Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Nat Chem Biol ; 9(10): 630-5, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23974117

RESUMEN

Melanopsin, expressed in a subset of retinal ganglion cells, mediates behavioral adaptation to ambient light and other non-image-forming photic responses. This has raised the possibility that pharmacological manipulation of melanopsin can modulate several central nervous system responses, including photophobia, sleep, circadian rhythms and neuroendocrine function. Here we describe the identification of a potent synthetic melanopsin antagonist with in vivo activity. New sulfonamide compounds inhibiting melanopsin (opsinamides) compete with retinal binding to melanopsin and inhibit its function without affecting rod- and cone-mediated responses. In vivo administration of opsinamides to mice specifically and reversibly modified melanopsin-dependent light responses, including the pupillary light reflex and light aversion. The discovery of opsinamides raises the prospect of therapeutic control of the melanopsin phototransduction system to regulate light-dependent behavior and remediate pathological conditions.


Asunto(s)
Fototransducción/efectos de los fármacos , Opsinas de Bastones/antagonistas & inhibidores , Bibliotecas de Moléculas Pequeñas/farmacología , Sulfonamidas/farmacología , Humanos , Estructura Molecular , Opsinas de Bastones/metabolismo , Bibliotecas de Moléculas Pequeñas/síntesis química , Bibliotecas de Moléculas Pequeñas/química , Relación Estructura-Actividad , Sulfonamidas/síntesis química , Sulfonamidas/química
2.
J Pharmacol Exp Ther ; 328(3): 900-11, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19098165

RESUMEN

Neuropeptide Y (NPY) regulates physiological processes via receptor subtypes (Y(1), Y(2), Y(4), Y(5), and y(6)). The Y(5) receptor is well known for its role in appetite. Based on expression in the limbic system, we hypothesized that the Y(5) receptor might also modulate stress sensitivity. We identified a novel Y(5) receptor-selective antagonist, Lu AA33810 [N-[[trans-4-[(4,5-dihydro[1]-benzothiepino[5,4-d]thiazol-2-yl)amino]cyclohexyl]methyl]-methanesulfonamide], that bound to cloned rat Y(5) receptors (K(i) = 1.5 nM) and antagonized NPY-evoked cAMP and calcium mobilization in vitro. Lu AA33810 (3-30 mg/kg p.o.) blocked feeding elicited by intracerebroventricular injection of the Y(5) receptor-selective agonist [cPP(1-7),NPY(19-23),Ala(31),Aib(32),Gln(34)]-hPancreatic Polypeptide in Sprague-Dawley rats. In vivo effects of Lu AA33810 were correlated with brain exposure > or = 50 ng/g and ex vivo Y(5) receptor occupancy of 22 to 95%. Lu AA33810 was subsequently evaluated in models of stress sensitivity. In Fischer 344 rats, Lu AA33810 (30 mg/kg p.o.) attenuated increases in plasma ACTH and corticosterone elicited by intracerebroventricular injection of [cPP(1-7),NPY(19-23),Ala(31),Aib(32),Gln(34)]-hPancreatic Polypeptide. In Sprague-Dawley rats subjected to the social interaction test, Lu AA33810 (3-30 mg/kg p.o.) produced anxiolytic-like effects after acute or chronic treatment. In Flinders sensitive line rats, chronic dosing of Lu AA33810 (10 mg/kg/day i.p.) produced anxiolytic-like effects in the social interaction test, plus antidepressant-like effects in the forced swim test. In Wistar rats exposed to chronic mild stress, chronic dosing of Lu AA33810 (3 and 10 mg/kg/day i.p.) produced antidepressant-like activity, i.e., normalization of stress-induced decrease in sucrose consumption. We propose that Y(5) receptors may function as part of an endogenous stress-sensing system to mediate social anxiety and reward or motivational deficits in selected rodent models.


Asunto(s)
Ansiolíticos/uso terapéutico , Antidepresivos/uso terapéutico , Benzotiepinas/uso terapéutico , Receptores de Neuropéptido Y/antagonistas & inhibidores , Estrés Psicológico/tratamiento farmacológico , Sulfonamidas/uso terapéutico , Tiazoles/uso terapéutico , Animales , Modelos Animales de Enfermedad , Masculino , Modelos Moleculares , Ratas , Ratas Endogámicas F344 , Ratas Wistar
3.
Circ Res ; 98(5): 659-66, 2006 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-16456099

RESUMEN

Cardiac automaticity is controlled by G protein-coupled receptors, such as adrenergic, muscarinic, and adenosine receptors. The strength and duration of G protein signaling is attenuated by regulator of G protein signaling (RGS) proteins acting as GTPase-activating proteins for Galpha subunits; however, little is known about the role of endogenous RGS proteins in cardiac function. We created point mutations in Galpha subunits that disrupt Galpha-RGS binding and introduced them into embryonic stem (ES) cells by homologous recombination. Spontaneously contacting cardiocytes derived from the ES cells were used to evaluate the role of endogenous RGS proteins in chronotropic regulation. The RGS-insensitive GalphaoG184S homozygous knock-in (GalphaoGS/GS) cells demonstrated enhanced adenosine A1 and muscarinic M2 receptor-mediated bradycardic responses. In contrast, Galphai2GS/GS cells showed enhanced responses to M2 but not A1 receptors. Similarly M2 but not A1 bradycardic responses were dramatically enhanced in Galphai2GS/GS mice. Blocking G protein-coupled inward rectifying K+ (GIRK) channels largely abolished the mutation-induced enhancement of the M2 receptor-mediated response but had a minimal effect on A1 responses. The Galphas-dependent stimulation of beating rate by the beta2 adrenergic receptor agonist procaterol was significantly attenuated in GalphaoGS/GS and nearly abolished in Galphai2GS/GS cells because of enhanced signaling via a pertussis toxin sensitive mechanism. Thus, endogenous RGS proteins potently reduce the actions of Galpha(i/o)-linked receptors on cardiac automaticity. Furthermore, M2 and A1 receptors differentially use Galphai2 and Galphao and associated downstream effectors. Thus, alterations in RGS function may play a role in pathophysiological processes and RGS proteins could represent novel cardiovascular therapeutic targets.


Asunto(s)
Adenosina/farmacología , Frecuencia Cardíaca/efectos de los fármacos , Agonistas Muscarínicos/farmacología , Proteínas RGS/fisiología , Animales , Canales de Potasio Rectificados Internamente Asociados a la Proteína G/fisiología , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/fisiología , Ratones , Miocitos Cardíacos/fisiología , Receptor Muscarínico M2/fisiología , Receptores Adrenérgicos alfa 1/fisiología , Receptores Adrenérgicos beta 2/fisiología
4.
J Hazard Mater ; 333: 265-274, 2017 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-28364689

RESUMEN

Natural products are becoming increasingly popular in multiple fields involving medicines, foods and beverages. However, due to the frequent occurrence of poisoning incidents, their toxicity and safety have caused a serious concern. Here we report a method of biosensor-based two-phase pharmacological profiling (BTPP) for discovery, monitor and control of receptor-targeted natural products. BTPP uses a resonant waveguide grating biosensor for label-free and non-invasive detection of intracellular dynamic mass redistribution (DMR), a phenomenon caused by protein relocalization after receptors receiving stimulation from toxicants. The method can not only facilitate the identification of hazardous materials but also quantify their bioactivity by EC50. As a proof of concept, the method was successfully applied to recognize Daturae Flos (DF), an herb that can antagonize muscarinic acetylcholine M2 receptor and further cause poisoning, from other easily confused species. BTPP combined with high performance liquid chromatography revealed that scopolamine and hyoscyamine in DF were the key marker compounds. Moreover, the method accurately picked out 2 M2 receptor antagonists from 25 natural compounds, displaying its potential in high-throughput screening. This study provides a systematic illustration about the establishment, applicability and advantages of BTPP, which contributes to the safety assessment of natural products in related fields.


Asunto(s)
Productos Biológicos/química , Cromatografía Líquida de Alta Presión/métodos , Datura/química , Sustancias Peligrosas/química , Animales , Productos Biológicos/toxicidad , Técnicas Biosensibles , Células CHO , Línea Celular , Cricetulus , Sustancias Peligrosas/toxicidad , Humanos , Hiosciamina/análisis , Hiosciamina/toxicidad , Simulación del Acoplamiento Molecular , Prueba de Estudio Conceptual , Receptor Muscarínico M2/antagonistas & inhibidores , Escopolamina/análisis , Escopolamina/toxicidad
5.
J Biomol Screen ; 10(2): 127-36, 2005 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15799956

RESUMEN

G-protein-coupled receptors (GPCRs) activate heterotrimeric G-proteins (G(i)-, G(s)-, G(q)-, or G(12)-like) to generate specific intracellular responses, depending on the receptor/G-protein coupling. The aim was to enable a majority of GPCRs to generate a predetermined output by signaling through a single G-protein-supported pathway. The authors focused on calcium responses as the output, then engineered Galpha(q) to promote promiscuous receptor interactions. Starting with a human Galpha(q) containing 5 Galpha(z) residues in the C-terminal receptor recognition domain (hGalpha(q/z5)), they evaluated agonist-stimulated calcium responses for 33 diverse GPCRs (G(i)-, G(s)-, and G(q)-coupled) and found 20 of 33 responders. In parallel, they tested Caenorhabditis elegans Galpha(q) containing 5 or 9 C-terminal Galpha(z) residues (cGalpha(q/z5), cGalpha(q/z9)). Signal detection was enhanced with cGalpha(q/z5) and cGalpha(q/z9) (yielding 25/33 and 26/33 responders, respectively). In a separate study of Galpha(s)-coupled receptors, the authors compared hGalpha(q/s5) versus hGalpha(q/s9), cGalpha(q/s9), andcGalphaq/s21 and observed optimal function with cGalpha(q/s9). Cotransfection of an engineered Galpha(q) "cocktail" (cGalpha(q/z5) plus cGalpha(q/s9)) provided a powerful and efficient screening platform. When the chimeras included N-terminal myristoylation sites (to promote membrane localization), calcium responses were sustained or improved, depending on the receptor. This approach toward a "universal functional assay" is particularly useful for orphan GPCRs whose signaling pathways are unknown.


Asunto(s)
Subunidades alfa de la Proteína de Unión al GTP Gq-G11/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Transducción de Señal , Secuencia de Aminoácidos , Animales , Células COS , Caenorhabditis elegans , Agonistas de los Canales de Cloruro , Canales de Cloruro/metabolismo , Chlorocebus aethiops , Electrofisiología , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/química , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/genética , Humanos , Datos de Secuencia Molecular , Oocitos/metabolismo , Receptores Acoplados a Proteínas G/genética , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Alineación de Secuencia , Transfección , Xenopus laevis
6.
Methods Enzymol ; 389: 229-43, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-15313569

RESUMEN

Regulator of G-protein signaling (RGS) proteins are very active GTPase-accelerating proteins (GAPs) in vitro and are expected to reduce signaling by G-protein coupled receptors in vivo. A novel method is presented to assess the in vivo role of RGS proteins in the function of a G protein in which Galpha subunits do not bind to RGS proteins or respond with enhanced GTPase activity. A point mutation in the switch I region of Galpha subunits (G184S Galpha(o) and G183S Galpha(i1)) blocks the interaction with RGS proteins but leaves intact the ability of Galpha to couple to betagamma subunits, receptors, and downstream effectors. Expression of the RGS-insensitive mutant G184S Galpha(o) in C6 glioma cells with the micro-opioid receptor dramatically enhances adenylylcyclase inhibition and activation of extracellular regulated kinase. Introducing the same G184S Galpha(o) protein into embryonic stem (ES) cells by gene targeting allows us to assess the functional importance of the endogenous RGS proteins using in vitro differentiation models and in intact mice. Using ES cell-derived cardiocytes, spontaneous and isoproterenol-stimulated beating rates were not different between wild-type and G184S Galpha(o) mutant cells; however, the bradycardiac response to adenosine A1 receptor agonists was enhanced significantly (seven-fold decrease EC50) in Galpha(o)RGSi mutant cells compared to wild-type Galpha(o), indicating a significant role of endogenous RGS proteins in cardiac automaticity regulation. The approach of using RGS-insensitive Galpha subunit knockins will reveal the role of RGS protein-mediated GAP activity in signaling by a given G(i/o) protein. This will reveal the full extent of RGS regulation and will not be confounded by redundancy in the function of multiple RGS proteins.


Asunto(s)
Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo , Proteínas de Unión al GTP/genética , Mutación Puntual , Proteínas RGS/metabolismo , Transducción de Señal , Animales , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/genética , Proteínas Activadoras de GTPasa/metabolismo , Humanos , Proteínas RGS/genética
7.
Methods Enzymol ; 389: 266-77, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-15313571

RESUMEN

One of the principal roles of the multifunctional regulator of G-protein signaling (RGS) proteins is to terminate G-protein-coupled receptor (GPCR) signaling by binding to the G-protein Galpha subunit, thus acting as GTPase-activating proteins (GAPs). In principle, then, selective inhibitors of this GAP function would have potential as therapeutic agents, as they could be used to augment the effects of endogenous or exogenous GPCR agonists. Using the published RGS4-G(ialpha1) X-ray structure, we have designed and synthesized a series of cyclic peptides, modeled on the G(ialpha) switch I region, that inhibit RGS4 GAP activity, presumably by blocking the interaction between RGS4 and G(ialpha1). These compounds should prove useful for elucidating RGS-mediated activity and serve as a starting point for the development of a novel class of therapeutic agent.


Asunto(s)
Péptidos Cíclicos/química , Péptidos Cíclicos/farmacología , Proteínas RGS/antagonistas & inhibidores , Secuencia de Aminoácidos , Cristalografía por Rayos X , Cisteína/química , Proteínas Activadoras de GTPasa/metabolismo , Concentración 50 Inhibidora , Modelos Moleculares , Péptidos Cíclicos/síntesis química , Proteínas RGS/metabolismo , Relación Estructura-Actividad
8.
Biochem Pharmacol ; 83(4): 435-42, 2012 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-21983034

RESUMEN

The serotonin transporter (SERT) functions to transport serotonin (5-HT) from the extracellular space into neurons to maintain homeostatic control of 5-HT. It is the molecular target for selective serotonin reuptake inhibitor (SSRI) antidepressants. Preclinical research has shown that some SERT inhibitors can bind to two distinct binding sites on the SERT, a primary high affinity binding site and a low affinity allosteric binding site. Mutational studies of the SERT and computational modeling methods with escitalopram resulted in the identification of key amino acid residues important for the function of the allosteric binding site. While this allosteric binding site appears to influence the clinical efficacy of escitalopram under physiological conditions, the molecular mechanism of this effect is still poorly understood and may involve a large network of protein-protein interactions with the SERT. Dynamic interfaces between the SERT and the SERT interacting proteins (SIPs) potentially influence not only the SERT on its uptake function, its regulation, and trafficking, but also on known as well as yet to be identified non-canonical signaling pathways through SIPs. In this commentary, we outline approaches in the areas of selective small-molecule allosteric compound discovery, biochemistry, in vivo genetic knock-in mouse models, as well as computational and structural biology. These studies of the intra-molecular allosteric modulation of the SERT in the context of the myriad of potential inter-molecular signaling interactions with SIPs may help uncover unknown physiological functions of the SERT.


Asunto(s)
Proteínas de Transporte de Serotonina en la Membrana Plasmática/fisiología , Serotonina/metabolismo , Regulación Alostérica , Unión Proteica , Inhibidores Selectivos de la Recaptación de Serotonina/metabolismo , Inhibidores Selectivos de la Recaptación de Serotonina/farmacología , Transducción de Señal
9.
Psychopharmacology (Berl) ; 219(1): 1-13, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21901317

RESUMEN

RATIONALE: Escitalopram is a widely used antidepressant for the treatment of patients with major depression. It is the pure S-enantiomer of racemic citalopram. Several clinical trials and meta-analyses indicate that escitalopram is quantitatively more efficacious than many other antidepressants with a faster onset of action. OBJECTIVE: This paper reviews current knowledge about the mechanism of action of escitalopram. RESULTS: The primary target for escitalopram is the serotonin transporter (SERT), which is responsible for serotonin (or 5-hydroxytryptamine [5-HT]) reuptake at the terminals and cell bodies of serotonergic neurons. Escitalopram and selective serotonin reuptake inhibitors bind with high affinity to the 5-HT binding site (orthosteric site) on the transporter. This leads to antidepressant effects by increasing extracellular 5-HT levels which enhance 5-HT neurotransmission. SERT also has one or more allosteric sites, binding to which modulates activity at the orthosteric binding site but does not directly affect 5-HT reuptake by the transporter. In vitro studies have shown that through allosteric binding, escitalopram decreases its own dissociation rate from the orthosteric site on the SERT. R-citalopram, the nontherapeutic enantiomer in citalopram, is also an allosteric modulator of SERT but can inhibit the actions of escitalopram by interfering negatively with its binding. Both nonclinical studies and some clinical investigations have demonstrated the cellular, neurochemical, neuroadaptive, and neuroplastic changes induced by escitalopram with acute and chronic administration. CONCLUSIONS: The findings from binding, neurochemical, and neurophysiological studies may provide a mechanistic rationale for the clinical difference observed with escitalopram compared to other antidepressant therapies.


Asunto(s)
Antidepresivos de Segunda Generación/metabolismo , Citalopram/metabolismo , Inhibidores Selectivos de la Recaptación de Serotonina/metabolismo , Proteínas de Transporte de Serotonina en la Membrana Plasmática/metabolismo , Regulación Alostérica/fisiología , Animales , Antidepresivos de Segunda Generación/uso terapéutico , Citalopram/uso terapéutico , Trastorno Depresivo/tratamiento farmacológico , Trastorno Depresivo/metabolismo , Humanos , Proteínas de Transporte de Serotonina en la Membrana Plasmática/genética , Inhibidores Selectivos de la Recaptación de Serotonina/uso terapéutico
10.
J Med Chem ; 54(9): 3206-21, 2011 May 12.
Artículo en Inglés | MEDLINE | ID: mdl-21486038

RESUMEN

The synthesis and structure-activity relationship of a novel series of compounds with combined effects on 5-HT(3A) and 5-HT(1A) receptors and on the serotonin (5-HT) transporter (SERT) are described. Compound 5m (Lu AA21004) was the lead compound, displaying high affinity for recombinant human 5-HT(1A) (K(i) = 15 nM), 5-HT(1B) (K(i) = 33 nM), 5-HT(3A) (K(i) = 3.7 nM), 5-HT(7) (K(i) = 19 nM), and noradrenergic ß(1) (K(i) = 46 nM) receptors, and SERT (K(i) = 1.6 nM). Compound 5m displayed antagonistic properties at 5-HT(3A) and 5-HT(7) receptors, partial agonist properties at 5-HT(1B) receptors, agonistic properties at 5-HT(1A) receptors, and potent inhibition of SERT. In conscious rats, 5m significantly increased extracellular 5-HT levels in the brain after acute and 3 days of treatment. Following the 3-day treatment (5 or 10 (mg/kg)/day) SERT occupancies were only 43% and 57%, respectively. These characteristics indicate that 5m is a novel multimodal serotonergic compound, and 5m is currently in clinical development for major depressive disorder.


Asunto(s)
Antidepresivos/síntesis química , Trastorno Depresivo Mayor/tratamiento farmacológico , Piperazinas/síntesis química , Sulfuros/síntesis química , Animales , Antidepresivos/química , Antidepresivos/farmacología , Línea Celular , Agonismo Parcial de Drogas , Estabilidad de Medicamentos , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Humanos , Técnicas In Vitro , Microsomas Hepáticos/metabolismo , Oocitos/efectos de los fármacos , Oocitos/fisiología , Piperazinas/química , Piperazinas/farmacología , Ensayo de Unión Radioligante , Ratas , Receptor de Serotonina 5-HT1A/metabolismo , Receptor de Serotonina 5-HT2C/metabolismo , Receptores de Serotonina/metabolismo , Receptores de Serotonina 5-HT1/metabolismo , Receptores de Serotonina 5-HT3/metabolismo , Proteínas Recombinantes/agonistas , Proteínas Recombinantes/antagonistas & inhibidores , Serotonina/metabolismo , Agonistas del Receptor de Serotonina 5-HT1/síntesis química , Agonistas del Receptor de Serotonina 5-HT1/química , Agonistas del Receptor de Serotonina 5-HT1/farmacología , Antagonistas del Receptor de Serotonina 5-HT3/síntesis química , Antagonistas del Receptor de Serotonina 5-HT3/química , Antagonistas del Receptor de Serotonina 5-HT3/farmacología , Proteínas de Transporte de Serotonina en la Membrana Plasmática/metabolismo , Inhibidores Selectivos de la Recaptación de Serotonina/síntesis química , Inhibidores Selectivos de la Recaptación de Serotonina/química , Inhibidores Selectivos de la Recaptación de Serotonina/farmacología , Relación Estructura-Actividad , Sulfuros/química , Sulfuros/farmacología , Vortioxetina , Xenopus
11.
Neurosci Lett ; 462(3): 207-12, 2009 Oct 25.
Artículo en Inglés | MEDLINE | ID: mdl-19616061

RESUMEN

The human serotonin transporter (hSERT) has primary and allosteric binding sites for escitalopram and R-citalopram. Previous studies have established that the interaction of these two compounds at a low affinity allosteric binding site of hSERT can affect the dissociation of [(3)H]escitalopram from hSERT. The allosteric binding site involves a series of residues in the 10th, 11th, and 12th trans-membrane domains of hSERT. The low affinity allosteric activities of escitalopram and R-citalopram are essentially eliminated in a mutant hSERT with changes in some of these residues, namely A505V, L506F, I507L, S574T, I575T, as measured in dissociation binding studies. We confirm that in association binding experiments, R-citalopram at clinically relevant concentrations reduces the association rate of [(3)H]escitalopram as a ligand to wild type hSERT. We demonstrate that the ability of R-citalopram to reduce the association rate of escitalopram is also abolished in the mutant hSERT (A505V, L506F, I507L, S574T, I575T), along with the expected disruption the low affinity allosteric function on dissociation binding. This suggests that the allosteric binding site mediates both the low affinity and higher affinity interactions between R-citalopram, escitalopram, and hSERT. Our data add an additional structural basis for the different efficacies of escitalopram compared to racemic citalopram reported in animal studies and clinical trials, and substantiate the hypothesis that hSERT has complex allosteric mechanisms underlying the unexplained in vivo activities of its inhibitors.


Asunto(s)
Citalopram/farmacología , Inhibidores Selectivos de la Recaptación de Serotonina/farmacología , Proteínas de Transporte de Serotonina en la Membrana Plasmática/metabolismo , Sitio Alostérico , Animales , Unión Competitiva , Línea Celular , Citalopram/química , Cricetinae , Cricetulus , Antagonismo de Drogas , Humanos , Cinética , Mutación , Ensayo de Unión Radioligante , Proteínas de Transporte de Serotonina en la Membrana Plasmática/genética , Inhibidores Selectivos de la Recaptación de Serotonina/química , Estereoisomerismo
12.
Proc Natl Acad Sci U S A ; 102(48): 17489-94, 2005 Nov 29.
Artículo en Inglés | MEDLINE | ID: mdl-16287967

RESUMEN

The neuropeptide galanin mediates its effects through the receptor subtypes Gal(1), Gal(2), and Gal(3) and has been implicated in anxiety- and depression-related behaviors. Nevertheless, the receptor subtypes relevant to these behaviors are not known because of the lack of available galanin-selective ligands. In this article, we use behavioral, neurochemical, and electrophysiological approaches to investigate the anxiolytic- and antidepressant-like effects of two potent small-molecule, Gal(3)-selective antagonists, SNAP 37889 and the more soluble analog SNAP 398299. Acute administration of SNAP 37889 or SNAP 398299 enhanced rat social interaction. Furthermore, acute SNAP 37889 was also shown to reduce guinea pig vocalizations after maternal separation, to attenuate stress-induced hyperthermia in mice, to increase punished drinking in rats, and to decrease immobility and increase swimming time during forced swim tests with rats. Moreover, SNAP 37889 increased the social interaction time after 14 days of treatment and maintained its antidepressant effects during forced swim tests with rats after 21 days of treatment. In microdialysis studies, SNAP 37889 partially antagonized the galanin-evoked reduction in hippocampal serotonin (5-hydroxytryptamine, 5-HT), as did the 5-HT(1A) receptor antagonist WAY100635. Their combination produced a complete reversal of the effect of galanin. SNAP 398299 partially reversed the galanin-evoked inhibition of dorsal raphe cell firing and galanin-evoked hyperpolarizing currents. These results indicate that Gal(3)-selective antagonists produce anxiolytic- and antidepressant-like effects, possibly by attenuating the inhibitory influence of galanin on 5-HT transmission at the level of the dorsal raphe nucleus.


Asunto(s)
Conducta Animal/efectos de los fármacos , Hipocampo/metabolismo , Indoles/farmacología , Pirrolidinas/farmacología , Receptor de Galanina Tipo 3/antagonistas & inhibidores , Análisis de Varianza , Animales , Línea Celular , Electrofisiología , Cobayas , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Actividad Motora/efectos de los fármacos , Piperazinas/farmacología , Piridinas/farmacología , Ratas , Ratas Sprague-Dawley , Serotonina/metabolismo , Conducta Social , Vocalización Animal/efectos de los fármacos
13.
Mol Pharmacol ; 61(2): 436-45, 2002 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-11809869

RESUMEN

Several previous human postmortem experiments have detected an increase in striatal [(3)H]WIN 35428 binding to the dopamine transporter (DAT) in chronic cocaine users. However, animal experiments have found considerable variability in DAT radioligand binding levels in brain after cocaine administration, perhaps caused by length and dose of treatment and type of radioligand used. The present experiments tested the hypothesis that [(3)H]WIN 35428 binding and [(3)H]dopamine uptake would be increased by exposure to cocaine through alterations in DAT cellular trafficking, rather than increased protein synthesis. Experiments were conducted in stably hDAT-transfected N2A cells and assessed the dose response and time course of cocaine effects on [(3)H]WIN 35428 binding to the DAT, [(3)H]dopamine uptake, measures of DAT protein and mRNA, as well as DAT subcellular location. Cocaine doses of 10(-6) M caused statistically significant increases in [(3)H]WIN 35428 binding and [(3)H]dopamine uptake after 12 and 3 h, respectively. Despite these increases in DAT function, there was no change in DAT total protein or mRNA. Immunofluorescence and biotinylation experiments indicated that cocaine treatment induced increases in plasma membrane DAT immunoreactivity and intracellular decreases. The present model system may further our understanding of regulatory alterations in DAT radioligand binding and function caused by cocaine exposure.


Asunto(s)
Cocaína/análogos & derivados , Cocaína/farmacología , Inhibidores de Captación de Dopamina/farmacología , Dopamina/metabolismo , Glicoproteínas de Membrana , Proteínas de Transporte de Membrana/metabolismo , Proteínas del Tejido Nervioso , Animales , Unión Competitiva , Transporte Biológico , Western Blotting , Membrana Celular/metabolismo , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática , Interacciones Farmacológicas , Humanos , Proteínas de Transporte de Membrana/genética , Ratones , Fosforilación , ARN Mensajero/efectos de los fármacos , ARN Mensajero/metabolismo , Ensayo de Unión Radioligante , Sistemas de Mensajero Secundario/fisiología , Serotonina/metabolismo , Fracciones Subcelulares , Transfección , Tritio , Células Tumorales Cultivadas
14.
J Biol Chem ; 278(11): 9418-25, 2003 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-12524446

RESUMEN

RGS (regulators of G protein signaling) proteins are GTPase-activating proteins for the Galpha subunits of heterotrimeric G proteins and act to regulate signaling by rapidly cycling G protein. RGS proteins may integrate receptors and signaling pathways by physical or kinetic scaffolding mechanisms. To determine whether this results in enhancement and/or selectivity of agonist signaling, we have prepared C6 cells stably expressing the mu-opioid receptor and either pertussis toxin-insensitive or RGS- and pertussis toxin-insensitive Galpha(o). We have compared the activation of G protein, inhibition of adenylyl cyclase, stimulation of intracellular calcium release, and activation of the ERK1/2 MAPK pathway between cells expressing mutant Galpha(o) that is either RGS-insensitive or RGS-sensitive. The mu-receptor agonist [d-Ala(2),MePhe(4),Gly(5)-ol]enkephalin and partial agonist morphine were much more potent and/or had an increased maximal effect in inhibiting adenylyl cyclase and in activating MAPK in cells expressing RGS-insensitive Galpha(o). In contrast, mu-opioid agonist increases in intracellular calcium were less affected. The results are consistent with the hypothesis that the GTPase-activating protein activity of RGS proteins provides a control that limits agonist action through effector pathways and may contribute to selectivity of activation of intracellular signaling pathways.


Asunto(s)
Adenilil Ciclasas/metabolismo , Calcio/metabolismo , Proteínas de Unión al GTP Heterotriméricas/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Proteínas RGS/química , Receptores Opioides mu/metabolismo , Transducción de Señal , Animales , Línea Celular , Membrana Celular/metabolismo , AMP Cíclico/metabolismo , Relación Dosis-Respuesta a Droga , Encefalina Ala(2)-MeFe(4)-Gli(5)/farmacología , Activación Enzimática , GTP Fosfohidrolasas/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gi-Go , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Proteínas de Unión al GTP Heterotriméricas/química , Ligandos , Sistema de Señalización de MAP Quinasas , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos , Morfina/farmacología , Toxina del Pertussis/farmacología , Fosforilación , Unión Proteica , Proteínas RGS/metabolismo , Proteínas RGS/farmacología , Ratas , Factores de Tiempo , Transfección , Células Tumorales Cultivadas
15.
J Biol Chem ; 278(9): 7278-84, 2003 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-12446706

RESUMEN

Regulators of G protein signaling (RGS) are GTPase-accelerating proteins (GAPs), which can inhibit heterotrimeric G protein pathways. In this study, we provide experimental and theoretical evidence that high concentrations of receptors (as at a synapse) can lead to saturation of GDP-GTP exchange making GTP hydrolysis rate-limiting. This results in local depletion of inactive heterotrimeric G-GDP, which is reversed by RGS GAP activity. Thus, RGS enhances receptor-mediated G protein activation even as it deactivates the G protein. Evidence supporting this model includes a GTP-dependent enhancement of guanosine 5'-3-O-(thio)triphosphate (GTPgammaS) binding to G(i) by RGS. The RGS domain of RGS4 is sufficient for this, not requiring the NH(2)- or COOH-terminal extensions. Furthermore, a kinetic model including only the GAP activity of RGS replicates the GTP-dependent enhancement of GTPgammaS binding observed experimentally. Finally in a Monte Carlo model, this mechanism results in a dramatic "spatial focusing" of active G protein. Near the receptor, G protein activity is maintained even with RGS due to the ability of RGS to reduce depletion of local Galpha-GDP levels permitting rapid recoupling to receptor and maintained G protein activation near the receptor. In contrast, distant signals are suppressed by the RGS, since Galpha-GDP is not depleted there. Thus, a novel RGS-mediated "kinetic scaffolding" mechanism is proposed which narrows the spatial range of active G protein around a cluster of receptors limiting the spill-over of G protein signals to more distant effector molecules, thus enhancing the specificity of G(i) protein signals.


Asunto(s)
Proteínas RGS/química , Proteínas RGS/metabolismo , Transducción de Señal , Animales , Células CHO , Cricetinae , Relación Dosis-Respuesta a Droga , GTP Fosfohidrolasas/metabolismo , Glutatión Transferasa/metabolismo , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Guanosina Difosfato/metabolismo , Guanosina Trifosfato/metabolismo , Hidrólisis , Cinética , Método de Montecarlo , Unión Proteica , Estructura Terciaria de Proteína , Proteínas Recombinantes de Fusión/metabolismo , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA