Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Eur J Med Chem ; 188: 111920, 2020 Feb 15.
Article in English | MEDLINE | ID: mdl-31901745

ABSTRACT

γ-Aminobutyric acid (GABA) uptake transporters are membrane transport proteins that are involved in the pathophysiology of a number of neurological disorders. Some types of chronic pain appear to result from the dysfunction of the GABAergic system. The deficiency of mouse GAT1 transporter (mGAT1) abolishes the nociceptive response, which means that mGAT1 inhibition is an appropriate medical approach to achieve analgesia. The mGAT4 transporter is the second most abundant GAT subtype in the brain; however, its physiological role has not yet been fully understood in the central nervous system. In this study, we examined whether the combination of mGAT1 and mGAT3/mGAT4 inhibition in a single molecule might lead to potentially synergistic effects improving analgesic activity to relieve neuropathic pain. To study this hypothesis, new GABA uptake inhibitors were designed, synthesized, and evaluated in terms of their activity and subtype selectivity for mGAT1-4. Among new functionalized amino acid derivatives of serine and GABA analogs, compounds with preferential mGAT3/4 inhibitory activity were discovered. Two selected hits (19b and 31c) were subjected to in vivo tests. We found a statistically significant antiallodynic activity in the von Frey test in diabetic and oxaliplatin-induced neuropathic pain model. The novel compounds (4-hydroxybutanoic, 4-hydroxypentanoic, and 4-aminobutanoic acid derivatives and serine analogs) provide new insights into the structure-activity relationship of mGAT3/mGAT4 inhibitors and indicate a new direction in the search for potential treatment of neuropathic pain of various origin.


Subject(s)
Analgesics/therapeutic use , GABA Plasma Membrane Transport Proteins/metabolism , GABA Uptake Inhibitors/therapeutic use , Hyperalgesia/drug therapy , Neuralgia/drug therapy , Pain Threshold/drug effects , Analgesics/chemical synthesis , Analgesics/metabolism , Animals , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/complications , GABA Plasma Membrane Transport Proteins/chemistry , GABA Uptake Inhibitors/chemical synthesis , GABA Uptake Inhibitors/metabolism , Hyperalgesia/chemically induced , Hyperalgesia/etiology , Male , Mice , Molecular Docking Simulation , Molecular Structure , Neuralgia/chemically induced , Neuralgia/etiology , Oxaliplatin , Protein Binding , Streptozocin , Structure-Activity Relationship
2.
ChemMedChem ; 14(5): 583-593, 2019 03 05.
Article in English | MEDLINE | ID: mdl-30663849

ABSTRACT

A screening of compound libraries based on nipecotic acid derivatives with lipophilic residues attached to the scarcely explored 5-position of the core structure was used for the search of new inhibitors of the γ-aminobutyric acid (GABA) transporter 1 (mGAT1). The generated compound libraries, which were based on hydrazone chemistry commonly used in dynamic combinatorial chemistry but rendered pseudostatic, were screened for their binding affinities toward mGAT1 by means of MS Binding Assays. With nipecotic acid derived hydrazone rac-16 h [rac-(3R,5S)-{5-[(E)-2-{[5-(2-phenylethynyl)thiophen-2-yl]methylidene}hydrazin-1-yl]piperidine-3-carboxylic acid}-sodium chloride (1/2)], one hit was found and evaluated displaying sub-micromolar potency (pKi =6.62±0.04) and a noncompetitive interaction mode at mGAT1. By bearing a 5-(2-phenylethynyl)thiophen-2-yl residue attached to the 5-position of nipecotic acid via a three-atom spacer, compound rac-16 h contains a structural moiety so far unprecedented for these kinds of bioactive molecules, and complements novel 5-substituted nipecotic acid derived ligands of mGAT1 revealed in a recently published screening campaign. This new class of ligands, with an inhibition mode distinct from that of benchmark mGAT1 inhibitors, could serve as research tools for investigations of mGAT1-mediated GABA transport.


Subject(s)
GABA Plasma Membrane Transport Proteins/metabolism , GABA Uptake Inhibitors/chemistry , Hydrazones/chemistry , Nipecotic Acids/chemistry , Small Molecule Libraries/chemistry , Spectrometry, Mass, Electrospray Ionization/methods , Binding, Competitive , Chromatography, High Pressure Liquid , Drug Evaluation, Preclinical/methods , GABA Uptake Inhibitors/metabolism , HEK293 Cells , Humans , Hydrazones/metabolism , Ligands , Molecular Structure , Small Molecule Libraries/metabolism , Structure-Activity Relationship
3.
Bioorg Med Chem ; 26(12): 3668-3687, 2018 07 23.
Article in English | MEDLINE | ID: mdl-29886082

ABSTRACT

In this study, we present the synthesis and structure-activity relationships (SAR) of novel N-substituted nipecotic acid derivatives closely related to (S)-SNAP-5114 (2) in the pursuit of finding new and potent mGAT4 selective inhibitors. By the use of iminium ion chemistry, a series of new N-substituted nipecotic acid derivatives containing a variety of heterocycles, and an alkyne spacer were synthesized. Biological evaluation of the prepared compounds showed, how the inhibitory potency and subtype selectivity for the murine GABA transporters (mGATs) were influenced by the performed modifications.


Subject(s)
Alkynes/chemistry , GABA Plasma Membrane Transport Proteins/chemistry , GABA Plasma Membrane Transport Proteins/metabolism , GABA Uptake Inhibitors/chemical synthesis , Nipecotic Acids/chemistry , Animals , GABA Uptake Inhibitors/metabolism , HEK293 Cells , Humans , Mice , Nipecotic Acids/metabolism , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/metabolism , Structure-Activity Relationship , gamma-Aminobutyric Acid/metabolism
4.
ACS Chem Neurosci ; 9(11): 2767-2773, 2018 11 21.
Article in English | MEDLINE | ID: mdl-29763549

ABSTRACT

In vivo positron emission tomography (PET) imaging of the γ-aminobutyric acid (GABA) receptor complex has been accomplished using radiolabeled benzodiazepine derivatives, but development of specific presynaptic radioligands targeting the neuronal membrane GABA transporter type 1 (GAT-1) has been less successful. The availability of new structure-activity studies of GAT-1 inhibitors and the introduction of a GAT-1 inhibitor (tiagabine, Gabatril) into clinical use prompted us to reinvestigate the syntheses of PET ligands for this transporter. Initial synthesis and rodent PET studies of N-[11C]methylnipecotic acid confirmed the low brain uptake of that small and polar molecule. The common design approach to improve blood-brain barrier permeability of GAT-1 inhibitors is the attachment of a large lipophilic substituent. We selected an unsymmetrical bis-aromatic residue attached to the ring nitrogen by a vinyl ether spacer from a series recently reported by Wanner and coworkers. Nucleophilic aromatic substitution of an aryl chloride precursor with [18F]fluoride was used to prepare the desired candidate radiotracer ( R, E/ Z)-1-(2-((4-fluoro-2-(4-[18F]fluorobenzoyl)styryl)oxy)ethyl)piperidine-3-carboxylic acid (( R, E/ Z)-[18F]10). PET studies in rats showed no brain uptake, which was not altered by pretreatment of animals with the P-glycoprotein inhibitor cyclosporine A, indicating efflux by Pgp was not responsible. Subsequent PET imaging studies of ( R, E/ Z)-[18F]10 in rhesus monkey brain showed very low brain uptake. Finally, to test if the free carboxylic acid group was the likely cause of poor brain uptake, PET studies were done using the ethyl ester derivative of ( R, E/ Z)-[18F]10. Rapid and significant monkey brain uptake of the ester was observed, followed by a slow washout over 90 min. The blood-brain barrier permeability of the ester supports a hypothesis that the free acid function limits brain uptake of nipecotic acid-based GAT-1 radioligands, and future radiotracer efforts should investigate the use of carboxylic acid bioisosteres.


Subject(s)
Blood-Brain Barrier/metabolism , Brain/metabolism , GABA Plasma Membrane Transport Proteins/metabolism , GABA Uptake Inhibitors/metabolism , Piperidines/metabolism , Radiopharmaceuticals/metabolism , ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , Animals , Brain/diagnostic imaging , Cyclosporine/pharmacology , Enzyme Inhibitors/pharmacology , Esters/metabolism , Fluorine Radioisotopes , Macaca mulatta , Permeability , Piperidines/chemical synthesis , Positron-Emission Tomography , Radiopharmaceuticals/chemical synthesis , Rats , Stereoisomerism , Tiagabine/metabolism
5.
Neurochem Res ; 43(2): 306-315, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29127598

ABSTRACT

Inhibitory signaling in the ventral tegmental area (VTA) is involved in the mechanism of action for many drugs of abuse. Although drugs of abuse have been shown to alter extracellular γ-aminobutyric acid (GABA) concentration in the VTA, knowledge on how uptake mechanisms are regulated in vivo is limited. Quantitative (no-net-flux) microdialysis is commonly used to examine the extracellular concentration and clearance of monoamine neurotransmitters, however it is unclear whether this method is sensitive to changes in clearance for amino acid neurotransmitters such as GABA. The purpose of this study was to determine whether changes in GABA uptake are reflected by in vivo extraction fraction within the VTA. Using quantitative (no-net-flux) microdialysis adapted for transient conditions, we examined the effects of local perfusion with the GABA uptake inhibitor, nipecotic acid, in the VTA of Long Evans rats. Basal extracellular GABA concentration and in vivo extraction fraction were 44.4 ± 1.9 nM (x-intercepts from 4 baseline regressions using a total of 24 rats) and 0.19 ± 0.01 (slopes from 4 baseline regressions using a total of 24 rats), respectively. Nipecotic acid (50 µM) significantly increased extracellular GABA concentration to 170 ± 4 nM and reduced in vivo extraction fraction to 0.112 ± 0.003. Extraction fraction returned to baseline following removal of nipecotic acid from the perfusate. Conventional microdialysis substantially underestimated the increase of extracellular GABA concentration due to nipecotic acid perfusion compared with that obtained from the quantitative analysis. Together, these results show that inhibiting GABA uptake mechanisms within the VTA alters in vivo extraction fraction measured using microdialysis and that in vivo extraction fraction may be an indirect measure of GABA clearance.


Subject(s)
Microdialysis , Neurons/metabolism , Ventral Tegmental Area/metabolism , gamma-Aminobutyric Acid/metabolism , Amino Acids/metabolism , Animals , Dopamine/metabolism , Extracellular Space/metabolism , GABA Uptake Inhibitors/metabolism , Microdialysis/methods , Nipecotic Acids/metabolism , Nucleus Accumbens/drug effects , Rats, Long-Evans
6.
ChemMedChem ; 12(5): 362-371, 2017 03 07.
Article in English | MEDLINE | ID: mdl-28125164

ABSTRACT

A new scaffold of highly potent and mGAT1-selective inhibitors has been developed. Compounds in this class are characterized by an alkyne-type spacer connecting nipecotic acid with an aromatic moiety. Preliminary evaluations made it apparent that a nipecotic acid derivative with an N-butynyl linker and a terminal 2-biphenyl residue exhibiting a binding affinity (pKi ) of 7.61±0.03 to mGAT1 and uptake inhibition (pIC50 ) of 7.00±0.06 selective for mGAT1 could serve as a hit compound. Docking calculations for compounds based on this structure in an hGAT1 homology modeling study indicated binding affinities similar to or even higher than that of the well-known mGAT1 inhibitor tiagabine. Synthesis of the designed compounds was readily carried out by two consecutive cross-coupling reactions, giving flexible access to variously substituted biphenyl subunits. With an appropriate substitution pattern of the biphenyl moiety, the binding affinity of enantiopure (R)-nipecotic acid derivatives to mGAT1 increased to pKi =8.33±0.01, and the uptake inhibitory potency up to pIC50 =7.72±0.02.


Subject(s)
GABA Plasma Membrane Transport Proteins/chemistry , GABA Uptake Inhibitors/chemistry , Nipecotic Acids/chemistry , Binding Sites , GABA Plasma Membrane Transport Proteins/metabolism , GABA Uptake Inhibitors/chemical synthesis , GABA Uptake Inhibitors/metabolism , HEK293 Cells , Humans , Inhibitory Concentration 50 , Molecular Docking Simulation , Nipecotic Acids/chemical synthesis , Nipecotic Acids/metabolism , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/metabolism , Protein Structure, Tertiary , Structure-Activity Relationship
7.
ChemMedChem ; 10(9): 1498-510, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26220444

ABSTRACT

γ-Aminobutyric acid (GABA) transporters (GATs) are promising drug targets for various diseases associated with imbalances in GABAergic neurotransmission. For the development of new drugs or pharmacological tools addressing GATs, screening techniques to identify new inhibitors and to characterize their potency at each GAT subtype are indispensable. By now, the technique by far dominating is based on radiolabeled GABA. We recently described "MS Transport Assays" for hGAT-1 by employing ((2) H6 )GABA as the substrate. In the present study, we applied this approach to all four human GAT subtypes and determined the KM values for GAT-mediated transport of ((2) H6 )GABA at each subtype. Furthermore, a comprehensive set of GAT inhibitors reflecting the whole range of potency and subtype selectivity known so far was evaluated for their potency. The comparison of pIC50 values obtained in conventional [(3) H]GABA uptake assays with those obtained in MS Transport Assays indicated the reliability of the latter. The MS Transport Assays enable a throughput similar to that of conventional radiometric transport assays performed in a 96-well format but avoid the use of radiolabeled substrates.


Subject(s)
GABA Plasma Membrane Transport Proteins/metabolism , GABA Uptake Inhibitors/pharmacology , High-Throughput Screening Assays/methods , Animals , Binding, Competitive , COS Cells , Carrier Proteins/genetics , Carrier Proteins/metabolism , Chlorocebus aethiops , Drug Evaluation, Preclinical/methods , GABA Plasma Membrane Transport Proteins/analysis , GABA Plasma Membrane Transport Proteins/genetics , GABA Uptake Inhibitors/chemistry , GABA Uptake Inhibitors/metabolism , Humans , Molecular Targeted Therapy/methods , Tandem Mass Spectrometry/methods , Workflow , gamma-Aminobutyric Acid/metabolism , gamma-Aminobutyric Acid/pharmacokinetics
8.
Epilepsia ; 56(7): 1141-51, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26040777

ABSTRACT

OBJECTIVE: Glutamate and γ-aminobutyric acid (GABA) are the key neurotransmitter systems in the cortical-thalamocortical network, involved in normal and pathologic oscillations such as spike-wave discharges (SWDs), which characterize different forms of absence epilepsy. Metabotropic glutamate (mGlu) and GABA receptors are widely expressed within this network. Herein, we examined the effects of two selective positive allosteric modulators (PAMs) of mGlu1 and mGlu5 receptors, the GABA reuptake inhibitor, tiagabine, and their interaction in the somatosensory cortex and thalamus on SWDs in WAG/Rij rats. METHODS: Male WAG/Rij rats were equipped with bilateral cannulas in the somatosensory cortex (S1po) or the ventrobasal (VB) thalamic nuclei, and with cortical electroencephalography (EEG) electrodes. Rats received a single dose of the mGlu1 receptor PAM, RO0711401, or the mGlu5 receptor PAM, VU0360172, various doses of tiagabine, or VU0360172 combined with tiagabine. RESULTS: Both PAMs suppressed SWDs regardless of the site of injection. Tiagabine enhanced SWDs when injected into the thalamus, but, unexpectedly, suppressed SWDs in a dose-dependent manner when injected into the cortex. Intracortical co-injection of VU0360172 and tiagabine produced slightly larger effects as compared to either VU0360172 or tiagabine alone. Intrathalamic co-injections of VU0360172 and subthreshold doses of tiagabine caused an antiabsence effect similar to that exhibited by VU0360172 alone in the first 10 min. At 30 min, however, the antiabsence effect of VU0360172 was prevented by subthreshold doses of tiagabine, and the combination produced a paradoxical proabsence effect at 40 and 50 min. SIGNIFICANCE: These data (1) show that mGlu1 and mGlu5 receptor PAMs reduce absence seizures acting at both thalamic and cortical levels; (2) demonstrate for the first time that tiagabine, despite its established absence-enhancing effect, reduces SWDs when injected into the somatosensory cortex; and (3) indicate that the efficacy of VU0360172 in the thalamus may be critically affected by the availability of (extra)synaptic GABA.


Subject(s)
Epilepsy, Absence/metabolism , GABA Uptake Inhibitors/metabolism , Receptor, Metabotropic Glutamate 5/metabolism , Receptors, Metabotropic Glutamate/metabolism , Somatosensory Cortex/metabolism , Thalamus/metabolism , Animals , Anticonvulsants/administration & dosage , Anticonvulsants/metabolism , Epilepsy, Absence/prevention & control , GABA Uptake Inhibitors/administration & dosage , Infusions, Intraventricular , Male , Rats , Rats, Transgenic , Receptor, Metabotropic Glutamate 5/agonists , Receptors, Metabotropic Glutamate/agonists , Somatosensory Cortex/drug effects , Thalamus/drug effects
9.
ChemMedChem ; 10(2): 396-410, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25369775

ABSTRACT

The objective of the present study was to transfer the concept of library screening by MS binding assays, so far applied to pseudostatic hydrazine libraries, to static oxime libraries to screen for new potent inhibitors of mGAT1, the most abundant GABA transporter in the central nervous system that represents a validated drug target for the treatment of epilepsy. Library generation was performed by reaction of guvacine derivatives possessing a hydroxylamine functionality with various sets of four aldehydes. After dilution, the libraries were screened by competitive MS binding assays. Deconvolution experiments allowed hits in the most active libraries to be identified, and they were resynthesized for biological evaluation. That way a series of compounds was identified that displayed binding affinities ≥8.00 (pKi ) at mGAT1, one of which was found to be the most potent mGAT1 inhibitor known to date in a functional GABA uptake assay with a pIC50 value of 8.27 ± 0.03.


Subject(s)
GABA Plasma Membrane Transport Proteins/metabolism , Oximes/chemistry , Combinatorial Chemistry Techniques , GABA Plasma Membrane Transport Proteins/chemistry , GABA Plasma Membrane Transport Proteins/genetics , GABA Uptake Inhibitors/chemical synthesis , GABA Uptake Inhibitors/chemistry , GABA Uptake Inhibitors/metabolism , HEK293 Cells , Humans , Kinetics , Oximes/chemical synthesis , Oximes/metabolism , Protein Binding , Structure-Activity Relationship , gamma-Aminobutyric Acid/chemistry , gamma-Aminobutyric Acid/metabolism
10.
J Pharm Pharmacol ; 65(4): 582-90, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23488788

ABSTRACT

OBJECTIVES: The pyridine alkaloid arecaidine is an ingredient of areca nut preparations. It is responsible for many physiological effects observed during areca nut chewing. However, the mechanism underlying its oral bioavailability has not yet been studied. We investigated whether the H⁺-coupled amino acid transporter 1 (PAT1, SLC36A1), which is expressed in the intestinal epithelium, accepts arecaidine, arecoline, isoguvacine and other derivatives as substrates. METHODS: Inhibition of L-[³H]proline uptake by arecaidine and derivatives was determined in Caco-2 cells expressing hPAT1 constitutively and in HeLa cells transiently transfected with hPAT1-cDNA. Transmembrane transport of arecaidine and derivatives was measured electrophysiologically in Xenopus laevis oocytes. KEY FINDINGS: Arecaidine, guvacine and isoguvacine but not arecoline strongly inhibited the uptake of L-[³H]proline into Caco-2 cells. Kinetic analyses revealed the competitive manner of L-proline uptake inhibition by arecaidine. In HeLa cells transfected with hPAT1-cDNA an affinity constant of 3.8 mm was obtained for arecaidine. Electrophysiological measurements at hPAT1-expressing X. laevis oocytes demonstrated that arecaidine, guvacine and isoguvacine are transported by hPAT1 in an electrogenic manner. CONCLUSION: We conclude that hPAT1 transports arecaidine, guvacine and isoguvacine across the apical membrane of enterocytes and that hPAT1 might be responsible for the intestinal absorption of these drug candidates.


Subject(s)
Amino Acid Transport Systems/metabolism , Areca/chemistry , Arecoline/analogs & derivatives , Enterocytes/metabolism , GABA Uptake Inhibitors/metabolism , Nuts/chemistry , Symporters/metabolism , Amino Acid Transport Systems/genetics , Animals , Arecoline/metabolism , Arecoline/pharmacology , Binding, Competitive , Biological Transport/drug effects , Caco-2 Cells , Drugs, Investigational/metabolism , Drugs, Investigational/pharmacology , Enterocytes/drug effects , Female , GABA Agonists/metabolism , GABA Agonists/pharmacology , GABA Uptake Inhibitors/pharmacology , HeLa Cells , Humans , Intestinal Absorption/drug effects , Isonicotinic Acids/metabolism , Isonicotinic Acids/pharmacology , Kinetics , Nicotinic Acids/metabolism , Nicotinic Acids/pharmacology , Oocytes/metabolism , Recombinant Proteins/metabolism , Symporters/genetics , Xenopus laevis
11.
Bioorg Med Chem ; 21(2): 470-84, 2013 Jan 15.
Article in English | MEDLINE | ID: mdl-23245753

ABSTRACT

A series of enantiomerically pure 4-hydroxy-4-(4-methoxyphenyl)-substituted proline and pyrrolidin-2-ylacetic acid derivatives have been synthesized starting from the respective N-protected 4-hydroxy derivatives via oxidation to the corresponding 4-oxo compounds, subsequent addition of organometallic reagents, final hydrolysis and deprotection. The major diastereoisomers obtained by the addition of the Grignard reagents were found to have opposite stereoconfigurations depending on whether cerium trichloride was present or absent as an additive. The final compounds were evaluated for their capability to inhibit the GABA transport proteins GAT1 and GAT3. 4-Hydroxyproline derivatives substituted with a tris(4-methoxyphenyl)methyloxyethyl residue at the nitrogen and a 4-methoxyphenyl group in 4-position showed, with the exception of the (2R,4R)-diastereomer, an improved inhibition at GAT3 compared to the derivatives missing the 4-methoxyphenyl group in 4-position. This may imply that an appropriate lipophilic group at the C-4 position of the proline moiety is beneficial for potent inhibition at GAT3.


Subject(s)
GABA Plasma Membrane Transport Proteins/metabolism , GABA Uptake Inhibitors/chemical synthesis , Proline/analogs & derivatives , Pyrrolidines/chemistry , Drug Evaluation, Preclinical , GABA Plasma Membrane Transport Proteins/chemistry , GABA Uptake Inhibitors/chemistry , GABA Uptake Inhibitors/metabolism , Proline/chemical synthesis , Protein Binding , Stereoisomerism , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...