Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 588
Filter
1.
J Enzyme Inhib Med Chem ; 36(1): 2016-2024, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34514924

ABSTRACT

Many studies have focussed on modulating the activity of γ-aminobutyric acid transaminase (GABA-T), a GABA-catabolizing enzyme, for treating neurological diseases, such as epilepsy and drug addiction. Nevertheless, human GABA-T synthesis and purification have not been established. Thus, biochemical and drug design studies on GABA-T have been performed by using porcine GABA-T mostly and even bacterial GABA-T. Here we report an optimised protocol for overexpression of 6xHis-tagged human GABA-T in human cells followed by a two-step protein purification. Then, we established an optimised human GABA-T (0.5 U/mg) activity assay. Finally, we compared the difference between human and bacterial GABA-T in sensitivity to two irreversible GABA-T inhibitors, gabaculine and vigabatrin. Human GABA-T in homodimeric form showed 70-fold higher sensitivity to vigabatrin than bacterial GABA-T in multimeric form, indicating the importance of using human GABA-T. In summary, our newly developed protocol can be an important first step in developing more effective human GABA-T modulators.


Subject(s)
4-Aminobutyrate Transaminase/biosynthesis , 4-Aminobutyrate Transaminase/isolation & purification , 4-Aminobutyrate Transaminase/antagonists & inhibitors , Drug Evaluation, Preclinical , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , High-Throughput Screening Assays , Humans , Recombinant Proteins/biosynthesis , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism
2.
ACS Chem Biol ; 16(4): 615-630, 2021 04 16.
Article in English | MEDLINE | ID: mdl-33735567

ABSTRACT

((S)-3-Amino-(difluoromethylenyl)cyclopent-1-ene-1-carboxylic acid (OV329) is a recently discovered inactivator of γ-aminobutyric acid aminotransferase (GABA-AT), which has 10 times better inactivation efficiency than its predecessor, CPP-115, despite the only structural difference being an endocyclic double bond in OV329. Both compounds are mechanism-based enzyme inactivators (MBEIs), which inactivate GABA-AT by a similar mechanism. Here, a combination of a variety of computational chemistry tools and experimental methods, including quantum mechanical (QM) calculations, molecular dynamic simulations, progress curve analysis, and deuterium kinetic isotope effect (KIE) experiments, are utilized to comprehensively study the mechanism of inactivation of GABA-AT by CPP-115 and OV329 and account for their experimentally obtained global kinetic parameters kinact and KI. Our first key finding is that the rate-limiting step of the inactivation mechanism is the deprotonation step, and according to QM calculations and the KIE experiments, kinact accurately represents the enhancement of the rate-limiting step for the given mechanism. Second, the present study shows that the widely used simple QM models do not accurately represent the geometric criteria that are present in the enzyme for the deprotonation step. In contrast, QM cluster models successfully represent both the ground state destabilization and the transition state stabilization, as revealed by natural bond orbital analysis. Furthermore, the globally derived KI values for both of the inactivators represent the inhibitor constants for the initial binding complexes (Kd) and indicate the inactivator competition with the substrate according to progress curve analysis and the observed binding isotope effect. The configurational entropy loss accounts for the difference in KI values between the inactivators. The approach we describe in this work can be employed to determine the validity of globally derived parameters in the process of MBEI optimization for given inactivation mechanisms.


Subject(s)
4-Aminobutyrate Transaminase/metabolism , Proline/analogs & derivatives , 4-Aminobutyrate Transaminase/antagonists & inhibitors , Catalysis , Kinetics , Molecular Dynamics Simulation , Proline/pharmacology , Quantum Theory , Reproducibility of Results
3.
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
4.
Mini Rev Med Chem ; 20(14): 1447-1460, 2020.
Article in English | MEDLINE | ID: mdl-32072906

ABSTRACT

BACKGROUND: Since their introduction in the virtual screening field, Receiver Operating Characteristic (ROC) curve-derived metrics have been widely used for benchmarking of computational methods and algorithms intended for virtual screening applications. Whereas in classification problems, the ratio between sensitivity and specificity for a given score value is very informative, a practical concern in virtual screening campaigns is to predict the actual probability that a predicted hit will prove truly active when submitted to experimental testing (in other words, the Positive Predictive Value - PPV). Estimation of such probability is however, obstructed due to its dependency on the yield of actives of the screened library, which cannot be known a priori. OBJECTIVE: To explore the use of PPV surfaces derived from simulated ranking experiments (retrospective virtual screening) as a complementary tool to ROC curves, for both benchmarking and optimization of score cutoff values. METHODS: The utility of the proposed approach is assessed in retrospective virtual screening experiments with four datasets used to infer QSAR classifiers: inhibitors of Trypanosoma cruzi trypanothione synthetase; inhibitors of Trypanosoma brucei N-myristoyltransferase; inhibitors of GABA transaminase and anticonvulsant activity in the 6 Hz seizure model. RESULTS: Besides illustrating the utility of PPV surfaces to compare the performance of machine learning models for virtual screening applications and to select an adequate score threshold, our results also suggest that ensemble learning provides models with better predictivity and more robust behavior. CONCLUSION: PPV surfaces are valuable tools to assess virtual screening tools and choose score thresholds to be applied in prospective in silico screens. Ensemble learning approaches seem to consistently lead to improved predictivity and robustness.


Subject(s)
Machine Learning , Quantitative Structure-Activity Relationship , 4-Aminobutyrate Transaminase/antagonists & inhibitors , 4-Aminobutyrate Transaminase/metabolism , Animals , Anticonvulsants/chemistry , Anticonvulsants/therapeutic use , Area Under Curve , Protozoan Proteins/antagonists & inhibitors , Protozoan Proteins/metabolism , ROC Curve , Seizures/drug therapy , Seizures/pathology , Trypanosoma/metabolism
5.
Mol Brain ; 13(1): 15, 2020 02 04.
Article in English | MEDLINE | ID: mdl-32019580

ABSTRACT

Anxiety disorder is highly prevalent worldwide and represents a chronic and functionally disabling condition, with high levels of psychological stress characterized by cognitive and physiological symptoms. Scopoletin (SP), a main active compound in Angelica dahurica, is traditionally used for the treatment of headache, rhinitis, pain, and other conditions. Here, we evaluated the effects of SP in a mouse model of complete Freund's adjuvant (CFA)-induced chronic inflammation anxiety. SP (2.0, 10.0, 50.0 mg/kg) administration for 2 weeks dose-dependently ameliorated CFA-induced anxiety-like behaviors in the open field test and elevated plus maze test. Moreover, we found that SP treatment inhibited microglia activation and decreased both peripheral and central IL-1ß, IL-6, and TNF-α levels in a dose-dependent manner. Additionally, the imbalance in excitatory/inhibitory receptors and neurotransmitters in the basolateral nucleus after CFA injection was also modulated by SP administration. Our findings indicate that the inhibition of the nuclear factor-kappa B and mitogen-activated protein kinase signaling pathways involving anti-inflammatory activities and regulation of the excitatory/inhibitory balance can be attributed to the anxiolytic effects of SP. Moreover, our molecular docking analyses show that SP also has good affinity for gamma-aminobutyric acid (GABA) transaminase and GABAA receptors. Therefore, these results suggest that SP could be a candidate compound for anxiolytic therapy and for use as a structural base for developing new drugs.


Subject(s)
Angelica/chemistry , Anti-Anxiety Agents/therapeutic use , Anxiety/drug therapy , Drugs, Chinese Herbal/therapeutic use , GABA-A Receptor Agonists/therapeutic use , Phytotherapy , Scopoletin/therapeutic use , 4-Aminobutyrate Transaminase/antagonists & inhibitors , Amygdala/chemistry , Amygdala/drug effects , Animals , Anti-Anxiety Agents/pharmacology , Anxiety/etiology , Cytokines/metabolism , Disease Models, Animal , Drugs, Chinese Herbal/pharmacology , Elevated Plus Maze Test , Freund's Adjuvant/toxicity , GABA-A Receptor Agonists/pharmacology , Inflammation/chemically induced , Inflammation/psychology , MAP Kinase Signaling System/drug effects , Male , Mice , Mice, Inbred C57BL , Microglia/drug effects , Models, Molecular , Molecular Docking Simulation , NF-kappa B/metabolism , Neurotransmitter Agents/metabolism , Open Field Test , Protein Conformation , Receptors, Neurotransmitter/metabolism , Scopoletin/pharmacology
6.
Pharmacol Rep ; 72(2): 322-330, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32048251

ABSTRACT

BACKGROUND: The goal of our study was to examine the long-term effect of vigabatrin (VGB), a γ-aminobutyric acid aminotransferase (GABA-AT) inhibitor on clonazepam (CLO), ethosuximide (ETX) and valproate (VPA) anticonvulsive activity against pentylenetetrazole (PTZ)-induced seizures in mice. METHODS: VGB was administered for 3 and 7 days. Convulsions were evoked by PTZ at its CD97 (99 mg/kg). The influence of CLO, ETX and VPA alone or in combination with VGB on motor performance and long-term memory was analyzed. γ-aminobutyric acid (GABA) concentration in mice brain and plasma as well as glutamate decarboxylase (GAD) activity was measured. RESULTS: After 3 days of treatment, VGB in doses up to 500 mg/kg increased PTZ-induced seizure threshold, whereas after 7 days VGB (at the dose of 125 mg/kg) inhibited clonic seizures in experimental mice. 7 days of VGB administration did not change the protective effect of CLO, ETX and VPA against PTZ-induced seizures. 7 days of VGB treatment at a subthreshold dose of 75 mg/kg decreased TD50 of ETX and CLO in the chimney test, but did not affect TD50 value for VPA. 7 days of VGB administration in combination with AEDs did not affect long-term memory in mice. VGB after 3 days or 7 days of administration increased brain GABA concentration. GAD activity was decreased after 3 and 7 days of VGB administration. CONCLUSIONS: The presented results confirm anticonvulsive activity of VGB through GABA metabolism alteration and suggest care when combining VGB with ETX or CLO in the therapy.


Subject(s)
4-Aminobutyrate Transaminase/antagonists & inhibitors , Anticonvulsants/pharmacology , Brain/drug effects , Seizures/drug therapy , Vigabatrin/pharmacology , gamma-Aminobutyric Acid/metabolism , Animals , Brain/metabolism , Clonazepam/pharmacology , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Therapy, Combination , Ethosuximide/pharmacology , Mice , Pentylenetetrazole/pharmacology , Seizures/metabolism , Time Factors , Valproic Acid/pharmacology , Vigabatrin/administration & dosage
7.
JCI Insight ; 4(18)2019 09 19.
Article in English | MEDLINE | ID: mdl-31534049

ABSTRACT

Recently we demonstrated that ablation of the DNA methyltransferase enzyme, Dnmt3b, resulted in catabolism and progression of osteoarthritis (OA) in murine articular cartilage through a mechanism involving increased mitochondrial respiration. In this study, we identify 4-aminobutyrate aminotransferase (Abat) as a downstream target of Dnmt3b. Abat is an enzyme that metabolizes γ-aminobutyric acid to succinate, a key intermediate in the tricarboxylic acid cycle. We show that Dnmt3b binds to the Abat promoter, increases methylation of a conserved CpG sequence just upstream of the transcriptional start site, and inhibits Abat expression. Dnmt3b deletion in articular chondrocytes results in reduced methylation of the CpG sequence in the Abat promoter, which subsequently increases expression of Abat. Increased Abat expression in chondrocytes leads to enhanced mitochondrial respiration and elevated expression of catabolic genes. Overexpression of Abat in murine knee joints via lentiviral injection results in accelerated cartilage degradation following surgical induction of OA. In contrast, lentiviral-based knockdown of Abat attenuates the expression of IL-1ß-induced catabolic genes in primary murine articular chondrocytes in vitro and also protects against murine articular cartilage degradation in vivo. Strikingly, treatment with the FDA-approved small-molecule Abat inhibitor, vigabatrin, significantly prevents the development of injury-induced OA in mice. In summary, these studies establish Abat as an important new target for therapies to prevent OA.


Subject(s)
4-Aminobutyrate Transaminase/genetics , Cartilage, Articular/injuries , DNA (Cytosine-5-)-Methyltransferases/metabolism , Osteoarthritis, Knee/pathology , Vigabatrin/pharmacology , 4-Aminobutyrate Transaminase/antagonists & inhibitors , 4-Aminobutyrate Transaminase/metabolism , Animals , Cartilage, Articular/cytology , Cartilage, Articular/drug effects , Cartilage, Articular/pathology , Cells, Cultured , Chondrocytes/cytology , Chondrocytes/drug effects , Chondrocytes/immunology , Chondrocytes/pathology , CpG Islands/genetics , DNA Methylation/genetics , Disease Models, Animal , Gene Knockdown Techniques , Humans , Interleukin-1beta/immunology , Interleukin-1beta/metabolism , Male , Mice , Mitochondria/metabolism , Osteoarthritis, Knee/drug therapy , Osteoarthritis, Knee/etiology , Oxidative Phosphorylation/drug effects , Primary Cell Culture , Promoter Regions, Genetic/genetics , Transcription Initiation Site , Transcription, Genetic , Vigabatrin/therapeutic use , DNA Methyltransferase 3B
8.
Neurochem Int ; 125: 151-162, 2019 05.
Article in English | MEDLINE | ID: mdl-30822440

ABSTRACT

The anticonvulsant vigabatrin (VGB; SabrilR) irreversibly inhibits GABA transaminase to increase neural GABA, yet its mechanism of retinal toxicity remains unclear. VGB is suggested to alter several amino acids, including homocarnosine, ß-alanine, ornithine, glycine, taurine, and 2-aminoadipic acid (AADA), the latter a homologue of glutamic acid. Here, we evaluate the effect of VGB on amino acid concentrations in mice, employing a continuous VGB infusion (subcutaneously implanted osmotic minipumps), dose-escalation paradigm (35-140 mg/kg/d, 12 days), and amino acid quantitation in eye, visual and prefrontal cortex, total brain, liver and plasma. We hypothesized that continuous VGB dosing would reveal numerous hitherto undescribed amino acid disturbances. Consistent amino acid elevations across tissues included GABA, ß-alanine, carnosine, ornithine and AADA, as well as neuroactive aspartic and glutamic acids, serine and glycine. Maximal increase of AADA in eye occurred at 35 mg/kg/d (41 ±â€¯2 nmol/g (n = 21, vehicle) to 60 ±â€¯8.5 (n = 8)), and at 70 mg/kg/d for brain (97 ±â€¯6 (n = 21) to 145 ±â€¯6 (n = 6)), visual cortex (128 ±â€¯6 to 215 ±â€¯19) and prefrontal cortex (124 ±â€¯11 to 200 ±â€¯13; mean ±â€¯SEM; p < 0.05), the first demonstration of tissue AADA accumulation with VGB in mammal. VGB effects on basic amino acids, including guanidino-species, suggested the capacity of VGB to alter urea cycle function and nitrogen disposal. The known toxicity of AADA in retinal glial cells highlights new avenues for assessing VGB retinal toxicity and other off-target effects.


Subject(s)
4-Aminobutyrate Transaminase/metabolism , Amino Acids/metabolism , Metabolome/physiology , Metabolomics/methods , Vigabatrin/pharmacology , 4-Aminobutyrate Transaminase/antagonists & inhibitors , Amino Acids/blood , Amino Acids/genetics , Animals , Anticonvulsants/pharmacology , Brain/drug effects , Brain/metabolism , Dose-Response Relationship, Drug , Liver/drug effects , Liver/metabolism , Male , Metabolome/drug effects , Mice , Mice, Inbred C57BL , Retina/drug effects , Retina/metabolism
9.
Pharmacol Res Perspect ; 7(1): e00456, 2019 02.
Article in English | MEDLINE | ID: mdl-30631446

ABSTRACT

Vigabatrin (VGB; (S)-(+)/(R)-(-) 4-aminohex-5-enoic acid), an antiepileptic irreversibly inactivating GABA transaminase (GABA-T), manifests use-limiting ocular toxicity. Hypothesizing that the active S enantiomer of VGB would preferentially accumulate in eye and visual cortex (VC) as one potential mechanism for ocular toxicity, we infused racemic VGB into mice via subcutaneous minipump at 35, 70, and 140 mg/kg/d (n = 6-8 animals/dose) for 12 days. VGB enantiomers, total GABA and ß-alanine (BALA), 4-guanidinobutyrate (4-GBA), and creatine were quantified by mass spectrometry in eye, brain, liver, prefrontal cortex (PFC), and VC. Plasma VGB concentrations increased linearly by dose (3 ± 0.76 (35 mg/kg/d); 15.1 ± 1.4 (70 mg/kg/d); 34.6 ± 3.2 µmol/L (140 mg/kg/d); mean ± SEM) with an S/R ratio of 0.74 ± 0.02 (n = 14). Steady state S/R ratios (35, 70 mg/kg/d doses) were highest in eye (5.5 ± 0.2; P < 0.0001), followed by VC (3.9 ± 0.4), PFC (3.6 ± 0.3), liver (2.9 ± 0.1), and brain (1.5 ± 0.1; n = 13-14 each). Total VGB content of eye exceeded that of brain, PFC and VC at all doses. High-dose VGB diminished endogenous metabolite production, especially in PFC and VC. GABA significantly increased in all tissues (all doses) except brain; BALA increases were confined to liver and VC; and 4-GBA was prominently increased in brain, PFC and VC (and eye at high dose). Linear correlations between enantiomers and GABA were observed in all tissues, but only in PFC/VC for BALA, 4-GBA, and creatine. Preferential accumulation of the VGB S isomer in eye and VC may provide new insight into VGB ocular toxicity.


Subject(s)
Anticonvulsants/pharmacokinetics , Vigabatrin/pharmacokinetics , Vision Disorders/prevention & control , 4-Aminobutyrate Transaminase/antagonists & inhibitors , Animals , Anticonvulsants/adverse effects , Anticonvulsants/chemistry , Drug Evaluation, Preclinical , Eye/drug effects , Eye/metabolism , Male , Mice , Mice, Inbred C57BL , Models, Animal , Stereoisomerism , Tissue Distribution , Vigabatrin/adverse effects , Vigabatrin/chemistry , Vision Disorders/chemically induced , Visual Cortex/drug effects , Visual Cortex/metabolism , Visual Fields/drug effects
10.
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
11.
Sci Rep ; 8(1): 13200, 2018 09 04.
Article in English | MEDLINE | ID: mdl-30181656

ABSTRACT

Metabolite-specific, scalar spin-spin coupling constant (J)-editing 1H MRS methods have become gold-standard for measuring brain γ-amino butyric acid (GABA) levels in human brain. Localized, two-dimensional (2D) 1H MRS technology offers an attractive alternative as it significantly alleviates the problem of severe metabolite signal overlap associated with standard 1D MRS and retains spectroscopic information for all MRS-detectable species. However, for metabolites found at low concentration, a direct, in vivo, comprehensive methods comparison is challenging and has not been reported to date. Here, we document an assessment of comparability between 2D 1H MRS and J-editing methods for measuring GABA in human brain. This clinical study is unique in that it involved chronic administration a GABA-amino transferase (AT) inhibitor (CPP-115), which induces substantial increases in brain GABA concentration, with normalization after washout. We report a qualitative and quantitative comparison between these two measurement techniques. In general, GABA concentration changes detected using J-editing were closely mirrored by the 2D 1H MRS time courses. The data presented are particularly encouraging considering recent 2D 1H MRS methodological advances are continuing to improve temporal resolution and spatial coverage for achieving whole-brain, multi-metabolite mapping.


Subject(s)
4-Aminobutyrate Transaminase/antagonists & inhibitors , Brain/metabolism , Enzyme Inhibitors/pharmacology , Proline/analogs & derivatives , gamma-Aminobutyric Acid/metabolism , Adolescent , Adult , Brain/drug effects , Humans , Male , Middle Aged , Proline/pharmacology , Proton Magnetic Resonance Spectroscopy/methods , Young Adult , gamma-Aminobutyric Acid/analysis
12.
Org Lett ; 20(15): 4589-4592, 2018 08 03.
Article in English | MEDLINE | ID: mdl-30009604

ABSTRACT

( S)-3-Amino-4-(difluoromethylenyl)cyclopent-1-ene-1-carboxylic acid (OV329, 1) is being developed for the treatment of epilepsy and addiction. The previous 14-step synthesis of OV329 was low yielding, involved an unselective α-elimination to form the cyclopentene, required the use of tert-butyllithium, and produced toxic selenium byproducts in the penultimate step. A new synthesis, which avoids the aforementioned issues, was carried out on large scale, reducing the step count from 14 to 9 steps and increasing the overall yield from 3.7% to 8.1%.


Subject(s)
4-Aminobutyrate Transaminase/antagonists & inhibitors , Cyclopentanes/chemistry , Enzyme Inhibitors/chemical synthesis , Transaminases/antagonists & inhibitors , Molecular Structure , Organometallic Compounds/chemistry , Phenylacetates/chemistry , Selenium Compounds/chemistry , Stereoisomerism , Structure-Activity Relationship
13.
Molecules ; 23(5)2018 05 09.
Article in English | MEDLINE | ID: mdl-29747438

ABSTRACT

γ-Aminobutyric acid (GABA) is the most important inhibitory neurotransmitter in the central nervous system, and a deficiency of GABA is associated with serious neurological disorders. Due to its low lipophilicity, there has been an intensive search for new molecules with increased lipophilicity to cross the blood-brain barrier to raise GABA concentrations. We have designed and evaluated in vitro and in silico some new analogues of GABA, where the nitrogen atom at the γ-position is embedded in heterocyclic scaffolds and determined their inhibitory potential over the GABA-AT enzyme from Pseudomonas fluorescens. These modifications lead to compounds with inhibitory activity as it occurs with compounds 18a and 19a. The construction of Pseudomonas fluorescens and human GABA-AT models were carried out by homology modeling. Docking assays were done for these compounds over the GABA-AT enzyme models where 19a showed a strong interaction with both GABA-AT enzymes.


Subject(s)
4-Aminobutyrate Transaminase/antagonists & inhibitors , Computer Simulation , Heterocyclic Compounds/chemistry , Heterocyclic Compounds/pharmacology , Models, Molecular , Pseudomonas fluorescens/enzymology , gamma-Aminobutyric Acid/analogs & derivatives , Catalytic Domain , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Heterocyclic Compounds/chemical synthesis , Humans , Hydrogen Bonding , Molecular Docking Simulation , Static Electricity
15.
Chem Rev ; 118(7): 4037-4070, 2018 04 11.
Article in English | MEDLINE | ID: mdl-29569907

ABSTRACT

When the brain concentration of the inhibitory neurotransmitter γ-aminobutyric acid (GABA) diminishes below a threshold level, the excess neuronal excitation can lead to convulsions. This imbalance in neurotransmission can be corrected by inhibition of the enzyme γ-aminobutyric acid aminotransferase (GABA-AT), which catalyzes the conversion of GABA to the excitatory neurotransmitter l-glutamic acid. It also has been found that raising GABA levels can antagonize the rapid elevation and release of dopamine in the nucleus accumbens, which is responsible for the reward response in addiction. Therefore, the design of new inhibitors of GABA-AT, which increases brain GABA levels, is an important approach to new treatments for epilepsy and addiction. This review summarizes findings over the last 40 or so years of mechanism-based inactivators (unreactive compounds that require the target enzyme to catalyze their conversion to the inactivating species, which inactivate the enzyme prior to their release) of GABA-AT with emphasis on their catalytic mechanisms of inactivation, presented according to organic chemical mechanism, with minimal pharmacology, except where important for activity in epilepsy and addiction. Patents, abstracts, and conference proceedings are not covered in this review. The inactivation mechanisms described here can be applied to the inactivations of a wide variety of unrelated enzymes.


Subject(s)
4-Aminobutyrate Transaminase/antagonists & inhibitors , Enzyme Inhibitors/chemistry , Epilepsy/drug therapy , Substance-Related Disorders/drug therapy , 4-Aminobutyrate Transaminase/metabolism , Animals , Drug Design , Enzyme Inhibitors/pharmacology , Humans , Models, Molecular
16.
Biochem J ; 475(4): 749-758, 2018 02 16.
Article in English | MEDLINE | ID: mdl-29339464

ABSTRACT

Propionic acidemia is the accumulation of propionate in blood due to dysfunction of propionyl-CoA carboxylase. The condition causes lethargy and striatal degeneration with motor impairment in humans. How propionate exerts its toxic effect is unclear. Here, we show that intravenous administration of propionate causes dose-dependent propionate accumulation in the brain and transient lethargy in mice. Propionate, an inhibitor of histone deacetylase, entered GABAergic neurons, as could be seen from increased neuronal histone H4 acetylation in the striatum and neocortex. Propionate caused an increase in GABA (γ-amino butyric acid) levels in the brain, suggesting inhibition of GABA breakdown. In vitro propionate inhibited GABA transaminase with a Ki of ∼1 mmol/l. In isolated nerve endings, propionate caused increased release of GABA to the extracellular fluid. In vivo, propionate reduced cerebral glucose metabolism in both striatum and neocortex. We conclude that propionate-induced inhibition of GABA transaminase causes accumulation of GABA in the brain, leading to increased extracellular GABA concentration, which inhibits neuronal activity and causes lethargy. Propionate-mediated inhibition of neuronal GABA transaminase, an enzyme of the inner mitochondrial membrane, indicates entry of propionate into neuronal mitochondria. However, previous work has shown that neurons are unable to metabolize propionate oxidatively, leading us to conclude that propionyl-CoA synthetase is probably absent from neuronal mitochondria. Propionate-induced inhibition of energy metabolism in GABAergic neurons may render the striatum, in which >90% of the neurons are GABAergic, particularly vulnerable to degeneration in propionic acidemia.


Subject(s)
4-Aminobutyrate Transaminase/antagonists & inhibitors , GABAergic Neurons/drug effects , Lethargy/metabolism , Propionates/administration & dosage , Propionic Acidemia/metabolism , 4-Aminobutyrate Transaminase/metabolism , Animals , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Corpus Striatum/pathology , GABAergic Neurons/metabolism , Glucose/metabolism , Histone Deacetylase Inhibitors/administration & dosage , Histone Deacetylases , Humans , Lethargy/chemically induced , Lethargy/physiopathology , Methylmalonyl-CoA Decarboxylase/metabolism , Mice , Mitochondria/drug effects , Mitochondria/metabolism , Neocortex/drug effects , Neocortex/metabolism , Neocortex/pathology , Propionic Acidemia/chemically induced , Propionic Acidemia/physiopathology , gamma-Aminobutyric Acid/metabolism
17.
Bioorg Chem ; 77: 56-67, 2018 04.
Article in English | MEDLINE | ID: mdl-29331765

ABSTRACT

Even after considerable advances in the field of epilepsy treatment, convulsions are inefficiently controlled by standard drug therapy. Herein, a series of pyrimidine-carbothioamide derivatives 4(a-t) was designed as anticonvulsant agents by doing some important structural modifications in well-known anticonvulsant drugs. Two classical animal models were used for the in vivo anticonvulsant screening, maximum electroshock seizure (MES) and subcutaneous pentylenetetrazole (scPTZ) models; followed by motor impairment study by rotarod method. The most active compound 4g effectively suppressed seizure effect in both the animal models with median doses of 15.6 mg/kg (MES ED50), 278.4 mg/kg (scPTZ ED50) and 534.4 mg/kg (TD50) with no sign of neurotoxicity. Furthermore, in vitro GABA-AT enzyme activity assay of 4g showed inhibitory potency (IC50) of 12.23 µM. The docking study also favored the animal studies.


Subject(s)
4-Aminobutyrate Transaminase/antagonists & inhibitors , Anticonvulsants/pharmacology , Enzyme Inhibitors/pharmacology , Pyrimidines/pharmacology , Seizures/drug therapy , Thioamides/pharmacology , 4-Aminobutyrate Transaminase/metabolism , Animals , Anticonvulsants/chemical synthesis , Anticonvulsants/chemistry , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Electroshock , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Male , Mice , Models, Molecular , Molecular Structure , Pentylenetetrazole , Pyrimidines/chemical synthesis , Pyrimidines/chemistry , Seizures/chemically induced , Structure-Activity Relationship , Thioamides/chemical synthesis , Thioamides/chemistry
18.
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
19.
Neuropsychopharmacology ; 43(3): 646-654, 2018 02.
Article in English | MEDLINE | ID: mdl-28741622

ABSTRACT

CPP-115, a next-generation γ-amino butyric acid (GABA)-aminotransferase (AT) inhibitor, shows comparable pharmacokinetics, improved safety and tolerability, and a more favorable toxicity profile when compared with vigabatrin. The pharmacodynamic characteristics of CPP-115 remain to be evaluated. The present study employed state-of-the-art proton magnetic resonance spectroscopy techniques to measure changes in brain GABA+ (the composite resonance of GABA, homocarnosine, and macromolecules) concentrations in healthy subjects receiving oral daily doses of CPP-115 or placebo. Six healthy adult males were randomized to receive either single daily 80 mg doses of CPP-115 (n=4) or placebo (n=2) for 6, 10, or 14 days. Metabolite-edited spectra and two-dimensional J-resolved spectroscopy data were acquired from the parietal-occipital cortex and supplementary motor area in all subjects. Four scans were performed in each subject that included a predrug baseline measure, two scans during the dosing timeframe, and a final scan that occurred 1 week after drug cessation. CPP-115 induced robust and significant increases in brain GABA+ concentrations that ranged between 52 and 141% higher than baseline values. Elevated GABA+ concentrations returned to baseline values following drug clearance. Subjects receiving placebo showed no significant changes in GABA+ concentration. CPP-115-induced changes were exclusive to GABA and homocarnosine, and CPP-115 afforded brain GABA+ concentration changes comparable to or greater than previous vigabatrin spectroscopy studies in healthy epilepsy-naive subjects. The return to baseline GABA+ concentration indicates the reversible GABA-AT resynthesis following drug washout. These preliminary data warrant further spectroscopy studies that characterize the acute pharmacodynamic effects of CPP-115 with additional dose-descending measures.


Subject(s)
4-Aminobutyrate Transaminase/antagonists & inhibitors , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Enzyme Inhibitors/pharmacology , Proline/analogs & derivatives , Adult , Double-Blind Method , Humans , Magnetic Resonance Imaging , Male , Proline/pharmacology , Proton Magnetic Resonance Spectroscopy , Time Factors
20.
Bioorg Chem ; 74: 166-178, 2017 10.
Article in English | MEDLINE | ID: mdl-28806600

ABSTRACT

Several new 5,6-dihydropyrimidine-2(1H)-thione derivatives have been prepared and investigated for their potencies for anticonvulsant activity against maximal electroshock (MES) and subcutaneous pentylenetetrazole (scPTZ) test in mice. The acute neurotoxicity was measured by rotarod test. Compounds 3c and 3l were found active in both of the animal models. Further, in vitro GABA-AT enzyme activity assay was carried out to investigate the possible mechanism of action through GABA-AT inhibition. The most potent compounds 3c and 3l showed inhibitory potency (IC50) of 18.42µM and 19.23µM, respectively. The molecular modeling was performed for all the synthesized compounds. The docking results were found in concordant with the observed animal studies.


Subject(s)
4-Aminobutyrate Transaminase/antagonists & inhibitors , Anticonvulsants/pharmacology , Drug Design , Enzyme Inhibitors/pharmacology , Pyrimidines/pharmacology , Seizures/drug therapy , Thiones/pharmacology , 4-Aminobutyrate Transaminase/metabolism , Animals , Anticonvulsants/chemical synthesis , Anticonvulsants/chemistry , Dose-Response Relationship, Drug , Electroshock , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Mice , Models, Molecular , Molecular Structure , Pentylenetetrazole , Pyrimidines/chemical synthesis , Pyrimidines/chemistry , Structure-Activity Relationship , Thiones/chemical synthesis , Thiones/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...