Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Nature ; 601(7893): 465-469, 2022 01.
Article in English | MEDLINE | ID: mdl-34937936

ABSTRACT

Hsp90 is a conserved and essential molecular chaperone responsible for the folding and activation of hundreds of 'client' proteins1-3. The glucocorticoid receptor (GR) is a model client that constantly depends on Hsp90 for activity4-9. GR ligand binding was previously shown to nr inhibited by Hsp70 and restored by Hsp90, aided by the co-chaperone p2310. However, a molecular understanding of the chaperone-mediated remodelling that occurs between the inactive Hsp70-Hsp90 'client-loading complex' and an activated Hsp90-p23 'client-maturation complex' is lacking for any client, including GR. Here we present a cryo-electron microscopy (cryo-EM) structure of the human GR-maturation complex (GR-Hsp90-p23), revealing that the GR ligand-binding domain is restored to a folded, ligand-bound conformation, while being simultaneously threaded through the Hsp90 lumen. In addition, p23 directly stabilizes native GR using a C-terminal helix, resulting in enhanced ligand binding. This structure of a client bound to Hsp90 in a native conformation contrasts sharply with the unfolded kinase-Hsp90 structure11. Thus, aided by direct co-chaperone-client interactions, Hsp90 can directly dictate client-specific folding outcomes. Together with the GR-loading complex structure12, we present the molecular mechanism of chaperone-mediated GR remodelling, establishing the first, to our knowledge, complete chaperone cycle for any Hsp90 client.


Subject(s)
Cryoelectron Microscopy , HSP90 Heat-Shock Proteins , Prostaglandin-E Synthases , Receptors, Glucocorticoid , HSP70 Heat-Shock Proteins/chemistry , HSP70 Heat-Shock Proteins/metabolism , HSP70 Heat-Shock Proteins/ultrastructure , HSP90 Heat-Shock Proteins/chemistry , HSP90 Heat-Shock Proteins/metabolism , HSP90 Heat-Shock Proteins/ultrastructure , Humans , Ligands , Molecular Chaperones/chemistry , Molecular Chaperones/metabolism , Molecular Chaperones/ultrastructure , Prostaglandin-E Synthases/chemistry , Prostaglandin-E Synthases/metabolism , Prostaglandin-E Synthases/ultrastructure , Protein Binding , Receptors, Glucocorticoid/chemistry , Receptors, Glucocorticoid/metabolism , Receptors, Glucocorticoid/ultrastructure
2.
Nat Commun ; 12(1): 828, 2021 02 05.
Article in English | MEDLINE | ID: mdl-33547294

ABSTRACT

The co-chaperone p23 is a central part of the Hsp90 machinery. It stabilizes the closed conformation of Hsp90, inhibits its ATPase and is important for client maturation. Yet, how this is achieved has remained enigmatic. Here, we show that a tryptophan residue in the proximal region of the tail decelerates the ATPase by allosterically switching the conformation of the catalytic loop in Hsp90. We further show by NMR spectroscopy that the tail interacts with the Hsp90 client binding site via a conserved helix. This helical motif in the p23 tail also binds to the client protein glucocorticoid receptor (GR) in the free and Hsp90-bound form. In vivo experiments confirm the physiological importance of ATPase modulation and the role of the evolutionary conserved helical motif for GR activation in the cellular context.


Subject(s)
Adenylyl Imidodiphosphate/chemistry , HSP90 Heat-Shock Proteins/chemistry , Molecular Chaperones/chemistry , Prostaglandin-E Synthases/chemistry , Receptors, Glucocorticoid/chemistry , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae/chemistry , Adenylyl Imidodiphosphate/metabolism , Amino Acid Sequence , Binding Sites , Cloning, Molecular , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , HSP90 Heat-Shock Proteins/genetics , HSP90 Heat-Shock Proteins/metabolism , Humans , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , Molecular Dynamics Simulation , Mutation , Nuclear Magnetic Resonance, Biomolecular , Prostaglandin-E Synthases/genetics , Prostaglandin-E Synthases/metabolism , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Receptors, Glucocorticoid/genetics , Receptors, Glucocorticoid/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid
3.
Protein J ; 40(1): 63-67, 2021 02.
Article in English | MEDLINE | ID: mdl-33403608

ABSTRACT

Prostaglandin E synthase (PGES) catalyzes the conversion of prostaglandin H2 to prostaglandin E2 in the presence of glutathione (GSH) in mammals. Amid the limited knowledge on prostaglandin and its related enzymes in insects, we recently identified PGES from the silkworm Bombyx mori (bmPGES) and determined its crystal structure complexed with GSH. In the current study, we investigated the substrate-binding site of bmPGES by site-directed mutagenesis and X-ray crystallography. We found that the residues Tyr107, Val155, Met159, and Glu203 are located in the catalytic pockets of bmPGES, and mutagenesis of each residue reduced the bmPGES activity. Our results suggest that these four residues contribute to the catalytic activity of bmPGES. Overall, this structure-function study holds implications in controlling pests by designing rational and efficient pesticides.


Subject(s)
Bombyx/chemistry , Dinoprostone/chemistry , Glutathione/chemistry , Insect Proteins/chemistry , Prostaglandin-E Synthases/chemistry , Amino Acid Motifs , Animals , Bombyx/enzymology , Catalytic Domain , Cloning, Molecular , Crystallography, X-Ray , Dinitrochlorobenzene/chemistry , Dinitrochlorobenzene/metabolism , Dinoprostone/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Glutathione/metabolism , Insect Proteins/genetics , Insect Proteins/metabolism , Models, Molecular , Mutagenesis, Site-Directed , Mutation , Prostaglandin-E Synthases/genetics , Prostaglandin-E Synthases/metabolism , Protein Binding , Protein Interaction Domains and Motifs , Protein Structure, Secondary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Structure-Activity Relationship , Substrate Specificity
4.
J Chem Inf Model ; 59(9): 3839-3845, 2019 09 23.
Article in English | MEDLINE | ID: mdl-31429562

ABSTRACT

Microsomal prostaglandin E2 synthase-1 (mPGES-1) is known as an ideal target for next-generation anti-inflammatory drugs to effectively and safely treat a variety of inflammation-related diseases. High-resolution X-ray crystal structures are available for human mPGES-1, but all in a closed conformation for a glutathione (GSH)-binding site. Here, we report an in silico observation of the desirable open conformation of mPGES-1 using a simple computational strategy with fully relaxed molecular dynamics simulations starting a high-resolution X-ray crystal structure in the closed conformation. The open conformation mainly exists in the apo-form. Once GSH enters the binding site, the binding site is closed and, thus, mPGES-1 becomes the closed conformation. According to the determined free energy profile, both the open and closed conformations can co-exist in solution with a thermodynamic equilibrium, and the conformational distribution is dependent on the GSH concentration. In addition, the cap domain responsible for the conformational transition is located right on the crystal packing interface, showing that only closed conformation is suitable for the crystal packing. All of the computational insights are consistent with reported experimental observations. The computationally simulated open conformation of mPGES-1 may serve as a new target state for the rational design of novel inhibitors of mPGES-1. We anticipate that a computational strategy similar to the one used in this study may also be used to explore open conformation starting from a crystal structure of the corresponding closed conformation with a ligand bound for other proteins.


Subject(s)
Computer Simulation , Glutathione/metabolism , Molecular Dynamics Simulation , Prostaglandin-E Synthases/chemistry , Prostaglandin-E Synthases/metabolism , Binding Sites , Humans , Protein Domains , Thermodynamics
5.
J Cell Biochem ; 120(11): 19064-19075, 2019 11.
Article in English | MEDLINE | ID: mdl-31257629

ABSTRACT

Co-chaperon p23 has been well established as molecular chaperon for the heat shock protein 90 (Hsp90) that further leads to immorality in cancer cells by providing defense against Hsp90 inhibitors, and as stimulating agent for generating overexpressed antiapoptotic proteins, that is, Hsp70 and Hsp27. The natural compounds such as catechins from Camellia sinensis (green tea) are also well known for inhibition activity against various cancer. However, molecular interaction profile and potential lead bioactive compounds against co-chaperon p23 from green tea are not yet reported. To this context, we study the various secondary metabolites of green tea against co-chaperon p23 using structure-based virtual screening from Traditional Chinese Medicine (TCM) database. Following 26 compounds were obtained from TCM database and further studied for extra precision molecular docking that showed binding score between -10.221 and -2.276 kcal/mol with co-chaperon p23. However, relative docking score to known inhibitors, that is, ailanthone (-4.54 kcal/mol) and gedunin ( 3.60 kcal/mol) along with ADME profile analysis concluded epicatechin (-7.013 kcal/mol) and cis-theaspirone (-4.495 kcal/mol) as potential lead inhibitors from green tea against co-chaperone p23. Furthermore, molecular dynamics simulation and molecular mechanics generalized born surface area calculations validated that epicatechin and cis-theaspirone have significantly occupied the active region of co-chaperone p23 by hydrogen and hydrophobic interactions with various residues including most substantial amino acids, that is, Thr90, Ala94, and Lys95. Hence, these results supported the fact that green tea contained potential compounds with an ability to inhibit the cancer by disrupting the co-chaperon p23 activity.


Subject(s)
Antineoplastic Agents, Phytogenic/chemistry , Camellia sinensis/chemistry , Molecular Docking Simulation , Molecular Dynamics Simulation , Phytochemicals/chemistry , Prostaglandin-E Synthases , Humans , Prostaglandin-E Synthases/antagonists & inhibitors , Prostaglandin-E Synthases/chemistry
6.
Biochemistry ; 58(24): 2715-2719, 2019 06 18.
Article in English | MEDLINE | ID: mdl-31181899

ABSTRACT

Despite its power in identifying highly potent ligands for select protein targets, conventional medicinal chemistry is limited by its low throughput and lack of proteomic selectivity information. We seek to develop a chemoproteomic approach for discovering covalent ligands for protein targets in an unbiased, high-throughput manner. Tripartite probe compounds composed of a heterocyclic core, an electrophilic "warhead", and an alkyne tag have been designed and synthesized for covalently labeling and identifying targets in cells. We have developed a novel condensation reaction to prepare 2-chloromethylquinoline (2-CMQ), an electrophilic heterocycle. These chloromethylquinolines potently and covalently bind to a number of cellular protein targets, including prostaglandin E synthase 2 (PTGES2), a critical regulator of cell proliferation, apoptosis, angiogenesis, inflammation, and immune surveillance. The 2-CMQs that we have developed here are novel PTGES2 binders that have the potential to serve as therapies for the treatment of human diseases such as inflammation.


Subject(s)
Molecular Probes/pharmacology , Prostaglandin-E Synthases/drug effects , Quinolines/pharmacology , Glutathione Transferase/chemistry , Glutathione Transferase/drug effects , HEK293 Cells , Heme Oxygenase (Decyclizing)/chemistry , Heme Oxygenase (Decyclizing)/drug effects , Humans , Molecular Probes/chemical synthesis , Molecular Probes/chemistry , Prostaglandin-E Synthases/chemistry , Proteome/chemistry , Proteomics/methods , Quinolines/chemical synthesis , Quinolines/chemistry
7.
Int J Biol Macromol ; 119: 352-359, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30031079

ABSTRACT

The huge therapeutic potential and the market share of painkillers are well-known. Due to the side effects associated with traditional NSAIDs and selective cyclooxygenase (COX-2) inhibitors, a new generation of painkillers is the need of the hour. In this regard, microsomal prostaglandin E synthase-1 (mPGES-1) offers great potential as an alternative drug target against inflammatory disorders. The present study is aimed at identifying the amino acids crucial in effective inhibitor binding at the mPGES-1 active site by performing molecular dynamics based studies on a series of 7-Phenyl-imidazoquinolin-4(5H)-one derivatives. Molecular dynamics (MD) simulations, MM-PBSA, per-residue energy decomposition and Dimensionality Reduction through Covariance matrix Transformation for Identification of Differences in dynamics (DIRECT-ID) analysis were performed to get insights into the structural details that can aid in novel drug design against mPGES-1. The high correlations of electrostatic and polar energy terms with biological activity highlight their importance and applicability in in silico screening studies. Further, per-residue energy decomposition studies revealed that Lys42, Arg52, Arg122, Pro124, Ser127, Val128 and Thr131 were contributing more towards inhibitor binding energy. The results clearly show that MM-PBSA can act as a filter in virtual screening experiments and can play major role in facilitating various mPGES-1 drug discovery studies.


Subject(s)
Adipates/chemistry , Catalytic Domain , Molecular Dynamics Simulation , Prostaglandin-E Synthases/chemistry , Succinates/chemistry , Ligands , Molecular Structure , Protein Binding
8.
Sci Rep ; 8(1): 5205, 2018 03 26.
Article in English | MEDLINE | ID: mdl-29581541

ABSTRACT

Human mPGES-1 is recognized as a promising target for next generation of anti-inflammatory drugs without the side effects of currently available anti-inflammatory drugs, and various inhibitors have been reported in the literature. However, none of the reported potent inhibitors of human mPGES-1 has shown to be also a potent inhibitor of mouse or rat mPGES-1, which prevents using the well-established mouse/rat models of inflammation-related diseases for preclinical studies. Hence, despite of extensive efforts to design and discover various human mPGES-1 inhibitors, the promise of mPGES-1 as a target for the next generation of anti-inflammatory drugs has never been demonstrated in any wild-type mouse/rat model using an mPGES-1 inhibitor. Here we report discovery of a novel type of selective mPGES-1 inhibitors potent for both human and mouse mPGES-1 enzymes through structure-based rational design. Based on in vivo studies using wild-type mice, the lead compound is indeed non-toxic, orally bioavailable, and more potent in decreasing the PGE2 (an inflammatory marker) levels compared to the currently available drug celecoxib. This is the first demonstration in wild-type mice that mPGES-1 is truly a promising target for the next generation of anti-inflammatory drugs.


Subject(s)
Anti-Inflammatory Agents/chemistry , Enzyme Inhibitors/chemistry , Inflammation/drug therapy , Prostaglandin-E Synthases/chemistry , Animals , Anti-Inflammatory Agents/administration & dosage , Enzyme Inhibitors/administration & dosage , Humans , Inflammation/genetics , Inflammation/pathology , Mice , Prostaglandin-E Synthases/antagonists & inhibitors , Rats , Structure-Activity Relationship
9.
Mol Inform ; 37(3)2018 03.
Article in English | MEDLINE | ID: mdl-28991413

ABSTRACT

The single-target drugs against the arachidonic acid inflammatory pathway are associated with serious side effects, hence, as a first step towards multi-target drugs, we have studied the pharmacophoric features common to the inhibitors of 5-lipoxygenase-activating protein (FLAP), microsomal prostaglandin E-synthase 1 (mPGES-1) and leukotriene A4 hydrolase (LTA4H). FLAP and mPGES-1 shared subfamily-specific positions (SSPs) and four mPGES-1 inhibitors binding to them mapped onto the pharmacophore derived from FLAP inhibitors (Ph-FLAP). The reactions of mPGES-1 and LTA4H had high structural similarity. The pharmacophore derived from two substrate mimic inhibitors of LTA4H (Ph-LTA4H) also mapped onto three mPGES-1 inhibitors. Screening of in-house database for Ph-FLAP and Ph-LTA4H identified one compound, C1. It inhibited the production of the mPGES-1 product, prostaglandin E2 (PGE2) by 97.8±1.6 % at 50 µM in HeLa cells and can be a starting point for designing molecules inhibiting all three targets simultaneously.


Subject(s)
5-Lipoxygenase-Activating Protein Inhibitors/chemistry , 5-Lipoxygenase-Activating Proteins/chemistry , Arachidonate 5-Lipoxygenase/chemistry , Lipoxygenase Inhibitors/chemistry , Molecular Docking Simulation , Prostaglandin-E Synthases/chemistry , 5-Lipoxygenase-Activating Protein Inhibitors/pharmacology , 5-Lipoxygenase-Activating Proteins/metabolism , Animals , Arachidonate 5-Lipoxygenase/metabolism , Binding Sites , Humans , Lipoxygenase Inhibitors/pharmacology , Prostaglandin-E Synthases/antagonists & inhibitors , Prostaglandin-E Synthases/metabolism , Protein Binding
10.
Eur J Med Chem ; 143: 1419-1427, 2018 Jan 01.
Article in English | MEDLINE | ID: mdl-29133047

ABSTRACT

mPGES-1, a glutathione-dependent membrane protein is involved in the last step of PGE2 production and has been well recognized as a strategic target for the development of anti-inflammatory and anti-cancer agents. It has been proven to selectively control the PGE2 levels induced by inflammatory stimuli, with neither affecting PGE2 constitutively produced, nor homeostatic prostanoids, so that its modulation can represent a better strategy to control PGE2 related disorders, compared to the use of the classical anti-inflammatory drugs, endowed with severe side effects. Despite the intensive research on the identification of potent mPGES-1 inhibitors as attractive candidates for drug development, none of the disclosed molecules, except for LY3023705, which recently entered clinical trials, are available for clinical use, therefore the discovery of new effective mPGES-1 inhibitors with increased drug-like properties are urgently needed. Continuing our work aimed at identifying new chemical platforms able to interact with this enzyme, here we describe the discovery of potent mPGES-1 modulators, featuring a 1-fluoro-2,4-dinitro-biphenyl-based scaffold, by processing and docking a small collection of synthetically accessible molecules, built around two main fragments, disclosed in our in silico screening. The top scoring hits obtained have been synthesized and tested, and five of the predicted compounds showed to potently inhibit mPGES-1 enzyme, without affecting COX enzymes activities.


Subject(s)
Drug Discovery , Enzyme Inhibitors/pharmacology , Prostaglandin-E Synthases/antagonists & inhibitors , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , Humans , Molecular Docking Simulation , Prostaglandin-E Synthases/chemistry , Prostaglandin-E Synthases/metabolism , Protein Conformation
11.
Mol Divers ; 21(3): 661-675, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28484935

ABSTRACT

Human microsomal prostaglandin [Formula: see text] synthase (mPGES)-1 is a promising drug target for inflammation and other diseases with inflammatory symptoms. In this work, we built classification models which were able to classify mPGES-1 inhibitors into two groups: highly active inhibitors and weakly active inhibitors. A dataset of 1910 mPGES-1 inhibitors was separated into a training set and a test set by two methods, by a Kohonen's self-organizing map or by random selection. The molecules were represented by different types of fingerprint descriptors including MACCS keys (MACCS), CDK fingerprints, Estate fingerprints, PubChem fingerprints, substructure fingerprints and 2D atom pairs fingerprint. First, we used a support vector machine (SVM) to build twelve models with six types of fingerprints and found that MACCS had some advantage over the other fingerprints in modeling. Next, we used naïve Bayes (NB), random forest (RF) and multilayer perceptron (MLP) methods to build six models with MACCS only and found that models using RF and MLP methods were better than NB. Finally, all the models with MACCS keys were used to make predictions on an external test set of 41 compounds. In summary, the models built with MACCS keys and using SVM, RF and MLP methods show good prediction performance on the test sets and the external test set. Furthermore, we made a structure-activity relationship analysis between mPGES-1 and its inhibitors based on the information gain of fingerprints and could pinpoint some key functional groups for mPGES-1 activity. It was found that highly active inhibitors usually contained an amide group, an aromatic ring or a nitrogen heterocyclic ring, and several heteroatoms substituents such as fluorine and chlorine. The carboxyl group and sulfur atom groups mainly appeared in weakly active inhibitors.


Subject(s)
Enzyme Inhibitors/chemistry , Prostaglandin-E Synthases/antagonists & inhibitors , Small Molecule Libraries/chemistry , Algorithms , Bayes Theorem , Computer Simulation , Enzyme Inhibitors/pharmacology , Humans , Models, Molecular , Prostaglandin-E Synthases/chemistry , Quantitative Structure-Activity Relationship , Small Molecule Libraries/pharmacology , Support Vector Machine
12.
Bioorg Med Chem Lett ; 27(6): 1478-1483, 2017 03 15.
Article in English | MEDLINE | ID: mdl-28190634

ABSTRACT

We describe a novel class of acidic mPGES-1 inhibitors with nanomolar enzymatic and human whole blood (HWB) potency. Rational design in conjunction with structure-based design led initially to the identification of anthranilic acid 5, an mPGES-1 inhibitor with micromolar HWB potency. Structural modifications of 5 improved HWB potency by over 1000×, reduced CYP2C9 single point inhibition, and improved rat clearance, which led to the selection of [(cyclopentyl)ethyl]benzoic acid compound 16 for clinical studies. Compound 16 showed an IC80 of 24nM for inhibition of PGE2 formation in vitro in LPS-stimulated HWB. A single oral dose resulted in plasma concentrations of 16 that exceeded its HWB IC80 in both rat (5mg/kg) and dog (3mg/kg) for over twelve hours.


Subject(s)
Benzoates/chemistry , Benzoates/pharmacology , Drug Discovery , Microsomes/drug effects , Prostaglandin-E Synthases/antagonists & inhibitors , Animals , Crystallography, X-Ray , Dogs , Microsomes/enzymology , Prostaglandin-E Synthases/chemistry , Rats
13.
SAR QSAR Environ Res ; 28(1): 17-39, 2017 Jan.
Article in English | MEDLINE | ID: mdl-28094550

ABSTRACT

COX-2 inhibitors exhibit anticancer effects in various cancer models but due to the adverse side effects associated with these inhibitors, targeting molecules downstream of COX-2 (such as mPGES-1) has been suggested. Even after calls for mPGES-1 inhibitor design, to date there are only a few published inhibitors targeting the enzyme and displaying anticancer activity. In the present study, we have deployed both ligand and structure-based drug design approaches to hunt novel drug-like candidates as mPGES-1 inhibitors. Fifty-four compounds with tested mPGES-1 inhibitory value were used to develop a model with four pharmacophoric features. 3D-QSAR studies were undertaken to check the robustness of the model. Statistical parameters such as r2 = 0.9924, q2 = 0.5761 and F test = 1139.7 indicated significant predictive ability of the proposed model. Our QSAR model exhibits sites where a hydrogen bond donor, hydrophobic group and the aromatic ring can be substituted so as to enhance the efficacy of the inhibitor. Furthermore, we used our validated pharmacophore model as a three-dimensional query to screen the FDA-approved Lopac database. Finally, five compounds were selected as potent mPGES-1 inhibitors on the basis of their docking energy and pharmacokinetic properties such as ADME and Lipinski rule of five.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Prostaglandin-E Synthases/antagonists & inhibitors , Prostaglandin-E Synthases/chemistry , Quantitative Structure-Activity Relationship , Drug Evaluation, Preclinical/methods , Models, Molecular , Models, Statistical
14.
Proteins ; 85(5): 812-826, 2017 05.
Article in English | MEDLINE | ID: mdl-27936510

ABSTRACT

We critically test and validate the CS-Rosetta methodology for de novo structure prediction of α-helical membrane proteins (MPs) from NMR data, such as chemical shifts and NOE distance restraints. By systematically reducing the number and types of NOE restraints, we focus on determining the regime in which MP structures can be reliably predicted and pinpoint the boundaries of the approach. Five MPs of known structure were used as test systems, phototaxis sensory rhodopsin II (pSRII), a subdomain of pSRII, disulfide binding protein B (DsbB), microsomal prostaglandin E2 synthase-1 (mPGES-1), and translocator protein (TSPO). For pSRII and DsbB, where NMR and X-ray structures are available, resolution-adapted structural recombination (RASREC) CS-Rosetta yields structures that are as close to the X-ray structure as the published NMR structures if all available NMR data are used to guide structure prediction. For mPGES-1 and Bacillus cereus TSPO, where only X-ray crystal structures are available, highly accurate structures are obtained using simulated NMR data. One main advantage of RASREC CS-Rosetta is its robustness with respect to even a drastic reduction of the number of NOEs. Close-to-native structures were obtained with one randomly picked long-range NOEs for every 14, 31, 38, and 8 residues for full-length pSRII, the pSRII subdomain, TSPO, and DsbB, respectively, in addition to using chemical shifts. For mPGES-1, atomically accurate structures could be predicted even from chemical shifts alone. Our results show that atomic level accuracy for helical membrane proteins is achievable with CS-Rosetta using very sparse NOE restraint sets to guide structure prediction. Proteins 2017; 85:812-826. © 2016 Wiley Periodicals, Inc.


Subject(s)
Archaeal Proteins/chemistry , Bacillus cereus/chemistry , Bacterial Proteins/chemistry , Carotenoids/chemistry , Carrier Proteins/chemistry , Halobacteriales/chemistry , Membrane Proteins/chemistry , Prostaglandin-E Synthases/chemistry , Algorithms , Amino Acid Motifs , Humans , Models, Molecular , Nuclear Magnetic Resonance, Biomolecular , Protein Conformation, alpha-Helical , Protein Interaction Domains and Motifs , Thermodynamics
15.
Int J Biol Macromol ; 88: 624-32, 2016 Jul.
Article in English | MEDLINE | ID: mdl-27012893

ABSTRACT

Microsomal prostaglandin E synthase-1 (mPGES-1) is a membrane protein which plays crucial role in arachidonic acid metabolism, in the catalysis of PGH2 to PGE2. It is a potential drug target involved in variety of human cancers and inflammatory disorders. In the present study we made an attempt to identify crucial amino acid residues involved in the effective binding of its inhibitors at the active site. Molecular docking and Structure Activity Relationship (SAR) studies were performed. In the present study 127 inhibitors having significant variability in parent scaffold were considered. The results clearly indicated that in the GSH and PGH2 binding site Arg70, Arg73, Asn74, Glu77, His113, Tyr117, Arg126, Ser127, Tyr130, Thr131 and Ala138 consistently form crucial interactions with inhibitors of different classes/scaffolds. These findings are consistent with that of existing reports on the active site residues pivotal at mPGES-1 active site. Further analysis suggested that out of all important amino acid residues identified; Arg73, Asn74, His113, Tyr117, Arg126, Ser127, Tyr130, Thr131 and Ala138 play a crucial role in hydrogen and π-π interactions. The identified amino acid residues can act as target sites for the design and development of drug candidates against mPGES-1.


Subject(s)
Enzyme Inhibitors/chemistry , Prostaglandin-E Synthases/antagonists & inhibitors , Prostaglandin-E Synthases/chemistry , Structure-Activity Relationship , Amino Acids/chemistry , Amino Acids/metabolism , Arachidonic Acid/chemistry , Arachidonic Acid/metabolism , Binding Sites/drug effects , Catalytic Domain , Dinoprostone/chemistry , Dinoprostone/metabolism , Enzyme Inhibitors/therapeutic use , Humans , Molecular Docking Simulation , Prostaglandin H2/chemistry , Prostaglandin H2/metabolism , Prostaglandin-E Synthases/metabolism
16.
J Med Chem ; 59(13): 5970-86, 2016 07 14.
Article in English | MEDLINE | ID: mdl-26791385

ABSTRACT

Microsomal prostaglandin E2 synthase (mPGES)-1 is responsible for the massive prostaglandin E2 (PGE2) formation during inflammation. Increasing evidence reveals mPGES-1 inhibitors as a safe alternative to nonsteroidal anti-inflammatory drugs. The first selective mPGES-1 inhibitors recently entered clinical trials. Major challenges for drug development have been the high plasma protein binding of lead structures, interspecies discrepancies, nuisance inhibition, sophisticated enzyme assays, and limited structural information about the mPGES-1 inhibitor binding site. Since most of these drawbacks could be solved during the past few years, we are standing at the threshold of a new era of mPGES-1-targeting anti-inflammatory drugs. This perspective introduces mPGES-1 as a key player within the network of eicosanoid biosynthesis and summarizes our current understanding of its structure and mechanism. Moreover, we present high-throughput and in silico screening techniques and discuss the structure-activity relationship and pharmacological potential of major mPGES-1 inhibitor classes in light of recent insights from pharmacophore models and cocrystallization studies.


Subject(s)
Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/pharmacology , Dinoprostone/immunology , Drug Design , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Prostaglandin-E Synthases/antagonists & inhibitors , Animals , Drug Evaluation, Preclinical/methods , High-Throughput Screening Assays/methods , Humans , Inflammation/drug therapy , Inflammation/immunology , Models, Molecular , Prostaglandin-E Synthases/chemistry , Prostaglandin-E Synthases/immunology , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...