Network


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

Hotspot


Dive into the research topics where Jean-Didier Maréchal is active.

Publication


Featured researches published by Jean-Didier Maréchal.


Journal of Biological Chemistry | 2005

Why is quinidine an inhibitor of cytochrome P450 2D6? The role of key active site residues in quinidine binding

Lesley A. McLaughlin; Mark J. I. Paine; Carol A. Kemp; Jean-Didier Maréchal; Jack U. Flanagan; Clive J. Ward; Michael J. Sutcliffe; Gordon C. K. Roberts; C. Roland Wolf

We have previously shown that Phe120, Glu216, and Asp301 in the active site of cytochrome P450 2D6 (CYP2D6) play a key role in substrate recognition by this important drug-metabolizing enzyme (Paine, M. J., McLaughlin, L. A., Flanagan, J. U., Kemp, C. A., Sutcliffe, M. J., Roberts, G. C., and Wolf, C. R. (2003) J. Biol. Chem. 278, 4021–4027 and Flanagan, J. U., Maréchal, J.-D., Ward, R., Kemp, C. A., McLaughlin, L. A., Sutcliffe, M. J., Roberts, G. C., Paine, M. J., and Wolf, C. R. (2004) Biochem. J. 380, 353–360). We have now examined the effect of mutations of these residues on interactions of the enzyme with the prototypical CYP2D6 inhibitor, quinidine. Abolition of the negative charge at either or both residues 216 and 301 decreased quinidine inhibition of bufuralol 1′-hydroxylation and dextromethorphan O-demethylation by at least 100-fold. The apparent dissociation constants (Kd) for quinidine binding to the wild-type enzyme and the E216D and D301E mutants were 0.25–0.50 μm. The amide substitution of Glu216 or Asp301 resulted in 30–64-fold increases in the Kd for quinidine. The double mutant E216Q/D301Q showed the largest decrease in quinidine affinity, with a Kd of 65 μm. Alanine substitution of Phe120, Phe481,or Phe483 had only a minor effect on the inhibition of bufuralol 1′-hydroxylation and dextromethorphan O-demethylation and on binding. In contrast to the wild-type enzyme, a number of the mutants studied were found to be able to metabolize quinidine. E216F produced O-demethylated quinidine, and F120A and E216Q/D301Q produced both O-demethylated quinidine and 3-hydroxyquinidine metabolites. Homology modeling and molecular docking were used to predict the modes of quinidine binding to the wild-type and mutant enzymes; these were able to rationalize the experimental observations.


FEBS Letters | 2009

Targeting STAT1 by myricetin and delphinidin provides efficient protection of the heart from ischemia/reperfusion-induced injury.

Tiziano M. Scarabelli; Sofia Mariotto; Safwat Abdel-Azeim; Kazuo Shoji; Elena Darra; Anastasis Stephanou; Carol Chen-Scarabelli; Jean-Didier Maréchal; Richard A. Knight; Anna Rosa Ciampa; Louis D. Saravolatz; Alessandra Carcereri de Prati; Zhaokan Yuan; Elisabetta Cavalieri; Marta Menegazzi; David S. Latchman; Cosimo Pizza; David Perahia; Hisanori Suzuki

Flavonoids exhibit a variety of beneficial effects in cardiovascular diseases. Although their therapeutic properties have been attributed mainly to their antioxidant action, they have additional protective mechanisms such as inhibition of signal transducer and activator of transcription 1 (STAT1) activation. Here, we have investigated the cardioprotective mechanisms of strong antioxidant flavonoids such as quercetin, myricetin and delphinidin. Although all of them protect the heart from ischemia/reperfusion‐injury, myricetin and delphinidin exert a more pronounced protective action than quercetin by their capacity to inhibit STAT1 activation. Biochemical and computer modeling analysis indicated the direct interaction between STAT1 and flavonoids with anti‐STAT1 activity.


Inorganica Chimica Acta | 2000

Amine conformational change and spin conversion induced by metal-assisted ligand oxidation: from the seven-coordinate iron(II)–TPAA complex to the two oxidized iron(II)–(py)3tren isomers. Characterization, crystal structures, and density functional study

Irène Morgenstern-Badarau; François Lambert; Jean Philippe Renault; Michèle Cesario; Jean-Didier Maréchal; Feliu Maseras

Abstract The tripodal heptadentate ligand TPAA (TPAA=tris[ N -(2-pyridylmethyl)-2-aminoethyl]amine) is found to undergo an iron(II)-assisted oxidative dehydrogenation with its three amine functions oxidized into three imine groups, giving the tripodal potentially heptadentate ligand (py) 3 tren ((py) 3 tren=tris[ N -(2-pyridylmethyl)-2-iminoethyl]amine). This oxidative process induces structural changes and spin conversion in the three identified iron(II) complexes, Fe–TPAA complex ( 1 ), Fe–py3tren complexes ( 2 ) and ( 3 ). X-ray crystallographic studies revealed differences in the coordination geometry of the bridging nitrogen atom and shifts in the coordination number from seven to six. Complex 1 is seven-coordinate and is characterized by a pyramidal environment of the tripodal centered nitrogen and a short N tripodal Fe distance equal to 2.504 A. Complex 3 is hexa-coordinate with a planar environment of the centered nitrogen located at 3.435 A from iron(II) and therefore not bound to the iron. Complex 2 which is an isomeric form of complex 3 is found to be a structural intermediate between complexes 2 and 3 with a pseudo-coordination of six as shown by the N tripodal Fe distance equal to 2.754 A. Electronic changes were recognized from NMR studies in solution and EPR and SQUID measurements of susceptibilities in solid state. Complex 1 is high-spin with the S =2 state characterized by a g factor equal to 2.25. Complex 3 is low-spin but its isomer, the intermediate complex 2 exhibits a temperature-dependent spin conversion from the S =2 high-spin form at room temperature to a lower spin that we provisionally identify as a S =1 intermediate spin form. DFT Becke3LYP calculations were carried out on the two isomeric complexes 2 and 3 . The planar complex was found to be 3.2 kcal mol −1 more stable than the pyramidal isomer, in agreement with the experiment.


ChemBioChem | 2012

Incorporation of manganese complexes into xylanase: new artificial metalloenzymes for enantioselective epoxidation.

Mathieu Allard; Claude Dupont; Victor Muñoz Robles; Nicolas Doucet; Agustí Lledós; Jean-Didier Maréchal; Agathe Urvoas; Jean-Pierre Mahy; Rémy Ricoux

Here we report the best artificial metalloenzyme to date for the selective oxidation of aromatic alkenes; it was obtained by noncovalent insertion of MnIII‐meso‐tetrakis(p‐carboxyphenyl)porphyrin [Mn(TpCPP), 1‐Mn] into a host protein, xylanase 10A from Streptomyces lividans (Xln10A). Two metallic complexes—N,N′‐ethylene bis(2‐hydroxybenzylimine)‐5,5′‐dicarboxylic acid MnIII [(Mn‐salen), 2‐Mn] and 1‐Mn—were associated with Xln10A, and the two hybrid biocatalysts were characterised by UV–visible spectroscopy, circular dichroism and molecular modelling. Only the artificial metalloenzyme based on 1‐Mn and Xln10A was studied for its catalytic properties in the oxidation of various substituted styrene derivatives by KHSO5: after optimisation, the 1‐Mn‐Xln10A artificial metalloenzyme was able to catalyse the oxidation of para‐methoxystyrene by KHSO5 with a 16 % yield and the best enantioselectivity (80 % in favour of the R isomer) ever reported for an artificial metalloenzyme.


Journal of the American Chemical Society | 2011

Three dimensional models of Cu(2+)-Aβ(1-16) complexes from computational approaches.

Jorge Alí-Torres; Jean-Didier Maréchal; Luis Rodríguez-Santiago; Mariona Sodupe

Elucidation of the coordination of metal ions to Aβ is essential to understand their role in its aggregation and to rationally design new chelators with potential therapeutic applications in Alzheimer disease. Because of that, in the last 10 years several studies have focused their attention in determining the coordination properties of Cu(2+) interacting with Aβ. However, more important than characterizing the first coordination sphere of the metal is the determination of the whole Cu(2+)-Aβ structure. In this study, we combine homology modeling (HM) techniques with quantum mechanics based approaches (QM) to determine plausible three-dimensional models for Cu(2+)-Aβ(1-16) with three histidines in their coordination sphere. We considered both ε and δ coordination of histidines 6, 13, and 14 as well as the coordination of different possible candidates containing oxygen as fourth ligand (Asp1, Glu3, Asp7, Glu11, and CO(Ala2)). Among the 32 models that enclose COO(-), the lowest energy structures correspond to [O(E3),N(δ)(H6),N(ε)(H13),N(ε)(H14)] (1), [O(E3),N(δ)(H6),N(δ)(H13),N(δ)(H14)] (2), and [O(D7),N(ε)(H6),N(δ)(H13),N(δ)(H14)] (3). The most stable model containing CO(Ala2) as fourth ligand in the Cu(2+) coordination sphere is [O(c)(A2),N(ε)(H6),N(δ)(H13),N(ε)(H14)] (4). An estimation of the relative stability between Glu3 (1) and CO(Ala2) (4) coordinated complexes seems to indicate that the preference for the latter coordination may be due to solvent effects. The present results also show the relationship between the peptidic and metallic moieties in defining the overall geometry of the complex and illustrate that the final stability of the complexes results from a balance between the metal coordination site and amyloid folding upon complexation.


Archives of Biochemistry and Biophysics | 2008

Insight into the apoptosis-inducing action of α-bisabolol towards malignant tumor cells: Involvement of lipid rafts and Bid

Elena Darra; Safwat Abdel-Azeim; Anna Manara; Kazuo Shoji; Jean-Didier Maréchal; Sofia Mariotto; Elisabetta Cavalieri; Luigi Perbellini; Cosimo Pizza; David Perahia; Massimo Crimi; Hisanori Suzuki

In a precedent report we showed that alpha-bisabolol, a sesquiterpene present widely in the plant kingdom, exerts a rapid and efficient apoptosis-inducing action selectively towards human and murine malignant glioblastoma cell lines through mitochondrial damage. The present study extends these data demonstrating the apoptosis-inducing action of alpha-bisabolol towards highly malignant human pancreatic carcinoma cell lines without affecting human fibroblast viability. The present study further shows the preferential incorporation of alpha-bisabolol to transformed cells through lipid rafts on plasma membranes and, thereafter, direct interaction between alpha-bisabolol and Bid protein, one of pro-apoptotic Bcl-2 family proteins, analyzed either by Surface Plasmon Resonance method or by intrinsic fluorescence measurement. Notions that lipid rafts are rich in plasma membranes of transformed cells and that Bid, richly present in lipid rafts, is deeply involved in lipid transport make highly credible the hypothesis that the molecular mechanism of alpha-bisabolol action may include its capacity to interact with Bid protein.


Drug Metabolism and Disposition | 2006

In silico and in vitro screening for inhibition of cytochrome P450 CYP3A4 by comedications commonly used by patients with cancer

Jean-Didier Maréchal; Jinglei Yu; Simon Brown; Iouri Kapelioukh; Elaine M. Rankin; C. Roland Wolf; Gordon C. K. Roberts; Mark J. I. Paine; Michael J. Sutcliffe

Cytochrome P450 3A4 (CYP3A4) is the major enzyme responsible for phase I drug metabolism of many anticancer agents. It is also a major route for metabolism of many drugs used by patients to treat the symptoms caused by cancer and its treatment as well as their other illnesses, for example, cardiovascular disease. To assess the ability to inhibit CYP3A4 of drugs most commonly used by our patients during cancer therapy, we have made in silico predictions based on the crystal structures of CYP3A4. From this set of 33 common comedicated drugs, 10 were predicted to be inhibitors of CYP3A4, with the antidiarrheal drug loperamide predicted to be the most potent. There was significant correlation (r2 = 0.75–0.66) between predicted affinity and our measured IC50 values, and loperamide was confirmed as a potent inhibitor (IC50 of 0.050 ± 0.006 μM). Active site docking studies predicted an orientation of loperamide consistent with formation of the major (N-demethylated) metabolite, where it interacts with the phenylalanine cluster and Arg-212 and Glu-374; experimental evidence for the latter interaction comes from the ∼12-fold increase in KM for loperamide observed for the Glu-374-Gln mutant. The commonly prescribed drugs loperamide, amitriptyline, diltiazem, domperidone, lansoprazole, omeprazole, and simvastatin were identified by our in silico and in vitro screens as relatively potent inhibitors of CYP3A4 that have the potential to interact with cytotoxic agents to cause adverse effects, highlighting the likelihood of drug-drug interactions affecting chemotherapy treatment.


Journal of Physical Chemistry A | 2008

How do azoles inhibit cytochrome P450 enzymes? A density functional study

Philip R. Balding; Cristina S. Porro; Kirsty J. McLean; Michael J. Sutcliffe; Jean-Didier Maréchal; Andrew W. Munro; Sam P. de Visser

To examine how azole inhibitors interact with the heme active site of the cytochrome P450 enzymes, we have performed a series of density functional theory studies on azole binding. These are the first density functional studies on azole interactions with a heme center and give fundamental insight into how azoles inhibit the catalytic function of P450 enzymes. Since azoles come in many varieties, we tested three typical azole motifs representing a broad range of azole and azole-type inhibitors: methylimidazolate, methyltriazolate, and pyridine. These structural motifs represent typical azoles, such as econazole, fluconazole, and metyrapone. The calculations show that azole binding is a stepwise mechanism whereby first the water molecule from the resting state of P450 is released from the sixth binding site of the heme to create a pentacoordinated active site followed by coordination of the azole nitrogen to the heme iron. This process leads to the breaking of a hydrogen bond between the resting state water molecule and the approaching inhibitor molecule. Although, formally, the water molecule is released in the first step of the reaction mechanism and a pentacoordinated heme is created, this does not lead to an observed spin state crossing. Thus, we show that release of a water molecule from the resting state of P450 enzymes to create a pentacoordinated heme will lead to a doublet to quartet spin state crossing at an Fe-OH(2) distance of approximately 3.0 A, while the azole substitution process takes place at shorter distances. Azoles bind heme with significantly stronger binding energies than a water molecule, so that these inhibitors block the catalytic cycle of the enzyme and prevent oxygen binding and the catalysis of substrate oxidation. Perturbations within the active site (e.g., a polarized environment) have little effect on the relative energies of azole binding. Studies with an extra hydrogen-bonded ethanol molecule in the model, mimicking the active site of the CYP121 P450, show that the resting state and azole binding structures are close in energy, which may lead to chemical equilibrium between the two structures, as indeed observed with recent protein structural studies that have demonstrated two distinct azole binding mechanisms to P450 heme.


Drug Metabolism and Disposition | 2008

Multiple Substrate Binding by Cytochrome P450 3A4: Estimation of the Number of Bound Substrate Molecules

Yury Kapelyukh; Mark J. I. Paine; Jean-Didier Maréchal; Michael J. Sutcliffe; C. Roland Wolf; Gordon C. K. Roberts

Cytochrome P450 3A4, a major drug-metabolizing enzyme in man, is well known to show non-Michaelis-Menten steady-state kinetics for a number of substrates, indicating that more than one substrate can bind to the enzyme simultaneously, but it has proved difficult to obtain reliable estimates of exactly how many substrate molecules can bind. We have used a simple method involving studies of the effect of large inhibitors on the Hill coefficient to provide improved estimates of substrate stoichiometry from simple steady-state kinetics. Using a panel of eight inhibitors, we show that at least four molecules of the widely used CYP3A4 substrate 7-benzyloxyquinoline can bind simultaneously to the enzyme. Computational docking studies show that this is consistent with the recently reported crystal structures of the enzyme. In the case of midazolam, which shows simple Michaelis-Menten kinetics, the inhibitor effects demonstrate that two molecules must bind simultaneously, consistent with earlier evidence, whereas for diltiazem, the experiments provide no evidence for the binding of more than one molecule. The consequences of this “inhibitor-induced cooperativity” for the prediction of pharmacokinetics and drug-drug interactions are discussed.


Journal of Physical Chemistry B | 2010

Insights into the Mechanism of Binding of Arachidonic Acid to Mammalian 15-Lipoxygenases

Lea Toledo; Laura Masgrau; Jean-Didier Maréchal; José M. Lluch; Àngels González-Lafont

Mammalian 15-lipoxygenases (15-LOs) are key pharmaceutical targets under strong investigation because of their implication in atherosclerosis and cancer. Here, we present an atomic-level study of the binding modes of arachidonic acid (AA) to rabbit reticulocyte 15-LO, with a particular insight into the 15-LO:AA complexes consistent with known catalytic activity. We take into account both ligand and protein flexibility, by combining protein-ligand docking techniques and molecular dynamics simulations. We have also performed in silico mutagenesis. Our results are in agreement with previous mutagenesis data, in particular with the importance of Arg403 on AA binding. Interestingly, our results also reveal a central role of Arg403 in the conformational change of the alpha2-helix observed upon ligand binding. That induced-fit effect could give a possible framework for the molecular explanation of the known allosteric effect and questions the suitability of the inhibitor-bound crystal structure for accepting AA. Accounting for these dynamical considerations might improve the drug design process.

Collaboration


Dive into the Jean-Didier Maréchal's collaboration.

Top Co-Authors

Avatar

Agustí Lledós

Autonomous University of Barcelona

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

David Perahia

École normale supérieure de Cachan

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Elisabeth Ortega-Carrasco

Autonomous University of Barcelona

View shared research outputs
Top Co-Authors

Avatar

Rémy Ricoux

University of Paris-Sud

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Mark J. I. Paine

Liverpool School of Tropical Medicine

View shared research outputs
Top Co-Authors

Avatar

Giuseppe Sciortino

Autonomous University of Barcelona

View shared research outputs
Researchain Logo
Decentralizing Knowledge