Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Inflammation ; 42(6): 2257-2266, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31489527

RESUMEN

Gut microbiota-derived metabolite trimethylamine N-oxide (TMAO) has recently been shown to promote inflammation in peripheral tissues and the central nervous system (CNS), contributing to the pathogenesis of various human diseases. Here, we examined whether the presence of high levels of circulating TMAO would influence central and peripheral inflammation and inflammatory hyperalgesia in a carrageenan (CG)-induced rat model of inflammation. Rats were treated with vehicle or TMAO in drinking water. After 2 weeks of treatment, rats received intraplantar injection of saline or CG into the hind paw. Acute nociception was unaltered in TMAO-treated rats that had elevated plasma TMAO. Following CG injection, TMAO-treated rats were significantly more sensitive to thermal and mechanical stimulation of the inflamed paw and displayed greater paw edema. Molecular studies revealed that CG injection induced increases in recruitment of neutrophils/macrophages in the paw and activation of microglia in the spinal cord, along with increased activation of nuclear factor (NF)-kB and production of proinflammatory mediators in both vehicle-treated rats and TMAO-treated rats. However, the increases in the above parameters were more pronounced in TMAO-treated rats. Moreover, TMAO treatment decreased protein levels of anti-inflammatory mediator regulator of G protein signaling (RGS)-10 in both saline-injected rats and CG-injected rats. These findings suggest that the presence of high levels of circulating TMAO downregulates anti-inflammatory mediator RGS10 in both peripheral tissues and the CNS, which may increase the susceptibility to inflammatory challenge-induced NF-kB activity, leading to greater increase in production of inflammatory mediators and consequent exacerbation of peripheral inflammation and inflammatory hyperalgesia.


Asunto(s)
Hiperalgesia/inducido químicamente , Inflamación/inducido químicamente , Metilaminas/sangre , Animales , Carragenina , Edema , Mediadores de Inflamación/metabolismo , Metilaminas/farmacología , FN-kappa B/metabolismo , Proteínas RGS/efectos de los fármacos , Ratas
2.
Eur J Neurosci ; 48(5): 2110-2117, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30103281

RESUMEN

This study assessed the role of regulator of G protein signaling 2 (RGS2) in nicotine-induced anxiolytic- and antidepressant-like effects using RGS2 wildtype (WT) and RGS2 knockout (KO) mice. RGS2 negatively regulates monoaminergic neurotransmission, which is implicated in the pathology of anxiety and depression. We hypothesized that deletion of RGS2 would enhance nicotine-induced anxiolytic- and antidepressant-like effects, which were assessed using the elevated plus maze and tail suspension tests, respectively. Anxiolytic-like effects were observed in both RGS2 WT and KO mice after administration of low dose of nicotine (0.05 mg/kg, base) compared to respective saline controls. Additionally, administration of nicotine (0.1 mg/kg, base) compared to saline resulted in anxiolytic-like effects in RGS2 KO mice, but not RGS2 WT mice, suggesting genetic deletion of RGS2 facilitated anxiolytic-like effects of nicotine. Administration of nicotine (0.5 and 1 mg/kg, base) compared to saline resulted in antidepressant-like effects in RGS2 WT mice. Antidepressant-like effects were observed in RGS2 KO mice only at the highest tested dose of nicotine (1 mg/kg, base) compared to saline controls, suggesting that genetic deletion of RGS2 decreased sensitivity to antidepressant-like effects of nicotine. Together, the data suggest that RGS2 differentially regulated nicotine-induced affective behavioral responses. These data suggest that individuals with RGS2 polymorphisms may experience differential affective responses to tobacco smoking, which may make them vulnerable to developing nicotine addiction.


Asunto(s)
Ansiolíticos/farmacología , Antidepresivos/farmacología , Ansiedad/tratamiento farmacológico , Proteínas RGS/efectos de los fármacos , Animales , Ansiedad/genética , Trastornos de Ansiedad/tratamiento farmacológico , Conducta Animal/efectos de los fármacos , Depresión/inducido químicamente , Depresión/tratamiento farmacológico , Trastorno Depresivo/tratamiento farmacológico , Aprendizaje por Laberinto/efectos de los fármacos , Ratones Noqueados , Nicotina/farmacología
3.
Am J Physiol Heart Circ Physiol ; 312(2): H189-H200, 2017 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-27881385

RESUMEN

Peroxisome proliferator-activated receptors (PPARs) are members of the nuclear hormone receptor superfamily of ligand-activated transcription factors, which is composed of three members encoded by distinct genes: PPARα, PPARß/δ, and PPARγ. The biological actions of PPARα and PPARγ and their potential as a cardiovascular therapeutic target have been extensively reviewed, whereas the biological actions of PPARß/δ and its effectiveness as a therapeutic target in the treatment of hypertension remain less investigated. Preclinical studies suggest that pharmacological PPARß/δ activation induces antihypertensive effects in direct [spontaneously hypertensive rat (SHR), ANG II, and DOCA-salt] and indirect (dyslipemic and gestational) models of hypertension, associated with end-organ damage protection. This review summarizes mechanistic insights into the antihypertensive effects of PPARß/δ activators, including molecular and functional mechanisms. Pharmacological PPARß/δ activation induces genomic actions including the increase of regulators of G protein-coupled signaling (RGS), acute nongenomic vasodilator effects, as well as the ability to improve the endothelial dysfunction, reduce vascular inflammation, vasoconstrictor responses, and sympathetic outflow from central nervous system. Evidence from clinical trials is also examined. These preclinical and clinical outcomes of PPARß/δ ligands may provide a basis for the development of therapies in combating hypertension.


Asunto(s)
Hipertensión/fisiopatología , PPAR delta/fisiología , PPAR-beta/fisiología , Vasodilatación/fisiología , Animales , Antihipertensivos/farmacología , Antihipertensivos/uso terapéutico , Presión Sanguínea/efectos de los fármacos , Presión Sanguínea/fisiología , Endotelio Vascular/fisiopatología , Ácidos Grasos/metabolismo , Regulación de la Expresión Génica , Humanos , Hipertensión/tratamiento farmacológico , Inflamación , PPAR delta/agonistas , PPAR delta/metabolismo , PPAR-beta/agonistas , PPAR-beta/metabolismo , Fenoxiacetatos/farmacología , Fenoxiacetatos/uso terapéutico , Proteínas RGS/efectos de los fármacos , Proteínas RGS/genética , Ratas , Ratas Endogámicas SHR , Sistema Nervioso Simpático/fisiopatología , Tiazoles/farmacología , Tiazoles/uso terapéutico , Vasoconstricción/efectos de los fármacos , Vasoconstricción/fisiología , Vasodilatación/efectos de los fármacos
4.
J Pharmacol Exp Ther ; 357(2): 311-9, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26941169

RESUMEN

Regulator of G protein signaling (RGS) proteins have emerged as novel drug targets since their discovery almost two decades ago. RGS2 has received particular interest in cardiovascular research due to its role in regulating Gqsignaling in the heart and vascular smooth muscle. RGS2(-/-)mice are hypertensive, prone to heart failure, and display accelerated kidney fibrosis. RGS2 is rapidly degraded through the proteasome, and human mutations leading to accelerated RGS2 protein degradation correlate with hypertension. Hence, stabilizing RGS2 protein expression could be a novel route in treating cardiovascular disease. We previously identified cardiotonic steroids, including digoxin, as selective stabilizers of RGS2 protein in vitro. In the current study we investigated the functional effects of digoxin-mediated RGS2 protein stabilization in vivo. Using freshly isolated myocytes from wild-type and RGS2(-/-)mice treated with vehicle or low-dose digoxin (2µg/kg/day for 7 days) we demonstrated that agonist-induced cAMP levels and cardiomyocyte contractility was inhibited by digoxin in wild-type but not in RGS2(-/-)mice. This inhibition was accompanied by an increase in RGS2 protein levels in cardiomyocytes as well as in whole heart tissue. Furthermore, digoxin had protective effects in a model of cardiac injury in wild-type mice and this protection was lost in RGS2(-/-)mice. Digoxin is the oldest known therapy for heart failure; however, beyond its activity at the Na(+)/K(+)-ATPase, the exact mechanism of action is not known. The current study adds a novel mechanism, whereby through stabilizing RGS2 protein levels digoxin could exert its protective effects in the failing heart.


Asunto(s)
Cardiotónicos/farmacología , Digoxina/farmacología , Cardiopatías/prevención & control , Proteínas RGS/biosíntesis , Animales , AMP Cíclico/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Contracción Miocárdica/efectos de los fármacos , Miocitos Cardíacos/efectos de los fármacos , Proteínas RGS/efectos de los fármacos , Receptores Acoplados a Proteínas G/efectos de los fármacos , Receptores Acoplados a Proteínas G/metabolismo , ATPasa Intercambiadora de Sodio-Potasio/genética , ATPasa Intercambiadora de Sodio-Potasio/metabolismo , Regulación hacia Arriba/efectos de los fármacos
5.
Asian Pac J Cancer Prev ; 13(4): 1097-104, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22799288

RESUMEN

BACKGROUND/AIM: Pristimerin isolated from Celastrus and Maytenus spp can inhibit proteasome activity. However, whether pristimerin can modulate cancer metastasis is unknown. METHODS: The impacts of pristimerin on the purified and intracellular chymotrypsin proteasomal activity, the levels of regulator of G protein signaling 4 (RGS 4) expression and breast cancer cell lamellipodia formation, and the migration and invasion were determined by enzymatic, Western blot, immunofluorescent, and transwell assays, respectively. RESULTS: We found that pristimerin inhibited human chymotrypsin proteasomal activity in MDA-MB-231 cells in a dose-dependent manner. Pristimerin also inhibited breast cancer cell lamellipodia formation, migration, and invasion in vitro by up-regulating RGS4 expression. Thus, knockdown of RGS4 attenuated pristimerin-mediated inhibition of breast cancer cell migration and invasion. Furthermore, pristimerin inhibited growth and invasion of implanted breast tumors in mice. CONCLUSION: Pristmerin inhibits proteasomal activity and increases the levels of RGS4, inhibiting the migration and invasion of breast cancer cells.


Asunto(s)
Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Movimiento Celular/efectos de los fármacos , Inhibidores de Proteasoma/farmacología , Proteínas RGS/metabolismo , Triterpenos/farmacología , Actinas/metabolismo , Análisis de Varianza , Animales , Antineoplásicos/farmacología , Neoplasias de la Mama/enzimología , Línea Celular Tumoral , Quimasas/efectos de los fármacos , Quimasas/metabolismo , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Leupeptinas/farmacología , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Invasividad Neoplásica , Triterpenos Pentacíclicos , Complejo de la Endopetidasa Proteasomal/efectos de los fármacos , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteínas RGS/efectos de los fármacos , Proteínas RGS/genética , Interferencia de ARN , Distribución Aleatoria , Carga Tumoral/efectos de los fármacos , Regulación hacia Arriba
6.
Curr Protoc Pharmacol ; Chapter 2: Unit2.8, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22382998

RESUMEN

"Regulator of G-protein Signaling" (RGS) proteins constitute a class of intracellular signaling regulators that accelerate GTP hydrolysis by heterotrimeric Gα subunits. In recent years, RGS proteins have emerged as potential drug targets for modulation by small molecules. Described in this unit are high-throughput screening procedures for identifying modulators of RGS protein-mediated GTPase acceleration (GAP activity), for assessment of RGS domain/Gα interactions (most avid in vitro when Gα is bound by aluminum tetrafluoride), and for validation of candidate GAP-modulatory molecules with the single-turnover GTP hydrolysis assay.


Asunto(s)
Bioensayo/métodos , Proteínas de Unión al GTP/metabolismo , Proteínas RGS/metabolismo , Transducción de Señal/fisiología , Cromatografía Liquida/métodos , Transferencia Resonante de Energía de Fluorescencia/métodos , Colorantes Fluorescentes , Subunidades alfa de la Proteína de Unión al GTP/aislamiento & purificación , Subunidades alfa de la Proteína de Unión al GTP/metabolismo , Proteínas de Unión al GTP/efectos de los fármacos , Proteínas Activadoras de GTPasa/metabolismo , Hidrólisis , Proteínas RGS/efectos de los fármacos , Transducción de Señal/efectos de los fármacos
7.
J Neurosci ; 31(15): 5617-24, 2011 Apr 13.
Artículo en Inglés | MEDLINE | ID: mdl-21490202

RESUMEN

The signaling molecule RGS9-2 is a potent modulator of G-protein-coupled receptor function in striatum. Our earlier work revealed a critical role for RGS9-2 in the actions of the µ-opioid receptor (MOR) agonist morphine. In this study, we demonstrate that RGS9-2 may act as a positive or negative modulator of MOR-mediated behavioral responses in mice depending on the agonist administered. Paralleling these findings we use coimmunoprecipitation assays to show that the signaling complexes formed between RGS9-2 and Gα subunits in striatum are determined by the MOR agonist, and we identify RGS9-2 containing complexes associated with analgesic tolerance. In striatum, MOR activation promotes the formation of complexes between RGS9-2 and several Gα subunits, but morphine uniquely promotes an association between RGS9-2 and Gαi3. In contrast, RGS9-2/Gαq complexes assemble after acute application of several MOR agonists but not after morphine application. Repeated morphine administration leads to the formation of distinct complexes, which contain RGS9-2, Gß5, and Gαq. Finally, we use simple pharmacological manipulations to disrupt RGS9-2 complexes formed during repeated MOR activation to delay the development of analgesic tolerance to morphine. Our data provide a better understanding of the brain-region-specific signaling events associated with opiate analgesia and tolerance and point to pharmacological approaches that can be readily tested for improving chronic analgesic responsiveness.


Asunto(s)
Analgésicos Opioides/farmacología , Cuerpo Estriado/fisiología , Proteínas RGS/fisiología , Animales , Arrestinas/genética , Arrestinas/fisiología , Western Blotting , Cuerpo Estriado/efectos de los fármacos , Tolerancia a Medicamentos , Quinasas MAP Reguladas por Señal Extracelular/genética , Quinasas MAP Reguladas por Señal Extracelular/fisiología , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/genética , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/metabolismo , Inmunoprecipitación , Ratones , Ratones Noqueados , Morfina/farmacología , Fosfolipasa C beta/metabolismo , Fosforilación , Proteínas RGS/efectos de los fármacos , Proteínas RGS/genética , Receptores Opioides mu/agonistas , Receptores Opioides mu/genética , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , beta-Arrestinas
8.
Prog Mol Biol Transl Sci ; 91: 81-119, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20691960

RESUMEN

Signaling via G-protein-coupled receptors (GPCRs) is central for the function of biological systems. Many clinically used drugs target GPCRs directly or target molecules involved in GPCR signaling. As an alternative to targeting receptors directly, one could modulate signaling cascades downstream of receptor activation. In recent years, there has been substantial interest in a family of proteins called regulators of G protein signaling (RGS) proteins. They modulate GPCR signaling by accelerating GTP hydrolysis on active Galpha subunits, thereby reducing the amplitude and duration of signaling. Modulating RGS activity would be a useful strategy to control GPCR signaling. An RGS inhibitor would be expected to enhance GPCR signaling and could do so in a tissue- or pathway-specific manner. Apart from the central GAP (GTPase accelerating protein) activity, many RGS proteins also have other functions like regulating protein-protein interactions, subcellular localization of signaling molecules, and protein translation. It is clear that these proteins serve important functions in a number of physiological and pathophysiological processes, and they are emerging as potential drug targets. This chapter gives an overview of what is currently known about biological functions of RGS proteins based on in vivo and in vitro data. We also summarize the current status in targeting RGS proteins in drug discovery.


Asunto(s)
Descubrimiento de Drogas , Proteínas RGS/efectos de los fármacos , Proteínas RGS/metabolismo , Animales , Sistema Cardiovascular/metabolismo , Sistema Nervioso Central/metabolismo , Humanos , Modelos Biológicos , Neoplasias/metabolismo , Proteínas RGS/química , Transducción de Señal/efectos de los fármacos , Homología Estructural de Proteína
9.
Eur J Pharmacol ; 601(1-3): 66-72, 2008 Dec 28.
Artículo en Inglés | MEDLINE | ID: mdl-18992738

RESUMEN

We have previously reported that black tea polyphenol theaflavin monogallate (TF-2) suppressed COX-2 and induced apoptosis in human colon cancer cells [Lu, J.B., Ho, C.-T., Ghai, G., Chen, K.Y., 2000. Differential effects of theaflavin monogallates on cell growth, apoptosis and Cox-2 gene expression in cancerous versus normal cells. Cancer Res. 60, 6465-6471.]. We now extended the study by using PCR-based differential display to search for genes that were selectively induced by TF-2. Here we report the identification of Regulator of G-binding protein signaling 10 (RGS10) as the target gene, which was induced as early as 4 h after the TF-2 treatment. We then examined the effect of TF-2 on several other RGS genes and found that, in addition to RGS10, TF-2 induced the expression of RGS14, but not RGS4. Other tea polyphenols, including theaflavin-3,3'-digallate (TF-3) and (-) epigallocatechin-3-gallate (EGCG), also induced the expression of RGS10 and RGS14, but not RGS4. However, theaflavin (TF-1), which does not contain the gallate moiety, was ineffective. These results showed for the first time that tea polyphenols can induce the expression of selective RGS genes and that the gallate moiety may be important in this induction. In view of the role of RGS in modulating G-protein mediated signal transduction pathways, our findings may be significant since dysregulation of G-signaling is prominently implicated in carcinogenesis.


Asunto(s)
Biflavonoides/farmacología , Catequina/farmacología , Ácido Gálico/análogos & derivados , Regulación de la Expresión Génica/efectos de los fármacos , Proteínas RGS/efectos de los fármacos , Té/química , Biflavonoides/aislamiento & purificación , Células CACO-2 , Catequina/análogos & derivados , Catequina/aislamiento & purificación , Neoplasias del Colon , Inducción Enzimática/efectos de los fármacos , Flavonoides/aislamiento & purificación , Flavonoides/farmacología , Ácido Gálico/aislamiento & purificación , Ácido Gálico/farmacología , Humanos , Fenoles/aislamiento & purificación , Fenoles/farmacología , Polifenoles , Proteínas RGS/genética , Factores de Tiempo
10.
J Neurochem ; 106(5): 2224-35, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18624922

RESUMEN

Somatostatin acts at five G protein-coupled receptors, sst(1)-sst(5). In mouse ischaemic retinas, the over-expression of sst(2) (as in sst(1) knock-out mice) results in the reduction of cell death and glutamate release. In this study, we reported that, in wild-type retinas, somatostatin, the multireceptor ligand pasireotide and the sst(2) agonist octreotide decreased ischaemia-induced cell death and that octreotide also decreased glutamate release. In contrast, cell death was increased by blocking sst(2) with cyanamide. In sst(2) over-expressing ischaemic retinas, somatostatin analogues increased cell death, and octreotide also increased glutamate release. To explain this reversal of the anti-ischaemic effect of somatostatin agonists in the presence of sst(2) over-expression, we tested sst(2) desensitisation because of internalisation or altered receptor function. We observed that (i) sst(2) was not internalised, (ii) among G protein-coupled receptor kinases (GRKs) and regulators of G protein signalling (RGSs), GRK1 and RGS1 expression increased following ischaemia, (iii) both GRK1 and RGS1 were down-regulated by octreotide in wild-type ischaemic retinas, (iv) octreotide down-regulated GRK1 but not RGS1 in sst(2) over-expressing ischaemic retinas. These results demonstrate that sst(2) activation protects against retinal ischaemia. However, in the presence of sst(2) over-expression sst(2) is functionally desensitised by agonists, possibly because of sustained RGS1 levels.


Asunto(s)
Isquemia Encefálica/metabolismo , Degeneración Nerviosa/metabolismo , Receptores de Somatostatina/agonistas , Retina/metabolismo , Enfermedades de la Retina/metabolismo , Somatostatina/análogos & derivados , Animales , Isquemia Encefálica/tratamiento farmacológico , Isquemia Encefálica/genética , Muerte Celular/efectos de los fármacos , Muerte Celular/fisiología , Cianamida/farmacología , Femenino , Quinasa 1 del Receptor Acoplado a Proteína-G/efectos de los fármacos , Quinasa 1 del Receptor Acoplado a Proteína-G/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Degeneración Nerviosa/tratamiento farmacológico , Degeneración Nerviosa/genética , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Octreótido/farmacología , Oligopéptidos/farmacología , Proteínas RGS/efectos de los fármacos , Proteínas RGS/metabolismo , Receptores de Somatostatina/genética , Retina/efectos de los fármacos , Retina/fisiopatología , Enfermedades de la Retina/tratamiento farmacológico , Enfermedades de la Retina/genética , Somatostatina/metabolismo
11.
Learn Mem ; 15(5): 281-9, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18441285

RESUMEN

Activation of beta-adrenergic receptors (beta-ARs) enhances hippocampal memory consolidation and long-term potentiation (LTP), a likely mechanism for memory storage. One signaling pathway linked to beta-AR activation is the cAMP-PKA pathway. PKA is critical for the consolidation of hippocampal long-term memory and for the expression of some forms of long-lasting hippocampal LTP. How does beta-AR activation affect the PKA-dependence, and persistence, of LTP elicited by distinct stimulation frequencies? Here, we use in vitro electrophysiology to show that patterns of stimulation determine the temporal phase of LTP affected by beta-AR activation. In addition, only specific patterns of stimulation recruit PKA-dependent LTP following beta-AR activation. Impairments of PKA-dependent LTP maintenance generated by pharmacologic or genetic deficiency of PKA activity are also abolished by concurrent activation of beta-ARs. Taken together, our data show that, depending on patterns of synaptic stimulation, activation of beta-ARs can gate the PKA-dependence and persistence of synaptic plasticity. We suggest that this may allow neuromodulatory receptors to fine-tune neural information processing to meet the demands imposed by numerous synaptic activity profiles. This is a form of "metaplasticity" that could control the efficacy of consolidation of hippocampal long-term memories.


Asunto(s)
Hipocampo/efectos de los fármacos , Péptidos y Proteínas de Señalización Intracelular/farmacología , Potenciación a Largo Plazo/efectos de los fármacos , Receptores Adrenérgicos beta/efectos de los fármacos , Agonistas Adrenérgicos beta/administración & dosificación , Agonistas Adrenérgicos beta/farmacología , Animales , Carbazoles/administración & dosificación , Carbazoles/farmacología , Proteínas Quinasas Dependientes de AMP Cíclico/antagonistas & inhibidores , Estimulación Eléctrica , Inhibidores Enzimáticos/administración & dosificación , Inhibidores Enzimáticos/farmacología , Femenino , Hipocampo/metabolismo , Péptidos y Proteínas de Señalización Intracelular/administración & dosificación , Isoproterenol/administración & dosificación , Isoproterenol/farmacología , Locus Coeruleus/efectos de los fármacos , Memoria/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Ratones Transgénicos , Red Nerviosa/efectos de los fármacos , Plasticidad Neuronal/efectos de los fármacos , Pirroles/administración & dosificación , Pirroles/farmacología , Proteínas RGS/efectos de los fármacos , Receptores Adrenérgicos beta/metabolismo , Transmisión Sináptica/efectos de los fármacos
12.
Eur J Neurosci ; 26(9): 2532-8, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17970732

RESUMEN

Ontogenetic treatment of rats with the dopamine D(2)-like receptor agonist quinpirole produces a significant increase in dopamine D(2) receptor sensitivity that persists throughout the animal's lifetime, a phenomenon known as D(2) priming. The present study was designed to investigate the effects of priming of the D(2) receptor on the expression of three different members of the regulator of G-protein signaling (RGS) family: Rgs4, Rgs9 and Rgs17. Male offspring were ontogenetically treated with quinpirole or saline from postnatal days (P)1-21 and raised to adulthood. On approximately P65, animals were given an acute quinipirole injection (0.1 mg/kg) and the number of yawns was recorded for 1 h after the injection. Yawning has been shown to be a behavioural event mediated by the dopamine D(2)/D(3) receptor. Animals ontogenetically treated with quinpirole demonstrated a significant 2.5-fold increase in yawning as compared to controls. Rgs transcripts were analysed through in situ hybridization several weeks later. Rats ontogenetically treated with quinpirole demonstrated a significant decrease in Rgs9 expression in the frontal cortex, but a more robust decrease in the striatum and nucleus accumbens as compared to controls. Regarding Rgs17, ontogenetic quinpirole produced a modest but significant increase in expression in the same brain areas. There were no significant differences in Rgs4 expression produced by drug treatment in any of the brain regions analysed. This study demonstrates that ontogenetic quinpirole treatment, which results in priming of the D(2) receptor, results in significant decreases in Rgs9, which has been shown to regulate G-protein coupling to D(2) receptors.


Asunto(s)
Encéfalo/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Quinpirol/farmacología , Proteínas RGS/efectos de los fármacos , Proteínas RGS/metabolismo , Receptores de Dopamina D2/efectos de los fármacos , Animales , Animales Recién Nacidos , Encéfalo/crecimiento & desarrollo , Encéfalo/metabolismo , Cuerpo Estriado/efectos de los fármacos , Cuerpo Estriado/crecimiento & desarrollo , Cuerpo Estriado/metabolismo , Dopamina/metabolismo , Agonistas de Dopamina/farmacología , Femenino , Lóbulo Frontal/efectos de los fármacos , Lóbulo Frontal/crecimiento & desarrollo , Lóbulo Frontal/metabolismo , Proteínas de Unión al GTP/efectos de los fármacos , Proteínas de Unión al GTP/metabolismo , Regulación de la Expresión Génica/fisiología , Masculino , Núcleo Accumbens/efectos de los fármacos , Núcleo Accumbens/crecimiento & desarrollo , Núcleo Accumbens/metabolismo , Proteínas RGS/genética , Ratas , Ratas Sprague-Dawley , Receptores de Dopamina D2/metabolismo , Receptores Acoplados a Proteínas G/efectos de los fármacos , Receptores Acoplados a Proteínas G/metabolismo , Tiempo , Bostezo/efectos de los fármacos , Bostezo/fisiología
13.
J Neurosci Res ; 85(11): 2385-99, 2007 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-17551986

RESUMEN

Activity-dependent gene expression is one of the key mechanisms of synaptic plasticity that form the basis of higher order functions such as learning and memory. In the present study, we surveyed for activity-dependent genes by analyzing gene expression changes accompanying reversible inhibition of synaptic activity by tetrodotoxin (TTX) using two types of DNA microarrays; our focused oligo DNA microarray "Synaptoarray" and the commercially available high-density array. Cerebral cortical cells from E18 rat embryos were cultured for 14 days to ensure synaptogenesis, then treated with 1 muM TTX for 48 hr without detectable effect on cell viability. Synaptic density estimated by the amount of Synapsin I and Synaptotagmin I was decreased 21-24% by TTX treatment, but recovered to the control level 48 hr after TTX withdrawal. Comparison of gene expression profiles by competitive hybridization of fluorescently labeled cRNA from TTX-treated and control cells showed an overall downregulation of the genes on the Synaptoarray by TTX-treatment with different recovery rates after TTX withdrawal. With 16 representative genes, microarray data were validated by real-time PCR analysis. Genes most severely downregulated by TTX and upregulated above the control level at 5 hr after TTX withdrawal were munc13-1 (involved in docking and priming of synaptic vesicles) and Shank2 (involved in the postsynaptic scaffold). In addition, comprehensive screening at 5 hr after TTX withdrawal using high density arrays resulted in additional identification of Rgs2, a regulator of trimeric G-protein signaling, as an activity-dependent gene. These three genes are thus likely to be key factors in the regulation of synaptic plasticity. (c) 2007 Wiley-Liss, Inc.


Asunto(s)
Expresión Génica/efectos de los fármacos , Neuronas/fisiología , Venenos/administración & dosificación , Sinapsis/genética , Tetrodotoxina/administración & dosificación , Animales , Western Blotting , Células Cultivadas , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/fisiología , Perfilación de la Expresión Génica , Inmunohistoquímica , Proteínas del Tejido Nervioso/biosíntesis , Proteínas del Tejido Nervioso/efectos de los fármacos , Plasticidad Neuronal/efectos de los fármacos , Plasticidad Neuronal/genética , Neuronas/efectos de los fármacos , Análisis de Secuencia por Matrices de Oligonucleótidos , Proteínas RGS/biosíntesis , Proteínas RGS/efectos de los fármacos , Ratas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sinapsis/efectos de los fármacos , Factores de Tiempo
14.
Synapse ; 61(5): 303-9, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17318883

RESUMEN

It is known that RGS9-2 gene knockout mice show supersensitivity to DA and have a marked elevation in the proportion of DA D2 receptors in the high-affinity state for DA (D2(High) receptors). As this is a similar profile to that observed in the CNS from subjects with schizophrenia, we examined whether postmortem CNS tissue from subjects with the disorder and brain striata from an animal model of psychosis or schizophrenia (the amphetamine-sensitized rat) had altered levels of RGS9-2. The mRNA for RGS9-2 in 29 control hippocampi was 0.185 +/- 0.015 fg per fg of beta-glucuronidase mRNA (average +/- SE), while that in 29 schizophrenia hippocampi was 0.145 +/- 0.015 fg per fg of beta-glucuronidase mRNA (average +/- SE), a reduction of 22%. Of the many receptor-regulating genes related to G proteins, and of 11 RGS genes, RGS9-2 was the most reduced in the amphetamine-sensitized rat striatum. The reduced levels of RGS9-2 expression in both an animal model of schizophrenia and a postmortem schizophrenia brain provide further evidence implicating RGS9-2 as a candidate gene in schizophrenia.


Asunto(s)
Anfetamina/farmacología , Encéfalo/metabolismo , Proteínas RGS/biosíntesis , Esquizofrenia/genética , Esquizofrenia/metabolismo , Animales , Autopsia , Encéfalo/efectos de los fármacos , Modelos Animales de Enfermedad , Dopamina/metabolismo , Dopaminérgicos/farmacología , Expresión Génica , Humanos , Masculino , Análisis de Secuencia por Matrices de Oligonucleótidos , Proteínas RGS/efectos de los fármacos , ARN Mensajero/análisis , Ratas , Ratas Sprague-Dawley , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Esquizofrenia/inducido químicamente
15.
Brain Res ; 1093(1): 161-6, 2006 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-16677622

RESUMEN

Prolonged over-exposure of rats to corticosterone attenuates 5-HT(1A)-receptor-mediated responses in hippocampal CA1 cells through an unknown mechanism, not involving downregulation of 5-HT(1A) receptor expression. We here tested if corticosterone changes 5-HT(1A) receptor function indirectly, by altering hippocampal mRNA expression of NCAM, SGK1, or RGS4, which all modulate 5-HT(1A) receptor function. We found that the expression of none of these candidates was affected by corticosterone treatment.


Asunto(s)
Antiinflamatorios/farmacología , Corticosterona/farmacología , Hipocampo/efectos de los fármacos , Proteínas Inmediatas-Precoces/efectos de los fármacos , Moléculas de Adhesión de Célula Nerviosa/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/efectos de los fármacos , Proteínas RGS/efectos de los fármacos , Animales , Hipocampo/metabolismo , Proteínas Inmediatas-Precoces/biosíntesis , Hibridación in Situ , Masculino , Moléculas de Adhesión de Célula Nerviosa/biosíntesis , Proteínas Serina-Treonina Quinasas/biosíntesis , Proteínas RGS/biosíntesis , ARN Mensajero/análisis , Ratas , Ratas Wistar , Factores de Tiempo
16.
J Neurochem ; 96(6): 1606-15, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16539683

RESUMEN

Administration of psychostimulants modulates mRNA of several regulators of guanine nucleotide-binding protein signaling (RGSs) proteins in the brain. In the present study, the regulation of amphetamine-induced decrease of RGS4 expression in the rat forebrain was evaluated. RGS4 mRNA was reduced by amphetamine in an inverse, dose-dependent manner. The lowest dose (2.5 mg/kg) decreased RGS4 mRNA in caudate putamen for up to 6 h after injection whereas the decrease in several frontal cortical areas was detected at 3 h only. Analysis of RGS4 immunoreactivity by western blotting revealed a decrease 3 h after amphetamine solely in the caudate putamen. Systemic administration of D(1) (SCH23390) or D(2) (eticlopride) receptor antagonists blocked amphetamine-induced locomotion but amphetamine augmented both the SCH23390-induced increase and the eticlopride-induced decrease in RGS4 mRNA in the caudate putamen. Further, the down-regulation of RGS4 immunoreactivity by eticlopride was robust whereas the effect of SCH23390 was blunted as compared with its effect on mRNA. These data suggest that, by decreasing RGS4 expression in the caudate putamen via D(1) receptors, acute amphetamine could disinhibit RGS4-sensitive guanine nucleotide-binding protein alpha-subunit i- and/or q-coupled signaling pathways and favor mechanisms that counterbalance D(1) receptor stimulation.


Asunto(s)
Anfetamina/farmacología , Dopamina/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Prosencéfalo/efectos de los fármacos , Proteínas RGS/efectos de los fármacos , Receptores Dopaminérgicos/metabolismo , Trastornos Relacionados con Anfetaminas/genética , Trastornos Relacionados con Anfetaminas/metabolismo , Trastornos Relacionados con Anfetaminas/fisiopatología , Animales , Benzazepinas/farmacología , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/metabolismo , Modelos Animales de Enfermedad , Antagonistas de Dopamina/farmacología , Relación Dosis-Respuesta a Droga , Regulación hacia Abajo/fisiología , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/metabolismo , Masculino , Neostriado/efectos de los fármacos , Neostriado/metabolismo , Prosencéfalo/metabolismo , Prosencéfalo/fisiopatología , Proteínas RGS/genética , Proteínas RGS/metabolismo , ARN Mensajero/efectos de los fármacos , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores Dopaminérgicos/efectos de los fármacos , Receptores de Dopamina D1/efectos de los fármacos , Receptores de Dopamina D1/metabolismo , Receptores de Dopamina D2/efectos de los fármacos , Receptores de Dopamina D2/metabolismo , Salicilamidas/farmacología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología
17.
Circ Res ; 96(4): 401-11, 2005 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-15746448

RESUMEN

Regulator of G-protein-signaling (RGS) proteins play a key role in the regulation of G-protein-coupled receptor (GPCR) signaling. The characteristic hallmark of RGS proteins is a conserved approximately 120-aa RGS region that confers on these proteins the ability to serve as GTPase-activating proteins (GAPs) for G(alpha) proteins. Most RGS proteins can serve as GAPs for multiple isoforms of G(alpha) and therefore have the potential to influence many cellular signaling pathways. However, RGS proteins can be highly regulated and can demonstrate extreme specificity for a particular signaling pathway. RGS proteins can be regulated by altering their GAP activity or subcellular localization; such regulation is achieved by phosphorylation, palmitoylation, and interaction with protein and lipid-binding partners. Many RGS proteins have GAP-independent functions that influence GPCR and non-GPCR-mediated signaling, such as effector regulation or action as an effector. Hence, RGS proteins should be considered multifunctional signaling regulators. GPCR-mediated signaling is critical for normal function in the cardiovascular system and is currently the primary target for the pharmacological treatment of disease. Alterations in RGS protein levels, in particular RGS2 and RGS4, produce cardiovascular phenotypes. Thus, because of the importance of GPCR-signaling pathways and the profound influence of RGS proteins on these pathways, RGS proteins are regulators of cardiovascular physiology and potentially novel drug targets as well.


Asunto(s)
Enfermedades Cardiovasculares/tratamiento farmacológico , Miocardio/metabolismo , Proteínas RGS/fisiología , Proteínas 14-3-3/metabolismo , Animales , Fármacos Cardiovasculares/farmacología , Fármacos Cardiovasculares/uso terapéutico , Enfermedades Cardiovasculares/metabolismo , Diseño de Fármacos , Subunidades alfa de la Proteína de Unión al GTP/metabolismo , Regulación de la Expresión Génica , Corazón/efectos de los fármacos , Humanos , Ratones , Familia de Multigenes , Unión Proteica , Isoformas de Proteínas/fisiología , Procesamiento Proteico-Postraduccional , Estructura Terciaria de Proteína , Proteínas RGS/efectos de los fármacos , Ratas , Receptores Acoplados a Proteínas G/fisiología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Fracciones Subcelulares/metabolismo , Especificidad por Sustrato
18.
Neuroscience ; 127(2): 261-7, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-15262317

RESUMEN

Desensitization of post-synaptic serotonin1A (5-HT1A) receptors may underlie the clinical improvement of neuropsychiatric disorders. In the hypothalamic paraventricular nucleus, Galphaz proteins mediate the 5-HT1A receptor-stimulated increases in hormone release. Regulator of G protein signaling-Z1 (RGSZ1) is a GTPase-activating protein selective for Galphaz proteins. RGSZ1 regulates the duration of interaction between Galphaz proteins and effector systems. The present investigation determined the levels of RGSZ1 in the hypothalamic paraventricular nucleus of rats subjected to four different treatment protocols that produce desensitization of 5-HT1A receptors. These protocols include: daily administration of beta estradiol 3-benzoate (estradiol) for 2 days; daily administration of fluoxetine for 3 and 14 days; daily administration of cocaine for 7 or 14 days; and acute administration of (+/-)-1-(2,5 dimethoxy-4-iodophenyl)-2-amino-propane HCl (DOI; a 5-HT2A/2C receptor agonist). Estradiol treatment was the only protocol that increased the levels of RGSZ1 protein in the hypothalamic paraventricular nucleus in a dose-dependent manner (46%-132% over control). Interestingly, previous experiments indicate that only estradiol produces a decreased Emax of 5-HT1A receptor-stimulation of hormone release, whereas fluoxetine, cocaine and DOI produce a shift to the right (increased ED50). Thus, the desensitization of 5-HT1A receptors by estradiol might be attributable to increased levels of RGSZ1 protein. These findings may provide insight into the adaptation of 5-HT1A receptor signaling during pharmacotherapies of mood disorders in women and the well-established gender differences in the vulnerability to depression.


Asunto(s)
Estrógenos/farmacología , Proteínas Activadoras de GTPasa/efectos de los fármacos , Proteínas Activadoras de GTPasa/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Núcleo Hipotalámico Paraventricular/efectos de los fármacos , Proteínas RGS/metabolismo , Receptor de Serotonina 5-HT1A/efectos de los fármacos , Adaptación Fisiológica/efectos de los fármacos , Adaptación Fisiológica/fisiología , Animales , Química Encefálica/genética , Cocaína/farmacología , Trastorno Depresivo/genética , Trastorno Depresivo/metabolismo , Trastorno Depresivo/fisiopatología , Relación Dosis-Respuesta a Droga , Resistencia a Medicamentos , Estrógenos/metabolismo , Femenino , Predisposición Genética a la Enfermedad/genética , Masculino , Proteínas de la Membrana/efectos de los fármacos , Proteínas del Tejido Nervioso/efectos de los fármacos , Núcleo Hipotalámico Paraventricular/metabolismo , Proteínas RGS/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Receptor de Serotonina 5-HT1A/metabolismo , Serotonina/metabolismo , Agonistas de Receptores de Serotonina/farmacología , Inhibidores Selectivos de la Recaptación de Serotonina/farmacología , Caracteres Sexuales , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/fisiología
19.
Eur J Neurosci ; 17(5): 971-80, 2003 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-12653973

RESUMEN

The present study explored a possible role for RGS (regulators of G protein signalling) proteins in the long term actions of morphine in the locus coeruleus (LC), a brainstem region implicated in opiate physical dependence and withdrawal. Morphine influences LC neurons through activation of micro -opioid receptors, which, being Gi/o-linked, would be expected to be modulated by RGS proteins. We focused on several RGS subtypes that are known to be expressed in this brain region. Levels of mRNAs encoding RGS2, -3, -4, -5, -7, -8 and -11 are unchanged following chronic morphine, but RGS2 and -4 mRNA levels are increased 2-3-fold 6 h following precipitation of opiate withdrawal. The increases in RGS2 and -4 mRNA peak after 6 h of withdrawal and return to control levels by 24 h. Immunoblot analysis of RGS4 revealed a striking divergence between mRNA and protein responses in LC: protein levels are elevated twofold following chronic morphine and decrease to control values by 6 h of withdrawal. In contrast, levels of RGS7 and -11 proteins, the only other subtypes for which antibodies are available, were not altered by these treatments. Intracellular application of wild-type RGS4, but not a GTPase accelerating-deficient mutant of RGS4, into LC neurons diminished electrophysiological responses to morphine. The observed subtype- and time-specific regulation of RGS4 protein and mRNA, and the diminished morphine-induced currents in the presence of elevated RGS4 protein levels, indicate that morphine induction of RGS4 could contribute to aspects of opiate tolerance and dependence displayed by LC neurons.


Asunto(s)
Locus Coeruleus/efectos de los fármacos , Morfina/farmacología , Narcóticos/farmacología , Neuronas/efectos de los fármacos , Proteínas RGS/efectos de los fármacos , Animales , Western Blotting , Inmunohistoquímica , Hibridación in Situ , Locus Coeruleus/metabolismo , Masculino , Potenciales de la Membrana/efectos de los fármacos , Naltrexona/farmacología , Antagonistas de Narcóticos/farmacología , Neuronas/metabolismo , Técnicas de Cultivo de Órganos , Técnicas de Placa-Clamp , Proteínas RGS/farmacología , ARN Mensajero/análisis , Ratas , Ratas Sprague-Dawley , Síndrome de Abstinencia a Sustancias/fisiopatología
20.
Biosci Rep ; 23(4): 213-24, 2003 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-14748540

RESUMEN

Regulator of G protein signaling (RGS) proteins are GTPase-activating proteins (GAP) for G protein alpha-subunits and are thought to be responsible for rapid deactivation of G protein mediated signaling pathway. In this present study, we demonstrate that PA is the most efficient candidate to inhibit GAP activity of RGS4. The functional significance of N-terminus of RGS4 in respose to PA-granted inhibition on GAP activity has been studied with the site mutation in the N-terminus of RGS4. These site-directed mutations in the N-terminal domain do not severely disrupt its association with liposomes of PA. However, RGS4L23E diminishes the inhibition of GAP activity by PA compared with the wild type RGS4, whereas RGSR22E abrogates the inhibitory effect by PA on GAP activity. The correspondent conformational discrepancy in the RGS domain of these mutants in the presence of PA vesicles was detected from fluorescence experiments. It is suggested that the functional pertinence between the N-terminus and RGS domain may be important to modulate PA-conferred inhibitory effect on its GAP activity.


Asunto(s)
Proteínas Activadoras de GTPasa/metabolismo , Mutación , Ácidos Fosfatidicos/farmacología , Proteínas RGS/genética , Proteínas RGS/metabolismo , Membrana Celular/metabolismo , Fluorescencia , Proteínas Activadoras de GTPasa/efectos de los fármacos , Conformación Proteica , Estructura Terciaria de Proteína , Proteínas RGS/química , Proteínas RGS/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...