 mmol) was added dropwise and the reaction mixture was stirred for 1h at −78 °C. A solution of aldehyde 11 (1 mmol) in dry diethyl ether (1.5 mL) was then added, and the reaction mixture was further stirred for 1 h at −78 °C and for 16 h at room temperature. The reaction was quenched with a saturated NH4Cl aqueous solution (5 mL), and the aqueous layer was collected and extracted with diethyl ether (2 x 10 mL). The combined organic layers were washed with brine (10 mL), dried over Na2SO4, and concentrated under reduced pressure. Purification by flash chromatography eluting with a mixture of EtOAc:petroleum ether (40–60 °C) 3:7 afforded the desired product. Yield 55%; Orange solid of low melting point; 1H NMR (400 MHz, CDCl3): δ = 7.98 (d, J = 8.1 Hz, 1H, ArH), 7.91−7.70 (m, 4H, 4 × ArH), 7.59 (s, 1H, ArH), 7.50−7.34 (m, 4H, ArH), 7.30 (d, J = 8.4 Hz, 1H, ArH), 5.10 (dd, J1 = 7.5, J2 = 4.7 Hz, 1H, CHOH), 3.83 (br s, 1H, OH), 2.77 (t, J = 7.6 Hz, 2H, CH2Ar), 2.14−1.93 (m, 2H, CH2), 1.84−1.70 (m, 2H, CH2), 1.70−1.50 (m, 2H, CH2); 13C NMR (100 MHz, CDCl3): δ = 176.8, 152.8, 140.0, 134.8, 133.7, 132.0, 127.9, 127.7, 127.5, 127.4, 126.4, 126.2, 125.9, 125.12, 125.09, 122.9, 121.9, 72.2, 38.0, 35.9, 31.1, 25.0; HRMS (ESI) [M+H]+ m/z: 348.1425; (calculated for [C22H22NOS]+ 348.1417).
General procedure for the synthesis of cyanohydrins 14a,b and 15.
To a stirred solution of aldehydes 13a,b, 11 (1 mmol) in CH2Cl2 (1.3 mL), a solution of NaHSO3 (1.5 mmol, 156 mg) in water (0.3 mL) was added at room temperature. After stirring for 30 min, the organic solvent was concentrated under reduced pressure, water (0.3 mL) was added, and the reaction mixture was cooled to 0 °C. Then, a solution of KCN (1.5 mmol, 98 mg) in water (0.3 mL) was added dropwise over 1 h, and the reaction mixture was left stirring for 16 h. After the completion of the reaction, CH2Cl2 (5 mL) was added to extract the product and the organic layer was washed with brine (10 mL), dried over Na2SO4 and concentrated under reduced pressure. Purification by flash chromatography eluting with the appropriate mixture of EtOAc:petroleum ether (40–60 °C) afforded the desired product.
2-Hydroxy-6-(4-methoxyphenyl)hexanenitrile (14a). Yield 78%; Colorless oil; 1H NMR (200 MHz, CDCl3): δ = 7.16 (d, J = 8.0 Hz, 2H, 2 × ArH), 6.90 (d, J = 8.0 Hz, 2H, 2 × ArH), 4.45 (q, J = 6.0 Hz, 1H, CHOH), 4.11 (br s, 1H, OH), 3.82 (s, 3H, OCH3), 2.62 (t, J = 6.0 Hz, 2H, CH2Ar), 1.93−1.83 (m, 2H, CH2), 1.72−1.55 (m, 4H, 2 × CH2); 13C NMR (50 MHz, CDCl3): δ = 157.2, 133.9, 128.9, 119.9, 113.4, 60.6, 54.9, 34.5, 34.3, 30.6, 23.8; MS (ESI) m/z (%): 237.2 [(M+NH4)+, 100].
2-Hydroxy-3-(4-methoxyphenethoxy)propanenitrile (14b). Yield 40%; Colorless oil; 1H NMR (200 MHz, CDCl3): δ = 7.14 (d, J = 8.6 Hz, 2H, 2 × ArH), 6.86 (d, J = 8.8 Hz, 2H, 2 × ArH), 4.56−4.45 (m, 1H, CHOH), 3.79 (s, 3H, OCH3), 3.77−3.65 (m, 4H, 2 × CH2), 2.86 (t, J = 6.9 Hz, 2H, CH2); HRMS (ESI) [M+Na]+ m/z: 244.0944; (calculated for [C12H15NNaO3]+ 244.0944).
2-Hydroxy-6-(naphthalen-2-yl)hexanenitrile (15). Yield 45%; Colorless oil; 1H NMR (200 MHz, CDCl3): δ = 7.93−7.28 (m, 7H, 7 × ArH), 4.39 (q, J = 6.2 Hz, 1H, CHOH), 3.41 (br s, 1H, OH), 2.81 (t, J = 7.4 Hz, 2H, CH2Ar), 1.82−1.68 (m, 4H, 2 × CH2), 1.59−1.55 (m, 2H, CH2); 13C NMR (50 MHz, CDCl3): δ = 139.4, 133.4, 131.8, 127.8, 127.5, 127.3, 127.1, 126.3, 125.9, 125.1, 120.0, 60.9, 35.6, 34.8, 30.4, 24.1; HRMS (ESI) [M+Na]+ m/z: 262.1202; (calculated for [C16H17NNaO]+ 262.1202).
General procedure for the synthesis of α-hydroxy benzoxazoles (16a,c,d) and α-hydroxy benzimidazoles (16b,e) from cyanohydrins.
To a stirred mixture of chloroform (0.5 M) and absolute ethanol (0.5 M) cooled at 0 °C, under an inert dry argon atmosphere, acetyl chloride (0.46 mL) was added dropwise over 15 min. Then, a solution of cyanohydrins 14a,b and 15 (1 mmol) in CHCl3 (0.5 M) was added and the reaction mixture was stirred at 0 °C for 1 h. The solvent was evaporated under reduced pressure and at a temperature not higher than 25 °C. The reaction mixture was then dissolved in absolute ethanol (1.2 M), 2-aminophenol (for the benzoxazole compounds 16a,c,d) or 2-phenylenediamine (for the benzimidazole compounds 16b,e) (1.1 mmol) was added and the final reaction mixture was refluxed under an inert argon atmosphere for 16 h. After completion of the reaction, the solvent was evaporated under reduced pressure. Purification by flash chromatography eluting with the appropriate mixture of EtOAc:petroleum ether (40–60 °C) afforded the desired product.
1-(Benzo[d]oxazol-2-yl)-5-(4-methoxyphenyl)pentan-1-ol (16a). Yield 57%; Pale yellow oil; 1H NMR (200 MHz, CDCl3): δ = 7.73−7.67 (m, 1H, ArH), 7.57−7.48 (m, 1H, ArH), 7.39−7.30 (m, 2H, 2 × ArH), 7.08 (d, J = 8.0 Hz, 2H, 2 × ArH), 6.81 (d, J = 8.0 Hz, 2H, 2 × ArH), 4.97 (t, J = 6.0 Hz, 1H, CHOH), 3.84 (br s, 1H, OH), 3.78 (s, 3H, OCH3), 2.57 (t, J = 6.0 Hz, 2H, CH2Ar), 2.15–1.94 (m, 2H, CH2), 1.73−1.47 (m, 4H, 2 × CH2); 13C NMR (50 MHz, CDCl3): δ = 167.8, 157.5, 150.6, 140.2, 134.3, 129.1, 125.1, 124.4, 119.8, 113.6, 110.7, 67.9, 55.1, 35.3, 34.7, 31.3, 24.5; MS (ESI) m/z (%): 312.2 [(M+H)+, 100].
1-(1H-Benzo[d]imidazol-2-yl)-5-(4-methoxyphenyl)pentan-1-ol (16b). Yield 37%; White solid; mp: 165–167 °C; 1H NMR (200 MHz, CD3OD): δ = 7.58−7.54 (m, 2H, 2 × ArH), 7.25−7.21 (m, 2H, 2 × ArH), 7.03 (d, J = 8.0 Hz, 2H, 2 × ArH), 6.75 (d, J = 8.0 Hz, 2H, 2 × ArH), 4.93 (t, J = 6.0 Hz, 1H, CHOH), 3.73 (s, 3H, OCH3), 2.54 (t, J = 6.0 Hz, 2H, CH2Ar), 2.05−1.91 (m, 2H, CH2), 1.66−1.40 (m, 4H, 2 × CH2); 13C NMR (50 MHz, CD3OD): δ = 159.3, 139.4, 135.8, 135.7, 130.4, 123.4, 115.8, 114.7, 69.5, 55.7, 37.8, 35.8, 32.7, 25.7; MS (ESI) m/z (%): 311.2 [(M+H)+, 100].
1-(Benzo[d]oxazol-2-yl)-2-(4-methoxyphenethoxy)ethan-1-ol (16c). Yield 65%; Pale yellow solid of low melting point; 1H NMR (200 MHz, CDCl3): δ = 7.80−7.69 (m, 1H, ArH), 7.56−7.47 (m, 1H, ArH), 7.38−7.30 (m, 2H, 2 × ArH), 7.03 (d, J = 8.5 Hz, 2H, 2 × ArH), 6.71 (d, J = 8.6 Hz, 2H, 2 × ArH), 5.12 (t, J = 4.9 Hz, 1H, CHOH), 4.01−3.91 (m, 2H, OCH2CH), 3.81−3.59 (m, 5H, OCH3, OCH2), 2.79 (t, J = 6.9 Hz, 2H, CH2); 13C NMR (50 MHz, CDCl3): δ = 165.0, 158.0, 157.6, 140.5, 130.5, 129.7, 125.2, 124.5, 120.1, 113.7, 110.8, 72.7, 72.5, 67.5, 55.1, 35.0; HRMS (ESI) [M+Na]+ m/z: 336.1201; (calculated for [C18H19NNaO4]+ 336.1206).
1-(Benzo[d]oxazol-2-yl)-5-(naphthalen-2-yl)pentan-1-ol (16d). Yield 53%; Orange oil; 1H NMR (200 MHz, CDCl3): δ = 7.94–7.20 (m, 11H, 11 × ArH, 5.08–4.93 (m, 1H, CHOH), 4.45 (br s, 1H, OH), 2.79 (t, J = 7.4 Hz, 2H, CH2Ar), 2.10 (m, 2H, CH2), 1.89−1.49 (m, 4H, 2 × CH2); 13C NMR (50 MHz, CDCl3): δ = 168.0, 150.5, 140.2, 139.8, 133.5, 131.8, 127.7, 127.5, 127.3, 127.2, 126.2, 125.8, 125.1, 125.0, 124.4, 119.8, 110.7, 67.8, 35.8, 35.2, 30.9, 24.7; HRMS (ESI) [M+Na]+ m/z: 354.1466; (calculated for [C22H21NNaO2]+ 354.1465).
1-(1H-Benzo[d]imidazol-2-yl)-5-(naphthalen-2-yl)pentan-1-ol (16e). Yield 43%; White solid; 1H NMR (200 MHz, CDCl3): δ = 7.82−7.03 (m, 11H, 11 × ArH), 4.85 (t, J = 6.6 Hz, 1H, CHOH), 4.41 (br s, 1H, OH), 2.65 (t, J = 7.3 Hz, 2H, CH2Ar), 2.03−1.76 (m, 2H, CH2), 1.74−1.25 (m, 4H, 2 × CH2); 13C NMR (50 MHz, CDCl3): δ = 157.4, 139.8, 137.6, 133.4, 131.7, 127.5, 127.4, 127.2, 127.1, 126.1, 125.6, 124.8, 122.3, 114.7, 68.1, 36.3, 35.7, 30.9, 24.7; HRMS (ESI) [M+H]+ m/z: 331.1805; (calculated for [C22H23N2O]+ 331.1805).
General procedure for the synthesis of O-acyl-amidoximes (20b,c).
To a stirred solution of amidoxime 19 (1.0 mmol) in dry CH2Cl2 (20 mL), benzoic acid (for benzoate group) or isobutyric anhydride (for isobutyrate group) (1 mmol, 102 mg) and N,N′-dicyclohexylcarbodiimide (DCC) (1.1 mmol, 227 mg) were added. The reaction mixture was stirred for 24 h at room temperature. After completion of the reaction the organic solvent was evaporated under reduced pressure. Purification by flash chromatography eluting with the appropriate mixture of EtOAc:petroleum ether (40–60 °C) afforded the desired product.
(Z)-N’-(Benzoyloxy)-2-((tert-butyldimethylsilyl)oxy)-6-(4-methoxyphenyl)hexanimidamide (20b). Yield 67%; 1H NMR (200 MHz, CDCl3): δ = 8.08−7.98 (m, 2H, 2 × ArH), 7.60−7.50 (m, 1H, ArH), 7.48−7.37 (m, 2H, 2 × ArH), 7.07 (d, J = 8.6 Hz, 2H, 2 × ArH), 6.79 (d, J = 8.6 Hz, 2H, 2 × ArH), 5.07 (s, 2H, NH2), 4.41 (t, J = 6.2 Hz, 1H, OCH), 3.75 (s, 3H, OCH3), 2.55 (t, J = 7.2 Hz, 2H, CH2Ar), 1.66−1.42 (m, 4H, 2 × CH2), 1.32−1.08 (m, 2H, CH2), 0.90 (s, 9H, 3 × CCH3), 0.09 (s, 6H, 2 × SiCH3); 13C NMR (50 MHz, CDCl3): δ = 164.2, 161.0, 157.8, 134.7, 133.1, 129.9, 129.6, 129.5, 128.6, 113.9, 70.5, 55.4, 37.8, 35.0, 31.7, 25.9, 24.9, 18.3, -4.9; HRMS (ESI) [M+H]+ m/z: 471.2670; (calculated for [C26H39N2O4Si]+ 471.2674).
(Z)-2-((tert-Butyldimethylsilyl)oxy)-N’-(isobutyryloxy)-6-(4-methoxyphenyl)hexanimidamide (20c). Yield 95%; 1H NMR (200 MHz, CDCl3): δ = 7.05 (d, J = 8.5 Hz, 2H, 2 × ArH), 6.79 (d, J = 8.6 Hz, 2H, 2 × ArH), 4.92 (s, 2H, NH2), 4.30 (t, J = 6.2 Hz, 1H, OCH), 3.76 (s, 3H, OCH3), [2.77−2.57 m, 1H, CH(CH3)2], 2.52 (t, J = 7.3 Hz, 2H, CH2Ar), 1.78−1.33 (m, 6H, 3 × CH2), 1.22 (d, J = 7.0 Hz, 6H, 2 × CHCH3), 0.87 (s, 9H, 3 × CCH3), 0.05 (s, 6H, 2 × SiCH3); 13C NMR (50 MHz, CDCl3): δ = 174.1, 160.1, 157.6, 134.6, 129.3, 113.7, 70.2, 55.2, 37.5, 34.8, 33.2, 31.5, 25.7, 24.7, 19.3, 18.0, -5.1; HRMS (ESI) [M+H]+ m/z: 437.2826; (calculated for [C23H41N2O4Si]+ 437.2830).
General procedure for the synthesis of α-hydroxy-oxadiazoles (21b,c).
To a stirred solution of O-acyl-amidoximes 20b,c (1.0 mmol) in dry toluene (3 mL) in a microwave vessel, tetrabutylammonium fluoride (TBAF) (1 M in THF, 1.0 mmol) was added. The reaction mixture was left stirring under microwave irradiation (initial setting at 90 W) for 1 h at 120 °C. The organic solvent was evaporated under reduced pressure. Purification by flash chromatography eluting with the appropriate mixture of EtOAc:petroleum ether (40–60 °C) afforded the desired product.
5-(4-Methoxyphenyl)-1-(5-phenyl-1,2,4-oxadiazol-3-yl)pentan-1-ol (21b). Yield 38%; White solid; mp: 88–90 °C; 1H NMR (200 MHz, CDCl3): δ = 8.26−8.05 (m, 2H, 2 × ArH), 7.67−7.46 (m, 3H, 3 × ArH), 7.08 (d, J = 8.6 Hz, 2H, 2 × ArH), 6.80 (d, J = 8.6 Hz, 2H, 2 × ArH), 4.93 (t, J = 6.6 Hz, 1H, CHOH), 3.76 (s, 3H, OCH3), 2.97 (br s, 1H, OH), 2.57 (t, J = 7.3 Hz, 2H, CH2Ar), 2.12−1.91 (m, 2H, CH2), 1.78−1.40 (m, 4H, 2 × CH2); 13C NMR (50 MHz, CDCl3): δ = 176.0, 172.8, 157.7, 134.6, 133.0, 129.3, 129.2, 128.3, 124.1, 113.8, 66.8, 55.3, 35.6, 34.9, 31.5, 24.8; HRMS (ESI) [M+H]+ m/z: 339.1700; (calculated for [C20H23N2O3]+ 339.1703).
1-(5-Isopropyl-1,2,4-oxadiazol-3-yl)-5-(4-methoxyphenyl)pentan-1-ol (21c). Yield 51%; Pale yellow oil; 1H NMR (200 MHz, CDCl3): δ = 7.05 (d, J = 8.7 Hz, 2H, 2 × ArH), 6.79 (d, J = 8.7 Hz, 2H, 2 × ArH), 4.80 (t, J = 6.6 Hz, 1H, CHOH), 3.75 (s, 3H, OCH3), 3.30−3.08 [m, 1H, CH(CH3)2], 2.83 (br s, 1H, OH), 2.54 (t, J = 7.4 Hz, 2H, CH2Ar), 1.98−1.83 (m, 2H, CH2), 1.75−1.47 (m, 4H, 2 × CH2), 1.37 (d, J = 7.0 Hz, 6H, 2 × CHCH3); 13C NMR (50 MHz, CDCl3): δ = 184.3, 171.9, 157.7, 134.6, 129.3, 113.8, 66.7, 55.3, 35.5, 34.9, 31.4, 27.6, 24.8, 20.2; HRMS (ESI) [M+H]+ m/z: 305.1858; (calculated for [C17H25N2O3]+ 305.1860).
General procedure for the oxidation of secondary alcohols to ketones (17ae, 22b,c).
To a stirred solution of α-hydroxy-heterocyclic compounds 16ae and 21b,c (1 mmol) in dry CH2Cl2 (0.2 M), under an inert argon atmosphere, Dess–Martin periodinane was added (1.3 mmol, 551 mg). The reaction mixture was stirred for 1 h and after completion of the reaction the solvent was evaporated under reduced pressure and Et2O (30 mL) was added. The organic phase was washed with saturated aqueous NaHCO3 (20 mL) containing Na2S2O3 (1.5 g, 9.5 mmol), H2O (20 mL), dried over Na2SO4, and the organic solvent was evaporated under reduced pressure. Purification by flash chromatography eluting with the appropriate mixture of EtOAc:petroleum ether (40–60 °C) afforded the desired product.
1-(Benzo[d]oxazol-2-yl)-5-(4-methoxyphenyl)pentan-1-one (17a). Yield 88%; White solid; mp: 59–61 °C; 1H NMR (200 MHz, CDCl3): δ = 7.90 (d, J = 7.4 Hz, 1H, ArH), 7.66 (d, J = 7.6 Hz, 1H, ArH), 7.59–7.38 (m, 2H, 2 × ArH), 7.11 (d, J = 8.5 Hz, 2H, 2 × ArH), 6.82 (d, J = 8.5 Hz, 2H, 2 × ArH), 3.77 (s, 3H, OCH3), 3.24 (t, J = 7.1 Hz, 2H, CH2CO), 2.63 (t, J = 7.3 Hz, 2H, CH2Ar), 1.95–1.60 (m, 4H, 2 × CH2); 13C NMR (50 MHz, CDCl3): δ = 190.2, 157.7, 157.2, 150.7, 140.5, 134.1, 129.3, 128.5, 125.8, 122.3, 113.7, 112.0, 55.3, 39.4, 34.7, 31.1, 23.4; HRMS (ESI) [M-H]- m/z: 308.1291; (calculated for [C19H18NO3]- 308.1292).
1-(1H-Benzo[d]imidazol-2-yl)-5-(4-methoxyphenyl)pentan-1-one (17b). Yield 79%; Colorless solid; mp: 101–103 °C; 1H NMR (200 MHz, CDCl3): δ = 10.64 (br s, 1H, NH), 8.00−7.82 (m, 1H, ArH), 7.63−7.31 (m, 3H, 3 × ArH), 7.10 (d, J = 8.4 Hz, 2H, 2 × ArH), 6.81 (d, J = 8.6 Hz, 2H, 2 × ArH), 3.77 (s, 3H), 3.34 (t, J = 7.2 Hz, 2H, CH2CO), 2.62 (t, J = 7.3 Hz, 2H, CH2Ar), 1.96−1.62 (m, 4H, 2 × CH2); 13C NMR (50 MHz, CDCl3): δ = 194.7, 157.8, 151.1, 147.6, 134.3, 129.4, 126.6, 124.0, 122.0, 114.1, 113.8, 112.3, 55.4, 38.2, 34.9, 31.3, 23.7; HRMS (ESI) [M-H] m/z: 307.1455; (calculated for [C19H19N2O2]- 308.1292).
1-(Benzo[d]oxazol-2-yl)-2-(4-methoxyphenethoxy)ethan-1-one (17c). Yield 35%; Pale yellow solid of low melting point; 1H NMR (500 MHz, CDCl3): δ = 7.87 (d, J = 8.0 Hz, 1H, ArH), 7.66 (d, J = 8.2 Hz, 1H, ArH), 7.55 (t, J = 7.8 Hz, 1H, ArH), 7.47 (t, J = 7.8 Hz, 1H, ArH), 7.18 (d, J = 8.5 Hz, 2H, 2 × ArH), 6.83 (d, J = 8.3 Hz, 2H, 2 × ArH), 5.01 (s, 2H, OCH2CO), 3.84 (t, J = 7.0 Hz, 2H, OCH2), 3.77 (s, 3H, OCH3), 2.97 (t, J = 7.0 Hz, 2H, CH2Ar); 13C NMR (125 MHz, CDCl3): δ = 186.4, 158.3, 155.5, 150.6, 140.4, 130.4, 130.0, 128.9, 126.1, 122.4, 114.0, 112.1, 73.8, 73.4, 55.4, 35.4; IR: v ˜ = 3091, 1718, 1612, 1515, 1453 cm-1; HRMS (ESI) [M+H]+ m/z: 312.1230; (calculated for [C18H18NO4]+ 312.1230); HRMS (ESI) [M+Na]+ m/z: 334.1051; (calculated for [C18H17NNaO4]+ 334.1050).
1-(Benzo[d]oxazol-2-yl)-5-(naphthalen-2-yl)pentan-1-one (17d). Yield 96%; White solid; 77−82 °C; 1H NMR (400 MHz, CDCl3): δ = 7.89 (d, J = 8.0 Hz, 1H, ArH), 7.84–7.72 (m, 3H, 3 × ArH), 7.69−7.60 (m, 2H, 2 × ArH), 7.56−7.38 (m, 4H, 4 × ArH), 7.34 (dd, J1 = 8.4, J2 = 1.4 Hz, 1H, ArH), 3.27 (t, J = 7.0 Hz, 2H, CH2CO), 2.86 (t, J = 7.2 Hz, 2H, CH2Ar), 1.98−1.79 (m, 4H, 2 × CH2); 13C NMR (100 MHz, CDCl3): δ = 190.1, 157.3, 150.8, 140.6, 139.6, 133.7, 132.1, 128.6, 128.0, 127.7, 127.5, 127.3, 126.5, 126.0, 125.8, 125.2, 122.3, 112.0, 39.4, 35.8, 30.7, 23.6; IR: v ˜ = 3049, 1701, 1601, 1534, 1506 cm-1; HRMS (ESI) [M+H]+ m/z: 330.1499; (calculated for [C22H20NO2]+ 330.1489).
1-(1H-Benzo[d]imidazol-2-yl)-5-(naphthalen-2-yl)pentan-1-one (17e). Yield 45%; White solid; mp: 127−132 °C; 1H NMR (400 MHz, CDCl3): δ = 10.24 (s, 1H, NH), 7.91 (d, J = 8.0 Hz, 1H, ArH), 7.82–7.73 (m, 3H, 3 × ArH), 7.62 (s, 1H, ArH), 7.53 (d, J = 7.9 Hz, 1H, ArH), 7.46−7.31 (m, 5H, 5 × ArH), 3.35 (t, J = 6.9 Hz, 2H, CH2CO), 2.85 (t, J = 7.2 Hz, 2H, CH2Ar), 1.96−1.80 (m, 4H, 2 × CH2); 13C NMR (50 MHz, CDCl3): δ = 194.4, 147.6, 143.5, 139.8, 133.7, 133.5, 132.1, 128.0, 127.7, 127.5, 127.4, 126.63, 126.56, 126.0, 125.2, 124.0, 122.1, 112.2, 38.2, 35.9, 30.9, 23.7; IR: v ˜ = 3286, 3055, 1679, 1598, 1512, 1401 cm−1; HRMS (ESI) [M+H]+ m/z: 329.1655; (calculated for [C22H21N2O]+ 329.1648).
5-(4-Methoxyphenyl)-1-(5-phenyl-1,2,4-oxadiazol-3-yl)pentan-1-one (22b). Yield 77%; White solid; 85−87 °C; 1H NMR (200 MHz, CDCl3): δ = 8.30−8.13 (m, 2H, 2 × ArH), 7.72−7.46 (m, 3H, 3 × ArH), 7.10 (d, J = 8.6 Hz, 2H, 2 × ArH), 6.82 (d, J = 8.7 Hz, 2H, 2 × ArH), 3.78 (s, 3H, OCH3), 3.14 (t, J = 7.1 Hz, 2H, CH2CO), 2.62 (t, J = 7.3 Hz, 2H, CH2Ar), 1.93–1.62 (m, 4H, 2 × CH2); 13C NMR (50 MHz, CDCl3): δ = 191.8, 177.2, 166.2, 157.9, 134.1, 133.6, 129.4, 128.6, 123.5, 113.9, 55.3, 40.7, 34.8, 31.1, 23.2; HRMS (ESI) [M+Na]+ m/z: 359.1363; (calculated for [C20H20N2NaO3]+ 359.1366).
1-(5-Isopropyl-1,2,4-oxadiazol-3-yl)-5-(4-methoxyphenyl)pentan-1-one (22c). Yield 45%; Colorless oil; 1H NMR (200 MHz, CDCl3): δ = 7.09 (d, J = 8.4 Hz, 2H, 2 × ArH), 6.81 (d, J = 8.4 Hz, 2H, 2 × ArH), 3.78 (s, 3H, OCH3), 3.38−3.21 [m, 1H, CH(CH3)2], 3.07 (t, J = 7.0 Hz, 2H, CH2CO), 2.59 (t, J = 7.2 Hz, 2H, CH2Ar), 1.84–1.61 (m, 4H, 2 × CH2), 1.44 (d, J = 7.0 Hz, 6H, 2 × CHCH3); 13C NMR (50 MHz, CDCl3): δ = 191.9, 185.7, 165.6, 157.9, 134.2, 129.4, 113.9, 55.4, 40.5, 34.8, 31.1, 27.7, 23.2, 20.2; HRMS (ESI) [M+Na]+ m/z: 325.1521; (calculated for [C17H22N2NaO3]+ 325.1523).

2.2. In Vitro Suppression of Cytokine-Triggered PGE2 Generation in Renal Mesangial Cells

2.2.1. Cell Culture

Rat renal mesangial cells (clone MZ B1) were isolated and characterized as previously described [22] and cultivated in medium consisting of RPMI 1640 supplemented with 10% fetal bovine serum, 10 mM Hepes, pH 7.4, 6 µg/mL bovine insulin, 5 mg/mL transferrin, 5 nM sodium selenite, 100 units/mL penicillin, and 100 µg/mL streptomycin. Prior to stimulation, cells were incubated for 4 h in DMEM containing 10 mM Hepes, pH 7.4, and 0.1 mg/mL fatty acid-free bovine serum albumin (BSA).

2.2.2. Quantification of Prostagladin E2

Confluent mesangial cells in 24-well plates were stimulated for 24 h in a total volume of 400 µL DMEM containing 0.1 mg/mL BSA with the stimuli and inhibitors as indicated in the figure legends. Thereafter, supernatants were removed and centrifuged for 5 min at 1000 x g. The supernatant was taken for PGE2 quantification using an enzyme-linked immunoassay (Enzo Life Sciences, Lörrach, Germany) following exactly the manufacturer’s recommendations.

2.2.3. Statistical Analysis

Statistical analysis of data was performed using one-way analysis of variance (ANOVA) followed by a Bonferroni’s post hoc test for multiple comparisons (GraphPad Prism version 5.00, San Diego, CA, USA). Half-maximal effective concentrations (EC50) of the compounds were calculated using the same software.

3. Results and Discussion

3.1. Synthesis of Inhibitors

Benzothiazolyl ketones were synthesized in two steps, most of them starting from the corresponding carboxylic acids, as shown in Figure 3. The first step involves the formation of the Weinreb amides 4af by coupling of carboxylic acids 3af with N,O-dimethylhydroxylamine hydrochloride using N-(3-dimethylaminopropyl)-N’-ethyl carbodiimide hydrochloride (WSCI·HCl) as the coupling agent [17,23]. Weinreb amides 4g,h were prepared from esters 5 and 6 using N,O-dimethylhydroxylamine hydrochloride, in the presence of isopropylmagnesium chloride (i-PrMgCl) [24]. A nucleophilic attack of benzothiazolyl lithium to Weinerb amides 4ah afforded the desired α-heterocyclic ketones 8ah (Figure 3).
Carboxylic acid 3d, required for the synthesis of 8d, was prepared in two steps via etherification of alcohol 9 with tert-butyl 2-bromoacetate, followed by deprotection using trifluoroacetic acid (TFA) (Figure 4).
α-Hydroxy-derivative 12 was synthesized through a reaction between aldehyde 11 and benzothiazolyl lithium (Figure 5) [25].
For the benzoxazoles and benzimidazoles 17ae, the synthesis started from the corresponding aldehydes 13a,b and 11, which were converted to the corresponding cyanohydrins 14a,b and 15. The formation of the heterocyclic rings was accomplished by the treatment of these compounds with 2-aminophenol (for the benzoxazole derivatives 16a,c,d) or 2-phenylenediamine (for the benzimidazole derivatives 16b,e) in the presence of acetyl chloride [26]. Hydroxy compounds 16ae were finally oxidized to the α-ketoheterocycles 17ae using Dess–Martin periodinane (Figure 6).
The synthesis of keto-1,2,4-oxadiazoles was accomplished following a previously published procedure, as depicted in Figure 7. Amidoxime 19 was synthesized from aldehyde 13a through the corresponding O-tert-bytyldimethylsilyl cyanide 18 [21]. It was then coupled with either pivalic acid [21], benzoic acid, or isobutyric anhydride, using N,N′-dicyclohexylcarbodiimide (DCC) as the coupling reagent to afford compounds 20ac. The cyclization of these O-acyl-amidoximes took place in the presence of tetrabutylammonium fluoride (TBAF) under microwave irradiation, giving the desired hydroxy-oxadiazole derivatives 21ac, which were then subjected to oxidation with Dess-Martin periodinane, providing the final α-keto-oxadiazoles 22ac (Figure 7).
In the 1H-NMR spectra of the final heterocyclic compounds, the most characteristic peaks are those corresponding to the protons of the aromatic fused ring, which are located closest to the heteroatoms. These protons are the most downfield shifted aromatic ones, appearing at 8.24−7.92 ppm in the case of α-ketobenzothiazoles 8ah, and at 7.90−7.31 in the case of α-ketobenzoxazoles 17a,c,d and α-ketobenzimidazoles 17b,e. In addition, the 1H-NMR spectra of α-ketobenzimidazoles 17b,e show a characteristic chemical shift of N-H above 10 ppm. In the 13C-NMR spectra of α-ketobenzothiazoles 8a,b,e,f, the carbon atom of the carbonyl group resonates at 195.5–194.4 ppm, while the presence of an oxygen or a sulfur atom at the β-position of the alkyl chain (compounds 8c,d,g,h) causes an upfield shift to 191.2−188.7 ppm. A characteristic chemical shift for the carbonyl carbon atom at 190.2−186.4 ppm, 194.7−194.4 ppm, and 191.9-191.8 ppm, is observed in the 13C-NMR spectra of α-ketobenzoxazoles 17a,c,d, α-ketobenzimidazoles 17b,e, and 22b,c, α-keto-1,2,4-oxadiazoles, respectively.

3.2. Study of the Suppression of PGE2 Generation in Mesangial Cells

Renal mesangial cells were chosen as a model to evaluate the ability of our synthetic compounds to suppress the production of PGE2, based on our previous studies on PLA2 inhibitors [15,16]. Mesangial cells located in the renal glomerulus are involved in various pathological processes, including inflammation, of the renal glomerulus. As shown by Huwiler et al. [27,28], different PLA2s operate in mesangial cells to initiate the generation of PGE2. Stimulation of rat renal mesangial cells by interleukin-1β (IL-1β) plus forskolin (Fκ) results in huge increase of PGE2 synthesis, as previously described [15,16,29]. All the synthetic compounds were tested at a concentration of 3 µM and the results are summarized in Table 1.
The α-ketobenzothiazolyl derivative GK181, where the naphthalene group was placed at a distance of four carbon atoms from the carbonyl group, exhibited 85% inhibition of PGE2 release at a concentration of 3 µM (entry 1). When the distance between the naphthalene and the heterocyclic group was reduced to two carbon atoms (GK517, entry 2), the activity was abolished. Similarly, GK489 (entry 3), where an oxygen atom was introduced at the β-carbon atom to the carbonyl did not present any inhibitory activity. The reduction of the carbonyl group of GK181 to the corresponding alcohol (GK490, entry 4) also destroyed the inhibitory activity, highlighting the importance of the carbonyl group. Replacement of the benzothiazolyl group of GK181 by a benzoxazolyl one (GK491, entry 5) led led to a slight decrease of the activity (77%) in comparison to GK181, while the corresponding benzimidazolyl derivative (GK492, entry 6) exhibited even lower activity (57%).
Then, the naphthalene group was replaced by a p-methoxy-phenyl group. Compound GK299 (entry 7) exhibited a slightly decreased inhibitory activity (79%) in comparison to GK181 (entry 1). Keeping constant the p-methoxy-phenyl group and its distance from the carbonyl (four carbon atoms), the effect of various heterocyclic rings was examined. In all cases (GK355, GK358, GK367 GK368, and GK369, entries 8-12, respectively), the inhibitory potency was either reduced or diminished. Only compounds GK355 and GK368 (entries 8 and 11, respectively) containing a benzoxazolyl or a phenyl substituted oxadiazolyl ring, respectively, presented inhibitory activity (25% and 68%, respectively). Compound GK358 (entry 9), containing a benzimidazolyl group; GK367 (entry 10); and GK369 (entry 12), containing either a tert-butyl or an isopropyl substituted oxadiazolyl ring, did not present any inhibitory activity. Taking into account the results obtained for the various heterocyclic systems, either for the naphthalene-containing compounds or for the p-methoxy-phenyl-containing compounds, it seems that the benzothiazolyl group is the optimum heterocyclic system. The benzoxazole derivative GK453 (entry 13), as well as the benzothiazole derivatives GK455 (entry 14), GK516 (entry 15), GK518 (entry 16), and GK519 (entry 17) were proven unable to cause any inhibition.
The activity of compounds GK181, GK299, and GK491, which exhibited the highest potency at 3 µM, was further explored at various concentrations and the results are shown in Figure 8. GK181, GK299, and GK491 compounds presented potent inhibition of PGE2 generation with EC50 values of 0.71 µM, 1.42 µM, and 0.79 µM, respectively. In conclusion, we have identified one α-ketobenzothiazolyl derivative (GK181) and one α-ketobenzoxazolyl derivative (GK491), being able to inhibit the generation of PGE2 in renal mesangial cells at a nanomolar level.

3.3. Docking Studies

We have previously shown that inhibition of secreted PLA2 is a possible path via which synthetic compounds may evoke the suppression of PGE2 release [15,16]. Thus, we performed docking calculations to understand how these compounds may interact with the active site of secreted GIIA sPLA2. AutoDock Vina [30] was used for docking the most potent compounds GK181 and GK491 in GIIA sPLA2. The crystal structure of the enzyme was retrieved from the Brookhaven Protein Databank (PDB: 1KQU).
As shown in Figure 9 for both GK181 and GK491, the naphthyl group is accommodated at the lipophilic pocket of the site and is involved in a T-shape interaction with His6. The carbonyl group points towards Ca2+ ion and is close enough to Gly29 to form a hydrogen bond. The extended heterocyclic aromatic system is involved in π–π interactions with the catalytic His47. These two models reproduce the key interactions, as known by co-crystallized ligands [31], and therefore may suggest the mode of interactions between either GK181 or GK491 and secreted sPLA2.

4. Conclusions

We present herein the synthesis of eight α-ketobenzothiazoles, using the reaction between benzothiazolyl lithium and the appropriate Weinreb amide as the key step. A series of α-ketobenzoxazoles, α-ketobenzimidazoles, and α-keto-1,2,4-oxadiazoles were also synthesized. All the synthetic heterocycles were evaluated for their ability to suppress the generation of PGE2 in renal mesangial cells after stimulation with IL-1 plus forskolin. Interestingly, two heterocycles were identified, which were found able to inhibit PGE2 formation at a nanomolar level. These structures may serve as leads for the development of novel potent inhibitors of PGE2 formation with potential anti-inflammatory and/or anticancer properties.

Supplementary Materials

The following are available online at https://www.mdpi.com/2218-273X/11/2/275/s1, Copies of 1H NMR and 13C NMR spectra of the final products.

Author Contributions

Conceptualization, G.K. and A.H.; methodology, A.P., M.A.T., M.E., M.M., C.M., S.V., and V.M.; investigation, A.P., M.A.T., M.E., M.M., C.M., S.V., and V.M.; writing—original draft preparation, G.K. and M.A.T.; writing—review and editing, G.K. and A.H. All authors have read and agreed to the published version of the manuscript.

Funding

This research was carried out within the framework of a Stavros Niarchos Foundation grant to the National and Kapodistrian University of Athens (G.K.). This research was funded by the Swiss National Science Foundation (310030_175561, to A.H.).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The data presented in this study are available on request from the corresponding author. The data are not publicly available due to privacy.

Acknowledgments

Evmorfia Kostaki is acknowledged for initial experiments. The authors thank Christos Pappas for recording IR spectra.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Funk, C.D. Prostaglandins and leukotrienes: Advances in eicosanoid biology. Science 2001, 294, 1871–1875. [Google Scholar] [CrossRef] [Green Version]
  2. Dennis, E.A.; Norris, P.C. Eicosanoid storm in infection and inflammation. Nat. Rev. Immunol. 2015, 15, 511–523. [Google Scholar] [CrossRef] [Green Version]
  3. Miller, S.B. Prostaglandins in health and disease: An overview. Semin. Arthritis Rheum. 2006, 36, 37–49. [Google Scholar] [CrossRef]
  4. Dennis, E.A.; Cao, J.; Hsu, Y.-H.; Magrioti, V.; Kokotos, G. Phospholipase A2 enzymes: Physical structure, biological function, disease implication, chemical inhibition, and therapeutic intervention. Chem. Rev. 2011, 111, 6130–6185. [Google Scholar] [CrossRef] [Green Version]
  5. Koeberle, A.; Laufer, S.A.; Werz, O. Design and development of microsomal prostaglandin E2 synthase-1 inhibitors: Challenges and future directions. J. Med. Chem. 2016, 59, 5970–5986. [Google Scholar] [CrossRef] [PubMed]
  6. Psarra, A.; Nikolaou, A.; Kokotou, M.G.; Limnios, D.; Kokotos, G. Microsomal prostaglandin E2 synthase-1 inhibitors: A patent review. Expert Opin. Ther. Pat. 2017, 27, 1047–1059. [Google Scholar] [CrossRef] [PubMed]
  7. Ricciotti, E.; FitzGerald, G.A. Prostaglandins and inflammation. Arterioscler. Thromb. Vasc. Biol. 2011, 31, 986–1000. [Google Scholar] [CrossRef]
  8. Nakanishi, M.; Rosenberg, D.W. Multifaceted roles of PGE2 in inflammation and cancer. Semin. Immunopathol. 2013, 35, 123–137. [Google Scholar] [CrossRef] [PubMed]
  9. Grösch, S.; Niederberger, E.; Geisslinger, G. Investigational drugs targeting the prostaglandin E2 signaling pathway for the treatment of inflammatory pain. Expert Opin. Invest. Drugs 2017, 26, 51–61. [Google Scholar] [CrossRef]
  10. Mizuno, R.; Kawada, K.; Sakai, Y. Prostaglandin E2/EP signaling in the tumor microenvironment of colorectal cancer. Int. J. Mol. Sci. 2019, 20, 6254. [Google Scholar] [CrossRef] [Green Version]
  11. Woolbright, B.L.; Pilbeam, C.C.; Taylor, J.A. Prostaglandin E2 as a therapeutic target in bladder cancer: From basic science to clinical trials. Prostaglandins Other Lipid Mediat. 2020, 148, 106409. [Google Scholar] [CrossRef] [PubMed]
  12. Ye, Y.; Wang, X.; Jeschke, U.; von Schönfeldt, V. COX-2-PGE2-EPs in gynecological cancers. Arch. Gynecol. Obstet. 2020, 301, 1365–1375. [Google Scholar] [CrossRef]
  13. Nikolaou, A.; Kokotou, M.G.; Vasilakaki, S.; Kokotos, G. Small-molecule inhibitors as potential therapeutics and as tools to understand the role of phospholipases A2. Biochim. Biophys. Acta (BBA)-Mol. Cell Biol. Lipids 2019, 1864, 941–956. [Google Scholar] [CrossRef]
  14. Ferrer, M.D.; Busquets-Cortés, C.; Capó, X.; Tejada, S.; Tur, J.A.; Pons, A.; Sureda, A. Cyclooxygenase-2 inhibitors as a therapeutic target in inflammatory diseases. Curr. Med. Chem. 2019, 26, 3225–3241. [Google Scholar] [CrossRef]
  15. Vasilakaki, S.; Barbayianni, E.; Magrioti, V.; Pastukhov, O.; Constantinou-Kokotou, V.; Huwiler, A.; Kokotos, G. Inhibitors of secreted phospholipase A2 suppress the release of PGE2 in renal mesangial cells. Bioorg. Med. Chem. 2016, 24, 3029–3034. [Google Scholar] [CrossRef] [PubMed]
  16. Vasilakaki, S.; Pastukhov, O.; Mavromoustakos, T.; Huwiler, A.; Kokotos, G. Small peptides able to suppress prostaglandin E2 generation in renal mesangial cells. Molecules 2018, 23, 158. [Google Scholar] [CrossRef] [Green Version]
  17. Kokotos, G.; Feuerherm, A.J.; Barbayianni, E.; Shah, I.; Sæther, M.; Magrioti, V.; Nguyen, T.; Constantinou-Kokotou, V.; Dennis, E.A.; Johansen, B. Inhibition of group IVA cytosolic phospholipase A2 by thiazolyl ketones in vitro, ex vivo, and in vivo. J. Med. Chem. 2014, 57, 7523–7535. [Google Scholar] [CrossRef]
  18. Mete, A.; Andrews, G.; Bernstein, M.; Connolly, S.; Hartopp, P.; Jackson, C.G.; Lewis, R.; Martin, I.; Murray, D.; Riley, R.; et al. Design of novel and potent cPLA inhibitors containing an α-methyl-2-ketothiazole as a metabolically stable serine trap. Bioorg. Med. Chem. Lett. 2011, 21, 3128–3133. [Google Scholar] [CrossRef] [PubMed]
  19. Johansen, B.; Kokotos, G.; Magrioti, V.; Tsakos, M. 2-Oxothiazole compounds and method of using same for chronic nflammatory disorders. US9597318B2, 21 March 2017. [Google Scholar]
  20. Aukrust, I.-R.; Barbayianni, E.; Johansen, B.; Kokotos, G.; Sandberg, M. Antiinflammatory and antitumor 2-oxothiazoles and 2-oxothiophenes compounds. WO2014118195A1, 7 August 2014. [Google Scholar]
  21. Mouchlis, V.D.; Limnios, D.; Kokotou, M.G.; Barbayianni, E.; Kokotos, G.; McCammon, J.A.; Dennis, E.A. Development of potent and selective inhibitors for group VIA calcium-independent phospholipase A2 guided by molecular dynamics and structure–activity relationships. J. Med. Chem. 2016, 59, 4403–4414. [Google Scholar] [CrossRef] [Green Version]
  22. Pfeilschifter, J.; Kurtz, A.; Bauer, C. Role of phospholipase C and protein kinase C in vasoconstrictor-induced prostaglandin synthesis in cultured rat renal mesangial cells. Biochem. J. 1986, 234, 125–130. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Angelastro, M.R.; Baugh, L.E.; Bey, P.; Burkhart, J.P.; Chen, T.-M.; Durham, S.L.; Hare, C.M.; Huber, E.W.; Janusz, M.J. Inhibition of human neutrophil elastase with peptidyl electrophilic ketones. 2. Orally active PG-Val-Pro-Val pentafluoroethyl ketones. J. Med. Chem. 1994, 37, 4538–4553. [Google Scholar] [CrossRef]
  24. Trost, B.M.; Gunzner, J.L. Total synthesis of deschlorocallipeltoside A. J. Am. Chem. Soc. 2001, 123, 9449–9450. [Google Scholar] [CrossRef]
  25. Chikashita, H.; Ishibaba, M.; Ori, K.; Itoh, K. General reactivity of 2-lithiobenzothiazole to various electrophiles and the use as a formyl anion equivalent in the synthesis of α-hydroxy carbonyl compounds. Bull. Chem. Soc. Jpn. 1988, 61, 3637–3648. [Google Scholar] [CrossRef] [Green Version]
  26. Edwards, P.D.; Meyer, E.F., Jr.; Vijayalakshmi, J.; Tuthill, P.A.; Andisik, D.A.; Gomes, B.; Strimpler, A. Design, synthesis, and kinetic evaluation of a unique class of elastase inhibitors, the peptidyl α-ketobenzoxazoles, and the x-ray crystal structure of the covalent complex between porcine pancreatic elastase and Ac-Ala-Pro-Val-2-benzoxazole. J. Am. Chem. Soc. 1992, 114, 1854–1863. [Google Scholar] [CrossRef]
  27. Huwiler, A.; Van Rossum, G.; Wartmann, M.; Pfeilschifter, J. Stimulation by extracellular ATP and UTP of the stress-activated protein kinase cascade in rat renal mesangial cells: Nucleotide-stimulated stress-activated protein kinases. Br. J. Pharmacol. 1997, 120, 807–812. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  28. Huwiler, A.; Staudt, G.; Kramer, R.M.; Pfeilschifter, J. Cross-talk between secretory phospholipase A2 and cytosolic phospholipase A2 in rat renal mesangial cells. Biochim. Biophys. Acta (BBA)-Lipids Lipid Metab. 1997, 1348, 257–272. [Google Scholar] [CrossRef]
  29. Huwiler, A.; Feuerherm, A.J.; Sakem, B.; Pastukhov, O.; Filipenko, I.; Nguyen, T.; Johansen, B. The ω3-polyunsaturated fatty acid derivatives AVX001 and AVX002 directly inhibit cytosolic phospholipase A2 and suppress PGE2 formation in mesangial cells: AVX compounds as novel cPLA2 inhibitors. Br. J. Pharmacol. 2012, 167, 1691–1701. [Google Scholar] [CrossRef] [Green Version]
  30. Trott, O.; Olson, A.J. AutoDock Vina: Improving the speed and accuracy of docking with a new scoring function, efficient optimization and multithreading. J. Comput. Chem. 2010, 31, 455–461. [Google Scholar] [CrossRef] [Green Version]
  31. Hansford, K.A.; Reid, R.C.; Clark, C.I.; Tyndall, J.D.A.; Whitehouse, M.W.; Guthrie, T.; McGeary, R.P.; Schafer, K.; Martin, J.L.; Fairlie, D.P. D-Tyrosine as a chiral precusor to potent inhibitors of human nonpancreatic secretory phospholipase A2 (IIa) with antiinflammatory activity. ChemBioChem 2003, 4, 181–185. [Google Scholar] [CrossRef]
Figure 1. Generation of PGE2 through metabolism of arachidonic acid.
Figure 1. Generation of PGE2 through metabolism of arachidonic acid.
Biomolecules 11 00275 g001
Figure 2. Examples of α-ketothiazoles exhibiting anti-inflammatory properties.
Figure 2. Examples of α-ketothiazoles exhibiting anti-inflammatory properties.
Biomolecules 11 00275 g002
Figure 3. Synthesis of α-ketobenzothiazoles 8ah. (a) HN(OMe)Me·HCl, WSCI·HCl, DMAP, NMM, CH2Cl2; (b) (i) HN(OMe)Me·HCl, dry THF, −20 °C; (ii) i-PrMgCl, -20 °C; and (c) (i) n-BuLi, dry Et2O, −78 °C; (ii) Weinreb amides 4ah, dry Et2O, −78 °C to rt.
Figure 3. Synthesis of α-ketobenzothiazoles 8ah. (a) HN(OMe)Me·HCl, WSCI·HCl, DMAP, NMM, CH2Cl2; (b) (i) HN(OMe)Me·HCl, dry THF, −20 °C; (ii) i-PrMgCl, -20 °C; and (c) (i) n-BuLi, dry Et2O, −78 °C; (ii) Weinreb amides 4ah, dry Et2O, −78 °C to rt.
Biomolecules 11 00275 g003
Figure 4. Synthesis of compound 3d. (a) BrCH2COOC(CH3)3, Bu4NHSO4, 50% NaOH, toluene; (b) 50% TFA, dry CH2Cl2.
Figure 4. Synthesis of compound 3d. (a) BrCH2COOC(CH3)3, Bu4NHSO4, 50% NaOH, toluene; (b) 50% TFA, dry CH2Cl2.
Biomolecules 11 00275 g004
Figure 5. Synthesis of compound 12. (a) Benzothiazole, n-BuLi, dry Et2O, −78 °C to rt.
Figure 5. Synthesis of compound 12. (a) Benzothiazole, n-BuLi, dry Et2O, −78 °C to rt.
Biomolecules 11 00275 g005
Figure 6. Synthesis of α-ketobenzoxazoles 17a,c,d and α-ketobenzimidazoles 17b,e. (a) (i) NaHSO3, CH2Cl2; (ii) KCN, H2O; (b) (i) CH3COCl, CHCl3/absolute EtOH; (ii) 2-aminophenol or 2-phenylenediamine, absolute EtOH; (c) Dess–Martin periodinane, CH2Cl2.
Figure 6. Synthesis of α-ketobenzoxazoles 17a,c,d and α-ketobenzimidazoles 17b,e. (a) (i) NaHSO3, CH2Cl2; (ii) KCN, H2O; (b) (i) CH3COCl, CHCl3/absolute EtOH; (ii) 2-aminophenol or 2-phenylenediamine, absolute EtOH; (c) Dess–Martin periodinane, CH2Cl2.
Biomolecules 11 00275 g006
Figure 7. Synthesis of α-keto-1,2,4-oxadiazoles 22ac. (a) TBDMSCN, KCN, 18-crown-6, dry CH2Cl2; (b) 50% aq. NH2OH, microwave irradiation 50 W, 120 °C; (c) pivalic acid (for pivalate group) or benzoic acid (for benzoate group) or isobutyric anhydride (for isobutyrate group), DCC, dry CH2Cl2; (d) TBAF, toluene, microwave irradiation 90 W, 120 °C; (e) Dess–Martin periodinane, CH2Cl2.
Figure 7. Synthesis of α-keto-1,2,4-oxadiazoles 22ac. (a) TBDMSCN, KCN, 18-crown-6, dry CH2Cl2; (b) 50% aq. NH2OH, microwave irradiation 50 W, 120 °C; (c) pivalic acid (for pivalate group) or benzoic acid (for benzoate group) or isobutyric anhydride (for isobutyrate group), DCC, dry CH2Cl2; (d) TBAF, toluene, microwave irradiation 90 W, 120 °C; (e) Dess–Martin periodinane, CH2Cl2.
Biomolecules 11 00275 g007
Figure 8. Effect of compounds (A) GK181, (B) GK299, and (C) GK491 on IL-1/Fk-stimulated PGE2 formation in mesangial cells. Cells were pretreated for 20 min with the indicated concentrations of GK compounds and then stimulated for 24 h in the absence (−) or presence (+) of 1 nM interleukin 1β(IL-1) plus 5 µM forskolin (Fk). Supernatants were taken for PGE2 quantification using an enzyme-linked immunoassay as described in the Methods section. Data are presented as % of maximal IL-1/Fk stimulation and are means S.D. (n = 3). *** p < 0.001 considered statistically significant when compared to the unstimulated samples; # p < 0.05, ## p < 0.01, ### p < 0.001 compared to the IL-1/Fk-stimulated samples.
Figure 8. Effect of compounds (A) GK181, (B) GK299, and (C) GK491 on IL-1/Fk-stimulated PGE2 formation in mesangial cells. Cells were pretreated for 20 min with the indicated concentrations of GK compounds and then stimulated for 24 h in the absence (−) or presence (+) of 1 nM interleukin 1β(IL-1) plus 5 µM forskolin (Fk). Supernatants were taken for PGE2 quantification using an enzyme-linked immunoassay as described in the Methods section. Data are presented as % of maximal IL-1/Fk stimulation and are means S.D. (n = 3). *** p < 0.001 considered statistically significant when compared to the unstimulated samples; # p < 0.05, ## p < 0.01, ### p < 0.001 compared to the IL-1/Fk-stimulated samples.
Biomolecules 11 00275 g008
Figure 9. Proposed binding modes of GK181 (left) and GK491 (right) in the active site of GIIA sPLA2 (PDB:1KQU). The purple ball is Ca2+, which coordinates with Asp48 and the glycine loop (Gly29, Gly31). The inhibitors are involved in π–π interactions with His6 and the catalytic His47.
Figure 9. Proposed binding modes of GK181 (left) and GK491 (right) in the active site of GIIA sPLA2 (PDB:1KQU). The purple ball is Ca2+, which coordinates with Asp48 and the glycine loop (Gly29, Gly31). The inhibitors are involved in π–π interactions with His6 and the catalytic His47.
Biomolecules 11 00275 g009
Table 1. Compounds tested for their inhibition of PGE2 generation at 3 μM.
Table 1. Compounds tested for their inhibition of PGE2 generation at 3 μM.
Entry.Compound
(Code Number)
Structure% Inhibition
(at 3 µM)
18a
(GK181)
Biomolecules 11 00275 i00185
28f
(GK517)
Biomolecules 11 00275 i002no inhibition
38d
(GK489)
Biomolecules 11 00275 i003no inhibition
412
(GK490)
Biomolecules 11 00275 i004no inhibition
517d
(GK491)
Biomolecules 11 00275 i00577
617e
(GK492)
Biomolecules 11 00275 i00657
78b
(GK299)
Biomolecules 11 00275 i00779
817a
(GK355)
Biomolecules 11 00275 i00825
917b
(GK358)
Biomolecules 11 00275 i009no inhibition
1022a
(GK367)
Biomolecules 11 00275 i010no inhibition
1122b
(GK368)
Biomolecules 11 00275 i01168
1222c
(GK369)
Biomolecules 11 00275 i012no inhibition
1317c
(GK453)
Biomolecules 11 00275 i013no inhibition
148c
(GK455)
Biomolecules 11 00275 i014no inhibition
158e
(GK516)
Biomolecules 11 00275 i015no inhibition
168g
(GK518)
Biomolecules 11 00275 i016no inhibition
178h
(GK519)
Biomolecules 11 00275 i017no inhibition
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Psarra, A.; Theodoropoulou, M.A.; Erhardt, M.; Mertiri, M.; Mantzourani, C.; Vasilakaki, S.; Magrioti, V.; Huwiler, A.; Kokotos, G. α-Ketoheterocycles Able to Inhibit the Generation of Prostaglandin E2 (PGE2) in Rat Mesangial Cells. Biomolecules 2021, 11, 275. https://doi.org/10.3390/biom11020275

AMA Style

Psarra A, Theodoropoulou MA, Erhardt M, Mertiri M, Mantzourani C, Vasilakaki S, Magrioti V, Huwiler A, Kokotos G. α-Ketoheterocycles Able to Inhibit the Generation of Prostaglandin E2 (PGE2) in Rat Mesangial Cells. Biomolecules. 2021; 11(2):275. https://doi.org/10.3390/biom11020275

Chicago/Turabian Style

Psarra, Anastasia, Maria A. Theodoropoulou, Martin Erhardt, Marina Mertiri, Christiana Mantzourani, Sofia Vasilakaki, Victoria Magrioti, Andrea Huwiler, and George Kokotos. 2021. "α-Ketoheterocycles Able to Inhibit the Generation of Prostaglandin E2 (PGE2) in Rat Mesangial Cells" Biomolecules 11, no. 2: 275. https://doi.org/10.3390/biom11020275

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop