Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cells ; 12(24)2023 12 14.
Artículo en Inglés | MEDLINE | ID: mdl-38132157

RESUMEN

Recently, a Y727C variant in the dual-specific 3',5'-cyclic nucleotide phosphodiesterase 11A (PDE11A-Y727C) was linked to increased sleep quality and reduced myopia risk in humans. Given the well-established role that the PDE11 substrates cAMP and cGMP play in eye physiology and sleep, we determined if (1) PDE11A protein is expressed in the retina or other eye segments in mice, (2) PDE11A-Y7272C affects catalytic activity and/or subcellular compartmentalization more so than the nearby suicide-associated PDE11A-M878V variant, and (3) Pde11a deletion alters eye growth or sleep quality in male and female mice. Western blots show distinct protein expression of PDE11A4, but not PDE11A1-3, in eyes of Pde11a WT, but not KO mice, that vary by eye segment and age. In HT22 and COS-1 cells, PDE11A4-Y727C reduces PDE11A4 catalytic activity far more than PDE11A4-M878V, with both variants reducing PDE11A4-cAMP more so than PDE11A4-cGMP activity. Despite this, Pde11a deletion does not alter age-related changes in retinal or lens thickness or axial length, nor vitreous or anterior chamber depth. Further, Pde11a deletion only minimally changes refractive error and sleep quality. That said, both variants also dramatically alter the subcellular compartmentalization of human and mouse PDE11A4, an effect occurring independently of dephosphorylating PDE11A4-S117/S124 or phosphorylating PDE11A4-S162. Rather, re-compartmentalization of PDE11A4-Y727C is due to the loss of the tyrosine changing how PDE11A4 is packaged/repackaged via the trans-Golgi network. Therefore, the protective impact of the Y727C variant may reflect a gain-of-function (e.g., PDE11A4 displacing another PDE) that warrants further investigation in the context of reversing/preventing sleep disturbances or myopia.


Asunto(s)
3',5'-GMP Cíclico Fosfodiesterasas , Miopía , Humanos , Masculino , Femenino , Animales , Ratones , 3',5'-GMP Cíclico Fosfodiesterasas/metabolismo , Hidrolasas Diéster Fosfóricas/genética , Hidrolasas Diéster Fosfóricas/metabolismo , Calidad del Sueño , Western Blotting
2.
bioRxiv ; 2023 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-38014312

RESUMEN

Recently, a Y727C variant in the dual-specific 3',5'-cyclic nucleotide phosphodiesterase 11A (PDE11A-Y727C) was linked to increased sleep quality and reduced myopia risk in humans. Given the well-established role that the PDE11 substrates cAMP and cGMP play in eye physiology and sleep, we determined if 1) PDE11A protein is expressed in the retina or other eye segments in mouse, 2) PDE11A-Y7272C affects catalytic activity and/or subcellular compartmentalization more so than the nearby suicide-associated PDE11A-M878V variant, and 3) Pde11a deletion alters eye growth or sleep quality in male and female mice. Western blots show distinct protein expression of PDE11A4, but not PDE11A1-3, in eyes of Pde11a WT-but not KO mice-that vary by eye segment and age. In HT22 and COS-1 cells, PDE11A4-Y727C reduces PDE11A4 catalytic activity far more than PDE11A4-M878V, with both variants reducing PDE11A4-cAMP more so than PDE11A4-cGMP activity. Despite this, Pde11a deletion does not alter age-related changes in retinal or lens thickness, axial length, nor vitreous or anterior chamber depth. Further, Pde11a deletion only minimally changes refractive error and sleep quality. That said, both variants also dramatically alter the subcellular compartmentalization of human and mouse PDE11A4, an effect occurring independently of dephosphorylating PDE11A4-S117/S124 or phosphorylating PDE11A4-S162. Rather, re-compartmentalization of PDE11A4-Y727C is due to the loss of the tyrosine changing how PDE11A4 is packaged/repackaged via the trans-Golgi network. Therefore, the protective impact of the Y727C variant may reflect a gain-of-function (e.g., PDE11A4 displacing another PDE) that warrants further investigation in the context of reversing/preventing sleep disturbances or myopia.

3.
Aging Cell ; 21(10): e13687, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-36073342

RESUMEN

In humans, associative memories are more susceptible to age-related cognitive decline (ARCD) than are recognition memories. Reduced cAMP/cGMP signaling in the hippocampus may contribute to ARCD. Here, we found that both aging and traumatic brain injury-associated dementia increased the expression of the cAMP/cGMP-degrading enzyme phosphodiesterase 11A (PDE11A) in the human hippocampus. Further, age-related increases in hippocampal PDE11A4 mRNA and protein were conserved in mice, as was the increased vulnerability of associative versus recognition memories to ARCD. Interestingly, mouse PDE11A4 protein in the aged ventral hippocampus (VHIPP) ectopically accumulated in the membrane fraction and filamentous structures we term "ghost axons." These age-related increases in expression were driven by reduced exoribonuclease-mediated degradation of PDE11A mRNA and increased PDE11A4-pS117/pS124, the latter of which also drove the punctate accumulation of PDE11A4. In contrast, PDE11A4-pS162 caused dispersal. Importantly, preventing age-related increases in PDE11 expression via genetic deletion protected mice from ARCD of short-term and remote long-term associative memory (aLTM) in the social transmission of food preference assay, albeit at the expense of recent aLTM. Further, mimicking age-related overexpression of PDE11A4 in CA1 of old KO mice caused aging-like impairments in CREB function and remote social-but not non-social-LTMs. RNA sequencing and phosphoproteomic analyses of VHIPP identified cGMP-PKG-as opposed to cAMP-PKA-as well as circadian entrainment, glutamatergic/cholinergic synapses, calcium signaling, oxytocin, and retrograde endocannabinoid signaling as mechanisms by which PDE11A deletion protects against ARCD. Together, these data suggest that PDE11A4 proteinopathies acutely impair signaling in the aged brain and contribute to ARCD of social memories.


Asunto(s)
3',5'-GMP Cíclico Fosfodiesterasas , Disfunción Cognitiva , 3',5'-GMP Cíclico Fosfodiesterasas/genética , 3',5'-GMP Cíclico Fosfodiesterasas/metabolismo , Anciano , Animales , Colinérgicos/metabolismo , Disfunción Cognitiva/metabolismo , Endocannabinoides/metabolismo , Exorribonucleasas/metabolismo , Hipocampo/metabolismo , Humanos , Ratones , Oxitocina/metabolismo , ARN Mensajero/metabolismo
4.
Biomolecules ; 11(2)2021 01 28.
Artículo en Inglés | MEDLINE | ID: mdl-33525455

RESUMEN

Tourette syndrome (TS) is a disabling neurodevelopmental disorder characterized by multiple, recurrent tics. The pharmacological treatment of TS is currently based on dopaminergic antagonists; however, these drugs are associated with extrapyramidal symptoms and other serious adverse events. Recent evidence suggests that positive allosteric modulators (PAMs) of GABAA receptors containing α6 subunits (α6 GABAARs) oppose the behavioral effects of dopamine. Building on this evidence, in the present study, we tested the efficacy of DK-I-56-1, a highly selective PAM for α6 GABAARs, in mouse models of TS exhibiting tic-related responses. DK-I-56-1 significantly reduced tic-like jerks and prepulse inhibition (PPI) deficits in D1CT-7 transgenic mice, a well-documented mouse model of TS. DK-I-56-1 also prevented the exacerbation of spontaneous eyeblink reflex induced by the potent dopamine D1 receptor agonist SKF 82958, a proxy for tic-like responses. We also showed that both systemic and prefrontal cortical administration of DK-I-56-1 countered the PPI disruption caused by SKF 82958. Although the effects of DK-I-56-1 were akin to those elicited by dopaminergic antagonists, this drug did not elicit extrapyramidal effects, as measured by catalepsy. These results point to α6 GABAAR PAMs as promising TS therapies with a better safety profile than dopaminergic antagonists.


Asunto(s)
Conducta Animal , Receptores de GABA-A/metabolismo , Síndrome de Tourette/genética , Síndrome de Tourette/inmunología , Animales , Benzazepinas/farmacología , Parpadeo , Cataplejía , Modelos Animales de Enfermedad , Dopamina/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Corteza Prefrontal/metabolismo , Reflejo de Sobresalto , Tics/complicaciones
6.
Mol Neuropsychiatry ; 5(2): 84-97, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-31192221

RESUMEN

Altered gamma-aminobutyric acid (GABA) function is consistently reported in psychiatric disorders, normal aging, and neurodegenerative disorders and reduced function of GABA interneurons is associated with both mood and cognitive symptoms. Benzodiazepines (BZ) have broad anxiolytic, but also sedative, anticonvulsant and amnesic effects, due to nonspecific GABA-A receptor (GABAA-R) targeting. Varying the profile of activity of BZs at GABAA-Rs is predicted to uncover additional therapeutic potential. We synthesized four novel imidazobenzodiazepine (IBZD) amide ligands and tested them for positive allosteric modulation at multiple α-GABAA-R (α-positive allosteric modulators), pharmacokinetic properties, as well as anxiolytic and antidepressant activities in adult mice. Efficacy at reversing stress-induced or age-related working memory deficits was assessed using a spontaneous alternation task. Diazepam (DZP) was used as a control. Three ligands (GL-II-73, GL-II-74, and GL-II-75) demonstrated adequate brain penetration and showed predictive anxiolytic and antidepressant efficacies. GL-II-73 and GL-II-75 significantly reversed stress-induced and age-related working memory deficits. In contrast, DZP displayed anxiolytic but no antidepressant effects or effects on working memory. We demonstrate distinct profiles of anxiolytic, antidepressant, and/or pro-cognitive activities of newly designed IBZD amide ligands, suggesting novel therapeutic potential for IBZD derivatives in depression and aging.

7.
Mol Pharm ; 15(5): 1766-1777, 2018 05 07.
Artículo en Inglés | MEDLINE | ID: mdl-29578347

RESUMEN

We describe lead compound MIDD0301 for the oral treatment of asthma based on previously developed positive allosteric α5ß3γ2 selective GABAA receptor (GABAAR) ligands. MIDD0301 relaxed airway smooth muscle at single micromolar concentrations as demonstrated with ex vivo guinea pig tracheal rings. MIDD0301 also attenuated airway hyperresponsiveness (AHR) in an ovalbumin murine model of asthma by oral administration. Reduced numbers of eosinophils and macrophages were observed in mouse bronchoalveolar lavage fluid without changing mucous metaplasia. Importantly, lung cytokine expression of IL-17A, IL-4, and TNF-α were reduced for MIDD0301-treated mice without changing antiinflammatory cytokine IL-10 levels. Automated patch clamp confirmed amplification of GABA induced current mediated by α1-3,5ß3γ2 GABAARs in the presence of MIDD0301. Pharmacodynamically, transmembrane currents of ex vivo CD4+ T cells from asthmatic mice were potentiated by MIDD0301 in the presence of GABA. The number of CD4+ T cells observed in the lung of MIDD0301-treated mice were reduced by an oral treatment of 20 mg/kg b.i.d. for 5 days. A half-life of almost 14 h was demonstrated by pharmacokinetic studies (PK) with no adverse CNS effects when treated mice were subjected to sensorimotor studies using the rotarod. PK studies also confirmed very low brain distribution. In conclusion, MIDD0301 represents a safe and improved oral asthma drug candidate that relaxes airway smooth muscle and attenuates inflammation in the lung leading to a reduction of AHR at a dosage lower than earlier reported GABAAR ligands.


Asunto(s)
Asma/tratamiento farmacológico , Inflamación/tratamiento farmacológico , Pulmón/efectos de los fármacos , Músculo Liso/efectos de los fármacos , Receptores de GABA-A/metabolismo , Animales , Asma/metabolismo , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Líquido del Lavado Bronquioalveolar/química , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD4-Positivos/metabolismo , Constricción , Citocinas/metabolismo , Eosinófilos/efectos de los fármacos , Eosinófilos/metabolismo , Femenino , Cobayas , Inflamación/metabolismo , Ligandos , Pulmón/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Músculo Liso/metabolismo , Ovalbúmina/metabolismo , Hipersensibilidad Respiratoria/metabolismo
8.
ACS Chem Neurosci ; 8(6): 1305-1312, 2017 06 21.
Artículo en Inglés | MEDLINE | ID: mdl-28150939

RESUMEN

Pain remains a challenging clinical condition and spinal GABAA receptors are crucial modulators of pain processing. α2/α3-subtype GABAA receptors mediate the analgesic actions of benzodiazepines. Positive allosteric modulators (PAMs) at α2/α3-subtype GABAA receptors may have analgesic potential. Here we report a new selective α2/α3-subtype GABAA receptor PAM in in vitro and in vivo pain assays. KRM-II-81 demonstrated similar efficacy at α1/α2/α3 GABAA receptors and negligible efficacy at α4/α5/α6 GABAA receptors, with α2 and α3-subtypes being 17- and 28-fold more potent than α1 subtypes in HEK-293T cells expressing GABAA receptors with different α subunits. In contrast, KRM-II-18B showed significant efficacy at α1/α2/α3/ α5 subtypes, with similar potency at α1/α2/α3 subtypes. Both PAMs and morphine dose-dependently decreased 0.6% acetic acid- and 0.32% lactic acid-induced writhing. The effects of both PAMs were reversed by the benzodiazepine receptor antagonist flumazenil, confirming their action at the benzodiazepine binding site of GABAA receptors. Both PAMS and morphine all dose-dependently reversed 0.32% lactic acid (but not 0.6% acetic acid) induced suppression of nesting behavior. Acetaminophen, but not the PAMs, reversed acid-depressed locomotor activity. Combined, these findings suggest that KRM-II-81 is a selective α2/α3 subtype GABAA PAM with significant antinociceptive effects in chemical stimulation-induced pain in mice.


Asunto(s)
Analgésicos/farmacología , Agonistas de Receptores de GABA-A/farmacología , Oxazoles/farmacología , Receptores de GABA-A/efectos de los fármacos , Regulación Alostérica/efectos de los fármacos , Sitio Alostérico/efectos de los fármacos , Animales , Células HEK293 , Humanos , Masculino , Ratones , Ratones Endogámicos ICR
9.
Neuropharmacology ; 99: 471-80, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26277340

RESUMEN

The kainate-type of ionotropic glutamate receptors are assembled from a combination of five different pore-forming subunits (GluK1-5), which confer distinct functional and pharmacological properties. These receptors are also modulated by co-assembly with the auxiliary subunits Neto1 and Neto2. To determine the impact of variation in subunit composition on the functional interaction between kainate receptors and Neto subunits, the Neto subunits were combined with either GluK1 or GluK2 in HEK-293T cells and responses to glutamate examined through patch-clamp recordings. Co-expression of GluK1 with either Neto1 or Neto2 caused a substantial increase in glutamate sensitivity and a slowing of the onset of desensitization at low agonist concentrations. However, at higher glutamate concentrations the primary effect of Neto2 was to slow the onset of desensitization, while that of Neto1 was to increase recovery from desensitization. In contrast, co-expression of Neto2 with GluK2 homomeric receptors had only modest effects on glutamate sensitivity, but increased the rate of recovery from desensitization as well as slowing its onset at all agonist concentrations. The properties of chimeric Neto1/Neto2 subunits suggested that the extracellular N-terminal region including the two CUB domains was largely responsible for the distinct regulatory effects of Neto1 and Neto2 on the desensitization properties of GluK1 homomeric receptors. These results further demonstrate that the functional effects of Neto subunits depend upon the subunit identity of both the auxiliary and the pore-forming subunits.


Asunto(s)
Ácido Glutámico/metabolismo , Proteínas de la Membrana/metabolismo , Receptores de Ácido Kaínico/metabolismo , Relación Dosis-Respuesta a Droga , Fármacos actuantes sobre Aminoácidos Excitadores/farmacología , Ácido Glutámico/farmacología , Células HEK293 , Humanos , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/fisiología , Proteínas de la Membrana/genética , Técnicas de Placa-Clamp , Receptores de Ácido Kaínico/genética , Receptores de N-Metil-D-Aspartato , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Receptor de Ácido Kaínico GluK2
10.
Neuropharmacology ; 85: 9-17, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24859608

RESUMEN

Ionotropic glutamate receptors (iGluRs) are responsible for fast excitatory neurotransmission in the mammalian brain, and are critical regulators of neuronal activity and synaptic plasticity. The three main types of iGluRs (AMPA, NMDA, and kainate receptors) are composed of distinct subunit populations. The tetrameric kainate receptors can be assembled from a combination of five different types of subunits (GluK1-GluK5). GluK1-3 subunits are able to produce functional homomeric receptors, while GluK4-5 are obligate heteromers, and must assemble with a GluK1-3 subunit. The neurotoxin domoate is widely used as an agonist at kainate-type receptors because it produces a less desensitizing response compared to glutamate. We have identified an additional, subunit-dependent action of domoate at recombinant kainate receptors. When applied to heteromeric GluK2/K5 receptors, domoate generates a small, long-lasting, tonic current. In addition, brief exposure to domoate inhibits the GluK5 subunit, preventing its activation by other agonists for several minutes. These characteristics are not associated with the GluK1, K2, or K4 subunits and can be prevented by a mutation in GluK5 that reduces agonist binding affinity. The results also show that the domoate-bound, GluK2/K5 heteromeric receptors can be fully activated by agonists acting through the GluK2 subunit, suggesting that the subunits within the tetramer can function independently to open the ion channel, and that the domoate-bound state is not a desensitized or blocked conformation. This study describes new properties associated with domoate action at kainate receptors, and further characterizes the distinct roles played by different subunits in heteromeric receptors.


Asunto(s)
Ácido Kaínico/análogos & derivados , Neurotoxinas/farmacología , Receptores de Ácido Kaínico/antagonistas & inhibidores , 6-Ciano 7-nitroquinoxalina 2,3-diona/farmacología , Sitios de Unión/genética , Sitios de Unión/fisiología , Antagonistas de Aminoácidos Excitadores/farmacología , Ácido Glutámico/metabolismo , Células HEK293 , Humanos , Ácido Kaínico/farmacología , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/fisiología , Mutación , Inhibición Neural/efectos de los fármacos , Inhibición Neural/fisiología , Técnicas de Placa-Clamp , Receptores de Ácido Kaínico/agonistas , Receptores de Ácido Kaínico/genética , Receptores de Ácido Kaínico/metabolismo , Transfección , Receptor de Ácido Kaínico GluK2
12.
Cell Mol Neurobiol ; 33(8): 1099-108, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23975096

RESUMEN

Trafficking of ionotropic glutamate receptors to the plasma membrane commonly requires occupation of the agonist binding sites. This quality control check does not typically involve receptor activation, as binding by competitive antagonists or to non-functional channels may also permit surface expression. The tetrameric kainate receptors can be assembled from five different subunits (GluK1-GluK5). While the "low-affinity" GluK1-3 subunits are able to produce functional homomeric receptors, the "high-affinity" GluK4 and GluK5 subunits require co-assembly with GluK1, 2, or 3 for surface expression. These two different types of subunits have distinct functional roles in the receptor. Therefore, we examined the relative importance of occupancy of the agonist site of the GluK2 or GluK5 subunit for surface expression of heteromeric receptors. We created subunits with a mutation within the S2 ligand-binding domain which decreased agonist affinity. Mutations at this site reduced functional surface expression of homomeric GluK2 receptors, but surface expression of these receptors could be increased with either a competitive antagonist or co-assembly with wild-type GluK5. In contrast, mutations in the GluK5 subunit reduced the production of functional heteromeric receptors at the membrane, and could not be rescued with either an antagonist or wild-type GluK2. These findings indicate that ligand binding to only the GluK5 subunit is both necessary and sufficient to allow trafficking of recombinant GluK2/K5 heteromers to the cell membrane, but that occupancy of the GluK2 site alone is not. Our results suggest a distinct role for the GluK5 subunit in regulating surface expression of heteromeric kainate receptors.


Asunto(s)
Membrana Celular/metabolismo , Multimerización de Proteína , Subunidades de Proteína/agonistas , Receptores de Ácido Kaínico/agonistas , Receptores de Ácido Kaínico/metabolismo , 6-Ciano 7-nitroquinoxalina 2,3-diona/farmacología , Secuencia de Aminoácidos , Animales , Sitios de Unión , Membrana Celular/efectos de los fármacos , Glutamatos/farmacología , Células HEK293 , Humanos , Ligandos , Datos de Secuencia Molecular , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Mutación/genética , Unión Proteica/efectos de los fármacos , Multimerización de Proteína/efectos de los fármacos , Subunidades de Proteína/química , Subunidades de Proteína/metabolismo , Ratas , Receptores de Ácido Kaínico/antagonistas & inhibidores , Receptores de Ácido Kaínico/química , Receptor de Ácido Kaínico GluK2
13.
J Pharmacol Exp Ther ; 347(1): 235-41, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23902941

RESUMEN

Many drugs used to treat anxiety are positive modulators of GABAA receptors, which mediate fast inhibitory neurotransmission. The GABAA receptors can be assembled from a combination of at least 16 different subunits. The receptor's subunit composition determines its pharmacologic and functional properties, and subunit expression varies throughout the brain. A primary goal for new treatments targeting GABAA receptors is the production of subunit-selective modulators acting upon a discrete population of receptors. The anxiolytic 4-amino-7-hydroxy-2-methyl-5,6,7,8,-tetrahydrobenzo[b]thieno[2,3-b]pyridine-3-carboxylic acid, but-2-ynyl ester (SB-205384) is widely considered to be selective for α3-containing GABAA receptors. However, it has been tested only on α1-, α2-, and α3-containing receptors. We examined the activity of SB-205384 at recombinant receptors containing the six different α subunits and found that receptors containing the α3, α5, and α6 subunits were potentiated by SB-205384, with the α6 subunit conferring the greatest responsiveness. Properties associated with chimeric α1/α6 subunits suggested that multiple structural domains influence sensitivity to SB-205384. Point mutations of residues within the extracellular N-terminal domain identified a leucine residue located in loop E of the agonist binding site as an important determinant of high sensitivity to modulation. In the α6 subunit the identity of this residue is species-dependent, with the leucine found in rat subunits but not in human. Our results indicate that SB-205384 is not an α3-selective modulator, and instead acts at several GABAA receptor isoforms. These findings have implications for the side-effect profile of this anxiolytic as well as for its use in neuronal and animal studies as a marker for contribution from α3-containing receptors.


Asunto(s)
Aminopiridinas/farmacología , Receptores de GABA-A/biosíntesis , Tiofenos/farmacología , Regulación Alostérica/efectos de los fármacos , Regulación Alostérica/fisiología , Animales , Regulación de la Expresión Génica , Células HEK293 , Humanos , Ratas , Proteínas Recombinantes/biosíntesis
14.
J Physiol ; 591(19): 4711-24, 2013 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-23798491

RESUMEN

The ionotropic glutamate receptors are primary mediators of fast excitatory neurotransmission, and their properties are determined both by their subunit composition and their association with auxiliary subunits. The neuropilin and tolloid-like 1 and 2 proteins (Neto1 and Neto2) have been recently identified as auxiliary subunits for kainate-type glutamate receptors. Heteromeric kainate receptors (KARs) can be assembled from varying combinations of low-affinity (GluK1-GluK3) and high-affinity (GluK4-GluK5) subunits. To better understand the functional impact of auxiliary subunits on KARs, we examined the effect of Neto1 on the responses of recombinant homomeric and heteromeric KARs to varying concentrations of glutamate. We found that co-expression of Neto1 with homomeric GluK2 receptors had a small effect on sensitivity of the receptors to glutamate, but decreased the onset of desensitization while speeding recovery from desensitization. In the absence of Neto1, addition of GluK5 subunits to form GluK2/GluK5 heteromeric receptors slowed the onset of desensitization at low glutamate concentrations, compared with GluK2 homomers. Co-expression of Neto1 with GluK2/GluK5 receptors further enhanced these effects, essentially eliminating desensitization at µm glutamate concentrations without altering the EC50 for activation by glutamate. In addition, a prominent rebound current was observed upon removal of the agonist. The rate of recovery from desensitization was increased to the same degree by Neto1 for both homomeric GluK2 and heteromeric GluK2/GluK5 receptors. Expression of Neto1 with GluK1/GluK5, GluK3/GluK5 or GluK2/GluK4 receptors produced qualitatively similar effects on whole-cell currents, suggesting that the impact of Neto1 on the desensitization properties of heteromeric receptors was not subunit dependent. These results provide greater insight into the functional effects of the auxiliary subunit Neto1 on both homomeric and heteromeric KARs. Alteration of the characteristics of desensitization at both sub-maximal and saturating glutamate concentrations could influence the responsiveness of these receptors to repeated stimuli. As a result, assembly of KARs with the Neto auxiliary subunits could change the kinetic properties of the neuronal response to glutamatergic input.


Asunto(s)
Proteínas de la Membrana/metabolismo , Multimerización de Proteína , Receptores de Ácido Kaínico/metabolismo , Potenciales de Acción , Animales , Ácido Glutámico/metabolismo , Células HEK293 , Humanos , Unión Proteica , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , Ratas , Receptores de N-Metil-D-Aspartato
15.
J Neurosci ; 32(37): 12928-33, 2012 Sep 12.
Artículo en Inglés | MEDLINE | ID: mdl-22973017

RESUMEN

Kainate receptors can be subject to voltage-dependent block by intracellular polyamines, which causes inward rectification of the current-voltage relationship. Sensitivity to polyamine block is largely determined by the identity of a residue within the pore domain that can be altered through RNA editing. This process causes replacement of the encoded glutamine(Q) with a positively charged arginine(R), eliminating polyamine inhibition and thus inward rectification. In neurons, kainate receptors can associate with the auxiliary subunits Neto1 or Neto2. These transmembrane proteins alter the trafficking, channel kinetics, and pharmacology of the receptors in a subunit-dependent manner. We found that coexpression of Neto subunits with recombinant GluK2(Q) kainate receptors greatly reduced inward rectification without altering calcium permeability. This effect was separate from modulation of channel kinetics, as mutations within the extracellular LDLa domain of the Neto proteins completely eliminated their effects on desensitization but only reduced their effects on rectification. Conversely, deletion of the intracellular C-terminal domain of Neto1 or Neto2 or neutralization of positively charged residues within this domain prevented the reduction in rectification but did not alter effects on channel kinetics. These results demonstrate new roles for Neto1 and Neto2 in regulating kainate receptor function and identify domains within these auxiliary subunits important for mediating their effects.


Asunto(s)
Calcio/metabolismo , Activación del Canal Iónico/fisiología , Potenciales de la Membrana/fisiología , Proteínas de la Membrana/química , Proteínas de la Membrana/metabolismo , Subunidades de Proteína , Receptores de N-Metil-D-Aspartato , Relación Estructura-Actividad
16.
Neuropharmacology ; 62(8): 2507-14, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22445602

RESUMEN

The National Center for Complementary and Alternative Medicine (NCCAM) estimates that nearly 40% of adults in the United States use alternative medicines, often in the form of an herbal supplement. Extracts from the tree bark of magnolia species have been used for centuries in traditional Chinese and Japanese medicines to treat a variety of neurological diseases, including anxiety, depression, and seizures. The active ingredients in the extracts have been identified as the bi-phenolic isomers magnolol and honokiol. These compounds were shown to enhance the activity of GABA(A) receptors, consistent with their biological effects. The GABA(A) receptors exhibit substantial subunit heterogeneity, which influences both their functional and pharmacological properties. We examined the activity of magnolol and honokiol at different populations of both neuronal and recombinant GABA(A) receptors to characterize their mechanism of action and to determine whether sensitivity to modulation was dependent upon the receptor's subunit composition. We found that magnolol and honokiol enhanced both phasic and tonic GABAergic neurotransmission in hippocampal dentate granule neurons. In addition, all recombinant receptors examined were sensitive to modulation, regardless of the identity of the α, ß, or γ subunit subtype, although the compounds showed particularly high efficacy at δ-containing receptors. This direct positive modulation of both synaptic and extra-synaptic populations of GABA(A) receptors suggests that supplements containing magnolol and/or honokiol would be effective anxiolytics, sedatives, and anti-convulsants. However, significant side-effects and risk of drug interactions would also be expected.


Asunto(s)
Compuestos de Bifenilo/farmacología , Medicamentos Herbarios Chinos/farmacología , GABAérgicos/farmacología , Hipocampo/efectos de los fármacos , Lignanos/farmacología , Neuronas/efectos de los fármacos , Receptores de GABA-A/metabolismo , Animales , Células HEK293 , Hipocampo/metabolismo , Humanos , Potenciales Postsinápticos Inhibidores/efectos de los fármacos , Neuronas/metabolismo , Ratas , Transmisión Sináptica/efectos de los fármacos
17.
Neuropsychiatr Dis Treat ; 7: 673-81, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22128252

RESUMEN

Lennox-Gastaut syndrome (LGS) is a devastating childhood epilepsy syndrome characterized by the occurrence of multiple types of seizures and cognitive decline. Most children suffer from frequent seizures that are refractory to current medical management. Recent clinical trials have suggested that addition of clobazam may improve the clinical outcome for some LGS patients. Although clobazam has been available for over five decades, it has only recently been approved by the US Food and Drug Administration for this indication. As a 1,5-benzodiazepine, clobazam is structurally related to the widely used 1,4-benzodiazepines, which include diazepam. Clobazam has been shown to modulate GABAergic neurotransmission by positive allosteric modulation of GABA(A) receptors, and to increase expression of transporters for both GABA and glutamate. The active metabolite n-desmethylclobazam (norclobazam) also modulates GABA(A) receptors, and the relative importance of these two compounds in the clinical effectiveness of clobazam remains an open question. Clinical trials involving clobazam as an addon therapy in a variety of pediatric epilepsy populations have found a significant improvement in seizure control. In patients with LGS, clobazam may have greatest efficacy for drop seizures. Longstanding clinical experience suggests that clobazam is a safe and well tolerated antiepileptic drug with infrequent and mild adverse effects. These results suggest that adjunctive treatment with clobazam may be a reasonable option for LGS patients, particularly those who are treatment-resistant.

18.
J Neurosci ; 31(47): 17113-22, 2011 Nov 23.
Artículo en Inglés | MEDLINE | ID: mdl-22114280

RESUMEN

Kainate receptors (KARs) have been implicated in a number of neurological disorders, including epilepsy. KARs are tetrameric, composed of a combination of GluK1-GluK5 subunits. We examined the contribution of GluK2 and GluK5 subunits to activation and desensitization of the heteromeric receptor. Heteromeric GluK2/K5 receptors expressed in HEK-293T cells showed markedly higher glutamate sensitivity than GluK2 homomers and did not desensitize at low glutamate concentrations. Mutation of residue E738 in GluK2 substantially lowered its glutamate sensitivity. However, heteromeric KARs containing this mutant GluK2 [GluK2(E738D)] assembled with wild-type GluK5 showed no change in glutamate EC(50) compared with wild-type heteromeric KARs. Instead, higher concentrations of glutamate were required to produce desensitization. This suggested that, within the heteromeric receptor, glutamate binding to the high-affinity GluK5 subunit alone was sufficient for channel activation but not desensitization, whereas agonist binding to the low-affinity GluK2 subunit was not necessary to open the channel but instead caused the channel to enter a closed, desensitized state. To test this hypothesis in wild-type receptors, we used the competitive antagonist kynurenate, which has higher affinity for the GluK2 than the GluK5 subunit. Coapplication of kynurenate with glutamate to heteromeric receptors reduced the onset of desensitization without affecting the peak current response, consistent with our hypothesis. Our results suggest that GluK2 and GluK5 subunits can be individually activated within the heteromeric receptor and that these subunits serve dramatically different functional roles.


Asunto(s)
Receptores de Ácido Kaínico/fisiología , Animales , Regulación de la Expresión Génica , Ácido Glutámico/fisiología , Células HEK293 , Humanos , Ácido Quinurénico/farmacología , Multimerización de Proteína/genética , Subunidades de Proteína/biosíntesis , Subunidades de Proteína/genética , Subunidades de Proteína/fisiología , Ratas , Receptores de Ácido Kaínico/biosíntesis , Receptores de Ácido Kaínico/genética , Receptor de Ácido Kaínico GluK2
19.
Neuropharmacology ; 61(4): 747-52, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21640733

RESUMEN

Histamine is an important wake-promoting neurotransmitter that activates seven-transmembrane G-protein coupled histamine receptors. However, histamine demonstrates target promiscuity, including direct interaction with the structurally unrelated glutamate (NMDA) and GABA(A) receptor channels. Previous work showed that histamine enhances the activity of recombinant GABA(A) receptor isoforms typically found in synaptic locations, although co-release of histamine and GABA is not known to occur in vivo. Here we used patch clamp recordings of various recombinant GABA(A) receptor isoforms (α1-6, ß1-3, γ1-3, δ) to test the hypothesis that histamine might show subunit preference under low GABA concentration (extrasynaptic) conditions. We found that histamine potentiated the whole-cell responses to GABA for all tested subunit combinations. However, the magnitude of enhancement was largest (∼400% of EC(10) GABA-evoked currents) with α4ß3 and α4ß3X isoforms, where X could be γ or δ. In contrast, histamine (1 mM) had small effects on prolonging deactivation of α4ß3γ2 receptors following brief (5 ms) pulses of 1 mM GABA. These findings suggest GABA-histamine cross-talk may occur preferentially at low GABA concentrations, which could theoretically be inhibitory (via enhancing tonic inhibition), directly excitatory (via enhancing presynaptic GABAergic signaling), or indirectly excitatory (via inhibiting GABAergic interneurons).


Asunto(s)
Histamina/farmacología , Subunidades de Proteína/agonistas , Subunidades de Proteína/fisiología , Receptores de GABA-A/fisiología , Vigilia/fisiología , Ácido gamma-Aminobutírico/farmacología , Relación Dosis-Respuesta a Droga , Células HEK293 , Humanos , Receptor Cross-Talk/efectos de los fármacos , Receptor Cross-Talk/fisiología
20.
Epilepsia ; 52 Suppl 2: 76-8, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21463286

RESUMEN

The anticonvulsant stiripentol (Diacomittm) has been shown to have a positive impact on control of seizures for many patients with Dravet syndrome. As with most antiepileptic drugs, stiripentol has multiple mechanisms of action. Its direct anticonvulsant activity is likely due to enhancement of inhibitory, γ-aminobutyric acid (GABA)ergic neurotransmission. Stiripentol was shown to increase the activity of both neuronal and recombinant GABA(A) receptors at clinically relevant concentrations. At recombinant receptors, stiripentol was found to act through a unique site in a subunit-dependent manner. Positive modulation by stiripentol was most effective at GABA(A) receptors containing an α3 subunit. The expression of the α3 subunit is developmentally regulated, with highest levels in the immature brain. This subunit selectivity may explain the greater clinical efficacy of stiripentol in childhood-onset epilepsies, including Dravet syndrome.


Asunto(s)
Anticonvulsivantes/farmacología , Dioxolanos/farmacología , Epilepsias Mioclónicas/tratamiento farmacológico , Agonistas de Receptores de GABA-A/farmacología , Receptores de GABA-A/fisiología , Anticonvulsivantes/química , Anticonvulsivantes/uso terapéutico , Dioxolanos/química , Dioxolanos/uso terapéutico , Epilepsias Mioclónicas/fisiopatología , Agonistas de Receptores de GABA-A/química , Agonistas de Receptores de GABA-A/uso terapéutico , Humanos , Síndrome
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...