Next Article in Journal
Ensemble Model Based on Stacked Long Short-Term Memory Model for Cycle Life Prediction of Lithium–Ion Batteries
Next Article in Special Issue
Synthesis of Novel Potent Biologically Active N-Benzylisatin-Aryl Hydrazones in Comparison with Lung Cancer Drug ‘Gefitinib’
Previous Article in Journal
Axial Vibration Characteristics of Fluid-Structure Interaction of an Aircraft Hydraulic Pipe Based on Modified Friction Coupling Model
Previous Article in Special Issue
In Vitro and In Silico Screening of 2,4,5-Trisubstituted Imidazole Derivatives as Potential Xanthine Oxidase and Acetylcholinesterase Inhibitors, Antioxidant, and Antiproliferative Agents
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Enamine Barbiturates and Thiobarbiturates as a New Class of Bacterial Urease Inhibitors

1
Department of Chemistry, College of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia
2
Department of Chemistry, Faculty of Science, Alexandria University, P.O. Box 426, Ibrahimia, Alexandria 21321, Egypt
3
Hussain Ebrahim Jamal Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
4
Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
5
KRISP—Kwazulu-Natal Research Innovation and Sequencing Platform, College of Health Sciences, University of KwaZulu-Natal, Westville, Durban 4001, South Africa
6
School of Chemistry and Physics, University of KwaZulu-Natal, University Road, Westville, Durban 4001, South Africa
7
Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Networking Centre on Bioengineering, Biomaterials and Nanomedicine, PCB, Baldiri Reixac 10, 08028 Barcelona, Spain
8
Department of Organic Chemistry, University of Barcelona, Martí i Franqués 1-11, 08028 Barcelona, Spain
*
Authors to whom correspondence should be addressed.
Appl. Sci. 2020, 10(10), 3523; https://doi.org/10.3390/app10103523
Submission received: 2 April 2020 / Revised: 15 May 2020 / Accepted: 15 May 2020 / Published: 20 May 2020

Abstract

:
Urease is a therapeutic target associated with several important diseases and health problems. Based on our previous work on the inhibition of glucosidase and other enzymes and exploiting the privileged structure assigned to the (thio)barbiturate (pyrimidine) scaffold, here we tested the capacity of two (thio)barbiturate-based compound collections to inhibit urease. Several compounds showed more activity than acetohydroxamic acid as a standard tested compound. In addition, by means of a conformational study and using the Density Functional Theory (DFT) method, we identified energetically low-lying conformers. Finally, we undertook a docking study to explore the binding mechanism of these new pyrimidine derivatives as urease inhibitors.

Graphical Abstract

1. Introduction

Many microorganisms use nitrogen from urease for growth (urea amidohydrolase, EC: 3.5.1.5). Urease is an enzyme that catalyzes the hydrolysis of urea into ammonia and carbon dioxide. There is a link between health complications and ammonia production by urease [1]. In animals and humans, low pH of the stomach allows microbial strains to survive, multiply, and grow, resulting in pyelonephritis, hepatic coma, gastric carcinoma, gastric lymphoma, kidney stones, and peptic ulcer complications [2,3]. The treatment of bacterial infection with therapeutics has often proven ineffective due to drug resistance. Thus, there is a clear need for alternatives or novel treatments. Given the involvement of ureases in various diseases, pharmaceutical research has channeled considerable efforts into discovering potent and safe urease inhibitors [4,5,6,7].
Molecules with a binding site that can chelate metals are an interesting challenge and could be a promising line of action to prevent the adverse effects of ureolytic bacterial infections in humans [8]. In this regard, many types of potent urease inhibitors have been designed [9], such as dihydropyrimidines [10], urea derivatives [11], semicarbazones [12], Schiff bases [13], hydroxamic acid derivatives [14], piperazines [15], biscoumarines [16], benzimidazoles [17], and sulfonamides [18].
Enaminone molecular scaffolds have been found in many synthetic drugs and natural products [19,20]. Specially, pyrimidine-based enamines have demonstrated an array of biological activities owing to the presence of the alkenylamine moiety (R2NCH = CH−) in their structures, and are capable of strong bonding with metal ion chelates in biological systems [21,22].
The barbituric and thiobarbituric acids [(thio)pyrimdine trione analog derivatives] have been reported urease inhibitors [12]. These can be considered privileged structures because they have antifungal [23], antimicrobial [24,25], anti-adiponectin [26], anti-sclerosis [27], anti-convulsing [28,29], antiglycation [30], and α-glucosidase inhibitory [30] properties. Several compounds A, B, and C have been reported, and showed anti-urease activity comparable to acetohydroxamic acid (Figure 1) [31,32].
In this context, and as a continuation of our extended medicinal chemistry program based on the barbituric and thiobarbituric acid moieties [30,33,34,35,36,37,38,39,40,41,42,43,44], here we examined the in vitro anti-urease activity of a collection of 1,3-dimethylbarbiturate-enamine compounds and their analogs, thiobarbiturate derivatives. In addition, molecular docking studies were performed to evaluate the molecular interactions of the newly synthesized compounds with selected drug targets (PDB ID 4GY7).

2. Materials and Methods

The synthesis and the full characterization of compounds 3a, 3d, 3h, 3j–p, 4, and 5 (Table 1) have been previously described by our group [30,33]. The rest of the compounds studied were prepared following the previously described method. The yields and full characterization are provided in the Supplementary materials.

2.1. In Vitro Urease Inhibition Assay Protocol

The urease inhibition assay was performed spectrophotometrically following the manufacturer’s instructions [45,46,47]. The source of urease is Jack bean Urease and the full protocol provided in the supplementary materials. In the present study, urease was preincubated with the inhibitors for a period of 15 min, which proved to be sufficient in our studies.

2.2. DFT Calculation

The molecular building and geometric optimization of all 18 compounds was performed by DFT using Gaussian 03W software package with basis set method B3LYP/6-31 + G(d,p). The Gauss-view program was used for molecular visualization. Geometrical, energetic and electronic parameters were calculated using structures optimized at 298 K and 1 atm. The Gibbs free energy equation (ΔG = −RT ln K) was used to search conformational space.

2.3. Molecular Docking

Molecular docking studies were conducted to rationalize the binding mode of the pyrimidine derivatives using the MOE package [Molecular Operating Environment, 2016.0810; Chemical Computing Group ULC, 1010 Sherbrooke St. West, Suite #910, Montreal, QC, Canada, H3A 2R7, 2017]. The three-dimensional structure of the urease protein was retrieved from the Protein Data Bank (PDB ID 4GY7) [48]. Water molecules were removed, and the protonation step was performed. Energy minimization using default parameters was performed to achieve a stable conformation. The optimized geometries of the pyrimidine derivatives were docked to the active site of the urease protein. For docking studies, an induced fit protocol with the triangular matcher placement method and London dG scoring function were used. The interactive docking procedure was performed for all the optimized conformers of each compound in the active site. A score was assigned to each docked compound based on its fitting in the binding cavity and its binding mode (docking study protocol are provided in the Supplementary materials).

3. Results and Discussion

3.1. Synthesis

Derivatives 3a–p, 4, and 5 (Table 1) were straightforwardly synthesized by nucleophilic substitution of the corresponding amines with enaminone scaffolds 1 [49] and 2 [50] in methanolic solution, following the reported method [30,33] (Scheme 1). Due to the potential tautomerism of these families of compounds, X-ray crystallography was carried out on two compounds (3b and 3k) to confirm the structures. (1H-NMR and 13C-NMR spectra, Figures S1–S21, and X-ray data of compounds 3b and 3k are provided in the Supplementary materials).

3.2. In Vitro Evaluation of Urease Inhibition

The capacity of synthesized derivatives of N,N-dimethylpyrimidine trione (3ai and 4) and N,N-diethyl-thio pyrimidine dione (3jp and 5) to inhibit urease in vitro was examined and compared with that of acetohydroxamic acid (AHA) (IC50 = 20± 0.4 µM), as a standard compound (Table 1).
Of note, the tested compounds showed potent urease inhibition activity, twelve of them (3a, 3d, 3e, 3f, 3h, 3j, 3k, 3l, 3m, 3n, 3o, and 4) exerting greater activity than the reference acetohydroxamic acid. The most active member between this series is compound 3h which the structure attributed the pyridine moiety. Separation the pyridine ring from the enaminone functionality in compound 3j with CH2 group, the activity has been decreased compared to the most active member 3h. On the other hands, compounds 3k, and 3o showed superior activity to their counterparts 3b and 3g. Indeed, 3l and 3o showed activity similar to that of 3d, and 3f. The chloro atom in para-position in compound 3m, the activity has been decreased compared to the ortho position of compounds 3f, and 3n. Compounds 3c, 3i and 3p with sulfonic, cyclic amide or sulfonamide functionalities shown less activity compared to standard drug. The cyclohexyl group in compound 3e increases the activity with 1.8 folds than the control. it was observed that in case of compounds 4 and 5 the presence of the N,N’-diethyl group and the S atom in the thiobarbituric derivative 5 make them more voluminous compared to its analogs N,N’-dimethyl and the “O” atom in the barbituric derivatives 4. Finally, compounds 3b, and 3g were not active.
However, there are some discrepancies regarding whether AHA is or is not a competitive inhibitor to the active site of the urease enzyme [51,52]. Therefore, some limitations could be applied, and we think that comparisons are important.

3.3. Docking Study

Molecular docking studies have the potential to identify novel drug-like molecules that display high binding affinity for the target protein, and they can facilitate the understanding of biological activity data with the purpose of designing new compounds with improved activity. Hence, we extended our study to explore the conformational space and binding orientation of the synthesized pyrimidine derivatives.
The binding conformations of these compounds were explored by MOE-Dock module implemented in the MOE program [53]. The optimized conformers obtained from the Gaussian program were docked to the active site of urease. The docking results indicated that all the conformers were well accommodated inside the active site and were stabilized by various hydrophilic, hydrophobic and van der Waals interactions. The residues involved in these interactions were Arg439, Ala440, His492, Asp494, His593, His594, Asp633 Ala636 and Met637. Compound 3h (IC50 = 6 ± 0.3 µM) showed strong inhibitory potential against urease as compared to 3b (inactive) due to methyl substitution at the para position of the pyridine ring. Compound 3h showed good interactions with the residues of the active site by coordinating with the bi-nickel center, via its carbonyl group at the pyrimidine ring and anchoring itself in a way that permitted strong interaction of the carbonyl group with the two nickel ions. Additionally, the two carbonyl groups on the pyrimidine ring acted as hydrogen bond acceptors and mediated hydrogen bond interaction with His492 (2.3 Å) and Arg439 (2.04 Å) of the binding pocket (Figure 2b).
Compound 3a (IC50 = 9 ± 2 µM), another active compound of the series, established various potential interactions with the active site of urease, as well as with nickel ions. The oxygen atom of the morpholine ring was involved in the chelation process with the nickel center in the catalytic site. This compound was further stabilized by two typical hydrogen bonds with His492 (2.8 Å) and Arg609 (3.0 Å). Apart from hydrogen bond interactions, a tetramine of the compound participated in the salt bridge interaction with the negatively charged Asp494 residue (Figure 2a). Compounds 3d and 3l (IC50 = 8 ± 0.3 µM) had almost similar structures, biological activities and patterns of binding interactions with the active site residues. The only difference was the interaction with metal ions. The lone pair of electrons for sulfur (C = S) of 3l mediated strong interaction with the two nickel ions and hydrogen bond interaction with Arg609 (2.4 Å) and Met637 (2.8 Å), while in case of 3d, the carbonyl group on the pyrimidine ring was involved in the interaction with the nickel ions as depicted in Figure 2c.
Compounds 3j (IC50 =10 ± 0.9 µM) and 3k (IC50 =11 ± 1 µM), which showed good biological activity, presented similar types of interactions. The additional effectiveness of 3j compared to 3k was due to the absence of a methyl group at the meta position of the benzene ring, which allowed the compound to establish close contacts with the active site residues (Figure 2d). The ring nitrogen of the pyridine is involved in two productive hydrogen bond interaction with His492 and Met637 at 2.54 and 3.4 Å, respectively. Moreover, the C = O of pyrimidine rind mediated a potential hydrogen bond interaction with the NH of His593 at 2.59 Å, while the compound was further stabilized through hydrophobic interactions with Ala440, Cme592, and His594.
The compounds with electron-withdrawing substitution, especially halogens, showed noteworthy inhibitory potential. For 3m, 3n, and 3o, the presence of halogen atoms with their respective position on the benzene ring affected the binding pattern and orientation of the compound within the pocket (Figure 2e). Compounds 3f (o-chloro phenyl) and 3o (o-iodo phenyl) with IC50 = 10 ± 0.6 µM showed two hydrogen bond interactions with His492 and Ala440 (Figure 2f), while 3m and 3n were stabilized by a single hydrogen bond with His492. These compounds also displayed hydrophobic interaction with Ala440, His593 and Met637. Compounds 3c (IC50 = 26 ± 1µM) and 3p (IC50 = 22 ± 0.8 µM), both with moderate biological activity, established two hydrogen bond interactions with His492 and Ala440. However, only one interaction with the metallocenter was observed (Figure 2g). The compounds with low biological activity, as compared to the standard and other pyrimidine derivatives such as 3i (IC50= 66 ± 2.4 µM) and 5 (IC50= 42 ± 2.3 µM), established single hydrogen bond interaction with the active site residue Arg439 while compound mostly established by hydrophobic interaction with the active site residues (Figure 2h). However, no interaction with nickel was observed for 3i.
Indeed, the urease inhibition capacity of the synthesized pyrimidine derivatives is attributed to the mutual contribution of the distinct substitutions they bear. However, interaction with the metallocenter and hydrophilic interaction with Ala440, His492, His593, and Met637 are found to be crucial for the activity of these compounds.

3.4. Density Functional Theory (DFT)

The physicochemical properties and frontier molecular orbitals (FMOs) of the new enaminone compounds play a crucial role in enhancing bioactivity. Khon-Sham’s DFT approach with the B3LYP method was used for geometry optimization [54]. The electron-donating and -withdrawing ability of a compound can be explained by its HOMO and LUMO. The higher the energy value of HOMO, the greater the electron-contributing ability of the compound. The energy difference between HOMO and LUMO is an established parameter to measure the electron conductivity or degree of intermolecular charge transfer, which also affects bioactivity [55]. In this study, to examine the urease inhibition capacity of the pyrimidine derivatives, we randomly selected compounds for comparison with DFT results.
The results of HOMO-LUMO energies were plotted against the biological activity of the pyrimidine derivatives (Table 2). Good correlation was observed between biological activity and the energies of the LUMO orbitals. Compounds 3h, 3k, 3f, and 3o showed significant LUMO energy of −1.89, −2.24, −1.96 and −2.26 eV, respectively.
The visualization of the HOMO-LUMO orbitals of 3h reflects the localization of FMO (frontier molecular orbital) (Figure 3). The negative and positive phases of orbitals are shown in green and red, respectively. For 3h, HOMO is localized on the pyrimidine ring, distal pyridine moiety and carbonyl group, whereas LUMO is on the pi-bond adjacent to the pyrimidine ring. In contrast, for 3o, electrons are delocalized mainly on the benzene ring with iodine group as a substituent. The influence of LUMO energy on the inhibitory activity of the compounds might be due to the presence of halogen substitution and the pyridine ring. The halogen substitution at ortho and para positions made a great contribution to the urease inhibition capacity of the derivatives. Notably, neither HOMO nor LUMO were located on the methyl group, which would explain why this group contributed the least to binding with the protein.
The energy gap for the most active compound 3h of the series was −4.35. A similar energy difference was observed for 3k, 3f, and 3o. The smaller the difference in HOMO-LUMO energies, the greater the chemical reactivity. For any potential interaction, electron transfer from high-lying HOMO to low-lying LUMO is always energetically favorable. Given this consideration, 3h, 3k, 3f, and 3o possess good activity, which correlated well with urease inhibitory activity.
Molecular electrostatic potentials (MEP) were run for compounds 3h, 3k, and 3o at B3LYP/6-31G (d, p), providing information about the sites reactive towards nucleophilic and electrophilic attack, together with hydrogen-bond interactions. The potential of electrostatic interaction at the surface is shown by blue, red and green, representing the sites for positive, negative and no electrostatic potential, respectively (Figure 4). Moreover, the positive and negative regions of the maps were responsible for the nucleophilic and electrophilic reactivity of the compounds.

4. Conclusions

Based on our previous work addressing the inhibition of glucosidase and taking advantage of the privileged structure associated with the (thio)barbiturate (pyrimidiene) scaffold, here we have identified compounds with high in vitro anti-urease activity. All conformations of the pyrimidine derivatives were optimized with the B3LYP/6–31G method. The DFT results for the compounds correlated well with the experimental data, which indicated that the presence of the pyridine ring and electron-withdrawing groups, especially halogens (compounds 3m, 3n, and 3o), play an important role in conferring urease inhibition activity. Moreover, the molecular docking studies further helped to explain the experimental results. Because of our findings, compounds 3a, 3d, 3h, 3k, and 3o emerge as potential leads for the development of urease inhibitors. In addition, the obtained results confirm that the use of privileged structures for medicinal chemistry programs is an excellent strategy for identifying hits and leads. Further investigation will be carried out in the future for urease enzyme competitive inhibition.

Supplementary Materials

The following are available online at https://www.mdpi.com/2076-3417/10/10/3523/s1. The details of the synthesis and characterization of the studied compounds; X-ray data of compounds 3b and 3k; biological activity assay; docking study protocol and copies of NMR spectra (Figures S1–S21) of the synthesized target compounds are provided as Supplementary materials.

Author Contributions

Conceptualization, A.B. and A.E.-F.; Data curation, S.A. and Z.U.-H.; Funding acquisition, A.M.A.-M.; Investigation, M.A. and A.B.; Methodology, M.A.; Software, S.Y., S.A. and Z.U.-H.; Supervision, A.E.-F., M.I.C. and F.A.; Visualization, A.M.A.-M., M.I.C. and B.G.d.l.T.; Writing—original draft, A.B.; Writing—review & editing, A.B., M.I.C., B.G.d.l.T. and F.A. All authors have read and agreed to the published version of the manuscript.

Funding

Deanship of Scientific Research at King Saud University, Saudi Arabia, research group No. (RGP-044).

Acknowledgments

The authors would like to extend their sincere appreciation to the Deanship of Scientific Research at King Saud University for supporting and funding the research group No. (RGP-044).

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Mobley, H.L.; Hausinger, R.P. Microbial ureases: Significance, regulation, and molecular characterization. Microbiol. Mol. Biol. Rev. 1989, 53, 85–108. [Google Scholar] [CrossRef] [Green Version]
  2. Zonia, L.E.; Stebbins, N.E.; Polacco, J.C. Essential role of urease in germination of nitrogen-limited Arabidopsis thaliana seeds. Plant Physiol. 1995, 107, 1097–1103. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Mulvaney, R.L.; Bremner, J.M. Soil Biochemistry; Paul, E.A., Ladd, J.N., Eds.; Marcel Dekker, Inc.: New York, NY, USA, 1981; pp. 153–196. [Google Scholar]
  4. Kataria, R.; Khatkar, A. In-silico Designing, ADMET Analysis, Synthesis and Biological Evaluation of Novel Derivatives of Diosmin Against Urease Protein and Helicobacter pylori Bacterium. Curr. Top. Med. Chem. 2019, 19, 2658–2675. [Google Scholar] [CrossRef] [PubMed]
  5. Li, W.Y.; Ni, W.W.; Ye, Y.X.; Fang, H.L.; Pan, X.M.; He, J.L.; Zhou, T.-L.; Yi, J.; Liu, S.-S.; Zhou, M.; et al. N-monoarylacetothioureas as potent urease inhibitors: Synthesis, SAR, and biological evaluation. J. Enzyme Inhib. Med. Chem. 2020, 35, 404–413. [Google Scholar] [CrossRef] [Green Version]
  6. Qamar, N.; Sultan, H.; Raheel, A.; Ashfaq, M.; Azmat, R.; Naz, R.; Raheela, L.M.; Khan, K.M.; Arshad, T. Heterochelates of metals as an effective anti-Urease agents couple with their docking studies. Pak. J. Pharm. Sci. 2019, 32, 1179–1183. [Google Scholar]
  7. Shah, S.R.; Shah, Z.; Khiat, M.; Khan, A.; Hill, L.R.; Khan, S.; Hussain, J.; Csuk, R.; Anwar, M.U.; Al-Harrasi, A. Complexes of N-and O-Donor Ligands as Potential Urease Inhibitors. ACS Omega 2020, 17, 10200–10206. [Google Scholar] [CrossRef]
  8. Hanif, M.; Saleem, M.; Hussain, M.T.; Rama, N.H.; Zaib, S.; Aslam MA, M.; Iqbal, J. Synthesis, urease inhibition, antioxidant and antibacterial studies of some 4-amino-5-aryl-3H-1,2,4-triazole-3-thiones and their 3,6-disubstituted 1,2,4-triazolo[3,4-b]1,3,4-thiadiazole derivatives. J. Braz. Chem. Soc. 2012, 23, 854–860. [Google Scholar] [CrossRef] [Green Version]
  9. Kafarski, P.; Talma, M. Recent advances in design of new urease inhibitors: A review. J. Adv. Res. 2018, 13, 101–112. [Google Scholar] [CrossRef]
  10. Shamim, S.; Khan, K.M.; Salar, U.; Ali, F.; Lodhi, M.A.; Taha, M.; Perveen, S. 5-Acetyl-6-methyl-4-aryl-3,4-dihydropyrimidin-2(1H)-ones: As potent urease inhibitors; synthesis, in vitro screening, and molecular modeling study. Bioorg. Chem. 2018, 76, 37–52. [Google Scholar] [CrossRef]
  11. Perveen, S.; Khan, K.M.; Lodhi, M.A.; Choudhary, M.I.; Voelter, W. Urease and α-chymotrypsin inhibitory effects of selected urea derivatives. Lett. Drug Des. Discov. 2008, 5, 401–405. [Google Scholar] [CrossRef]
  12. Pervez, H.; Chohan, Z.H.; Ramzan, M.; Nasim FU, H.; Khan, K.M. Synthesis and biological evaluation of some new N(4)-substituted isatin-3-thiosemicarbazones. J. Enzyme Inhib. Med. Chem. 2009, 24, 437–446. [Google Scholar] [CrossRef] [PubMed]
  13. Aslam MA, S.; Mahmood, S.U.; Shahid, M.; Saeed, A.; Iqbal, J. Synthesis, biological assay in vitro and molecular docking studies of new Schiff base derivatives as potential urease inhibitors. Eur. J. Med. Chem. 2011, 46, 5473–5479. [Google Scholar] [CrossRef] [PubMed]
  14. Arora, R.; Issar, U.; Kakkar, R. In silico study of the active site of Helicobacter pylori urease and its inhibition by hydroxamic acids. J. Mol. Graph. Model. 2018, 83, 64–73. [Google Scholar] [CrossRef] [PubMed]
  15. Taha, M.; Wadood, A. Synthesis and molecular docking study of piperazine derivatives as potent urease inhibitors. Bioorg. Chem. 2018, 78, 411–417. [Google Scholar]
  16. Khan, K.M.; Iqbal, S.; Lodhi, M.A.; Maharvi, G.M.; Choudhary, M.I.; Perveen, S. Biscoumarin: New class of urease inhibitors; economical synthesis and activity. Bioorg. Med. Chem. 2004, 12, 1963–1968. [Google Scholar] [CrossRef]
  17. Menteşe, E.; Bektaş, H.; Sokmen, B.B.; Emirik, M.; Çakır, D.; Kahveci, B. Synthesis and molecular docking study of some 5,6-dichloro-2-cyclopropyl-1H-benzimidazole derivatives bearing triazole, oxadiazole, and imine functionalities as potent inhibitors of urease. Bioorg. Med. Chem. Lett. 2017, 27, 3014–3018. [Google Scholar]
  18. Saeed, A.; Mahmood, S.U.; Rafiq, M.; Ashraf, Z.; Jabeen, F.; Seo, S.Y. Iminothiazoline-sulfonamide hybrids as Jack bean urease inhibitors; synthesis, kinetic mechanism and computational molecular modeling. Chem. Biol. Drug. Des. 2016, 87, 434–443. [Google Scholar] [CrossRef]
  19. Mabkhot, Y.N.; Barakat, A.; Yousuf, S. Substituted thieno [2, 3-b] thiophenes and related congeners: Synthesis, β-glucuronidase inhibition activity, crystal structure, and POM analyses. Bioorg. Med. Chem. 2014, 22, 6715–6725. [Google Scholar] [CrossRef]
  20. Mabkhot, Y.N.; Al-Majid, A.M.; Barakat, A.; Alshahrani, S.; Siddiqui, Y. 1, 1′-(3-Methyl-4-phenylthieno [2–b] thiophene-2, 5-diyl) diethanone as a building block in heterocyclic synthesis. novel synthesis of some pyrazole and pyrimidine derivatives. Molecules 2011, 16, 6502–6511. [Google Scholar] [CrossRef] [Green Version]
  21. Fırıncı, R.; Fırıncı, E.; Başbülbül, G.; Dabanca, M.B.; Celepci, D.B.; Günay, M.E. Enamines of 1, 3-dimethylbarbiturates and their symmetrical palladium (II) complexes: Synthesis, characterization and biological activity. Trans. Met. Chem. 2019, 44, 391–397. [Google Scholar]
  22. Cox, D.S.; Scott, K.R.; Gao, H.; Eddington, N.D. Effect of P-glycoprotein on the pharmacokinetics and tissue distribution of enaminone anticonvulsants: Analysis by population and physiological approaches. J. Pharmacol. Exp. Ther. 2002, 302, 1096–1104. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Kidwai, M.; Thakur, R.; Mohan, R. Ecofriendly synthesis of novel antifungal (thio)- barbituric acid derivatives. Acta Chim. Solv. 2005, 52, 88–92. [Google Scholar]
  24. Figueiredo, J.; Serrano, J.L.; Cavalheiro, E.; Keurulainen, L.; Yli-Kauhaluoma, J.; Moreira, V.M.; Almeida, P. Trisubstituted barbiturates and thiobarbiturates: Synthesis and biological evaluation as xanthine oxidase inhibitors, antioxidants, antibacterial and anti-proliferative agents. Eur. J. Med. Chem. 2018, 143, 829–842. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Dabholkar, V.V.; Ravi, T.D. Synthesis of Biginelli products of thiobarbituric acids and their antimicrobial activity. J. Serb. Chem. Soc. 2010, 75, 1033–1040. [Google Scholar] [CrossRef]
  26. Ma, L.; Li, S.; Zheng, H. Synthesis and biological activity of novel barbituric and thiobarbituric acid derivatives against non-alcoholic fatty liver disease. Eur. J. Med. Chem. 2011, 46, 2003. [Google Scholar] [CrossRef] [PubMed]
  27. Khan, K.M.; Ali, M.; Farooqui, T.A.; Khan, M.; Taha, M.; Perveen, S. An improved method for the synthesis of 5- arylidene barbiturates using BiCl3. J. Chem. Soc. Pak. 2009, 31, 823–828. [Google Scholar]
  28. Archana, S.V.K.; Kumar, A. Synthesis of some newer derivatives of substitute quinazolinonyl-2-oxo/thiobarbituric acid as potent anticonvulsant agents. Bioorg. Med. Chem. 2004, 12, 1257–1264. [Google Scholar] [CrossRef]
  29. Hassan, M.; Khan Faidallah, K.A. Synthesis and biological evaluation of new barbituric and thiobarbituric acid fluoro analogs of benzenesulfonamides as antidiabetic and antibacterial agents. J. Fluorine Chem. 2012, 142, 96–104. [Google Scholar]
  30. Ali, M.; Barakat, A.; El-Faham, A.; Al-Rasheed, H.H.; Dahlous, K.; Al-Majid, A.M.; Choudhary, M.I. Synthesis and characterisation of thiobarbituric acid enamine derivatives, and evaluation of their α-glucosidase inhibitory and anti-glycation activity. Enzyme Inhib. Med. Chem. 2020, 35, 692–701. [Google Scholar] [CrossRef]
  31. Puerta, D.T.; Cohen, S.M.A. A bioinorganic perspective on matrix metalloproteinase inhibition. Curr. Topic. Med. Chem. 2004, 4, 1551–1573. [Google Scholar] [CrossRef]
  32. Hameed, A.; Al-Rashida, M.; Uroos, M. A patent update on therapeutic applications of urease inhibitors (2012–2018). Expert Opin. Rapeutic Pat. 2019, 29, 181–189. [Google Scholar] [CrossRef] [PubMed]
  33. Barakat, A.; Soliman, S.M.; Ali, M.; Elmarghany, A.; Al-Majid, A.M.; Yousuf, S.; El-Faham, A. Synthesis, crystal structure, evaluation of urease inhibition potential and the docking studies of cobalt(III) complex based on barbituric acid Schiff base ligand. Inorg. Chim. Acta. 2020, 503, 119405. [Google Scholar] [CrossRef]
  34. Barakat, A.; Ali, M.; Al-Majid, A.M.; Yousuf, S.; Choudhary, M.I.; Khalil, R.; Ul-Haq, Z. Synthesis of thiobarbituric acid derivatives: In vitro α-glucosidase inhibition and molecular docking studies. Bioorg. Chem. 2017, 75, 99–105. [Google Scholar] [CrossRef] [PubMed]
  35. Barakat, A.; Al-Majid, A.M.; Al-Najjar, H.J.; Mabkhot, Y.N.; Javaid, S.; Yousuf, S.; Choudhary, M.I. Zwitterionic pyrimidinium adducts as antioxidants with therapeutic potential as nitric oxide scavenger. Eur. J. Med. Chem. 2014, 84, 146. [Google Scholar] [CrossRef] [PubMed]
  36. Barakat, A.; Islam, M.S.; Al-Majid, A.M.; Ghabbour, H.A.; Fun, H.K.; Javed, K.; Wadood, A. Synthesis, in vitro biological activities and in silico study of dihydropyrimidines derivatives. Bioorg.Med. Chem. 2015, 23, 6740. [Google Scholar] [CrossRef] [PubMed]
  37. Barakat, A.; Al-Majid, A.M.; Soliman, S.M.; Lotfy, G.; Ghabbour, H.A.; Fun, H.K.; Sloop, J.C. New diethyl ammonium salt of thiobarbituric acid derivative: Synthesis, molecular structure investigations and docking studies. Molecules 2015, 20, 20642–20658. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  38. Barakat, A.; Al-Majid, A.M.; Lotfy, G.; Arshad, F.; Yousuf, S.; Choudhary, M.I.; Ul-Haq, Z. Synthesis and Dynamics Studies of Barbituric Acid Derivatives as Urease Inhibitors. Chem. Cent. J. 2015, 9, 63. [Google Scholar] [CrossRef] [Green Version]
  39. Badria, F.A.; Atef, S.; Al-Majid, A.M.; Ali, M.; Elshaier, Y.A.; Ghabbour, H.A.; Barakat, A. Synthesis and inhibitory effect of some indole-pyrimidine-based hybrid heterocycles on α-glucosidase and α-amylase as potential hypoglycemic agents. ChemistryOpen 2019, 8, 1288–1297. [Google Scholar] [CrossRef]
  40. Barakat, A.; Islam, M.S.; Al-Majid, A.M.; Ghabbour, H.A.; Yousuf, S.; Ashraf, M.; Ul-Haq, Z. Synthesis of pyrimidine-2,4,6-trione derivatives: Anti-oxidant, Anti-cancer, α-glucosidase, ß-glucuronidase inhibiton and their molecular docking studies. Bioorg. Chem. 2016, 86, 72–79. [Google Scholar] [CrossRef]
  41. Barakat, A.; Soliman, S.M.; Elshaier, Y.A.M.M. Molecular structure investigation, hypoglycemic/anticancer and docking studies of 5-((4-fluorophenyl)(2-hydroxy-6-oxocyclohex-1-en-1-yl)methyl)-6-hydroxy-1,3-dimethylpyrimidine-2,4(1H,3H)-dione. J. Mol. Struc. 2017, 1134, 99–111. [Google Scholar] [CrossRef]
  42. Ul-Haq, Z.; Ashraf, S.; Al-Majid, A.M.; Barakat, A. 3D-QSAR Studies on Barbituric Acid Derivatives as Urease Inhibitors and the Effect of Charges on the Quality of a Model. Int. J. Mol. Sci. 2016, 17, 657. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Khan, K.M.; Rahim, F.; Khan, A. Synthesis and structure–activity relationship of thiobarbituric acid derivatives as potent inhibitors of urease. Bioorg. Med. Chem. 2014, 22, 4119. [Google Scholar] [CrossRef] [PubMed]
  44. Altowyan, M.S.; Barakat, A.; Soliman, S.M.; Al-Majid, A.M.; Ali, M.; Elshaier, Y.A.; Ghabbour, H.A. A New Barbituric Acid Derivatives as Reactive Oxygen Scavenger: Experimental and Theoretical Investigations. J. Mol. Struc. 2019, 1175, 524–535. [Google Scholar] [CrossRef]
  45. Weatherburn, M.W. Phenol-hypochlorite reaction for determination of ammonia. Anal. Chem. 1967, 39, 971. [Google Scholar] [CrossRef]
  46. Mabkhot, Y.N.; Aldawsari, F.D.; Al-Showiman, S.S.; Barakat, A.; Soliman, S.M.; Choudhary, M.I.; Hadda, T.B. Novel enaminone derived from thieno [2–b] thiene: Synthesis, x-ray crystal structure, HOMO, LUMO, NBO analyses and biological activity. Chem. Cent. J. 2015, 9, 24. [Google Scholar] [CrossRef] [Green Version]
  47. Khan, M.K.; Saify, Z.S.; Arif Lodhi, M.; Butt, N.; Perveen, S.; Murtaza Maharvi, G.; Atta-Ur-Rahman. Piperidines: A new class of urease inhibitors. Nat. Prod. Res. 2006, 20, 523. [Google Scholar] [CrossRef]
  48. Begum, A.; Banumathi, S.; Choudhary, M.I.; Betzel, C. Crystallographic structure analysis of urease from Jack bean (Canavalia ensiformis) at 1.49 A Resolution. Macromolecules 2012. [Google Scholar] [CrossRef]
  49. Kulinich, A.V.; Derevyanko, N.A.; Ishchenko, A.A. Synthesis and spectral properties of cyanine dyes-derivatives of 10,10-dimethyl-7,8,9,10-tetrahydro-6H-pyrido[1–a]indolium. J. Photochem. Photobiol. 2008, 198, 119–125. [Google Scholar] [CrossRef]
  50. Gorobets, N.Y.; Yousefi, B.H.; Belaj, F.; Kappe, C.O. Rapid microwave-assisted solution phase synthesis of substituted 2-pyridone libraries. Tetrahedron 2004, 60, 8633–8644. [Google Scholar] [CrossRef]
  51. Kumar, S.; Kayastha, A.M. Acetohydroxamic Acid—A Competitive Inhibitor of Urease from Soybean “Glycine max”. J. Proteins Proteom. 2013, 1, 3–8. [Google Scholar]
  52. Krajewska, B.; Zaborska, W. Jack bean urease: The effect of active-site binding inhibitors on the reactivity of enzyme thiol groups. Bioorg. Chem. 2007, 35, 355–365. [Google Scholar] [CrossRef] [PubMed]
  53. Chemical Computing Group. Molecular Operating Environment (MOE) 2016.0810; Chemical Computing Group Inc.: Montreal, QC, Canada, 2016; Available online: https://www.chemcomp.com/Products.htm (accessed on 19 May 2020).
  54. Rauf, A.; Shahzad, S.; Bajda, M.; Yar, M.; Ahmed, F.; Hussain, N.; Jończyk, J. Design and synthesis of new barbituric-and thiobarbituric acid derivatives as potent urease inhibitors: Structure activity relationship and molecular modeling studies. Bioorg. Med. Chem. 2015, 23, 6049–6058. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Stephens, P.J.; Devlin, F.J.; Chabalowski, C.F.N.; Frisch, M.J. Ab initio calculation of vibrational absorption and circular dichroism spectra using density functional force fields. J. Phys. Chem. 1994, 98, 11623–11627. [Google Scholar] [CrossRef]
Figure 1. Some examples of bacterial urease inhibitors.
Figure 1. Some examples of bacterial urease inhibitors.
Applsci 10 03523 g001
Scheme 1. Synthesis of 3a–p, 4, and 5.
Scheme 1. Synthesis of 3a–p, 4, and 5.
Applsci 10 03523 sch001
Figure 2. Depiction of the docking results of low energy conformer: (a) 3a, (b) 3h, (c) 3d, (d) 3j, (e) 3m, (f) 3f, (g) 3p, and (h) 5. The key residues are presented as sticks models and nickel atoms are shown as green circles.
Figure 2. Depiction of the docking results of low energy conformer: (a) 3a, (b) 3h, (c) 3d, (d) 3j, (e) 3m, (f) 3f, (g) 3p, and (h) 5. The key residues are presented as sticks models and nickel atoms are shown as green circles.
Applsci 10 03523 g002
Figure 3. The optimized geometries and surfaces of the HOMO (Highest occupied molecular orbital)-LUMO (Lowest unoccupied molecular orbital) of 3h, 3k and 3o obtained at the B3LYP/6-31G (d, p) level.
Figure 3. The optimized geometries and surfaces of the HOMO (Highest occupied molecular orbital)-LUMO (Lowest unoccupied molecular orbital) of 3h, 3k and 3o obtained at the B3LYP/6-31G (d, p) level.
Applsci 10 03523 g003
Figure 4. The molecular electrostatic potential (MEP) of 3h, 3k, and 3o.
Figure 4. The molecular electrostatic potential (MEP) of 3h, 3k, and 3o.
Applsci 10 03523 g004
Table 1. Urease inhibition capacity of compounds 3a–p, 4, and 5.
Table 1. Urease inhibition capacity of compounds 3a–p, 4, and 5.
CompoundStructureUrease Inhibition IC50 ± SEM [µM]
3a Applsci 10 03523 i0019 ± 2
3b Applsci 10 03523 i002NA
3c Applsci 10 03523 i00326 ± 1
3d Applsci 10 03523 i0048 ± 0.3
3e Applsci 10 03523 i00511 ± 0.3
3f Applsci 10 03523 i00610 ± 0.6
3g Applsci 10 03523 i007NA
3h Applsci 10 03523 i0086.4 ± 0.3
3i Applsci 10 03523 i00966 ± 2.4
3j Applsci 10 03523 i01010 ± 0.9
3k Applsci 10 03523 i01111 ± 1.2
3l Applsci 10 03523 i0128 ± 0.4
3m Applsci 10 03523 i01315 ± 1.2
3n Applsci 10 03523 i01411 ± 1.1
3o Applsci 10 03523 i0159 ± 0.3
3p Applsci 10 03523 i01622 ± 0.8
4 Applsci 10 03523 i01710 ± 1.2
5 Applsci 10 03523 i01842 ± 2.3
STDAcetohydroxamic Acid (AHA)20 ± 0.4
Table 2. Theoretical results of pyrimidine derivatives.
Table 2. Theoretical results of pyrimidine derivatives.
CompoundIC50HOMO (eV)LUMO (eV)Egap (eV)
3a9 ± 2−5.79330622969−1.21770997548−4.57559625421
3d8 ± 0.3−5.88881823−1.70370550982−4.76988562237
3f10 ± 0.6−6.2202530345−1.96248588673−4.25776714777
3h6 ± 0.3−6.24583174683−1.88955934518−4.35627240165
3k11 ± 1−5.70922301574−2.24439595033−3.46482706541
3o9 ± 0.3−5.75357759138−2.25963433214−3.49394325924

Share and Cite

MDPI and ACS Style

Ali, M.; Barakat, A.; El-Faham, A.; Al-Majid, A.M.; Yousuf, S.; Ashraf, S.; Ul-Haq, Z.; Choudhary, M.I.; de la Torre, B.G.; Albericio, F. Enamine Barbiturates and Thiobarbiturates as a New Class of Bacterial Urease Inhibitors. Appl. Sci. 2020, 10, 3523. https://doi.org/10.3390/app10103523

AMA Style

Ali M, Barakat A, El-Faham A, Al-Majid AM, Yousuf S, Ashraf S, Ul-Haq Z, Choudhary MI, de la Torre BG, Albericio F. Enamine Barbiturates and Thiobarbiturates as a New Class of Bacterial Urease Inhibitors. Applied Sciences. 2020; 10(10):3523. https://doi.org/10.3390/app10103523

Chicago/Turabian Style

Ali, M., Assem Barakat, Ayman El-Faham, Abdullah Mohammed Al-Majid, Sammer Yousuf, Sajda Ashraf, Zaheer Ul-Haq, M. Iqbal Choudhary, Beatriz G. de la Torre, and Fernando Albericio. 2020. "Enamine Barbiturates and Thiobarbiturates as a New Class of Bacterial Urease Inhibitors" Applied Sciences 10, no. 10: 3523. https://doi.org/10.3390/app10103523

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