Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 303
Filter
1.
Molecules ; 29(16)2024 Aug 10.
Article in English | MEDLINE | ID: mdl-39202871

ABSTRACT

Staphylococcus aureus (S. aureus) is a major bacterial infection in humans, leading to severe disease and causing death. The stagnation of antibiotic development in recent decades has made it difficult to combat drug-resistant infections. In this study, we performed an in silico structure-based drug screening (SBDS) targeting the S. aureus MurE (saMurE) enzyme involved in cell wall synthesis of S. aureus. saMurE is an enzyme that is essential for the survival of S. aureus but not present in humans. SBDS identified nine saMurE inhibitor candidates, Compounds 1-9, from a structural library of 154,118 compounds. Among them, Compound 2 showed strong antibacterial activity against Staphylococcus epidermidis (S. epidermidis) used as a model bacterium. Amino acid sequence homology between saMurE and S. epidermidis MurE is 87.4%, suggesting that Compound 2 has a similar inhibitory effect on S. aureus. Compound 2 showed an IC50 value of 301 nM for S. epidermidis in the dose-dependent growth inhibition assay. Molecular dynamics simulation showed that Compound 2 binds stably to both S. aureus MurD and S. aureus MurF, suggesting that it is a potential multi-pharmacological pharmacological inhibitor. The structural and bioactivity information of Compound 2, as well as its potential multiple-target activity, could contribute to developing new antimicrobial agents based on MurE inhibition.


Subject(s)
Anti-Bacterial Agents , Drug Evaluation, Preclinical , Staphylococcus aureus , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Staphylococcus aureus/drug effects , Staphylococcus aureus/enzymology , Staphylococcus epidermidis/drug effects , Staphylococcus epidermidis/enzymology , Microbial Sensitivity Tests , Molecular Docking Simulation , Computer Simulation , Drug Discovery , Structure-Activity Relationship , Peptide Synthases/antagonists & inhibitors , Peptide Synthases/metabolism , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/chemistry , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Humans , Molecular Dynamics Simulation
2.
Mol Pharmacol ; 106(4): 173-187, 2024 Sep 17.
Article in English | MEDLINE | ID: mdl-39048308

ABSTRACT

Folate-dependent one-carbon (C1) metabolism encompasses distinct cytosolic and mitochondrial pathways connected by an interchange among serine, glycine, and formate. In both the cytosol and mitochondria, folates exist as polyglutamates, with polyglutamylation catalyzed by folylpolyglutamate synthetase (FPGS), including cytosolic and mitochondrial isoforms. Serine is metabolized by serine hydroxymethyltransferase (SHMT)2 in the mitochondria and generates glycine and C1 units for cellular biosynthesis in the cytosol. AGF347 is a novel pyrrolo[3,2-day]pyrimidine antifolate that targets SHMT2 in the mitochondria and SHMT1 and de novo purine biosynthesis in the cytosol. FPGS is expressed in primary pancreatic cancer specimens, and FPGS levels correlate with in vitro efficacies of AGF347 toward human pancreatic cancer cells. MIA PaCa-2 pancreatic cancer cells with CRISPR knockout of FPGS were engineered to express doxycycline-inducible FPGS exclusively in the cytosol (cFPGS) or in both the cytosol and mitochondria (mFPGS). Folate and AGF347 accumulations increased in both the cytosol and mitochondria with increased mFPGS but were restricted to the cytosol with cFPGS. AGF347-Glu5 inhibited SHMT2 ∼19-fold greater than AGF347 By metabolomics analysis, mFPGS stimulated the C1 flux from serine in the mitochondria and de novo purine and dTTP synthesis far greater than cFPGS. mFPGS enhanced in vitro inhibition of MIA PaCa-2 cell proliferation by AGF347 (∼30-fold) more than cFPGS (∼4.9-fold). Similar results were seen with other pyrrolo[3,2-d]pyrimidine antifolates (AGF291, AGF320); however, elevated mFPGS adversely impacted inhibition by the nonclassical SHMT2/SHMT1 inhibitor SHIN1. These results suggest a critical role of mFPGS levels in determining antitumor efficacies of mitochondrial-targeted pyrrolo[3,2-d]pyrimidine antifolates for pancreatic cancer. SIGNIFICANCE STATEMENT: AGF347 is a novel pyrrolo[3,2-d]pyrimidine antifolate that targets serine hydroxymethyltransferase (SHMT)2 in the mitochondria and SHMT1 and de novo purine biosynthesis in the cytosol. AGF347 accumulation increases with folylpolyglutamate synthetase (FPGS) levels in both the cytosol and mitochondria. Increased mitochondrial FPGS stimulated one-carbon metabolic fluxes in the cytosol and mitochondria and substantially enhanced in vitro inhibition of pancreatic cancer cells by AGF347. Mitochondrial FPGS levels play important roles in determining the antitumor efficacies of pyrrolo[3,2-d]pyrimidine antifolates for pancreatic cancer.


Subject(s)
Cytosol , Folic Acid Antagonists , Mitochondria , Peptide Synthases , Humans , Peptide Synthases/metabolism , Peptide Synthases/antagonists & inhibitors , Cytosol/metabolism , Mitochondria/metabolism , Mitochondria/drug effects , Folic Acid Antagonists/pharmacology , Cell Line, Tumor , Carbon/metabolism , Antineoplastic Agents/pharmacology , Glycine Hydroxymethyltransferase/metabolism , Glycine Hydroxymethyltransferase/antagonists & inhibitors , Glycine Hydroxymethyltransferase/genetics , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Folic Acid/metabolism
3.
J Biomol Struct Dyn ; 40(21): 11405-11417, 2022.
Article in English | MEDLINE | ID: mdl-34348086

ABSTRACT

Acinetobacter baumannii is an extremely dangerous multidrug-resistant (MDR) gram-negative pathogen which poses a serious life-threatening risk in immunocompromised patients. Phosphopantothenoyl cysteine synthetase (PPCS) catalyzes the formation of an amide bond between L-cysteine and phosphopantothenic acid (PPA) to form 4'- Phosphopantothenoylcysteine during Coenzyme A (CoA) biosynthesis. CoA is a crucial cofactor for cellular survival and inhibiting its synthesis will result in cell death. Bacterial PPCS differs from eukaryotic PPCS in a number of ways like it exists as a C-terminal domain of a PPCDC/PPCS fusion protein whereas eukaryotic PPCS exists as an independent protein. This difference makes it an attractive drug target. For which a conventional iterative approach of SBDD (structure-based drug design) was used, which began with three-dimensional structure prediction of AbPPCS using PHYRE 2.0. A database of FDA-approved compounds (Drug Bank) was then screened against the target of interest by means of docking score and glide energy, leading to the identification of 6 prominent drug candidates. The shortlisted 6 molecules were further subjected to all-atom MD simulation studies in explicit-solvent conditions (using AMBER force field). The MD simulation studies revealed that the ligands DB65103, DB449108 and DB443210, maintained several H-bonds with intense van der Waals contacts at the active site of the protein with high binding free energies: -11.42 kcal/mol, -10.49 kcal/mol and -10.98 kcal/mol, respectively, calculated via MM-PBSA method. Overall, binding of these compounds at the active site was found to be the most stable and robust highlighting the potential of these compounds to serve as antibacterials.Communicated by Ramaswamy H. Sarma.


Subject(s)
Acinetobacter baumannii , Peptide Synthases , Acinetobacter baumannii/drug effects , Catalytic Domain , Coenzyme A/metabolism , Ligands , Molecular Docking Simulation , Molecular Dynamics Simulation , Peptide Synthases/antagonists & inhibitors
4.
Molecules ; 26(21)2021 Oct 25.
Article in English | MEDLINE | ID: mdl-34770835

ABSTRACT

Staphylococcus aureus is an opportunistic pathogen that can cause fatal bacterial infections. MurD catalyzes the formation of peptide bond between UDP-N-acetylehyl-l-alanine and d-glutamic acid, which plays an important role in the synthesis of peptidoglycan and the formation of cell wall by S. aureus. Because S. aureus is resistant to most existing antibiotics, it is necessary to develop new inhibitors. In this study, Schrodinger 11.5 Prime homology modeling was selected to prepare the protein model of MurD enzyme, and its structure was optimized. We used a virtual screening program and similarity screening to screen 47163 compounds from three marine natural product libraries to explore new inhibitors of S. aureus. ADME provides analysis of the physicochemical properties of the best performing compounds during the screening process. To determine the stability of the docking effect, a 100 ns molecular dynamics was performed to verify how tightly the compound was bound to the protein. By docking analysis and molecular dynamics analysis, both 46604 and 46608 have strong interaction with the docking pocket, have good pharmacological properties, and maintain stable conformation with the target protein, so they have a chance to become drugs for S. aureus. Through virtual screening, similarity screening, ADME study and molecular dynamics simulation, 46604 and 46608 were selected as potential drug candidates for S. aureus.


Subject(s)
Anti-Bacterial Agents/pharmacology , Aquatic Organisms/chemistry , Biological Products/pharmacology , Enzyme Inhibitors/pharmacology , Peptide Synthases/antagonists & inhibitors , Staphylococcus aureus/drug effects , Staphylococcus aureus/enzymology , Amino Acid Sequence , Anti-Bacterial Agents/chemistry , Biological Products/chemistry , Chemical Phenomena , Drug Discovery , Drug Evaluation, Preclinical , Enzyme Inhibitors/chemistry , Molecular Docking Simulation , Molecular Dynamics Simulation , Peptide Synthases/chemistry , Small Molecule Libraries , Structure-Activity Relationship
5.
ACS Chem Biol ; 16(11): 2401-2414, 2021 11 19.
Article in English | MEDLINE | ID: mdl-34582681

ABSTRACT

The pantothenate analogue hopantenate (HoPan) is widely used as a modulator of coenzyme A (CoA) levels in cell biology and disease models─especially for pantothenate kinase associated neurodegeneration (PKAN), a genetic disease rooted in impaired CoA metabolism. This use of HoPan was based on reports that it inhibits pantothenate kinase (PanK), the first enzyme of CoA biosynthesis. Using a combination of in vitro enzyme kinetic studies, crystal structure analysis, and experiments in a typical PKAN cell biology model, we demonstrate that instead of inhibiting PanK, HoPan relies on it for metabolic activation. Once phosphorylated, HoPan inhibits the next enzyme in the CoA pathway─phosphopantothenoylcysteine synthetase (PPCS)─through formation of a nonproductive substrate complex. Moreover, the obtained structure of the human PPCS in complex with the inhibitor and activating nucleotide analogue provides new insights into the catalytic mechanism of PPCS enzymes─including the elusive binding mode for cysteine─and reveals the functional implications of mutations in the human PPCS that have been linked to severe dilated cardiomyopathy. Taken together, this study demonstrates that the molecular mechanism of action of HoPan is more complex than previously thought, suggesting that the results of studies in which it is used as a tool compound must be interpreted with care. Moreover, our findings provide a clear framework for evaluating the various factors that contribute to the potency of CoA-directed inhibitors, one that will prove useful in the future rational development of potential therapies of both human genetic and infectious diseases.


Subject(s)
Coenzyme A/metabolism , Enzyme Inhibitors/pharmacology , Pantothenic Acid/analogs & derivatives , Peptide Synthases/antagonists & inhibitors , gamma-Aminobutyric Acid/analogs & derivatives , Amino Acid Sequence , Amino Acid Substitution , Animals , Cells, Cultured , Crystallization , Drosophila melanogaster , Kinetics , Molecular Conformation , Pantothenic Acid/pharmacology , Peptide Synthases/metabolism , Substrate Specificity , gamma-Aminobutyric Acid/pharmacology
6.
Angew Chem Int Ed Engl ; 60(41): 22207-22211, 2021 10 04.
Article in English | MEDLINE | ID: mdl-34396662

ABSTRACT

Peptidyl asparaginyl ligases (PALs) are powerful tools for peptide macrocyclization. Herein, we report that a derivative of Asn, namely Nγ -hydroxyasparagine or Asn(OH), is an unnatural P1 substrate of PALs. By Asn(OH)-mediated cyclization, we prepared cyclic peptides as new matrix metalloproteinase 2 (MMP2) inhibitors displaying the hydroxamic acid moiety of Asn(OH) as the key pharmacophore. The most potent cyclic peptide (Ki =2.8±0.5 nM) was built on the hyperstable tetracyclic scaffold of rhesus theta defensin-1. The Asn(OH) residue in the cyclized peptides can also be readily oxidized to Asp. By this approach, we synthesized several bioactive Asp-containing cyclic peptides (MCoTI-II, kB2, SFTI, and integrin-targeting RGD peptides) that are otherwise difficult targets for PAL-catalyzed cyclization owing to unfavorable kinetics of the P1-Asp substrates. This study demonstrates that substrate engineering is a useful strategy to expand the application of PAL ligation in the synthesis of therapeutic cyclic peptides.


Subject(s)
Amino Acids/pharmacology , Asparagine/pharmacology , Enzyme Inhibitors/pharmacology , Peptide Synthases/antagonists & inhibitors , Peptides, Cyclic/pharmacology , Amino Acids/chemistry , Asparagine/chemistry , Enzyme Inhibitors/chemistry , Peptide Synthases/metabolism , Peptides, Cyclic/chemistry , Substrate Specificity
7.
Eur J Med Chem ; 222: 113568, 2021 Oct 15.
Article in English | MEDLINE | ID: mdl-34118719

ABSTRACT

TUBERCULOSIS: (TB) transmitted by Mycobacterium tuberculosis (Mtb) is one of the top 10 causes of death globally. Currently, the widespread occurrence of resistance toward Mtb strains is becoming a significant concern to public health. This scenario exaggerated the need for the discovery of novel targets and their inhibitors. Targeting the "Mtb cell wall peptidoglycan synthesis" is an attractive strategy to overcome drug resistance. Mur enzymes (MurA-MurF) play essential roles in the peptidoglycan synthesis by catalyzing the ligation of key amino acid residues to the stem peptide. These enzymes are unique and confined to the eubacteria and are absent in humans, representing potential targets for anti-tubercular drug discovery. Mtb Mur ligases with the same catalytic mechanism share conserved amino acid regions and structural features that can conceivably exploit for the designing of the inhibitors, which can simultaneously target more than one isoforms (MurC-MurF) of the enzyme. In light of these findings in the current review, we have discussed the recent advances in medicinal chemistry of Mtb Mur enzymes (MurA-MurF) and their inhibitors, offering attractive multi-targeted strategies to combat the problem of drug-resistant in M. tuberculosis.


Subject(s)
Antitubercular Agents/pharmacology , Enzyme Inhibitors/pharmacology , Mycobacterium tuberculosis/enzymology , Peptide Synthases/antagonists & inhibitors , Peptidoglycan/pharmacology , Antitubercular Agents/chemical synthesis , Antitubercular Agents/chemistry , Cell Wall/drug effects , Cell Wall/metabolism , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Molecular Structure , Mycobacterium tuberculosis/cytology , Peptide Synthases/metabolism , Peptidoglycan/chemistry
8.
ACS Infect Dis ; 7(6): 1666-1679, 2021 06 11.
Article in English | MEDLINE | ID: mdl-33939919

ABSTRACT

Coenzyme A (CoA) is a ubiquitous cofactor present in all living cells and estimated to be required for up to 9% of intracellular enzymatic reactions. Mycobacterium tuberculosis (Mtb) relies on its own ability to biosynthesize CoA to meet the needs of the myriad enzymatic reactions that depend on this cofactor for activity. As such, the pathway to CoA biosynthesis is recognized as a potential source of novel tuberculosis drug targets. In prior work, we genetically validated CoaBC as a bactericidal drug target in Mtb in vitro and in vivo. Here, we describe the identification of compound 1f, a small molecule inhibitor of the 4'-phosphopantothenoyl-l-cysteine synthetase (PPCS; CoaB) domain of the bifunctional Mtb CoaBC, and show that this compound displays on-target activity in Mtb. Compound 1f was found to inhibit CoaBC uncompetitively with respect to 4'-phosphopantothenate, the substrate for the CoaB-catalyzed reaction. Furthermore, metabolomic profiling of wild-type Mtb H37Rv following exposure to compound 1f produced a signature consistent with perturbations in pantothenate and CoA biosynthesis. As the first report of a direct small molecule inhibitor of Mtb CoaBC displaying target-selective whole-cell activity, this study confirms the druggability of CoaBC and chemically validates this target.


Subject(s)
Mycobacterium tuberculosis , Peptide Synthases/antagonists & inhibitors , Coenzyme A , Cysteine/analogs & derivatives , Mycobacterium tuberculosis/enzymology , Mycobacterium tuberculosis/genetics , Pantothenic Acid/analogs & derivatives , Peptide Synthases/genetics
9.
Bioorg Med Chem Lett ; 40: 127966, 2021 05 15.
Article in English | MEDLINE | ID: mdl-33744441

ABSTRACT

Antibiotic resistance represents one of the biggest public health challenges in the last few years. Mur ligases (MurC-MurF) are involved in the synthesis of UDP-N-acetylmuramyl-pentapeptide, the main building block of bacterial peptidoglycan polymer. They are essential for the survival of bacteria and therefore important antibacterial targets. We report herein the synthesis and structure-activity relationships of Mur ligases inhibitors with an azastilbene scaffold. Several compounds showed promising inhibitory potencies against multiple ligases and one compound also possessed moderate antibacterial activity. These results represent a solid ground for further development and optimization of structurally novel antimicrobial agents to combat the rising bacterial resistance.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacterial Proteins/antagonists & inhibitors , Benzylidene Compounds/pharmacology , Enzyme Inhibitors/pharmacology , Peptide Synthases/antagonists & inhibitors , Pyridines/pharmacology , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/metabolism , Bacterial Proteins/metabolism , Benzylidene Compounds/chemical synthesis , Benzylidene Compounds/metabolism , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/metabolism , Molecular Docking Simulation , Molecular Structure , Peptide Synthases/metabolism , Protein Binding , Pyridines/chemical synthesis , Pyridines/metabolism , Staphylococcus aureus/drug effects , Staphylococcus aureus/enzymology , Structure-Activity Relationship
10.
Chem Pharm Bull (Tokyo) ; 69(2): 222-225, 2021.
Article in English | MEDLINE | ID: mdl-33518604

ABSTRACT

The gatekeeping adenylation (A) domain of the non-ribosomal peptide synthetase (NRPS) selectively incorporates specific proteinogenic/non-proteinogenic amino acid into a growing peptide chain. The EntE of the enterobactin NRPS is a discrete aryl acid A-domain with 2,3-dihydroxybenzoic acid (DHB) substrate specificity. Reprogrammed EntE N235G variant possesses an enlarged substrate recognition site, and is capable of accepting non-native aryl acids. Biochemical characterization of this unique substrate recognition site should provide a better understanding of activi-site microenvironments. Here, we synthesized a non-hydrolysable adenylate analogue with 2-aminobenzoic acid (2-ABA), 3-aminobenzoic acid (3-ABA), and 4-aminobenzoic acid (4-ABA) and used them to calculate the apparent inhibition constants (Kiapp.). Dose-response experiments using 3-ABA-sulfamoyladenosine (AMS) provided Kiapp. values of 596 nM for wild-type EntE and 2.4 nM for the N235G variants. These results suggest that 3-amino group of benzoic acid plays an important role in substrate recognition by the N235G variant. These findings would help designing aryl acid substrates with substituents at the 2- and 3-positions.


Subject(s)
Molecular Dynamics Simulation , Peptide Synthases/metabolism , 4-Aminobenzoic Acid/chemistry , 4-Aminobenzoic Acid/metabolism , Binding Sites , Enterobactin/chemistry , Enterobactin/metabolism , Kinetics , Mutagenesis, Site-Directed , Peptide Synthases/antagonists & inhibitors , Peptide Synthases/genetics , Protein Binding , Protein Domains , Substrate Specificity
11.
Nat Commun ; 12(1): 143, 2021 01 08.
Article in English | MEDLINE | ID: mdl-33420031

ABSTRACT

Coenzyme A (CoA) is a fundamental co-factor for all life, involved in numerous metabolic pathways and cellular processes, and its biosynthetic pathway has raised substantial interest as a drug target against multiple pathogens including Mycobacterium tuberculosis. The biosynthesis of CoA is performed in five steps, with the second and third steps being catalysed in the vast majority of prokaryotes, including M. tuberculosis, by a single bifunctional protein, CoaBC. Depletion of CoaBC was found to be bactericidal in M. tuberculosis. Here we report the first structure of a full-length CoaBC, from the model organism Mycobacterium smegmatis, describe how it is organised as a dodecamer and regulated by CoA thioesters. A high-throughput biochemical screen focusing on CoaB identified two inhibitors with different chemical scaffolds. Hit expansion led to the discovery of potent and selective inhibitors of M. tuberculosis CoaB, which we show to bind to a cryptic allosteric site within CoaB.


Subject(s)
Antitubercular Agents/pharmacology , Bacterial Proteins/antagonists & inhibitors , Carboxy-Lyases/antagonists & inhibitors , Mycobacterium smegmatis/enzymology , Mycobacterium tuberculosis/drug effects , Peptide Synthases/antagonists & inhibitors , Allosteric Regulation/drug effects , Allosteric Site/drug effects , Antitubercular Agents/therapeutic use , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Bacterial Proteins/ultrastructure , Carboxy-Lyases/genetics , Carboxy-Lyases/metabolism , Carboxy-Lyases/ultrastructure , Coenzyme A/biosynthesis , Crystallography, X-Ray , Enzyme Assays , Gene Knockdown Techniques , High-Throughput Screening Assays , Humans , Microbial Sensitivity Tests , Mycobacterium tuberculosis/enzymology , Mycobacterium tuberculosis/genetics , Peptide Synthases/genetics , Peptide Synthases/metabolism , Peptide Synthases/ultrastructure , Tuberculosis/drug therapy , Tuberculosis/microbiology
12.
Mini Rev Med Chem ; 21(18): 2778-2787, 2021.
Article in English | MEDLINE | ID: mdl-32634082

ABSTRACT

INTRODUCTION: Brucella melitensis is a facultative intracellular bacterial pathogen that causes abortion in goats and sheep and Malta fever in humans. In humans, chronic infection occurs through contact with infected animals or their waste products. METHODS: The subtractive genomic approach is considered as a powerful and useful method for the identification of potential drug and vaccine targets. In this study, an attempt has been made through a subtractive proteomic strategy to identify novel drug targets in Brucella melitensis strains. Total 2604 core proteins of 56 strains of B. melitensis were taken, of which 545 non-human homologs were found to be essential for pathogen growth. Metabolic pathway analysis of these essential proteins revealed that 129 proteins are exclusively involved in 21 unique metabolic pathways in B. melitensis reference strain. RESULTS: Of these, 31 proteins were found to be involved in 10 metabolic pathways that are unique to the pathogen. We selected Nitrate reductase subunit-ß, Urease subunit α-2, Pantoate-ß-alanine ligase, Isochorismatase, 2-dehydro-3-deoxyphosphooctonate aldolase and Serine O-acetyltransferase as drug targets in Brucella melitensis strains. Among these druggable targets, we selected only Pantoate-ß- alanine ligase as high confidence target based on intensive literature curation, which is nonhomologous to the human gut metagenome involved in biosynthesis of secondary metabolites pathway. Pantothenate synthetase is the best chemotherapeutic target to combat Brucellulosis. CONCLUSION: Furthermore, in vitro and in vivo validation is needed for the evaluation of lead compounds against Brucella melitensis strains.


Subject(s)
Bacterial Proteins/antagonists & inhibitors , Brucella melitensis/drug effects , Brucella melitensis/genetics , Drug Discovery , Genome, Bacterial , Proteome , Proteomics , Animals , Bacterial Proteins/genetics , Brucella melitensis/classification , Brucella melitensis/enzymology , Brucellosis/microbiology , Brucellosis/veterinary , Humans , Molecular Targeted Therapy , Peptide Synthases/antagonists & inhibitors , Peptide Synthases/genetics
13.
J Mol Model ; 26(11): 304, 2020 Oct 17.
Article in English | MEDLINE | ID: mdl-33068184

ABSTRACT

Acinetobacter baumannii, an opportunistic bacterium of the multidrug-resistant (MDR) ESKAPE family of pathogens, is responsible for 2-10% infections associated with all gram-negative bacteria. The hospital-acquired nosocomial infections caused by A.baumannii include deadly diseases like ventilator-associated pneumonia, bacteremia, septicemia and urinary tract infections (UTI). Over the last 3 years, it has evolved into multiple strains demonstrating high antibiotic resistance against a wide array of antibiotics. Hence, it becomes imperative to identify novel drug-like molecules to treat such infections effectively. UDP-N-acetylmuramoyl-L-alanine-D-glutamate ligase (MurD) is an essential enzyme of the Mur family which is responsible for peptidoglycan biosynthesis, making it a unique and ideal drug target. Initially, a homology modelling approach was employed to predict the three-dimensional model of MurD from A. baumannii using MurD from Escherichia coli (PDB ID: 4UAG) as a suitable structural template. Subsequently, an optimised model of MurD was subjected to virtual high-throughput screening (vHTS) against a ZINC library of ~ 642,759 commercially available molecules to identify promising lead compounds demonstrating high binding affinities towards it. From the screening process, four promising molecules were identified based on the estimated binding affinities (ΔG), estimated inhibition constants (Ki), catalytic residue interactions and drug-like properties, which were then subjected to molecular dynamics (MD) simulation studies to reflect the physiological state of protein molecules in vivo equivalently. The binding free energies of the selected MurD-ligand complexes were also calculated using MM/PBSA (molecular mechanics with Poisson-Boltzmann and surface area solvation) approach. Finally, the global dynamics along with binding free energy analysis suggested that ZINC19221101 (ΔG = - 62.6 ± 5.6 kcal/mol) and ZINC12454357 (ΔG = - 46.1 ± 2.6 kcal/mol) could act as most promising candidates for inhibiting the function of MurD ligase and aid in drug discovery and development against A.baumannii. Graphical abstract.


Subject(s)
Acinetobacter baumannii/enzymology , Drug Evaluation, Preclinical , Enzyme Inhibitors/analysis , Enzyme Inhibitors/pharmacology , Molecular Dynamics Simulation , Peptide Synthases/antagonists & inhibitors , Peptide Synthases/chemistry , Amino Acid Sequence , Enzyme Inhibitors/chemistry , Hydrogen Bonding , Ligands , Molecular Docking Simulation , Protein Domains , Sequence Homology, Amino Acid , Thermodynamics
14.
Molecules ; 25(21)2020 Oct 26.
Article in English | MEDLINE | ID: mdl-33114668

ABSTRACT

The synthesis of hitherto unknown 5'-deoxy-5'-(4-substituted-1,2,3-triazol-1-yl)-uridine and its evaluation, through an one-pot screening assay, against MurA-F enzymes involved in Mycobacterium tuberculosis (Mtb), are described. Starting from UDP-N-acetylmuramic acid (UDP-MurNAc), the natural substrate involved in the peptidoglycan biosynthesis, our strategy was to substitute the diphosphate group of UDP-MurNAc by a 1,2,3-triazolo spacer under copper-catalyzed azide-alkyne cycloaddition conditions. The structure-activity relationship was discussed and among the 23 novel compounds developed, N-acetylglucosamine analogues 11c and 11e emerged as the best inhibitors against the Mtb MurA-F enzymes reconstruction pathway with an inhibitory effect of 56% and 50%, respectively, at 100 µM. Both compounds are selective inhibitors of Mtb MurE, the molecular docking and molecular dynamic simulation suggesting that 11c and 11e are occupying the active site of Mtb MurE ligase.


Subject(s)
Drug Design , Mycobacterium tuberculosis/enzymology , Peptide Synthases/antagonists & inhibitors , Triazoles/chemistry , Uridine/chemical synthesis , Uridine/pharmacology , Catalytic Domain , Chemistry Techniques, Synthetic , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Molecular Docking Simulation , Peptide Synthases/chemistry , Peptide Synthases/metabolism , Uridine/chemistry
15.
Life Sci ; 262: 118466, 2020 Dec 01.
Article in English | MEDLINE | ID: mdl-32961233

ABSTRACT

INTRODUCTION: UDP-N-acetylmuramic-alanine ligase (MurC) is an enzyme catalyzing the addition of L-alanine to UDP-acetylmuramoyl nucleotide precursor in Mycobacterium tuberculosis (M. tuberculosis). This enzyme is a prerequisite for the biosynthesis of the peptidoglycans in M. tuberculosis. AIM: This study aimed to identify the novel inhibitors of MurC using in silico approach. MATERIALS AND METHODS: The three dimensional (3D) structure of MurC was determined using comparative modeling and based on the template obtained from Haemophilus influenza (1P31). The structural analysis of the model structure shown that three residues (Lys126, Glu170, and Glu358) are critical for in the catalytic activity of the enzyme, and their inhibition will block the function of the enzyme. Ten thousand and ninety-five (10095) compounds obtained through virtual screening against Zinc and PubChem databases based on their ability to bind to MurC with minimum binding energies. These ligands screened for the physicochemical properties, molecular docking, and pharmacokinetic analyses. FINDING: Six compounds had desirable physicochemical and pharmacokinetic properties with excellent binding energy ranged between -12.27 and -10.09 kcal/mol. These compounds subjected to Molecular Dynamic (MD) Simulation and Molecular Mechanics Generalized Born Surface Area (MM-GBSA) analyses. The outcome of the analysis revealed that four ligands (PubChem1548994, ZINC11882115, ZINC22241774, and ZINC12330603) formed a stable conformation in the substrate-binding site of the protein during the 50 ns MD simulation. CONCLUSION: Therefore, the ligands mentioned above might regard as novel inhibitors of M. tuberculosis which requires further in vitro and in vivo validation.


Subject(s)
Antitubercular Agents/pharmacology , Molecular Dynamics Simulation , Mycobacterium tuberculosis/drug effects , Peptide Synthases/antagonists & inhibitors , Antitubercular Agents/chemistry , Antitubercular Agents/pharmacokinetics , Binding Sites , Ligands , Molecular Docking Simulation , Mycobacterium tuberculosis/enzymology , Peptide Synthases/chemistry , Protein Binding
16.
Eur J Med Chem ; 200: 112444, 2020 Aug 15.
Article in English | MEDLINE | ID: mdl-32497961

ABSTRACT

d-Alanyl-d-alanine ligase (Ddl) is a validated and attractive target among the bacterial enzymes involved in peptidoglycan biosynthesis. In the present work, we investigated the pharmacomodulations of the benzoylthiosemicarbazide scaffold to identify new Ddl inhibitors with antibacterial potency. Five novel series of thiosemicarbazide analogues, 1,2,4-thiotriazole-3-thiones, 1,3,4-thiadiazoles, phenylthiosemicarbazones, diacylthiosemicarbazides and thioureas were synthesized via straightforward procedures, then tested against Ddl and on susceptible or resistant bacterial strains. Among these, the thiosemicarbazone and thiotriazole were identified as the most promising scaffolds with Ddl inhibition potency in the micromolar range. Antimicrobial evaluation of salicylaldehyde-4(N)-(3,4-dichlorophenyl) thiosemicarbazone 33, one of the best compounds in our study, revealed interesting antimicrobial activities with values of 3.12-6.25 µM (1.06-2.12 µg/mL) against VRE strains and 12.5-25.0 µM (4.25-8.50 µg/mL) towards MRSA and VRSA strains. A detailed mechanistic study was conducted on the Ddl inhibitors 4-(3,4-dichlorophenyl)-5-(2-hydroxyphenyl)-2,4-dihydro-3H-1,2,4-triazole-3-thione 20 and compound 33, and revealed a bactericidal effect at 5 × MIC concentration after 7 h and 24 h, respectively, and a bacteriostatic effect at 1 × MIC or 2 × MIC without any sign of bacterial membrane disruption at these lower concentrations. Finally, 20 and 33 were proved to target Ddl in bacterio via intracellular LC-MS dosage of d-Ala, l-Ala and d-Ala-d-Ala. Although, at this stage, our results indicate that other mechanisms might be involved to explain the antimicrobial potency of our compounds, their ability to inhibit the growth of strains resistant to usual antibiotics, as well as strains that express alternative ligases, sets the stage for the development of new antimicrobial agents potentially less sensitive to resistance mechanisms.


Subject(s)
Anti-Bacterial Agents/pharmacology , Enzyme Inhibitors/pharmacology , Escherichia/drug effects , Peptide Synthases/antagonists & inhibitors , Staphylococcus aureus/drug effects , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/chemistry , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Microbial Sensitivity Tests , Molecular Structure , Peptide Synthases/metabolism , Staphylococcus aureus/enzymology , Structure-Activity Relationship
17.
SLAS Discov ; 25(1): 70-78, 2020 01.
Article in English | MEDLINE | ID: mdl-31597510

ABSTRACT

The rapid rise in the emergence of multidrug-resistant (MDR) and extensively drug-resistant (XDR) strains of Mycobacterium tuberculosis (Mtb) mandates the discovery of novel tuberculosis (TB) drugs. Mur enzymes, which are identified as essential proteins in Mtb and catalyze the cytoplasmic steps in the peptidoglycan biosynthetic pathway, are considered potential drug targets. However, none of the clinical drugs have yet been developed against these enzymes. Hence, the aim of this study was to identify novel inhibitors of Mur enzymes in Mycobacterium tuberculosis. We screened an antitubercular compound library of 684 compounds, using MurB and MurE enzymes of the Mtb Mur pathway as drug targets. For experimental validation, the top hits obtained on in silico screening were screened in vitro, using Mtb Mur enzyme-specific assays. In all, seven compounds were found to show greater than 50% inhibition, with the highest inhibition observed at 77%, and the IC50 for these compounds was found to be in the range of 28-50 µM. Compound 5175112 showed the lowest IC50 (28.69 ± 1.17 µM), and on the basis of (1) the binding affinity, (2) the stability of interaction noted on molecular dynamics simulation, and (3) an in vitro assay, MurE appeared to be its target enzyme. We believe that the overall strategy followed in this study and the results obtained are a good starting point for developing Mur enzyme-specific Mtb inhibitors.


Subject(s)
Antitubercular Agents/pharmacology , Bacterial Proteins/antagonists & inhibitors , Drug Evaluation, Preclinical , Mycobacterium tuberculosis/drug effects , Mycobacterium tuberculosis/enzymology , Peptide Synthases/antagonists & inhibitors , Small Molecule Libraries , Antitubercular Agents/chemistry , Binding Sites , Drug Evaluation, Preclinical/methods , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Humans , Molecular Conformation , Molecular Docking Simulation , Molecular Dynamics Simulation , Protein Binding
18.
Biochem J ; 476(21): 3125-3139, 2019 11 15.
Article in English | MEDLINE | ID: mdl-31488574

ABSTRACT

CoaBC, part of the vital coenzyme A biosynthetic pathway in bacteria, has recently been validated as a promising antimicrobial target. In this work, we employed native ion mobility-mass spectrometry to gain structural insights into the phosphopantothenoylcysteine synthetase domain of E. coli CoaBC. Moreover, native mass spectrometry was validated as a screening tool to identify novel inhibitors of this enzyme, highlighting the utility and versatility of this technique both for structural biology and for drug discovery.


Subject(s)
Carboxy-Lyases/chemistry , Drug Evaluation, Preclinical/methods , Escherichia coli Proteins/chemistry , Escherichia coli/enzymology , Mass Spectrometry/methods , Multienzyme Complexes/chemistry , Peptide Synthases/chemistry , Carboxy-Lyases/antagonists & inhibitors , Carboxy-Lyases/metabolism , Dimerization , Enzyme Inhibitors/chemistry , Escherichia coli/chemistry , Escherichia coli/drug effects , Escherichia coli/genetics , Escherichia coli Proteins/antagonists & inhibitors , Escherichia coli Proteins/metabolism , Kinetics , Multienzyme Complexes/antagonists & inhibitors , Multienzyme Complexes/metabolism , Peptide Synthases/antagonists & inhibitors , Peptide Synthases/metabolism , Protein Domains
19.
J Recept Signal Transduct Res ; 39(3): 283-293, 2019 Jun.
Article in English | MEDLINE | ID: mdl-31538846

ABSTRACT

A new series of benzothiazol-2-ylcarbamodithioate functional compounds 5a-f has been designed, synthesized and characterized by spectral data. These compounds were screened for their in vitro antibacterial activity against strains of Staphylococcus aureus (NCIM 5021, NCIM 5022 and methicillin-resistant isolate 43300), Bacillus subtilis (NCIM 2545), Escherichia coli (NCIM 2567), Klebsiella pneumoniae (NCIM 2706) and Psudomonas aeruginosa (NCIM 2036). Compounds 5a and 5d exhibited significant activity against all the tested bacterial strains. Specifically, compounds 5a and 5d showed potent activity against K. pneumoniae (NCIM 2706), while compound 5a also displayed potent activity against S. aureus (NCIM 5021). Compound 5d showed minimum IC50 value of 13.37 µM against S. aureus MurD enzyme. Further, the binding interactions of compounds 5a-f in the catalytic pocket have been investigated using the extra-precision molecular docking and binding free energy calculation by MM-GBSA approach. A 30 ns molecular dynamics simulation of 5d/modeled S. aureus MurD enzyme was performed to determine the stability of the predicted binding conformation.


Subject(s)
Benzothiazoles/chemical synthesis , Benzothiazoles/pharmacology , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , Molecular Docking Simulation , Molecular Dynamics Simulation , Peptide Synthases/antagonists & inhibitors , Staphylococcus aureus/enzymology , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacokinetics , Anti-Bacterial Agents/pharmacology , Benzothiazoles/chemistry , Benzothiazoles/pharmacokinetics , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacokinetics , Inhibitory Concentration 50 , Microbial Sensitivity Tests , Peptide Synthases/metabolism , Staphylococcus aureus/drug effects , Thermodynamics
20.
Med Hypotheses ; 131: 109305, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31443754

ABSTRACT

Infections due to resistant bacteria are the life-threatening and leading cause of mortality worldwide. The current therapy for bacterial infections includes treatment with various drugs and antibiotics. The misuse and over usage of these antibiotics leads to bacterial resistance. There are several mechanisms by which bacteria exhibit resistance to some antibiotics. These include drug inactivation or modification, elimination of antibiotics through efflux pumps, drug target alteration, and modification of metabolic pathway. However, it is difficult to treat infections caused by resistant bacteria by conventional existing therapy. In the present study binding affinities of some glitazones against ParE and MurE bacterial enzymes are investigated by in silico methods. As evident by extra-precision docking and binding free energy calculation (MM-GBSA) results, rivoglitazone exhibited higher binding affinity against both ParE and MurE enzymes compared to all other selected compounds. Further molecular dynamic (MD) simulations were performed to validate the stability of rivoglitazone/4MOT and rivoglitazone/4C13 complexes and to get insight into the binding mode of inhibitor. Thus, we hypothesize that structural modifications of the rivoglitazone scaffold can be useful for the development of an effective antibacterial agent.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacterial Infections/drug therapy , DNA Topoisomerase IV/antagonists & inhibitors , Peptide Synthases/antagonists & inhibitors , Thiazolidinediones/pharmacology , Thiazolidines/pharmacology , Anti-Bacterial Agents/chemistry , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/chemistry , DNA Topoisomerase IV/chemistry , Drug Resistance, Microbial , Humans , Hydrogen Bonding , Molecular Docking Simulation , Molecular Dynamics Simulation , Peptide Synthases/chemistry , Structure-Activity Relationship , Thiazolidinediones/chemistry , Thiazolidines/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL