Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Protein Sci ; 33(3): e4844, 2023 Nov 27.
Article in English | MEDLINE | ID: mdl-38009704

ABSTRACT

Aminoacyl-tRNA synthetases (aaRSs) establish the genetic code. Each aaRS covalently links a given canonical amino acid to a cognate set of tRNA isoacceptors. Glycyl tRNA aminoacylation is unusual in that it is catalyzed by different aaRSs in different lineages of the Tree of Life. We have investigated the phylogenetic distribution and evolutionary history of bacterial glycyl tRNA synthetase (bacGlyRS). This enzyme is found in early diverging bacterial phyla such as Firmicutes, Acidobacteria, and Proteobacteria, but not in archaea or eukarya. We observe relationships between each of six domains of bacGlyRS and six domains of four different RNA-modifying proteins. Component domains of bacGlyRS show common ancestry with (i) the catalytic domain of class II tRNA synthetases; (ii) the HD domain of the bacterial RNase Y; (iii) the body and tail domains of the archaeal CCA-adding enzyme; (iv) the anti-codon binding domain of the arginyl tRNA synthetase; and (v) a previously unrecognized domain that we call ATL (Ancient tRNA latch). The ATL domain has been found thus far only in bacGlyRS and in the universal alanyl tRNA synthetase (uniAlaRS). Further, the catalytic domain of bacGlyRS is more closely related to the catalytic domain of uniAlaRS than to any other aminoacyl tRNA synthetase. The combined results suggest that the ATL and catalytic domains of these two enzymes are ancestral to bacGlyRS and uniAlaRS, which emerged from common protein ancestors by bricolage, stepwise accumulation of protein domains, before the last universal common ancestor of life.

2.
Molecules ; 28(22)2023 Nov 10.
Article in English | MEDLINE | ID: mdl-38005242

ABSTRACT

Lysosome-targeting chimeras (LYTACs) have recently been developed to facilitate the lysosomal degradation of specific extracellular and transmembrane molecular targets. However, the LYTAC particles described to date are based on glycopeptide conjugates, which are difficult to prepare and produce on a large scale. Here, we report on the development of pure protein LYTACs based on the non-glycosylated IGF2 peptides, which can be readily produced in virtually any facility capable of monoclonal antibody production. These chimeras utilize the IGF2R/CI-M6PR pathway for lysosomal shuttling and, in our illustrative example, target programmed death ligand 1 (PD-L1), eliciting physiological effects analogous to immune checkpoint blockade. Results from in vitro assays significantly exceed the effects of anti-PD-L1 antibodies alone.


Subject(s)
Antibodies, Monoclonal , Peptides , Peptides/chemistry , Antibodies, Monoclonal/metabolism , Glycopeptides/metabolism , Membrane Proteins/metabolism , Lysosomes/metabolism
3.
J Physiol ; 601(9): 1655-1673, 2023 05.
Article in English | MEDLINE | ID: mdl-36625071

ABSTRACT

The Transient Receptor Potential Vanilloid 4 (TRPV4) channel has been shown to function in many physiological and pathophysiological processes. Despite abundant information on its importance in physiology, very few endogenous agonists for this channel have been described, and very few underlying mechanisms for its activation have been clarified. TRPV4 is expressed by several types of cells, such as vascular endothelial, and skin and lung epithelial cells, where it plays pivotal roles in their function. In the present study, we show that TRPV4 is activated by lysophosphatidic acid (LPA) in both endogenous and heterologous expression systems, pinpointing this molecule as one of the few known endogenous agonists for TRPV4. Importantly, LPA is a bioactive glycerophospholipid, relevant in several physiological conditions, including inflammation and vascular function, where TRPV4 has also been found to be essential. Here we also provide mechanistic details of the activation of TRPV4 by LPA and another glycerophospholipid, lysophosphatidylcholine (LPC), and show that LPA directly interacts with both the N- and C-terminal regions of TRPV4 to activate this channel. Moreover, we show that LPC activates TRPV4 by producing an open state with a different single-channel conductance to that observed with LPA. Our data suggest that the activation of TRPV4 can be finely tuned in response to different endogenous lipids, highlighting this phenomenon as a regulator of cell and organismal physiology. KEY POINTS: The Transient Receptor Potential Vaniloid (TRPV) 4 ion channel is a widely distributed protein with important roles in normal and disease physiology for which few endogenous ligands are known. TRPV4 is activated by a bioactive lipid, lysophosphatidic acid (LPA) 18:1, in a dose-dependent manner, in both a primary and a heterologous expression system. Activation of TRPV4 by LPA18:1 requires residues in the N- and C-termini of the ion channel. Single-channel recordings show that TRPV4 is activated with a decreased current amplitude (conductance) in the presence of lysophosphatidylcholine (LPC) 18:1, while LPA18:1 and GSK101 activate the channel with a larger single-channel amplitude. Distinct single-channel amplitudes produced by LPA18:1 and LPC18:1 could differentially modulate the responses of the cells expressing TRPV4 under different physiological conditions.


Subject(s)
Transient Receptor Potential Channels , TRPV Cation Channels/metabolism , Lysophosphatidylcholines/pharmacology , Lysophospholipids/pharmacology
4.
Molecules ; 27(7)2022 Apr 06.
Article in English | MEDLINE | ID: mdl-35408761

ABSTRACT

The SARS-CoV-2 virus, since its appearance in 2019, has caused millions of cases and deaths. To date, there is no effective treatment or a vaccine that is fully protective. Despite the efforts made by governments and health institutions around the globe to control its propagation, the evolution of the virus has accelerated, diverging into hundreds of variants. However, not all of them are variants of concern (VoC's). VoC's have appeared in different regions and throughout the two years of the pandemic they have spread around the world. Specifically, in South America, the gamma variant (previously known as P.1) appeared in early 2021, bringing with it a second wave of infections. This variant contains the N501Y, E484K and K417T mutations in the receptor binding domain (RBD) of the spike protein. Although these mutations have been described experimentally, there is still no clarity regarding their role in the stabilization of the complex with the human angiotensin converting enzyme 2 (hACE-2) receptor. In this article we dissect the influence of mutations on the interaction with the hACE-2 receptor using molecular dynamics and estimations of binding affinity through a screened version of the molecular mechanics Poisson Boltzmann surface area (MM-PBSA) and interaction entropy. Our results indicate that mutations E484K and K417T compensate each other in terms of binding affinity, while the mutation N501Y promotes a more convoluted effect. This effect consists in the adoption of a cis configuration in the backbone of residue Y495 within the RBD, which in turn promotes polar interactions with the hACE-2 receptor. These results not only correlate with experimental observations and complement previous knowledge, but also expose new features associated with the specific contribution of concerned mutations. Additionally, we propose a recipe to assess the residue-specific contribution to the interaction entropy.


Subject(s)
Angiotensin-Converting Enzyme 2 , COVID-19 , SARS-CoV-2 , Angiotensin-Converting Enzyme 2/genetics , Humans , Molecular Dynamics Simulation , Mutation , Protein Binding/genetics , SARS-CoV-2/genetics , Spike Glycoprotein, Coronavirus/metabolism
5.
Proteins ; 88(9): 1169-1179, 2020 09.
Article in English | MEDLINE | ID: mdl-32112578

ABSTRACT

Internal structure similarity in proteins can be observed at the domain and subdomain levels. From an evolutionary perspective, structurally similar elements may arise divergently by gene duplication and fusion events but may also be the product of convergent evolution under physicochemical constraints. The characterization of proteins that contain repeated structural elements has implications for many fields of protein science including protein domain evolution, structure classification, structure prediction, and protein engineering. FiRES (Find Repeated Elements in Structure) is an algorithm that relies on a topology-independent structure alignment method to identify repeating elements in protein structure. FiRES was tested against two hand curated databases of protein repeats: MALIDUP, for very divergent duplicated domains; and RepeatsDB for short tandem repeats. The performance of FiRES was compared to that of lalign, RADAR, HHrepID, CE-symm, ReUPred, and Swelfe. FiRES was the method that most accurately detected proteins either with duplicated domains (accuracy = 0.86) or with multiple repeated units (accuracy = 0.92). FiRES is a new methodology for the discovery of proteins containing structurally similar elements. The FiRES web server is publicly available at http://fires.ifc.unam.mx. The scripts, results, and benchmarks from this study can be downloaded from https://github.com/Claualvarez/fires.


Subject(s)
Algorithms , Proteins/chemistry , Software , Structural Homology, Protein , Amino Acid Sequence , Benchmarking , Databases, Protein , Evolution, Molecular , Gene Duplication , Protein Structure, Secondary
6.
Nucleic Acids Res ; 47(12): 6425-6438, 2019 07 09.
Article in English | MEDLINE | ID: mdl-30997498

ABSTRACT

Ribonucleoprotein (RNP) complexes and RNA-processing enzymes are attractive targets for antibiotic development owing to their central roles in microbial physiology. For many of these complexes, comprehensive strategies to identify inhibitors are either lacking or suffer from substantial technical limitations. Here, we describe an activity-binding-structure platform for bacterial ribonuclease P (RNase P), an essential RNP ribozyme involved in 5' tRNA processing. A novel, real-time fluorescence-based assay was used to monitor RNase P activity and rapidly identify inhibitors using a mini-helix and a pre-tRNA-like bipartite substrate. Using the mini-helix substrate, we screened a library comprising 2560 compounds. Initial hits were then validated using pre-tRNA and the pre-tRNA-like substrate, which ultimately verified four compounds as inhibitors. Biolayer interferometry-based binding assays and molecular dynamics simulations were then used to characterize the interactions between each validated inhibitor and the P protein, P RNA and pre-tRNA. X-ray crystallographic studies subsequently elucidated the structure of the P protein bound to the most promising hit, purpurin, and revealed how this inhibitor adversely affects tRNA 5' leader binding. This integrated platform affords improved structure-function studies of RNA processing enzymes and facilitates the discovery of novel regulators or inhibitors.


Subject(s)
Anthraquinones/pharmacology , Drug Evaluation, Preclinical , Enzyme Inhibitors/pharmacology , Ribonuclease P/antagonists & inhibitors , Anthraquinones/chemistry , Anthraquinones/metabolism , Binding Sites , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , Fluorescent Dyes , Fluorometry , Hematoxylin/analogs & derivatives , Hematoxylin/chemistry , Hematoxylin/metabolism , Hematoxylin/pharmacology , Molecular Dynamics Simulation , RNA Precursors/metabolism , RNA Processing, Post-Transcriptional , RNA, Transfer/metabolism , Ribonuclease P/chemistry , Ribonuclease P/metabolism , Small Molecule Libraries
7.
Biochemistry ; 58(12): 1679-1688, 2019 03 26.
Article in English | MEDLINE | ID: mdl-30848586

ABSTRACT

The molecular determinants of substrate specificity and selectivity in the proprotein convertase (PC) family of proteases are poorly understood. Here we demonstrate that the natural serpin family inhibitor, serpin B8, is a specific and selective inhibitor of furin relative to the other PCs of the constitutive protein secretion pathway, PC4, PC5, PACE4, and PC7 (PC4-PC7, respectively), and identify reactive-site (P6-P5' residues) and exosite elements of the serpin that contribute to this specificity and selectivity through studies of chimeras of serpin B8 and α1PDX, an engineered serpin inhibitor of furin. Kinetic studies revealed that the specificity and selectivity of the serpin chimeras for inhibiting PCs were determined by P6-P5 and P3-P2 residue-dependent recognition of the P4Arg-X-X-P1Arg PC consensus sequence and exosite-dependent recognition of the reactive loop P2' residue of the chimeras by the PCs. Both productive and nonproductive binding of the chimeras to PC4-PC7 but not to furin contributed to a decreased specificity for inhibiting PC4-PC7 and an increased selectivity for inhibiting furin. Molecular dynamics simulations suggested that nonproductive binding of the chimeras to the PCs was correlated with a greater conformational variability of the catalytic sites of PC4-PC7 relative to that of furin. Our findings suggest a new approach for designing selective inhibitors of PCs using α1PDX as a scaffold, as evidenced by our ability to engineer highly specific and selective inhibitors of furin and PC4-PC7.


Subject(s)
Furin/chemistry , Serpins/chemistry , alpha 1-Antitrypsin/chemistry , Catalytic Domain , Enzyme Assays , Furin/antagonists & inhibitors , Humans , Ligands , Molecular Dynamics Simulation , Protein Engineering , Serpins/genetics , Substrate Specificity , alpha 1-Antitrypsin/genetics
8.
Proc Natl Acad Sci U S A ; 115(7): E1437-E1445, 2018 02 13.
Article in English | MEDLINE | ID: mdl-29382749

ABSTRACT

Dipeptidyl peptidases 8 and 9 are intracellular N-terminal dipeptidyl peptidases (preferentially postproline) associated with pathophysiological roles in immune response and cancer biology. While the DPP family member DPP4 is extensively characterized in molecular terms as a validated therapeutic target of type II diabetes, experimental 3D structures and ligand-/substrate-binding modes of DPP8 and DPP9 have not been reported. In this study we describe crystal and molecular structures of human DPP8 (2.5 Å) and DPP9 (3.0 Å) unliganded and complexed with a noncanonical substrate and a small molecule inhibitor, respectively. Similar to DPP4, DPP8 and DPP9 molecules consist of one ß-propeller and α/ß hydrolase domain, forming a functional homodimer. However, they differ extensively in the ligand binding site structure. In intriguing contrast to DPP4, where liganded and unliganded forms are closely similar, ligand binding to DPP8/9 induces an extensive rearrangement at the active site through a disorder-order transition of a 26-residue loop segment, which partially folds into an α-helix (R-helix), including R160/133, a key residue for substrate binding. As vestiges of this helix are also seen in one of the copies of the unliganded form, conformational selection may contributes to ligand binding. Molecular dynamics simulations support increased flexibility of the R-helix in the unliganded state. Consistently, enzyme kinetics assays reveal a cooperative allosteric mechanism. DPP8 and DPP9 are closely similar and display few opportunities for targeted ligand design. However, extensive differences from DPP4 provide multiple cues for specific inhibitor design and development of the DPP family members as therapeutic targets or antitargets.


Subject(s)
Dipeptidases/chemistry , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/chemistry , Homeostasis/physiology , Protein Conformation , Binding Sites , Catalytic Domain , Crystallography, X-Ray , Dipeptidases/metabolism , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/metabolism , Humans , Molecular Structure , Protein Domains
9.
Proc Natl Acad Sci U S A ; 113(40): 11196-11201, 2016 10 04.
Article in English | MEDLINE | ID: mdl-27647913

ABSTRACT

Proprotein convertases (PCs) are highly specific proteases required for the proteolytic modification of many secreted proteins. An unbalanced activity of these enzymes is connected to pathologies like cancer, atherosclerosis, hypercholesterolaemia, and infectious diseases. Novel protein crystallographic structures of the prototypical PC family member furin in different functional states were determined to 1.8-2.0 Å. These, together with biochemical data and modeling by molecular dynamics calculations, suggest essential elements underlying its unusually high substrate specificity. Furin shows a complex activation mechanism and exists in at least four defined states: (i) the "off state," incompatible with substrate binding as seen in the unliganded enzyme; (ii) the active "on state" seen in inhibitor-bound furin; and the respective (iii) calcium-free and (iv) calcium-bound forms. The transition from the off to the on state is triggered by ligand binding at subsites S1 to S4 and appears to underlie the preferential recognition of the four-residue sequence motif of furin. The molecular dynamics simulations of the four structural states reflect the experimental observations in general and provide approximations of the respective stabilities. Ligation by calcium at the PC-specific binding site II influences the active-site geometry and determines the rotamer state of the oxyanion hole-forming Asn295, and thus adds a second level of the activity modulation of furin. The described crystal forms and the observations of different defined functional states may foster the development of new tools and strategies for pharmacological intervention targeting furin.


Subject(s)
Furin/chemistry , Furin/metabolism , Calcium/metabolism , Catalytic Domain , Crystallography, X-Ray , Furin/antagonists & inhibitors , Humans , Ligands , Molecular Dynamics Simulation , Principal Component Analysis , Protein Conformation , Static Electricity , Structural Homology, Protein , Structure-Activity Relationship , Substrate Specificity
10.
J Biol Chem ; 291(28): 14430-46, 2016 Jul 08.
Article in English | MEDLINE | ID: mdl-27226617

ABSTRACT

Glycyl tRNA synthetase (GlyRS) provides a unique case among class II aminoacyl tRNA synthetases, with two clearly widespread types of enzymes: a dimeric (α2) species present in some bacteria, archaea, and eukaryotes; and a heterotetrameric form (α2ß2) present in most bacteria. Although the differences between both types of GlyRS at the anticodon binding domain level are evident, the extent and implications of the variations in the catalytic domain have not been described, and it is unclear whether the mechanism of amino acid recognition is also dissimilar. Here, we show that the α-subunit of the α2ß2 GlyRS from the bacterium Aquifex aeolicus is able to perform the first step of the aminoacylation reaction, which involves the activation of the amino acid with ATP. The crystal structure of the α-subunit in the complex with an analog of glycyl adenylate at 2.8 Å resolution presents a conformational arrangement that properly positions the cognate amino acid. This work shows that glycine is recognized by a subset of different residues in the two types of GlyRS. A structural and sequence analysis of class II catalytic domains shows that bacterial GlyRS is closely related to alanyl tRNA synthetase, which led us to define a new subclassification of these ancient enzymes and to propose an evolutionary path of α2ß2 GlyRS, convergent with α2 GlyRS and divergent from AlaRS, thus providing a possible explanation for the puzzling existence of two proteins sharing the same fold and function but not a common ancestor.


Subject(s)
Glycine-tRNA Ligase/chemistry , Phylogeny , Bacteria/enzymology , Crystallography, X-Ray , Models, Molecular , Protein Conformation
11.
Chemistry ; 21(47): 16887-94, 2015 Nov 16.
Article in English | MEDLINE | ID: mdl-26541354

ABSTRACT

The recent discovery of c-Jun NH2-terminal kinase JNK1 suppression by natural quercetagetin (1) is a promising lead for the development of novel anticancer agents. Using both X-ray structure and docking analyses we predicted that 5'-hydroxy- (2) and 5'-hydroxymethyl-quercetagetin (3) would inhibit JNK1 more actively than the parent compound 1. Notably, our drug design was based on the active enzyme-ligand complex as opposed to the enzyme's relatively open apo structure. In this paper we test our theoretical predictions, aided by docking-model experiments, and report the first synthesis and biological evaluation of quercetagetin analogues 2 and 3. As calculated, both compounds strongly suppress JNK1 activity. The IC50 values were determined to be 3.4 µM and 12.2 µM, respectively, which shows that 2 surpasses the potency of the parent compound 1 (IC50 =4.6 µM). Compound 2 was also shown to suppress matrix metalloproteinase-1 expression with high specificity after UV irradiation.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Chromones/chemistry , Chromones/pharmacology , Mitogen-Activated Protein Kinase 8/chemistry , Biological Factors , Chromones/metabolism , Drug Design , Flavones , Humans , Inhibitory Concentration 50 , Mitogen-Activated Protein Kinase 8/metabolism , Ultraviolet Rays
12.
Elife ; 42015 Feb 23.
Article in English | MEDLINE | ID: mdl-25706231

ABSTRACT

Actin filament dynamics govern many key physiological processes from cell motility to tissue morphogenesis. A central feature of actin dynamics is the capacity of filaments to polymerize and depolymerize at their ends in response to cellular conditions. It is currently thought that filament kinetics can be described by a single rate constant for each end. In this study, using direct visualization of single actin filament elongation, we show that actin polymerization kinetics at both filament ends are strongly influenced by the binding of proteins to the lateral filament surface. We also show that the pointed-end has a non-elongating state that dominates the observed filament kinetic asymmetry. Estimates of flexibility as well as effects on fragmentation and growth suggest that the observed kinetic diversity arises from structural alteration. Tuning elongation kinetics by exploiting the malleability of the filament structure may be a ubiquitous mechanism to generate a rich variety of cellular actin dynamics.


Subject(s)
Actin Cytoskeleton/metabolism , Carrier Proteins/metabolism , Microscopy, Fluorescence/methods , Polymerization , Actin Cytoskeleton/ultrastructure , Actinin/metabolism , Actins/metabolism , Adenosine Triphosphate/metabolism , Algorithms , Animals , Cell Adhesion Molecules/metabolism , Chickens , Filamins/metabolism , Kinetics , Microfilament Proteins/metabolism , Microscopy, Electron , Models, Biological , Monte Carlo Method , Myosins/metabolism , Phosphoproteins/metabolism , Protein Binding , Sf9 Cells , Spodoptera
13.
Proc Natl Acad Sci U S A ; 111(26): 9479-84, 2014 Jul 01.
Article in English | MEDLINE | ID: mdl-24979800

ABSTRACT

Two clusters of configurations of the main proteolytic subunit ß5 were identified by principal component analysis of crystal structures of the yeast proteasome core particle (yCP). The apo-cluster encompasses unliganded species and complexes with nonpeptidic ligands, and the pep-cluster comprises complexes with peptidic ligands. The murine constitutive CP structures conform to the yeast system, with the apo-form settled in the apo-cluster and the PR-957 (a peptidic ligand) complex in the pep-cluster. In striking contrast, the murine immune CP classifies into the pep-cluster in both the apo and the PR-957-liganded species. The two clusters differ essentially by multiple small structural changes and a domain motion enabling enclosure of the peptidic ligand and formation of specific hydrogen bonds in the pep-cluster. The immune CP species is in optimal peptide binding configuration also in its apo form. This favors productive ligand binding and may help to explain the generally increased functional activity of the immunoproteasome. Molecular dynamics simulations of the representative murine species are consistent with the experimentally observed configurations. A comparison of all 28 subunits of the unliganded species with the peptidic liganded forms demonstrates a greatly enhanced plasticity of ß5 and suggests specific signaling pathways to other subunits.


Subject(s)
Models, Molecular , Proteasome Endopeptidase Complex/chemistry , Proteasome Endopeptidase Complex/immunology , Signal Transduction/genetics , Animals , Crystallization , Ligands , Mice , Molecular Dynamics Simulation , Molecular Structure , Oligopeptides/metabolism , Principal Component Analysis , Protein Binding , Protein Conformation , Species Specificity , Yeasts
14.
J Chem Inf Model ; 54(5): 1401-11, 2014 May 27.
Article in English | MEDLINE | ID: mdl-24796936

ABSTRACT

In this study, we propose a novel approach to evaluate virtual screening (VS) experiments based on the analysis of docking output data. This approach, which we refer to as docking data feature analysis (DDFA), consists of two steps. First, a set of features derived from the docking output data is computed and assigned to each molecule in the virtually screened library. Second, an artificial neural network (ANN) analyzes the molecule's docking features and estimates its activity. Given the simple architecture of the ANN, DDFA can be easily adapted to deal with information from several docking programs simultaneously. We tested our approach on the Directory of Useful Decoys (DUD), a well-established and highly accepted VS benchmark. Outstanding results were obtained by DDFA not only in comparison with the conventional rankings of the docking programs used in this work but also with respect to other methods found in the literature. Our approach performs with similar good results as the best available methods, which, however, also require substantially more computing time, economic resources, and/or expert intervention. Taken together, DDFA represents an automatic and highly attractive methodology for VS.


Subject(s)
Drug Evaluation, Preclinical/methods , Molecular Docking Simulation/methods , Area Under Curve , Neural Networks, Computer , ROC Curve , User-Computer Interface
15.
Structure ; 21(12): 2143-51, 2013 Dec 03.
Article in English | MEDLINE | ID: mdl-24207125

ABSTRACT

Reactivation of p53 by release of the functional protein from its inhibition by MDM2 provides an efficient, nongenotoxic approach to a wide variety of cancers. We present the cocrystal structures of two complexes of MDM2 with inhibitors based on 6-chloroindole scaffolds. Both molecules bound to a distinct conformational state of MDM2 with nM-µM affinities. In contrast to other structurally characterized antagonists, which mimic three amino acids of p53 (Phe19, Trp23, and Leu26), the compounds induced an additional hydrophobic pocket on the MDM2 surface and unveiled a four-point binding mode. The enlarged interaction interface of the inhibitors resulted in extension of small molecules binding toward the "lid" segment of MDM2 (residues 19-23)--a nascent element that interferes with p53 binding. As supported by protein engineering and molecular dynamics studies, employing these unstable elements of MDM2 provides an efficient and yet unexplored alternative in development of MDM2-p53 association inhibitors.


Subject(s)
Dipeptides/chemistry , Hydroxamic Acids/chemistry , Proto-Oncogene Proteins c-mdm2/chemistry , Tryptophan/analogs & derivatives , Tumor Suppressor Protein p53/chemistry , Crystallography, X-Ray , Humans , Molecular Dynamics Simulation , Molecular Structure , Nuclear Magnetic Resonance, Biomolecular , Protein Interaction Domains and Motifs , Proto-Oncogene Proteins c-mdm2/antagonists & inhibitors , Tryptophan/chemistry , Tumor Suppressor Protein p53/antagonists & inhibitors
16.
J Mol Biol ; 425(2): 411-23, 2013 Jan 23.
Article in English | MEDLINE | ID: mdl-23142567

ABSTRACT

c-Jun NH2-terminal kinases (JNKs) and phosphatidylinositol 3-kinase (PI3-K) play critical roles in chronic diseases such as cancer, type II diabetes, and obesity. We describe here the binding of quercetagetin (3,3',4',5,6,7-hydroxyflavone), related flavonoids, and SP600125 to JNK1 and PI3-K by ATP-competitive and immobilized metal ion affinity-based fluorescence polarization assays and measure the effect of quercetagetin on JNK1 and PI3-K activities. Quercetagetin attenuated the phosphorylation of c-Jun and AKT, suppressed AP-1 and NF-κB promoter activities, and also reduced cell transformation. It attenuated tumor incidence and reduced tumor volumes in a two-stage skin carcinogenesis mouse model. Our crystallographic structure determination data show that quercetagetin binds to the ATP-binding site of JNK1. Notably, the interaction between Lys55, Asp169, and Glu73 of JNK1 and the catechol moiety of quercetagetin reorients the N-terminal lobe of JNK1, thereby improving compatibility of the ligand with its binding site. The results of a theoretical docking study suggest a binding mode of PI3-K with the hydroxyl groups of the catechol moiety forming hydrogen bonds with the side chains of Asp964 and Asp841 in the p110γ catalytic subunit. These interactions could contribute to the high inhibitory activity of quercetagetin against PI3-K. Our study suggests the potential use of quercetagetin in the prevention or therapy of cancer and other chronic diseases.


Subject(s)
Adenosine Triphosphate/metabolism , Cell Transformation, Neoplastic/drug effects , Chromones/chemistry , Chromones/pharmacology , Mitogen-Activated Protein Kinase 8/antagonists & inhibitors , Skin Neoplasms/prevention & control , Adaptor Proteins, Signal Transducing/metabolism , Animals , Anthracenes/pharmacology , Binding, Competitive , Crystallography, X-Ray , Flavones , Humans , Mice , Mice, Hairless , Mitogen-Activated Protein Kinase 8/metabolism , Models, Molecular , Molecular Dynamics Simulation , NF-kappa B/genetics , NF-kappa B/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation/drug effects , Protein Conformation , Proto-Oncogene Proteins c-akt/metabolism , Skin Neoplasms/etiology , Transcription Factor AP-1/metabolism , Ultraviolet Rays/adverse effects
SELECTION OF CITATIONS
SEARCH DETAIL
...