9 research outputs found

    Evaluation of quercetin as a potential β-lactamase CTX-M-15 inhibitor via the molecular docking, dynamics simulations, and MMGBSA

    Get PDF
    Antimicrobial resistance (AMR) threatens millions of people around the world and has been declared a global risk by the World Economic Forum. One of the important AMR mechanisms in Enterobacteriaceae is the production of extended-spectrum β-lactamases. The most common ESBL, CTX-M β-lactamases, is spread to the world by CTX-M-15 and CTX-M-14. Sulbactam, clavulanic acid, and tazobactam are first-generation β-lactamase inhibitors and avibactam is a new non-β-lactam β-lactamase inhibitor. We studied that avibactam, sulbactam, clavulanic acid, tazobactam, and quercetin natural flavonoids were docked to target protein CTXM-15. Subsequently, the complexes were simulated using the molecular dynamics simulations method during 100 ns for determining the final binding positions of ligands. Clavulanic acid left CTX-M-15 and other ligands remained in the binding site after the simulation. The estimated binding energies were calculated during 100 ns simulation by the MMGBSA-MMPBSA method. The estimated free binding energies of avibactam, sulbactam, quercetin, tazobactam, and clavulanic acid were sorted as –33.61 kcal/mol, –16.04 kcal/mol, –14 kcal/mol, –12.68 kcal/mol, and –2.95 kcal/mol. As a result of both final binding positions and free binding energy calculations, Quercetin may be evaluated an alternative candidate and a more potent β-lactamases inhibitor for new antimicrobial combinations to CTX-M-15. The results obtained in silico studies are predicted to be a preliminary study for in vitro studies for quercetin and similar bioactive natural compounds. These studies are notable for the discovery of natural compounds that can be used in the treatment of infections caused by β-lactamase-producing pathogens

    Targeting SARS-CoV-2 Nsp12/Nsp8 interactioninterface with approved and investigational drugs:an in silico structure-based approach

    Get PDF
    In this study, the Nsp12–Nsp8 complex of SARS-CoV-2 was targeted with structure-based and com-puter-aided drug design approach because of its vital role in viral replication. Sequence analysis ofRNA-dependent RNA polymerase (Nsp12) sequences from 30,366 different isolates were analysed forpossible mutations. FDA-approved and investigational drugs were screened for interaction with bothmutant and wild-type Nsp12–Nsp8 interfaces. Sequence analysis revealed that 70.42% of Nsp12sequences showed conserved P323L mutation, located in the Nsp8 binding cleft. Compounds werescreened for interface interaction, any with XP GScores lower than 7.0kcal/mol were considered aspossible interface inhibitors. RX-3117 (fluorocyclopentenyl cytosine) and Nebivolol had the highestbinding affinities in both mutant and wild-type enzymes, therefore they were selected and resultantprotein–ligand complexes were simulated for analysis of stability over 100ns. Although the selectedligands had partial mobility in the binding cavity, they were not removed from the binding pocketafter 100ns. The ligand RX-3117 remained in the same position in the binding pocket of the mutantand wild-type enzyme after 100ns MD simulation. However, the ligand Nebivolol folded andembedded in the binding pocket of mutant Nsp12 protein. Overall, FDA-approved and investigationaldrugs are able to bind to the Nsp12–Nsp8 interaction interface and prevent the formation of theNsp12–Nsp8 complex. Interruption of viral replication by drugs proposed in this study should be fur-ther tested to pave the way forin vivostudies towards the treatment of COVID-19

    Evaluation of the potency of FDA-approved drugs on wild type and mutant SARS-CoV-2 helicase (Nsp13)

    Get PDF
    SARS-CoV-2 has caused COVID-19 outbreak with nearly 2 M infected people and over 100K death worldwide, until middle of April 2020. There is no confirmed drug for the treatment of COVID-19 yet. As the disease spread fast and threaten human life, repositioning of FDA approved drugs may provide fast options for treatment. In this aspect, structure-based drug design could be applied as a powerful approach in distinguishing the viral drug target regions from the host. Evaluation of variations in SARS-CoV-2 genome may ease finding specific drug targets in the viral genome. In this study, 3458 SARS-CoV-2 genome sequences isolated from all around the world were analyzed. Incidence of C17747T and A17858G mutations were observed to be much higher than others and they were on Nsp13, a vital enzyme of SARS-CoV-2. Effect of these mutations was evaluated on protein-drug interactions using in silico methods. The most potent drugs were found to interact with the key and neighbor residues of the active site responsible from ATP hydrolysis. As result, cangrelor, fludarabine, folic acid and polydatin were determined to be the most potent drugs which have potency to inhibit both the wild type and mutant SARS-CoV-2 helicase. Clinical data supporting these findings would be important towards overcoming COVID-19

    Targeting SARS-CoV-2 Nsp12/Nsp8 interaction interface with approved and investigational drugs: anin silicostructure-based approach

    Get PDF
    In this study, the Nsp12-Nsp8 complex of SARS-CoV-2 was targeted with structure-based and computer-aided drug design approach because of its vital role in viral replication. Sequence analysis of RNA-dependent RNA polymerase (Nsp12) sequences from 30,366 different isolates were analysed for possible mutations. FDA-approved and investigational drugs were screened for interaction with both mutant and wild-type Nsp12-Nsp8 interfaces. Sequence analysis revealed that 70.42% of Nsp12 sequences showed conserved P323L mutation, located in the Nsp8 binding cleft. Compounds were screened for interface interaction, any with XP GScores lower than -7.0 kcal/mol were considered as possible interface inhibitors. RX-3117 (fluorocyclopentenyl cytosine) and Nebivolol had the highest binding affinities in both mutant and wild-type enzymes, therefore they were selected and resultant protein-ligand complexes were simulated for analysis of stability over 100 ns. Although the selected ligands had partial mobility in the binding cavity, they were not removed from the binding pocket after 100 ns. The ligand RX-3117 remained in the same position in the binding pocket of the mutant and wild-type enzyme after 100 ns MD simulation. However, the ligand Nebivolol folded and embedded in the binding pocket of mutant Nsp12 protein. Overall, FDA-approved and investigational drugs are able to bind to the Nsp12-Nsp8 interaction interface and prevent the formation of the Nsp12-Nsp8 complex. Interruption of viral replication by drugs proposed in this study should be further tested to pave the way forin vivostudies towards the treatment of COVID-19

    The role of Acinetobacter baumannii CarO outer membrane protein in carbapenems influx

    No full text
    The gram-negative strain Acinetobacter baumannii is a cocobacillus, non-motile and aerobic organism that is often found in nosocomial infections. Many institutions worldwide such as WHO are grappling with antibiotic resistance Acinetobacter baumannii. Therefore, in recent years, there have been many studies in the literature about antibiotic resistance mechanisms. We studied the specificity of carbapenems for CarO, an outer membrane protein associated with Imipenem-resistance that was strongly related to a decrease in CarO expression level or changes in protein structure. The specificity of five different Carbapenems, Imipenem, Biapenem, Ertapenem, Faropenem, and Meropenem, against the Acinetobacter baumannii ATCC-17978 CarO protein, as well as the specificity of Imipenem for five different types Type-1, Type-2, Type-3, Type-4, and ATCC-17978 CarO protein, were investigated using computational methods. In this study, homology modelling, molecular docking, membrane-protein complex building, and 800 ns long MD simulation methods were followed. The interactions of imipenem with the extracellular region of five different forms of CarO protein were investigated in this study, as well as five different antibiotic binding profiles to the model organism ATCC-17978 CarO protein. The mechanism of CarO influx has been revealed with this study at the molecular level and this data is intended to be used in future research, mutagenesis, and clinical trials

    In vitro inhibition studies of coumarin derivatives on Bos taurus enolase and elucidating their interaction by molecular docking, molecular dynamics simulations and MMGB(PB)SA binding energy calculation

    No full text
    Tropical theileriosis is among the most common vector-borne diseases and caused by Theileria parasites. Theileria annulata is an obligate intracellular protozoan parasite and transmitted to especially Bos taurus and Bos indicus by Hyalomma tick vectors. C8 ([4-(3,4-dimethoxyphenyl)-6,7-dihydroxy-2H-chromen-2-one); C9 (4-(3,4-dihydroxyphenyl)-7,8 dihydroxy-2H-chromen-2-one); C21 (4-(3,4-dihydroxyphenyl)-6,7-dihydroxy-2H-chromen-2 one) were identified as potent Theileria annulata enolase (TaEno) inhibitors in our previous studies. An ideal drug compound must inhibit the target parasite enzyme without inhibiting its homolog in the host. In this study, the inhibitory effect of the compounds previously evaluated on TaEno were tested on the host Bos taurus enolase (BtEno3) by in vitro studies. The interactions of enzyme-coumarin and enzyme-coumarin-substrate by in silico studies were also performed. All of the coumarin derivatives tested showed very low inhibitory effects on B. taurus enolase; 36,87% inhibition at 100 μM concentration for C8, 8,13% inhibition at 100 μM concentration for C9 and 77,69 μM of IC50 value for C21. In addition, these three coumarin derivatives and substrate 2PG were docked into the BtEno3 using molecular docking methods. Molecular interactions between enolase-coumarin and enolase-coumarin-substrate complexes were analyzed using molecular dynamics simulation methods for 100 ns. Estimated free energy of bindings of the substrate 2PG and coumarin derivatives to the BtEno3 were calculated by MM-GB(PB)SA methods. In comparison to the inhibition studies performed on TaEno, C8 and C9 coumarin derivatives remain the possible inhibitor candidates as they inhibit the host enolase at very high concentrations. These two promising compounds will be further analyzed by in vitro and in vivo studies towards developing an alternative drug against tropical theileriosis

    In vitro and in silico evaluation of some plant extracts and phytocompounds against multidrug-resistant Gram-negative bacteria

    No full text
    The spread of multidrug-resistant Gram-negative (MDR) bacteria is a global public health problem, as infections caused by MDR Gram-negative bacteria are difcult to treat. New antibiotic agents need to be developed to overcome this problem, and phytochemicals show promise at this point. In this study, methanol extracts were prepared from cinnamon, thyme, nettle, white tea, rosehip, and antibacterial activity of the methanol extracts was studied against two MDR Gram-Negative bacteria (K. pneumoniae and A. baumannii) by broth microdilution method. The MICs of methanol extracts of cinnamon, rosehip, thyme, white tea for A. baumannii were found as 0.015125 g/ml, 0.07825 g/ml, 0.030625 g/ml, 0.00796875 g/ml, respectively. It was found that only cinnamon methanol extract had antibacterial activity in the used extract concentrations against K. pneumoniae and the MIC value was 0.0605 g/ml. The efects of plant methanol extract with antibacterial activity and imipenem combinations were studied in vitro using the checkerboard method. The FIC Indexes were obtained from the checkerboard results and it was observed that the combination of methanol extract and imipenem showed an antagonistic or additive/indiferent efect but not a synergistic efect. We evaluated the binding afnity of epigallocatechin 3-gallate, quercetin, cinnamaldehyde, carvacrol, and thymol phytocompounds using in silico methods, which are well known as a phytocompounds in white tea, cinnamon, thyme, nettle, and rosehip and have antibacterial activities. The results suggested that these phytocompounds should be supported with in vivo and in vitro experiments to investigate their potential for being inhibitor candidates

    Hit identification against peptidyl-prolyl isomerase of Theileria annulata by combined virtual high-throughput screening and molecular dynamics simulation approach

    No full text
    The numerical calculations reported in this paper were fully/partially performed at TUBITAK ULAKBIM, High Performance and Grid Computing Center (TRUBA resources).Theileria annulata secretes peptidyl prolyl isomerase enzyme (TaPIN1) to manipulate the host cell oncogenic signaling pathway by disrupting the tumor suppressor F-box and WD repeat domain-containing 7 (FBW7) protein level leading to an increased level of c-Jun proto-oncogene. Buparvaquone is a hydroxynaphthoquinone anti-theilerial drug and has been used to treat theileriosis. However, TaPIN1 contains the A53 P mutation that causes drug resistance. In this study, potential TaPIN1 inhibitors were investigated using a library of naphthoquinone derivatives. Comparative models of mutant (m) and wild type (wt) TaPIN1 were predicted and energy minimization was followed by structure validation. A naphthoquinone (hydroxynaphthalene-1,2-dione, hydroxynaphthalene-1,4-dione) and hydroxynaphthalene-2,3-dione library was screened by Schrödinger Glide HTVS, SP and XP docking methodologies and the docked compounds were ranked by the Glide XP scoring function. The two highest ranked docked compounds Compound 1 (4-hydroxy-3-[3,4,5-trihydroxy-6-(hydroxymethyl)oxan-2-yl]oxynaphthalene-1,2-dione) and Compound 2 (6-acetyl-1,4,5,7,8-pentahydroxynaphthalene-2,3-dione) were used for further molecular dynamics (MD) simulation studies. The MD results showed that ligand Compound 1 was located in the active site of both mTaPIN1 and wtTaPIN1 and could be proposed as a potential inhibitor by acting as a substrate antagonist. However, ligand Compound 2 was displaced away from the binding pocket of wtTaPIN1 but was located near the active site binding pocket of mTaPIN1 suggesting that could be selectively evaluated as a potential inhibitor against the mTaPIN1. Compound 1 and Compound 2 ligands are potential inhibitors but Compound 2 is suggested as a better inhibitor for mTaPIN1. These ligands could also further evaluated as potential inhibitors against human peptidyl prolyl isomerase which causes cancer in humans by using the same mechanism as TaPIN1

    Heterologous expression, biochemical characterisation and computational analysis of Bacteroides fragilis enolase

    No full text
    Bacteriodes fragilis is an anaerobic bacterium found in the human intestinal flora. In this study, BfEno was targeted with a structure-based drug design approach because inhibition of this enzyme may prevent both the aerobic and anaerobic pathways due to its role in the glycolytic pathway. First, the gene encoding BfEno was cloned, expressed and the protein produced over 95% purity. The Km and Vmax values of BfEno were determined as 314.9 µM and 256.2 µmol/min.mg, respectively. Drug-like chemicals were retrieved from the ZINC database for high-throughput virtual screening analyses. As a result of screening study, the ZINC91441604 has been proposed to bind to the active site of the enzyme and remain stable. The same compound exhibited weak binding to the human enolases than the bacterial enolase. Hence, ZINC91441604 may be proposed as a novel candidate for further in vitro and in vivo drug analysis towards the treatment of B. fragilis infections
    corecore