Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
bioRxiv ; 2024 Mar 28.
Article in English | MEDLINE | ID: mdl-37732175

ABSTRACT

Rapid and high-fidelity phosphorylation of two serines (S32 and S36) of IκBα by a prototype Ser/Thr kinase IKK2 is critical for fruitful canonical NF-κB activation. Here, we report that IKK2 is a dual specificity Ser/Thr kinase that autophosphorylates itself at tyrosine residues in addition to its activation loop serines. Mutation of one such tyrosine, Y169, located in proximity to the active site, to phenylalanine, renders IKK2 inactive for phosphorylation of S32 of IκBα. Surprisingly, auto-phosphorylated IKK2 relayed phosphate group(s) to IκBα without ATP when ADP is present. We also observed that mutation of K44, an ATP-binding lysine conserved in all protein kinases, to methionine renders IKK2 inactive towards specific phosphorylation of S32 or S36 of IκBα, but not non-specific substrates. These observations highlight an unusual evolution of IKK2, in which autophosphorylation of tyrosine(s) in the activation loop and the invariant ATP-binding K44 residue define its signal-responsive substrate specificity ensuring the fidelity of NF-κB activation.

2.
Carcinogenesis ; 43(6): 571-583, 2022 06 27.
Article in English | MEDLINE | ID: mdl-35165685

ABSTRACT

Eucalyptol (EU) is a monoterpenoid found as an active compound of many plants such as bay leaves, cardamom and is also found as a major constituent in eucalyptus oil. Although the anticancer activity of eucalyptol (EU) has been reported in a few cancer cell lines, its effect on tumor metastasis has not been studied so far. Here, we have shown that the EU has anti-metastatic activity against skin cancer cells in vitro and in vivo. EU decreases migration and invasion of skin cancer cells. Further, it reduces the expression of mesenchymal markers vimentin, snail, slug, twist, and induces the expression of epithelial marker, E-cadherin which indicates that it reverses the epithelial to mesenchymal transition. Gelatin zymography shows that the EU reduces the activity of MMP2 and MMP9. Furthermore signaling study by molecular docking and western blotting shows that EU modulates PI3K/Akt/mTOR signaling pathway. The reduction in the expression of PI3K/Akt/mTOR was enhanced by the use of the PI3K inhibitor, LY294002. In vivo, the anti-metastatic potential of EU was confirmed in C57BL/6 mouse. In conclusion, the EU inhibits migration and invasion of skin cancer by modulating PI3K/Akt/mTOR pathway both in in vitro and in vivo and might provide a new therapeutic approach in skin cancer.


Subject(s)
Phosphatidylinositol 3-Kinases , Skin Neoplasms , Animals , Cell Line, Tumor , Cell Movement , Cell Proliferation , Epithelial-Mesenchymal Transition , Eucalyptol/pharmacology , Mice , Mice, Inbred C57BL , Molecular Docking Simulation , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Skin Neoplasms/drug therapy , TOR Serine-Threonine Kinases/metabolism
3.
J Mol Struct ; 1243: 130854, 2021 Nov 05.
Article in English | MEDLINE | ID: mdl-34121768

ABSTRACT

The recently emerged SARS-CoV2 caused a major pandemic of coronavirus disease (COVID-19). Non structural protein 1 (nsp1) is found in all beta coronavirus that cause severe respiratory disease. This protein is considered as a virulence factor and has an important role in pathogenesis. This study aims to elucidate the structural conformations of nsp1 to aid in the prediction of epitope sites and identification of important residues for targeted therapy against COVID-19. In this study, molecular modelling coupled with molecular dynamics simulations were performed to analyse the conformational landscape of nsp1 homologs of SARS-CoV1, SARS-CoV2 and MERS-CoV. Principal component analysis escorted by free energy landscape revealed that SARS-CoV2 nsp1 protein shows greater flexibility compared to SARS-CoV1 and MERS-CoV nsp1. Sequence comparison reveals that 28 mutations are present in SARS-CoV2 nsp1 protein compared to SARS-CoV1 nsp1. Several B-cell and T-cell epitopes were identified by an immunoinformatics approach. SARS-CoV2 nsp1 protein binds with the interface region of the palm and finger domain of POLA1 via hydrogen bonding and salt bridge interactions. Taken together, these in silico findings may help in the development of therapeutics specific against COVID-19.

4.
J Med Chem ; 63(24): 15621-15638, 2020 12 24.
Article in English | MEDLINE | ID: mdl-33296601

ABSTRACT

Since inception, the magic bullets developed against leishmaniasis traveled a certain path and then dropped down due to either toxicity or the emergence of resistance. The route of administration is also an important concern. We developed a series of water-soluble ferrocenylquinoline derivatives, targeting Leishmania donovani, among which CQFC1 showed the highest efficacy even in comparison to other drugs, in use or used, both in oral and intramuscular routes. It did not induce any toxicity to splenocytes and on hematopoiesis, induced protective cytokines, and did not hamper the drug-metabolizing enzymes in hosts. It acts through the reduction and the inhibition of parasites' survival enzyme trypanothione reductase of replicating amastigotes in hosts' reticuloendothelial tissues. Unlike conventional drugs, this molecule did not induce the resistance-conferring genes in laboratory-maintained resistant L. donovani lines. Experimentally, this easily bioavailable preclinical drug candidate overcame all of the limitations causing the discontinuation of the other conventional antileishmanial drugs.


Subject(s)
Antiprotozoal Agents/chemistry , Leishmania donovani/enzymology , NADH, NADPH Oxidoreductases/antagonists & inhibitors , Protozoan Proteins/antagonists & inhibitors , Quinolines/chemistry , Administration, Oral , Animals , Antiprotozoal Agents/metabolism , Antiprotozoal Agents/pharmacology , Antiprotozoal Agents/therapeutic use , Binding Sites , Disease Models, Animal , Drug Design , Drug Resistance/drug effects , Ferrous Compounds/chemistry , Half-Life , Leishmania donovani/drug effects , Leishmaniasis, Visceral/drug therapy , Metallocenes/chemistry , Mice , Molecular Docking Simulation , Mononuclear Phagocyte System/metabolism , Mononuclear Phagocyte System/parasitology , NADH, NADPH Oxidoreductases/metabolism , Protozoan Proteins/metabolism , Quinolines/metabolism , Quinolines/pharmacology , Quinolines/therapeutic use , Reactive Oxygen Species/metabolism , Solubility , Structure-Activity Relationship
5.
Chemosphere ; 248: 126037, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32018111

ABSTRACT

This study was focused on gaining insights into the mechanism by which the herbicide- Spectracide®, induces oxidative stress and alters behavior in Drosophila melanogaster. Exposure to Spectracide® (50%) significantly (p < 0.05) reduced the negative geotaxis response, jumping behavior and dampened locomotor activity rhythm in adult flies compared to non-exposed flies. Protein carbonyl levels indicative of oxidative damage increased significantly coupled with down-regulation of Sniffer gene expression encoding carbonyl reductase (CR) and its activity in Spectracide®-exposed flies. In silico modeling analysis revealed that the active ingredients of Spectracide® (atrazine, diquat dibromide, fluazifop-p-butyl, and dicamba) have significant binding affinity to the active site of CR enzyme, with atrazine having comparatively greater affinity. Our results suggest a mechanism by which ingredients in Spectracide® induce oxidative damage by competitive binding to the active site of a protective enzyme and impair its ability to prevent damage to proteins thereby leading to deficits in locomotor behavior in Drosophila.


Subject(s)
Herbicides/toxicity , Models, Molecular , Alcohol Oxidoreductases/metabolism , Animals , Atrazine/toxicity , Behavior, Animal/drug effects , Drosophila melanogaster/genetics , Gene Expression , Locomotion/drug effects , Oxidation-Reduction , Oxidative Stress/genetics
6.
J Biomol Struct Dyn ; 38(7): 2080-2092, 2020 Apr.
Article in English | MEDLINE | ID: mdl-31184526

ABSTRACT

Human meprin-ß, a zinc metalloprotease belonging to the astacin family, have been found to be associated with many pathological conditions like inflammatory bowel disease, fibrosis and neurodegenerative disease. The inhibition of meprin-ß by various inhibitors, both macromolecular and small molecules, is crucial in the control of several diseases. Human fetuin-A, a negative acute phase protein involved in inflammatory disease, has recently been identified as an endogenous inhibitor for meprin-ß. In this computational study, an integrated in silico approach was performed using existing structural information of meprin-ß coupled with ab initio modelling of human fetuin-A to predict a rational model of the complex through protein-protein docking. Further, the models were optimized and validated to generate an ensemble of conformations through extensive molecular dynamics simulation. Virtual alanine scanning mutagenesis was explored to identify hotspot residues on both proteins significant for protein-protein interaction (PPI). The results of the study provide structural insight into PPI between meprin-ß and fetuin-A which can be useful in designing molecules to modulate meprin-ß activity. Communicated by Ramaswamy H. Sarma.


Subject(s)
Metalloendopeptidases , Neurodegenerative Diseases , alpha-2-HS-Glycoprotein , Humans , Metalloendopeptidases/metabolism , Metalloproteases , Molecular Dynamics Simulation , alpha-2-HS-Glycoprotein/metabolism
7.
Comput Biol Chem ; 80: 292-306, 2019 Jun.
Article in English | MEDLINE | ID: mdl-31054542

ABSTRACT

Human meprin beta metalloprotease, a small subgroup of the astacin family, is a potent drug target for the treatment of several disorders such as fibrosis, neurodegenerative disease in particular Alzheimer and inflammatory bowel diseases. In this study, a ligand-based pharmacophore approach has been used for the selection of potentially active compounds to understand the inhibitory activities of meprin-ß by using the sulfonamide scaffold based inhibitors. Using this dataset, a pharmacophore model (Hypo1) was selected on the basis of a highest correlation coefficient (0.959), lowest total cost (105.89) and lowest root mean square deviation (1.31 Å) values. All the pharmacophore hypotheses generated from the candidate inhibitors comprised four features: two hydrogen-bond acceptor, one hydrogen-bond donor and one zinc binder feature. The best validated pharmacophore model (Hypo1) was used for virtual screening of compounds from several databases. The selective hit compounds were filtered by drug likeness property, acceptable ADMET profile, molecular docking and DFT study. Molecular dynamic simulations with the final 10 hit compounds revealed that a large number of non-covalent interactions were formed with the active site and specificity sub-pockets of the meprin beta metalloprotease. This study assists in the development of the new lead molecules as well as gives a better understanding of their interaction with meprin-ß.


Subject(s)
Metalloendopeptidases/chemistry , Protease Inhibitors/chemistry , Sulfonamides/chemistry , Catalytic Domain , Datasets as Topic , Density Functional Theory , Drug Design , Humans , Ligands , Metalloendopeptidases/metabolism , Models, Chemical , Molecular Docking Simulation , Molecular Dynamics Simulation , Protease Inhibitors/metabolism , Protein Binding , Sulfonamides/metabolism
8.
J Biomol Struct Dyn ; 37(2): 440-453, 2019 Feb.
Article in English | MEDLINE | ID: mdl-29343185

ABSTRACT

Meprins are a group of zinc metalloproteases of the astacin family which play a pivotal role in several physiological and pathologocal diseases. The inhibition of the meprins by various inhibitors, macromolecular and small molecules, is crucial in the control of several diseases. Human cystatinC, an amyloidogenic protein, is reported to be an endogenous inhibitor of meprin-α. In this computational study, we elucidate a rational model for meprinα-cystatinC complex using protein-protein docking. The complex model as well as the unbound form was evaluated by molecular dynamics simulation. A simulation study revealed higher stability of the complex owing to the presence of several interactions. Virtual alanine mutagenesis helps in identifying the hotspots on both proteins. Based on the frequency of occurrence of hotspot amino acids, it was possible to enumerate the important amino acids primarily responsible for protein stability present at the amino-terminal end of cystatin. Finally, pharmacophore elucidation carried out based on the information obtained from a series of small molecular inhibitors against meprin-α can be utilized in future for rational drug design and therapy.


Subject(s)
Metalloendopeptidases/chemistry , Protein Interaction Domains and Motifs , Drug Discovery , Enzyme Stability , Humans , Hydrogen Bonding , Hydrophobic and Hydrophilic Interactions , Ligands , Metalloendopeptidases/metabolism , Molecular Docking Simulation , Molecular Dynamics Simulation , Protein Binding , Protein Subunits , Reproducibility of Results , Structure-Activity Relationship
9.
J Biomol Struct Dyn ; 37(11): 2801-2822, 2019 Jul.
Article in English | MEDLINE | ID: mdl-30101692

ABSTRACT

Two novel copper (II) substituted thiosemicarbazone Schiff base complexes [Cu(L1)(µ-SCN)]n(NO3)2 (1) and [Cu2(µ-SCN)(SCN)(L2)2](NO3) (2) have been synthesized by condensing substituted thiosemicarbazides like 4-methyl-3-thiosemicarbazide or 4-ethyl-3-thiosemicarbazide with 2-acetylpyridine. Both the metal complexes 1 and 2 are characterized using different spectroscopic techniques like IR, UV-Vis, ESR spectroscopy followed by elemental analysis, cyclic voltammetric measurement and single crystal X-ray structure analysis. X-ray crystal structure analysis reveal that complex 1 is polymeric while complex 2 is dimeric in nature. The coordination geometry around Cu(II) are square pyramidal in which thiosemicarbazone Schiff base ligand coordinate to the central Cu(II) atom in tridentate fashion. The prominent interaction patterns of 1 and 2 with CT-DNA were examined by employing electronic absorption and emission spectral titrations, cyclic voltammetry and viscosity measurements. All the results show that CT-DNA binds with both copper (II) complexes 1 and 2. Furthermore, protein binding ability in vitro of complexes 1 and 2 with both BSA and HSA were carried out using multispectroscopic techniques and a static quenching pattern was observed in both cases. Molecular docking study was employed to ascertain the exact mechanism of action of 1 and 2 with DNA and protein molecules (BSA and HSA). In vitro cytotoxicity activity of complexes 1 and 2 toward AGS and A549 was evaluated using MTT assay which demonstrates that both complexes 1 and 2 have superior prospectus to act as anticancer agents. Communicated by Ramaswamy H. Sarma.


Subject(s)
Coordination Complexes/pharmacology , Copper/chemistry , DNA/metabolism , Molecular Docking Simulation , Serum Albumin, Bovine/metabolism , Serum Albumin, Human/metabolism , Thiosemicarbazones/chemistry , A549 Cells , Animals , Binding Sites , Cattle , Cell Cycle/drug effects , Cell Proliferation/drug effects , Coordination Complexes/chemistry , Crystallography, X-Ray , DNA/chemistry , Hemolysis/drug effects , Humans , Ligands , Protein Binding , Protein Conformation , Schiff Bases , Serum Albumin, Bovine/chemistry , Serum Albumin, Human/chemistry , Spectrometry, Fluorescence/methods
10.
Sci Rep ; 8(1): 11784, 2018 08 06.
Article in English | MEDLINE | ID: mdl-30082709

ABSTRACT

Japanese Encephalitis Virus (JEV), a globally important pathogen, belongs to the family Flaviviridae, is transmitted between vertebrate hosts by mosquitoes, principally by Culex tritaeniorhynchus. The E-glycoprotein of the virus mediates its attachment to the host cell receptors. In this study, we cloned and purified JEV E-glycoprotein in pET28a vector using E. coli BL21 (DE3) cells. A pull down assay was performed using plasma membrane fraction of BALB/c mouse brain and E-glycoprotein as a bait protein. 2-Dimensional Gel Electrophoresis based separation of the interacting proteins was analyzed by mass spectrometry. Among all the identified partners of E-glycoprotein, PLVAP (Plasmalemma vesicle associated protein) and GKN3 (Gastrokine3) showed significant up-regulation in both JEV infected mouse brain and neuro2a cells. In-silico studies also predicted significant interaction of these receptors with E-glycoprotein. Additionally, overexperssion and silencing of these receptors resulted in increase and reduction in viral load respectively, suggesting them as two critical cellular receptors governing JEV entry and propagation in neurons. In support, we observed significant expression of PLVAP but not GKN3 in post-mortem autopsied human brain tissue. Our results establish two novel receptor proteins in neurons in case of JEV infection, thus providing potential targets for antiviral research.


Subject(s)
Brain/metabolism , Carrier Proteins/metabolism , Encephalitis Virus, Japanese/pathogenicity , Membrane Proteins/metabolism , Animals , Cell Line, Tumor , Electrophoresis, Gel, Two-Dimensional , Encephalitis Virus, Japanese/metabolism , Encephalitis, Japanese/metabolism , Encephalitis, Japanese/virology , Female , Male , Mass Spectrometry , Mice , Mice, Inbred BALB C , Neurons/metabolism , Viral Envelope Proteins/metabolism
11.
Spectrochim Acta A Mol Biomol Spectrosc ; 175: 191-199, 2017 Mar 15.
Article in English | MEDLINE | ID: mdl-28039847

ABSTRACT

An environment sensitive fluorophore, 4-(5-(4-(dimethylamino)phenyl)oxazol-2-yl)benzoic acid (DMOBA), that closely mimics biologically active 2,5-disubstituited oxazoles has been designed to probe two homologous serum proteins, human serum albumin (HSA) and bovine serum albumin (BSA) by means of photophysical and molecular modeling studies. This fluorescent analogue exhibits solvent polarity sensitive fluorescence due to an intramolecular charge transfer in the excited state. In comparison to water, the steady state emission spectra of DMOBA in BSA is characterized by a greater blue shift (~10nm) and smaller Stokes' shift (~5980cm-1) in BSA than HSA (Stokes'shift~6600cm-1), indicating less polar and more hydrophobic environment of the dye in the former than the latter. The dye-protein binding interactions are remarkably stronger for BSA than HSA which is evident from higher value of the association constant for the DMOBA-BSA complex (Ka~5.2×106M-1) than the DMOBA-HSA complex (Ka~1.0×106M-1). FÓ§rster resonance energy transfer studies revealed remarkably less efficient energy transfer (8%) between the donor tryptophans in BSA and the acceptor DMOBA dye than that (30%) between the single tryptophan moiety in HSA and the dye, which is consistent with a much larger distance between the donor (tryptophan)-acceptor (dye) pair in BSA (34.5Å) than HSA (25.4Å). Site specific competitive binding assays have confirmed on the location of the dye in Sudlow's site II of BSA and in Sudlow's site I of HSA, respectively. Molecular modeling studies have shown that the fluorescent analogue is tightly packed in the binding site of BSA due to strong steric complementarity, where, binding of DMOBA to BSA is primarily dictated by the van der Waals and hydrogen bonding interactions. In contrast, in HSA the steric complementarity is less significant and binding is primarily guided by polar interactions and van der Waals interactions appear to be less significant in the formation of the HSA-DMOBA complex. Electrostatic interactions contribute significantly in the binding of DMOBA to HSA (-2.09kcal/mol) compared to BSA (-0.47kcal/mol). Electrostatic surface potential calculation reveals that the DMOBA binding site within HSA is highly charged compared to BSA.


Subject(s)
Environment , Fluorescent Dyes/chemistry , Models, Molecular , Oxazoles/chemistry , Serum Albumin, Bovine/analysis , Serum Albumin, Human/analysis , Animals , Anisotropy , Binding Sites , Cattle , Fluorescence Resonance Energy Transfer , Humans , Kinetics , Molecular Docking Simulation , Serum Albumin, Bovine/chemistry , Serum Albumin, Human/chemistry , Spectrometry, Fluorescence , Spectrophotometry, Ultraviolet , Static Electricity , Thermodynamics , Time Factors
12.
Comb Chem High Throughput Screen ; 19(3): 246-58, 2016.
Article in English | MEDLINE | ID: mdl-26875789

ABSTRACT

Human meprin-α and-ß are important regulators of angiogenesis, cancer, inflammation, fibrosis, and neurodegenerative diseases and hence important therapeutic targets. Meprins are the only astacin proteases that are expressed in membrane-bound and secreted form. The cleavage specificity of human meprins is similar in certain cases but differs markedly in others. The inhibitor selectivity of human meprins is controlled by the specific residues involved in binding at the active-site cleft of the proteases. Meprins are inhibited by various small molecular inhibitors as well as macromolecular endogenous inhibitors, making them good drug targets. In the current study, molecular dynamics simulation was performed for 10 ns on ten systems consisting of two apoenzymes of meprin -α/ß and eight complexes of human meprin-α and -ß complexed to four inhibitors with different metal binding moieties and comparable Ki values. These simulation studies helped to elucidate the molecular details of how several parameters influence protein-inhibitor binding affinity. Analysis of the interaction energies of the protein-inhibitor complexes revealed the diverse binding nature of this series of inhibitors. Several structural segments of human meprins exhibited certain conformational changes during the simulation time course. Among the inhibitors studied captopril had a different disposition in the meprin-bound complexes compared to the other three inhibitors, namely Pro- Leu-Gly-hydroxamate, galardin and EDTA. Comparison of the interaction energies for each system helped us to conclude that the hydroxamic acid-based inhibitors are the most potent inhibitors of meprins.


Subject(s)
Metalloendopeptidases/antagonists & inhibitors , Molecular Dynamics Simulation , Protease Inhibitors/pharmacology , Binding Sites/drug effects , Dose-Response Relationship, Drug , Humans , Molecular Docking Simulation , Molecular Structure , Protease Inhibitors/chemistry , Structure-Activity Relationship
13.
Antimicrob Agents Chemother ; 60(5): 2696-708, 2016 05.
Article in English | MEDLINE | ID: mdl-26883702

ABSTRACT

In our previous report, we showed that astrakurkurone, a triterpene isolated from the Indian mushroom Astraeus hygrometricus (Pers.) Morgan, induced reactive oxygen species, leading to apoptosis in Leishmania donovani promastigotes, and also was effective in inhibiting intracellular amastigotes at the 50% inhibitory concentration of 2.5 µg/ml. The aim of the present study is to characterize the associated immunomodulatory potentials and cellular activation provided by astrakurkurone, leading to effective antileishmanial activity in vitro and in vivo Astrakurkurone-mediated antileishmanial activity was evaluated by real-time PCR and flow cytometry. The involvement of Toll-like receptor 9 (TLR9) was studied by in vitro assay in the presence of a TLR9 agonist and antagonist and by in silico modeling of a three-dimensional structure of the ectodomain of TLR9 and its interaction with astrakurkurone. Astrakurkurone caused a significant increase in TLR9 expression of L. donovani-infected macrophages along with the activation of proinflammatory responses. The involvement of TLR9 in astrakurkurone-mediated amastigote killing has been evidenced from the fact that a TLR9 agonist (CpG, ODN 1826) in combination with astrakurkurone enhanced the amastigote killing, while a TLR9 antagonist (bafilomycin A1) alone or in combination with astrakurkurone curbed the amastigote killing, which could be further justified by in silico evidence of docking between mouse TLR9 and astrakurkurone. Astrakurkurone was found to reduce the parasite burden in vivo by inducing protective cytokines, gamma interferon and interleukin 17. Moreover, astrakurkurone was nontoxic toward peripheral blood mononuclear cells of immunocompromised patients with visceral leishmaniasis. Astrakurkurone, a nontoxic antileishmanial, enhances the immune efficiency of host cells, leading to parasite clearance in vitro and in vivo.


Subject(s)
Antiprotozoal Agents/therapeutic use , Leishmaniasis, Visceral/drug therapy , Leishmaniasis, Visceral/immunology , Toll-Like Receptor 9/metabolism , Triterpenes/therapeutic use , Agaricales/chemistry , Animals , Antiprotozoal Agents/immunology , Blotting, Western , Flow Cytometry , Immunity, Cellular/drug effects , Macrolides/therapeutic use , Macrophages/drug effects , Macrophages/metabolism , Male , Mice , Mice, Inbred BALB C , NF-kappa B/metabolism , Reactive Oxygen Species/metabolism , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Toll-Like Receptor 9/agonists , Toll-Like Receptor 9/antagonists & inhibitors , Triterpenes/immunology
14.
J Biomol Struct Dyn ; 32(12): 1969-78, 2014 Dec.
Article in English | MEDLINE | ID: mdl-24279637

ABSTRACT

Meprins are complex and highly glycosylated multi-domain enzymes that require post-translational modifications to reach full activity. Meprins are metalloproteases of the astacin family characterized by a conserved zinc-binding motif (HExxHxxGFxHExxRxDR). Human meprin-α and -ß protease subunits are 55% identical at the amino acid level, however the substrate and peptide bond specificities vary markedly. Current work focuses on the critical amino acid residues in the non-primed subsites of human meprins-α and -ß involved in inhibitor/ligand binding. To compare the molecular events underlying ligand affinity, homology modeling of the protease domain of humep-α and -ß based on the astacin crystal structure followed by energy minimization and molecular dynamics simulation of fully solvated proteases with inhibitor Pro-Leu-Gly-hydroxamate in S subsites were performed. The solvent accessible surface area curve shows a decrease in solvent accessibility values at specific residues upon inhibitor binding. The potential energy, total energy, H-bond interactions, root mean square deviation and root mean square fluctuation plot reflect the subtle differences in the S subsite of the enzymes which interact with different residues at P site of the inhibitor.


Subject(s)
Hydroxamic Acids/chemistry , Tiopronin/chemistry , Amino Acid Sequence , Binding Sites , Humans , Hydrogen Bonding , Ligands , Molecular Dynamics Simulation , Molecular Sequence Data , Protein Stability , Sequence Analysis, Protein
SELECTION OF CITATIONS
SEARCH DETAIL
...