Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Sci Rep ; 7: 42231, 2017 02 08.
Article in English | MEDLINE | ID: mdl-28176876

ABSTRACT

Listeriolysin O (LLO) is a cytolysin capable of forming pores in cholesterol-rich lipid membranes of host cells. It is conveniently suited for engineering a pH-governed responsiveness, due to a pH sensor identified in its structure that was shown before to affect its stability. Here we introduced a new level of control of its hemolytic activity by making a variant with hemolytic activity that was pH-dependent. Based on detailed structural analysis coupled with molecular dynamics and mutational analysis, we found that the bulky side chain of Tyr406 allosterically affects the pH sensor. Molecular dynamics simulation further suggested which other amino acid residues may also allosterically influence the pH-sensor. LLO was engineered to the point where it can, in a pH-regulated manner, perforate artificial and cellular membranes. The single mutant Tyr406Ala bound to membranes and oligomerized similarly to the wild-type LLO, however, the final membrane insertion step was pH-affected by the introduced mutation. We show that the mutant toxin can be activated at the surface of artificial membranes or living cells by a single wash with slightly acidic pH buffer. Y406A mutant has a high potential in development of novel nanobiotechnological applications such as controlled release of substances or as a sensor of environmental pH.


Subject(s)
Bacterial Toxins/metabolism , Heat-Shock Proteins/metabolism , Hemolysin Proteins/metabolism , Pore Forming Cytotoxic Proteins/metabolism , Protein Engineering/methods , Animals , Bacterial Toxins/chemistry , Caco-2 Cells , Cell Membrane Permeability , Heat-Shock Proteins/chemistry , Hemolysin Proteins/chemistry , Humans , Hydrogen-Ion Concentration , Membrane Lipids/metabolism , Molecular Dynamics Simulation , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Pore Forming Cytotoxic Proteins/chemistry , Protein Domains , Sheep
2.
J Phys Chem B ; 120(44): 11419-11427, 2016 11 10.
Article in English | MEDLINE | ID: mdl-27734680

ABSTRACT

Monoamine oxidases (MAOs) A and B are flavoenzymes responsible for the metabolism of biogenic amines, such as dopamine, serotonin, and noradrenaline (NA), which is why they have been extensively implicated in the etiology and course of various neurodegenerative disorders and, accordingly, used as primary pharmacological targets to treat these debilitating cognitive diseases. The precise chemical mechanism through which MAOs regulate the amine concentration, which is vital for the development of novel inhibitors, is still not unambiguously determined in the literature. In this work, we present atomistic empirical valence bond simulations of the rate-limiting step of the MAO-A-catalyzed NA (norepinephrine) degradation, involving hydride transfer from the substrate α-methylene group to the flavin moiety of the flavin adenine dinucleotide prosthetic group, employing the full dimensionality and thermal fluctuations of the hydrated enzyme, with extensive configurational sampling. We show that MAO-A lowers the free energy of activation by 14.3 kcal mol-1 relative to that of the same reaction in aqueous solution, whereas the calculated activation free energy of ΔG‡ = 20.3 ± 1.6 kcal mol-1 is found to be in reasonable agreement with the correlated experimental value of 16.5 kcal mol-1. The results presented here strongly support the fact that both MAO-A and MAO-B isoforms function by the same hydride-transfer mechanism. We also considered a few point mutations of the "aromatic cage" tyrosine residue (Tyr444Phe, Tyr444Leu, Tyr444Trp, Tyr444His, and Tyr444Glu), and the calculated changes in the reaction barriers are in agreement with the experimental values, thus providing further support to the proposed mechanism.


Subject(s)
Molecular Dynamics Simulation , Monoamine Oxidase/metabolism , Norepinephrine/metabolism , Biocatalysis , Molecular Conformation , Monoamine Oxidase/chemistry , Norepinephrine/chemistry , Quantum Theory
3.
PLoS One ; 11(5): e0154002, 2016.
Article in English | MEDLINE | ID: mdl-27159606

ABSTRACT

In this article we report a combined experimental and computational study concerning the effects of deuteration on the binding of histamine and two other histaminergic agonists to 3H-tiotidine-labeled histamine H2 receptor in neonatal rat astrocytes. Binding affinities were measured by displacing radiolabeled tiotidine from H2 receptor binding sites present on cultured neonatal rat astrocytes. Quantum-chemical calculations were performed by employing the empirical quantization of nuclear motion within a cluster model of the receptor binding site extracted from the homology model of the entire H2 receptor. Structure of H2 receptor built by homology modelling is attached in the supporting information (S1 Table) Experiments clearly demonstrate that deuteration affects the binding by increasing the affinity for histamine and reducing it for 2-methylhistamine, while basically leaving it unchanged for 4-methylhistamine. Ab initio quantum-chemical calculations on the cluster system extracted from the homology H2 model along with the implicit quantization of the acidic N-H and O-H bonds demonstrate that these changes in the binding can be rationalized by the altered strength of the hydrogen bonding upon deuteration known as the Ubbelohde effect. Our computational analysis also reveals a new mechanism of histamine binding, which underlines an important role of Tyr250 residue. The present work is, to our best knowledge, the first study of nuclear quantum effects on ligand receptor binding. The ligand H/D substitution is relevant for therapy in the context of perdeuterated and thus more stable drugs that are expected to enter therapeutic practice in the near future. Moreover, presented approach may contribute towards understanding receptor activation, while a distant goal remains in silico discrimination between agonists and antagonists based on the receptor structure.


Subject(s)
Deuterium/chemistry , Histamine/metabolism , Animals , Ligands , Quantum Theory , Rats , Receptors, Histamine/metabolism
4.
Mol Neurobiol ; 53(5): 3400-3415, 2016 07.
Article in English | MEDLINE | ID: mdl-26081152

ABSTRACT

We collected experimental kinetic rate constants for chemical processes responsible for the development and progress of neurodegeneration, focused on the enzymatic and non-enzymatic degradation of amine neurotransmitters and their reactive and neurotoxic metabolites. A gross scheme of neurodegeneration on the molecular level is based on two pathways. Firstly, reactive species oxidise heavy atom ions, which enhances the interaction with alpha-synuclein, thus promoting its folding to the beta form and giving rise to insoluble amyloid plaques. The latter prevents the function of vesicular transport leading to gradual neuronal death. In the second pathway, radical species, OH(·) in particular, react with the methylene groups of the apolar part of the lipid bilayer of either the cell or mitochondrial wall, resulting in membrane leakage followed by dyshomeostasis, loss of resting potential and neuron death. Unlike all other central neural system (CNS)-relevant biogenic amines, dopamine and noradrenaline are capable of a non-enzymatic auto-oxidative reaction, which produces hydrogen peroxide. This reaction is not limited to the mitochondrial membrane where scavenging enzymes, such as catalase, are located. On the other hand, dopamine and its metabolites, such as dopamine-o-quinone, dopaminechrome, 5,6-dihydroxyindole and indo-5,6-quinone, also interact directly with alpha-synuclein and reversibly inhibit plaque formation. We consider the role of the heavy metal ions, selected scavengers and scavenging enzymes, and discuss the relevance of certain foods and food supplements, including curcumin, garlic, N-acetyl cysteine, caffeine and red wine, as well as the long-term administration of non-steroid anti-inflammatory drugs and occasional tobacco smoking, that could all act toward preventing neurodegeneration. The current analysis can be employed in developing strategies for the prevention and treatment of neurodegeneration, and, hopefully, aid in the building of an overall kinetic molecular model of neurodegeneration itself.


Subject(s)
Nerve Degeneration/metabolism , Animals , Free Radicals/metabolism , Glutamic Acid/metabolism , Humans , Inflammation/pathology , Kinetics , Polyamines/metabolism
5.
Blood ; 124(26): 3896-904, 2014 Dec 18.
Article in English | MEDLINE | ID: mdl-25359991

ABSTRACT

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.


Subject(s)
Lymphoma/genetics , Mutation , Myeloid Differentiation Factor 88/genetics , Allosteric Site , Cell Line, Tumor , HEK293 Cells , Heterozygote , Humans , Inflammation , Luminescence , Microscopy, Confocal , Molecular Dynamics Simulation , Phenotype , Polymerase Chain Reaction , Protein Structure, Tertiary , RNA, Small Interfering/metabolism , Receptors, Interleukin-1/metabolism , Signal Transduction
6.
Proteins ; 82(12): 3347-55, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25220264

ABSTRACT

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.


Subject(s)
Dopamine/metabolism , Models, Molecular , Monoamine Oxidase/metabolism , Biocatalysis , Catalytic Domain , Cluster Analysis , Databases, Protein , Dopamine/chemistry , Energy Transfer , Flavin-Adenine Dinucleotide/chemistry , Flavin-Adenine Dinucleotide/metabolism , Humans , Kinetics , Lysine/chemistry , Molecular Dynamics Simulation , Monoamine Oxidase/chemistry , Protein Conformation , Quantum Theory
7.
J Phys Chem B ; 118(16): 4326-32, 2014 Apr 24.
Article in English | MEDLINE | ID: mdl-24678966

ABSTRACT

Monoamine oxidases (MAO) A and B are important flavoenzymes involved in the metabolism of amine neurotransmitters. Orru et al. ( J. Neural Transm. 2013 , 120 , 847 - 851 ) recently presented experimental results that have challenged the prevailing assumption that MAO A and MAO B employ an identical catalytic mechanism. We compared the spatial configuration of ionizable groups in both isozymes and estimated the time-averaged electrostatic potential by calculating the pKa values of five active site residues. Superimposition of both experimental structures shows very close overlap and the RMSD in placements of ionizable groups within 16 Å of the reaction center is only 0.847 Å. This similarity is also reflected in the calculated pKa values, where the largest difference between the MAO A and MAO B pKa values was found for residues Tyr188 in MAO B and the corresponding Tyr197 in MAO A assuming 1.23 units. The pKa values for the other four studied residues differ by less than 0.75 units. The results show that the electrostatic preorganizations in both active sites are very similar, supporting the idea that both enzymes work by the same mechanism.


Subject(s)
Monoamine Oxidase/chemistry , Static Electricity , Catalytic Domain , Dopamine/chemistry , Isoenzymes/chemistry , Molecular Dynamics Simulation , Molecular Structure , Tyrosine/chemistry
8.
J Neural Transm (Vienna) ; 120(6): 875-82, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23546802

ABSTRACT

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.


Subject(s)
Clorgyline/pharmacology , Monoamine Oxidase Inhibitors/chemistry , Monoamine Oxidase Inhibitors/pharmacology , Monoamine Oxidase/drug effects , Monoamine Oxidase/metabolism , Computers, Molecular , Humans , Models, Chemical
9.
Int J Mol Sci ; 13(2): 1269-1283, 2012.
Article in English | MEDLINE | ID: mdl-22408390

ABSTRACT

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.


Subject(s)
DNA/chemistry , Flavonoids/chemistry , Glycine max/chemistry , Guanine/chemistry
10.
J Chem Theory Comput ; 8(10): 3864-70, 2012 Oct 09.
Article in English | MEDLINE | ID: mdl-26593027

ABSTRACT

Monoamine oxidase (MAO), which exists in two isozymic forms, MAO A and MAO B, is an important flavoenzyme responsible for the metabolism of amine neurotransmitters such as dopamine, serotonin, and norepinephrine. Despite extensive research effort, neither the catalytic nor the inhibition mechanisms of MAO have been completely understood. There has also been dispute with regard to the protonation state of the substrate upon entering the active site, as well as the identity of residues that are important for the initial deprotonation of irreversible acetylenic inhibitors, in accordance with the recently proposed mechanism. Therefore, in order to investigate features essential for the modes of action of MAO, we have calculated pKa values of three relevant tyrosine residues in the MAO B active site, with and without dopamine bound as the substrate (as well as the pKa of the dopamine itself in the active site). The calculated pKa values for Tyr188, Tyr398, and Tyr435 in the complex are found to be shifted upward to 13.0, 13.7, and 14.7, respectively, relative to 10.1 in aqueous solution, ruling out the likelihood that they are viable proton acceptors. The altered tyrosine pKa values could be rationalized as an interplay of two opposing effects: insertion of positively charged bulky dopamine that lowers tyrosine pKa values, and subsequent removal of water molecules from the active site that elevates tyrosine pKa values, in which the latter prevails. Additionally, the pKa value of the bound dopamine (8.8) is practically unchanged compared to the corresponding value in aqueous solution (8.9), as would be expected from a charged amine placed in a hydrophobic active site consisting of aromatic moieties. We also observed potentially favorable cation-π interactions between the -NH3(+) group on dopamine and aromatic moieties, which provide a stabilizing effect to the charged fragment. Thus, we offer here theoretical evidence that the amine is most likely to be present in the active site in its protonated form, which is similar to the conclusion from experimental studies of MAO A (Jones et al. J. Neural Trans.2007, 114, 707-712). However, the free energy cost of transferring the proton from the substrate to the bulk solvent is only 1.9 kcal mol(-1), leaving open the possibility that the amine enters the chemical step in its neutral form. In conjunction with additional experimental and computational work, the data presented here should lead toward a deeper understanding of mechanisms of the catalytic activity and irreversible inhibition of MAO B, which can allow for the design of novel and improved MAO B inhibitors.

SELECTION OF CITATIONS
SEARCH DETAIL
...