Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where A. Peon is active.

Publication


Featured researches published by A. Peon.


ChemMedChem | 2010

Understanding the Key Factors that Control the Inhibition of Type II Dehydroquinase by (2R)-2- Benzyl-3-Dehydroquinic Acids.

A. Peon; José M. Otero; L. Tizon; Verónica F. V. Prazeres; Antonio L. Llamas-Saiz; Gavin C. Fox; Mark J. van Raaij; Heather K. Lamb; Alastair R. Hawkins; Federico Gago; Luis Castedo; Concepción González-Bello

The binding mode of several substrate analogues, (2R)‐2‐benzyl‐3‐dehydroquinic acids 4, which are potent reversible competitive inhibitors of type II dehydroquinase (DHQ2), the third enzyme of the shikimic acid pathway, has been investigated by structural and computational studies. The crystal structures of Mycobacterium tuberculosis and Helicobacter pylori DHQ2 in complex with one of the most potent inhibitor, p‐methoxybenzyl derivative 4 a, have been solved at 2.40 Å and 2.75 Å, respectively. This has allowed the resolution of the M. tuberculosis DHQ2 loop containing residues 20–25 for the first time. These structures show the key interactions of the aromatic ring in the active site of both enzymes and additionally reveal an important change in the conformation and flexibility of the loop that closes over substrate binding. The loop conformation and the binding mode of compounds 4 b–d has been also studied by molecular dynamics simulations, which suggest that the benzyl group of inhibitors 4 prevent appropriate orientation of the catalytic tyrosine of the loop for proton abstraction and disrupts its basicity.


Organic and Biomolecular Chemistry | 2012

Synthesis of 3-alkyl enol mimics inhibitors of type II dehydroquinase: factors influencing their inhibition potency

Beatriz Blanco; A. Sedes; A. Peon; Heather K. Lamb; Alastair R. Hawkins; Luis Castedo; Concepción González-Bello

Several 3-alkylaryl mimics of the enol intermediate in the reaction catalyzed by type II dehydroquinase were synthesized to investigate the effect on the inhibition potency of replacing the oxygen atom in the side chain by a carbon atom. The length and the rigidity of the spacer was also studied. The inhibitory properties of the reported compounds against type II dehydroquinase from Mycobacterium tuberculosis and Helicobacter pylori are also reported. The binding modes of these analogs in the active site of both enzymes were studied by molecular docking using GOLD 5.0 and dynamic simulations studies.


Biochemical Journal | 2014

Mechanistic insight into the reaction catalysed by bacterial type II dehydroquinases.

Claire Coderch; Emilio Lence; A. Peon; Heather K. Lamb; Alastair R. Hawkins; Federico Gago; Concepción González-Bello

DHQ2 (type II dehydroquinase), which is an essential enzyme in Helicobacter pylori and Mycobacterium tuberculosis and does not have any counterpart in humans, is recognized to be an attractive target for the development of new antibacterial agents. Computational and biochemical studies that help understand in atomic detail the catalytic mechanism of these bacterial enzymes are reported in the present paper. A previously unknown key role of certain conserved residues of these enzymes, as well as the structural changes responsible for triggering the release of the product from the active site, were identified. Asp89*/Asp88* from a neighbouring enzyme subunit proved to be the residue responsible for the deprotonation of the essential tyrosine to afford the catalytic tyrosinate, which triggers the enzymatic process. The essentiality of this residue is supported by results from site-directed mutagenesis. For H. pylori DHQ2, this reaction takes place through the assistance of a water molecule, whereas for M. tuberculosis DHQ2, the tyrosine is directly deprotonated by the aspartate residue. The participation of a water molecule in this deprotonation reaction is supported by solvent isotope effects and proton inventory studies. MD simulation studies provide details of the required motions for the catalytic turnover, which provides a complete overview of the catalytic cycle. The product is expelled from the active site by the essential arginine residue and after a large conformational change of a loop containing two conserved arginine residues (Arg109/Arg108 and Arg113/Arg112), which reveals a previously unknown key role for these residues. The present study highlights the key role of the aspartate residue whose blockage could be useful in the rational design of inhibitors and the mechanistic differences between both enzymes.


Biochemical Journal | 2014

Insights into substrate binding and catalysis in bacterial type I dehydroquinase

M. Maneiro; A. Peon; Emilio Lence; José M. Otero; Van Raaij Mj; Paul Thompson; Alastair R. Hawkins; Concepción González-Bello

Structural, biochemical and computational studies to study substrate binding and the role of the conserved residues of the DHQ1 (type I dehydroquinase) enzyme active site are reported in the present paper. The crystal structure of DHQ1 from Salmonella typhi in complex with (2R)-2-methyl-3-dehydroquinic acid, a substrate analogue, was solved at 1.5 Å. The present study reveals a previously unknown key role for conserved Glu46, Phe145 and Met205 and Gln236, Pro234 and Ala233 residues, with the latter three being located in the flexible substrate-covering loop. Gln236 was shown to be responsible for the folding of this loop and for the dramatic reduction of its flexibility, which triggers active site closure. Glu46 was found to be key in bringing the substrate close to the lysine/histidine catalytic pocket to initiate catalysis. The present study could be useful in the rational design of inhibitors of this challenging and recognized target for the development of novel herbicides and antimicrobial agents.


Journal of Medicinal Chemistry | 2014

Exploring the Water-Binding Pocket of the Type II Dehydroquinase Enzyme in the Structure-Based Design of Inhibitors

Beatriz Blanco; A. Sedes; A. Peon; José M. Otero; Mark J. van Raaij; Paul Thompson; Alastair R. Hawkins; Concepción González-Bello

Structural and computational studies to explore the WAT1 binding pocket in the structure-based design of inhibitors against the type II dehydroquinase (DHQ2) enzyme are reported. The crystal structures of DHQ2 from M. tuberculosis in complex with four of the reported compounds are described. The electrostatic interaction observed between the guanidinium group of the essential arginine and the carboxylate group of one of the inhibitors in the reported crystal structures supports the recently suggested role of this arginine as the residue that triggers the release of the product from the active site. The results of the structural and molecular dynamics simulation studies revealed that the inhibitory potency is favored by promoting interactions with WAT1 and the residues located within this pocket and, more importantly, by avoiding situations where the ligands occupy the WAT1 binding pocket. The new insights can be used to advantage in the structure-based design of inhibitors.


ChemMedChem | 2013

Comparative binding energy COMBINE analysis for understanding the binding determinants of type II dehydroquinase inhibitors.

A. Peon; Claire Coderch; Federico Gago; Concepción González-Bello

Herein we report comparative binding energy (COMBINE) analyses to derive quantitative structure–activity relationship (QSAR) models that help rationalize the determinants of binding affinity for inhibitors of type II dehydroquinase (DHQ2), the third enzyme of the shikimic acid pathway. Independent COMBINE models were derived for Helicobacter pylori and Mycobacterium tuberculosis DHQ2, which is an essential enzyme in both these pathogenic bacteria that has no counterpart in human cells. These studies quantify the importance of the hydrogen bonding interactions between the ligands and the water molecule involved in the DHQ2 reaction mechanism. They also highlight important differences in the ligand interactions with the interface pocket close to the active site that could provide guides for future inhibitor design.


ACS Chemical Biology | 2013

Mechanistic Basis of the Inhibition of Type II Dehydroquinase by (2S)- and (2R)-2-Benzyl-3-Dehydroquinic Acids.

Emilio Lence; L. Tizon; José M. Otero; A. Peon; Verónica F. V. Prazeres; Antonio L. Llamas-Saiz; Gavin C. Fox; Mark J. van Raaij; Heather K. Lamb; Alastair R. Hawkins; Concepción González-Bello

The structural changes caused by the substitution of the aromatic moiety in (2S)-2-benzyl-3-dehydroquinic acids and its epimers in C2 by electron-withdrawing or electron-donating groups in type II dehydroquinase enzyme from M. tuberculosis and H. pylori has been investigated by structural and computational studies. Both compounds are reversible competitive inhibitors of this enzyme, which is essential in these pathogenic bacteria. The crystal structures of M. tuberculosis and H. pylori in complex with (2S)-2-(4-methoxy)benzyl- and (2S)-2-perfluorobenzyl-3-dehydroquinic acids have been solved at 2.0, 2.3, 2.0, and 1.9 Å, respectively. The crystal structure of M. tuberculosis in complex with (2R)-2-(benzothiophen-5-yl)methyl-3-dehydroquinic acid is also reported at 1.55 Å. These crystal structures reveal key differences in the conformation of the flexible loop of the two enzymes, a difference that depends on the presence of electron-withdrawing or electron-donating groups in the aromatic moiety of the inhibitors. This loop closes over the active site after substrate binding, and its flexibility is essential for the function of the enzyme. These differences have also been investigated by molecular dynamics simulations in an effort to understand the significant inhibition potency differences observed between some of these compounds and also to obtain more information about the possible movements of the loop. These computational studies have also allowed us to identify key structural factors of the H. pylori loop that could explain its reduced flexibility in comparison to the M. tuberculosis loop, specifically by the formation of a key salt bridge between the side chains of residues Asp18 and Arg20.


ChemMedChem | 2017

Reducing the Flexibility of Type II Dehydroquinase for Inhibition: A Fragment‐Based Approach and Molecular Dynamics Study

A. Peon; Adrián Robles; Beatriz Blanco; Marino Convertino; Paul Thompson; Alastair R. Hawkins; Amedeo Caflisch; Concepción González-Bello

A multidisciplinary approach was used to identify and optimize a quinazolinedione‐based ligand that would decrease the flexibility of the substrate‐covering loop (catalytic loop) of the type II dehydroquinase from Helicobacter pylori. This enzyme, which is essential for the survival of this bacterium, is involved in the biosynthesis of aromatic amino acids. A computer‐aided fragment‐based protocol (ALTA) was first used to identify the aromatic fragments able to block the interface pocket that separates two neighboring enzyme subunits and is located at the active site entrance. Chemical modification of its non‐aromatic moiety through an olefin cross‐metathesis and Seebachs self‐reproduction of chirality synthetic principle allowed the development of a quinazolinedione derivative that disables the catalytic loop plasticity, which is essential for the enzyme′s catalytic cycle. Molecular dynamics simulations revealed that the ligand would force the catalytic loop into an inappropriate arrangement for catalysis by strong interactions with the catalytic tyrosine and by expelling the essential arginine out of the active site.


Organic and Biomolecular Chemistry | 2015

Irreversible covalent modification of type I dehydroquinase with a stable Schiff base

L. Tizon; M. Maneiro; A. Peon; José M. Otero; Emilio Lence; Sergio Poza; Mark J. van Raaij; Paul Thompson; Alastair R. Hawkins; Concepción González-Bello


Archive | 2016

Crystal structure of Mycobacterium tuberculosis Type II Dehydroquinase D88N mutant inhibited by a 3-dehydroquinic acid derivative

José M. Otero; Antonio L. Llamas-Saiz; C. Santiago; Heather K. Lamb; Alastair R. Hawkins; M. Maneiro; A. Peon; Concepción González-Bello; M.J. van Raaij

Collaboration


Dive into the A. Peon's collaboration.

Top Co-Authors

Avatar

Concepción González-Bello

University of Santiago de Compostela

View shared research outputs
Top Co-Authors

Avatar

José M. Otero

University of Santiago de Compostela

View shared research outputs
Top Co-Authors

Avatar

Antonio L. Llamas-Saiz

University of Santiago de Compostela

View shared research outputs
Top Co-Authors

Avatar

M. Maneiro

University of Santiago de Compostela

View shared research outputs
Top Co-Authors

Avatar

Emilio Lence

University of Santiago de Compostela

View shared research outputs
Top Co-Authors

Avatar

A. Sedes

University of Santiago de Compostela

View shared research outputs
Top Co-Authors

Avatar

Beatriz Blanco

University of Santiago de Compostela

View shared research outputs
Top Co-Authors

Avatar

L. Tizon

University of Santiago de Compostela

View shared research outputs
Top Co-Authors

Avatar

Mark J. van Raaij

Spanish National Research Council

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge