Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 59
Filter
Add more filters











Publication year range
1.
Int J Biol Macromol ; 276(Pt 1): 133732, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39002919

ABSTRACT

γ-aminobutyric acid (GABA) plays an important role in anti-anxiety by inhibiting neurotransmitter in the central nervous system (CNS) of mammals, which is generated in the germinating seeds. The key enzymes activity of GABA metabolism pathway and nutrients content in hemp seeds during germination were studied after treated with ultrasound and CaCl2. The mechanism of exogenous stress on key enzymes in GABA metabolism pathway was investigated by molecular dynamics simulation. The results showed that ultrasonic combined with 1.5 mmol·L-1CaCl2 significantly increased the activities of glutamate decarboxylase (GAD) and GABA transaminase (GABA-T) in seeds, and promoted the conversion of glutamate to GABA, resulting in the decrease of glutamate content and the accumulation of GABA. Molecular dynamics simulations revealed that Ca2+ environment enhanced the activity of GAD and GABA-T enzymes by altering their secondary structure, exposing their hydrophobic residues. Ultrasound, germination and CaCl2 stress improved the nutritional value of hemp seeds.


Subject(s)
Calcium Chloride , Cannabis , Germination , Seeds , Cannabis/metabolism , Cannabis/chemistry , Germination/drug effects , Seeds/drug effects , Seeds/growth & development , Seeds/metabolism , Calcium Chloride/pharmacology , Calcium Chloride/chemistry , Ultrasonic Waves , gamma-Aminobutyric Acid/metabolism , Glutamate Decarboxylase/metabolism , Molecular Dynamics Simulation , 4-Aminobutyrate Transaminase/metabolism , 4-Aminobutyrate Transaminase/chemistry
2.
J Mol Model ; 26(9): 228, 2020 Aug 11.
Article in English | MEDLINE | ID: mdl-32780180

ABSTRACT

Modulation of γ-aminobutyric acid (GABA) levels has been required in various disorders. GABA itself cannot be directly introduced into central nervous system (CNS) because of the blood brain barrier; inhibition of GABA aminotransferase (GABA-AT), which degrades GABA in CNS, has been the target for the modulation of GABA levels in CNS. Given that root extract of valerian (Valeriana officinalis) has been used for millennia as anti-anxiolytic and sedative, in silico approach was carried out to investigate valerian compounds exhibiting GABA-AT inhibiting activity. The 3D structure of human GABA-AT was created from pig crystal structure via homology modeling. Inhibition of GABA-AT by 18 valerian compounds was analyzed using molecular docking and molecular dynamics simulations and compared with known GABA-AT inhibitors such as vigabatrin and valproic acid. Isovaleric acid and didrovaltrate exhibited GABA-AT inhibiting activity in computational analysis, albeit less potent compared with vigabatrin. However, multiple compounds with low activity may have additive effects when the total extract of valeriana root was used in traditional usage. In addition, isovaleric acid shares similar backbone structure to GABA, suggesting that isovaleric acid might be a valuable starting structure for the development of more efficient GABA-AT inhibitors for disorders related with low level of GABA in the CNS.


Subject(s)
4-Aminobutyrate Transaminase/chemistry , Enzyme Inhibitors/chemistry , Molecular Docking Simulation , Molecular Dynamics Simulation , Plant Extracts/chemistry , Valerian/chemistry , 4-Aminobutyrate Transaminase/antagonists & inhibitors , Amino Acid Sequence , Binding Sites , Catalytic Domain , Enzyme Inhibitors/pharmacology , Ligands , Molecular Conformation , Molecular Structure , Plant Extracts/pharmacology , Protein Binding , Structure-Activity Relationship
3.
Biosci Biotechnol Biochem ; 84(1): 118-125, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31516066

ABSTRACT

We developed an enzymatic assay system enabling easy quantification of 4-aminobutyric acid (GABA). The reaction of GABA aminotransferase obtained from Streptomyces decoyicus NBRC 13977 was combined to those of the previously developed glutamate assay system using glutamate oxidase and peroxidase. The three-enzyme system allowing GABA-dependent dye formation due to the oxidative coupling between 4-aminoantipyrine and Trinder's reagent enabled accurate quantification of 0.2 - 150 mg/L GABA. A pretreatment mixture consisting of glutamate oxidase, ascorbate oxidase and catalase eliminating glutamate, ascorbate, and hydrogen peroxide, respectively, was also prepared to remove those inhibitory substances from samples. Thus, constructed assay kit was used to measure the GABA content in tomato samples. The results were almost the same as that obtained by the conventional method using liquid chromatography-tandem mass spectrometry. The kit will become a promising tool especially for the on-site measurement of GABA content in agricultural products.


Subject(s)
4-Aminobutyrate Transaminase/chemistry , Amino Acid Oxidoreductases/chemistry , Colorimetry/methods , Enzyme Assays/methods , Peroxidase/chemistry , gamma-Aminobutyric Acid/analysis , Ampyrone/chemistry , Ascorbate Oxidase/chemistry , Catalase/chemistry , Chromatography, Liquid , Enzyme Assays/economics , Ferric Compounds/chemistry , Glutamic Acid/chemistry , Hydrogen Peroxide/chemistry , Solanum lycopersicum/chemistry , Oxidative Coupling , Recombinant Proteins , Streptomyces/enzymology , Tandem Mass Spectrometry
4.
Bioorg Chem ; 88: 102832, 2019 07.
Article in English | MEDLINE | ID: mdl-31102809

ABSTRACT

Epilepsy is a group of neurological disorders characterized by recurrent seizures that disturbs about 60 million people worldwide. In this article, a novel series of 3,4,5-trimethoxycinnamic acid (TMCA) ester derivatives 1-35 were designed inspired from the traditional Chinese herb pair drugs Polygala tenuifolia and Gastrodia elata and synthesized followed by in vivo and in silico evaluation of their anticonvulsant potential. All the synthesized derivatives were biologically evaluated for their anticonvulsant potential using two acute model of seizures induced in mice, the maximal electroshock (MES) and sc-pentylenetetrazole (PTZ) models. Simultaneously, the motor impairment as a surrogate of acute neurotoxicity and in vitro screening of cytotoxicity against HepG-2 cells line were assessed through the rotarod performance test and CCK-8 assay, respectively. In addition, the physicochemical and pharmacokinetic parameters of the active compounds were determined. Our results showed that compounds 5, 7, 8, 13, 20, 25, 28, 30 and 32 exhibited preferable anticonvulsant activity in primary evaluation, with compounds 28 and 32 being the most promising anticonvulsant agents in according to results of subsequent pharmacology and toxicity evaluation. Additionally, the molecular modeling experiments predicted good binding interactions of part of the obtained active molecules with the gamma-aminobutyric acid (GABA) transferas. Therefore, it could be concluded that the synthesized derivatives 28 and 32 would represent useful lead compounds for further investigation in the development of anticonvulsant agents.


Subject(s)
Anticonvulsants/therapeutic use , Cinnamates/therapeutic use , Seizures/drug therapy , 4-Aminobutyrate Transaminase/chemistry , 4-Aminobutyrate Transaminase/metabolism , Animals , Anticonvulsants/chemical synthesis , Anticonvulsants/metabolism , Anticonvulsants/pharmacology , Binding Sites , Cinnamates/chemical synthesis , Cinnamates/metabolism , Cinnamates/pharmacology , Drug Design , Epilepsy/drug therapy , Gastrodia/chemistry , Hep G2 Cells , Humans , Male , Mice , Molecular Docking Simulation , Molecular Structure , Pentylenetetrazole , Polygala/chemistry , Protein Binding , Seizures/chemically induced , Structure-Activity Relationship , Swine
5.
Biochem Biophys Res Commun ; 514(3): 601-606, 2019 06 30.
Article in English | MEDLINE | ID: mdl-31072617

ABSTRACT

γ-Aminobutyrate (GABA), a four carbon non-protein amino acid, is used by some microorganisms as a source of carbon and/or nitrogen. Corynebacterium glutamicum has an incomplete GABA shunt that lacks a glutamate decarboxylase coding gene for the conversion of glutamate to GABA. Recently, a novel GABA assimilation system was identified in C. glutamicum. In the cell, GABA aminotransferase (GABA-AT) is the first step of GABA assimilation in the process of utilizing GABA as a carbon and/or nitrogen source. In this study, we report the crystal structure of CgGABA-AT in complex with PLP-GABA. We used structural studies and site-directed mutagenesis experiments to identify the key residues that contribute to the formation of the active site. Furthermore, based on structural comparisons and amino acid sequence alignment, we demonstrate the differences between the GABA-ATs of bacteria, fungi, and animals.


Subject(s)
4-Aminobutyrate Transaminase/chemistry , 4-Aminobutyrate Transaminase/metabolism , Corynebacterium glutamicum/enzymology , Pyridoxal Phosphate/metabolism , gamma-Aminobutyric Acid/metabolism , Amino Acid Sequence , Catalytic Domain , Crystallography, X-Ray , Models, Molecular , Structural Homology, Protein
6.
Molecules ; 23(11)2018 Nov 15.
Article in English | MEDLINE | ID: mdl-30445747

ABSTRACT

We have previously reported the synthesis, in vitro and in silico activities of new GABA analogues as inhibitors of the GABA-AT enzyme from Pseudomonas fluorescens, where the nitrogen atom at the γ-position is embedded in heterocyclic scaffolds. With the goal of finding more potent inhibitors, we now report the synthesis of a new set of GABA analogues with a broader variation of heterocyclic scaffolds at the γ-position such as thiazolidines, methyl-substituted piperidines, morpholine and thiomorpholine and determined their inhibitory potential over the GABA-AT enzyme from Pseudomonas fluorescens. These structural modifications led to compound 9b which showed a 73% inhibition against this enzyme. In vivo studies with PTZ-induced seizures on male CD1 mice show that compound 9b has a neuroprotective effect at a 0.50 mmole/kg dose. A QSAR study was carried out to find the molecular descriptors associated with the structural changes in the GABA scaffold to explain their inhibitory activity against GABA-AT. Employing 3D molecular descriptors allowed us to propose the GABA analogues enantiomeric active form. To evaluate the interaction with Pseudomonas fluorescens and human GABA-AT by molecular docking, the constructions of homology models was carried out. From these calculations, 9b showed a strong interaction with both GABA-AT enzymes in agreement with experimental results and the QSAR model, which indicates that bulky ligands tend to be the better inhibitors especially those with a sulfur atom on their structure.


Subject(s)
4-Aminobutyrate Transaminase/antagonists & inhibitors , 4-Aminobutyrate Transaminase/chemistry , Molecular Docking Simulation , Molecular Dynamics Simulation , Quantitative Structure-Activity Relationship , gamma-Aminobutyric Acid/chemistry , gamma-Aminobutyric Acid/pharmacology , Enzyme Activation , Humans , Ligands , Magnetic Resonance Spectroscopy , Molecular Structure , Pseudomonas fluorescens/enzymology , gamma-Aminobutyric Acid/analogs & derivatives
7.
Luminescence ; 33(4): 722-730, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29653023

ABSTRACT

γ-Aminobutyric acid (GABA) is a well-known neurotransmitter that regulates inhibitory neurotransmission in the mammalian central nervous system and participates in several processes outside the brain. A reliable quantification method is needed to determine its role in different physiological and pathological conditions. However, GABA measurements have several challenges because GABA is neither fluorescent nor electroactive, and it is difficult to detect using enzymatic reactions because no oxidases or dehydrogenases have been identified. Several methods have been developed to quantify GABA concentrations based on the instrumentation available, the sensitivity required, and the volume of samples analyzed. Most of these methods use high-performance liquid chromatography (HPLC). Here, we describe a method for quantifying GABA concentrations in small volume samples using enzymatically-induced electrochemiluminescence with the well-known GABAse complex, which produces glutamate for use in a luminescent reaction with glutamate oxidase and luminol in an electrochemiluminescence cell. The luminescence obtained was proportional to the GABA concentrations in the micromolar range (1-1000), with linear r2 values > 0.95. GABA standards were treated with the enzymatic reactors to generate glutamate (Glu), which was measured simultaneously with an HPLC technique, to validate this new procedure. The assay was further used to determine GABA concentrations in hippocampal extracts. This alternative may be used to quantify GABA levels in fluid samples, such as microdialysates, other perfusates and tissue extracts. Thus, the method presented here is a good alternative for monitoring GABA levels with good sensitivity compared with the traditional methods that are still in use.


Subject(s)
4-Aminobutyrate Transaminase/metabolism , Aldehyde Oxidoreductases/metabolism , Electrochemical Techniques , Luminescence , gamma-Aminobutyric Acid/analysis , 4-Aminobutyrate Transaminase/chemistry , Aldehyde Oxidoreductases/chemistry , Animals , Male , Rats , Rats, Wistar , Regression Analysis , gamma-Aminobutyric Acid/metabolism
8.
J Am Chem Soc ; 140(6): 2151-2164, 2018 02 14.
Article in English | MEDLINE | ID: mdl-29381352

ABSTRACT

γ-Aminobutyric acid (GABA) is the major inhibitory neurotransmitter in the central nervous system. Inhibition of GABA aminotransferase (GABA-AT), a pyridoxal 5'-phosphate (PLP)-dependent enzyme that degrades GABA, has been established as a possible strategy for the treatment of substance abuse. The raised GABA levels that occur as a consequence of this inhibition have been found to antagonize the rapid release of dopamine in the ventral striatum (nucleus accumbens) that follows an acute challenge by an addictive substance. In addition, increased GABA levels are also known to elicit an anticonvulsant effect in patients with epilepsy. We previously designed the mechanism-based inactivator (1S,3S)-3-amino-4-difluoromethylenyl-1-cyclopentanoic acid (2), now called CPP-115, that is 186 times more efficient in inactivating GABA-AT than vigabatrin, the only FDA-approved drug that is an inactivator of GABA-AT. CPP-115 was found to have high therapeutic potential for the treatment of cocaine addiction and for a variety of epilepsies, has successfully completed a Phase I safety clinical trial, and was found to be effective in the treatment of infantile spasms (West syndrome). Herein we report the design, using molecular dynamics simulations, synthesis, and biological evaluation of a new mechanism-based inactivator, (S)-3-amino-4-(difluoromethylenyl)cyclopent-1-ene-1-carboxylic acid (5), which was found to be almost 10 times more efficient as an inactivator of GABA-AT than CPP-115. We also present the unexpected crystal structure of 5 bound to GABA-AT, as well as computational analyses used to assist the structure elucidation process. Furthermore, 5 was found to have favorable pharmacokinetic properties and low off-target activities. In vivo studies in freely moving rats showed that 5 was dramatically superior to CPP-115 in suppressing the release of dopamine in the corpus striatum, which occurs subsequent to either an acute cocaine or nicotine challenge. Compound 5 also attenuated increased metabolic demands (neuronal glucose metabolism) in the hippocampus, a brain region that encodes spatial information concerning the environment in which an animal receives a reinforcing or aversive drug. This multidisciplinary computational design to preclinical efficacy approach should be applicable to the design and improvement of mechanism-based inhibitors of other enzymes whose crystal structures and inactivation mechanisms are known.


Subject(s)
4-Aminobutyrate Transaminase/antagonists & inhibitors , Drug Design , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Proline/analogs & derivatives , 4-Aminobutyrate Transaminase/chemistry , 4-Aminobutyrate Transaminase/metabolism , Animals , Brain/drug effects , Brain/metabolism , Catalytic Domain/drug effects , Crystallography, X-Ray , Dopamine/metabolism , Dopamine Antagonists/chemistry , Dopamine Antagonists/pharmacokinetics , Dopamine Antagonists/pharmacology , Enzyme Inhibitors/pharmacokinetics , Glucose/metabolism , Humans , Male , Models, Molecular , Proline/chemistry , Proline/pharmacokinetics , Proline/pharmacology , Rats , Rats, Sprague-Dawley , gamma-Aminobutyric Acid/metabolism
9.
Biochemistry ; 56(37): 4951-4961, 2017 09 19.
Article in English | MEDLINE | ID: mdl-28816437

ABSTRACT

Potent mechanism-based inactivators can be rationally designed against pyridoxal 5'-phosphate (PLP)-dependent drug targets, such as ornithine aminotransferase (OAT) or γ-aminobutyric acid aminotransferase (GABA-AT). An important challenge, however, is the lack of selectivity toward other PLP-dependent, off-target enzymes, because of similarities in mechanisms of all PLP-dependent aminotransferase reactions. On the basis of complex crystal structures, we investigate the inactivation mechanism of OAT, a hepatocellular carcinoma target, by (1R,3S,4S)-3-amino-4-fluorocyclopentane-1-carboxylic acid (FCP), a known inactivator of GABA-AT. A crystal structure of OAT and FCP showed the formation of a ternary adduct. This adduct can be rationalized as occurring via an enamine mechanism of inactivation, similar to that reported for GABA-AT. However, the crystal structure of an off-target, PLP-dependent enzyme, aspartate aminotransferase (Asp-AT), in complex with FCP, along with the results of attempted inhibition assays, suggests that FCP is not an inactivator of Asp-AT, but rather an alternate substrate. Turnover of FCP by Asp-AT is also supported by high-resolution mass spectrometry. Amid existing difficulties in achieving selectivity of inactivation among a large number of PLP-dependent enzymes, the obtained results provide evidence that a desirable selectivity could be achieved, taking advantage of subtle structural and mechanistic differences between a drug-target enzyme and an off-target enzyme, despite their largely similar substrate binding sites and catalytic mechanisms.


Subject(s)
4-Aminobutyrate Transaminase/antagonists & inhibitors , Aspartate Aminotransferases/antagonists & inhibitors , Cycloleucine/analogs & derivatives , Enzyme Inhibitors/pharmacology , Models, Molecular , Ornithine-Oxo-Acid Transaminase/antagonists & inhibitors , Pyridoxal Phosphate/metabolism , 4-Aminobutyrate Transaminase/chemistry , 4-Aminobutyrate Transaminase/metabolism , Aspartate Aminotransferases/chemistry , Aspartate Aminotransferases/genetics , Aspartate Aminotransferases/metabolism , Binding Sites , Catalytic Domain , Crystallography, X-Ray , Cycloleucine/chemistry , Cycloleucine/metabolism , Cycloleucine/pharmacology , Databases, Chemical , Databases, Protein , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , Escherichia coli Proteins/antagonists & inhibitors , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Humans , Ligands , Molecular Conformation , Ornithine-Oxo-Acid Transaminase/chemistry , Ornithine-Oxo-Acid Transaminase/genetics , Ornithine-Oxo-Acid Transaminase/metabolism , Protein Conformation , Pyridoxal Phosphate/chemistry , Pyridoxamine/chemistry , Pyridoxamine/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Structural Homology, Protein , Substrate Specificity
10.
Acta Pharm ; 66(3): 353-72, 2016 Sep 01.
Article in English | MEDLINE | ID: mdl-27383885

ABSTRACT

A series of N-(2-(benzoyl/4-chlorobenzoyl)-benzofuran- 3-yl)-2-(substituted)-acetamide derivatives (4a-l, 5a-l) was synthesized in good yield. All synthesized compounds were in agreement with elemental and spectral data. The anticonvulsant activity of all synthesized compounds was assessed against the maximal electroshock induced seizures (MES) model in mice. Neurotoxicity was evaluated using the rotarod method. The majority of compounds exhibited anticonvulsant activity at a dose of 30 mg kg-1 body mass during 0.5-4 h, indicating their ability to prevent seizure spread at low doses. Relative to phenytoin, [N-(2-(4-chlorobenzoyl)benzofuran-3-yl)-2-(cyclohexyl( methyl) amino)-acetamide] (5i) and [N-(2-(4-chlorobenzoyl)benzofuran-3-yl)-2-(4-methylpiperidin-1- yl)-acetamide] (5c) demonstrated comparable relative anticonvulsant potency of 0.74 and 0.72, respectively, whereas [(N-(2-(4-chlorobenzoyl)benzofuran-3-yl)-2-(4-(furan-2-carbonyl)-piperazin-1-yl)-acetamide] (5f) exhibited the lowest relative potency of 0.16. The ALD50 of tested compounds ranged from 1.604 to 1.675 mmol kg-1 body mass. The ED50 of synthesized compounds ranged from 0.055 to 0.259 mmol kg-1 (~23.4 to 127.6 mg kg-1) body mass. The pharmacophore mapping of the examined compounds on standard drugs (phenobarbital, phenytoin, ralitolin and carbamazepine) strongly suggests that these compounds may exert their anticonvulsant activity via the same established mechanism as that of known drugs.


Subject(s)
4-Aminobutyrate Transaminase/metabolism , Anticonvulsants/therapeutic use , Benzofurans/therapeutic use , Drug Design , Models, Molecular , Seizures/prevention & control , 4-Aminobutyrate Transaminase/chemistry , Acetamides/adverse effects , Acetamides/chemistry , Acetamides/metabolism , Acetamides/therapeutic use , Animals , Anticonvulsants/adverse effects , Anticonvulsants/chemistry , Anticonvulsants/metabolism , Benzofurans/adverse effects , Benzofurans/chemistry , Benzofurans/metabolism , Binding Sites , Cerebellum/drug effects , Cerebellum/metabolism , Dose-Response Relationship, Drug , GABA Agonists/adverse effects , GABA Agonists/chemistry , GABA Agonists/metabolism , GABA Agonists/therapeutic use , Glycine/adverse effects , Glycine/analogs & derivatives , Glycine/chemistry , Glycine/metabolism , Glycine/therapeutic use , Lethal Dose 50 , Male , Medulla Oblongata/drug effects , Medulla Oblongata/metabolism , Mesencephalon/drug effects , Mesencephalon/metabolism , Mice , Molecular Docking Simulation , Neurons/drug effects , Neurons/metabolism , Neurotoxicity Syndromes/etiology , Neurotoxicity Syndromes/metabolism , Rats, Wistar , Sus scrofa , gamma-Aminobutyric Acid/chemistry , gamma-Aminobutyric Acid/metabolism
11.
Biomed Pharmacother ; 81: 402-410, 2016 Jul.
Article in English | MEDLINE | ID: mdl-27261619

ABSTRACT

The unequivocal hypotheses about anticonvulsant activity of valproic acid (VPA) have always been a basic hurdle in designing next generation neurotherapeutics, particularly the anti-epileptic drugs. The present study reports about a comprehensive in-silico investigation into qualitative and quantitative binding of VPA and corresponding natural ligands of four major enzymes involved in neurotransmissions, namely-GABA transaminase (GABAt), α-keto glutarate dehydrogenase (α-KGDH), Succinate Semialdehyde dehydrogenase (SSADH) and Glutamate Decarboxylase (GAD), respectively. The molecular docking analyses revealed that VPA inhibits GABAt and α-KGDH through allosteric while SSADH through competitive mode of binding. There is an observed elevation in binding of glutamate over GAD in the presence of VPA. The docking inhibition constant (Ki) of VPA to all the studied enzymatic receptors were observed to be well below the therapeutic concentration of VPA in blood, except for α-KGDH, thus favouring GABAergic over glutamatergic mode of anticonvulsant activity of VPA. The report is probably the first comprehensive in-silico molecular study about VPA action.


Subject(s)
4-Aminobutyrate Transaminase/metabolism , Anticonvulsants/pharmacology , Computer Simulation , Succinate-Semialdehyde Dehydrogenase/metabolism , Valproic Acid/pharmacology , 4-Aminobutyrate Transaminase/chemistry , Anticonvulsants/chemistry , Binding Sites , Glutamate Decarboxylase/metabolism , Glutamates/metabolism , Humans , Hydrogen Bonding , Ketoglutarate Dehydrogenase Complex/metabolism , Ketoglutaric Acids/metabolism , Ligands , Molecular Docking Simulation , Structural Homology, Protein , Succinate-Semialdehyde Dehydrogenase/chemistry , Valproic Acid/chemistry , gamma-Aminobutyric Acid/metabolism
12.
Proteins ; 84(7): 875-91, 2016 07.
Article in English | MEDLINE | ID: mdl-26800298

ABSTRACT

The pyridoxal 5-phosphate (PLP) cofactor is a significant organic molecule in medicinal chemistry. It is often found covalently bound to lysine residues in proteins to form PLP dependent enzymes. An example of this family of PLP dependent enzymes is γ-aminobutyric acid aminotransferase (GABA-AT) which is responsible for the degradation of the neurotransmitter GABA. Its inhibition or inactivation can be used to prevent the reduction of GABA concentration in brain which is the source of several neurological disorders. As a test case for PLP dependent enzymes, we have performed molecular dynamics simulations of GABA-AT to reveal the roles of the protein residues and its cofactor. Three different states have been considered: the apoenzyme, the holoenzyme, and the inactive state obtained after the suicide inhibition by vigabatrin. Different protonation states have also been considered for PLP and two key active site residues: Asp298 and His190. Together, 24 independent molecular dynamics trajectories have been simulated for a cumulative total of 2.88 µs. Our results indicate that, unlike in aqueous solution, the PLP pyridine moiety is protonated in GABA-AT. This is a consequence of a pKa shift triggered by a strong charge-charge interaction with an ionic "diad" formed by Asp298 and His190 that would help the activation of the first half-reaction of the catalytic mechanism in GABA-AT: the conversion of PLP to free pyridoxamine phosphate (PMP). In addition, our MD simulations exhibit additional strong hydrogen bond networks between the protein and PLP: the phosphate group is held in place by the donation of at least three hydrogen bonds while the carbonyl oxygen of the pyridine ring interacts with Gln301; Phe181 forms a π-π stacking interaction with the pyridine ring and works as a gate keeper with the assistance of Val300. All these interactions are hypothesized to help maintain free PMP in place inside the protein active site to facilitate the second half-reaction in GABA-AT: the regeneration of PLP-bound GABA-AT (i.e., the holoenzyme). Proteins 2016; 84:875-891. © 2016 Wiley Periodicals, Inc.


Subject(s)
4-Aminobutyrate Transaminase/antagonists & inhibitors , 4-Aminobutyrate Transaminase/metabolism , Enzyme Inhibitors/pharmacology , GABA Agents/pharmacology , Vigabatrin/pharmacology , 4-Aminobutyrate Transaminase/chemistry , Animals , Catalytic Domain/drug effects , Molecular Docking Simulation , Molecular Dynamics Simulation , Pyridoxal Phosphate/metabolism , Pyridoxamine/analogs & derivatives , Pyridoxamine/metabolism , Swine , gamma-Aminobutyric Acid/metabolism
13.
J Enzyme Inhib Med Chem ; 31(2): 295-301, 2016.
Article in English | MEDLINE | ID: mdl-25807299

ABSTRACT

The γ-aminobutyrate (GABA)-degradative enzyme GABA aminotransferase (GABA-AT) is regarded as an attractive target to control GABA levels in the central nervous system: this has important implications in the treatment of several neurological disorders and drug dependencies. We have investigated the ability of newly synthesized compounds to act as GABA-AT inhibitors. These compounds have a unique bicyclic structure: the carbocyclic ring bears the GABA skeleton, while the fused 3-Br-isoxazoline ring contains an electrophilic warhead susceptible of nucleophilic attack by an active site residue of the target enzyme. Out of the four compounds tested, only the one named (+)-3 was found to significantly inhibit mammalian GABA-AT in vitro. Docking studies, performed on the available structures of GABA-AT, support the experimental findings: out of the four tested compounds, only (+)-3 suitably orients the electrophilic 3-Br-isoxazoline warhead towards the active site nucleophilic residue Lys329, thereby explaining the irreversible inhibition of GABA-AT observed experimentally.


Subject(s)
4-Aminobutyrate Transaminase/antagonists & inhibitors , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , 4-Aminobutyrate Transaminase/chemistry , 4-Aminobutyrate Transaminase/metabolism , Amino Acids/chemistry , Amino Acids/pharmacology , Animals , Catalytic Domain , Chemistry Techniques, Synthetic , Drug Evaluation, Preclinical/methods , Enzyme Inhibitors/chemical synthesis , Molecular Docking Simulation , Structure-Activity Relationship
14.
ACS Chem Biol ; 10(9): 2087-98, 2015 Sep 18.
Article in English | MEDLINE | ID: mdl-26110556

ABSTRACT

When γ-aminobutyric acid (GABA), the major inhibitory neurotransmitter in the mammalian central nervous system, falls below a threshold level, seizures occur. One approach to raise GABA concentrations is to inhibit GABA aminotransferase (GABA-AT), a pyridoxal 5'-phosphate-dependent enzyme that degrades GABA. We have previously developed (1S,3S)-3-amino-4-difluoromethylene-1-cyclopentanoic acid (CPP-115), which is 186 times more efficient in inactivating GABA-AT than vigabatrin, the only FDA-approved inactivator of GABA-AT. We also developed (E)- and (Z)-(1S,3S)-3-amino-4-fluoromethylenyl-1-cyclopentanoic acid (1 and 2, respectively), monofluorinated analogs of CPP-115, which are comparable to vigabatrin in inactivating GABA-AT. Here, we report the mechanism of inactivation of GABA-AT by 1 and 2. Both produce a metabolite that induces disruption of the Glu270-Arg445 salt bridge to accommodate interaction between the metabolite formyl group and Arg445. This is the second time that Arg445 has interacted with a ligand and is involved in GABA-AT inactivation, thereby confirming the importance of Arg445 in future inactivator design.


Subject(s)
4-Aminobutyrate Transaminase/antagonists & inhibitors , 4-Aminobutyrate Transaminase/metabolism , Enzyme Inhibitors/pharmacology , Proline/analogs & derivatives , 4-Aminobutyrate Transaminase/chemistry , Animals , Crystallography, X-Ray , Enzyme Activation/drug effects , Enzyme Inhibitors/chemistry , Models, Molecular , Proline/chemistry , Proline/pharmacology , Swine
15.
J Biochem ; 158(3): 225-34, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25911692

ABSTRACT

Bacillus subtilis GabR is a transcriptional regulator consisting of a helix-turn-helix N-terminal DNA-binding domain, a pyridoxal 5'-phosphate (PLP)-binding C-terminal domain that has a structure homologous to aminotransferases, and a linker of 29 amino acid residues. In the presence of γ-aminobutyrate (GABA), GabR activates the transcription of gabT and gabD, which encode GABA aminotransferase and succinate semialdehyde dehydrogenase, respectively. We expressed N-terminal and C-terminal domain fragments (named N'-GabR and C'-GabR) in Escherichia coli cells, and obtained N'-GabR as a soluble monomer and C'-GabR as a soluble dimer. Spectroscopic studies suggested that C'-GabR contains PLP and binds to d-Ala, ß-Ala, d-Asn and d-Gln, as well as GABA, although the intact GabR binds only to GABA. N'-GabR does not bind to the DNA fragment containing the GabR-binding sequence regardless of the presence or absence of C'-GabR. A fusion protein consisting of N'-GabR and 2-aminoadipate aminotransferase of Thermus thermophilus bound to the DNA fragment. These results suggested that each domain of GabR could be an independent folding unit. The C-terminal domain provides the N-terminal domain with DNA-binding ability via dimerization. The N-terminal domain controls the ligand specificity of the C-terminal domain. Connection by the linker is indispensable for the mutual interaction of the domains.


Subject(s)
Bacillus subtilis/genetics , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Pyridoxal Phosphate/metabolism , Trans-Activators/chemistry , Trans-Activators/genetics , gamma-Aminobutyric Acid/metabolism , 4-Aminobutyrate Transaminase/chemistry , 4-Aminobutyrate Transaminase/genetics , Bacillus subtilis/enzymology , Escherichia coli , Gene Expression Regulation, Bacterial , Helix-Turn-Helix Motifs , Protein Folding , Protein Structure, Tertiary , Pyridoxal Phosphate/genetics , Structural Homology, Protein , Succinate-Semialdehyde Dehydrogenase/chemistry , Succinate-Semialdehyde Dehydrogenase/genetics , gamma-Aminobutyric Acid/genetics
16.
J Am Chem Soc ; 137(13): 4525-33, 2015 Apr 08.
Article in English | MEDLINE | ID: mdl-25781189

ABSTRACT

Low levels of γ-aminobutyric acid (GABA), one of two major neurotransmitters that regulate brain neuronal activity, are associated with many neurological disorders, such as epilepsy, Parkinson's disease, Alzheimer's disease, Huntington's disease, and cocaine addiction. One of the main methods to raise the GABA level in human brain is to use small molecules that cross the blood-brain barrier and inhibit the activity of γ-aminobutyric acid aminotransferase (GABA-AT), the enzyme that degrades GABA. We have designed a series of conformationally restricted tetrahydrothiophene-based GABA analogues with a properly positioned leaving group that could facilitate a ring-opening mechanism, leading to inactivation of GABA-AT. One compound in the series is 8 times more efficient an inactivator of GABA-AT than vigabatrin, the only FDA-approved inactivator of GABA-AT. Our mechanistic studies show that the compound inactivates GABA-AT by a new mechanism. The metabolite resulting from inactivation does not covalently bind to amino acid residues of GABA-AT but stays in the active site via H-bonding interactions with Arg-192, a π-π interaction with Phe-189, and a weak nonbonded S···O═C interaction with Glu-270, thereby inactivating the enzyme.


Subject(s)
4-Aminobutyrate Transaminase/metabolism , Drug Design , Enzyme Inhibitors/pharmacology , Thiophenes/pharmacology , 4-Aminobutyrate Transaminase/chemistry , Enzyme Activation/drug effects , Enzyme Inhibitors/chemistry , Kinetics , Models, Molecular , Protein Conformation , Thiophenes/chemistry , gamma-Aminobutyric Acid/chemistry , gamma-Aminobutyric Acid/metabolism
17.
J Am Chem Soc ; 137(7): 2628-40, 2015 Feb 25.
Article in English | MEDLINE | ID: mdl-25616005

ABSTRACT

γ-Aminobutyric acid aminotransferase (GABA-AT) is a pyridoxal 5'-phosphate (PLP)-dependent enzyme that degrades GABA, the principal inhibitory neurotransmitter in mammalian cells. When the concentration of GABA falls below a threshold level, convulsions can occur. Inhibition of GABA-AT raises GABA levels in the brain, which can terminate seizures as well as have potential therapeutic applications in treating other neurological disorders, including drug addiction. Among the analogues that we previously developed, (1S,3S)-3-amino-4-difluoromethylene-1-cyclopentanoic acid (CPP-115) showed 187 times greater potency than that of vigabatrin, a known inactivator of GABA-AT and approved drug (Sabril) for the treatment of infantile spasms and refractory adult epilepsy. Recently, CPP-115 was shown to have no adverse effects in a Phase I clinical trial. Here we report a novel inactivation mechanism for CPP-115, a mechanism-based inactivator that undergoes GABA-AT-catalyzed hydrolysis of the difluoromethylene group to a carboxylic acid with concomitant loss of two fluoride ions and coenzyme conversion to pyridoxamine 5'-phosphate (PMP). The partition ratio for CPP-115 with GABA-AT is about 2000, releasing cyclopentanone-2,4-dicarboxylate (22) and two other precursors of this compound (20 and 21). Time-dependent inactivation occurs by a conformational change induced by the formation of the aldimine of 4-aminocyclopentane-1,3-dicarboxylic acid and PMP (20), which disrupts an electrostatic interaction between Glu270 and Arg445 to form an electrostatic interaction between Arg445 and the newly formed carboxylate produced by hydrolysis of the difluoromethylene group in CPP-115, resulting in a noncovalent, tightly bound complex. This represents a novel mechanism for inactivation of GABA-AT and a new approach for the design of mechanism-based inactivators in general.


Subject(s)
4-Aminobutyrate Transaminase/antagonists & inhibitors , 4-Aminobutyrate Transaminase/metabolism , Enzyme Inhibitors/pharmacology , Proline/analogs & derivatives , 4-Aminobutyrate Transaminase/chemistry , Enzyme Activation/drug effects , Fluorine/metabolism , Humans , Models, Molecular , Proline/pharmacology , Protein Conformation , Pyridoxal Phosphate/metabolism
18.
Med Res Rev ; 35(2): 286-305, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25145640

ABSTRACT

Ornithine aminotransferase (OAT) and γ-aminobutyric acid aminotransferase (GABA-AT) are classified under the same evolutionary subgroup and share a large portion of structural, functional, and mechanistic features. Therefore, it is not surprising that many molecules that bind to GABA-AT also bind well to OAT. Unlike GABA-AT, OAT had not been viewed as a potential therapeutic target until recently; consequently, the number of therapeutically viable molecules that target OAT is very limited. In this review the two enzymes are compared with respect to their active-site structures, catalytic and inactivation mechanisms, and selective inhibitors. Insight is offered that could aid in the design and development of new selective inhibitors of OAT for the treatment of cancer.


Subject(s)
4-Aminobutyrate Transaminase/metabolism , Antineoplastic Agents/pharmacology , Drug Design , Ornithine-Oxo-Acid Transaminase/metabolism , 4-Aminobutyrate Transaminase/antagonists & inhibitors , 4-Aminobutyrate Transaminase/chemistry , Amino Acid Sequence , Animals , Catalytic Domain , Humans , Ornithine-Oxo-Acid Transaminase/antagonists & inhibitors , Ornithine-Oxo-Acid Transaminase/chemistry , Substrate Specificity/drug effects
19.
Cent Nerv Syst Agents Med Chem ; 14(1): 39-42, 2014.
Article in English | MEDLINE | ID: mdl-25277157

ABSTRACT

Convulsion generally occurs as a result of the diminishing concentration of GABA below a threshold level in the brain. This degradation pathway of GABA is catalyzed by the γ-aminobutyric acid amino transferase. The objective of the current study is to propose the binding interaction of 3a, 4-Dihydro-3-H-indeno [1, 2-C] pyrazole-2-Carboxamide/ Carbothioamides anticonvulsant analogs with a three-dimensional structural model of the γ -aminobutyric acid amino transferase. For a flexible type of molecular docking, we proposed that these molecules could successfully bind to the active pocket of the enzyme with good predicted affinities in comparison to standard vigabatrin. In this series, 4b, 4c, 4i, 4f and 4a showed significant binding free energy of -9.64, -9.31, -9.01, -8.99 and -8.29 with predicted inhibitory constant values of 0.086, 0.149, 0.237, 0.257 and 0.831 µM, respectively.


Subject(s)
4-Aminobutyrate Transaminase/chemistry , 4-Aminobutyrate Transaminase/metabolism , Anticonvulsants/chemistry , Anticonvulsants/metabolism , Crystallography , Humans , Molecular Docking Simulation , Protein Binding/physiology
20.
Arch Pharm (Weinheim) ; 347(12): 958-68, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25251582

ABSTRACT

New hydrazone incorporated triazines were designed and synthesized using an appropriate synthetic route with regard to essential pharmacophores, and evaluated for their anticonvulsant activity through maximal electroshock seizure (MES) and subcutaneous pentylenetetrazole-induced seizure (scPTZ) screenings. Among the tested compounds, 4-[{2-(5-(3-chlorobenzyl)-3-phenyl-1,2,4-triazine-6-yl)hydrazono}methyl]-N,N-dimethylaniline 6k (MES ED50 54.31, scPTZ ED50 92.01) and 4-[{2-(5-(4-chlorobenzyl)-3-phenyl-1,2,4-triazine-6-yl)hydrazono}methyl]-N,N-dimethylaniline 6r (MES ED50 46.05, scPTZ ED50 83.90) emerged as the most active anticonvulsant agents having GABAergic effects. Compounds 6k and 6r also showed lesser CNS depressant effect than the standard drug carbamazepine. To obtain further insights into the binding interactions of these molecules, molecular docking studies were carried out.


Subject(s)
Anticonvulsants/chemical synthesis , Anticonvulsants/pharmacology , Drug Design , Hydrazones/chemical synthesis , Hydrazones/pharmacology , Seizures/prevention & control , Triazines/chemical synthesis , Triazines/pharmacology , 4-Aminobutyrate Transaminase/chemistry , 4-Aminobutyrate Transaminase/metabolism , Animals , Anticonvulsants/metabolism , Binding Sites , Catalytic Domain , Computer-Aided Design , Disease Models, Animal , Electroshock , Female , Humans , Hydrazones/metabolism , Male , Mice , Molecular Docking Simulation , Molecular Structure , Pentylenetetrazole , Protein Binding , Protein Conformation , Seizures/etiology , Seizures/physiopathology , Structure-Activity Relationship , Triazines/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL