Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
Biophys J ; 112(8): 1645-1653, 2017 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-28445755

RESUMO

Ryanodine (Ryd) irreversibly targets ryanodine receptors (RyRs), a family of intracellular calcium release channels essential for many cellular processes ranging from muscle contraction to learning and memory. Little is known of the atomistic details about how Ryd binds to RyRs. In this study, we used all-atom molecular dynamics simulations with both enhanced and bidirectional sampling to gain direct insights into how Ryd interacts with major residues in RyRs that were experimentally determined to be critical for its binding. We found that the pyrrolic ring of Ryd displays preference for the R4892AGGG-F4921 residues in the cavity of RyR1, which explain the effects of the corresponding mutations in RyR2 in experiments. Particularly, the mutant Q4933A (or Q4863A in RyR2) critical for both the gating and Ryd binding not only has significantly less interaction with Ryd than the wild-type, but also yields more space for Ryd and water molecules in the cavity. These results describe clear binding modes of Ryd in the RyR cavity and offer structural mechanisms explaining functional data collected on RyR blockade.


Assuntos
Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo , Rianodina/metabolismo , Animais , Sítios de Ligação , Simulação de Acoplamento Molecular , Simulação de Dinâmica Molecular , Mutação , Estrutura Secundária de Proteína , Rianodina/química , Canal de Liberação de Cálcio do Receptor de Rianodina/química , Canal de Liberação de Cálcio do Receptor de Rianodina/genética , Termodinâmica , Água/química
2.
J Physiol ; 595(14): 4695-4723, 2017 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-28516454

RESUMO

KEY POINTS: This study represents a first step toward predicting mechanisms of sex-based arrhythmias that may lead to important developments in risk stratification and may inform future drug design and screening. We undertook simulations to reveal the conditions (i.e. pacing, drugs, sympathetic stimulation) required for triggering and sustaining reentrant arrhythmias. Using the recently solved cryo-EM structure for the Eag-family channel as a template, we revealed potential interactions of oestrogen with the pore loop hERG mutation (G604S). Molecular models suggest that oestrogen and dofetilide blockade can concur simultaneously in the hERG channel pore. ABSTRACT: Female sex is a risk factor for inherited and acquired long-QT associated torsade de pointes (TdP) arrhythmias, and sympathetic discharge is a major factor in triggering TdP in female long-QT syndrome patients. We used a combined experimental and computational approach to predict 'the perfect storm' of hormone concentration, IKr block and sympathetic stimulation that induces arrhythmia in females with inherited and acquired long-QT. More specifically, we developed mathematical models of acquired and inherited long-QT syndrome in male and female ventricular human myocytes by combining effects of a hormone and a hERG blocker, dofetilide, or hERG mutations. These 'male' and 'female' model myocytes and tissues then were used to predict how various sex-based differences underlie arrhythmia risk in the setting of acute sympathetic nervous system discharge. The model predicted increased risk for arrhythmia in females when acute sympathetic nervous system discharge was applied in the settings of both inherited and acquired long-QT syndrome. Females were predicted to have protection from arrhythmia induction when progesterone is high. Males were protected by the presence of testosterone. Structural modelling points towards two plausible and distinct mechanisms of oestrogen action enhancing torsadogenic effects: oestradiol interaction with hERG mutations in the pore loop containing G604 or with common TdP-related blockers in the intra-cavity binding site. Our study presents findings that constitute the first evidence linking structure to function mechanisms underlying female dominance of arousal-induced arrhythmias.


Assuntos
Nível de Alerta/fisiologia , Arritmias Cardíacas/fisiopatologia , Modelos Biológicos , Agonistas Adrenérgicos beta/farmacologia , Animais , Antiarrítmicos/farmacologia , Estradiol/farmacologia , Canais de Potássio Éter-A-Go-Go/fisiologia , Feminino , Cobaias , Isoproterenol/farmacologia , Masculino , Simulação de Acoplamento Molecular , Miócitos Cardíacos/fisiologia , Fenetilaminas/farmacologia , Caracteres Sexuais , Sulfonamidas/farmacologia
3.
Biochim Biophys Acta Proteins Proteom ; 1865(11 Pt B): 1643-1653, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28847523

RESUMO

The rapid development of experimental and computational techniques has changed fundamentally our understanding of cellular-membrane transport. The advent of powerful computers and refined force-fields for proteins, ions, and lipids has expanded the applicability of Molecular Dynamics (MD) simulations. A myriad of cellular responses is modulated through the binding of endogenous and exogenous ligands (e.g. neurotransmitters and drugs, respectively) to ion channels. Deciphering the thermodynamics and kinetics of the ligand binding processes to these membrane proteins is at the heart of modern drug development. The ever-increasing computational power has already provided insightful data on the thermodynamics and kinetics of drug-target interactions, free energies of solvation, and partitioning into lipid bilayers for drugs. This review aims to provide a brief summary about modeling approaches to map out crucial binding pathways with intermediate conformations and free-energy surfaces for drug-ion channel binding mechanisms that are responsible for multiple effects on cellular functions. We will discuss post-processing analysis of simulation-generated data, which are then transformed to kinetic models to better understand the molecular underpinning of the experimental observables under the influence of drugs or mutations in ion channels. This review highlights crucial mathematical frameworks and perspectives on bridging different well-established computational techniques to connect the dynamics and timescales from all-atom MD and free energy simulations of ion channels to the physiology of action potentials in cellular models. This article is part of a Special Issue entitled: Biophysics in Canada, edited by Lewis Kay, John Baenziger, Albert Berghuis and Peter Tieleman.


Assuntos
Canais Iônicos/química , Bicamadas Lipídicas/química , Simulação de Dinâmica Molecular , Neurotransmissores/química , Termodinâmica , Animais , Humanos
4.
Biophys J ; 108(6): 1414-1424, 2015 Mar 24.
Artigo em Inglês | MEDLINE | ID: mdl-25809254

RESUMO

Congenital and acquired (drug-induced) forms of the human long-QT syndrome are associated with alterations in Kv11.1 (hERG) channel-controlled repolarizing IKr currents of cardiac action potentials. A mandatory drug screen implemented by many countries led to a discovery of a large group of small molecules that can activate hERG currents and thus may act as potent antiarrhythmic agents. Despite significant progress in identification of channel activators, little is known about their mechanism of action. A combination of electrophysiological studies with molecular and kinetic modeling was used to examine the mechanism of a model activator (NS1643) action on the hERG channel and its L529I mutant. The L529I mutant has gating dynamics similar to that of wild-type while its response to application of NS1643 is markedly different. We propose a mechanism compatible with experiments in which the model activator binds to the closed (C3) and open states (O). We suggest that NS1643 is affecting early gating transitions, probably during movements of the voltage sensor that precede the opening of the activation gate.


Assuntos
Cresóis/farmacologia , Canais de Potássio Éter-A-Go-Go/metabolismo , Moduladores de Transporte de Membrana/farmacologia , Modelos Moleculares , Compostos de Fenilureia/farmacologia , Linhagem Celular , Canal de Potássio ERG1 , Canais de Potássio Éter-A-Go-Go/genética , Humanos , Cinética , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Mutação , Técnicas de Patch-Clamp , Transfecção
5.
Biophys J ; 108(6): 1400-1413, 2015 Mar 24.
Artigo em Inglês | MEDLINE | ID: mdl-25809253

RESUMO

Activators of hERG1 such as NS1643 are being developed for congenital/acquired long QT syndrome. Previous studies identify the neighborhood of L529 around the voltage-sensor as a putative interacting site for NS1643. With NS1643, the V1/2 of activation of L529I (-34 ± 4 mV) is similar to wild-type (WT) (-37 ± 3 mV; P > 0.05). WT and L529I showed no difference in the slope factor in the absence of NS1643 (8 ± 0 vs. 9 ± 0) but showed a difference in the presence of NS1643 (9 ± 0.3 vs. 22 ± 1; P < 0.01). Voltage-clamp-fluorimetry studies also indicated that in L529I, NS1643 reduces the voltage-sensitivity of S4 movement. To further assess mechanism of NS1643 action, mutations were made in this neighborhood. NS1643 shifts the V1/2 of activation of both K525C and K525C/L529I to hyperpolarized potentials (-131 ± 4 mV for K525C and -120 ± 21 mV for K525C/L529I). Both K525C and K525C/K529I had similar slope factors in the absence of NS1643 (18 ± 2 vs. 34 ± 5, respectively) but with NS1643, the slope factor of K525C/L529I increased from 34 ± 5 to 71 ± 10 (P < 0.01) whereas for K525C the slope factor did not change (18 ± 2 at baseline and 16 ± 2 for NS1643). At baseline, K525R had a slope factor similar to WT (9 vs. 8) but in the presence of NS1643, the slope factor of K525R was increased to 24 ± 4 vs. 9 ± 0 mV for WT (P < 0.01). Molecular modeling indicates that L529I induces a kink in the S4 voltage-sensor helix, altering a salt-bridge involving K525. Moreover, docking studies indicate that NS1643 binds to the kinked structure induced by the mutation with a higher affinity. Combining biophysical, computational, and electrophysiological evidence, a mechanistic principle governing the action of some activators of hERG1 channels is proposed.


Assuntos
Cresóis/metabolismo , Cresóis/farmacologia , Canais de Potássio Éter-A-Go-Go/metabolismo , Compostos de Fenilureia/metabolismo , Compostos de Fenilureia/farmacologia , Animais , Sítios de Ligação , Linhagem Celular , Canal de Potássio ERG1 , Canais de Potássio Éter-A-Go-Go/genética , Fluorometria , Humanos , Potenciais da Membrana/fisiologia , Simulação de Acoplamento Molecular , Simulação de Dinâmica Molecular , Mutação , Oócitos , Técnicas de Patch-Clamp , Estrutura Secundária de Proteína , Transfecção , Xenopus laevis
6.
J Mol Cell Cardiol ; 85: 71-8, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25986146

RESUMO

In Europe, ivabradine has recently been approved to treat patients with angina who have intolerance to beta blockers and/or heart failure. Ivabradine is considered to act specifically on the sinoatrial node by inhibiting the If current (the funny current) to slow automaticity. However, in vitro studies show that ivabradine prolongs phase 3 repolarization in ventricular tissue. No episodes of Torsades de Pointes have been reported in randomized clinical studies. The objective of this study is to assess whether ivabradine blocked the hERG1 current. In the present study we discovered that ivabradine prolongs action potential and blocks the hERG current over a range of concentrations overlapping with those required to block HCN4. Ivabradine produced tonic, rather than use-dependent block. The mutation Y652A significantly suppressed pharmacologic block of hERG by ivabradine. Disruption of C-type inactivation also suppressed block of hERG1 by ivabradine. Molecular docking and molecular dynamics simulations indicate that ivabradine may access the inner cavity of the hERG1 via a lipophilic route and has a well-defined binding site in the closed state of the channel. Structural organization of the binding pockets for ivabradine is discussed. Ivabradine blocks hERG and prolongs action potential duration. Our study is potentially important because it indicates the need for active post marketing surveillance of ivabradine. Importantly, proarrhythmia of a number of other drugs has only been discovered during post marketing surveillance.


Assuntos
Benzazepinas/farmacologia , Canais de Potássio Éter-A-Go-Go/antagonistas & inibidores , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização/antagonistas & inibidores , Miócitos Cardíacos/fisiologia , Bloqueadores dos Canais de Potássio/farmacologia , Potenciais de Ação , Animais , Benzazepinas/química , Sítios de Ligação , Relação Dose-Resposta a Droga , Canal de Potássio ERG1 , Canais de Potássio Éter-A-Go-Go/química , Humanos , Concentração Inibidora 50 , Ivabradina , Bicamadas Lipídicas/química , Camundongos , Simulação de Acoplamento Molecular , Miócitos Cardíacos/efeitos dos fármacos , Bloqueadores dos Canais de Potássio/química , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína
7.
Proc Natl Acad Sci U S A ; 109(39): 15722-7, 2012 Sep 25.
Artigo em Inglês | MEDLINE | ID: mdl-23019356

RESUMO

The DNA nucleotide thymidylate is synthesized by the enzyme thymidylate synthase, which catalyzes the reductive methylation of deoxyuridylate using the cofactor methylene-tetrahydrofolate (CH(2)H(4)folate). Most organisms, including humans, rely on the thyA- or TYMS-encoded classic thymidylate synthase, whereas, certain microorganisms, including all Rickettsia and other pathogens, use an alternative thyX-encoded flavin-dependent thymidylate synthase (FDTS). Although several crystal structures of FDTSs have been reported, the absence of a structure with folates limits understanding of the molecular mechanism and the scope of drug design for these enzymes. Here we present X-ray crystal structures of FDTS with several folate derivatives, which together with mutagenesis, kinetic analysis, and computer modeling shed light on the cofactor binding and function. The unique structural data will likely facilitate further elucidation of FDTSs' mechanism and the design of structure-based inhibitors as potential leads to new antimicrobial drugs.


Assuntos
Proteínas de Bactérias/química , Ácido Fólico/química , Rickettsia/enzimologia , Timidilato Sintase/química , Sítios de Ligação , Cristalografia por Raios X , Estrutura Terciária de Proteína
8.
Biochim Biophys Acta ; 1834(9): 1722-38, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23470499

RESUMO

In this work we review the application of classical and quantum-mechanical atomistic computer simulation tools to the investigation of small ligand interaction with globins. In the first part, studies of ligand migration, with its connection to kinetic association rate constants (kon), are presented. In the second part, we review studies for a variety of ligands such as O2, NO, CO, HS(-), F(-), and NO2(-) showing how the heme structure, proximal effects, and the interactions with the distal amino acids can modulate protein ligand binding. The review presents mainly results derived from our previous works on the subject, in the context of other theoretical and experimental studies performed by others. The variety and extent of the presented data yield a clear example of how computer simulation tools have, in the last decade, contributed to our deeper understanding of small ligand interactions with globins. This article is part of a Special Issue entitled: Oxygen Binding and Sensing Proteins.


Assuntos
Simulação por Computador , Globinas/química , Globinas/metabolismo , Animais , Humanos , Ligantes , Teoria Quântica
9.
J Am Chem Soc ; 134(3): 1738-45, 2012 Jan 25.
Artigo em Inglês | MEDLINE | ID: mdl-22171795

RESUMO

A significant contemporary question in enzymology involves the role of protein dynamics and hydrogen tunneling in enhancing enzyme catalyzed reactions. Here, we report a correlation between the donor-acceptor distance (DAD) distribution and intrinsic kinetic isotope effects (KIEs) for the dihydrofolate reductase (DHFR) catalyzed reaction. This study compares the nature of the hydride-transfer step for a series of active-site mutants, where the size of a side chain that modulates the DAD (I14 in E. coli DHFR) is systematically reduced (I14V, I14A, and I14G). The contributions of the DAD and its dynamics to the hydride-transfer step were examined by the temperature dependence of intrinsic KIEs, hydride-transfer rates, activation parameters, and classical molecular dynamics (MD) simulations. Results are interpreted within the framework of the Marcus-like model where the increase in the temperature dependence of KIEs arises as a direct consequence of the deviation of the DAD from its distribution in the wild type enzyme. Classical MD simulations suggest new populations with larger average DADs, as well as broader distributions, and a reduction in the population of the reactive conformers correlated with the decrease in the size of the hydrophobic residue. The more flexible active site in the mutants required more substantial thermally activated motions for effective H-tunneling, consistent with the hypothesis that the role of the hydrophobic side chain of I14 is to restrict the distribution and dynamics of the DAD and thus assist the hydride-transfer. These studies establish relationships between the distribution of DADs, the hydride-transfer rates, and the DAD's rearrangement toward tunneling-ready states. This structure-function correlation shall assist in the interpretation of the temperature dependence of KIEs caused by mutants far from the active site in this and other enzymes, and may apply generally to C-H→C transfer reactions.


Assuntos
Escherichia coli/enzimologia , Tetra-Hidrofolato Desidrogenase/metabolismo , Domínio Catalítico , Escherichia coli/química , Escherichia coli/genética , Cinética , Modelos Moleculares , Mutação , Tetra-Hidrofolato Desidrogenase/química , Tetra-Hidrofolato Desidrogenase/genética
10.
Chemistry ; 17(15): 4145-56, 2011 Apr 04.
Artigo em Inglês | MEDLINE | ID: mdl-21404343

RESUMO

The nitroprusside ion [Fe(CN)(5)NO](2-) (NP) reacts with excess HS(-) in the pH range 8.5-12.5, in anaerobic medium ("Gmelin" reaction). The progress of the addition process of HS(-) into the bound NO(+) ligand was monitored by stopped-flow UV/Vis/EPR and FTIR spectroscopy, mass spectrometry, and chemical analysis. Theoretical calculations were employed for the characterization of the initial adducts and reaction intermediates. The shapes of the absorbance-time curves at 535-575 nm depend on the pH and concentration ratio of the reactants, R=[HS(-)]/[NP]. The initial adduct [Fe(CN)(5)N(O)SH](3-) (AH, λ(max) ≈570 nm) forms in the course of a reversible process, with k(ad)=190±20 M(-1)s(-1) , k(-ad)=0.3±0.05 s(-1) . Deprotonation of AH (pK(a)=10.5±0.1, at 25.0 °C, I=1 M), leads to [Fe(CN)(5)N(O)S](4-) (A, λ(max)=535 nm, ε=6000±300 M(-1) cm(-1) ). [Fe(CN)(5)NO](.)(3-) and HS(2)(.)(2-) radicals form through the spontaneous decomposition of AH and A. The minor formation of the [Fe(CN)(5)NO](3-) ion equilibrates with [Fe(CN)(4)NO](2-) through cyanide labilization, and generates the "g=2.03" iron-dinitrosyl, [Fe(NO)(2)(SH)(2)](-) , which is labile toward NO release. Alternative nucleophilic attack of HS(-) on AH and A generates the reactive intermediates [Fe(CN)(5)N(OH)(SH)(2)](3-) and [Fe(CN)(5)N(OH)(S)(SH)](4-) , respectively, which decompose through multielectronic nitrosyl reductions, leading to NH(3) and hydrogen disulfide, HS(2)(-) . N(2)O is also produced at pH≥11. Biological relevance relates to the role of NO, NO(-) , and other bound intermediates, eventually able to be released to the medium and rapidly trapped by substrates. Structure and reactivity comparisons of the new nitrososulfide ligands with free and bound nitrosothiolates are provided.


Assuntos
Compostos Ferrosos/química , Sulfeto de Hidrogênio/química , Óxidos de Nitrogênio/química , Nitroprussiato/química , Concentração de Íons de Hidrogênio , Cinética , Ligantes , Estrutura Molecular , Oxirredução , Espectrofotometria Ultravioleta , Estereoisomerismo
12.
Neurosci Lett ; 700: 70-77, 2019 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-29758301

RESUMO

Human-ether-a-go-go-related channel (hERG) is a voltage gated potassium channel (Kv11.1) abundantly expressed in heart and brain tissues. In addition to playing an important role in mediation of repolarizing K+ currents (IKr) in Action Potential (AP), hERG is notorious for its propensity to interact with various medications. The drug-induced block of K+ currents across hERG channel are strongly associated with dysrhythmic conditions collectively known as drug-induced long-QT-syndrome. The recent availability of the high-resolution Cryo-EM structures for the hERG channel has provided unique opportunity to resolve structural mechanisms involved into the process of voltage-gating of hERG channels, map various roles played by components of ventricular and neuronal membranes and then to connect it to cellular pathways through which diverse chemical compounds might be affecting function of the channel. Specifically, lipids and lipid derivatives such as polyunsaturated fatty acids (PUFAs), ceramides and steroids have been shown to directly interact with the lipid facing amino acids in various Kv channels including hERG. In this review, possible lipophilic pathways of hERG activators and blockers, together with the existence of fenestration windows and effects of PUFAs, ceramides and steroids are explored throughout different sections. Finally, the interplay between long QT inducing drugs and phospholipidosis is briefly discussed.


Assuntos
Canal de Potássio ERG1/fisiologia , Lipídeos/fisiologia , Ceramidas/metabolismo , Ceramidas/farmacologia , Canal de Potássio ERG1/agonistas , Canal de Potássio ERG1/antagonistas & inibidores , Canal de Potássio ERG1/química , Ácidos Graxos Insaturados/metabolismo , Ácidos Graxos Insaturados/farmacologia , Humanos , Síndrome do QT Longo/etiologia , Lipídeos de Membrana/fisiologia , Simulação de Acoplamento Molecular , Transdução de Sinais , Esteroides/metabolismo , Esteroides/farmacologia
13.
Biochemistry ; 47(37): 9793-802, 2008 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-18717599

RESUMO

There is recent evidence suggesting that nitrite anion (NO 2 (-)) represents the major intravascular NO storage molecule whose transduction to NO is facilitated by a reduction mechanism catalyzed by deoxygenated hemoglobin (deoxy-Hb). In this work, we provide a detailed microscopic study of deoxy-Hb nitrite reductase (NIR) activity by combining classical molecular dynamics and hybrid quantum mechanical-molecular mechanical simulations. Our results point out that two alternative mechanisms could be operative and suggest that the most energetic barriers should stem from either reprotonation of the distal histidine or NO dissociation from the ferric heme. In the first proposed mechanism, which is similar to that proposed for bacterial NIRs, nitrite anion or nitrous acid coordinates to the heme through the N atom. This pathway involves HisE7 in a one or two proton transfer process, depending on whether the active species is nitrite anion or nitrous acid, to yield an intermediate Fe(III)NO species which eventually dissociates leading to NO and methemoglobin. In the second mechanism, the nitrite anion coordinates to the heme through the O atom. This pathway requires only one proton transfer from HisE7 and leads directly to the formation of a hydroxo Fe(III) complex and NO.


Assuntos
Ânions/metabolismo , Hemoglobinas/química , Hemoglobinas/metabolismo , Óxido Nítrico/metabolismo , Nitritos/química , Nitritos/metabolismo , Ânions/química , Sítios de Ligação , Catálise , Histidina/química , Histidina/metabolismo , Humanos , Ligantes , Modelos Moleculares , Óxido Nítrico/química , Nitrito Redutases/química , Nitrito Redutases/metabolismo , Conformação Proteica
14.
Front Physiol ; 9: 207, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29706893

RESUMO

IKr is the rapidly activating component of the delayed rectifier potassium current, the ion current largely responsible for the repolarization of the cardiac action potential. Inherited forms of long QT syndrome (LQTS) (Lees-Miller et al., 1997) in humans are linked to functional modifications in the Kv11.1 (hERG) ion channel and potentially life threatening arrhythmias. There is little doubt now that hERG-related component of IKr in the heart depends on the tetrameric (homo- or hetero-) channels formed by two alternatively processed isoforms of hERG, termed hERG1a and hERG1b. Isoform composition (hERG1a- vs. the b-isoform) has recently been reported to alter pharmacologic responses to some hERG blockers and was proposed to be an essential factor pre-disposing patients for drug-induced QT prolongation. Very little is known about the gating and pharmacological properties of two isoforms in heart membranes. For example, how gating mechanisms of the hERG1a channels differ from that of hERG1b is still unknown. The mechanisms by which hERG 1a/1b hetero-tetramers contribute to function in the heart, or what role hERG1b might play in disease are all questions to be answered. Structurally, the two isoforms differ only in the N-terminal region located in the cytoplasm: hERG1b is 340 residues shorter than hERG1a and the initial 36 residues of hERG1b are unique to this isoform. In this study, we combined electrophysiological measurements for HEK cells, kinetics and structural modeling to tease out the individual contributions of each isoform to Action Potential formation and then make predictions about the effects of having various mixture ratios of the two isoforms. By coupling electrophysiological data with computational kinetic modeling, two proposed mechanisms of hERG gating in two homo-tetramers were examined. Sets of data from various experimental stimulation protocols (HEK cells) were analyzed simultaneously and fitted to Markov-chain models (M-models). The minimization procedure presented here, allowed assessment of suitability of different Markov model topologies and the corresponding parameters that describe the channel kinetics. The kinetics modeling pointed to key differences in the gating kinetics that were linked to the full channel structure. Interactions between soluble domains and the transmembrane part of the channel appeared to be critical determinants of the gating kinetics. The structures of the full channel in the open and closed states were compared for the first time using the recent Cryo-EM resolved structure for full open hERG channel and an homology model for the closed state, based on the highly homolog EAG1 channel. Key potential interactions which emphasize the importance of electrostatic interactions between N-PAS cap, S4-S5, and C-linker are suggested based on the structural analysis. The derived kinetic parameters were later used in higher order models of cells and tissue to track down the effect of varying the ratios of hERG1a and hERG1b on cardiac action potentials and computed electrocardiograms. Simulations suggest that the recovery from inactivation of hERG1b may contribute to its physiologic role of this isoform in the action potential. Finally, the results presented here contribute to the growing body of evidence that hERG1b significantly affects the generation of the cardiac Ikr and plays an important role in cardiac electrophysiology. We highlight the importance of carefully revisiting the Markov models previously proposed in order to properly account for the relative abundance of the hERG1 a- and b- isoforms.

15.
Curr Top Med Chem ; 17(23): 2681-2702, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28413954

RESUMO

The rapid delayed rectifier current IKr is one of the major K+ currents involved in repolarization of the human cardiac action potential. Various inherited or drug-induced forms of the long QT syndrome (LQTS) in humans are linked to functional and structural modifications in the IKr conducting channels. IKr is carried by the potassium channel Kv11.1 encoded by the gene KCNH2 (commonly referred to as human ether-a-go-go-related gene or hERG) [1, 2]. The first necessary step for predicting emergent drug effects on the heart is determining and modeling the binding thermodynamics and kinetics of primary and major off-target drug interactions with subcellular targets. The bulk of drugs that target hERG channels are known to have complex interactions at the atomic scale. Accordingly, one of the goals for this review is to provide comprehensive guide in the universe of computational models aiming to refine our understanding of structure-function relations in Kv11.1 and its isoforms. The special emphasis is placed on the mapping of drug binding sites and tentative mechanisms of channel inhibition and activation by drugs. An overview over recent structural models and mapping of binding sites for blockers and activators of IKr current along with the discussion on agreements and discrepancies among different models is presented. There is an apparent reciprocity or feedback loop between drug binding and action potential of the cardiac myocytes. Thus one has to connect drug binding to a particular receptor so that its functional consequences impact on the action potential duration. The natural pathway is to develop multi-scale models that connect between receptor and cellular scales. The potential for such multi-scale model development is discussed through the lens of common gating models. Accordingly, the second part of this review covers an ongoing development of the kinetic models of gating transitions and cardiac ion currents carried by hERG channels with and without drug bound.


Assuntos
Canais de Potássio Éter-A-Go-Go/química , Canais de Potássio Éter-A-Go-Go/metabolismo , Canais de Potássio Éter-A-Go-Go/antagonistas & inibidores , Coração/efeitos dos fármacos , Humanos , Modelos Moleculares
16.
Sci Rep ; 6: 32536, 2016 10 12.
Artigo em Inglês | MEDLINE | ID: mdl-27731415

RESUMO

Mutations that reduce inactivation of the voltage-gated Kv11.1 potassium channel (hERG) reduce binding for a number of blockers. State specific block of the inactivated state of hERG block may increase risks of drug-induced Torsade de pointes. In this study, molecular simulations of dofetilide binding to the previously developed and experimentally validated models of the hERG channel in open and open-inactivated states were combined with voltage-clamp experiments to unravel the mechanism(s) of state-dependent blockade. The computations of the free energy profiles associated with the drug block to its binding pocket in the intra-cavitary site display startling differences in the open and open-inactivated states of the channel. It was also found that drug ionization may play a crucial role in preferential targeting to the open-inactivated state of the pore domain. pH-dependent hERG blockade by dofetilie was studied with patch-clamp recordings. The results show that low pH increases the extent and speed of drug-induced block. Both experimental and computational findings indicate that binding to the open-inactivated state is of key importance to our understanding of the dofetilide's mode of action.


Assuntos
Proteínas de Ligação a DNA/química , Canal de Potássio ERG1/química , Canais de Potássio Éter-A-Go-Go/química , Fenetilaminas/química , Sulfonamidas/química , Sítios de Ligação , Proteínas de Ligação a DNA/genética , Canal de Potássio ERG1/genética , Eletrofisiologia , Canais de Potássio Éter-A-Go-Go/genética , Humanos , Concentração de Íons de Hidrogênio , Mutação , Técnicas de Patch-Clamp , Bloqueadores dos Canais de Potássio/química , Bloqueadores dos Canais de Potássio/farmacologia , Conformação Proteica
17.
PLoS One ; 9(9): e105553, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25191697

RESUMO

One of the main culprits in modern drug discovery is apparent cardiotoxicity of many lead-candidates via inadvertent pharmacologic blockade of K+, Ca2+ and Na+ currents. Many drugs inadvertently block hERG1 leading to an acquired form of the Long QT syndrome and potentially lethal polymorphic ventricular tachycardia. An emerging strategy is to rely on interventions with a drug that may proactively activate hERG1 channels reducing cardiovascular risks. Small molecules-activators have a great potential for co-therapies where the risk of hERG-related QT prolongation is significant and rehabilitation of the drug is impractical. Although a number of hERG1 activators have been identified in the last decade, their binding sites, functional moieties responsible for channel activation and thus mechanism of action, have yet to be established. Here, we present a proof-of-principle study that combines de-novo drug design, molecular modeling, chemical synthesis with whole cell electrophysiology and Action Potential (AP) recordings in fetal mouse ventricular myocytes to establish basic chemical principles required for efficient activator of hERG1 channel. In order to minimize the likelihood that these molecules would also block the hERG1 channel they were computationally engineered to minimize interactions with known intra-cavitary drug binding sites. The combination of experimental and theoretical studies led to identification of functional elements (functional groups, flexibility) underlying efficiency of hERG1 activators targeting binding pocket located in the S4-S5 linker, as well as identified potential side-effects in this promising line of drugs, which was associated with multi-channel targeting of the developed drugs.


Assuntos
Desenho de Fármacos , Canais de Potássio Éter-A-Go-Go/química , Modelos Moleculares , Potenciais de Ação/efeitos dos fármacos , Sítios de Ligação , Cresóis/síntese química , Cresóis/química , Cresóis/farmacologia , Bases de Dados de Produtos Farmacêuticos , Relação Dose-Resposta a Droga , Canal de Potássio ERG1 , Canais de Potássio Éter-A-Go-Go/agonistas , Canais de Potássio Éter-A-Go-Go/antagonistas & inibidores , Canais de Potássio Éter-A-Go-Go/metabolismo , Humanos , Ligantes , Conformação Molecular , Simulação de Acoplamento Molecular , Compostos de Fenilureia/síntese química , Compostos de Fenilureia/química , Compostos de Fenilureia/farmacologia , Ligação Proteica , Bibliotecas de Moléculas Pequenas
18.
Biophys Rev ; 6(1): 27-46, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-28509962

RESUMO

Thiol redox chemical reactions play a key role in a variety of physiological processes, mainly due to the presence of low-molecular-weight thiols and cysteine residues in proteins involved in catalysis and regulation. Specifically, the subtle sensitivity of thiol reactivity to the environment makes the use of simulation techniques extremely valuable for obtaining microscopic insights. In this work we review the application of classical and quantum-mechanical atomistic simulation tools to the investigation of selected relevant issues in thiol redox biochemistry, such as investigations on (1) the protonation state of cysteine in protein, (2) two-electron oxidation of thiols by hydroperoxides, chloramines, and hypochlorous acid, (3) mechanistic and kinetics aspects of the de novo formation of disulfide bonds and thiol-disulfide exchange, (4) formation of sulfenamides, (5) formation of nitrosothiols and transnitrosation reactions, and (6) one-electron oxidation pathways.

19.
Chem Commun (Camb) ; 46(47): 8974-6, 2010 Dec 21.
Artigo em Inglês | MEDLINE | ID: mdl-20972508

RESUMO

Comparison of the nature of hydride transfer in wild-type and active site mutant (I14A) of dihydrofolate reductase suggests that the size of this side chain at position 14 modulates H-tunneling.


Assuntos
Alanina/química , Isoleucina/química , Tetra-Hidrofolato Desidrogenase/metabolismo , Substituição de Aminoácidos , Catálise , Domínio Catalítico , Escherichia coli/enzimologia , Cinética , Simulação de Dinâmica Molecular , Mutação , NADP/química , NADP/metabolismo , Tetra-Hidrofolato Desidrogenase/genética
20.
Inorg Chem ; 47(11): 4723-33, 2008 Jun 02.
Artigo em Inglês | MEDLINE | ID: mdl-18465851

RESUMO

In this work, we present a complete and detailed experimental characterization and theoretical study of a variety of coordinated S-nitrosothiols (RSNOs), such as cysteine derivatives, mercaptosuccinic acid, benzyl thiol, and phenyl thiol. Some of them are extremely unstable and sensitive in free form. Strikingly, in contrast with free S-nitrosothiols, we found that, upon coordination to iridium, they become very stable even in aqueous solutions. The study of these coordinated complexes provides further insight on the elucidation of structural aspects dealing with the nature of the S-N bond in RSNOs, a fact which still remains a matter of controversy.


Assuntos
S-Nitrosotióis/química , Água/química , Cristalografia por Raios X , Cisteína/química , Ésteres/química , Espectroscopia de Ressonância Magnética , Compostos de Potássio/química , Solubilidade , Espectrometria de Massas por Ionização por Electrospray , Espectrofotometria Ultravioleta , Espectroscopia de Infravermelho com Transformada de Fourier , Tiomalatos/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA