271
Views
0
CrossRef citations to date
0
Altmetric
Research Articles

Identification of novel natural MurD ligase inhibitors as potential antimicrobial agents targeting Acinetobacter baumannii: In silico screening and biological evaluation

ORCID Icon, , ORCID Icon, ORCID Icon, ORCID Icon & ORCID Icon
Pages 14051-14066 | Received 06 Jul 2021, Accepted 26 Oct 2021, Published online: 12 Nov 2021

References

  • Ahmad, S., Murtaza, U. A., Raza, S., & Azam, S. S. (2019). Blocking the catalytic mechanism of MurC ligase enzyme from Acinetobacter baumannii: An in Silico guided study towards the discovery of natural antibiotics. Journal of Molecular Liquids, 281, 117–133. https://doi.org/10.1016/j.molliq.2019.02.051
  • Alsan, M., & Klompas, M. (2010). Acinetobacter baumannii: An emerging and important pathogen. Journal of Clinical Outcomes Management: JCOM, 17(8), 363–369.
  • Altschul, S. F., Gish, W., Miller, W., Myers, E. W., & Lipman, D. J. (1990). Basic local alignment search tool. Journal of Molecular Biology, 215(3), 403–410. https://doi.org/10.1016/S0022-2836(05)80360-2
  • Amera, G. M., Khan, R. J., Pathak, A., Jha, R. K., Muthukumaran, J., & Singh, A. K. (2020). Computer aided ligand based screening for identification of promising molecules against enzymes involved in peptidoglycan biosynthetic pathway from Acinetobacter baumannii. Microbial Pathogenesis, 147, 104205. https://doi.org/10.1016/j.micpath.2020.104205
  • Amera, G. M., Khan, R. J., Pathak, A., Kumar, A., & Singh, A. K. (2019). Structure based in-silico study on UDP-N-acetylmuramoyl-L-alanyl-D-glutamate-2,6-diaminopimelate ligase (MurE) from Acinetobacter baumannii as a drug target against nosocomial infections. Informatics in Medicine Unlocked, 16, 100216. https://doi.org/10.1016/j.imu.2019.100216
  • Bailly, C. (2019). Cepharanthine: An update of its mode of action, pharmacological properties and medical applications. Phytomedicine: International Journal of Phytotherapy and Phytopharmacology, 62, 152956. https://doi.org/10.1016/j.phymed.2019.152956
  • Barreteau, H., Sosič, I., Turk, S., Humljan, J., Tomašić, T., Zidar, N., Hervé, M., Boniface, A., Peterlin-Mašič, L., Kikelj, D., Mengin-Lecreulx, D., Gobec, S., & Blanot, D. (2012). MurD enzymes from different bacteria: Evaluation of inhibitors. Biochemical Pharmacology, 84(5), 625–632. https://doi.org/10.1016/j.bcp.2012.06.006
  • Bertrand, J. A., Auger, G., Fanchon, E., Martin, L., Blanot, D., van Heijenoort, J., & Dideberg, O. (1997). Crystal structure of UDP-N-acetylmuramoyl-L-alanine:D-glutamate ligase from Escherichia coli. The EMBO Journal, 16(12), 3416–3425. https://doi.org/10.1093/emboj/16.12.3416
  • Bertrand, J. A., Auger, G., Martin, L., Fanchon, E., Blanot, D., Le Beller, D., van Heijenoort, J., & Dideberg, O. (1999). Determination of the MurD mechanism through crystallographic analysis of enzyme complexes. Journal of Molecular Biology, 289(3), 579–590. https://doi.org/10.1006/jmbi.1999.2800
  • Bugg, T. D. H., Braddick, D., Dowson, C. G., & Roper, D. I. (2011). Bacterial cell wall assembly: Still an attractive antibacterial target. Trends in Biotechnology, 29(4), 167–173. https://doi.org/10.1016/j.tibtech.2010.12.006
  • Cerqueira, G. M., & Peleg, A. Y. (2011). Insights into Acinetobacter baumannii pathogenicity. IUBMB Life, 63(12), 1055–1060. https://doi.org/10.1002/iub.533
  • Chang, H. R., Vladoianu, I. R., & Pechère, J. C. (1990). Effects of ampicillin, ceftriaxone, chloramphenicol, pefloxacin and trimethoprim-sulphamethoxazole on Salmonella typhi within human monocyte-derived macrophages. The Journal of Antimicrobial Chemotherapy, 26(5), 689–694. https://doi.org/10.1093/jac/26.5.689
  • Cheng, T., Li, Q., Zhou, Z., Wang, Y., & Bryant, S. H. (2012). Structure-based virtual screening for drug discovery: A problem-centric review. The AAPS Journal, 14(1), 133–141. https://doi.org/10.1208/s12248-012-9322-0
  • Dai, Y., Harinantenaina, L., Bowman, J. D., Da Fonseca, I. O., Brodie, P. J., Goetz, M., Cassera, M. B., & Kingston, D. G. I. (2014). Isolation of antiplasmodial anthraquinones from Kniphofia ensifolia, and synthesis and structure-activity relationships of related compounds. Bioorganic & Medicinal Chemistry, 22(1), 269–276. https://doi.org/10.1016/j.bmc.2013.11.032
  • Dallakyan, S., & Olson, A. J. (2015). Small-molecule library screening by docking with PyRx. Methods in Molecular Biology (Clifton, NJ), 1263, 243–250. https://doi.org/10.1007/978-1-4939-2269-7_19
  • Dannenberg, J. J. (1998). An introduction to hydrogen bonding by George A. Jeffrey (University of Pittsburgh). Oxford University Press: New York and Oxford. 1997. ix + 303 pp. $60.00. ISBN 0-19-509549-9. Journal of the American Chemical Society, 120(22), 5604–5604. https://doi.org/10.1021/ja9756331
  • Dong, C.-L., Li, L.-X., Cui, Z.-H., Chen, S.-W., Xiong, Y. Q., Lu, J.-Q., Liao, X.-P., Gao, Y., Sun, J., & Liu, Y.-H. (2017). Synergistic effect of pleuromutilins with other antimicrobial agents against Staphylococcus aureus in vitro and in an experimental galleria mellonella model. Frontiers in Pharmacology, 8, 553. https://doi.org/10.3389/fphar.2017.00553
  • Eisenberg, D., Lüthy, R., & Bowie, J. U. (1997). VERIFY3D: Assessment of protein models with three-dimensional profiles. Methods in Enzymology, 277, 396–404. https://doi.org/10.1016/s0076-6879(97)77022-8
  • Feher, M., & Schmidt, J. M. (2003). Property distributions: Differences between drugs, natural products, and molecules from combinatorial chemistry. Journal of Chemical Information and Computer Sciences, 43(1), 218–227. https://doi.org/10.1021/ci0200467
  • Fiser, A., Do, R. K., & Sali, A. (2000). Modeling of loops in protein structures. Protein Science, 9(9), 1753–1773. https://doi.org/10.1110/ps.9.9.1753
  • Gilson, M. K., & Honig, B. (1988). Calculation of the total electrostatic energy of a macromolecular system: Solvation energies, binding energies, and conformational analysis. Proteins, 4(1), 7–18. https://doi.org/10.1002/prot.340040104
  • Gimeno, A., Ojeda-Montes, M. J., Tomás-Hernández, S., Cereto-Massagué, A., Beltrán-Debón, R., Mulero, M., Pujadas, G., & Garcia-Vallvé, S. (2019). The light and dark sides of virtual screening: What is there to know? International Journal of Molecular Sciences, 20(6), 1375. https://doi.org/10.3390/ijms20061375
  • Harder, E., Damm, W., Maple, J., Wu, C., Reboul, M., Xiang, J. Y., Wang, L., Lupyan, D., Dahlgren, M. K., Knight, J. L., Kaus, J. W., Cerutti, D. S., Krilov, G., Jorgensen, W. L., Abel, R., & Friesner, R. A. (2016). OPLS3: A force field providing broad coverage of drug-like small molecules and proteins. Journal of Chemical Theory and Computation, 12(1), 281–296. https://doi.org/10.1021/acs.jctc.5b00864
  • Harvey, A. L. (2008). Natural products in drug discovery. Drug Discovery Today, 13(19–20), 894–901. https://doi.org/10.1016/j.drudis.2008.07.004
  • Hess, B., Bekker, H., Berendsen, H. J. C., & Fraaije, J. G. E. M. (1997). LINCS: A linear constraint solver for molecular simulations. Journal of Computational Chemistry, 18(12), 1463–1472. https://doi.org/10.1002/(SICI)1096-987X(199709)18:12 < 1463::AID-JCC4 > 3.0.CO;2-H
  • Jha, R. K., Khan, R. J., Amera, G. M., Singh, E., Pathak, A., Jain, M., Muthukumaran, J., & Singh, A. K. (2020). Identification of promising molecules against MurD ligase from Acinetobacter baumannii: Insights from comparative protein modelling, virtual screening, molecular dynamics simulations and MM/PBSA analysis. Journal of Molecular Modeling, 26(11), 304. https://doi.org/10.1007/s00894-020-04557-4
  • Kaur, N., Khokhar, M., Jain, V., Bharatam, P. V., Sandhir, R., & Tewari, R. (2013). Identification of druggable targets for Acinetobacter baumannii via subtractive genomics and plausible inhibitors for MurA and MurB. Applied Biochemistry and Biotechnology, 171(2), 417–436. https://doi.org/10.1007/s12010-013-0372-2
  • Kotnik, M., Humljan, J., Contreras-Martel, C., Oblak, M., Kristan, K., Hervé, M., Blanot, D., Urleb, U., Gobec, S., Dessen, A., & Solmajer, T. (2007). Structural and functional characterization of enantiomeric glutamic acid derivatives as potential transition state analogue inhibitors of MurD ligase. Journal of Molecular Biology, 370(1), 107–115. https://doi.org/10.1016/j.jmb.2007.04.048
  • Kumari, R., Kumar, R., Consortium, O. S. D. D., & Lynn, A. (2014). g_mmpbsa—A GROMACS Tool for High-Throughput MM-PBSA Calculations (world) [Research-article]. American Chemical Society. https://doi.org/10.1021/ci500020m
  • Laskowski, R. A., Rullmannn, J. A., MacArthur, M. W., Kaptein, R., & Thornton, J. M. (1996). AQUA and PROCHECK-NMR: Programs for checking the quality of protein structures solved by NMR. Journal of Biomolecular NMR, 8(4), 477–486.
  • Liu, C., Chang, Y., Xu, Y., Luo, Y., Wu, L., Mei, Z., Li, S., Wang, R., & Jia, X. (2018). Distribution of virulence-associated genes and antimicrobial susceptibility in clinical Acinetobacter baumannii isolates. Oncotarget, 9(31), 21663–21673. https://doi.org/10.18632/oncotarget.24651
  • Miyasaki, Y., Rabenstein, J. D., Rhea, J., Crouch, M.-L., Mocek, U. M., Kittell, P. E., Morgan, M. A., Nichols, W. S., Benschoten, M. M. V., Hardy, W. D., & Liu, G. Y. (2013). Isolation and characterization of antimicrobial compounds in plant extracts against multidrug-resistant Acinetobacter baumannii. PLOS One, 8(4), e61594. https://doi.org/10.1371/journal.pone.0061594
  • Montefour, K., Frieden, J., Hurst, S., Helmich, C., Headley, D., Martin, M., & Boyle, D. A. (2008). Acinetobacter baumannii: An emerging multidrug-resistant pathogen in critical care. Critical Care Nurse, 28(1), 15–25.
  • Moraes, G. L., Gomes, G. C., de Sousa, P. R. M., Alves, C. N., Govender, T., Kruger, H. G., Maguire, G. E. M., Lamichhane, G., & Lameira, J. (2015). Structural and functional features of enzymes of Mycobacterium tuberculosis peptidoglycan biosynthesis as targets for drug development. Tuberculosis (Edinburgh, Scotland), 95(2), 95–111. https://doi.org/10.1016/j.tube.2015.01.006
  • Morris, G. M., Huey, R., Lindstrom, W., Sanner, M. F., Belew, R. K., Goodsell, D. S., & Olson, A. J. (2009). AutoDock4 and AutoDockTools4: Automated docking with selective receptor flexibility. Journal of Computational Chemistry, 30(16), 2785–2791. https://doi.org/10.1002/jcc.21256
  • Pires, D. E. V., Blundell, T. L., & Ascher, D. B. (2015). pkCSM: Predicting small-molecule pharmacokinetic and toxicity properties using graph-based signatures. Journal of Medicinal Chemistry, 58(9), 4066–4072. https://doi.org/10.1021/acs.jmedchem.5b00104
  • Pronk, S., Páll, S., Schulz, R., Larsson, P., Bjelkmar, P., Apostolov, R., Shirts, M. R., Smith, J. C., Kasson, P. M., van der Spoel, D., Hess, B., & Lindahl, E. (2013). GROMACS 4.5: A high-throughput and highly parallel open source molecular simulation toolkit. Bioinformatics (Oxford, England), 29(7), 845–854. https://doi.org/10.1093/bioinformatics/btt055
  • QikProp. (2017). Schrödinger Release 2017-1: QikProp. Schrödinger, LLC.
  • Sangshetti, J. N., Joshi, S. S., Patil, R. H., Moloney, M. G., & Shinde, D. B. (2017). Mur Ligase inhibitors as anti-bacterials: A comprehensive review. Current Pharmaceutical Design, 23(21), 3164–3196. https://doi.org/10.2174/1381612823666170214115048
  • Santajit, S., & Indrawattana, N. (2016). Mechanisms of antimicrobial resistance in ESKAPE pathogens. BioMed Research International, 2016, 2475067. https://doi.org/10.1155/2016/2475067
  • Šink, R., Barreteau, H., Patin, D., Mengin-Lecreulx, D., Gobec, S., & Blanot, D. (2013). MurD enzymes: Some recent developments. Biomolecular Concepts, 4(6), 539–556. https://doi.org/10.1515/bmc-2013-0024
  • Skariyachan, S., Manjunath, M., & Bachappanavar, N. (2019). Screening of potential lead molecules against prioritised targets of multi-drug-resistant-Acinetobacter baumannii - insights from molecular docking, molecular dynamic simulations and in vitro assays. Journal of Biomolecular Structure & Dynamics, 37(5), 1146–1169. https://doi.org/10.1080/07391102.2018.1451387
  • Skariyachan, S., Muddebihalkar, A. G., Badrinath, V., Umashankar, B., Eram, D., Uttarkar, A., & Niranjan, V. (2020). Natural epiestriol-16 act as potential lead molecule against prospective molecular targets of multidrug resistant Acinetobacter baumannii-Insight from in silico modelling and in vitro investigations. Infection, Genetics and Evolution: Journal of Molecular Epidemiology and Evolutionary Genetics in Infectious Diseases, 82, 104314. https://doi.org/10.1016/j.meegid.2020.104314
  • Skariyachan, S., Taskeen, N., Ganta, M., & Venkata Krishna, B. (2019). Recent perspectives on the virulent factors and treatment options for multidrug-resistant Acinetobacter baumannii. Critical Reviews in Microbiology, 45(3), 315–333. https://doi.org/10.1080/1040841X.2019.1600472
  • Sorokina, M., & Steinbeck, C. (2020). Review on natural products databases: Where to find data in 2020. Journal of Cheminformatics, 12(1), 20. https://doi.org/10.1186/s13321-020-00424-9
  • Sosič, I., Barreteau, H., Simčič, M., Sink, R., Cesar, J., Zega, A., Grdadolnik, S. G., Contreras-Martel, C., Dessen, A., Amoroso, A., Joris, B., Blanot, D., & Gobec, S. (2011). Second-generation sulfonamide inhibitors of D-glutamic acid-adding enzyme: Activity optimisation with conformationally rigid analogues of D-glutamic acid. European Journal of Medicinal Chemistry, 46(7), 2880–2894. https://doi.org/10.1016/j.ejmech.2011.04.011
  • Sterling, T., & Irwin, J. J. (2015). ZINC 15-ligand discovery for everyone. Journal of Chemical Information and Modeling, 55(11), 2324–2337. https://doi.org/10.1021/acs.jcim.5b00559
  • Tomašić, T., Kovač, A., Simčič, M., Blanot, D., Grdadolnik, S. G., Gobec, S., Kikelj, D., & Peterlin Mašič, L. (2011). Novel 2-thioxothiazolidin-4-one inhibitors of bacterial MurD ligase targeting d-Glu- and diphosphate-binding sites. European Journal of Medicinal Chemistry, 46(9), 3964–3975. https://doi.org/10.1016/j.ejmech.2011.05.070
  • Wang, H., Cheng, H., Wang, F., Wei, D., & Wang, X. (2010). An improved 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) reduction assay for evaluating the viability of Escherichia coli cells. Journal of Microbiological Methods, 82(3), 330–333. https://doi.org/10.1016/j.mimet.2010.06.014
  • Werneck, J. S., Picão, R. C., Carvalhaes, C. G., Cardoso, J. P., & Gales, A. C. (2011). OXA-72-producing Acinetobacter baumannii in Brazil: A case report. The Journal of Antimicrobial Chemotherapy, 66(2), 452–454. https://doi.org/10.1093/jac/dkq462
  • Wiederstein, M., & Sippl, M. J. (2007). ProSA-web: Interactive web service for the recognition of errors in three-dimensional structures of proteins. Nucleic Acids Research, 35(Web Server issue), W407–410. https://doi.org/10.1093/nar/gkm290
  • Zidar, N., Tomašić, T., Šink, R., Rupnik, V., Kovač, A., Turk, S., Patin, D., Blanot, D., Contreras Martel, C., Dessen, A., Müller Premru, M., Zega, A., Gobec, S., Peterlin Mašič, L., & Kikelj, D. (2010). Discovery of Novel 5-benzylidenerhodanine and 5-benzylidenethiazolidine-2,4-dione inhibitors of MurD ligase. Journal of Medicinal Chemistry, 53(18), 6584–6594. https://doi.org/10.1021/jm100285g

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.