Identification of Natural Compounds as CTX-M-15 Inhibitors for the Management of Multidrug-Resistant Bacteria: An in-silico study

Mohammad Zubair Alam, Absarul Haque

Abstract


Background: Antibiotic resistance is a major global threat to the efficacy of bacterial infection treatment. Resistance to beta-lactam antibiotics in bacteria is primarily caused by the production of extended-spectrum-lactamases, with the CTX-M variant, particularly CTX-M-15, being the most common. The need for an effective CTX-M-15 inhibitor is currently pressing.

Methods: This study screened a library of natural compounds from the ZINC database against the CTX-M-15 protein using the PyRx 0.8 tool. The SwissADME web platform was used to predict the ADMET properties of the five most promising compounds.

Result: The identified hits compounds, ZINC1857626342, ZINC403692, ZINC408773, ZINC57926, and ZINC790938591 exhibited strong binding with CTX-M-15. These compounds interacted with crucial catalytic site residues in the CTX-M-15 protein, particularly Ser70 and Ser130. Notably, the binding energies of these compounds were higher than those of the reference compound avibactam. Furthermore, they exhibited pharmacologically favorable characteristics.

Conclusion: These compounds show promise as potential CTX-M-15 inhibitors to combat bacterial resistance. However, more experimental research is needed to optimize these compounds for their role as CTX-M-15 inhibitors.

Full Text:

PDF

References


Roca I, Akova M, Baquero F, Carlet J, Cavaleri M, et al. The global threat of antimicrobial resistance: science for intervention. New Microbes and New Infections, (2015); 6: 22-29.

Mancuso G, Midiri A, Gerace E, Biondo C. Bacterial Antibiotic Resistance: The Most Critical Pathogens. Pathogens, (2021); 10(10): 1310.

Larsson DGJ, Flach CF. Antibiotic resistance in the environment. Nature Reviews Microbiology, (2022); 20(5): 257-269.

Shakil S, Rizvi SMD, Greig NH. High Throughput Virtual Screening and Molecular Dynamics Simulation for Identifying a Putative Inhibitor of Bacterial CTX-M-15. Antibiotics (Basel), (2021); 10(5): 474.

Yasir M, Ajlan AM, Shakil S, Jiman-Fatani AA, Almasaudi SB, et al. Molecular characterization, antimicrobial resistance and clinico-bioinformatics approaches to address the problem of extended-spectrum beta-lactamase-producing Escherichia coli in western Saudi Arabia. Scientific Reports, (2018); 8(1): 14847.

Bush K, Courvalin P, Dantas G, Davies J, Eisenstein B, et al. Tackling antibiotic resistance. Nature Reviews Microbiology, (2011); 9(12): 894-896.

Bush K, Bradford PA. Epidemiology of beta-Lactamase-Producing Pathogens. Clinical Microbiology Reviews, (2020); 33(2): e00047-19.

Peirano G, Pitout JDD. Extended-Spectrum beta-Lactamase-Producing Enterobacteriaceae: Update on Molecular Epidemiology and Treatment Options. Drugs, (2019); 79(14): 1529-1541.

Smet A, Martel A, Persoons D, Dewulf J, Heyndrickx M, et al. Broad-spectrum beta-lactamases among Enterobacteriaceae of animal origin: molecular aspects, mobility and impact on public health. FEMS Microbiology Reviews, (2010); 34(3): 295-316.

Bevan ER, Jones AM, Hawkey PM. Global epidemiology of CTX-M beta-lactamases: temporal and geographical shifts in genotype. Journal of Antimicrobial Chemotherapy, (2017); 72(8): 2145-2155.

Quinones D, Aung MS, Carmona Y, Gonzalez MK, Pereda N, et al. High Prevalence of CTX-M Type Extended-Spectrum Beta-Lactamase Genes and Detection of NDM-1 Carbapenemase Gene in Extraintestinal Pathogenic Escherichia coli in Cuba. Pathogens, (2020); 9(1): 65.

Mwangi J, Hao X, Lai R, Zhang ZY. Antimicrobial peptides: new hope in the war against multidrug resistance. Zoological Research, (2019); 40(6): 488-505.

Sabe VT, Ntombela T, Jhamba LA, Maguire GEM, Govender T, et al. Current trends in computer aided drug design and a highlight of drugs discovered via computational techniques: A review. European Journal of Medicinal Chemistry, (2021); 224: 113705.

Giordano D, Biancaniello C, Argenio MA, Facchiano A. Drug Design by Pharmacophore and Virtual Screening Approach. Pharmaceuticals (Basel), (2022); 15(5): 646.

Reddy AS, Pati SP, Kumar PP, Pradeep HN, Sastry GN. Virtual screening in drug discovery -- a computational perspective. Current Protein & Peptide Science, (2007); 8(4): 329-351.

Cavasotto CN, Orry AJ. Ligand docking and structure-based virtual screening in drug discovery. Current Topics in Medicinal Chemistry, (2007); 7(10): 1006-1014.

Good AC, Cheney DL. Analysis and optimization of structure-based virtual screening protocols (1): exploration of ligand conformational sampling techniques. Journal of Molecular Graphics and Modelling, (2003); 22(1): 23-30.

Irwin JJ, Shoichet BK. ZINC--a free database of commercially available compounds for virtual screening. Journal of Chemical Information and Modeling, (2005); 45(1): 177-182.

Dallakyan S, Olson AJ. Small-molecule library screening by docking with PyRx. Methods in Molecular Biology, (2015); 1263: 243-250.

Daina A, Michielin O, Zoete V. SwissADME: a free web tool to evaluate pharmacokinetics, drug-likeness and medicinal chemistry friendliness

of small molecules. Scientific Reports, (2017); 7: 42717.

Drawz SM, Papp-Wallace KM, Bonomo RA. New beta-lactamase inhibitors: a therapeutic renaissance in an MDR world. Antimicrobial Agents and Chemotherapy, (2014); 58(4): 1835-1846.

Pinzi L, Rastelli G. Molecular Docking: Shifting Paradigms in Drug Discovery. International Journal of Molecular Sciences, (2019); 20(18): 4331.

Gupta M, Sharma R, Kumar A. Docking techniques in pharmacology: How much promising? Computational Biology and Chemistry, (2018); 76: 210-217.

Meng XY, Zhang HX, Mezei M, Cui M. Molecular docking: a powerful approach for structure-based drug discovery. Current Computer-Aided Drug Design, (2011); 7(2): 146-157.

Sait KHW, Mashraqi M, Khogeer AA, Alzahrani O, Anfinan NM, et al. Molecular docking analysis of HER-2 inhibitor from the ZINC database as anticancer agents. Bioinformation, (2020); 16(11): 882-887.

Sait KHW, Alam Q, Anfinan N, Al-Ghamdi O, Malik A, et al. Structure-based virtual screening and molecular docking for the identification of potential novel EGFRkinase inhibitors against ovarian cancer. Bioinformation, (2019); 15(4): 287-294.

Faheem M, Rehman MT, Danishuddin M, Khan AU. Biochemical characterization of CTX-M-15 from Enterobacter cloacae and designing a novel non-beta-lactam-beta-lactamase inhibitor. PLoS One, (2013); 8(2): e56926.

Lahiri SD, Mangani S, Durand-Reville T, Benvenuti M, De Luca F, et al. Structural insight into potent broad-spectrum inhibition with reversible recyclization mechanism: avibactam in complex with CTX-M-15 and Pseudomonas aeruginosa AmpC beta-lactamases. Antimicrobial Agents and Chemotherapy, (2013); 57(6): 2496-2505.

King DT, King AM, Lal SM, Wright GD, Strynadka NC. Molecular Mechanism of Avibactam-Mediated beta-Lactamase Inhibition. ACS Infectious Diseases, (2015); 1(4): 175-184.

Winkler ML, Papp-Wallace KM, Taracila MA, Bonomo RA. Avibactam and inhibitor-resistant SHV beta-lactamases. Antimicrobial Agents and Chemotherapy, (2015); 59(7): 3700-3709.

Larregieu CA, Benet LZ. Drug discovery and regulatory considerations for improving in-silico and in vitro predictions that use Caco-2 as a surrogate for human intestinal permeability measurements. The AAPS Journal, (2013); 15(2): 483-497.

Rossiter SE, Fletcher MH, Wuest WM. Natural Products as Platforms To Overcome Antibiotic Resistance. Chemical Reviews, (2017); 117(19): 12415-12474.

Newman DJ, Cragg GM. Natural Products as Sources of New Drugs over the Nearly Four Decades from 01/1981 to 09/2019. Journal of Natural Products, (2020); 83(3): 770-803.

Mayegowda SB, Ng M, Alghamdi S, Atwah B, Alhindi Z, et al. Role of Antimicrobial Drug in the Development of Potential Therapeutics. Evidence-Based Complementary and Alternative Medicine, (2022); 2500613.


Refbacks

  • There are currently no refbacks.