Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Front Psychiatry ; 15: 1365231, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38979499

RESUMEN

Background: Neurodevelopmental disorders (NDDs) can cause debilitating impairments in social cognition and aberrant functional connectivity in large-scale brain networks, leading to social isolation and diminished everyday functioning. To facilitate the treatment of social impairments, animal models of NDDs that link N- methyl-D-aspartate receptor (NMDAR) hypofunction to social deficits in adulthood have been used. However, understanding the etiology of social impairments in NDDs requires investigating social changes during sensitive windows during development. Methods: We examine social behavior during adolescence using a translational mouse model of NMDAR hypofunction (SR-/-) caused by knocking out serine racemase (SR), the enzyme needed to make D-serine, a key NMDAR coagonist. Species-typical social interactions are maintained through brain-wide neural activation patterns; therefore, we employed whole-brain cFos activity mapping to examine network-level connectivity changes caused by SR deletion. Results: In adolescent SR-/- mice, we observed disinhibited social behavior toward a novel conspecific and rapid social habituation toward familiar social partners. SR-/- mice also spent more time in the open arm of the elevated plus maze which classically points to an anxiolytic behavioral phenotype. These behavioral findings point to a generalized reduction in anxiety-like behavior in both social and non-social contexts in SR-/- mice; importantly, these findings were not associated with diminished working memory. Inter-regional patterns of cFos activation revealed greater connectivity and network density in SR-/- mice compared to controls. Discussion: These results suggest that NMDAR hypofunction - a potential biomarker for NDDs - can lead to generalized behavioral disinhibition in adolescence, potentially arising from disrupted communication between and within salience and default mode networks.

2.
Neurosci Biobehav Rev ; 161: 105651, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38579901

RESUMEN

GABA is the primary inhibitory neurotransmitter in the adult brain and through its actions on GABAARs, it protects against excitotoxicity and seizure activity, ensures temporal fidelity of neurotransmission, and regulates concerted rhythmic activity of neuronal populations. In the developing brain, the development of GABAergic neurons precedes that of glutamatergic neurons and the GABA system serves as a guide and framework for the development of other brain systems. Despite this early start, the maturation of the GABA system also continues well into the early postnatal period. In this review, we organize evidence around two scenarios based on the essential and protracted nature of GABA system development: 1) disruptions in the development of the GABA system can lead to large scale disruptions in other developmental processes (i.e., GABA as the cause), 2) protracted maturation of this system makes it vulnerable to the effects of developmental insults (i.e., GABA as the effect). While ample evidence supports the importance of GABA/GABAAR system in both scenarios, large gaps in existing knowledge prevent strong mechanistic conclusions.


Asunto(s)
Encéfalo , Ácido gamma-Aminobutírico , Ácido gamma-Aminobutírico/metabolismo , Humanos , Animales , Encéfalo/crecimiento & desarrollo , Encéfalo/metabolismo , Receptores de GABA-A/metabolismo , Neuronas GABAérgicas/fisiología , Neuronas GABAérgicas/metabolismo
3.
bioRxiv ; 2023 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-37398055

RESUMEN

The biological significance of a small supernumerary marker chromosome that results in dosage alterations to chromosome 9p24.1, including triplication of the GLDC gene encoding glycine decarboxylase, in two patients with psychosis is unclear. In an allelic series of copy number variant mouse models, we identify that triplication of Gldc reduces extracellular glycine levels as determined by optical fluorescence resonance energy transfer (FRET) in dentate gyrus (DG) but not in CA1, suppresses long-term potentiation (LTP) in mPP-DG synapses but not in CA3-CA1 synapses, reduces the activity of biochemical pathways implicated in schizophrenia and mitochondrial bioenergetics, and displays deficits in prepulse inhibition, startle habituation, latent inhibition, working memory, sociability and social preference. Our results thus provide a link between a genomic copy number variation, biochemical, cellular and behavioral phenotypes, and further demonstrate that GLDC negatively regulates long-term synaptic plasticity at specific hippocampal synapses, possibly contributing to the development of neuropsychiatric disorders.

4.
Front Psychiatry ; 13: 1060949, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36713896

RESUMEN

Benzodiazepines have been in use for over half a century. While they remain highly prescribed, their unfavorable side-effect profile and abuse liability motivated a search for alternatives. Most of these efforts focused on the development of benzodiazepine-like drugs that are selective for specific GABAA receptor subtypes. While there is ample evidence that subtype-selective GABAA receptor ligands have great potential for providing symptom relief without typical benzodiazepine side-effects, it is less clear whether subtype-selective targeting strategies can also reduce misuse and abuse potential. This review focuses on the three benzodiazepine properties that are relevant to the DSM-5-TR criteria for Sedative, Hypnotic, or Anxiolytic Use Disorder, namely, reinforcing properties of benzodiazepines, maladaptive behaviors related to benzodiazepine use, and benzodiazepine tolerance and dependence. We review existing evidence regarding the involvement of different GABAA receptor subtypes in each of these areas. The reviewed studies suggest that α1-containing GABAA receptors play an integral role in benzodiazepine-induced plasticity in reward-related brain areas and might be involved in the development of tolerance and dependence to benzodiazepines. However, a systematic comparison of the contributions of all benzodiazepine-sensitive GABAA receptors to these processes, a mechanistic understanding of how the positive modulation of each receptor subtype might contribute to the brain mechanisms underlying each of these processes, and a definitive answer to the question of whether specific chronic modulation of any given subtype would result in some or all of the benzodiazepine effects are currently lacking from the literature. Moreover, how non-selective benzodiazepines might lead to the maladaptive behaviors listed in DSM and how different GABAA receptor subtypes might be involved in the development of these behaviors remains unexplored. Considering the increasing burden of benzodiazepine abuse, the common practice of benzodiazepine misuse that leads to severe dependence, and the current efforts to generate side-effect free benzodiazepine alternatives, there is an urgent need for systematic, mechanistic research that provides a better understanding of the brain mechanisms of benzodiazepine misuse and abuse, including the involvement of specific GABAA receptor subtypes in these processes, to establish an informed foundation for preclinical and clinical efforts.

5.
Neuropsychopharmacology ; 46(12): 2197-2206, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34408277

RESUMEN

Brain α2-containing GABAA receptors play a critical role in the modulation of anxiety- and fear-like behavior. However, it is unknown whether these receptors also play a role in modulating resilience to chronic stress, and in which brain areas and cell types such an effect would be mediated. We evaluated the role of α2-containing GABAA receptors following chronic social defeat stress using male mice deficient in the α2 subunit globally or conditionally in dopamine D1- or D2-receptor-expressing neurons, e.g., within the nucleus accumbens (NAc). In addition, we examined the effect of the lack of the α2 subunit on intermediates of the glutathione synthesis pathway. We found that α2-containing GABAA receptors on D2-receptor-positive but not on D1-receptor-positive neurons promote resiliency to chronic social defeat stress, as reflected in social interaction tests. The pro-resiliency effects of α2-containing GABAA receptors on D2-receptor-positive neurons do not appear to be directly related to alterations in anxiety-like behavior, as reflected in the elevated plus-maze, light-dark box, and novel open field tests. Increases in indices of oxidative stress-reflected by increases in cystathionine levels and reductions in GSH/GSSG ratios-were found in the NAc and prefrontal cortex but not in the hippocampus of mice lacking α2-containing GABAA receptors. We conclude that α2-containing GABAA receptors within specific brain areas and cell populations promote stress resiliency independently of direct effects on anxiety-like behaviors. A potential mechanism contributing to this increased resiliency is the protection that α2-containing GABAA receptors provide against oxidative stress in NAc and the prefrontal cortex.


Asunto(s)
Ansiedad , Receptores de GABA-A/metabolismo , Receptores de GABA , Animales , Masculino , Ratones , Ratones Endogámicos C57BL , Receptores de Dopamina D1/metabolismo , Ácido gamma-Aminobutírico
6.
Learn Mem ; 27(10): 423-428, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32934095

RESUMEN

Reduction in the expression or function of α5-subunit-containing GABAA receptors (α5GABAARs) leads to improvement in several hippocampus-dependent memory domains. However, studies thus far mostly lack anatomical specificity in terms of neuronal circuits and populations. We demonstrate that mice with a selective knockdown of α5GABAARs in CA1 pyramidal neurons (α5CA1KO mice) show improved spatial and trace fear-conditioning memory. Unexpectedly, α5CA1KO mice were comparable to controls in contextual fear-conditioning but showed an impairment in context discrimination, suggesting fine-tuning of activity in CA1 pyramidal cell dendrites through α5-mediated inhibition might be necessary for distinguishing highly similar contexts.


Asunto(s)
Región CA1 Hipocampal/fisiología , Memoria/fisiología , Receptores de GABA-A/fisiología , Animales , Condicionamiento Clásico/fisiología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Prueba del Laberinto Acuático de Morris/fisiología
7.
Trends Pharmacol Sci ; 39(8): 710-732, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29903580

RESUMEN

In the past 20 years we have learned a great deal about GABAA receptor (GABAAR) subtypes, and which behaviors are regulated or which drug effects are mediated by each subtype. However, the question of where GABAARs involved in specific drug effects and behaviors are located in the brain remains largely unanswered. We review here recent studies taking a circuit pharmacology approach to investigate the functions of GABAAR subtypes in specific brain circuits controlling fear, anxiety, learning, memory, reward, addiction, and stress-related behaviors. The findings of these studies highlight the complexity of brain inhibitory systems and the importance of taking a subtype-, circuit-, and neuronal population-specific approach to develop future therapeutic strategies using cell type-specific drug delivery.


Asunto(s)
Benzodiazepinas/farmacología , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Receptores de GABA-A/clasificación , Ácido gamma-Aminobutírico/farmacología , Animales , Agonistas de Receptores de GABA-A/farmacología , Antagonistas de Receptores de GABA-A/farmacología , Humanos , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Receptores de GABA-A/metabolismo
9.
Behav Brain Res ; 332: 172-179, 2017 08 14.
Artículo en Inglés | MEDLINE | ID: mdl-28587819

RESUMEN

Deficits in neuronal inhibition via gamma-aminobutyric acid (GABA) type A receptors (GABAA-Rs) are implicated in the pathophysiology of major depressive disorder and the therapeutic effects of current antidepressant treatments, however, the relevant GABAA-R subtype as defined by its alpha subunit is still unknown. We previously reported anxiety- and depressive-like behavior in alpha2+/- and alpha2-/- mice, respectively (Vollenweider, 2011). We sought to determine whether this phenotype could be reversed by chronic antidepressant treatment. Adult male mice received 4 or 8mg/kg fluoxetine or 53mg/kg desipramine in their drinking water for four weeks before undergoing behavioral testing. In the novelty suppressed feeding test, desipramine had anxiolytic-like effects reducing the latencies to bite and to eat the pellet in both wild-type and alpha2+/- mice. Surprisingly, 4mg/kg fluoxetine had anxiogenic-like effects in alpha2+/- mice increasing latency to bite and to eat while 8mg/kg fluoxetine increased the latency to eat in both wild-type and alpha2+/- mice. In the forced swim and tail suspension tests, chronic desipramine treatment increased latency to immobility in wild-type and alpha2-/- mice. In contrast, chronic fluoxetine treatment increased immobility in alpha2-/- mice in both tasks while generally having no effect in wild-type mice. These findings suggest that in preclinical paradigms of anxiety and behavioral despair the antidepressant-like effects of desipramine are independent of alpha2-containing GABAA-Rs, while a reduction in alpha2 expression leads to an increased sensitivity to anxiogenic- and prodepressant-like effects with chronic fluoxetine treatment, pointing to a potential role of alpha2-containing GABAA-Rs in the response to serotonin-selective antidepressants.


Asunto(s)
Inhibidores de Captación Adrenérgica/farmacología , Antidepresivos/farmacología , Desipramina/farmacología , Fluoxetina/farmacología , Receptores de GABA-A/deficiencia , Inhibidores Selectivos de la Recaptación de Serotonina/farmacología , Inhibidores de Captación Adrenérgica/sangre , Animales , Antidepresivos/sangre , Ansiedad/tratamiento farmacológico , Ansiedad/metabolismo , Depresión/tratamiento farmacológico , Depresión/metabolismo , Desipramina/sangre , Relación Dosis-Respuesta a Droga , Conducta Alimentaria/efectos de los fármacos , Conducta Alimentaria/fisiología , Fluoxetina/análogos & derivados , Fluoxetina/sangre , Masculino , Ratones de la Cepa 129 , Ratones Noqueados , Actividad Motora/efectos de los fármacos , Actividad Motora/fisiología , Fenotipo , Receptores de GABA-A/genética , Inhibidores Selectivos de la Recaptación de Serotonina/sangre
10.
Neuropsychopharmacology ; 41(10): 2492-501, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27067130

RESUMEN

Benzodiazepines have been widely used for their anxiolytic actions. However, the contribution of GABAA receptor subtypes to anxiolysis is still controversial. Studies with mutant mice harboring diazepam-insensitive α-subunits α1, α2, α3, or α5 have revealed that α2-containing GABAA receptors (α2-GABAARs) are required for diazepam-induced anxiolysis, with no evidence for an involvement of any other α-subunit, whereas TP003, described as a selective modulator of α3-containing GABAA receptors, was shown to be anxiolytic. Here, we describe a novel, systematic approach to evaluate the role of positive allosteric modulation of each of the four diazepam-sensitive α-subtypes in anxiety-related behavioral paradigms. By combining H to R point mutations in three out of the four diazepam-sensitive α-subunits in mice with a 129X1/SvJ background, diazepam becomes a subtype-specific modulator of the remaining non-mutated α-subtype. Modulation of α5-GABAARs, but not of α2-GABAARs, increased the time in the light side of the light-dark box as well as open-arm exploration in the elevated plus maze. In contrast, modulation of α3-GABAARs decreased open-arm exploration, whereas modulation of α2-GABAARs increased time in the center in the open-field test. Modulation of any single α-subtype had no effect on stress-induced hyperthermia. Our results provide evidence that modulation of α5-GABAARs elicits anxiolytic-like actions, whereas our data do not provide evidence for an anxiolytic-like action of α3-GABAARs. Thus, α5-GABAARs may be suitable targets for novel anxiolytic drugs.


Asunto(s)
Ansiolíticos/uso terapéutico , Ansiedad/tratamiento farmacológico , Ansiedad/genética , Farmacogenética , Receptores de GABA-A/metabolismo , Animales , Autorradiografía , Adaptación a la Oscuridad/efectos de los fármacos , Adaptación a la Oscuridad/genética , Modelos Animales de Enfermedad , Conducta Exploratoria/efectos de los fármacos , Fiebre/tratamiento farmacológico , Fiebre/etiología , Agonistas de Receptores de GABA-A/farmacología , Agonistas de Receptores de GABA-A/uso terapéutico , Imidazoles/farmacología , Imidazoles/uso terapéutico , Aprendizaje por Laberinto/efectos de los fármacos , Ratones , Ratones Transgénicos , Mutación/genética , Piridinas/farmacología , Piridinas/uso terapéutico , Receptores de GABA-A/genética , Estadísticas no Paramétricas , Estrés Psicológico/complicaciones
11.
Elife ; 5: e14120, 2016 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-26971710

RESUMEN

Recent findings indicate a high level of specialization at the level of microcircuits and cell populations within brain structures with regards to the control of fear and anxiety. The hippocampus, however, has been treated as a unitary structure in anxiety and fear research despite mounting evidence that different hippocampal subregions have specialized roles in other cognitive domains. Using novel cell-type- and region-specific conditional knockouts of the GABAA receptor α2 subunit, we demonstrate that inhibition of the principal neurons of the dentate gyrus or CA3 via α2-containing GABAA receptors (α2GABAARs) is required to suppress anxiety, while the inhibition of CA1 pyramidal neurons is required to suppress fear responses. We further show that the diazepam-modulation of hippocampal theta activity shows certain parallels with our behavioral findings, suggesting a possible mechanism for the observed behavioral effects. Thus, our findings demonstrate a double dissociation in the regulation of anxiety versus fear by hippocampal microcircuitry.


Asunto(s)
Ansiedad , Mapeo Encefálico , Miedo , Hipocampo/fisiología , Animales , Ratones , Red Nerviosa , Receptores de GABA-A/genética
12.
J Neurosci ; 35(40): 13698-712, 2015 Oct 07.
Artículo en Inglés | MEDLINE | ID: mdl-26446222

RESUMEN

Interference between similar or overlapping memories formed at different times poses an important challenge on the hippocampal declarative memory system. Difficulties in managing interference are at the core of disabling cognitive deficits in neuropsychiatric disorders. Computational models have suggested that, in the normal brain, the sparse activation of the dentate gyrus granule cells maintained by tonic inhibitory control enables pattern separation, an orthogonalization process that allows distinct representations of memories despite interference. To test this mechanistic hypothesis, we generated mice with significantly reduced expression of the α5-containing GABAA (α5-GABAARs) receptors selectively in the granule cells of the dentate gyrus (α5DGKO mice). α5DGKO mice had reduced tonic inhibition of the granule cells without any change in fast phasic inhibition and showed increased activation in the dentate gyrus when presented with novel stimuli. α5DGKO mice showed impairments in cognitive tasks characterized by high interference, without any deficiencies in low-interference tasks, suggesting specific impairment of pattern separation. Reduction of fast phasic inhibition in the dentate gyrus through granule cell-selective knock-out of α2-GABAARs or the knock-out of the α5-GABAARs in the downstream CA3 area did not detract from pattern separation abilities, which confirms the anatomical and molecular specificity of the findings. In addition to lending empirical support to computational hypotheses, our findings have implications for the treatment of interference-related cognitive symptoms in neuropsychiatric disorders, particularly considering the availability of pharmacological agents selectively targeting α5-GABAARs. SIGNIFICANCE STATEMENT: Interference between similar memories poses a significant limitation on the hippocampal declarative memory system, and impaired interference management is a cognitive symptom in many disorders. Thus, understanding mechanisms of successful interference management or processes that can lead to interference-related memory problems has high theoretical and translational importance. This study provides empirical evidence that tonic inhibition in the dentate gyrus (DG), which maintains sparseness of neuronal activation in the DG, is essential for management of interference. The specificity of findings to tonic, but not faster, more transient types of neuronal inhibition and to the DG, but not the neighboring brain areas, is presented through control experiments. Thus, the findings link interference management to a specific mechanism, proposed previously by computational models.


Asunto(s)
Giro Dentado/citología , Memoria/fisiología , Inhibición Neural/genética , Neuronas/fisiología , Receptores de GABA-A/metabolismo , Animales , Discriminación en Psicología/fisiología , Conducta Exploratoria/fisiología , Agonistas del GABA/farmacocinética , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Imidazoles/farmacocinética , Técnicas In Vitro , Potenciales Postsinápticos Inhibidores/efectos de los fármacos , Potenciales Postsinápticos Inhibidores/genética , Aprendizaje por Laberinto/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuronas/efectos de los fármacos , Proteínas Proto-Oncogénicas c-fos/metabolismo , Receptores de GABA-A/genética , Reconocimiento en Psicología/fisiología , Natación/psicología
13.
J Neurosci ; 35(26): 9707-16, 2015 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-26134653

RESUMEN

Previous experiments using genetic and pharmacological manipulations have provided strong evidence that etomidate impairs synaptic plasticity and memory by modulating α5-subunit containing GABAA receptors (α5-GABAARs). Because α5-GABAARs mediate tonic inhibition (TI) in hippocampal CA1 pyramidal cells and etomidate enhances TI, etomidate enhancement of TI in pyramidal cells has been proposed as the underlying mechanism (Martin et al., 2009). Here we tested this hypothesis by selectively removing α5-GABAARs from pyramidal neurons (CA1-pyr-α5-KO) and comparing the ability of etomidate to enhance TI and block LTP in fl-α5 (WT), global-α5-KO (gl-α5-KO), and CA1-pyr-α5-KO mice. Etomidate suppressed LTP in slices from WT and CA1-pyr-α5-KO but not gl-α5-KO mice. There was a trend toward reduced TI in both gl-α5-KO and CA1-pyr-α5-KO mice, but etomidate enhanced TI to similar levels in all genotypes. The dissociation between effects of etomidate on TI and LTP in gl-α5-KO mice indicates that increased TI in pyramidal neurons is not the mechanism by which etomidate impairs LTP and memory. Rather, the ability of etomidate to block LTP in WT and CA1-pyr-α5-KO mice, but not in gl-α5-KO mice, points toward α5-GABAARs on nonpyramidal cells as the essential effectors controlling plasticity in this in vitro model of learning and memory.


Asunto(s)
Etomidato/farmacología , Hipocampo/citología , Hipnóticos y Sedantes/farmacología , Potenciación a Largo Plazo/efectos de los fármacos , Neuronas/efectos de los fármacos , Receptores de GABA-A/metabolismo , Animales , Biofisica , Estimulación Eléctrica , Antagonistas de Aminoácidos Excitadores/farmacología , Antagonistas del GABA/farmacología , Técnicas In Vitro , Ácido Quinurénico , Potenciación a Largo Plazo/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuronas/fisiología , Picrotoxina/farmacología , Receptores de GABA-A/genética , Ácido gamma-Aminobutírico/farmacología
14.
J Neurosci ; 34(10): 3653-67, 2014 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-24599464

RESUMEN

Transient receptor potential (TRP) channels are abundant in the brain where they regulate transmission of sensory signals. The expression patterns of different TRPC subunits (TRPC1, 4, and 5) are consistent with their potential role in fear-related behaviors. Accordingly, we found recently that mutant mice lacking a specific TRP channel subunit, TRPC5, exhibited decreased innate fear responses. Both TRPC5 and another member of the same subfamily, TRPC4, form heteromeric complexes with the TRPC1 subunit (TRPC1/5 and TRPC1/4, respectively). As TRP channels with specific subunit compositions may have different functional properties, we hypothesized that fear-related behaviors could be differentially controlled by TRPCs with distinct subunit arrangements. In this study, we focused on the analysis of mutant mice lacking the TRPC4 subunit, which, as we confirmed in experiments on control mice, is expressed in brain areas implicated in the control of fear and anxiety. In behavioral experiments, we found that constitutive ablation of TRPC4 was associated with diminished anxiety levels (innate fear). Furthermore, knockdown of TRPC4 protein in the lateral amygdala via lentiviral-mediated gene delivery of RNAi mimicked the behavioral phenotype of constitutive TRPC4-null (TRPC4(-/-)) mouse. Recordings in brain slices demonstrated that these behavioral modifications could stem from the lack of TRPC4 potentiation in neurons in the lateral nucleus of the amygdala through two Gαq/11 protein-coupled signaling pathways, activated via Group I metabotropic glutamate receptors and cholecystokinin 2 receptors, respectively. Thus, TRPC4 and the structurally and functionally related subunit, TRPC5, may both contribute to the mechanisms underlying regulation of innate fear responses.


Asunto(s)
Amígdala del Cerebelo/metabolismo , Ansiedad/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/fisiología , Canales Catiónicos TRPC/deficiencia , Animales , Ansiedad/genética , Ansiedad/psicología , Regulación hacia Abajo/genética , Potenciales Evocados Somatosensoriales/genética , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/genética , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Canales Catiónicos TRPC/biosíntesis
15.
Neuropsychopharmacology ; 39(8): 1805-15, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24553732

RESUMEN

Despite long-standing concerns regarding the abuse liability of benzodiazepines, the mechanisms underlying properties of benzodiazepines that may be relevant to abuse are still poorly understood. Earlier studies showed that compounds selective for α1-containing GABAA receptors (α1GABAARs) are abused by humans and self-administered by animals, and that these receptors may underlie a preference for benzodiazepines as well as neuroplastic changes observed in the ventral tegmental area following benzodiazepine administration. There is some evidence, however, that even L-838, 417, a compound with antagonistic properties at α1GABAARs and agonistic properties at the other three benzodiazepine-sensitive GABAA receptor subtypes, is self-administered, and that the α2GABAARs may have a role in benzodiazepine-induced reward enhancement. Using a two-bottle choice drinking paradigm to evaluate midazolam preference and an intracranial self-stimulation (ICSS) paradigm to evaluate the impact of midazolam on reward enhancement, we demonstrated that mice carrying a histidine-to-arginine point mutation in the α2 subunit which renders it insensitive to benzodiazepines (α2(H101R) mice) did not prefer midazolam and did not show midazolam-induced reward enhancement in ICSS, in contrast to wild-type controls, suggesting that α2GABAARs are necessary for the reward enhancing effects and preference for oral benzodiazepines. Through a viral-mediated knockdown of α2GABAARs in the nucleus accumbens (NAc), we demonstrated that α2 in the NAc is necessary for the preference for midazolam. Findings imply that α2GABAARs in the NAc are involved in at least some reward-related properties of benzodiazepines, which might partially underlie repeated drug-taking behavior.


Asunto(s)
Conducta de Elección/efectos de los fármacos , Moduladores del GABA/farmacología , Midazolam/farmacología , Núcleo Accumbens/efectos de los fármacos , Receptores de GABA-A/fisiología , Recompensa , Animales , Masculino , Ratones , Ratones Transgénicos
17.
Pharmacol Ther ; 136(2): 142-52, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22921455

RESUMEN

GABA(A) receptors have important physiological functions, as revealed by pharmacological studies and experiments involving gene-targeted mouse models, and are the target of widely used drugs such as the benzodiazepines. In this review, we are summarizing current knowledge about the function of α2-containing GABA(A) receptors, a receptor subtype representing approximately 15-20% of all GABA(A) receptors. This receptor subtype mediates anxiolytic-like, reward-enhancing, and antihyperalgesic actions of diazepam, and has antidepressant-like properties. Secondary insufficiency of α2-containing GABA(A) receptors has been postulated to play a role in the pathogenesis of schizophrenia, and may be involved in cognitive impairment in other disorders. Moreover, polymorphisms in the GABRA2 gene encoding the GABA(A) receptor α2 subunit have been found to be linked to chronic alcohol dependence and to polydrug abuse. Thus, α2-containing GABA(A) receptors are involved in the regulation and/or modulation of emotional behaviors and of chronic pain, and appear to be a valid target for novel therapeutic approaches for the treatment of anxiety, depression, schizophrenia and chronic pain.


Asunto(s)
Enfermedades del Sistema Nervioso Central/tratamiento farmacológico , Agonistas de Receptores de GABA-A/uso terapéutico , Receptores de GABA-A/fisiología , Alcoholismo/tratamiento farmacológico , Alcoholismo/etiología , Animales , Ansiedad/tratamiento farmacológico , Ansiedad/etiología , Dolor Crónico/tratamiento farmacológico , Dolor Crónico/etiología , Trastornos del Conocimiento/tratamiento farmacológico , Trastornos del Conocimiento/etiología , Depresión/tratamiento farmacológico , Depresión/etiología , Humanos , Polimorfismo Genético , Piridazinas/uso terapéutico , Receptores de GABA-A/genética , Esquizofrenia/tratamiento farmacológico , Esquizofrenia/etiología , Trastornos Relacionados con Sustancias/tratamiento farmacológico , Trastornos Relacionados con Sustancias/etiología , Triazoles/uso terapéutico
18.
Neuropsychopharmacology ; 37(11): 2531-40, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22763624

RESUMEN

Benzodiazepines such as diazepam are widely prescribed as anxiolytics and sleep aids. Continued use of benzodiazepines, however, can lead to addiction in vulnerable individuals. Here, we investigate the neural mechanisms of the behavioral effects of benzodiazepines using the intracranial self-stimulation (ICSS) test, a procedure with which the reward-enhancing effects of these drugs can be measured. Benzodiazepines bind nonselectively to several different GABA(A) receptor subtypes. To elucidate the α subunit(s) responsible for the reward-enhancing effects of benzodiazepines, we examined mice carrying a histidine-to-arginine point mutation in the α1, α2, or α3 subunit, which renders the targeted subunit nonresponsive to diazepam, other benzodiazepines and zolpidem. In wild-type and α1-point-mutated mice, diazepam caused a dose-dependent reduction in ICSS thresholds (reflecting a reward-enhancing effect) that is comparable to the reduction observed following cocaine administration. This effect was abolished in α2- and α3-point-mutant mice, suggesting that these subunits are necessary for the reward-enhancing action of diazepam. α2 Subunits appear to be particularly important, since diazepam increased ICSS thresholds (reflecting an aversive-like effect) in α2-point-mutant animals. Zolpidem, an α1-preferring benzodiazepine-site agonist, had no reward-enhancing effects in any genotype. Our findings implicate α2 and α3 subunit containing GABA(A) receptors as key mediators of the reward-related effects of benzodiazepines. This finding has important implications for the development of new medications that retain the therapeutic effects of benzodiazepines but lack abuse liability.


Asunto(s)
Conducta Animal/efectos de los fármacos , Benzodiazepinas/farmacología , Encéfalo/efectos de los fármacos , Mutación Puntual/genética , Receptores de GABA-A/fisiología , Recompensa , Análisis de Varianza , Animales , Conducta Animal/fisiología , Encéfalo/fisiología , Cocaína/administración & dosificación , Inhibidores de Captación de Dopamina/administración & dosificación , Relación Dosis-Respuesta a Droga , Estimulación Eléctrica , GABAérgicos/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Subunidades de Proteína/genética , Receptores de GABA-A/genética , Esquema de Refuerzo , Autoestimulación/efectos de los fármacos
19.
Neuropharmacology ; 63(2): 250-8, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22465203

RESUMEN

GABA(A) receptor modulating drugs such as benzodiazepines (BZs) have been used to treat anxiety disorders for over five decades. In order to determine whether the same or different GABA(A) receptor subtypes are necessary for the anxiolytic-like action of BZs in unconditioned anxiety and conditioned fear models, we investigated the role of different GABA(A) receptor subtypes by challenging wild type, α1(H101R), α2(H101R) and α3(H126R) mice bred on the C57BL/6J background with diazepam or chlordiazepoxide in the elevated plus maze and the fear-potentiated startle paradigms. Both drugs significantly increased open arm exploration in the elevated plus maze in wild type, α1(H101R) and α3(H126R), but this effect was abolished in α2(H101R) mice; these were expected results based on previous published results. In contrast, while administration of diazepam and chlordiazepoxide significantly attenuated fear-potentiated startle (FPS) in wild type mice and α3(H126R) mice, the fear-reducing effects of these drugs were absent in both α1(H101R) and α2(H101R) point mutants, indicating that both α1- and α2-containing GABA(A) receptors are necessary for BZs to exert their effects on conditioned fear responses. Our findings illustrate both an overlap and a divergence between the GABA(A) receptor subtype requirements for the impact of BZs, specifically that both α1- and α2-containing GABA(A) receptors are necessary for BZs to reduce conditioned fear whereas only α2-containing GABA(A) receptors are needed for BZ-induced anxiolysis in unconditioned tests of anxiety. This raises the possibility that GABAergic pharmacological interventions for specific anxiety disorders can be differentially tailored.


Asunto(s)
Ansiolíticos/uso terapéutico , Ansiedad/tratamiento farmacológico , Clordiazepóxido/uso terapéutico , Condicionamiento Psicológico/efectos de los fármacos , Diazepam/uso terapéutico , Miedo/efectos de los fármacos , Subunidades de Proteína/metabolismo , Receptores de GABA-A/metabolismo , Animales , Ansiolíticos/farmacología , Ansiedad/genética , Ansiedad/metabolismo , Conducta Animal/efectos de los fármacos , Conducta Animal/fisiología , Clordiazepóxido/farmacología , Condicionamiento Psicológico/fisiología , Diazepam/farmacología , Miedo/fisiología , Masculino , Ratones , Ratones Noqueados , Ratones Mutantes , Subunidades de Proteína/genética , Receptores de GABA-A/genética , Reflejo de Sobresalto/efectos de los fármacos , Reflejo de Sobresalto/fisiología
20.
Psychopharmacology (Berl) ; 216(4): 557-67, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21424237

RESUMEN

RATIONALE AND OBJECTIVES: Somatostatin (SST) isoforms, SST 14 and SST 28, inhibit regulatory hormones in the periphery (e.g., growth hormone) and are widely distributed in the brain. In recent experiments, intracerebroventricular (ICV) SST produced anxiolytic-like effects in both behavioral and electrophysiological models. The sites of action of these anxiolytic effects in the brain, however, and the relative contributions of SST 14 and SST 28 to these effects are unknown. MATERIALS AND METHODS: Anxiolytic effects were assessed in the plus-maze and shock-probe tests after (1) intra-amygdalar microinfusion of SST 14 (0.5 or 3 µg per hemisphere) or SST 28 (3 µg per hemisphere), (2) intra-septal microinfusion of SST 14 (0.5 or 1.5 µg per hemisphere) or SST 28 (1.5 µg per hemisphere), or (3) intra-striatal microinfusion of SST 14 (3 µg per hemisphere). RESULTS: Intra-amygdalar and intra-septal microinfusions of SST 14 and SST 28 produced robust anxiolytic-like effects in the behavioral tests, unlike intra-striatal microinfusions. The magnitude of the anxiolytic effects in the amygdala and septum were comparable to those found previously with ICV SST 14, ICV L-779976, an SST (sst2) receptor agonist, and ICV diazepam, a classical benzodiazepine anxiolytic. CONCLUSIONS: SST receptors in the septum and amygdala are responsive to both SST 14 and SST 28, but not those in the striatum. Although no obvious differences in the anxiolytic-like effects of the isoforms were detected, quantitative or even qualitative differences in their specific anxiolytic effects may occur in different sub-regions of the septum and amygdala, as has been found for benzodiazepine anxiolytics.


Asunto(s)
Conducta Animal/efectos de los fármacos , Aprendizaje por Laberinto/efectos de los fármacos , Receptores de Somatostatina/efectos de los fármacos , Somatostatina/farmacología , Amígdala del Cerebelo/efectos de los fármacos , Amígdala del Cerebelo/metabolismo , Animales , Cuerpo Estriado/efectos de los fármacos , Cuerpo Estriado/metabolismo , Relación Dosis-Respuesta a Droga , Fenómenos Electrofisiológicos , Hormonas/administración & dosificación , Hormonas/química , Hormonas/farmacología , Masculino , Microinyecciones , Isoformas de Proteínas , Ratas , Ratas Sprague-Dawley , Receptores de Somatostatina/metabolismo , Tabique del Cerebro/efectos de los fármacos , Tabique del Cerebro/metabolismo , Somatostatina/administración & dosificación , Somatostatina/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...