Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Brain Commun ; 6(2): fcae110, 2024.
Article in English | MEDLINE | ID: mdl-38650830

ABSTRACT

We have previously characterized the molecular mechanisms for variants in γ-aminobutyric acid transporter 1-encoding solute carrier family 6-member 1 (SLC6A1) in vitro and concluded that a partial or complete loss of γ-aminobutyric acid uptake due to impaired protein trafficking is the primary aetiology. Impairment of γ-aminobutyric acid transporter 1 function could cause compensatory changes in the expression of γ-aminobutyric acid receptors, which, in turn, modify disease pathophysiology and phenotype. Here we used different approaches including radioactive 3H γ-aminobutyric acid uptake in cells and synaptosomes, immunohistochemistry and confocal microscopy as well as brain slice surface protein biotinylation to characterize Slc6a1+/A288V and Slc6a1+/S295L mice, representative of a partial or a complete loss of function of SLC6A1 mutations, respectively. We employed the γ-aminobutyric acid transporter 1-specific inhibitor [3H]tiagabine binding and GABAA receptor subunit-specific radioligand binding to profile the γ-aminobutyric acid transporter 1 and GABAA receptor expression in major brain regions such as cortex, cerebellum, hippocampus and thalamus. We also determined the total and surface expression of γ-aminobutyric acid transporter 1, γ-aminobutyric acid transporter 3 and expression of GABAA receptor in the major brain regions in the knockin mice. We found that γ-aminobutyric acid transporter 1 protein was markedly reduced in cortex, hippocampus, thalamus and cerebellum in both mutant mouse lines. Consistent with the findings of reduced γ-aminobutyric acid uptake for both γ-aminobutyric acid transporter 1(A288V) and γ-aminobutyric acid transporter 1(S295L), both the total and the γ-aminobutyric acid transporter 1-mediated 3H γ-aminobutyric acid reuptake was reduced. We found that γ-aminobutyric acid transporter 3 is only abundantly expressed in the thalamus and there was no compensatory increase of γ-aminobutyric acid transporter 3 in either of the mutant mouse lines. γ-Aminobutyric acid transporter 1 was reduced in both somatic regions and nonsomatic regions in both mouse models, in which a ring-like structure was identified only in the Slc6a1+/A288V mouse, suggesting more γ-aminobutyric acid transporter 1 retention inside endoplasmic reticulum in the Slc6a1+/A288V mouse. The [3H]tiagabine binding was similar in both mouse models despite the difference in γ-aminobutyric acid uptake function and γ-aminobutyric acid transporter 1 protein expression for both mutations. There were no differences in GABAA receptor subtype expression, except for a small increase in the expression of α5 subunits of GABAA receptor in the hippocampus of Slc6a1S295L homozygous mice, suggesting a potential interaction between the expression of this GABAA receptor subtype and the mutant γ-aminobutyric acid transporter 1. The study provides the first comprehensive characterization of the SLC6A1 mutations in vivo in two representative mouse models. Because both γ-aminobutyric acid transporter 1 and GABAA receptors are targets for anti-seizure medications, the findings from this study can help guide tailored treatment options based on the expression and function of γ-aminobutyric acid transporter 1 and GABAA receptor in SLC6A1 mutation-mediated neurodevelopmental and epileptic encephalopathies.

2.
Nat Struct Mol Biol ; 30(12): 1936-1946, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37903907

ABSTRACT

α5 subunit-containing γ-aminobutyric acid type A (GABAA) receptors represent a promising drug target for neurological and neuropsychiatric disorders. Altered expression and function contributes to neurodevelopmental disorders such as Dup15q and Angelman syndromes, developmental epilepsy and autism. Effective drug action without side effects is dependent on both α5-subtype selectivity and the strength of the positive or negative allosteric modulation (PAM or NAM). Here we solve structures of drugs bound to the α5 subunit. These define the molecular basis of binding and α5 selectivity of the ß-carboline, methyl 6,7-dimethoxy-4-ethyl-ß-carboline-3-carboxylate (DMCM), type II benzodiazepine NAMs, and a series of isoxazole NAMs and PAMs. For the isoxazole series, each molecule appears as an 'upper' and 'lower' moiety in the pocket. Structural data and radioligand binding data reveal a positional displacement of the upper moiety containing the isoxazole between the NAMs and PAMs. Using a hybrid molecule we directly measure the functional contribution of the upper moiety to NAM versus PAM activity. Overall, these structures provide a framework by which to understand distinct modulator binding modes and their basis of α5-subtype selectivity, appreciate structure-activity relationships, and empower future structure-based drug design campaigns.


Subject(s)
Receptors, GABA-A , gamma-Aminobutyric Acid , Receptors, GABA-A/metabolism , Isoxazoles/pharmacology
3.
J Neurodev Disord ; 14(1): 10, 2022 02 05.
Article in English | MEDLINE | ID: mdl-35123401

ABSTRACT

BACKGROUND: There are currently no pharmacological therapies to address the intellectual disability associated with Down syndrome. Excitatory/inhibitory imbalance has been hypothesized to contribute to impairments in cognitive functioning in Down syndrome. Negative modulation of the GABAA-α5 receptor is proposed as a mechanism to attenuate GABAergic function and restore the excitatory/inhibitory balance. METHODS: Basmisanil, a selective GABAA-α5 negative allosteric modulator, was evaluated at 120 mg or 240 mg BID (80 or 160 mg for 12-13 years) in a 6-month, randomized, double-blind, placebo-controlled phase II trial (Clematis) for efficacy and safety in adolescents and young adults with Down syndrome. The primary endpoint was based on a composite analysis of working memory (Repeatable Battery for the Assessment of Neuropsychological Scale [RBANS]) and independent functioning and adaptive behavior (Vineland Adaptive Behavior Scales [VABS-II] or the Clinical Global Impression-Improvement [CGI-I]). Secondary measures included the Behavior Rating Inventory of Executive Functioning-Preschool (BRIEF-P), Clinical Evaluation of Language Fundamentals (CELF-4), and Pediatric Quality of Life Inventory (Peds-QL). EEG was conducted for safety monitoring and quantitatively analyzed in adolescents. RESULTS: Basmisanil was safe and well-tolerated; the frequency and nature of adverse events were similar in basmisanil and placebo arms. EEG revealed treatment-related changes in spectral power (increase in low ~ 4-Hz and decrease in high ~ 20-Hz frequencies) providing evidence of functional target engagement. All treatment arms had a similar proportion of participants showing above-threshold improvement on the primary composite endpoint, evaluating concomitant responses in cognition and independent functioning (29% in placebo, 20% in low dose, and 25% in high dose). Further analysis of the individual measures contributing to the primary endpoint revealed no difference between placebo and basmisanil-treated groups in either adolescents or adults. There were also no differences across the secondary endpoints assessing changes in executive function, language, or quality of life. CONCLUSIONS: Basmisanil did not meet the primary efficacy objective of concomitant improvement on cognition and adaptive functioning after 6 months of treatment, despite evidence for target engagement. This study provides key learnings for future clinical trials in Down syndrome. TRIAL REGISTRATION: The study was registered on December 31, 2013, at clinicaltrials.gov as NCT02024789.


Subject(s)
Down Syndrome , Intellectual Disability , Adolescent , Child , Child, Preschool , Down Syndrome/complications , Down Syndrome/drug therapy , Humans , Intellectual Disability/complications , Intellectual Disability/drug therapy , Morpholines , Oxazoles , Pyridines , Quality of Life , Treatment Outcome , Young Adult , gamma-Aminobutyric Acid/therapeutic use
4.
Cell Rep ; 37(1): 109768, 2021 10 05.
Article in English | MEDLINE | ID: mdl-34610304

ABSTRACT

GABA can depolarize immature neurons close to the action potential (AP) threshold in development and adult neurogenesis. Nevertheless, GABAergic synapses effectively inhibit AP firing in newborn granule cells of the adult hippocampus as early as two weeks post-mitosis. The underlying mechanisms are largely unclear. Here, we analyze GABAergic inputs in newborn hippocampal granule cells mediated by soma-targeting parvalbumin and dendrite-targeting somatostatin interneurons. Surprisingly, both interneuron subtypes activate α5-subunit-containing GABAA receptors (α5-GABAARs) in young neurons, showing a nonlinear voltage dependence with increasing conductance around the AP threshold. By contrast, in mature cells, parvalbumin interneurons mediate linear GABAergic synaptic currents lacking α5-subunits, while somatostatin interneurons continue to target nonlinear α5-GABAARs. Computational modeling shows that the voltage-dependent amplification of α5-GABAAR opening in young neurons is crucial for inhibition of AP firing to generate balanced and sparse firing activity, even with depolarized GABA reversal potential.


Subject(s)
Action Potentials , Hippocampus/metabolism , Receptors, GABA-A/metabolism , Action Potentials/drug effects , Animals , Electric Stimulation , Female , Hippocampus/cytology , Interneurons/metabolism , Male , Membrane Potentials , Mice , Mice, Transgenic , Optogenetics/methods , Parvalbumins/genetics , Parvalbumins/metabolism , Protein Subunits/genetics , Protein Subunits/metabolism , Receptors, GABA-A/genetics , Receptors, N-Methyl-D-Aspartate/metabolism , Somatostatin/genetics , Somatostatin/metabolism , Synapses/metabolism , gamma-Aminobutyric Acid/metabolism , gamma-Aminobutyric Acid/pharmacology
5.
Sci Rep ; 11(1): 7700, 2021 04 08.
Article in English | MEDLINE | ID: mdl-33833333

ABSTRACT

GABAA-α5 subunit-containing receptors have been shown to play a key modulatory role in cognition and represent a promising drug target for cognitive dysfunction, as well as other disorders. Here we report on the preclinical and early clinical profile of a novel GABAA-α5 selective negative allosteric modulator (NAM), basmisanil, which progressed into Phase II trials for intellectual disability in Down syndrome and cognitive impairment associated with schizophrenia. Preclinical pharmacology studies showed that basmisanil is the most selective GABAA-α5 receptor NAM described so far. Basmisanil bound to recombinant human GABAA-α5 receptors with 5 nM affinity and more than 90-fold selectivity versus α1, α2, and α3 subunit-containing receptors. Moreover, basmisanil inhibited GABA-induced currents at GABAA-α5 yet had little or no effect at the other receptor subtypes. An in vivo occupancy study in rats showed dose-dependent target engagement and was utilized to establish the plasma exposure to receptor occupancy relationship. At estimated receptor occupancies between 30 and 65% basmisanil attenuated diazepam-induced spatial learning impairment in rats (Morris water maze), improved executive function in non-human primates (object retrieval), without showing anxiogenic or proconvulsant effects in rats. During the Phase I open-label studies, basmisanil showed good safety and tolerability in healthy volunteers at maximum GABAA-α5 receptor occupancy as confirmed by PET analysis with the tracer [11C]-Ro 15-4513. An exploratory EEG study provided evidence for functional activity of basmisanil in human brain. Therefore, these preclinical and early clinical studies show that basmisanil has an ideal profile to investigate potential clinical benefits of GABAA-α5 receptor negative modulation.


Subject(s)
GABA-A Receptor Agonists/pharmacology , Receptors, GABA-A/drug effects , Allosteric Regulation , Animals , Brain/diagnostic imaging , Brain/drug effects , Brain/metabolism , Female , HEK293 Cells , Healthy Volunteers , Humans , Learning/drug effects , Macaca fascicularis , Positron-Emission Tomography , Radioligand Assay , Rats , Rats, Sprague-Dawley , Receptors, GABA-A/chemistry , Receptors, GABA-A/metabolism , Xenopus laevis
6.
J Neurosci ; 39(26): 5210-5221, 2019 06 26.
Article in English | MEDLINE | ID: mdl-31000585

ABSTRACT

Down syndrome (DS) or Trisomy 21 is a developmental disorder leading to cognitive deficits, including disruption of hippocampus-dependent learning and memory. Enhanced inhibition has been suggested to underlie these deficits in DS based on studies using the Ts65Dn mouse model. Here we show that, in this mouse model, GABAergic synaptic inhibition onto dendrites of hippocampal pyramidal cells is increased. By contrast, somatic inhibition was not altered. In addition, synaptic NMDAR currents were reduced. Furthermore, dendritic inhibition was mediated via nonlinear α5-subunit containing GABAARs that closely matched the kinetics and voltage dependence of NMDARs. Thus, enhanced dendritic inhibition and reduced NMDA currents strongly decreased burst-induced NMDAR-mediated depolarization and impaired LTP induction. Finally, selective reduction of α5-GABAAR-mediated inhibition rescued both burst-induced synaptic NMDAR activation and synaptic plasticity. These results demonstrate that reduced synaptic NMDAR activation and synaptic plasticity in the Ts65Dn mouse model of DS can be corrected by specifically targeting nonlinear dendritic inhibition.SIGNIFICANCE STATEMENT Mild to moderate intellectual disability is a prominent feature of Down syndrome. Previous studies in mouse models suggest that increased synaptic inhibition is a main factor for decreased synaptic plasticity, the cellular phenomenon underlying memory. The present study shows that increased inhibition specifically onto dendrites together with reduced NMDAR content in excitatory synapses may be the cause. Reducing a slow nonlinear component that is specific to dendritic inhibitory inputs and mediated by α5 subunit-containing GABAA receptors rescues both NMDAR activation and synaptic plasticity.


Subject(s)
Dendrites/physiology , Down Syndrome/physiopathology , Long-Term Potentiation/physiology , Neural Inhibition/physiology , Pyramidal Cells/physiology , Receptors, N-Methyl-D-Aspartate/metabolism , Animals , Disease Models, Animal , Down Syndrome/metabolism , Male , Membrane Potentials/physiology , Mice , Synapses/physiology , Synaptic Transmission/physiology
7.
Biol Psychiatry ; 85(9): 752-759, 2019 05 01.
Article in English | MEDLINE | ID: mdl-30826071

ABSTRACT

BACKGROUND: Angelman syndrome (AS) is a severe neurodevelopmental disorder caused by either disruptions of the gene UBE3A or deletion of chromosome 15 at 15q11-q13, which encompasses UBE3A and several other genes, including GABRB3, GABRA5, GABRG3, encoding gamma-aminobutyric acid type A receptor subunits (ß3, α5, γ3). Individuals with deletions are generally more impaired than those with other genotypes, but the underlying pathophysiology remains largely unknown. Here, we used electroencephalography (EEG) to test the hypothesis that genes other than UBE3A located on 15q11-q13 cause differences in pathophysiology between AS genotypes. METHODS: We compared spectral power of clinical EEG recordings from children (1-18 years of age) with a deletion genotype (n = 37) or a nondeletion genotype (n = 21) and typically developing children without Angelman syndrome (n = 48). RESULTS: We found elevated theta power (peak frequency: 5.3 Hz) and diminished beta power (peak frequency: 23 Hz) in the deletion genotype compared with the nondeletion genotype as well as excess broadband EEG power (1-32 Hz) peaking in the delta frequency range (peak frequency: 2.8 Hz), shared by both genotypes but stronger for the deletion genotype at younger ages. CONCLUSIONS: Our results provide strong evidence for the contribution of non-UBE3A neuronal pathophysiology in deletion AS and suggest that hemizygosity of the GABRB3-GABRA5-GABRG3 gene cluster causes abnormal theta and beta EEG oscillations that may underlie the more severe clinical phenotype. Our work improves the understanding of AS pathophysiology and has direct implications for the development of AS treatments and biomarkers.


Subject(s)
Angelman Syndrome/genetics , Angelman Syndrome/physiopathology , Brain Waves , Cerebral Cortex/physiopathology , Adolescent , Beta Rhythm , Child , Child, Preschool , Delta Rhythm , Electroencephalography , Genotype , Humans , Infant , Phenotype , Theta Rhythm
8.
J Vis Exp ; (138)2018 08 16.
Article in English | MEDLINE | ID: mdl-30175997

ABSTRACT

This manuscript presents a step-by-step protocol for screening compounds at gamma-aminobutyric acid type A (GABAA) receptors and its use towards the identification of novel molecules active in preclinical assays from an in vitro recombinant receptor to their pharmacological effects at native receptors in rodent brain slices. For compounds binding at the benzodiazepine site of the receptor, the first step is to set up a primary screen that consists of developing radioligand binding assays on cell membranes expressing the major GABAA subtypes. Then, taking advantage of the heterologous expression of rodent and human GABAA receptors in Xenopus oocytes or HEK 293 cells, it is possible to explore, in electrophysiological assays, the physiological properties of the different receptor subtypes and the pharmacological properties of the identified compounds. The Xenopus oocyte system will be presented here, starting with the isolation of the oocytes and their microinjection with different mRNAs, up to the pharmacological characterization using two-electrode voltage clamps. Finally, recordings conducted in rodent brain slices will be described that are used as a secondary physiological test to assess the activity of molecules at their native receptors in a well-defined neuronal circuit. Extracellular recordings using population responses of multiple neurons are demonstrated together with the drug application.


Subject(s)
Drug Discovery/methods , Receptors, GABA/metabolism , Synaptic Transmission/genetics , Animals , HEK293 Cells , Humans , Neurons/metabolism
9.
Nat Commun ; 9(1): 3576, 2018 09 03.
Article in English | MEDLINE | ID: mdl-30177704

ABSTRACT

Dendrite-targeting GABAergic interneurons powerfully control postsynaptic integration, synaptic plasticity, and learning. However, the mechanisms underlying the efficient GABAergic control of dendritic electrogenesis are not well understood. Using subtype-selective blockers for GABAA receptors, we show that dendrite-targeting somatostatin interneurons and NO-synthase-positive neurogliaform cells preferentially activate α5-subunit- containing GABAA receptors (α5-GABAARs), generating slow inhibitory postsynaptic currents (IPSCs) in hippocampal CA1 pyramidal cells. By contrast, only negligible contribution of these receptors could be found in perisomatic IPSCs, generated by fast-spiking parvalbumin interneurons. Remarkably, α5-GABAAR-mediated IPSCs were strongly outward-rectifying generating 4-fold larger conductances above -50 mV than at rest. Experiments and modeling show that synaptic activation of these receptors can very effectively control voltage-dependent NMDA-receptor activation as well as Schaffer-collateral evoked burst firing in pyramidal cells. Taken together, nonlinear-rectifying α5-GABAARs with slow kinetics match functional NMDA-receptor properties and thereby mediate powerful control of dendritic postsynaptic integration and action potential firing by dendrite-targeting interneurons.


Subject(s)
Dendrites/metabolism , Inhibitory Postsynaptic Potentials , Interneurons/metabolism , Pyramidal Cells/metabolism , Receptors, GABA-A/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Synapses/metabolism , Action Potentials , Animals , CA1 Region, Hippocampal/cytology , Mice , Mice, Transgenic , Parvalbumins , Patch-Clamp Techniques , Synaptic Transmission
10.
Mol Neurobiol ; 55(6): 4745-4762, 2018 Jun.
Article in English | MEDLINE | ID: mdl-28717969

ABSTRACT

Trisomy 21 or Down syndrome (DS) is the most common cause of intellectual disability of a genetic origin. The Ts65Dn (TS) mouse, which is the most commonly used and best-characterized mouse model of DS, displays many of the cognitive, neuromorphological, and biochemical anomalies that are found in the human condition. One of the mechanisms that have been proposed to be responsible for the cognitive deficits in this mouse model is impaired GABA-mediated inhibition. Because of the well-known modulatory role of GABAA α5 subunit-containing receptors in cognitive processes, these receptors are considered to be potential targets for improving the intellectual disability in DS. The chronic administration of GABAA α5-negative allosteric modulators has been shown to be procognitive without anxiogenic or proconvulsant side effects. In the present study, we use a genetic approach to evaluate the contribution of GABAA α5 subunit-containing receptors to the cognitive, electrophysiological, and neuromorphological deficits in TS mice. We show that reducing the expression of GABAA α5 receptors by deleting one or two copies of the Gabra5 gene in TS mice partially ameliorated the cognitive impairments, improved long-term potentiation, enhanced neural differentiation and maturation, and normalized the density of the GABAergic synapse markers. Reducing the gene dosage of Gabra5 in TS mice did not induce motor alterations and anxiety or affect the viability of the mice. Our results provide further evidence of the role of GABAA α5 receptor-mediated inhibition in cognitive impairment in the TS mouse model of DS.


Subject(s)
Cognition , Down Syndrome/pathology , Down Syndrome/physiopathology , Electrophysiological Phenomena , Hippocampus/pathology , Hippocampus/physiopathology , Receptors, GABA-A/metabolism , Animals , Cell Count , Cell Differentiation , Cell Proliferation , Crosses, Genetic , Disease Models, Animal , Female , Gene Dosage , Male , Mice, Knockout , Neurons/pathology , Receptors, GABA-A/genetics , Time Factors
11.
Rev. Síndr. Down ; 35(135): 125-134, dic. 2017. ilus, tab
Article in Spanish | IBECS | ID: ibc-170217

ABSTRACT

Pese al creciente número de ensayos clínicos desarrollados para evaluar la cognición en el síndrome de Down, sus resultados para identificar intervenciones eficaces han sido muy limitados hasta la fecha. Las intervenciones en los modelos animales, con frecuencia muy favorables, no se han visto reflejadas en los ensayos clínicos. Esta revisión describe los resultados de los principales ensayos realizados. Ofrece consideraciones a los investigadores y describe estrategias a la industria farmacéutica para que se implique crecientemente en el descubrimiento de fármacos en el síndrome de Down


Although an increasing number of clinical trials have been developed for cognition in Down syndrome, there has been limited success to date in identifying effective interventions. This review describes the progression from pre-clinical studies with mouse models to human clinical trials research using pharmacological interventions to improve cognition and adaptive functioning in Down syndrome. We also provide considerations for investigators when conducting human clinical trials and describe strategies for the pharmaceutical industry to advance the field in drug discovery for Down syndrome


Subject(s)
Humans , Cognition Disorders/drug therapy , Adjustment Disorders/drug therapy , Down Syndrome/drug therapy , Cognition , Adaptation, Psychological , Rivastigmine/pharmacokinetics , Piracetam/pharmacokinetics , Memantine/pharmacokinetics , Drugs, Investigational
12.
Am J Med Genet A ; 173(11): 3029-3041, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28884975

ABSTRACT

Although an increasing number of clinical trials have been developed for cognition in Down syndrome, there has been limited success to date in identifying effective interventions. This review describes the progression from pre-clinical studies with mouse models to human clinical trials research using pharmacological interventions to improve cognition and adaptive functioning in Down syndrome. We also provide considerations for investigators when conducting human clinical trials and describe strategies for the pharmaceutical industry to advance the field in drug discovery for Down syndrome. Future research focusing on earlier pharmaceutical interventions, development of appropriate outcome measures, and greater collaboration between industry, academia, advocacy, and regulatory groups will be important for addressing limitations from prior studies and developing potential effective interventions for cognition in Down syndrome.


Subject(s)
Cognition/drug effects , Cognitive Dysfunction/drug therapy , Down Syndrome/drug therapy , Animals , Cognitive Dysfunction/genetics , Cognitive Dysfunction/physiopathology , Down Syndrome/genetics , Down Syndrome/physiopathology , Humans , Mice
13.
Biochem Pharmacol ; 115: 10-7, 2016 Sep 01.
Article in English | MEDLINE | ID: mdl-27002180

ABSTRACT

GABAA receptors are ligand-gated ion channels that form a fundamental component of inhibitory neurotransmission in the central and peripheral nervous systems. However, since the initial recordings of inhibitory electrical activity of neurons in response to GABA, these receptors have been found to play a more complex role and can, under some circumstances, function in an excitatory manner. This has been demonstrated via electrophysiological recordings conducted in both mature and developing neurons from different brain regions, as well as in various subcellular locations such as dendritic and axonal membranes. The balance between the inhibitory and excitatory effects mediated by GABAA receptor activation depends not only on multiple factors that govern the equilibrium of the transmembrane chloride gradient, but also on bicarbonate concentration. Moreover, electrophysiological and fluorescence measurements have revealed that a spatial distribution of the chloride gradient exists within neurons, which locally influences the effects mediated by GABAA receptor activation. In recent years, it has also become apparent that intra-neuronal chloride concentration is partially regulated by cation-chloride co-transporters (CCCs), in particular NKCC1 and KCC2. The aim of the present commentary is to discuss, in light of the latest findings, potential implications of the tight spatial and temporal regulation of chloride equilibrium in health and disease, as well as its relevance for the therapeutic effects of molecules acting at GABAA receptors.


Subject(s)
Receptors, GABA-A/metabolism , Synaptic Transmission , Animals , Humans , Ion Transport , Potassium Chloride/metabolism , Receptors, GABA-A/chemistry , Receptors, GABA-A/drug effects , Sodium Chloride Symporters/metabolism
14.
J Neurosci ; 33(9): 3953-66, 2013 Feb 27.
Article in English | MEDLINE | ID: mdl-23447605

ABSTRACT

Down syndrome (DS) is associated with neurological complications, including cognitive deficits that lead to impairment in intellectual functioning. Increased GABA-mediated inhibition has been proposed as a mechanism underlying deficient cognition in the Ts65Dn (TS) mouse model of DS. We show that chronic treatment of these mice with RO4938581 (3-bromo-10-(difluoromethyl)-9H-benzo[f]imidazo[1,5-a][1,2,4]triazolo[1,5-d][1,4]diazepine), a selective GABA(A) α5 negative allosteric modulator (NAM), rescued their deficits in spatial learning and memory, hippocampal synaptic plasticity, and adult neurogenesis. We also show that RO4938581 normalized the high density of GABAergic synapse markers in the molecular layer of the hippocampus of TS mice. In addition, RO4938581 treatment suppressed the hyperactivity observed in TS mice without inducing anxiety or altering their motor abilities. These data demonstrate that reducing GABAergic inhibition with RO4938581 can reverse functional and neuromorphological deficits of TS mice by facilitating brain plasticity and support the potential therapeutic use of selective GABA(A) α5 NAMs to treat cognitive dysfunction in DS.


Subject(s)
Down Syndrome/complications , Down Syndrome/pathology , Hippocampus/pathology , Learning Disabilities/drug therapy , Neurons/physiology , Receptors, GABA-A/metabolism , Acoustic Stimulation , Analysis of Variance , Animals , Benzodiazepines/pharmacology , Benzodiazepines/therapeutic use , Biophysics , Carrier Proteins/metabolism , Cell Count , Cell Proliferation/drug effects , Cues , Disease Models, Animal , Down Syndrome/drug therapy , Electric Stimulation , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/genetics , Exploratory Behavior/drug effects , GABA Modulators/pharmacology , GABA Modulators/therapeutic use , Glutamate Decarboxylase/metabolism , Hippocampus/drug effects , Hyperkinesis/drug therapy , Hyperkinesis/etiology , Imidazoles/pharmacology , Imidazoles/therapeutic use , Ki-67 Antigen , Learning Disabilities/etiology , Long-Term Potentiation/drug effects , Long-Term Potentiation/genetics , Male , Maze Learning/drug effects , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurogenesis/drug effects , Neurogenesis/genetics , Neurons/drug effects , Protein Binding/drug effects , Protein Binding/genetics , Psychomotor Performance/drug effects , Reaction Time/drug effects , Reflex/drug effects , Reflex/genetics , Reflex, Startle/drug effects , Rotarod Performance Test , Seizures/etiology , Sensory Gating/drug effects , Tritium/pharmacokinetics , Vesicular Inhibitory Amino Acid Transport Proteins/metabolism
15.
PLoS One ; 7(12): e51041, 2012.
Article in English | MEDLINE | ID: mdl-23236427

ABSTRACT

Chloride channels represent a group of targets for major clinical indications. However, molecular screening for chloride channel modulators has proven to be difficult and time-consuming as approaches essentially rely on the use of fluorescent dyes or invasive patch-clamp techniques which do not lend themselves to the screening of large sets of compounds. To address this problem, we have developed a non-invasive optical method, based on digital holographic microcopy (DHM), allowing monitoring of ion channel activity without using any electrode or fluorescent dye. To illustrate this approach, GABA(A) mediated chloride currents have been monitored with DHM. Practically, we show that DHM can non-invasively provide the quantitative determination of transmembrane chloride fluxes mediated by the activation of chloride channels associated with GABA(A) receptors. Indeed through an original algorithm, chloride currents elicited by application of appropriate agonists of the GABA(A) receptor can be derived from the quantitative phase signal recorded with DHM. Finally, chloride currents can be determined and pharmacologically characterized non-invasively simultaneously on a large cellular sampling by DHM.


Subject(s)
Chloride Channels/metabolism , Membrane Potentials/physiology , Receptors, GABA-A/metabolism , Chlorides/metabolism , HEK293 Cells , Holography/methods , Humans , Ion Channel Gating/drug effects , Ion Channel Gating/physiology , Membrane Potentials/drug effects , Microscopy/methods , Sodium-Potassium-Chloride Symporters/metabolism , Solute Carrier Family 12, Member 2 , Symporters/metabolism , Transfection , gamma-Aminobutyric Acid/pharmacology , K Cl- Cotransporters
16.
Bioorg Med Chem Lett ; 19(20): 5958-61, 2009 Oct 15.
Article in English | MEDLINE | ID: mdl-19740657

ABSTRACT

In a search for GABAA alpha5 ligands that combine high subtype binding selectivity with a marked inverse agonism imidazo[1,5-a][1,2,4]-triazolo[1,5-d][1,4]benzodiazepines were identified as a promising class. A short tandem reaction allowed rapid access to this chemical series, thereby facilitating rapid SAR generation which guided the optimization process. Two compounds (10e and 11f) were found to be active in an in vivo paradigm for cognitive improvement.


Subject(s)
Anticonvulsants/chemistry , Benzodiazepines/chemistry , Cognition Disorders/drug therapy , Receptors, GABA-A/metabolism , Triazoles/chemistry , Adjuvants, Anesthesia/pharmacology , Animals , Anticonvulsants/chemical synthesis , Anticonvulsants/pharmacokinetics , Benzodiazepines/chemical synthesis , Benzodiazepines/pharmacokinetics , Benzodiazepines/pharmacology , Cell Line , Drug Inverse Agonism , GABA-A Receptor Agonists , Humans , Memory, Short-Term/drug effects , Microsomes, Liver/metabolism , Rats , Scopolamine/pharmacology , Structure-Activity Relationship , Triazoles/chemical synthesis , Triazoles/pharmacology
17.
Bioorg Med Chem Lett ; 19(20): 5940-4, 2009 Oct 15.
Article in English | MEDLINE | ID: mdl-19762240

ABSTRACT

Lead optimisation of the imidazo[1,5-a][1,2,4]-triazolo[1,5-d][1,4]benzodiazepine class led to the identification of two clinical leads [RO4882224 (11) and RO4938581 (44)] functioning as novel potent and selective GABAA alpha5 inverse agonists. The unique pharmacological profiles and optimal pharmacokinetic profiles resulted in in vivo activity in selected cognition models.


Subject(s)
Anticonvulsants/pharmacokinetics , Benzodiazepines/pharmacokinetics , Cognition Disorders/drug therapy , Imidazoles/pharmacokinetics , Receptors, GABA-A/metabolism , Triazoles/pharmacokinetics , Animals , Anticonvulsants/chemical synthesis , Anticonvulsants/chemistry , Benzodiazepines/chemical synthesis , Benzodiazepines/chemistry , Cell Line , Disease Models, Animal , Drug Discovery , Drug Inverse Agonism , GABA-A Receptor Agonists , Humans , Imidazoles/chemical synthesis , Imidazoles/chemistry , Mice , Protein Binding , Rats , Seizures/chemically induced , Seizures/drug therapy , Structure-Activity Relationship , Triazoles/chemical synthesis , Triazoles/chemistry
18.
Bioorg Med Chem Lett ; 19(19): 5746-52, 2009 Oct 01.
Article in English | MEDLINE | ID: mdl-19726184

ABSTRACT

Through iterative design cycles we have discovered a number of novel new classes where the imidazo[1,5-a][1,2,4]-triazolo[1,5-d][1,4]benzodiazepine was deemed the most promising GABA(A) alpha5 inverse agonist class with potential for cognitive enhancement. This class combines a modest subtype binding selectivity with inverse agonism and has the most favourable molecular properties for further lead optimisation towards a central nervous system (CNS) acting medicine.


Subject(s)
Benzodiazepines/chemistry , Nootropic Agents/chemistry , Receptors, GABA-A/metabolism , Triazoles/chemistry , Animals , Benzodiazepines/chemical synthesis , Benzodiazepines/pharmacology , Drug Discovery , Drug Inverse Agonism , GABA-A Receptor Agonists , Humans , Nootropic Agents/chemical synthesis , Nootropic Agents/pharmacology , Oocytes/drug effects , Triazoles/chemical synthesis , Triazoles/pharmacology , Xenopus laevis
19.
Psychopharmacology (Berl) ; 202(1-3): 207-23, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18936916

ABSTRACT

RATIONALE: GABAA alpha5 subunit-containing receptors are primarily expressed in the hippocampus and their role in learning and memory has been demonstrated recently by both genetic and pharmacological approaches. OBJECTIVES: The objective of the study is to evaluate the cognitive effects of a novel GABAA alpha5 receptor inverse agonist, RO4938581 in rats and monkeys. MATERIALS AND METHODS: The in vitro profile was determined using radioligand binding and electrophysiological assays for the GABAA alpha1, alpha2, alpha3, and alpha5 receptors. Long-term potentiation (LTP) was performed in mouse hippocampal slices. Cognitive effects were assessed in rats in the delayed match to position (DMTP) task and the Morris water maze. In monkeys, the object retrieval task was used. Pro-convulsant and anxiogenic potentials were evaluated in mice and rats. In vivo receptor occupancy was determined using [3H]-RO0154513. RESULTS: RO4938581 is a potent inverse agonist at the GABAA alpha5 receptor, with both binding and functional selectivity, enhancing hippocampal LTP. RO4938581 reversed scopolamine-induced working memory impairment in the DMTP task (0.3-1 mg/kg p.o.) and diazepam-induced spatial learning impairment (1-10 mg/kg p.o.). RO4938581 improved executive function in monkeys (3-10 mg/kg p.o.). Importantly, RO4938581 showed no anxiogenic and pro-convulsive potential. RO4938581 dose-dependently bound to GABAA alpha5 receptors and approximately 30% receptor occupancy was sufficient to produce enhanced cognition in the rat. CONCLUSIONS: The data further support the potential of GABAA alpha5 receptors as a target for cognition-enhancing drugs. The dual binding and functional selectivity offers an ideal profile for cognition-enhancing effects without the unwanted side effects associated with activity at other GABAA receptor subtypes.


Subject(s)
Benzodiazepines/pharmacology , Cognition/drug effects , Imidazoles/pharmacology , Nootropic Agents/pharmacology , Receptors, GABA-A/drug effects , Animals , Cell Line , Dose-Response Relationship, Drug , Female , Haplorhini , Hippocampus/drug effects , In Vitro Techniques , Learning/drug effects , Long-Term Potentiation/drug effects , Male , Membranes/drug effects , Memory/drug effects , Mice , Mice, Inbred DBA , Motor Activity/drug effects , Patch-Clamp Techniques , Plasmids , Rats , Rats, Wistar , Receptors, GABA-A/genetics , Receptors, GABA-A/metabolism , Recombinant Proteins , Seizures/chemically induced
SELECTION OF CITATIONS
SEARCH DETAIL
...