Next Article in Journal
2-(3,4-Dimethoxyphenyl)-N-(4-methoxyphenyl)-1-propyl-1H-benzo[d]imidazole-5-carboxamide
Previous Article in Journal
2-(3,5-Dimethyl-1H-pyrazol-1-yl)thiazolo[4,5-b]pyridine
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Communication

A-Ring-Modified Triterpenoids and Their Spermidine–Aldimines with Strong Antibacterial Activity

by
Oxana B. Kazakova
1,*,
Jean Michel Brunel
2,*,
Elmira F. Khusnutdinova
1,
Sophie Negrel
2,
Gulnara V. Giniyatullina
1,
Tatyana V. Lopatina
1 and
Anastasiya V. Petrova
1
1
Ufa Institute of Chemistry of the Ufa Federal Research Centre of the Russian Academy of Sciences, 71 pr. Oktyabrya, Ufa 450054, Russia
2
Aix Marseille Université, INSERM, SSA, MCT, 13385 Marseille, France
*
Authors to whom correspondence should be addressed.
Molbank 2019, 2019(3), M1078; https://0-doi-org.brum.beds.ac.uk/10.3390/M1078
Submission received: 2 August 2019 / Revised: 3 September 2019 / Accepted: 5 September 2019 / Published: 6 September 2019
(This article belongs to the Section Natural Products)

Abstract

:
Synthesis of A-ring-modified lupane, oleanane and ursane type triterpenoid conjugates with spermidine through an aldimine linkage or diethylentriamine via an amide bond is described. These derivatives were evaluated for their in vitro antimicrobial properties against human pathogens. Except for derivatives 1 and 7, all compounds have moderate to weak minimum inhibitory concentrations (MICs) against Gram-positive Staphylococcus aureus bacteria, with MICs varying from 3.125 to 200 µM. Compound 11 is efficient against Escherichia coli and Pseudomonas aeruginosa, with MICs of 25 and 50 µM, respectively, while all other derivatives do not possess important antimicrobial activities against these Gram-negative bacteria.

Graphical Abstract

1. Introduction

Pentacyclic triterpenoids are widely represented in natural products and are useful substrates for the synthesis of various important bioactive compounds [1,2,3]. Conjugation of a triterpenoid scaffold with amines, amino acids, and polyamines resulted in a series of anticancer, antimicrobial and antiviral agents [4,5,6,7,8]. Chemical antibiotics were one of the great health successes of the 20th century leading to a huge decrease in both morbidity and mortality from bacterial infections. However, this has led to high levels of inappropriate prescribing, which has contributed to a recent rise in the number of antibiotic-resistant bacteria. As a result, multidrug-resistant bacteria such as vancomycin-resistant Enterococcus (VRE), methicillin-resistant Staphylococcus aureus (MRSA), and multidrug-resistant Pseudomonas aeruginosa (MRPA) have appeared [9]. In this context, new types of antibiotics such as antibiotic peptides, lipids, and alkaloids have been isolated as host defense agents from diverse animal species [10,11,12]. Among these substances, two water-soluble cationic steroids, squalamine, and trodusquemine, have been identified from the dogfish shark Squalus acanthias, exhibiting excellent antimicrobial activities (Figure 1) [13,14,15].
Since that time, the synthesis of the triterpenoid analog of squalamine has been accomplished [16], and novel steroidal polyamines, such as Claramine, that exhibit a broad-spectrum antimicrobial bimodal activity, have been discovered [17,18]. Polyamines (putrestycine, spermidine, spermine, etc.) play an important role in the physiology of plants and live organisms. They are polycations that interact with negatively charged molecules such as DNA, RNA, and proteins. Betulinic acid-based amides with putrescine and spermine demonstrate a high level of cytotoxicity and antimicrobial activity [19]. At the same time, there are no reports on the synthesis of spermidine conjugates connected to the triterpenoid core via the aldimine group.
In this work, the first synthesis of triterpenoid spermidino-aldimines, as well as some derivatives with an A-azepano-ring and C28-(diethylentriamino)-amides is described and the study of their antimicrobial activity is presented.

2. Results and Discussion

2.1. Chemistry

The synthesis of a series of nitrogen-containing triterpenoids is presented in Scheme 1 and Scheme 2. Azepanobetulin 1 was obtained by several stages from naturally-occurring betulin [20]. The following p-TsOH catalyzed rearrangement of compound 1 resulted in azepanoallobetulin 2 with a yield of 86%. Reduction of lupane A-azepanono-C28-nitryl 3 [21] and 2-cyano-3,4-seco-ursolic acid 4 [22] with LiAlH4 under reflux in tetrahydrofurane led to methylenamino-derivatives 5 and 6. Betulin and C28-hydroxy-triterpenoids 1 or 6 were converted to corresponding aldehydes and were reacted with spermidine in MeOH under reflux to obtain aldimines 79 in yields of 65–72%. The reaction of ursonic or oleanonic acid chlorides with diethylenetriamine led to amides 10 and 11 in yields of 85 and 73%, respectively. The data for the latter amide have been previously presented [23].
The structure of the compounds was confirmed by NMR spectroscopy. The 13C-NMR spectra of azepanes 2, 5 and 8 showed signals of C-3 at dC 62–63 ppm. For aldimines 79, the signals of the –CH=N– function were detected at δ 169–173 ppm (13C-NMR) and at δ 7.3–7.8 ppm (1H-NMR). In the spectra of compound 10, the signal of amide function C(O)NH was detected at δ 178.9 ppm (13C-NMR) and at δ 6.61 ppm (1H-NMR). Finally, the presence of polyamine moieties at position C-28 of the triterpene core in the structure of compounds 711 was confirmed by the characteristic signals of the aminomethylene groups –CH2NH– at δ 2.58–3.70 ppm (1H-NMR) (see Supplementary Materials).

2.2. Biology

All the synthesized compounds were screened for antimicrobial activity against both Gram-positive and Gram-negative bacterial strains and found to possess activities (Table 1). Thus, except for derivatives 1 and 7, all compounds have moderate to weak minimum inhibitory concentrations against Gram-positive S. aureus bacteria with MICs varying from 3.125 to 200 µM. Nevertheless, compound 11 is efficient against E. coli and P. aeruginosa, with MICs of 25 and 50 µM, respectively, while all other derivatives do not possess important antimicrobial activities against these Gram-negative bacteria. It is noteworthy that the minimum bactericidal activity remains of interest for all products active against S. aureus. These data clearly suggest that the nature of the triterpenoid involved plays an important role in the potential activities of our compounds.

3. Materials and Methods

3.1. General Methods and Physical Measurements

The spectra were recorded at the Center for the Collective Use ‘Chemistry’ of the Ufa Institute of Chemistry of the Ufa Federal Research Centre of the Russian Academy of Sciences. 1H and 13C-NMR spectra were recorded on a “Bruker AM-500” (Bruker, Billerica, MA, USA, 500 and 125.5 MHz respectively, δ, ppm, Hz) in CDCl3, internal standard tetramethylsilane. Mass spectra were obtained on a liquid chromatograph–mass spectrometer LCMS-2010 EV (Shimadzu, Kyoto, Japan). Melting points were detected on a micro table “Rapido PHMK05“ (Nagema, Dresden, Germany). Optical rotations were measured on a polarimeter “Perkin-Elmer 241 MC” (PerkinElmer, Waltham, MA, USA) in a tube length of 1 dm. Elemental analysis was performed on a Euro EA-3000 CHNS analyzer (Eurovector, Milan, Italy); the main standard is acetanilide. Thin-layer chromatography analyses were performed on Sorbfil plates (Sorbpolimer, Krasnodar, Russian Federation), using the solvent system chloroform–ethyl acetate, 40:1. Substances were detected by 10% H2SO4 with subsequent heating to 100–120 °C for 2–3 min. Betulonic aldehyde [24], compounds 1 [20], 3 [21], ursonic acid and 4 [22], 11 [23] were obtained according to the methods described previously.

3.2. Synthesis of 3-Deoxy-3a-homo-3a-aza-19β,28-epoxy-18α-oleanane (2)

A mixture of compound 1 (220 mg, 0.5 mmol) and a catalytic amount of p-TsOH (8.6 mg, 0.05 mmol) in dry CHCl3 (25 mL) was heated under reflux for 4 h, and then the solvent was evaporated under reduced pressure. The product was purified by Al2O3 column chromatography using CHCl3 and a mixture of CHCl3–EtOH (100:1) as eluents to afford compound 2 as a beige powder (190 mg, 86%): [α]D20 +54 (c 0.05, CHCl3), m.p. 261 °C. 1H-NMR (δ, ppm, CDCl3, 125.5 MHz): 3.62 and 3.45 (2H, both d, H-28, J = 7.7 Hz), 3.41 (1H, s, H-19), 3.21–3.16 (1H, m, H3b), 2.91–2.82 (1H, m, H3a), 2.02–1.16 (26H, m, CH, CH2), 1.42, 1.29, 1.01, 0.92, 0.81, 0.74, 0.69 (21H, all s, 7CH3). 13C-NMR (δ, ppm, CDCl3, 500 MHz): 90.0 (C-19), 72.4 (C-28), 63.0 (C-3), 54.5 (C-9), 48.2 (C-18), 46.6 (C-4), 41.6 (C-5), 41.2, 40.9, 39.9, 36.5, 36.1, 34.4, 33.0, 32.5, 31.5, 28.6, 27.7, 26.6, 26.2, 25.8, 24.4, 22.9, 22.8, 22.0, 21.6, 19.9, 16.2, 15.9, 13.1. Anal. Calcd for C30H51NO: C, 81.57; H, 11.64; N, 3.17. Found: C, 81.60; H, 11.54; N, 3.16. MS (APCI): m/z [M + H]+ 442.75, calcd for C30H51NO: 441.74.

3.3. Synthesis of 5 and 6

A mixture of compound 3 [21] (225 mg, 0.5 mmol) or compound 4 [22] (226 mg, 0.5 mmol) and LiAlH4 (230 mg, 0.65 mmol) in dry THF (20 mL) was refluxed for 3 h and then poured into a 5% HCl solution (100 mL). The crude product was extracted with CHCl3 (3 × 40 mL), and then the organic layer was washed with H2O, dried over CaCl2 and evaporated under reduced pressure. The product was purified by Al2O3 column chromatography using CHCl3 and a mixture of CHCl3–EtOH (100:1; 50:1, 25:1, 10:1) as eluents to afford compound 5 (187 mg, 85%) or compound 6 (174 mg, 79%) as white powders.
3-Deoxy-3a-homo-3a-aza-lup-20(29)-en-28-methylenamine (5): [α]D20 +96 (c 0.05, CHCl3), m.p. 246 °C. 1H-NMR (δ, ppm, CDCl3, 125.5 MHz): 4.68 and 4.58 (2H, both s, J = 2.0 Hz, H-29), 3.80–3.78 (1H, m, H3b), 3.35–3.29 (1H, m, H3a), 2.90–2.78 (2H, m, H-28), 2.40−1.65 (28H, m, CH, CH2), 1.65 (3H, s, H-30), 1.41, 1.17, 1.05, 0.99, 0.85 (15H, all s, 5CH3). 13C-NMR (δ, ppm, CDCl3, 500 MHz): 151.0 (C-20), 109.2 (C-29), 63.2 (C-3), 60.5, 54.4, 48.5, 47.8, 47.6, 47.4 (C-28), 43.0, 41.2, 40.9, 40.7, 37.7, 34.0, 29.7, 29.0, 27.9, 26.9, 25.8, 23.0, 22.9, 22.7, 22.0, 21.6, 21.3, 19.1, 16.6, 16.4, 14.5. Anal. Calcd for C30H52N2: C, 81.75; H, 11.89; N, 6.36. Found: C, 81.69; H, 11.84; N, 6.30. MS (APCI): m/z [M + H]+ 441.73, calcd for C30H52N2: 440.75.
3-Amino-3,4-seco-28-hydroxy-urs-12(13)-en (6): [α]D20 +54 (c 0.05, CHCl3), m.p. 195 °C. 1H-NMR (δ, ppm, CDCl3+C5D5N, 125.5 MHz): 5.10 (1H, s, H-12), 4.82 and 4.78 (2H, both d, H-23, J = 2.8 Hz), 3.20 and 3.40 (2H, both d, H-28), 3.10 (2H, m, CH2NH2), 2.15−1.00 (25H, m, CH and CH2), 1.65 (3H, s, CH3, H-24), 1.22, 1.00, 0.92, 0.78, 0.66 (15H, all s, 5CH3). 13C-NMR (δ, ppm, CDCl3+C5D5N, 500 MHz): 148.7 (C-4), 139.2 (C-13), 123.3 (C-12), 115.4 (C-23), 69.9 (C-28), 51.8, 48.4, 43.9, 43.6, 41.8, 40.9, 40.6, 39.4, 38.7, 37.7, 35.8, 34.8, 32.9, 32.6, 32.5, 27.4, 27.1, 25.8, 25.2, 25.1, 24.6, 23.8, 23.0, 21.2, 18.1. Anal. Calcd for C30H51NO: C, 81.57; H, 11.64; N, 3.17. Found: C, 81.49; H, 11.70; N, 3.09. MS (APCI): m/z [M + H]+ 442.73, calcd for C30H51NO: 441.74.

3.4. Synthesis of Compounds 79

To a solution of compound 1 (221 mg, 0.5 mmol) or compound 6 (220 mg, 0.5 mmol) in CH2Cl2 (25 mL) pyridinium chlorochromate (PCC) (8.6 mg, 1.2 mmol) was added and the mixture was stirred at room temperature until the starting material disappeared (monitored by TLC). After completion of the reaction, the mixture was passed through Al2O3, the organic phase was washed with H2O (2 × 30 mL), dried over CaCl2 and evaporated under reduced pressure to yield crude aldehydes. Then, to a solution of betulonic aldehyde (110 mg, 0.25 mmol) [24] or freshly prepared above aldehydes in anhydrous MeOH (25 mL), spermidine (0.05 mL, 0.25 mmol)) was added and the reaction mixture was heated under reflux for 8 h, then poured into H2O (50 mL) and the precipitate was filtered off, washed with water, and dried in air. The products were chromatographed over Al2O3 using CHCl3 and a mixture of CHCl3–EtOH (100:1; 50:1) as eluents to afford compounds 7 (100 mg, 65%) as a yellow powder, 8 (102 mg, 72%) as a beige powder or 9 (109 mg, 70%) as an orange powder.
3-Oxo-lup-20(29)-en-28-N-(4-aminobutyl)-N-propylamin-28-imine (7): [α]D20 +3 (c 0.25, CHCl3), m.p. 201 °C. 1H-NMR (δ, ppm, CDCl3, 125.5 MHz): 7.82 (1H, s, CH=N), 4.62 and 4.73 (2H, both d, J = 2.0 Hz, H-29), 3.50‒2.68 (8H, m, 4CH2N), 2.10−1.73 (34H, m, CH, CH2), 1.72 (3H, s, H-30), 1.09, 1.08, 1.01, 0.98, 0.89 (15H, all s, 5CH3). 13C-NMR (δ, ppm, CDCl3, 500 MHz): 219.8 (C-3), 170.9 (C-28), 150.0 (C-20), 110.1 (C-29), 57.3, 55.1, 49.8, 49.5, 49.3, 48.9, 48.0, 47.9, 47.4, 41.4, 41.2, 39.9, 39.6, 39.0, 37.7, 37.5, 34.3, 33.6, 32.2, 31.9, 30.8, 30.7, 29.0, 28.1, 27.5, 26.7, 25.5, 21.5, 21.0, 19.2, 16.6, 15.7, 14.9. Anal. Calcd for C37H63N3O: C, 78.53; H, 11.22; N, 7.43. Found: C, 78.49; H, 11.25; N, 7.40. MS (APCI): m/z [M + H]+ 566.80, calcd for C37H63N3O: 565.92.
3-Deoxy-3a-homo-3a-aza-lup-20(29)-en-28-N-(4-aminobutyl)-N-propylamin-28-imine (8): [α]D20 +85 (c 0.1, CHCl3), m.p. 185 °C. 1H-NMR (δ, ppm, CDCl3, 500 MHz): 7.78 (1H, s, CH=N), 4.60 and 4.72 (2H, both d, J = 2.0 Hz, H-29), 3.48–3.32 (2H, m, H3), 2.80–2.58 (8H, m, 4CH2N), 2.10−1.42 (35H, m, CH, CH2), 1.72 (3H, s, H-30), 1.09, 1.08, 1.01, 0.98, 0.89 (15H, all s, 5CH3). 13C-NMR (δ, ppm, CDCl3, 125.5 MHz): 169.1 (C-28), 150.2 (C-20), 109.8 (C-29), 62.3 (C-3), 58.0, 55.0, 52.2, 50.0, 48.7, 48.1, 47.8, 47.7, 42.9, 42.5, 41.4, 41.3, 39.2, 39.1, 37.4, 34.3, 34.0, 32.0, 31.5, 31.0, 30.1, 29.7, 29.4, 29.1, 28.0, 27.3, 26.9, 26.3, 23.2, 19.2, 16.8, 16.6, 14.3. Anal. Calcd for C37H66N4: C, 78.38; H, 11.73; N, 9.88. Found: C, 78.32; H, 11.70; N, 9.85. MS (APCI): m/z [M + H]+ 567.93, calcd for C37H66N4: 566.95.
3-Amino-3,4-seco-urs-12(13)-en-28-N-(3-aminopropyl)-N-(butane-1,4-diamine)-N-propylamin-28-imine (9): [α]D20 +91 (c 0.1, CHCl3), m.p. 145 °C. 1H-NMR (δ, ppm, CDCl3, 125.5 MHz): 7.31 (1H, s, CH=N, H-28), 5.10 (1H, s, H-12), 4.79 and 4.59 (2H, both d, H-23, J = 2.8 Hz), 3.68−2.58 (8H, m, 4CH2N), 2.00−1.63 (36H, m, CH and CH2), 1.62 (3H, s, CH3, H-24), 1.18, 0.99, 0.90, 0.74, 0.65 (15H, all s, 5CH3). 13C-NMR (δ, ppm, CDCl3, 500 MHz): 173.6 (C-28), 147.9 (C-4), 139.5 (C-13), 125.2 (C-12), 113.2 (C-23), 54.2, 52.2, 50.4, 42.6, 41.6, 41.4, 40.6, 39.7, 39.1, 38.3, 37.7, 36.5, 35.3, 33.9, 32.3, 31.8, 31.6, 30.6, 29.7, 29.3, 28.0, 26.0, 25.6, 24.4, 23.6, 23.3, 23.0, 21.9, 21.3, 20.7, 20.1, 19.9. Anal. Calcd for C37H66N4: Found: C, 78.38; H, 11.73; N, 9.88. Found: C, 78.29; H, 11.84; N, 9.79. MS (APCI): m/z [M + H]+ 567.94, calcd for C37H66N4: 566.95.

3.5. Synthesis of 3-oxo-urs-12(13)-en-28-N-(2-((2-aminoethyl)amino)ethyl)-2-ethylamide (10)

A solution of ursonic acid [22] (225 mg, 0.5 mmol) in dry CH2Cl2 (20 mL) and (COCl)2 (1.5 mmol, 0.13 mL) was stirred at room temperature for 2 h and then was concentrated to dryness under reduced pressure. A resulting ursonic acid chloride (235 mg, 0.5 mmol) was dissolved in dry CH2Cl2 (30 mL) and treated with diethylenetriamine (0.05 mL, 0.5 mmol) and Et3N (0.07 mL, 0.5 mmol). It was then stirred at room temperature for 3 h, washed with 5% HCl solution (2 × 50 mL) and H2O (50 mL), dried over CaCl2, and the solvent was removed under reduced pressure. The product was chromatographed over a column of Al2O3 using CHCl3 and a mixture of CHCl3–EtOH (100:1; 70:1, 40:1) as eluents to afford compound 10 (229 mg, 85%) as a yellow powder. [α]D20 +35 (c 0.1, CHCl3), m.p. 178 °C. 1H-NMR (δ, ppm, CDCl3, 125.5 MHz): 6.61 (1H, s, CONH), 5.30 (1H, s, H-12), 4.50−4.25 (3H, m, NH, NH2), 3.70−2.70 (8H, m, 4CH2), 2.55−1.36 (23H, m, CH and CH2), 1.35, 1.19, 1.10, 0.99, 0.89, 0.75, 0.65 (21H, all s, 7CH3). 13C-NMR (δ, ppm, CDCl3, 500 MHz): 217.6 (C-3), 178.9 (C-28), 139.4 (C-13), 126.8 (C-12), 58.1, 55.2, 54.9, 53.4, 52.5, 48.3, 47.4, 46.7, 42.2, 39.7, 39.5, 38.9, 37.3, 36.6, 34.1, 32.6, 30.9, 28.1, 27.8, 26.6, 24.8, 24.6, 23.5, 23.3, 21.2, 19.6, 19.1, 18.8, 18.4, 17.2. Anal. Calcd for C34H57N3O2: C, 75.65; H, 10.64; N, 7.78. Found: C, 75.59; H, 10.58; N, 7.70. MS (APCI): m/z [M + H]+ 540.82, calcd for C34H57N3O2: 539.84.

4. Biology Methods

Determination of Minimal Inhibitory Concentrations

The antimicrobial activity of the compounds 1, 2, 5, and 711 was studied by determination of minimal inhibitory concentrations (MICs) according to the NCCLS guidelines M7-A2 using the microbroth dilution methods. All of the strains were issued from the Institute Pasteur collection (Paris, France). The bacteria strains were grown on trypticase soy agar (Becton Dickinson) at 37 °C for 24 h (E. coli ATCC25299, S. aureus DSM789, P. aeruginosa (PA01)) in MHII broth for P. aeruginosa, E. coli and S. aureus. Inocula were prepared in MHII by adjusting the turbidity at 623 nm to obtain 106 CFU/mL.
Antimicrobial activities of the compounds were determined by using a broth microdilution method performed in sterile 96-well microplates. All compounds were solubilized in methanol at a concentration of 5 mg/mL and were transferred to each microplate well (in all cases concentrations of the desired molecules in methanol did not exceed 2% of the total proportion). In order to obtain a two-fold serial dilution, 100 µL of broth and 100 µL of inocula containing 5 × 105–106 CFU of each bacteria were added to each well. Several wells were reserved for positive controls, inoculum viability, and solvent effect. After 24 h incubation, growth was assayed by absorbance measurement at 623 nm with an IEMS Labsystem automatic plate reader. MIC was defined for each agent from triplicate observations as the lowest concentration of compound allowing no visible growth.

5. Conclusions

Finally, a series of triterpenoid C-28 polyamine conjugates or A-azepanes was synthesized and evaluated for their antimicrobial activity. Among them, oleanonic acid amide with diethylentriamine appears to be an interesting antimicrobial candidate against both Gram-positive and Gram-negative bacteria. Current studies are underway to evaluate the potentiality of such derivatives in vivo, by determining the cytotoxicity of these compounds, establishing the mechanism of action of this new class of antimicrobial agents and designing more active derivatives by varying the nature of the polyamine chain involved.

Supplementary Materials

The following are available online, Figures S1–S18: 1H and 13C spectra for compounds 1, 2, 511, Scheme S1: Full synthetic route from betulin to compounds 1, 2 and 8 and from betulonic aldehyde to compounds 3, 5 and 7, Scheme S2: Full synthetic route from ursonic and oleanonic acids to compounds 4, 6 and 911.

Author Contributions

O.B.K. brought the idea, managed the research, conducted some synthetic experiments and prepared the manuscript; S.N. conducted some biological experiments, E.F.K. managed the research and prepared the manuscript; G.V.G. and T.V.L. conducted some synthetic experiments; A.V.P. made structure elucidation and prepared the manuscript; J.M.B. brought the idea, conducted biological experiments and prepared the manuscript.

Funding

The synthesis of compounds 13, 5 and 8 was supported by the Russian Foundation for Basic Research (project no. 18-33-00364 mol_a to Tatyana V. Lopatina).

Acknowledgments

This work was supported by the Ministry of Education of the Russian Federation, State task projects no. AAAA-A17-117011910023-2 and AAAA-A19-119020890014-7. The study of antimicrobial activity was supported by Aix-Marseille Université.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Salvador, J.A.R.; Leal, A.S.; Valdeira, A.S.; Gonçalves, B.M.F.; Alho, D.P.S.; Figueiredo, S.A.C.; Silvestre, S.M.; Mendes, V.I.S. Oleanane-, ursane-, and quinone methide friedelane-type triterpenoid derivatives: Recent advances in cancer treatment. Eur. J. Med. Chem. 2017, 142, 95–130. [Google Scholar] [CrossRef] [PubMed]
  2. Sousa, J.L.C.; Freire, C.S.R.; Silvestre, A.J.D.; Silva, A.M.S. Recent developments in the functionalization of betulinic acid and its natural analogues: A route to new bioactive compounds. Molecules 2019, 24, 355. [Google Scholar] [CrossRef] [PubMed]
  3. Kvasnica, M.; Urban, M.; Dickinson, N.J.; Sarek, J. Pentacyclic triterpenoids with nitrogen- and sulfur-containing heterocycles: Synthesis and medicinal significance. Nat. Prod. Rep. 2015, 32, 1303–1330. [Google Scholar] [CrossRef] [PubMed]
  4. Smirnova, I.E.; Kazakova, O.B. Structure – Anti-influenza Type a Activity Relationship among a Series of Nitrogen Lupane Triterpenoids. Nat. Prod. Commun. 2018, 13, 1267–1270. [Google Scholar] [CrossRef]
  5. Bildziukevich, U.; Rarova, L.; Janovska, L.; Saman, D.; Wimmer, Z. Enhancing effect of cystamine in its amides with betulinic acid as antimicrobial and antitumor agent in vitro. Steroids 2019, 148, 91–98. [Google Scholar] [CrossRef] [PubMed]
  6. Khusnutdinova, E.F.; Apryshko, G.N.; Petrova, A.V.; Kukovinets, O.S.; Kazakova, O.B. The synthesis and selective cytotoxicity of new Mannich bases derivatives of 19- and 28-alkynyltriterpenoids. Russ. J. Bioorg. Chem. 2018, 1, 123–127. [Google Scholar] [CrossRef]
  7. Bildziukevich, U.; Kaletova, E.; Saman, D.; Sievanen, E.; Kolehmainen, E.T.; Slouf, M.; Wimmer, Z. Spectral and microscopic study of self-assambly of novel cationic spermine amides of betulinic acid. Steroids 2017, 117, 90–96. [Google Scholar] [CrossRef]
  8. Kahnt, M.; Hoenke, S.; Fischer, L.; Al-Harrasi, A.; Csuk, R. Synthesis and cytotoxicity evaluation of DOTA-conjugates of ursolic acid. Molecules 2019, 24, 2254. [Google Scholar] [CrossRef]
  9. Foster, T.J. The Staphylococcus aureus “superbug”. J. Clin. Investig. 2004, 114, 1693–1696. [Google Scholar] [CrossRef]
  10. Boman, H.G. Antibacterial peptides: Key components needed in immunity. Cell 1991, 65, 205–207. [Google Scholar] [CrossRef]
  11. Zasloff, M. Antibacterial molecules from frogs, sharks and man. In Phylogenetic Perspective in Immunity: The Insect-Host Defense; Hoffmann, J., Natori, S., Janeway, C., Eds.; RG Landes Biomedical Publisher: Austin, TX, USA, 1994; pp. 31–41. [Google Scholar]
  12. Cho, J.; Kim, Y. Sharks: A potential source of antiangiogenic factors and tumor treatments. Marine Biotechnology 2002, 4, 521–525. [Google Scholar] [CrossRef] [PubMed]
  13. Moore, K.S.; Wehrli, S.; Roder, H.; Rogers, M.; Forrest, J.N., Jr.; McCrimmon, D.; Zasloff, M. Squalamine: An aminosterol antibiotic from the shark. Proc. Natl. Acad. Sci. USA 1993, 90, 1354–1358. [Google Scholar] [CrossRef] [PubMed]
  14. Brunel, J.M.; Letourneux, Y. Recent advances in the synthesis of spermine and spermidine analogs of the shark aminosterol squalamine. Eur. J. Org. Chem. 2003, 3897–3907. [Google Scholar] [CrossRef]
  15. Alhanout, K.; Rolain, J.M.; Brunel, J.M. Squalamine as an example of a new potent antimicrobial agent class: A critical review. Curr. Med. Chem. 2010, 17, 3909–3917. [Google Scholar] [CrossRef] [PubMed]
  16. Kazakova, O.B.; Giniyatullina, G.V.; Medvedeva, N.I.; Tolstikov, G.A. Synthesis of a triterpene-spermidine conjugate. Russ. J. Bioorg. Chem. 2012, 45, 1366–1369. [Google Scholar] [CrossRef]
  17. Djouhri-Bouktab, L.; Vidal, N.; Rolain, J.M.; Brunel, J.M. Synthesis of new 3,20-bispolyaminosteroid squalamine analogues and evaluation of their antimicrobial activities. J. Med. Chem. 2011, 54, 7417–7421. [Google Scholar] [CrossRef]
  18. Blanchet, M.; Borselli, D.; Rodallec, A.; Peiretti, F.; Vidal, N.; Bolla, J.M.; Digiorgio, C.; Morrison, K.R.; Wuest, W.M.; Brunel, J.M. Claramines: A new class of broad-spectrum antimicrobial agents with bimodal activity. Chem. Med. Chem. 2018, 13, 1018–1027. [Google Scholar] [CrossRef]
  19. Bildziukevich, U.; Vida, N.; Rarova, L.; Kolar, M.; Saman, D.; Havlicek, L.; Drasar, P.; Wimmer, Z. Polyamine derivatives of betulinic acid and β-sitosterol: A comparative investigation. Steroids 2015, 100, 27–35. [Google Scholar] [CrossRef]
  20. Medvedeva, N.I.; Kazakova, O.B.; Lopatina, T.V.; Smirnova, I.E.; Giniyatullina, G.V.; Baikova, I.P.; Kataev, V.E. Synthesis and antimycobacterial activity of triterpenic A-ring azepanes. Eur. J. Med. Chem. 2018, 143, 464–472. [Google Scholar] [CrossRef]
  21. Khusnutdinova, E.F.; Petrova, A.V.; Thu, H.N.; Tu, A.L.; Thanh, T.N.; Thi, C.B.; Babkov, D.A.; Kazakova, O.B. Structural modifications of 2,3-indolobetulinic acid: Design and synthesis of highly potent α-glucosidase inhibitors. Bioorg. Chem. 2019, 88, 102957. [Google Scholar] [CrossRef]
  22. Dalla-Vechia, L.; Dassonville-Klimpt, A.; Grellier, P.; Sonnet, P.; Gosmann, G.; Gnoatto, S.C.B. The Beckmann rearrangement applied to ursolic acid with antimalarial activity in medicinal chemistry studies. Lett. Org. Chem. 2012, 9, 92–95. [Google Scholar] [CrossRef]
  23. Giniyatullina, G.V.; Kazakova, O.B.; Salimova, E.V.; Tolstikov, G.A. Synthesis of new betulonic and oleanolic acid amides. Chem. Nat. Compd. 2011, 47, 68–72. [Google Scholar] [CrossRef]
  24. Flekhter, O.B.; Ashavina, O.Y.; Boreko, E.I.; Karachurina, L.T.; Pavlova, N.I.; Kabal’nova, N.N.; Savinova, O.V.; Galin, F.Z.; Nikolaeva, S.N.; Zarudii, F.S.; et al. Synthesis of 3-acetylbetulinic acid and betulonic aldehydes according to Svern and the pharmacological activity of related oximes. Pharm. Chem. J. 2002, 36, 303–306. [Google Scholar] [CrossRef]
Figure 1. Structure of squalamine and trodusquemine.
Figure 1. Structure of squalamine and trodusquemine.
Molbank 2019 m1078 g001
Scheme 1. Synthesis of lupane type derivatives. Reagents and conditions: a. TsOH, CHCl3, reflux, 4 h; b. PCC, CH2Cl2, rt, 0.5 h; c. NH2(CH2)3NH(CH2)4NH2, MeOH, reflux, 8 h; d. LiAlH4, THF, reflux, 3 h.
Scheme 1. Synthesis of lupane type derivatives. Reagents and conditions: a. TsOH, CHCl3, reflux, 4 h; b. PCC, CH2Cl2, rt, 0.5 h; c. NH2(CH2)3NH(CH2)4NH2, MeOH, reflux, 8 h; d. LiAlH4, THF, reflux, 3 h.
Molbank 2019 m1078 sch001
Scheme 2. Synthesis of oleanane and ursane type derivatives. Reagents and conditions: a. LiAlH4, THF, reflux, 3 h; b. i PCC, CH2Cl2, rt, 0.5 h, ii NH2(CH2)3NH(CH2)4NH2, MeOH, reflux, 8 h; c. i (COCl)2, Et3N, CH2Cl2, 20 °C, 2 h, ii diethylenetriamine, CH2Cl2, Et3N, 20 °C, 3 h.
Scheme 2. Synthesis of oleanane and ursane type derivatives. Reagents and conditions: a. LiAlH4, THF, reflux, 3 h; b. i PCC, CH2Cl2, rt, 0.5 h, ii NH2(CH2)3NH(CH2)4NH2, MeOH, reflux, 8 h; c. i (COCl)2, Et3N, CH2Cl2, 20 °C, 2 h, ii diethylenetriamine, CH2Cl2, Et3N, 20 °C, 3 h.
Molbank 2019 m1078 sch002
Table 1. Minimum inhibitory concentrations and minimum bactericidal concentration of compounds 1, 2, 5, and 711.
Table 1. Minimum inhibitory concentrations and minimum bactericidal concentration of compounds 1, 2, 5, and 711.
CompoundMIC (µM)/MBC (µM)
S. aureus
DSM789
E. coli
ATCC25299
P. aeruginosa
PA01
1>200/>200>200/>200>200/>200
26.25/25>200/>200>200/>200
512.5/25>200/>200>200/>200
7>200/>200>200/>200>200/>200
83.125/6.25>200/>200>200/>200
912.5/25200/>200200/>200
1012.5/25200/>200200/>200
1112.5/2525/2550/>200
Ciprofloxacine0.200.200.20
Vancomycine0.80>200>200

Share and Cite

MDPI and ACS Style

Kazakova, O.B.; Brunel, J.M.; Khusnutdinova, E.F.; Negrel, S.; Giniyatullina, G.V.; Lopatina, T.V.; Petrova, A.V. A-Ring-Modified Triterpenoids and Their Spermidine–Aldimines with Strong Antibacterial Activity. Molbank 2019, 2019, M1078. https://0-doi-org.brum.beds.ac.uk/10.3390/M1078

AMA Style

Kazakova OB, Brunel JM, Khusnutdinova EF, Negrel S, Giniyatullina GV, Lopatina TV, Petrova AV. A-Ring-Modified Triterpenoids and Their Spermidine–Aldimines with Strong Antibacterial Activity. Molbank. 2019; 2019(3):M1078. https://0-doi-org.brum.beds.ac.uk/10.3390/M1078

Chicago/Turabian Style

Kazakova, Oxana B., Jean Michel Brunel, Elmira F. Khusnutdinova, Sophie Negrel, Gulnara V. Giniyatullina, Tatyana V. Lopatina, and Anastasiya V. Petrova. 2019. "A-Ring-Modified Triterpenoids and Their Spermidine–Aldimines with Strong Antibacterial Activity" Molbank 2019, no. 3: M1078. https://0-doi-org.brum.beds.ac.uk/10.3390/M1078

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