Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
Pharmacol Res ; 110: 10-24, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27157251

RESUMEN

Efavirenz is highly effective at suppressing HIV-1, and the WHO guidelines list it as a component of the first-line antiretroviral (ARV) therapies for treatment-naïve patients. Though the pharmacological basis is unclear, efavirenz is commonly associated with a risk for neuropsychiatric adverse events (NPAEs) when taken at the prescribed dose. In many patients these NPAEs appear to subside after several weeks of treatment, though long-term studies show that in some patients the NPAEs persist. In a recent study focusing on the abuse potential of efavirenz, its receptor psychopharmacology was reported to include interactions with a number of established molecular targets for known drugs of abuse, and it displayed a prevailing behavioral profile in rodents resembling an LSD-like activity. In this report, we discovered interactions with additional serotonergic targets that may be associated with efavirenz-induced NPAEs. The most robust interactions were with 5-HT3A and 5-HT6 receptors, with more modest interactions noted for the 5-HT2B receptor and monoamine oxidase A. From a molecular mechanistic perspective, efavirenz acts as a 5-HT6 receptor inverse agonist of Gs-signaling, 5-HT2A and 5-HT2C antagonist of Gq-signaling, and a blocker of the 5-HT3A receptor currents. Efavirenz also completely or partially blocks agonist stimulation of the M1 and M3 muscarinic receptors, respectively. Schild analysis suggests that efavirenz competes for the same site on the 5-HT2A receptor as two known hallucinogenic partial agonists (±)-DOI and LSD. Prolonged exposure to efavirenz reduces 5-HT2A receptor density and responsiveness to 5-HT. Other ARVs such as zidovudine, nevirapine and emtricitabine did not share the same complex pharmacological profile as efavirenz, though some of them weakly interact with the 5-HT6 receptor or modestly block GABAA currents.


Asunto(s)
Fármacos Anti-VIH/toxicidad , Benzoxazinas/toxicidad , Encéfalo/efectos de los fármacos , Infecciones por VIH/tratamiento farmacológico , VIH-1/efectos de los fármacos , Receptores de Serotonina/efectos de los fármacos , Inhibidores de la Transcriptasa Inversa/toxicidad , Antagonistas de la Serotonina/toxicidad , Alquinos , Animales , Fármacos Anti-VIH/metabolismo , Benzoxazinas/metabolismo , Unión Competitiva , Encéfalo/metabolismo , Células CHO , Señalización del Calcio/efectos de los fármacos , Cricetulus , Ciclopropanos , Relación Dosis-Respuesta a Droga , Agonismo Parcial de Drogas , Cobayas , Células HEK293 , Infecciones por VIH/diagnóstico , Infecciones por VIH/virología , VIH-1/patogenicidad , Células HeLa , Humanos , Potenciales de la Membrana , Inhibidores de la Monoaminooxidasa/toxicidad , Unión Proteica , Ensayo de Unión Radioligante , Receptores Muscarínicos/efectos de los fármacos , Receptores Muscarínicos/genética , Receptores Muscarínicos/metabolismo , Receptores de Serotonina/genética , Receptores de Serotonina/metabolismo , Inhibidores de la Transcriptasa Inversa/metabolismo , Factores de Tiempo , Transfección
2.
Nat Commun ; 15(1): 5176, 2024 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-38890279

RESUMEN

The longevity of grafts remains a major challenge in allogeneic transplantation due to immune rejection. Systemic immunosuppression can impair graft function and can also cause severe adverse effects. Here, we report a local immuno-protective strategy to enhance post-transplant persistence of allografts using a mesenchymal stem cell membrane-derived vesicle (MMV)-crosslinked hydrogel (MMV-Gel). MMVs are engineered to upregulate expression of Fas ligand (FasL) and programmed death ligand 1 (PD-L1). The MMVs are retained within the hydrogel by crosslinking. The immuno-protective microenvironment of the hydrogel protects allografts by presenting FasL and PD-L1. The binding of these ligands to T effector cells, the dominant contributors to graft destruction and rejection, results in apoptosis of T effector cells and generation of regulatory T cells. We demonstrate that implantation with MMV-Gel prolongs the survival and function of grafts in mouse models of allogeneic pancreatic islet cells and skin transplantation.


Asunto(s)
Proteína Ligando Fas , Hidrogeles , Trasplante de Islotes Pancreáticos , Ratones Endogámicos C57BL , Trasplante de Piel , Linfocitos T Reguladores , Trasplante Homólogo , Animales , Hidrogeles/química , Ratones , Proteína Ligando Fas/metabolismo , Proteína Ligando Fas/inmunología , Linfocitos T Reguladores/inmunología , Trasplante de Islotes Pancreáticos/métodos , Trasplante de Piel/métodos , Antígeno B7-H1/metabolismo , Antígeno B7-H1/inmunología , Células Madre Mesenquimatosas/inmunología , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/efectos de los fármacos , Ratones Endogámicos BALB C , Supervivencia de Injerto/efectos de los fármacos , Supervivencia de Injerto/inmunología , Rechazo de Injerto/prevención & control , Rechazo de Injerto/inmunología , Humanos , Masculino , Apoptosis/efectos de los fármacos
3.
Sci Adv ; 10(11): eadk2444, 2024 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-38478602

RESUMEN

Cancer vaccines show huge potential for cancer prevention and treatment. However, their efficacy remains limited due to weak immunogenicity regarding inefficient stimulation of cytotoxic T lymphocyte (CTL) responses. Inspired by the unique characteristic and biological function of high-density lipoprotein (HDL), we here develop an HDL-mimicking nanovaccine with the commendable lymph-targeted capacity to potently elicit antitumor immunity using lipid nanoparticle that is co-loaded with specific cancer cytomembrane harboring a collection of tumor-associated antigens and an immune adjuvant. The nanoparticulate impact is explored on the efficiency of lymphatic targeting and dendritic cell uptake. The optimized nanovaccine promotes the co-delivery of antigens and adjuvants to lymph nodes and maintains antigen presentation of dendritic cells, resulting in long-term immune surveillance as the elevated frequency of CTLs within lymphoid organs and tumor tissue. Immunization of nanovaccine suppresses tumor formation and growth and augments the therapeutic efficacy of checkpoint inhibitors notably on the high-stemness melanoma in the mouse models.


Asunto(s)
Melanoma , Nanopartículas , Neoplasias , Animales , Ratones , Nanovacunas , Linfocitos T Citotóxicos , Melanoma/patología , Antígenos de Neoplasias , Adyuvantes Inmunológicos/farmacología , Inmunoterapia/métodos , Ratones Endogámicos C57BL
4.
Neurobiol Dis ; 59: 18-25, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23845275

RESUMEN

Vascular dementia ranks as the second leading cause of dementia in the United States. However, its underlying pathophysiological mechanism is not fully understood and no effective treatment is available. The purpose of the current study was to evaluate long-term cognitive deficits induced by transient middle cerebral artery occlusion (tMCAO) in rats and to investigate the underlying mechanism. Sprague-Dawley rats were subjected to tMCAO or sham surgery. Behavior tests for locomotor activity and cognitive function were conducted at 7 or 30days after stroke. Hippocampal long term potentiation (LTP) and involvement of GABAergic neurotransmission were evaluated at 30days after sham surgery or stroke. Immunohistochemistry and Western blot analyses were conducted to determine the effect of tMCAO on cell signaling in the hippocampus. Transient MCAO induced a progressive deficiency in spatial performance. At 30days after stroke, no neuron loss or synaptic marker change in the hippocampus were observed. LTP in both hippocampi was reduced at 30days after stroke. This LTP impairment was prevented by blocking GABAA receptors. In addition, ERK activity was significantly reduced in both hippocampi. In summary, we identified a progressive decline in spatial learning and memory after ischemic stroke that correlates with suppression of hippocampal LTP, elevation of GABAergic neurotransmission, and inhibition of ERK activation. Our results indicate that the attenuation of GABAergic activity or enhancement of ERK/MAPK activation in the hippocampus might be potential therapeutic approaches to prevent or attenuate cognitive impairment after ischemic stroke.


Asunto(s)
Trastornos del Conocimiento/etiología , Regulación de la Expresión Génica/fisiología , Infarto de la Arteria Cerebral Media/complicaciones , Transducción de Señal/fisiología , Animales , Estimulantes del Sistema Nervioso Central/farmacología , Trastornos del Conocimiento/patología , Modelos Animales de Enfermedad , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Potenciales Postsinápticos Excitadores/fisiología , Lateralidad Funcional , Hipocampo/fisiopatología , Técnicas In Vitro , Masculino , Aprendizaje por Laberinto/fisiología , Proteínas de la Membrana/metabolismo , Actividad Motora/fisiología , Proteínas del Tejido Nervioso/metabolismo , Picrotoxina/farmacología , Ratas , Ratas Sprague-Dawley , Factores de Tiempo , Percepción Visual/fisiología
5.
Pharmacology ; 92(1-2): 84-9, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23942137

RESUMEN

SV 293 [1-([5-methoxy-1H-indol-3-yl]methyl)-4-(4-[methylthio]​phenyl)piperidin-4-ol] binds with 100-fold higher affinity to human D2 receptors compared to the human D3 and D4 dopamine receptor subtypes. We investigated the intrinsic efficacy of this compound at the D2 dopamine receptor subtype using both: (1) a forskolin-dependent adenylyl cyclase inhibition assay and (2) an electrophysiological assay for evaluating coupling to G-protein-coupled inwardly rectifying potassium channels. In both assays SV 293 was found to be a neutral antagonist capable of blocking the effects of the full D2-like receptor agonist quinpirole. Based upon these results we propose that SV 293 is a useful pharmacological tool that can be used for both in vitro and in vivo studies to investigate the role of D2-like dopamine receptor subtypes in neurological, neuropsychiatric and movement disorders where dopaminergic pathways have been implicated.


Asunto(s)
Antagonistas de Dopamina/farmacología , Antagonistas de los Receptores de Dopamina D2 , Indoles/farmacología , Piperidinas/farmacología , Inhibidores de Adenilato Ciclasa , Animales , Línea Celular Tumoral , Colforsina/farmacología , Agonistas de Dopamina/farmacología , Canales de Potasio Rectificados Internamente Asociados a la Proteína G/fisiología , Humanos , Ratones , Quinpirol/farmacología , Receptores de Dopamina D2/fisiología
6.
Proc Natl Acad Sci U S A ; 105(39): 15148-53, 2008 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-18815371

RESUMEN

L-type voltage-gated Ca(2+)channels (VGCC) play an important role in dendritic development, neuronal survival, and synaptic plasticity. Recent studies have demonstrated that the gonadal steroid estrogen rapidly induces Ca(2+) influx in hippocampal neurons, which is required for neuroprotection and potentiation of LTP. The mechanism by which estrogen rapidly induces this Ca(2+) influx is not clearly understood. We show by electrophysiological studies that extremely low concentrations of estrogens acutely potentiate VGCC in hippocampal neurons, hippocampal slices, and HEK-293 cells transfected with neuronal L-type VGCC, in a manner that was estrogen receptor (ER)-independent. Equilibrium, competitive, and whole-cell binding assays indicate that estrogen directly interacts with the VGCC. Furthermore, a L-type VGCC antagonist to the dihydropyridine site displaced estrogen binding to neuronal membranes, and the effects of estrogen were markedly attenuated in a mutant, dihydropyridine-insensitive L-type VGCC, demonstrating a direct interaction of estrogens with L-type VGCC. Thus, estrogen-induced potentiation of calcium influx via L-type VGCC may link electrical events with rapid intracellular signaling seen with estrogen exposure leading to modulation of synaptic plasticity, neuroprotection, and memory formation.


Asunto(s)
Canales de Calcio Tipo L/metabolismo , Estrógenos/metabolismo , Neuronas/metabolismo , Animales , Canales de Calcio Tipo L/genética , Línea Celular , Estradiol/metabolismo , Estradiol/farmacología , Estrógenos/farmacología , Humanos , Mutación , Neuronas/efectos de los fármacos , Ratas , Receptores de Estrógenos/genética , Receptores de Estrógenos/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
7.
Behav Brain Res ; 378: 112278, 2020 01 27.
Artículo en Inglés | MEDLINE | ID: mdl-31629836

RESUMEN

Stroke leads to devastating outcomes including impairments of sensorimotor and cognitive function that may be long lasting. New intervention strategies are needed to overcome the long-lasting effects of ischemic injury. Previous studies determined that treatment with 5-methoxyindole-2-carboxylic acid (MICA) conferred chemical preconditioning and neuroprotection against stroke. The purpose of the current study was to determine whether the preconditioning can lead to functional improvements after stroke (done by transient middle cerebral artery occlusion). After 4 weeks of MICA feeding, half the rats underwent ischemic injury, while the other half remained intact. After one week recovery, all the rats were tested for motor and cognitive function (rotorod and water maze). At the time of euthanasia, measurements of long-term potentiation (LTP) were performed. While stroke injury led to motor and cognitive dysfunction, MICA supplementation did not reverse these impairments. However, MICA supplementation did improve stroke-related impairments in hippocampal LTP. The dichotomy of the outcomes suggest that more studies are needed to determine optimum duration and dosage for MICA to lead to substantial motor and cognitive improvements, along with LTP change and neuroprotection.


Asunto(s)
Hipocampo/efectos de los fármacos , Indoles/farmacología , Accidente Cerebrovascular Isquémico/tratamiento farmacológico , Accidente Cerebrovascular Isquémico/prevención & control , Accidente Cerebrovascular Isquémico/fisiopatología , Potenciación a Largo Plazo/efectos de los fármacos , Aprendizaje por Laberinto/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Desempeño Psicomotor/efectos de los fármacos , Recuperación de la Función/efectos de los fármacos , Animales , Conducta Animal/efectos de los fármacos , Suplementos Dietéticos , Dihidrolipoamida Deshidrogenasa/efectos de los fármacos , Modelos Animales de Enfermedad , Indoles/administración & dosificación , Infarto de la Arteria Cerebral Media/complicaciones , Accidente Cerebrovascular Isquémico/etiología , Masculino , Fármacos Neuroprotectores/administración & dosificación , Ratas , Ratas Sprague-Dawley
8.
Artículo en Inglés | MEDLINE | ID: mdl-32194202

RESUMEN

To determine whether (+)-catharanthine induces sedative- or anxiolytic/anxiogenic-like activity in male mice, proper animal paradigms were used. The results showed that (+)-catharanthine induces sedative-like activity in the 63-72 mg/Kg dose range in a flumazenil-insensitive manner, but neither this effect nor anxiolytic/anxiogenic-like activity was observed at lower doses. To determine the underlying molecular mechanism of the sedative-like activity, electrophysiological and radioligand binding experiments were performed with (+)-catharanthine and (±)-18-methoxycoronaridine [(±)-18-MC] on GABAA (GABAARs) and glycine receptors (GlyRs). Coronaridine congeners both activated and potentiated a variety of human (h) GABAARs, except hρ1. (+)-Catharanthine-induced potentiation followed this receptor selectivity (EC50's in µM): hα1ß2 (4.6 ± 0.8) > hα2ß2γ2 (12.6 ± 3.8) ~ hα1ß2γ2 (14.4 ± 4.6) indicating that both α1 and α2 are equally important, whereas γ2 is not necessary. (+)-Catharanthine was >2-fold more potent and efficient than (±)-18-MC at hα1ß2γ2. (+)-Catharanthine also potentiated, whereas (±)-18-MC inhibited, hα1 GlyRs with very low potency. Additional [3H]-flunitrazepam competition binding experiments using rat cerebellum membranes clearly demonstrated that these ligands do not bind to the benzodiazepine site. This is supported by the observed activity at hα1ß2 (lacking the BDZ site) and similar effects between α1- and α2-containing GABAARs. Our study shows, for the first time, that (+)-catharanthine induced sedative-like effects in mice, and coronaridine congeners potentiated human α1ß2γ2, α1ß2, and hα2ß2γ2, but not ρ1, GABAARs, both in a benzodiazepine-insensitive fashion, whereas only (+)-catharanthine slightly potentiated GlyRs.


Asunto(s)
Benzodiazepinas/metabolismo , Hipnóticos y Sedantes/metabolismo , Ibogaína/análogos & derivados , Ibogaína/metabolismo , Receptores de GABA-A/metabolismo , Animales , Benzodiazepinas/farmacología , Relación Dosis-Respuesta a Droga , Agonistas de Receptores de GABA-A/metabolismo , Agonistas de Receptores de GABA-A/farmacología , Células HEK293 , Humanos , Hipnóticos y Sedantes/farmacología , Ibogaína/farmacología , Locomoción/efectos de los fármacos , Locomoción/fisiología , Masculino , Aprendizaje por Laberinto/fisiología , Ratones
9.
Neurosci Lett ; 431(2): 184-9, 2008 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-18162311

RESUMEN

The presence of phenylalanine (F) at the 6' position of transmembrane domain 2 (TM2) in the alpha4 subunit of alpha4beta2 nicotinic receptors enhances desensitization. As the GABA A receptor affords the ability to study the influence of as few as one and as many as five Fs at this position, we have used it to investigate potential subunit- and stoichiometry-dependent effects of the TM2 6'F mutation on desensitization. Whereas the presence of one F at this position decreased extent of desensitization, desensitization was increased in all configurations that included two or more Fs at the TM2 6' position; desensitization was particularly rapid with 3 or 4 F residues present. Our results demonstrate the ability of F residues at the TM2 6' position to modulate desensitization is likely conserved in the cys-loop family of ligand-gated ion channels. Moreover, our findings demonstrate both stoichiometric- and subunit-dependent effects of the ability of this mutation to regulate desensitization in GABA A receptors.


Asunto(s)
Mutación/fisiología , Fenilalanina/genética , Receptores de GABA-A/fisiología , Procesos Estocásticos , Secuencia de Aminoácidos , Animales , Línea Celular Transformada , Estimulación Eléctrica/métodos , Humanos , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/fisiología , Potenciales de la Membrana/efectos de la radiación , Técnicas de Placa-Clamp/métodos , Estructura Terciaria de Proteína/fisiología , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , Ratas , Receptores de GABA-A/genética , Transfección/métodos , Ácido gamma-Aminobutírico/farmacología
10.
Neuropharmacology ; 133: 171-180, 2018 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-29378213

RESUMEN

This study aimed to address the mechanisms and reinforcing effects of three synthetic cathinone analogs of MDMA commonly reported in "Ecstasy" formulations: methylone, butylone, and pentylone. Whole-cell patch clamp techniques were used to assess the mechanism of each compound at the dopamine and serotonin transporters. Separate groups of rats were trained to discriminate methamphetamine, DOM, or MDMA from vehicle. Substitution studies were performed in each group and antagonism studies with SCH23390 were performed against each compound that produced substitution. Self-administration of each compound was evaluated under a progressive ratio schedule of reinforcement. Each compound produced an inward current at the serotonin transporter, but little or no current at the dopamine transporter. Each of the test compounds substituted fully for the discriminative stimulus effects of methamphetamine, methylone and butylone substituted partially for DOM and fully for MDMA, whereas pentylone failed to substitute for DOM and substituted only partially for MDMA. SCH23390 fully and dose-dependently attenuated methamphetamine-appropriate responding produced by each test compound, but was least potent against pentylone. MDMA-appropriate responding was minimally affected by SCH23390. Each test compound was robustly self-administered with pentylone producing the greatest self-administration at the doses tested. Given the prevalence of synthetic cathinones in "Ecstasy" formulations, these data indicate that adulterated "Ecstasy" formulations may drive more compulsive drug use than those containing only MDMA.


Asunto(s)
Alucinógenos/farmacología , N-Metil-3,4-metilenodioxianfetamina/farmacología , Refuerzo en Psicología , Trastornos Relacionados con Sustancias/fisiopatología , Animales , Benzazepinas/farmacología , Estimulantes del Sistema Nervioso Central/farmacología , Condicionamiento Operante/efectos de los fármacos , Modelos Animales de Enfermedad , Antagonistas de Dopamina/farmacología , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/genética , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/metabolismo , Combinación de Medicamentos , Células HEK293 , Humanos , Masculino , Metanfetamina/análogos & derivados , Metanfetamina/farmacología , N-Metil-3,4-metilenodioxianfetamina/análogos & derivados , Técnicas de Placa-Clamp , Ratas , Ratas Sprague-Dawley , Autoadministración , Serotonina/farmacología , Trastornos Relacionados con Sustancias/etiología
11.
Nat Commun ; 9(1): 2082, 2018 05 25.
Artículo en Inglés | MEDLINE | ID: mdl-29802295

RESUMEN

Acid-sensing ion channels (ASICs) evolved to sense changes in extracellular acidity with the divalent cation calcium (Ca2+) as an allosteric modulator and channel blocker. The channel-blocking activity is most apparent in ASIC3, as removing Ca2+ results in channel opening, with the site's location remaining unresolved. Here we show that a ring of rat ASIC3 (rASIC3) glutamates (Glu435), located above the channel gate, modulates proton sensitivity and contributes to the formation of the elusive Ca2+ block site. Mutation of this residue to glycine, the equivalent residue in chicken ASIC1, diminished the rASIC3 Ca2+ block effect. Atomistic molecular dynamic simulations corroborate the involvement of this acidic residue in forming a high-affinity Ca2+ site atop the channel pore. Furthermore, the reported observations provide clarity for past controversies regarding ASIC channel gating. Our findings enhance understanding of ASIC gating mechanisms and provide structural and energetic insights into this unique calcium-binding site.


Asunto(s)
Canales Iónicos Sensibles al Ácido/química , Sitios de Unión/fisiología , Calcio/metabolismo , Activación del Canal Iónico/fisiología , Canales Iónicos Sensibles al Ácido/genética , Canales Iónicos Sensibles al Ácido/metabolismo , Animales , Células CHO , Cationes Bivalentes/metabolismo , Cricetulus , Ácido Glutámico/genética , Ácido Glutámico/metabolismo , Glicina/genética , Glicina/metabolismo , Simulación de Dinámica Molecular , Mutagénesis Sitio-Dirigida , Dominios Proteicos/fisiología , Relación Estructura-Actividad
12.
Neuropharmacology ; 52(8): 1606-15, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17459427

RESUMEN

We previously identified H109 of the glycine alpha1 subunit as a putative proton binding site. In the present studies, we explored additional proton binding site(s) as well as the mechanism underlying modulation of glycine receptors by protons. Whole-cell glycine currents were recorded from HEK 293 cells transiently expressing wild type or mutant glycine receptors. Individual mutation of 3 of 4 remaining extracellular histidine residue into alanine (i.e., alpha1 H107A, H215A or H419A), reduced the receptor sensitivity to protons to a varying extent. In contrast, mutation of alpha1 H201A did not affect proton sensitivity. Double, triple or quadruple histidine mutation of these residues caused a further reduction of proton sensitivity, suggesting multiple binding sites for proton action on glycine receptors. Furthermore, the substitution T133A, which mediates Zn(2+) inhibition, virtually abolished the proton effect on peak amplitude and current kinetics of glycine response. Replacement of T with S on position 133 partially restored receptor sensitivity to protons, suggesting the hydroxyl group of residue T133 is essential for proton-mediated modulation. In heteromeric alpha1beta receptors, mutations beta H132A and S156A, which correspond to H109 and T133 of the alpha1 subunit, respectively, also affected proton inhibition. In conclusion, multiple extracellular histidine residues (H107, H109, H215 and H419) and threonine residues of the alpha1 and beta Zn(2+) coordination sites are critical for modulation of the glycine receptor by protons.


Asunto(s)
Protones , Receptores de Glicina/química , Línea Celular Transformada , Clonación Molecular , Relación Dosis-Respuesta en la Radiación , Estimulación Eléctrica/métodos , Glicina/farmacología , Histidina/metabolismo , Humanos , Concentración de Iones de Hidrógeno , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/fisiología , Potenciales de la Membrana/efectos de la radiación , Mutagénesis/fisiología , Técnicas de Placa-Clamp , Receptores de Glicina/genética , Transfección , Zinc/farmacología
13.
Neuropharmacology ; 119: 100-110, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28390894

RESUMEN

Methylene blue (MB) is commonly used in diagnostic procedures and is also used to treat various medical conditions. Neurological effects of MB have been reported in clinical observations and experimental studies. Thus the modulation of GABAA receptor function by MB was investigated. Whole-cell GABA-activated currents were recorded from HEK293 cells expressing various GABAA receptor subunit configurations. MB inhibition of GABA currents was apparent at 3 µM, and it had an IC50 of 31 µM in human α1ß2γ2 receptors. The MB action was rapid and reversible. MB inhibition was not mediated via the picrotoxin site, as a mutation (T6'F of the ß2 subunit) known to confer resistance to picrotoxin had no effect on MB-induced inhibition. Blockade of GABAA receptors by MB was demonstrated across a range of receptors expressing varying subunits, including those expressed at extrasynaptic sites. The sensitivity of α1ß2 receptors to MB was similar to that observed in α1ß2γ2 receptors, indicating that MB's action via the benzodiazepine or Zn2+ site is unlikely. MB-induced inhibition of GABA response was competitive with respect to GABA. Furthermore, mutation of α1 F64 to A and ß2 Y205 to F in the extracellular N-terminus, both residues which are known to comprise GABA binding pocket, remarkably diminished MB inhibition of GABA currents. These data suggest that MB inhibits GABAA receptor function by direct or allosteric interaction with the GABA binding site. Finally, in mouse hippocampal CA1 pyramidal neurons, MB inhibited GABA-activated currents as well as GABAergic IPSCs. We demonstrate that MB directly inhibits GABAA receptor function, which may underlie some of the effects of MB on the CNS.


Asunto(s)
Sitios de Unión/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Azul de Metileno/farmacología , Neuronas/efectos de los fármacos , Receptores de GABA-A/metabolismo , Animales , Animales Recién Nacidos , Sitios de Unión/genética , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/fisiología , Células HEK293 , Hipocampo/citología , Humanos , Técnicas In Vitro , Potenciales Postsinápticos Inhibidores/efectos de los fármacos , Potenciales Postsinápticos Inhibidores/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Neuronas/metabolismo , Unión Proteica/efectos de los fármacos , Ratas , Receptores de GABA-A/genética , Bloqueadores de los Canales de Sodio/farmacología , Tetrodotoxina/farmacología , Ácido gamma-Aminobutírico/metabolismo , Ácido gamma-Aminobutírico/farmacología
14.
Neuropharmacology ; 121: 167-178, 2017 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-28456686

RESUMEN

Efavirenz is a widely prescribed medicine used to treat type 1 human immunodeficiency virus (HIV-1), the most prevalent pathogenic strain of the virus responsible for the acquired immune deficiency syndrome (AIDS) pandemic. Under prescribed dosing conditions, either alone or in combination therapy, efavirenz-induced CNS disturbances are frequently reported. Efavirenz was recently reported to interact in a similar concentration range with a number of receptors, transporters and ion channels including recombinant rat α1ß2γ2 GABAA receptors whose actions were potentiated (Gatch et al., 2013; Dalwadi et al., 2016). Now we report on the molecular mechanism of efavirenz on GABAA receptors as a function of concentration and subunit composition via whole-cell recordings of GABA-activated currents from HEK293 cells expressing varying subunit configurations of GABAA receptors. Efavirenz elicited dual effects on the GABA response; it allosterically potentiated currents at low concentrations, whereas it inhibited currents at higher concentrations. The allosteric potentiating action on GABAA receptors was pronounced in the α1ß2γ2, α2ß2γ2 and α4ß2γ2 configurations, greatly diminished in the α6ß2γ2 configuration, and completely absent in the α3ß2γ2 or α5ß2γ2 configuration. In stark contrast, the inhibitory modulation of efavirenz at higher concentrations was evident in all subunit configurations examined. Moreover, efavirenz-induced modulatory effects were dependent on GABA concentration ([GABA]), with a pronounced impact on currents activated by low [GABA] but little effect at saturating [GABA]. Mutation of a highly-conserved threonine to phenylalanine in transmembrane domain 2 of the α1 subunit abolished the inhibitory effect of efavirenz in α1ß2 receptors. Finally, mutations of any of the three conserved extracellular residues in α1/2/4 subunits to the conserved residues at the corresponding positions in α3/5 subunits (i.e., R84P, M89L or I120L) completely eliminated the potentiating effect of efavirenz in α1ß2γ2 configuration. These findings demonstrate that efavirenz's positive allosteric modulation of the GABAA receptor is mediated via a novel allosteric site associated with the extracellular domain of the receptor.


Asunto(s)
Benzoxazinas/farmacología , Receptores de GABA-A/metabolismo , Inhibidores de la Transcriptasa Inversa/farmacología , Alquinos , Regulación Alostérica , Animales , Ciclopropanos , Diazepam/farmacología , Relación Dosis-Respuesta a Droga , Flumazenil/farmacología , Moduladores del GABA/farmacología , Células HEK293 , Humanos , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/genética , Mutagénesis/genética , Técnicas de Placa-Clamp , Dominios Proteicos/genética , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , Ratas , Receptores de GABA-A/genética , Transfección , Ácido gamma-Aminobutírico/farmacología
15.
Neuropharmacology ; 51(4): 701-8, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16842826

RESUMEN

Endogenous divalent cations Cu2+ and Zn2+ suppress the activity of glycine receptors (glyRs). Whereas residues critical for the effects of Zn2+ on glyRs have been identified, little is known about the determinants of Cu2+-mediated inhibition. In the present studies, we have assessed the potential commonality of Zn2+ and Cu2+-mediated inhibition of glyRs. Cu2+ potently inhibited recombinant human glycine alpha1 receptors, with an IC50 of 4.1+/-0.7 microM. Systematic mutation of extracellular histidine residues revealed that mutation H215A greatly reduced the inhibitory modulation by Cu2+. Substitution of H215 with C produced receptors with Cu2+ sensitivity similar to the wild type. Furthermore, modification of H215C with a thio-specific reagent, [2-(trimethylammonium)ethyl] methanethiosulfonate bromide (MTSET), reduced Cu2+ sensitivity of H215C receptors. However, mutation of other extracellular histidine residues including H107 and H109, which are known inhibitory Zn2+coordination sites, failed to influence inhibition of glycine currents by Cu2+. Moreover, mutation to alanine of two threonine residues (T112, T133) critical for Zn2+ inhibition had no effect (T133A) or only partial inhibitory effects (T112A) on Cu2+-induced inhibition. The double mutation, T112A/H215A, caused greater effects on Cu2+-mediated inhibition than either mutation alone. In addition, the glycine currents recorded from T112A/H215A mutant receptors were significantly potentiated by low concentrations of Cu2+. Our results have identified critical determinants of Cu2+-mediated inhibition of glyRs. Moreover, we demonstrate for the first time a clear difference in residues responsible for Cu2+-mediated compared to Zn2+-mediated inhibition of glyRs.


Asunto(s)
Cobre/metabolismo , Receptores de Glicina/química , Receptores de Glicina/fisiología , Línea Celular Transformada , Clonación Molecular/métodos , Cobre/farmacología , Relación Dosis-Respuesta a Droga , Interacciones Farmacológicas , Glicina/farmacología , Histidina/genética , Histidina/metabolismo , Humanos , Concentración 50 Inhibidora , Potenciales de la Membrana/efectos de los fármacos , Mesilatos/farmacología , Mutagénesis/fisiología , Técnicas de Placa-Clamp/métodos , Reactivos de Sulfhidrilo/farmacología , Transfección/métodos , Zinc/farmacología
16.
Brain Res ; 1086(1): 1-8, 2006 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-16626662

RESUMEN

Amphibian oocyte and mammalian heterologous expression systems are often used to investigate the function of recombinant ion channels using electrophysiological techniques. Although both systems have yielded important information, the results obtained in these systems are sometimes conflicting. Oocytes and mammalian cells differ in their physiological temperature requirements. While room temperature is within the physiological temperature range for oocytes, this temperature is far below that required by mammalian cells. Since electrophysiological studies are often performed in both oocytes and mammalian cells at room temperature, we sought to determine if recording temperature could be a factor in some disparate results obtained in these cell types. For these studies, we examined phorbol ester modulation of GABA(A) and glycine receptors. Consistent with the literature, at room temperature, PMA (phorbol 12-myristate 13-acetate) produced a large reproducible decrease in the peak amplitude of GABA and glycine-gated currents in Xenopus oocytes. In contrast, PMA was ineffective in modulating these heterologously expressed receptors at room temperature in human embryonic kidney (HEK) 293 cells. However, when electrophysiological experiments were performed at 35 degrees C in HEK 293 cells, PMA decreased the function of these receptors. Our results indicate that the temperature at which electrophysiological studies are conducted is an important experimental variable. To determine the extent to which electrophysiological recordings are performed at physiological temperatures in HEK 293 cells, a PubMed search was conducted using the search terms "patch clamp" and "HEK" for the years 2003-2004. This search revealed that only 15% of the patch clamp studies were reported to have been conducted in the temperature range of 32-37 degrees C. The results of our study indicate that temperature is an important experimental variable that requires rational consideration in the design of electrophysiological experiments.


Asunto(s)
Activación del Canal Iónico/fisiología , Potenciales de la Membrana/fisiología , Proteína Quinasa C/metabolismo , Temperatura , Análisis de Varianza , Animales , Línea Celular , Interacciones Farmacológicas , Estimulación Eléctrica/métodos , Activación Enzimática/efectos de los fármacos , Femenino , Glicina/farmacología , Humanos , Activación del Canal Iónico/efectos de los fármacos , Activación del Canal Iónico/efectos de la radiación , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/efectos de la radiación , Oocitos , Técnicas de Placa-Clamp/métodos , Ésteres del Forbol/farmacología , Receptores de GABA/fisiología , Receptores de Serotonina/fisiología , Serotonina/farmacología , Transfección/métodos , Xenopus , Ácido gamma-Aminobutírico/farmacología
17.
Endocrinology ; 157(5): 2067-79, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-26990062

RESUMEN

Progesterone (P4) exerts robust cytoprotection in brain slice cultures (containing both neurons and glia), yet such protection is not as evident in neuron-enriched cultures, suggesting that glia may play an indispensable role in P4's neuroprotection. We previously reported that a membrane-associated P4 receptor, P4 receptor membrane component 1, mediates P4-induced brain-derived neurotrophic factor (BDNF) release from glia. Here, we sought to determine whether glia are required for P4's neuroprotection and whether glia's roles are mediated, at least partially, via releasing soluble factors to act on neighboring neurons. Our data demonstrate that P4 increased the level of mature BDNF (neuroprotective) while decreasing pro-BDNF (potentially neurotoxic) in the conditioned media (CMs) of cultured C6 astrocytes. We examined the effects of CMs derived from P4-treated astrocytes (P4-CMs) on 2 neuronal models: 1) all-trans retinoid acid-differentiated SH-SY5Y cells and 2) mouse primary hippocampal neurons. P4-CM increased synaptic marker expression and promoted neuronal survival against H2O2. These effects were attenuated by Y1036 (an inhibitor of neurotrophin receptor [tropomysin-related kinase] signaling), as well as tropomysin-related kinase B-IgG (a more specific inhibitor to block BDNF signaling), which pointed to BDNF as the key protective component within P4-CM. These findings suggest that P4 may exert its maximal protection by triggering a glia-neuron cross talk, in which P4 promotes mature BDNF release from glia to enhance synaptogenesis as well as survival of neurons. This recognition of the importance of glia in mediating P4's neuroprotection may also inform the design of effective therapeutic methods for treating diseases wherein neuronal death and/or synaptic deficits are noted.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/metabolismo , Proteínas de la Membrana/metabolismo , Neuroglía/metabolismo , Neuronas/metabolismo , Progesterona/farmacología , Receptores de Progesterona/metabolismo , Transducción de Señal/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Astrocitos/citología , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Hipocampo/citología , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Humanos , Ratones , Neuroglía/citología , Neuroglía/efectos de los fármacos , Neuronas/citología , Neuronas/efectos de los fármacos , Propionatos/farmacología , Tiazolidinas/farmacología
18.
Neuropharmacology ; 48(2): 181-94, 2005 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15695157

RESUMEN

The modulation of GABAA receptors by protein kinase C is complex and involves effects on both ion channel function and receptor trafficking. Although PKC regulates receptor cell surface expression the mechanism is not well understood. Using immunofluorescence studies in HEK 293 cells, we demonstrate that activation of PKC by the phorbol ester PMA promotes receptor endocytosis and is dependent on the presence of a gamma subunit. This endocytosis is blocked by the dominant negative dynamin mutant K44A indicating that PKC-induced receptor endocytosis involves the dynamin endocytic pathway. Mutation of a dileucine motif within the receptor beta2 subunit inhibits the effect of PKC activation on receptor endocytosis. Using patch clamp analysis, we show that PKC activation produces a robust inhibition of GABA-gated chloride currents in cells expressing wildtype GABAA receptors, but it is ineffective in modulating receptors lacking the dileucine motif. Furthermore, the introduction into the patch pipette of a 10-amino acid peptide corresponding to the dileucine motif present in the receptor beta2 subunit prevents PKC modulation of wildtype recombinant receptors. Furthermore, in cerebral cortical neuronal slices inclusion of this peptide in the patch pipette prevents PKC modulation of native GABAA receptors. Using limited chymotrypsin digestion assays, we also show that PKC increases receptor internalization in primary cultures of cerebral cortical neurons. Lastly, PKC inhibitors do not block constitutive receptor endocytosis or affect GABA-gated chloride currents suggesting that PKC-dependent phosphorylation is not required for GABAA receptor endocytosis but plays a modulatory role in the process.


Asunto(s)
Endocitosis/fisiología , Leucina/metabolismo , Mutación , Proteína Quinasa C/metabolismo , Receptores de GABA-A/metabolismo , Animales , Línea Celular , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/metabolismo , Dipéptidos/genética , Dipéptidos/metabolismo , Dipéptidos/farmacología , Endocitosis/efectos de los fármacos , Femenino , Humanos , Técnicas In Vitro , Leucina/genética , Leucina/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Embarazo , Ratas , Ratas Sprague-Dawley , Receptores de GABA-A/genética
19.
Br J Pharmacol ; 172(10): 2519-31, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25573298

RESUMEN

BACKGROUND AND PURPOSE: Cognitive deficits in patients with Alzheimer's disease, Parkinson's disease, traumatic brain injury and stroke often involve alterations in cholinergic signalling. Currently available therapeutic drugs provide only symptomatic relief. Therefore, novel therapeutic strategies are needed to retard and/or arrest the progressive loss of memory. EXPERIMENTAL APPROACH: Scopolamine-induced memory impairment provides a rapid and reversible phenotypic screening paradigm for cognition enhancement drug discovery. Male C57BL/6J mice given scopolamine (1 mg·kg(-1) ) were used to evaluate the ability of LS-1-137, a novel sigma (σ1) receptor-selective agonist, to improve the cognitive deficits associated with muscarinic antagonist administration. KEY RESULTS: LS-1-137 is a high-affinity (Ki = 3.2 nM) σ1 receptor agonist that is 80-fold selective for σ1, compared with σ2 receptors. LS-1-137 binds with low affinity at D2-like (D2, D3 and D4) dopamine and muscarinic receptors. LS-1-137 was found to partially reverse the learning deficits associated with scopolamine administration using a water maze test and an active avoidance task. LS-1-137 treatment was also found to trigger the release of brain-derived neurotrophic factor from rat astrocytes. CONCLUSIONS AND IMPLICATIONS: The σ1 receptor-selective compound LS-1-137 may represent a novel candidate cognitive enhancer for the treatment of muscarinic receptor-dependent cognitive deficits.


Asunto(s)
Acetanilidas/farmacología , Acetanilidas/uso terapéutico , Trastornos del Conocimiento/inducido químicamente , Trastornos del Conocimiento/tratamiento farmacológico , Antagonistas Muscarínicos/farmacología , Piperidinas/farmacología , Piperidinas/uso terapéutico , Receptores sigma/agonistas , Receptores sigma/metabolismo , Animales , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Reacción de Prevención/efectos de los fármacos , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Ligandos , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Ratones , Ratas , Escopolamina/farmacología , Receptor Sigma-1
20.
Neuropsychopharmacology ; 38(12): 2373-84, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23702798

RESUMEN

Anecdotal reports have surfaced concerning misuse of the HIV antiretroviral medication efavirenz ((4S)-6-chloro-4-(2-cyclopropylethynyl)-4-(trifluoromethyl)-2,4-dihydro-1H-3,1-benzoxazin-2-one) by HIV patients and non-infected teens who crush the pills and smoke the powder for its psychoactive effects. Molecular profiling of the receptor pharmacology of efavirenz pinpointed interactions with multiple established sites of action for other known drugs of abuse including catecholamine and indolamine transporters, and GABAA and 5-HT(2A) receptors. In rodents, interaction with the 5-HT(2A) receptor, a primary site of action of lysergic acid diethylamine (LSD), appears to dominate efavirenz's behavioral profile. Both LSD and efavirenz reduce ambulation in a novel open-field environment. Efavirenz occasions drug-lever responding in rats discriminating LSD from saline, and this effect is abolished by selective blockade of the 5-HT(2A) receptor. Similar to LSD, efavirenz induces head-twitch responses in wild-type, but not in 5-HT(2A)-knockout, mice. Despite having GABAA-potentiating effects (like benzodiazepines and barbiturates), and interactions with dopamine transporter, serotonin transporter, and vesicular monoamine transporter 2 (like cocaine and methamphetamine), efavirenz fails to maintain responding in rats that self-administer cocaine, and it fails to produce a conditioned place preference. Although its molecular pharmacology is multifarious, efavirenz's prevailing behavioral effect in rodents is consistent with LSD-like activity mediated via the 5-HT(2A) receptor. This finding correlates, in part, with the subjective experiences in humans who abuse efavirenz and with specific dose-dependent adverse neuropsychiatric events, such as hallucinations and night terrors, reported by HIV patients taking it as a medication.


Asunto(s)
Fármacos Anti-VIH/toxicidad , Benzoxazinas/toxicidad , Alucinógenos/toxicidad , Dietilamida del Ácido Lisérgico/toxicidad , Receptor de Serotonina 5-HT2A/metabolismo , Agonistas del Receptor de Serotonina 5-HT2/metabolismo , Alquinos , Animales , Conducta Animal/efectos de los fármacos , Condicionamiento Psicológico/efectos de los fármacos , Ciclopropanos , Discriminación en Psicología , Masculino , Ratones , Ratas , Ratas Sprague-Dawley , Receptores de Superficie Celular/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA