Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 64
Filtrar
1.
Int J Mol Sci ; 23(3)2022 Feb 08.
Artigo em Inglês | MEDLINE | ID: mdl-35163835

RESUMO

Histamine levels in the human brain are controlled by rather peculiar metabolic pathways. In the first step, histamine is enzymatically methylated at its imidazole Nτ atom, and the produced N-methylhistamine undergoes an oxidative deamination catalyzed by monoamine oxidase B (MAO-B), as is common with other monoaminergic neurotransmitters and neuromodulators of the central nervous system. The fact that histamine requires such a conversion prior to oxidative deamination is intriguing since MAO-B is known to be relatively promiscuous towards monoaminergic substrates; its in-vitro oxidation of N-methylhistamine is about 10 times faster than that for histamine, yet this rather subtle difference appears to be governing the decomposition pathway. This work clarifies the MAO-B selectivity toward histamine and N-methylhistamine by multiscale simulations of the rate-limiting hydride abstraction step for both compounds in the gas phase, in aqueous solution, and in the enzyme, using the established empirical valence bond methodology, assisted by gas-phase density functional theory (DFT) calculations. The computed barriers are in very good agreement with experimental kinetic data, especially for relative trends among systems, thereby reproducing the observed MAO-B selectivity. Simulations clearly demonstrate that solvation effects govern the reactivity, both in aqueous solution as well as in the enzyme although with an opposing effect on the free energy barrier. In the aqueous solution, the transition-state structure involving histamine is better solvated than its methylated analog, leading to a lower barrier for histamine oxidation. In the enzyme, the higher hydrophobicity of N-methylhistamine results in a decreased number of water molecules at the active side, leading to decreased dielectric shielding of the preorganized catalytic electrostatic environment provided by the enzyme. This renders the catalytic environment more efficient for N-methylhistamine, giving rise to a lower barrier relative to histamine. In addition, the transition state involving N-methylhistamine appears to be stabilized by the surrounding nonpolar residues to a larger extent than with unsubstituted histamine, contributing to a lower barrier with the former.


Assuntos
Histamina/metabolismo , Metilistaminas/metabolismo , Monoaminoxidase/metabolismo , Encéfalo/metabolismo , Simulação por Computador , Teoria da Densidade Funcional , Histamina/química , Humanos , Interações Hidrofóbicas e Hidrofílicas , Metilistaminas/química , Estrutura Molecular , Oxirredução , Especificidade por Substrato
2.
Molecules ; 27(19)2022 Oct 09.
Artigo em Inglês | MEDLINE | ID: mdl-36235246

RESUMO

Monoamine oxidases (MAOs) are an important group of enzymes involved in the degradation of neurotransmitters and their imbalanced mode of action may lead to the development of various neuropsychiatric or neurodegenerative disorders. In this work, we report the results of an in-depth computational study in which we performed a static and a dynamic analysis of a series of substituted ß-carboline natural products, found mainly in roasted coffee and tobacco smoke, that bind to the active site of the MAO-A isoform. By applying molecular docking in conjunction with structure-based pharmacophores and molecular dynamics simulations coupled with dynamic pharmacophores, we extensively investigated the geometric aspects of MAO-A binding. To gain insight into the energetics of binding, we used the linear interaction energy (LIE) method and determined the key anchors that allow productive ß-carboline binding to MAO-A. The results presented herein could be applied in the rational structure-based design and optimization of ß-carbolines towards preclinical candidates that would target the MAO-A enzyme and would be applicable especially in the treatment of mental disorders such as depression.


Assuntos
Inibidores da Monoaminoxidase , Poluição por Fumaça de Tabaco , Carbolinas/farmacologia , Café , Humanos , Simulação de Acoplamento Molecular , Monoaminoxidase/metabolismo , Inibidores da Monoaminoxidase/química , Inibidores da Monoaminoxidase/farmacologia , Relação Estrutura-Atividade
3.
Phys Chem Chem Phys ; 23(46): 26459-26467, 2021 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-34806105

RESUMO

The origin of the immense catalytic power of enzymes remains one of the biggest unresolved questions in biochemistry, with electrostatics being one of the main contenders. Herein, we report results that not only confirm that electrostatics is the driving force behind enzyme catalysis, but also that it is capable of tuning subtle differences in the catalytic performance between structurally similar enzymes, as demonstrated using the example of isoenzymes, monoamine oxidases A and B. Using our own computationally efficient multiscale model [A. Prah, et al., ACS Catal., 2019, 9, 1231] we analyzed the rate-limiting step of the reaction between phenylethylamine and both isoenzymes and deduced that the electrostatic environment provided by isoenzyme B has a perceivably higher catalytic influence on all the considered parameters of the reaction (energy barrier, charge transfer, dipole moment, and HOMO-LUMO gap). This is in full agreement with the available experimental kinetic data and with our own simulations of the reaction in question. In-depth analysis of individual amino acid contributions of both isoenzymes to the barrier (based on the interaction between the electric field provided by the enzyme and the dipole moment of the reacting moiety) shows that the majority of the difference between the isoenzymes can be attributed to a small number of sizable differences between the aligned amino acid pairs, whereas in most of the pairs the difference in contribution to the barrier is vanishingly small. These results suggest that electrostatics largely controls the substrate selectivity of enzymes and validates our approach as being capable of discerning fine nuances in the selectivity of structurally related isoenzymes.


Assuntos
Teoria da Densidade Funcional , Monoaminoxidase/metabolismo , Biocatálise , Isoenzimas/química , Isoenzimas/metabolismo , Monoaminoxidase/química , Eletricidade Estática
4.
Phys Chem Chem Phys ; 22(13): 6838-6847, 2020 Apr 06.
Artigo em Inglês | MEDLINE | ID: mdl-32191250

RESUMO

The kinetic isotope effect (KIE) is arguably the most established experimental observable reflecting nuclear quantum effects in enzymatic reactions. The role of nuclear quantum effects in enzymes is rather intriguing and has long been a source of profound investigations. Herein, we present a computational study of monoamine oxidase A (MAO A) enzyme and its substrate phenylethylamine, focusing on the impact of nuclear quantum effects on the reaction free energy barrier. Two distinct schemes of quantization of nuclear motion were used, one being the established Quantum Classical Path (QCP) approach, and the other our own code for quantum treatment along the selected nuclear coordinate (hydrogen transfer coordinate) which reasonably mimics the reaction coordinate. In excellent agreement with the experimental value of 8.5 ± 0.3, H/D KIE was computed to 8.66, corresponding to the D-H barrier difference of 1.28 kcal mol-1. The magnitude of KIE implies that nuclear quantum effects probably have only a minor role in the reaction, which is in accordance with the features of potentials computed along the reaction coordinate and with the pertinent energy levels and wavefunctions. The computed H/D KIE for the same reaction in aqueous solution and in the gas phase was fairly similar to the one in the enzyme, suggesting that the role of tunneling in the catalytic function of MAO A is insignificant. The agreement between the computed and observed KIE supported by analysis of nuclear quantum effects implicitly validates the assumed hydride transfer reaction mechanism.


Assuntos
Simulação por Computador , Monoaminoxidase/metabolismo , Fenetilaminas/metabolismo , Catálise , Isótopos/química , Cinética , Teoria Quântica
5.
Int J Mol Sci ; 21(17)2020 Aug 26.
Artigo em Inglês | MEDLINE | ID: mdl-32858935

RESUMO

Monoamine oxidases (MAOs) catalyze the degradation of a very broad range of biogenic and dietary amines including many neurotransmitters in the brain, whose imbalance is extensively linked with the biochemical pathology of various neurological disorders, and are, accordingly, used as primary pharmacological targets to treat these debilitating cognitive diseases. Still, despite this practical significance, the precise molecular mechanism underlying the irreversible MAO inhibition with clinically used propargylamine inhibitors rasagiline and selegiline is still not unambiguously determined, which hinders the rational design of improved inhibitors devoid of side effects current drugs are experiencing. To address this challenge, we present empirical valence bond QM/MM simulations of the rate-limiting step of the MAO inhibition involving the hydride anion transfer from the inhibitor α-carbon onto the N5 atom of the flavin adenin dinucleotide (FAD) cofactor. The proposed mechanism is strongly supported by the obtained free energy profiles, which confirm a higher reactivity of selegiline over rasagiline, while the calculated difference in the activation Gibbs energies of ΔΔG‡ = 3.1 kcal mol-1 is found to be in very good agreement with that from the measured literature kinact values that predict a 1.7 kcal mol-1 higher selegiline reactivity. Given the similarity with the hydride transfer mechanism during the MAO catalytic activity, these results verify that both rasagiline and selegiline are mechanism-based irreversible inhibitors and offer guidelines in designing new and improved inhibitors, which are all clinically employed in treating a variety of neuropsychiatric and neurodegenerative conditions.


Assuntos
Indanos/farmacologia , Inibidores da Monoaminoxidase/farmacologia , Monoaminoxidase/metabolismo , Selegilina/farmacologia , Domínio Catalítico/efeitos dos fármacos , Simulação por Computador , Transferência de Energia , Flavina-Adenina Dinucleotídeo/metabolismo , Humanos , Indanos/química , Modelos Moleculares , Estrutura Molecular , Monoaminoxidase/química , Inibidores da Monoaminoxidase/química , Conformação Proteica , Selegilina/química
6.
Molecules ; 25(24)2020 Dec 18.
Artigo em Inglês | MEDLINE | ID: mdl-33353215

RESUMO

We used a range of computational techniques to reveal an increased histamine affinity for its H2 receptor upon deuteration, which was interpreted through altered hydrogen bonding interactions within the receptor and the aqueous environment preceding the binding. Molecular docking identified the area between third and fifth transmembrane α-helices as the likely binding pocket for several histamine poses, with the most favorable binding energy of -7.4 kcal mol-1 closely matching the experimental value of -5.9 kcal mol-1. The subsequent molecular dynamics simulation and MM-GBSA analysis recognized Asp98 as the most dominant residue, accounting for 40% of the total binding energy, established through a persistent hydrogen bonding with the histamine -NH3+ group, the latter further held in place through the N-H∙∙∙O hydrogen bonding with Tyr250. Unlike earlier literature proposals, the important role of Thr190 is not evident in hydrogen bonds through its -OH group, but rather in the C-H∙∙∙π contacts with the imidazole ring, while its former moiety is constantly engaged in the hydrogen bonding with Asp186. Lastly, quantum-chemical calculations within the receptor cluster model and utilizing the empirical quantization of the ionizable X-H bonds (X = N, O, S), supported the deuteration-induced affinity increase, with the calculated difference in the binding free energy of -0.85 kcal mol-1, being in excellent agreement with an experimental value of -0.75 kcal mol-1, thus confirming the relevance of hydrogen bonding for the H2 receptor activation.


Assuntos
Teoria da Densidade Funcional , Histamina/química , Simulação de Acoplamento Molecular , Simulação de Dinâmica Molecular , Receptores Histamínicos H2/química , Sítios de Ligação , Ligação de Hidrogênio , Estrutura Molecular , Termodinâmica
7.
Molecules ; 24(23)2019 Nov 28.
Artigo em Inglês | MEDLINE | ID: mdl-31795294

RESUMO

Monoamine oxidase A (MAO A) is a well-known enzyme responsible for the oxidative deamination of several important monoaminergic neurotransmitters. The rate-limiting step of amine decomposition is hydride anion transfer from the substrate α-CH2 group to the N5 atom of the flavin cofactor moiety. In this work, we focus on MAO A-catalyzed benzylamine decomposition in order to elucidate nuclear quantum effects through the calculation of the hydrogen/deuterium (H/D) kinetic isotope effect. The rate-limiting step of the reaction was simulated using a multiscale approach at the empirical valence bond (EVB) level. We applied path integral quantization using the quantum classical path method (QCP) for the substrate benzylamine as well as the MAO cofactor flavin adenine dinucleotide. The calculated H/D kinetic isotope effect of 6.5 ± 1.4 is in reasonable agreement with the available experimental values.


Assuntos
Benzilaminas/química , Deutério/química , Hidrogênio/química , Monoaminoxidase/química , Algoritmos , Catálise , Cinética , Modelos Moleculares , Modelos Teóricos , Oxirredução
8.
Phys Chem Chem Phys ; 20(6): 4181-4188, 2018 Feb 07.
Artigo em Inglês | MEDLINE | ID: mdl-29360121

RESUMO

This work investigates the Y326I point mutation effect on the kinetics of oxidative deamination of phenylethylamine (PEA) catalyzed by the monoamine oxidase B (MAO B) enzyme. PEA is a neuromodulator capable of affecting the plasticity of the brain and is responsible for the mood enhancing effect caused by physical exercise. Due to a similar functionality, PEA is often regarded as an endogenous amphetamine. The rate limiting step of the deamination was simulated at the multiscale level, employing the Empirical Valence Bond approach for the quantum treatment of the involved valence states, whereas the environment (solvated protein) was represented with a classical force field. A comparison of the reaction free energy profiles delivered by simulation of the reaction in the wild type MAO B and its Y326I mutant yields an increase in the barrier by 1.06 kcal mol-1 upon mutation, corresponding to a roughly 6-fold decrease in the reaction rate. This is in excellent agreement with the experimental kinetic studies. Inspection of simulation trajectories reveals possible sources of the point mutation effect, namely vanishing favorable electrostatic interactions between PEA and a Tyr326 side chain and an increased amount of water molecules at the active site due to the replacement of tyrosine by a less spacious isoleucine residue, thereby increasing the dielectric shielding of the catalytic environment provided by the enzyme.


Assuntos
Anfetamina/metabolismo , Monoaminoxidase/metabolismo , Anfetamina/química , Sítios de Ligação , Biocatálise , Domínio Catalítico , Desaminação , Cinética , Monoaminoxidase/química , Monoaminoxidase/genética , Fenetilaminas/química , Fenetilaminas/metabolismo , Mutação Puntual , Especificidade por Substrato
9.
Proteins ; 85(12): 2170-2178, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28836294

RESUMO

This work scrutinizes kinetics of decomposition of adrenaline catalyzed by monoamine oxidase (MAO) A and B enzymes, a process controlling the levels of adrenaline in the central nervous system and other tissues. Experimental kinetic data for MAO A and B catalyzed decomposition of adrenaline are reported only in the form of the maximum reaction rate. Therefore, we estimated the experimental free energy barriers form the kinetic data of closely related systems using regression method, as was done in our previous study. By using multiscale simulation on the Empirical Valence Bond (EVB) level, we studied the chemical reactivity of the MAO A catalyzed decomposition of adrenaline and we obtained a value of activation free energy of 17.3 ± 0.4 kcal/mol. The corresponding value for MAO B is 15.7 ± 0.7 kcal/mol. Both values are in good agreement with the estimated experimental barriers of 16.6 and 16.0 kcal/mol for MAO A and MAO B, respectively. The fact that we reproduced the kinetic data and preferential catalytic effect of MAO B over MAO A gives additional support to the validity of the proposed hydride transfer mechanism. Furthermore, we demonstrate that adrenaline is preferably involved in the reaction in a neutral rather than in a protonated form due to considerably higher barriers computed for the protonated adrenaline substrate. The results are discussed in the context of chemical mechanism of MAO enzymes and possible applications of multiscale simulation to rationalize the effects of MAO activity on adrenaline level.


Assuntos
Epinefrina/química , Flavinas/química , Monoaminoxidase/química , Prótons , Domínio Catalítico , Humanos , Hidrólise , Isoenzimas/química , Cinética , Simulação de Acoplamento Molecular , Termodinâmica
10.
Blood ; 124(26): 3896-904, 2014 Dec 18.
Artigo em Inglês | MEDLINE | ID: mdl-25359991

RESUMO

Myeloid differentiation 88 (MyD88) is the key signaling adapter of Toll-like and interleukin-1 receptors. Recurrent lymphoma-associated mutations, particularly Leu265Pro (L265P), within the MyD88 Toll/interleukin-1 receptor (TIR) domain sustain lymphoma cell survival due to constitutive nuclear factor κB signaling. We found that mutated TIR domains displayed an intrinsic propensity for augmented oligomerization and spontaneous formation of cytosolic Myddosome aggregates in lymphoma cell lines, mimicking the effect of dimerized TIR domains. Blocking of MyD88 oligomerization induced apoptosis. The L265P TIR domain can recruit the endogenous wild-type MyD88 for oligomer formation and hyperactivity. Molecular dynamics simulations and analysis of additional mutations suggest that constitutive activity is caused by allosteric oligomerization.


Assuntos
Linfoma/genética , Mutação , Fator 88 de Diferenciação Mieloide/genética , Sítio Alostérico , Linhagem Celular Tumoral , Células HEK293 , Heterozigoto , Humanos , Inflamação , Luminescência , Microscopia Confocal , Simulação de Dinâmica Molecular , Fenótipo , Reação em Cadeia da Polimerase , Estrutura Terciária de Proteína , RNA Interferente Pequeno/metabolismo , Receptores de Interleucina-1/metabolismo , Transdução de Sinais
11.
Phys Chem Chem Phys ; 18(19): 13346-56, 2016 05 21.
Artigo em Inglês | MEDLINE | ID: mdl-27121693

RESUMO

The I335Y point mutation effect on the kinetics of phenylethylamine decomposition catalyzed by monoamine oxidase A was elucidated by means of molecular simulation. The established empirical valence bond methodology was used in conjunction with the free energy perturbation sampling technique and a classical force field representing the state of reactants and products. The methodology allows for the simulation of chemical reactions, in the present case the breaking of the α-C-H bond in a phenylethylamine substrate and the subsequent hydrogen transfer to the flavin cofactor, resulting in the formation of the N-H bond on flavin. The empirical parameters were calibrated against the experimental data for the simulated reaction in a wild type protein and then used for the calculation of the reaction free energy profile in the I335Y mutant. In very good agreement with the measured kinetic data, mutation increases the free energy barrier for the rate limiting step by slightly more than 1 kcal mol(-1) and consequently decreases the rate constant by about an order of magnitude. The magnitude of the computed effect slightly varies with simulation settings, but always remains in reasonable agreement with the experiment. Analysis of trajectories reveals a major change in the interaction between phenyl rings of the substrate and the neighboring Phe352 residue upon the I335Y mutation due to the increased local polarity, leading to an attenuated quadrupole interaction between the rings and destabilization of the transition state. Additionally, the increased local polarity in the mutant allows for a larger number of water molecules to be present near the active site, effectively shielding the catalytic effect of the enzyme and contributing to the increased barrier.


Assuntos
Monoaminoxidase/química , Fenetilaminas/química , Mutação Puntual , Catálise , Domínio Catalítico , Ativação Enzimática , Flavinas/química , Cinética , Modelos Moleculares , Oxirredução
12.
Proteins ; 82(12): 3347-55, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25220264

RESUMO

Monoamine oxidases (MAOs) A and B are flavoenzymes responsible for the metabolism of biogenic amines such as dopamine, serotonin and noradrenaline. In this work, we present a comprehensive study of the rate-limiting step of dopamine degradation by MAO B, which consists in the hydride transfer from the methylene group of the substrate to the flavin moiety of the FAD prosthetic group. This article builds on our previous quantum chemical study of the same reaction using a cluster model (Vianello et al., Eur J Org Chem 2012; 7057), but now considering the full dimensionality of the hydrated enzyme with extensive configurational sampling. We show that MAO B is specifically tuned to catalyze the hydride transfer step from the substrate to the flavin moiety of the FAD prosthetic group and that it lowers the activation barrier by 12.3 kcal mol⁻¹ compared to the same reaction in aqueous solution, a rate enhancement of more than nine orders of magnitude. Taking into account the deprotonation of the substrate prior to the hydride transfer reaction, the activation barrier in the enzyme is calculated to be 16.1 kcal mol⁻¹, in excellent agreement with the experimental value of 16.5 kcal mol⁻¹. Additionally, we demonstrate that the protonation state of the active site residue Lys296 does not have an influence on the hydride transfer reaction.


Assuntos
Dopamina/metabolismo , Modelos Moleculares , Monoaminoxidase/metabolismo , Biocatálise , Domínio Catalítico , Análise por Conglomerados , Bases de Dados de Proteínas , Dopamina/química , Transferência de Energia , Flavina-Adenina Dinucleotídeo/química , Flavina-Adenina Dinucleotídeo/metabolismo , Humanos , Cinética , Lisina/química , Simulação de Dinâmica Molecular , Monoaminoxidase/química , Conformação Proteica , Teoria Quântica
13.
J Chem Inf Model ; 54(4): 1254-67, 2014 Apr 28.
Artigo em Inglês | MEDLINE | ID: mdl-24628082

RESUMO

Predicting the endocrine disruption potential of compounds is a daunting but essential task. Here we report a new tool for this purpose that we have termed Endocrine Disruptome. It is a free and simple-to-use Web service that runs on an open source platform called Docking interface for Target Systems (DoTS). The molecular docking is handled via AutoDock Vina. Compounds are docked to 18 integrated and well-validated crystal structures of 14 different human nuclear receptors: androgen receptor; estrogen receptors α and ß; glucocorticoid receptor; liver X receptors α and ß; mineralocorticoid receptor; peroxisome proliferator activated receptors α, ß/δ, and γ; progesterone receptor; retinoid X receptor α; and thyroid receptors α and ß. Endocrine Disruptome is free of charge and available at http://endocrinedisruptome.ki.si.


Assuntos
Disruptores Endócrinos/toxicidade , Receptores Citoplasmáticos e Nucleares/metabolismo , Disruptores Endócrinos/metabolismo , Humanos , Simulação de Acoplamento Molecular , Ligação Proteica , Interface Usuário-Computador
14.
Biomolecules ; 14(4)2024 Mar 30.
Artigo em Inglês | MEDLINE | ID: mdl-38672440

RESUMO

This study assessed the suitability of the complementarity-determining region 2 (CDR2) of the nanobody (Nb) as a template for the derivation of nanobody-derived peptides (NDPs) targeting active-state ß2-adrenergic receptor (ß2AR) conformation. Sequences of conformationally selective Nbs favoring the agonist-occupied ß2AR were initially analyzed by the informational spectrum method (ISM). The derived NDPs in complex with ß2AR were subjected to protein-peptide docking, molecular dynamics (MD) simulations, and metadynamics-based free-energy binding calculations. Computational analyses identified a 25-amino-acid-long CDR2-NDP of Nb71, designated P4, which exhibited the following binding free-energy for the formation of the ß2AR:P4 complex (ΔG = -6.8 ± 0.8 kcal/mol or a Ki = 16.5 µM at 310 K) and mapped the ß2AR:P4 amino acid interaction network. In vitro characterization showed that P4 (i) can cross the plasma membrane, (ii) reduces the maximum isoproterenol-induced cAMP level by approximately 40% and the isoproterenol potency by up to 20-fold at micromolar concentration, (iii) has a very low affinity to interact with unstimulated ß2AR in the cAMP assay, and (iv) cannot reduce the efficacy and potency of the isoproterenol-mediated ß2AR/ß-arrestin-2 interaction in the BRET2-based recruitment assay. In summary, the CDR2-NDP, P4, binds preferentially to agonist-activated ß2AR and disrupts Gαs-mediated signaling.


Assuntos
Peptídeos , Receptores Adrenérgicos beta 2 , Anticorpos de Domínio Único , Humanos , Sequência de Aminoácidos , Regiões Determinantes de Complementaridade/química , AMP Cíclico/metabolismo , Simulação de Acoplamento Molecular , Simulação de Dinâmica Molecular , Peptídeos/química , Peptídeos/farmacologia , Peptídeos/metabolismo , Ligação Proteica , Conformação Proteica , Receptores Adrenérgicos beta 2/metabolismo , Receptores Adrenérgicos beta 2/química , Anticorpos de Domínio Único/química , Anticorpos de Domínio Único/farmacologia , Anticorpos de Domínio Único/metabolismo
15.
Chem Res Toxicol ; 26(1): 106-11, 2013 Jan 18.
Artigo em Inglês | MEDLINE | ID: mdl-23198967

RESUMO

Bisphenol A is a monomer used in the production of polycarbonate plastics, epoxy resins, and flame retardants. It is an endocrine disruptor with a variety of other effects, including genotoxicity. Oxidative metabolism of bisphenol A yields electophilic bisphenol A-3,4-quinone (BPAQ), which may cause genotoxicity. To determine the genotoxic potential of bisphenol A, the mechanism of the reaction between the BPAQ and deoxyadenosine (dA) was studied in detail. The most probable reaction pathway was determined using quantum chemical methods. Our results demonstrate that the rate limiting step is Michael addition between BPAQ and dA, the main product being the unstable N7-modified adduct that rapidly undergoes depurination. In addition, our calculations provide strong evidence for protonation of the adducts prior to depurination, which indicates pH dependence of the reaction. The calculated activation barrier for Michael addition is 28.7 kcal/mol, indicating that the reaction with dA is very slow. Comparison with the activation energy of 23.1 kcal/mol for the corresponding deoxyguanosine reaction indicates that most of the DNA damage by BPAQ will occur at the guanine site. The detoxification reactions with glutathione compete with reactions between BPAQ and DNA. The calculated free energy of activation for the reaction with glutathione is significantly lower than that for the corresponding reaction with dA. This indicates that BPAQ will preferably react with glutathione and will only react with DNA when the level of glutathione in the cell is low.


Assuntos
Compostos Benzidrílicos/química , Benzoquinonas/química , Adutos de DNA/química , Desoxiadenosinas/química , Glutationa/química , Modelos Moleculares , Fenóis/química , Teoria Quântica
16.
J Neural Transm (Vienna) ; 120(6): 875-82, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23546802

RESUMO

Density functional theory calculations were employed to investigate the nature of chemical bond formation between the flavin co-factor of the enzyme monoamine oxidase (MAO) and its irreversible acetylenic inhibitor clorgyline in its terminally deprotonated anionic form. Since MAOs regulate the level of neurotransmitters in living cells, this reaction is pharmacologically relevant for treating depression and other mood disorders. The results revealed that this pathway is associated with the activation free energy of ΔG act (#) = 17.4 kcal mol(-1), which, together with our previous results, suggests that clorgyline is intrinsically a more effective MAO inhibitor than antiparkinsonian drugs rasagiline and selegiline considering the preferred MAO isoforms in each case, thus displaying a trend in agreement with experimental data. The reaction is facilitated by the pronounced electrophilic character of the flavin moiety, due to its ability to efficiently accommodate excess negative charge from the approaching anionic inhibitor through resonance effect. The investigated mechanism was additionally validated by the inspection of the geometry of the flavin moiety in the formed adduct, which exhibit distortion from planarity consistent with experimental observations. These results offer valuable insight for mechanistic studies on other flavoenzymes and for the design of new antidepressants and antiparkinsonian drugs.


Assuntos
Clorgilina/farmacologia , Inibidores da Monoaminoxidase/química , Inibidores da Monoaminoxidase/farmacologia , Monoaminoxidase/efeitos dos fármacos , Monoaminoxidase/metabolismo , Computadores Moleculares , Humanos , Modelos Químicos
17.
Comput Biol Med ; 165: 107375, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37611421

RESUMO

BACKGROUND: The aim of the present study was to predict the time to onset and duration of action of two local anesthetics (lidocaine and bupivacaine) based on experimental dimensions of a typical nerve and experimental octanol/water partition coefficients. METHODS: We began our compilation of experimental data with a numerical solution of the Smoluchowski equation for the transfer of lidocaine and bupivacaine across the axon membrane in the region of the node of Ranvier (axolemma) and across the Schwann cell. The difference between the aqueous and lipid environments of the neuron was simulated by including the coordinate-dependent chemical potential. In the second step, the permeation rates calculated using the diffusion equation were used to solve a system of four ordinary differential equations. This approach allowed us to simulate the cellular environment for a longer time and to compare our model with pharmacokinetic properties (time to onset and duration of action) of local anesthetics from the literature. The behavior of local anesthetics under physiological conditions and in case of local acidosis was also simulated. RESULTS: We demonstrated that local anesthetics cross the axolemma in a time span of less than 1 µs. The time to onset of action, controlled by diffusion from the epineurium to an axon with a typical distance of 500 µm, was 167 s and 186 s for lidocaine and bupivacaine, respectively. The calculated half-life, which is a measure of the duration of action, was 41 min and 328 min for lidocaine and bupivacaine, respectively. CONCLUSIONS: Duration of action is controlled by the storage capacity of lipophilic compartments around the axon, which is higher for bupivacaine but lower in local acidosis. For the latter case, the literature, including textbooks, provides a misinterpretation, namely that protonated species cannot penetrate the membrane.


Assuntos
Bupivacaína , Lidocaína , Bupivacaína/farmacocinética , Lidocaína/farmacocinética , Anestésicos Locais/farmacocinética , Fibras Nervosas Mielinizadas
18.
Protein J ; 42(3): 229-238, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-37289420

RESUMO

Hydrazoic acid (HN3) and its deprotonated form azide ion (N3-) (AHA) are toxic because they inhibit the cytochrome c oxidase complex IV (CoX IV) embedded in the inner mitochondrial membrane that forms part of the enzyme complexes involved in cellular respiration. Critical to its toxicity is the inhibition of CoX IV in the central nervous system and cardiovascular system. Hydrazoic acid is an ionizable species and its affinity for membranes, and the associated permeabilities, depend on the pH values of aqueous media on both sides of the membranes. In this article, we address the permeability of AHA through the biological membrane. In order to understand the affinity of the membrane for the neutral and ionized form of azide, we measured the octanol/water partition coefficients at pH values of 2.0 and 8.0, which are 2.01 and 0.00034, respectively. Using a Parallel Artificial Membrane Permeability Assay (PAMPA) experiment, we measured the effective permeability through the membrane, which is logPe - 4.97 and - 5.26 for pH values of 7.4 and pH 8.0, respectively. Experimental permeability was used to validate theoretical permeability, which was estimated by numerically solving a Smoluchowski equation for AHA diffusion through the membrane. We demonstrated that the rate of permeation through the cell membrane of 8.46·104 s-1 is much higher than the rate of the chemical step of CoX IV inhibition by azide of 200 s-1. The results of this study show that transport through the membrane does not represent the rate-limiting step and therefore does not control the rate of CoX IV inhibition in the mitochondria. However, the observed dynamics of azide poisoning is controlled by circulatory transport that takes place on a time scale of minutes.


Assuntos
Azidas , Membranas Artificiais , Azidas/metabolismo , Membrana Celular/metabolismo , Octanóis/química , Permeabilidade , Concentração de Íons de Hidrogênio
19.
Int J Mol Sci ; 13(2): 1269-1283, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22408390

RESUMO

Genistein, daidzein, glycitein and quercetin are flavonoids present in soybean and other vegetables in high amounts. These flavonoids can be metabolically converted to more active forms, which may react with guanine in the DNA to form complexes and can lead to DNA depurination. We assumed two ultimate carcinogen forms of each of these flavonoids, diol epoxide form and diketone form. Density functional theory (DFT) and Hartree-Fock (HF) methods were used to study the reaction thermodynamics between active forms of flavonoids and DNA guanine. Solvent reaction field method of Tomasi and co-workers and the Langevin dipoles method of Florian and Warshel were used to calculate the hydration free energies. Activation free energy for each reaction was estimated using the linear free energy relation. Our calculations show that diol epoxide forms of flavonoids are more reactive than the corresponding diketone forms and are hence more likely flavonoid ultimate carcinogens. Genistein, daidzein and glycitein show comparable reactivity while quercetin is less reactive toward DNA.


Assuntos
DNA/química , Flavonoides/química , Glycine max/química , Guanina/química
20.
Comput Struct Biotechnol J ; 20: 1254-1263, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35228857

RESUMO

Although COVID-19 has been primarily associated with pneumonia, recent data show that its causative agent, the SARS-CoV-2 coronavirus, can infect many vital organs beyond the lungs, including the heart, kidneys and the brain. The literature agrees that COVID-19 is likely to have long-term mental health effects on infected individuals, which signifies a need to understand the role of the virus in the pathophysiology of brain disorders that is currently unknown and widely debated. Our docking and molecular dynamics simulations show that the affinity of the spike protein from the wild type (WT) and the South African B.1.351 (SA) variant towards MAO enzymes is comparable to that for its ACE2 receptor. This allows for the WT/SA⋅⋅⋅MAO complex formation, which changes MAO affinities for their neurotransmitter substrates, thereby impacting their metabolic conversion and misbalancing their levels. Knowing that this fine regulation is strongly linked with the etiology of various brain pathologies, these results are the first to highlight the possibility that the interference with the brain MAO catalytic activity is responsible for the increased neurodegenerative illnesses following a COVID-19 infection, thus placing a neurobiological link between these two conditions in the spotlight. Since the obtained insight suggests that a more contagious SA variant causes even larger disturbances, and with new and more problematic strains likely emerging in the near future, we firmly advise that the presented prospect of the SARS-CoV-2 induced neurological complications should not be ignored, but rather requires further clinical investigations to achieve an early diagnosis and timely therapeutic interventions.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA