525
Views
7
CrossRef citations to date
0
Altmetric
Research Article

Glutamine analogues containing a keto function – novel inhibitors of fungal glucosamine-6-phosphate synthase

, , &
Pages 439-447 | Received 30 May 2005, Accepted 20 Jun 2005, Published online: 04 Oct 2008

Abstract

A series of novel inhibitors of glucosamine-6-phosphate synthase, analogues of AADP and BADP, have been synthesized and their inhibitory, lipophilic and antifungal properties have been tested. The improvement in lipophilicity has not much affected the antifungal activity of the new compounds. Dipeptides containing norvaline and selected inhibitors have shown substantial activity against S. cerevisiae and C. glabrata and only poor activity against C. albicans strain. These peptides do not seem to be toxic towards human cells.

Introduction

Glucosamine-6-phosphate synthase (GlcN-6-P synthase – EC 2.6.1.16) is a key enzyme that catalyses the formation of D-glucosamine-6-phosphate (GlcN-6-P) from D-fructose-6-phosphate using L-glutamine as the ammonia source [Citation1]. GlcN-6-P is then transformed into uridine-5′-N-acetylglucosamine (UDP-GlcN) which is an important aminosugar utilized in the biosynthesis of various cellular structures – lipopolisaccharides and peptidoglycan in bacteria, mannan and chitin in fungi, and glycoproteins in mammals. The enzyme may be considered as a selective molecular target since substantial differences in physiological consequences of GlcN-6-P synthase inhibition is observed in these organisms. In fungal cells, the GlcN-6-P synthase inhibition resulted in cell agglutination and lysis whereas in mammalian cells temporary inhibition of the enzyme is not lethal due to the rapid turnover of the mammalian gene encoding GlcN-6-P synthase. Therefore GlcN-6- synthase has been proposed as a potential target for design of antibacterial and especially antifungal agents. In antifungal chemotherapy, on the contrary to antibacterial chemotherapy there is still lack of safe and effective drugs [Citation2]. GlcN-6-P synthase is inactivated by a number of electrophilic reagents, however, only selective and irreversible inhibitors may be of chemotherapeutic value. Of the GlcN-6-P synthase inhibitors, analogues of one of the enzyme substrates – glutamine, e.g. N3-(4-methoxyfumaroyl)-(S)-2,3-diaminopropanoic acid (FMDP), N3-(trans-epoxysuccinamoyl)-(S)-2,3-diaminopropanoic acid (EADP) have been shown to be strong and selective inactivators of fungal GlcN-6-P synthase [Citation3,Citation4]. Although these inhibitors displayed excellent inhibitory properties, they exhibit poor activity against whole cells. High polarity of the inhibitors effectively hampered their transport inside the cells. However, these inhibitors in the latent form – most commonly as peptides [Citation5,Citation6] can be recognized by peptide permeases and transported into the cells. Lipophilic FMDP-peptides [Citation7] and diffusible FMDP analogues [Citation8] also exerted antifungal activity. Development of inhibitor with enhanced lipophilicity that can enter the fungal cells by free diffusion may also be another possible solution to the GlcN-6-P synthase inhibitor transport problem. Previously, we have synthesised novel inhibitors and inactivators of fungal GlcN-6-P synthase containing a keto function, i.e. N3-trans-4-oxo-2-pentenoyl-l-2,3-diaminopropanoic acid (AADP) and N3-trans-4-oxo-4-phenyl-2-butenoyl-L-2,3-diaminopropanoic acid (BADP) and demonstrated their inhibitory activity against fungal GlcN-6-P synthase [Citation9]. Based on this observation, we decided to synthesise a series of novel BADP and AADP analogues with enhanced lipophilicity and to evaluate these compounds as GlcN-6-P synthase inhibitors ().

Figure 1 Structures of new inhibitors.

Figure 1 Structures of new inhibitors.

We have also determined the inhibitory activity of the new inhibitors against glucosamine-6-phosphate synthase, their affinity to biological membranes and their antifungal activity. Moreover, we have synthesised three peptides (7b, s, u) containing novel inhibitors (1 b, s, u) and norvaline in the N-terminal position, and tested their in vitro activity. Previously, we have demonstated that peptides containing norvaline and GlcN-6-P synthase inhibitors displayed excellent antifungal activity [Citation5,Citation6].

Experimental

Chemistry

(E)-4-oxo-4-(2′-pyrrolyl)-2-butenoic acid (2s)

A solution of 2-acetylpyrrole (1.0 g; 9.2 mmol), glyoxylic acid monohydrate (1.69 g, 9.2 mmol) in 2 M NaOH (20 ml) and methanol (6 ml) was heated for 5 h at 50°C. The reaction mixture was then acidified with 2 M HCl and evaporated to a small volume. The concentrated solution was extracted with diethyl ether (5 × 30 ml), dried over MgSO4 and the solvents were evaporated leaving an oily residue which was crystallized from diethyl ether /hexane to yield 2s as a solid (0.45 g, 30%). 1H NMR (500 MHz, CDCl3) δ = 6.65–6.66 (m, 1H), 7.01 (d, 1H), 7.42 (d, 1H), 7.73 (m, 1H), 7.86 (d, 1H). MS(FAB) m/z: 166 (MH+)

(E)-3-(2′-ethyl-1′,3′-dioxolan-2′-yl)-2-propenoic acid (3u)

To a solution of (E)-4-oxo-2-hexenoic acid (0.40 g, 3.2 mmol) in benzene (20 ml) (CARE–Carcinogenic) triethylorthoformate (1.74 ml, 32 mmol), ethylene glycol (2.6 ml, 16 mmol) and p-toluenesulfonic acid (0.08 g, 0.4 mmol) were added. The mixture was heated at 35°C for 24 h, then diluted with ethyl acetate (20 ml) and concentrated NaCl solution (20 ml) and extracted with ethyl acetate (3 × 50 ml). The combined organic phase was dried over MgSO4, the solvents were evaporated and the residue was dissolved in methanol (40 ml) and treated with 2 M KOH solution (10 ml) at room temperature. After 24 h the solution was acidified to pH 4 with conc. HCl end extracted with CH2Cl2. The organic phase was dried, the solvents were evaporated and the residue was purified by column chromatography on silica gel gel (CHCl3–AcOH, 60:1) to give 3u (0.35 g, 75%) as white crystals. M.p. = 63–64°C; 1H NMR (500 MHz, CDCl3) δ = 0.94 (t, 3H), 1.78 (q, 2H), 3.88–3.93 (m, 2H), 3.96–4.00 (m, 2H), 6.10 (d, 1H), 6.84 (d, 1H). MS(FAB) m/z: 173 (MH+)

N-succinimidoyl ester of (E)-4-(4′-methyl)phenyl-4-oxo-2-butenoic acid (4a)

DCC (0.45 g, 2.2 mmol) dissolved in THF (10 ml) was slowly added to a solution of (E)-4-(4′-methyl)phenyl-4-oxo-2-butenoic acid 2a (0.38 g, 2 mmol) and N-hydroxysuccinimide (0.23 g, 2 mmol) in freshly distilled THF (10 ml) at 0°C. After 24 h the precipitate formed was filtered off, the solution was concentrated and a few drops of acetic acid were added and the mixture kept for 24 h. Then the rest of the dicyclohexylurea was filtered off and the residue was crystallised from THF/diethyl ether giving 4a (0.46 g, 80%) as yellow crystals. M.p. = 137–141°C; 1H NMR (200 MHz, CDCl3) δ = 2.48 (t, 3H), 2.93 (s, 4H), 7.04–7.11 (d, 1H), 7.34–7.38 (d, 2H), 7.92–7.96 (d, 2H), 8.12–8.20 (d, 1H). MS(FAB) m/z 288 (MH+)

(2S)-2-[(N-tert-butoxycarbonyl)amino]-3-{[(2E)-4-oxo-4-(4-methylphenyl)-2-butenoyl]amino}propanoic acid (5a)

(2S)-3-amino-2-[(N-tert-butoxycarbonyl)amino]propanoic acid (2.04 g, 10 mmol) and NaHCO3 (0.84 g, 10 mmol) were dissolved in water-methanol solution (1:1, 20 ml). Then the N-succinimidoyl ester of (E)-4-(4′-methyl)phenyl-4-oxo-2-butenoic acid (2.73 g, 10 mmol) was added with stirring at 0°C and the reaction mixture was left overnight. The solvents were removed in vacuo. The residue was dissolved in 10 ml of water, acidified with 1 M KHSO4 to pH 2 and extracted with ethyl acetate (3 × 30 ml). The organic phase was dried and the solvent was evaporated leaving an oily residue which was crystallised from ethyl ether/hexane to give 5a (3.0 g, 63%) as a white solid. 1H NMR (200 MHz, CDCl3) δ = 1.46 (s, 9H) 2.44 (s, 3H), 3.77–3.94 (m, 2H), 4,40–4.44 (m, 1H), 6.0–6.1 (m, 1H), 6.98–7.06 (d, 1H), 7.30 (d, 2H), 7.90–7.99 (m, 3H). MS(FAB) m/z: 377 (MH+)

(2S)-2-amino-3-{[(2E)-4-oxo-4-(4′-methyl)phenyl-2-butenoyl]amino}propanoic acid trifluoroacetate (1a)

Compound 5a was dissolved in cold trifluoroacetic acid (10 ml) and kept for 1 h. TFA was removed in vacuo, the residue was triturated with diethyl ether and the precipitate was filtered off and dried over KOH pellets to give 1a (2.13 g, 55%) as an amorphous powder (c = 1, DMSO), 1H NMR (200 MHz, DMSO-d6) δ = 2.48 (s, 3H), 3.4–3.8 (m, 3H), 6.96 (d, 1H), 7.39 (d, 2H), 7.76 (d, 1H), 7.93 (d, 1H), 8.8–8.9 (m, 1H). MS(FAB) m/z: 252 (MH+).

(2S)-2-[(N-tert-butoxycarbonyl)norvalyl]amino-3-{[(E)-4-oxo-4-(4′-ethyl)phenyl-2-butenoyl]amino}-propanoic acid (6b)

Compound 6b was synthesized from (2S)-3-amino-2-{[(N-tert-butoxycarbonyl)norvalyl]amino}propanoic acid 5a (0.303 g, 1 mmol) and the N-succinimidoyl ester of (E)-4-oxo-4-(4′-ethyl)phenyl-2-butenoic acid (4b) (0.301 g, 1 mmol). It was purified on silica gel (CHCl3–MeOH–AcOH, 15:1:0.05) to afford 6b (0.25 g, 51%) as an oily residue. 1H NMR (500 MHz, CDCl3) δ = 0.90–0.95 (m, 3H), 1.25 (t, 3H), 1.40–1.44 (m, 11H), 1.52–1.75 (m, 2H), 2.70 (q, 2H), 3.14–3.29 (m, 2H), 3.99–4.02 (m, 1H), 4.32–4.35 (m, 1H), 7.05 (d, 1H), 7.30 (m, 2H), 7.91–7.94 (m, 3H). MS(FAB) m/z: 490 (MH+)

(2S)-2-(norvalyl)amino-3-{[(E)-4-oxo-4-(4′-ethyl)phenyl-2-butenoyl]amino}propanoic acid trifluoroacetate (7b)

Compound 6b was treated with trifluoroacetic acid (10 ml) for 0.5 h at 0°C. TFA was evaporated in vacuo and the residue was triturated with ethyl ether, the precipitate was filtered off, dried over KOH pellets giving 7b (0.23, 91%) as an amorphous powder, (c = 1, H2O), 1H NMR (200 MHz, D2O) δ = 0.83–0.93 (m, 3H), 1.14 (t, 3H), 1.21–1.41 (m, 2H), 1.76–1.84 (m, 2H), 2.59–2.73 (m, 2H), 3.7–3.9 (m, 2H), 3.95–4.05 (m, 1H), 4.6–4.7 (m, 1H), 6.88 (d, 1H), 7.37–7.39 (m, 2H), 7.77 (d, 1H), 7.87–7.90 (m, 2H). MS(FAB) m/z: 351(MH+).

Biological preparation of the crude enzyme (cell free extract) and determination of glucosamine-6 phosphate synthase activity

Candida albicans glucosamine-6-phosphate synthase cell free extract was prepared by the previously described procedure [Citation10]. The standard incubation mixture contained: d-fructose-6-phsphate (15 mM), l-glutamine (10 mM), EDTA (1 mM), potassium phosphate buffer (25 mM, pH 7.0), inhibitor at an appropriate concentration and enzymatic protein (0.005–0.01 mg m l− 1) in a total volume of 0.4 ml. The mixtures were incubated at 37°C for 30 min. The reaction was stopped by heating at 100°C for 1 min. The activity of glucosamine-6-phosphate synthase was determined according to the modified Elson-Morgan method [Citation11].

Inactivation of glucosamine-6-phosphate synthase

Incubation mixtures containing: glucosamine-6-phosphate synthase from C. albicans (0.005–0.01 mg), albumin (1 mg/ml), phosphate buffer (25 mM, pH 7.0) EDTA (1 mM) and inactivators at various concentration in a total volume of 1 ml, were incubated at 25°C. After 5, 10, 15 and 30 min, aliquots (200 μl) were withdrawn from the reaction mixture and applied to the top of a small 1-ml column packed with Sephadex G-25 (equilibrated with 25 mM potassium phosphate buffer pH 7.0) and centrifuged (500 × g for 1 min at 4°C). Under these condition the unbound inhibitor was separated from the enzyme and the protein was recovered in clean test-tubes. Appropriate aliquots were used for the determination of the residual activity of glucosamine-6-phosphate synthase using the standard method. The GlcN-6-P synthase activity for various inhibitory concentrations was plotted versus time and the kinetic parameters were determined following the literature procedure [Citation12].

Determination of the affinity to the artificial biological membrane

Interactions between the test compounds and immobilised artificial biological membrane were investigated using HPLC column IAM PC DD2 (Regis Technologies, Inc., Morton Grove, IL, USA) [Citation13]. The column dimensions were 3 cm × 4.6 mm, particle diameter 300 Å. The chromatographic system consisted of a Model L-6200 A, Model L-4250 UV/VIS detector and Model D-2500 integrator (all from Merck-Hitachi, Vienna, Austria). Potassium phosphate buffer (0.1 M) was used as the mobile phase; the injection volume was 10 μl; the flow rate was 1 ml/min; the samples were detected at 220 nm. The dead volume of the column was determined by the retention time of citric acid (aqueous solution, 50 mg/ml) and was used to calculate capacity factors

Antifungal susceptibility tests

Minimal inhibitory concentrations (MIC's) of the examined compounds were determined by the serial twofold dilution microtiter plate method, in the minimal liquid Yeast Nitrogen Base (YNB) medium without amino acids and ammonium sulphate containing 2% glucose and l-proline (4 mg ml− 1). Wells containing serially diluted test compounds and control were inoculated with 104 cells ml− 1 of an overnight culture of fungal cells and the microtiter plates were incubated for 24 h at 30°C. Fungal growth was measured using the microplate reader (Labsystems, Multiscan Bichromatic) at λ = 595 nm. The MIC was defined as the inhibitor concentration preventing at least 80% of fungal growth, as compared to the inhibitor-free control.

2.2.5 Disc diffusion competition assay

The minimal solid YNB medium without amino acids and ammonium sulphate containing 2% glucose, 4 mg ml− 1 of l-proline and 2% agar was liquefied by warming to 100°C and then chilled to 50°C. The semi-liquid medium was inoculated with 3 × 105 cells ml− 1 of an overnight culture of fungal cells and poured into Petri dishes and left for solidification. Sterile filter discs, 6 mm diameter, were saturated with 10 μl portion of sterile aqueous solutions containing either 200 μg of GlcNAc or 100 μg of one of the test compounds. The discs were placed onto the agar medium surface. In each plate, the GlcNAc saturated disc was flanked by four discs containing test compounds placed at a distance of 20 mm. Plates were incubated at 30°C for 48 h. Growth inhibition zones appearing around the discs containing the test compounds were evaluated and observed distortions were assessed quantitatively.

Cell lines

Human promyelocytic leukemia sensitive cell line HL-60 (Kansas State University, Manhattan, KS, USA) was grown in RPMI 1640 medium supplemented with 10% FBS penicillin G (100 000 units/L), streptomycin (100 mg/L). The cell line was grown in a controlled (air-5% CO2) humidified atmosphere at 37°C and was transplanted three times a week. For the experiments the cells in logarithmic growth were suspended in the growth medium to give a final required density.

In vitro cytotoxic evaluation

Cells of the required density were seeded and different concentrations of the test compounds were added. The experiments were carried out in a controlled (air-5% CO2) humidified atmosphere at 37°C. The exposure time was 72 h. The cytotoxic activity (IC50 values) of the compounds was defined as their in vitro concentrations causing 50% inhibition of cell growth after continuous exposure to the test compound, as measured by cell counting with a Z2 Cell Analyzer (Beckman Coulter). Results are given as the mean of at least three independent experiments ± standard error of the mean (SEM).

Results and discussion

Synthesis

The novel GlcN-6-P synthase inhibitors (indicated by Arabic numbers) were synthesised starting from suitably substituted at the C4 position (E)-4-oxo-2-butenoic acids, which were obtained using three different routes (Scheme ).

Scheme 1 Syntheses of (E)-4-oxo-2-butenoic acids (2). i) AlCl3; ii) H+ for 2 f–q and 2 t; 1. H+2. OH for 2 r; OH for 2 s; iii) 1. BuLi, 2. H2O, 3. MnO2, 4. HBraq/dioxane.

Scheme 1 Syntheses of (E)-4-oxo-2-butenoic acids (2). i) AlCl3; ii) H+ for 2 f–q and 2 t; 1. H+2. OH− for 2 r; OH− for 2 s; iii) 1. BuLi, 2. H2O, 3. MnO2, 4. HBraq/dioxane.

Derivatives with the alkyl group in the para position of the phenyl ring, 2 a–e, were obtained applying the Friedel-Crafts alkylation reaction of an appropriate alkylbenzene with maleic acid anhydride according to the published procedures Citation14-16 and described as Method A (see ).

Table I.  Properties of (E)-4-oxo-2-butenoic acids.

All the other (E)-4-oxo-4-phenyl-2-butenoic acids 2 fq and compounds where the phenyl ring was replaced by furyl 2 r, pyrrolyl 2 s or tert-butyl 2 t group were prepared by condensation of an appropriate methyl ketone with glyoxylic acid (Method B, ) [Citation17,Citation18]. The three acids 2 ux were prepared via condensation of the appropriate aldehyde with propionlaldehyde diethyl acetal followed by oxidation and hydrolysis (Method C, )[Citation19]. The keto function in these acids was first converted into a diacetal by reacting with ethylene glycol [Citation20] to obtain the protected compounds 3 ux for further syntheses (Scheme and ).

Scheme 2 Syntheses of keto protected derivatives of alkenoic acids.

Scheme 2 Syntheses of keto protected derivatives of alkenoic acids.

Table II.  Properties of keto protected derivatives of alkenoic acids.

The crystalline acids were then activated with N-hydroxysuccinimide (HOSu) and dicyclohexylcarbodiimide (DCC) [Citation25] according to the procedure described for the preparation of 4a to give the active esters in good yields (Scheme and ) and condensed with N2-tert-butoxycarbonyl-(S)-2,3-diaminopropanoic acid to afford the protected inhibitors ().

Scheme 3 Syntheses of new compounds. i) HOSu, DCC, ii) 1. NH2CH2CH(NHBoc)COONa, 2. H+, iii) TFA, iv) 1. NH2CH2CH(BocNvaNH)COONa, 2. H+.

Scheme 3 Syntheses of new compounds. i) HOSu, DCC, ii) 1. NH2CH2CH(NHBoc)COONa, 2. H+, iii) TFA, iv) 1. NH2CH2CH(BocNvaNH)COONa, 2. H+.

Table III.  Properties of N-succinimidoyl active esters.

Table IV.  Properties of protected inhibitors.

Deprotection of the amino group and the keto function was performed simultaneously in anhydrous trifluoroacetic acid, and the final compounds were precipitated with diethyl ether ().

Table V.  Properties of final inhibitors.

The furyl derivative 1r was additionally purified on Sephadex LH-20 in methanol. Three inhibitors 1 b, 1s and 1u were then combined with the N-hydoxysuccinimide ester of N-(tert-butoxycarbonyl)-l-norvaline to obtain the protected dipeptides 6b, 6s and 6u (Scheme and ).

Table VI.  Properties of new peptides.

Final deprotection of the Boc groups afforded dipeptides 7 b, 7s and 7u in good yields (). All new compounds were fully characterised by 1H NMR and mass spectrum analyses.

GlcN-6-P synthase inhibition studies

All the newly synthesised inhibitors as well as the parent compounds AADP and BADP were tested against purified glucosamine-6-phosphate synthase from Candida albicans [Citation26]. Their ability to inhibit this enzyme was measured by determining a concentration which caused 50% inhibition of the enzyme[Citation27]. Kinetic parameters of the inactivation by compounds 1ax, AADP and BADP are summarized in . For all the tested compounds, a time-dependent loss of enzyme activity (plots not shown) was observed. Therefore, these novel compounds are irreversible agents.

Table VII.  Inhibitory and inactivatory data for new compounds in respect to C. albicans GlcN-6-P synthase and affinity to biological membranes.

It is apparent from inspection of the Table that as the size of the substituents on the aromatic ring increases, the Kinact values, in general for most of the compounds, also increase, suggesting there is a steric effect involved in binding to the active site, since Kinact value reflects affinity of an inhibitor to the enzyme active site. Inhibitors with longer aliphatic chains were found to be poorly soluble in water making determination of Kinact for compound 1e impossible. The only analogue with a 4-ethyl substituent, 1b, showed inhibitory potency against glucosamine-6-phosphate synthase comparable to that of the parent compound BADP. Compounds with ortho substituted phenyl ring seems to be less reactive (higher T values) than para analogues. On the other hand, analogues with an aliphatic chain, 1t and 1w, displayed lowest inhibitory activity in the series although their affinity to the active site was markedly improved in comparison to other inhibitors with aliphatic chains. The inactivation potencies (k2/Kinact) of the examined inhibitors were also lowered in comparison to BADP. Probably, the compounds with a substituted phenyl ring may not be properly fitted into the enzyme binding pocket, and may disturb the enzyme conformation before its inactivation. These discrepancies observed between reactivity and affinity should be explained in further studies.

3.3 Membrane affinity

For all these compounds their ability to diffuse through biological membrane was measured. This property was investigated using a HPLC chromatographic column IAM (immobilized artificial membrane) PC DD 2 with stationary phase mimicking the cell membrane. The retention times were measured and affinity to the biological membrane was expressed as (). These values describe not only lipophilic properties but also hydrogen bond formation or electrostatic interactions. The highest values were determined for compounds with linear aliphatic chain substituents, however, these compounds show only moderate affinity to the biological membrane.

Antifungalal activity

Minimal inhibitory concentration (MIC's) were determined in YNB medium with glutamine and proline as carbon source. presents activity against a few species. Most of the synthesised inhibitors exhibited rather poor antifungal activity with MIC's in the range from 62.5 μg/ml to 1000 μg/ml and their activity is not correlated with apparent lipophilicity. Compound 1c with a n-butyl substituent on the aromatic ring and with 1.078 displayed almost the same antifungal activity as the lead compound (BADP) with a lower Much lower would hamper diffusion of the inhibitor into the microbial cells and higher would cause the retainment of the tested compound in the cytoplasmic membrane. On the other hand inhibitors with aliphatic chains e.g. 1x with 0.324 containing a n-butyl group and AADP itself with as low as − 1.415 showed also similar antifungal properties. Unexpectedly high activity of AADP against S. cerevisiae suggests rather different than the facilitated diffusion transport mechanism in this case. All compounds may be considered as only moderately lipophilic and therefore may not be able to cross the cell membrane by free diffusion. Furthermore, these inhibitors are polar compounds still bearing a net charge, and probably this factor also influences their diffusion across the membrane. Inhibitor 1 e containing a large substituent on the aromatic ring, is only sparingly soluble in buffers used in the experiments, so making determination impossible.

Table VIII.  Antifungal activity of new inhibitors.

The peptides 7b, 7s and 7u with new inhibitors were tested against a few fungal stains (). Such compounds are transported into the cell not by diffusion but through peptide permeases [Citation28]. For comparative purposes LysNvaFMDP, known as one of the most active peptide-containing strong GlcN-6-P synthase inhibitor, was also used. Surprisingly, the new peptides show high activity against S. cerevisiae and C. glabrata, but no activity against C. albicans was detected. Such results suggest that the new peptides are not transported into C. albicans cells via peptide permeases.

Table IX.  Antifungal activity of new peptides.

The activity of the new inhibitors and their peptides was also tested against GlcN-6-P synthase in cell free extract (). For the compounds with the aromatic phenyl rings i.e., 1b and 7b no significant differences in activities were observed. On the other hand, the inhibitory activity of the peptides 7s and 7u were much higher then those of inhibitors 1s and 1u respectively.

Table X.  Peptides and inhibitors activity in cell-free extracts.

Moreover, dipeptide NvaFMDP was also tested, but it showed relatively poor activity in cell-free extract, probably due to the instability of its methyl ester group, that resulted in the formation of inactive inhibitor. FMDP itself, however, under the experimental condition displayed strongest inhibitory potencies. Such results suggest that in the cell free extract either peptides (but not inhibitors) were activated or, after the hydrolysis of the peptide bond, the inhibitor was in a more preferable conformation for inactivation of GlcN-6-P synthase. For the peptides 7 s and 7 u experiments on Petri plates were carried out to examine reversal of inhibition in presence of N-acetylglucosamine. If the enzyme Glc-6-P synthase was the only molecular target for the new peptides, addition of the enzymatic reaction product would result in complete prevention of anticandidal activity of the tested peptide, as took place for LysNvaFMDP. However, for the new peptides 7s and 7u no such effect was observed, which suggests that glucosamine-6-phosphate synthase is not the only molecular target for the new peptides.

Cytotoxic evaluation

Peptides 7 s and 7 u were tested against human tumour cells () and no toxic effects were observed even at high concentrations. Therefore, these compounds may be regarded as non-toxic to normal human cells.

Table XI.  Activity of tested peptides against HL-60 human promyelocytic leukaemia cells.

Conclusions

We have synthesised twenty-three novel GlcN-6-P synthase inhibitors with enhanced lipophilicity in comparison to BADP and AADP. These compounds displayed however, weak or medium inhibitory properties. Peptides containing the novel inhibitors (with pyrrolyl 7 s and ethyl 7 u substituents adjacent to the keto group) were highly active against C. glabrata and S. cerevisiae strains. High anticandidal activity and lack of disturbance of the growth in the inhibition zone in the presence of GlcNAc suggested that GlcN-6-P synthase is not the only molecular target for the new peptides, as it is for FMDP peptides. Further studies to elucidate the mechanism of the antifungal action of the new compounds are in progress.

Acknowledgements

This work was supported by the Ministry of Science and Information Society Technologies, Grant No. 2PO5F 014 26 and in part Grant No. 3PO5F02324.

References

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.