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










Base de dados
Intervalo de ano de publicação
1.
Obesity (Silver Spring) ; 21(7): 1406-15, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23671037

RESUMO

OBJECTIVE: Investigation was conducted to understand the mechanism of action of diacylglycerol acyltransferase 1 (DGAT1) using small molecules DGAT1 inhibitors, compounds K and L. DESIGN AND METHODS: Biochemical and stable-label tracer approaches were applied to interrogate the functional activities of compounds K and L on TG synthesis and changes of carbon flow. Energy homeostasis and gut peptide release upon DGAT1 inhibition was conducted in mouse and dog models. RESULTS: Compounds K and L, dose-dependently inhibits post-prandial TG excursion in mouse and dog models. Weight loss studies in WT and Dgat1(-/-) mice, confirmed that the effects of compound K on body weight loss is mechanism-based. Compounds K and L altered incretin peptide release following oral fat challenge. Immunohistochemical studies with intestinal tissues demonstrate lack of detectable DGAT1 immunoreactivity in enteroendocrine cells. Furthermore, (13) C-fatty acid tracing studies indicate that compound K inhibition of DGAT1 increased the production of phosphatidyl choline (PC). CONCLUSION: Treatment with DGAT1 inhibitors improves lipid metabolism and body weight. DGAT1 inhibition leads to enhanced PC production via alternative carbon channeling. Immunohistological studies suggest that DGAT1 inhibitor's effects on plasma gut peptide levels are likely via an indirect mechanism. Overall these data indicate a translational potential towards the clinic.


Assuntos
Peso Corporal/efeitos dos fármacos , Diacilglicerol O-Aciltransferase/metabolismo , Trato Gastrointestinal/efeitos dos fármacos , Animais , Composição Corporal , Cromatografia Líquida , Diacilglicerol O-Aciltransferase/antagonistas & inibidores , Diacilglicerol O-Aciltransferase/genética , Modelos Animais de Doenças , Cães , Células Enteroendócrinas/efeitos dos fármacos , Células Enteroendócrinas/metabolismo , Fezes/química , Trato Gastrointestinal/metabolismo , Ginsenosídeos/farmacologia , Células HT29 , Hormônios/metabolismo , Humanos , Imuno-Histoquímica , Lactonas/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Orlistate , Período Pós-Prandial/efeitos dos fármacos , Espectrometria de Massas em Tandem , Triglicerídeos/sangue
2.
PLoS One ; 6(11): e27984, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22132185

RESUMO

Regulator of G protein signaling 9-2 (RGS9-2) is a protein that is highly enriched in the striatum, a brain region that mediates motivation, movement and reward responses. We identified a naturally occurring 5 nucleotide deletion polymorphism in the human RGS9 gene and found that the mean body mass index (BMI) of individuals with the deletion was significantly higher than those without. A splicing reporter minigene assay demonstrated that the deletion had the potential to significantly decrease the levels of correctly spliced RGS9 gene product. We measured the weights of rats after virally transduced overexpression of RGS9-2 or the structurally related RGS proteins, RGS7, or RGS11, in the nucleus accumbens (NAc) and observed a reduction in body weight after overexpression of RGS9-2 but not RGS7 or 11. Conversely, we found that the RGS9 knockout mice were heavier than their wild-type littermates and had significantly higher percentages of abdominal fat. The constituent adipocytes were found to have a mean cross-sectional area that was more than double that of corresponding cells from wild-type mice. However, food intake and locomotion were not significantly different between the two strains. These studies with humans, rats and mice implicate RGS9-2 as a factor in regulating body weight.


Assuntos
Peso Corporal/genética , Estudos de Associação Genética , Proteínas RGS/genética , Adipócitos/metabolismo , Adipócitos/patologia , Tecido Adiposo/patologia , Animais , Sequência de Bases , Índice de Massa Corporal , Feminino , Genes Reporter/genética , Humanos , Gordura Intra-Abdominal/metabolismo , Gordura Intra-Abdominal/patologia , Íntrons/genética , Masculino , Camundongos , Camundongos Knockout , Dados de Sequência Molecular , Atividade Motora , Núcleo Accumbens/metabolismo , Splicing de RNA/genética , Ratos , Deleção de Sequência/genética , Redução de Peso/genética
3.
J Neurosci ; 31(15): 5617-24, 2011 Apr 13.
Artigo em Inglês | MEDLINE | ID: mdl-21490202

RESUMO

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.


Assuntos
Analgésicos Opioides/farmacologia , Corpo Estriado/fisiologia , Proteínas RGS/fisiologia , Animais , Arrestinas/genética , Arrestinas/fisiologia , Western Blotting , Corpo Estriado/efeitos dos fármacos , Tolerância a Medicamentos , MAP Quinases Reguladas por Sinal Extracelular/genética , MAP Quinases Reguladas por Sinal Extracelular/fisiologia , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/genética , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/metabolismo , Imunoprecipitação , Camundongos , Camundongos Knockout , Morfina/farmacologia , Fosfolipase C beta/metabolismo , Fosforilação , Proteínas RGS/efeitos dos fármacos , Proteínas RGS/genética , Receptores Opioides mu/agonistas , Receptores Opioides mu/genética , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , beta-Arrestinas
4.
Invest Ophthalmol Vis Sci ; 51(2): 686-93, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19797210

RESUMO

PURPOSE: In the Gbeta5(-/-) mouse, the electroretinogram (ERG) b-wave is absent, and the R7 subfamily of regulators of G protein signaling (RGS), which includes RGS6, -7, -9, and -11, is downregulated. Mutant mouse strains deficient in RGS7 or -11 were characterized, and the SG711 strain which is deficient in both proteins was examined, to learn whether the loss of some of these RGS proteins causes the absence of the ERG b-wave. METHODS: Antibodies to RGS7 and -11 were generated to determine their expression levels and localizations in retinas with various genetic backgrounds by Western blot analysis and immunohistochemistry, respectively. The implicit times and amplitudes of ERG a- and b-waves were analyzed to examine photoreceptor and bipolar cell functions. RESULTS: RGS7 and -11 co-localized to the dendritic tips of the ON-bipolar cells. In the RGS11(-/-) mouse, the level of RGS7 protein increased. However, the level of RGS11 protein remained unchanged in the RGS7 mutant mouse, where a truncated RGS7 protein was expressed due to the deletion of exon 10. In the SG711 mouse retina, the Gbeta5-S protein level was reduced. The ERG b-wave of SG711 mice was markedly delayed. In contrast, RGS11(-/-) mice showed a moderately delayed b-wave, whereas the RGS7 mutant mice showed normal ERG responses. CONCLUSIONS: The data demonstrate the presence of a delayed ERG b-wave in SG711 mice and a functionally redundant role for RGS11 and -7 at the tips of ON-bipolar cell dendrites. These results suggest that RGS11 or -7 works as the major physiological GAP (GTPase acceleration protein) for Galphao1 in ON-bipolar cells.


Assuntos
Dendritos/fisiologia , Proteínas RGS/fisiologia , Células Bipolares da Retina/fisiologia , Animais , Western Blotting , Eletrorretinografia , Técnica Indireta de Fluorescência para Anticorpo , Subunidades beta da Proteína de Ligação ao GTP/genética , Proteínas Ativadoras de GTPase/metabolismo , Genótipo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Mutantes , Estimulação Luminosa , Células Fotorreceptoras Retinianas Bastonetes/fisiologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Regulação para Cima , Visão Ocular/fisiologia
5.
J Neurosci ; 28(34): 8517-28, 2008 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-18716210

RESUMO

Epidemiological studies suggest that chronic use of nonsteroidal anti-inflammatory drugs lowers the incidence of Parkinson's disease (PD) in humans and implicate neuroinflammatory processes in the death of dopamine (DA) neurons. Here, we demonstrate that regulator of G-protein signaling 10 (RGS10), a microglia-enriched GAP (GTPase accelerating protein) for Galpha subunits, is an important regulator of microglia activation. Flow-cytometric and immunohistochemical analyses indicated that RGS10-deficient mice displayed increased microglial burden in the CNS, and exposure to chronic systemic inflammation induced nigral DA neuron loss measured by unbiased stereology. Primary microglia isolated from brains of RGS10-deficient mice displayed dysregulated inflammation-related gene expression profiles under basal and stimulated conditions in vitro compared with that of primary microglia isolated from wild-type littermates. Similarly, knockdown of RGS10 in the BV2 microglia cell line resulted in dysregulated inflammation-related gene expression, overproduction of tumor necrosis factor (TNF), and enhanced neurotoxic effects of BV2 microglia on the MN9D dopaminergic cell line that could be blocked by addition of the TNF decoy receptor etanercept. Importantly, ablation of RGS10 in MN9D dopaminergic cells further enhanced their vulnerability to microglial-derived death-inducing inflammatory mediators, suggesting a role for RGS10 in modulating the sensitivity of dopaminergic neurons against inflammation-mediated cell death. Together, our findings indicate that RGS10 limits microglial-derived TNF secretion and regulates the functional outcome of inflammatory stimuli in the ventral midbrain. RGS10 emerges as a novel drug target for prevention of nigrostriatal pathway degeneration, the neuropathological hallmark of PD.


Assuntos
Dopamina/metabolismo , Inflamação/fisiopatologia , Mesencéfalo/fisiopatologia , Microglia , Neurônios , Proteínas RGS/metabolismo , Animais , Encéfalo/patologia , Linhagem Celular , Sobrevivência Celular , Corpo Estriado/metabolismo , Corpo Estriado/patologia , Citocinas/biossíntese , Suscetibilidade a Doenças , Citometria de Fluxo , Regulação da Expressão Gênica , Imuno-Histoquímica , Inflamação/complicações , Inflamação/patologia , Mediadores da Inflamação/metabolismo , Mesencéfalo/metabolismo , Mesencéfalo/patologia , Camundongos , Camundongos Knockout , Microglia/efeitos dos fármacos , Microglia/metabolismo , Degeneração Neural/etiologia , Neurônios/metabolismo , Proteínas RGS/deficiência , Substância Negra/metabolismo , Substância Negra/patologia , Fator de Necrose Tumoral alfa/metabolismo
6.
Eur J Neurosci ; 27(9): 2433-43, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18461718

RESUMO

Chronic stress exposure alters the central noradrenergic neurons originating from the locus coeruleus (LC). Previously, we demonstrated that evoked increases in the firing rate of LC neurons and their release of norepinephrine are enhanced following chronic cold exposure. In the present studies, we tested the hypothesis that reduced feedback inhibition of LC neurons might underlie these alterations in LC activity by examining the effect of alpha(2)-autoreceptor stimulation on LC activity in chronically stressed rats using in vivo and in vitro single unit recordings. Given that regulators of G-protein signaling (RGS) proteins can impact the coupling of alpha(2)-autoreceptors to downstream signaling cascades, we also explored the expression of several RGS proteins following chronic stress exposure. We observed that the alpha(2)-autoreceptor-evoked inhibition of LC neurons was reduced and that the expression of RGS7 was increased following chronic stress exposure. Finally, we demonstrated that intracellular administration of RGS7 via patch clamp electrodes mimicked the stress-induced decrease in clonidine-evoked autoreceptor-mediated inhibition. These novel data provide a mechanism to explain how chronic stress-induced alterations in receptor coupling can result in changes in alpha(2)-autoreceptor control of noradrenergic function throughout the central nervous system, potentially leading to alterations in anxiety-related behaviors, and may suggest novel therapeutic targets for the treatment of mood and anxiety disorders.


Assuntos
Locus Cerúleo/metabolismo , Neurônios/metabolismo , Proteínas RGS/biossíntese , Receptores Adrenérgicos alfa 2/metabolismo , Estresse Psicológico/fisiopatologia , Animais , Autorreceptores/metabolismo , Western Blotting , Temperatura Baixa , Eletrofisiologia , Expressão Gênica , Masculino , Norepinefrina/metabolismo , Ratos , Ratos Sprague-Dawley , Tempo
7.
Brain Res ; 1190: 78-85, 2008 Jan 23.
Artigo em Inglês | MEDLINE | ID: mdl-18073128

RESUMO

RGS9-2 is a striatum-enriched protein that negatively modulates dopamine and opioid receptor signaling. We examined the role of RGS9-2 in modulating complex behavior. Genetic deletion of RGS9-2 does not lead to global impairments, but results in selective abnormalities in certain behavioral domains. RGS9 knockout (KO) mice have decreased motor coordination on the accelerating rotarod and deficits in working memory as measured in the delayed-match-to-place version of the water maze. In contrast, RGS9 KO mice exhibit normal locomotor activity, anxiety-like behavior, cue and contextual fear conditioning, startle threshold, and pre-pulse inhibition. These studies are the first to describe a role for RGS9-2 in motor coordination and working memory and implicate RGS9-2 as a potential therapeutic target for motor and cognitive dysfunction.


Assuntos
Ataxia/metabolismo , Comportamento Animal/fisiologia , Aprendizagem em Labirinto/fisiologia , Proteínas RGS/metabolismo , Reflexo de Sobressalto/fisiologia , Animais , Ataxia/genética , Condicionamento Clássico/fisiologia , Inibição Psicológica , Masculino , Análise por Pareamento , Memória de Curto Prazo/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Atividade Motora/fisiologia , Proteínas RGS/genética , Reflexo de Sobressalto/genética , Teste de Desempenho do Rota-Rod , Método Simples-Cego
8.
J Neurosci ; 27(52): 14338-48, 2007 Dec 26.
Artigo em Inglês | MEDLINE | ID: mdl-18160641

RESUMO

Chronic L-dopa treatment of Parkinson's disease (PD) often leads to debilitating involuntary movements, termed L-dopa-induced dyskinesia (LID), mediated by dopamine (DA) receptors. RGS9-2 is a GTPase accelerating protein that inhibits DA D2 receptor-activated G proteins. Herein, we assess the functional role of RGS9-2 on LID. In monkeys, Western blot analysis of striatal extracts shows that RGS9-2 levels are not altered by MPTP-induced DA denervation and/or chronic L-dopa administration. In MPTP monkeys with LID, striatal RGS9-2 overexpression--achieved by viral vector injection into the striatum--diminishes the involuntary movement intensity without lessening the anti-parkinsonian effects of the D1/D2 receptor agonist L-dopa. In contrasts, in these animals, striatal RGS9-2 overexpression diminishes both the involuntary movement intensity and the anti-parkinsonian effects of the D2/D3 receptor agonist ropinirole. In unilaterally 6-OHDA-lesioned rats with LID, we show that the time course of viral vector-mediated striatal RGS9-2 overexpression parallels the time course of improvement of L-dopa-induced involuntary movements. We also find that unilateral 6-OHDA-lesioned RGS9-/- mice are more susceptible to L-dopa-induced involuntary movements than unilateral 6-OHDA-lesioned RGS9+/+ mice, albeit the rotational behavior--taken as an index of the anti-parkinsonian response--is similar between the two groups of mice. Together, these findings suggest that RGS9-2 plays a pivotal role in LID pathophysiology. However, the findings also suggest that increasing RGS9-2 expression and/or function in PD patients may only be a suitable therapeutic strategy to control involuntary movements induced by nonselective DA agonist such as L-dopa.


Assuntos
Di-Hidroxifenilalanina/efeitos adversos , Dopaminérgicos/efeitos adversos , Discinesias/etiologia , Discinesias/fisiopatologia , Proteínas RGS/metabolismo , Comportamento Estereotipado/fisiologia , Análise de Variância , Animais , Comportamento Animal/efeitos dos fármacos , Comportamento Animal/fisiologia , Corpo Estriado/metabolismo , Corpo Estriado/fisiopatologia , Modelos Animais de Doenças , Discinesias/terapia , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/fisiologia , Intoxicação por MPTP/tratamento farmacológico , Macaca fascicularis , Camundongos , Camundongos Knockout , Oxidopamina/farmacologia , Proteínas RGS/administração & dosagem , Comportamento Estereotipado/efeitos dos fármacos , Simpatolíticos/farmacologia
9.
J Neurochem ; 103(2): 617-25, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17725581

RESUMO

Regulators of G-protein signaling (RGS) 9-2 is a striatal enriched protein that controls G protein coupled receptor signaling duration by accelerating Galpha subunit guanosine triphosphate hydrolysis. We have previously demonstrated that mice lacking the RGS9 gene show enhanced morphine analgesia and delayed development of tolerance. Here we extend these studies to understand the mechanism via which RGS9-2 modulates opiate actions. Our data suggest that RGS9-2 prevents several events triggered by mu-opioid receptor (MOR) activation. In transiently transfected PC12 cells, RGS9-2 delays agonist induced internalization of epitope HA-tagged mu-opioid receptor. This action of RGS9-2 requires localization of the protein near the cell membrane. Co-immunoprecipitation studies reveal that RGS9-2 interacts with HA-tagged mu-opioid receptor, and that this interaction is enhanced by morphine treatment. In addition, morphine promotes the association of RGS9-2 with another essential component of MOR desensitization, beta-arrestin-2. We also show that over-expression of RGS9-2 prevents opiate-induced extracellular signal-regulated kinase phosphorylation. Our data indicate that RGS9-2 plays an essential role in opiate actions, by negatively modulating MOR downstream signaling as well as the rate of MOR endocytosis.


Assuntos
Proteínas RGS/fisiologia , Receptores Opioides mu/fisiologia , Animais , Arrestinas/metabolismo , Comportamento Animal/fisiologia , Western Blotting , Química Encefálica/efeitos dos fármacos , Células Cultivadas , Endocitose/efeitos dos fármacos , Ensaio de Imunoadsorção Enzimática , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Imunofluorescência , Imuno-Histoquímica , Imunoprecipitação , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Morfina/farmacologia , Entorpecentes/farmacologia , Medição da Dor/efeitos dos fármacos , Fosforilação , Receptores Opioides mu/agonistas , beta-Arrestina 2 , beta-Arrestinas
10.
Comp Hepatol ; 5: 8, 2006 Nov 23.
Artigo em Inglês | MEDLINE | ID: mdl-17123436

RESUMO

BACKGROUND: Heterotrimeric G protein signaling in liver helps maintain carbohydrate and lipid homeostasis. G protein signaling is activated by binding of extracellular ligands to G protein coupled receptors and inhibited inside cells by regulators of G protein signaling (RGS) proteins. RGS proteins are GTPase activating proteins, and thereby regulate Gi and/or Gq class G proteins. RGS gene expression can be induced by the ligands they feedback regulate, and RGS gene expression can be used to mark tissues and cell-types when and where Gi/q signaling occurs. We characterized the expression of mouse RGS genes in liver during fasting and refeeding to identify novel signaling pathways controlling changes in liver metabolism. RESULTS: Rgs16 is the only RGS gene that is diurnally regulated in liver of ad libitum fed mice. Rgs16 transcription, mRNA and protein are up regulated during fasting and rapidly down regulated after refeeding. Rgs16 is expressed in periportal hepatocytes, the oxygen-rich zone of the liver where lipolysis and gluconeogenesis predominates. Restricting feeding to 4 hr of the light phase entrained Rgs16 expression in liver but did not affect circadian regulation of Rgs16 expression in the suprachiasmatic nuclei (SCN). CONCLUSION: Rgs16 is one of a subset of genes that is circadian regulated both in SCN and liver. Rgs16 mRNA expression in liver responds rapidly to changes in feeding schedule, coincident with key transcription factors controlling the circadian clock. Rgs16 expression can be used as a marker to identify and investigate novel G-protein mediated metabolic and circadian pathways, in specific zones within the liver.

11.
J Neurochem ; 96(6): 1606-15, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16539683

RESUMO

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.


Assuntos
Anfetamina/farmacologia , Dopamina/metabolismo , Regulação para Baixo/efeitos dos fármacos , Prosencéfalo/efeitos dos fármacos , Proteínas RGS/efeitos dos fármacos , Receptores Dopaminérgicos/metabolismo , Transtornos Relacionados ao Uso de Anfetaminas/genética , Transtornos Relacionados ao Uso de Anfetaminas/metabolismo , Transtornos Relacionados ao Uso de Anfetaminas/fisiopatologia , Animais , Benzazepinas/farmacologia , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/metabolismo , Modelos Animais de Doenças , Antagonistas de Dopamina/farmacologia , Relação Dose-Resposta a Droga , Regulação para Baixo/fisiologia , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/metabolismo , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/metabolismo , Masculino , Neostriado/efeitos dos fármacos , Neostriado/metabolismo , Prosencéfalo/metabolismo , Prosencéfalo/fisiopatologia , Proteínas RGS/genética , Proteínas RGS/metabolismo , RNA Mensageiro/efeitos dos fármacos , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores Dopaminérgicos/efeitos dos fármacos , Receptores de Dopamina D1/efeitos dos fármacos , Receptores de Dopamina D1/metabolismo , Receptores de Dopamina D2/efeitos dos fármacos , Receptores de Dopamina D2/metabolismo , Salicilamidas/farmacologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia
12.
J Comp Neurol ; 481(3): 299-313, 2005 Jan 17.
Artigo em Inglês | MEDLINE | ID: mdl-15593368

RESUMO

The regulator of G protein signaling type 10 (RGS10) modulates Galphai/o signaling by means of its GTPase accelerating activity and is abundantly expressed in brain and in immune tissues. To elucidate RGS10 function in the nervous system, we mapped RGS10 protein in rat and mouse brain using light microscopic (LM) and electron microscopic (EM) immunohistochemical techniques. The LM showed that RGS10-like immunoreactivity (LIR) labels all cellular subcompartments of neurons and microglia, including their nuclei. There were several differences between RGS10-LIR distributions in rat and mouse, the most striking of which were the far denser immunoreactivity in rat dentate gyrus and dorsal raphe. The EM analysis corroborated and extended our findings from LM. Thus, EM confirmed the presence of dense RGS10-LIR in the euchromatin compartment of nuclei. The EM analysis also resolved dense staining on terminals at symmetric synapses onto pyramidal cell somata. Dual immunofluorescence showed that forebrain interneurons densely labeled with RGS10-LIR partially colocalized with parvalbumin-LIR. Dual-labeling histochemistry in caudoputamen demonstrated that densely labeled striatal cells were biased to the indirect-projecting output pathway. Dual-labeling immunofluorescence also showed that densely labeled RGS10-LIR cells in the dentate gyrus subgranular zone were not proliferating but that newly born cells could differentiate to express RGS10-LIR. Taken together, these data support a role for RGS10 in diverse processes that include modulation of pre- and postsynaptic G-protein signaling. Moreover, enrichment of RGS10 in transcriptionally active regions of the nucleus suggests an unforeseen role of RGS10 in modulating gene expression.


Assuntos
Encéfalo/metabolismo , Camundongos Endogâmicos C57BL/metabolismo , Neurônios/metabolismo , Proteínas RGS/metabolismo , Ratos Sprague-Dawley/metabolismo , Sinapses/metabolismo , Animais , Encéfalo/ultraestrutura , Diferenciação Celular/fisiologia , Núcleo Celular/metabolismo , Núcleo Celular/ultraestrutura , Giro Denteado/metabolismo , Giro Denteado/ultraestrutura , Proteínas de Ligação ao GTP/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Endogâmicos C57BL/anatomia & histologia , Microglia/metabolismo , Microglia/ultraestrutura , Microscopia Eletrônica de Transmissão , Neurônios/ultraestrutura , Parvalbuminas/metabolismo , Células Piramidais/metabolismo , Células Piramidais/ultraestrutura , Núcleos da Rafe/metabolismo , Núcleos da Rafe/ultraestrutura , Ratos , Ratos Sprague-Dawley/anatomia & histologia , Transdução de Sinais/fisiologia , Especificidade da Espécie , Células-Tronco/metabolismo , Células-Tronco/ultraestrutura , Sinapses/ultraestrutura
13.
Methods Enzymol ; 389: 205-29, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15313568

RESUMO

To elucidate the functional role of regulators of G-protein signaling (RGS) in vivo, it will be critical to (i) determine how RGS activity is altered in response to a variety of manipulations and (ii) observe how the system is changed when RGS protein function is altered genetically. To facilitate studies of dynamic regulation of RGS protein activity, this article describes detailed methods for radioisotopic in situ hybridization for semiquantitative analyses of RGS mRNA abundances. Toward characterizing the functional differences in mice with genetically altered RGS activities, this article describes a subset of behavioral tests suitable for assaying sensitivities to drugs of abuse. These protocols should provide valuable guidance for investigators to establish these methodologies independently in their own laboratories and, over time, increase our understanding of RGS function in vivo.


Assuntos
Comportamento Animal/efeitos dos fármacos , Drogas Ilícitas/metabolismo , Hibridização In Situ , Fenótipo , Proteínas RGS/genética , RNA Mensageiro/análise , Animais , Ansiedade/induzido quimicamente , Encéfalo/metabolismo , Avaliação Pré-Clínica de Medicamentos , Regulação da Expressão Gênica , Aprendizagem/efeitos dos fármacos , Camundongos , Camundongos Mutantes , Camundongos Transgênicos , Morfina/metabolismo , Atividade Motora/efeitos dos fármacos
14.
J Biol Chem ; 279(4): 2593-9, 2004 Jan 23.
Artigo em Inglês | MEDLINE | ID: mdl-14604980

RESUMO

Regulators of G protein signaling (RGS proteins) constitute a family of newly appreciated components of G protein-mediated signal transduction. With few exceptions, most information available on mammalian RGS proteins was gained by transfection/overexpression or in vitro experiments, with relatively little known about the endogenous counterparts. Transfection studies, typically of tagged RGS proteins, have been conducted to overcome the low natural abundance of endogenous RGS proteins. Because transfection studies can lead to imprecise or erroneous conclusions, we have developed antibodies of high specificity and sensitivity to focus study on endogenous proteins. Expression of both RGS4 and RGS7 was detected in rat brain tissue and cultured PC12 and AtT-20 cells. Endogenous RGS4 presented as a single 27-28-kDa protein. By contrast, cultured cells transfected with a plasmid encoding RGS4 expressed two observable forms of the protein, apparently due to utilization of distinct sites of initiation of protein synthesis. Subcellular localization of endogenous RGS4 revealed predominant association with membrane fractions, rather than in cytosolic fractions, where most heterologously expressed RGS4 has been found. Endogenous levels of RGS7 exceeded RGS4 by 30-40-fold, and studies of cultured cells revealed regulatory differences between the two proteins. We observed that RGS4 mRNA and protein were concomitantly augmented with increased cell density and decreased by exposure of PC12M cells to nerve growth factor, whereas RGS7 was unaffected. Endogenous RGS7 was relatively stable, whereas proteolysis of endogenous RGS4 was a strong determinant of its lower level expression and short half-life. Although we searched without finding evidence for regulation of RGS4 proteolysis, the possibility remains that alterations in the degradation of this protein could provide a means to promptly alter patterns of signal transduction.


Assuntos
Proteínas de Ligação ao GTP/biossíntese , Proteínas RGS/biossíntese , Animais , Encéfalo/metabolismo , Linhagem Celular , Regulação da Expressão Gênica , Humanos , Ratos , Transdução de Sinais
15.
Proc Natl Acad Sci U S A ; 100(23): 13656-61, 2003 Nov 11.
Artigo em Inglês | MEDLINE | ID: mdl-14595021

RESUMO

Regulators of G protein signaling (RGS) are a family of proteins known to accelerate termination of effector stimulation after G protein receptor activation. RGS9-2, a brain-specific splice variant of the RGS9 gene, is highly enriched in striatum and also expressed at much lower levels in periaqueductal gray and spinal cord, structures known to mediate various actions of morphine and other opiates. Morphine exerts its acute rewarding and analgesic effects by activation of inhibitory guanine nucleotide-binding regulatory protein-coupled opioid receptors, whereas chronic morphine causes addiction, tolerance to its acute analgesic effects, and profound physical dependence by sustained activation of these receptors. We show here that acute morphine administration increases expression of RGS9-2 in NAc and the other CNS regions, whereas chronic exposure decreases RGS9-2 levels. Mice lacking RGS9 show enhanced behavioral responses to acute and chronic morphine, including a dramatic increase in morphine reward, increased morphine analgesia with delayed tolerance, and exacerbated morphine physical dependence and withdrawal. These findings establish RGS9 as a potent negative modulator of opiate action in vivo, and suggest that opiate-induced changes in RGS9 levels contribute to the behavioral and neural plasticity associated with chronic opiate administration.


Assuntos
Entorpecentes/metabolismo , Proteínas RGS/fisiologia , Processamento Alternativo , Analgésicos Opioides/farmacologia , Animais , Western Blotting , Sistema Nervoso Central/metabolismo , Relação Dose-Resposta a Droga , Técnicas de Transferência de Genes , Imuno-Histoquímica , Hibridização In Situ , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Morfina/metabolismo , Morfina/farmacologia , Ratos , Ratos Sprague-Dawley , Medula Espinal/patologia , Temperatura , Fatores de Tempo
16.
Neuron ; 38(6): 941-52, 2003 Jun 19.
Artigo em Inglês | MEDLINE | ID: mdl-12818179

RESUMO

Regulators of G protein signaling (RGS) modulate heterotrimeric G proteins in part by serving as GTPase-activating proteins for Galpha subunits. We examined a role for RGS9-2, an RGS subtype highly enriched in striatum, in modulating dopamine D2 receptor function. Viral-mediated overexpression of RGS9-2 in rat nucleus accumbens (ventral striatum) reduced locomotor responses to cocaine (an indirect dopamine agonist) and to D2 but not to D1 receptor agonists. Conversely, RGS9 knockout mice showed heightened locomotor and rewarding responses to cocaine and related psychostimulants. In vitro expression of RGS9-2 in Xenopus oocytes accelerated the off-kinetics of D2 receptor-induced GIRK currents, consistent with the in vivo data. Finally, chronic cocaine exposure increased RGS9-2 levels in nucleus accumbens. Together, these data demonstrate a functional interaction between RGS9-2 and D2 receptor signaling and the behavioral actions of psychostimulants and suggest that psychostimulant induction of RGS9-2 represents a compensatory adaptation that diminishes drug responsiveness.


Assuntos
Dopamina/metabolismo , Canais de Potássio Corretores do Fluxo de Internalização , Proteínas RGS/fisiologia , Transdução de Sinais , Animais , Comportamento Animal/efeitos dos fármacos , Estimulantes do Sistema Nervoso Central/farmacologia , Cocaína/farmacologia , Corpo Estriado/fisiologia , Condutividade Elétrica , Feminino , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G , Expressão Gênica , Masculino , Camundongos , Camundongos Knockout , Atividade Motora/efeitos dos fármacos , Núcleo Accumbens/química , Oócitos/metabolismo , Canais de Potássio/fisiologia , Proteínas RGS/análise , Proteínas RGS/deficiência , Proteínas RGS/genética , Ratos , Ratos Sprague-Dawley , Receptores de Dopamina D2/fisiologia , Transfecção , Xenopus
17.
Eur J Neurosci ; 17(5): 971-80, 2003 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-12653973

RESUMO

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.


Assuntos
Locus Cerúleo/efeitos dos fármacos , Morfina/farmacologia , Entorpecentes/farmacologia , Neurônios/efeitos dos fármacos , Proteínas RGS/efeitos dos fármacos , Animais , Western Blotting , Imuno-Histoquímica , Hibridização In Situ , Locus Cerúleo/metabolismo , Masculino , Potenciais da Membrana/efeitos dos fármacos , Naltrexona/farmacologia , Antagonistas de Entorpecentes/farmacologia , Neurônios/metabolismo , Técnicas de Cultura de Órgãos , Técnicas de Patch-Clamp , Proteínas RGS/farmacologia , RNA Mensageiro/análise , Ratos , Ratos Sprague-Dawley , Síndrome de Abstinência a Substâncias/fisiopatologia
18.
J Neurochem ; 82(4): 828-38, 2002 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12358788

RESUMO

G protein-coupled receptor (GPCR) signaling cascades may be key substrates for the antidepressant effects of chronic electroconvulsive seizures (ECS). To better understand changes in these signaling pathways, alterations in levels of mRNA's encoding regulators of G protein signaling (RGS) protein subtypes-2, -4, -7, -8 and -10 were evaluated in rat brain using northern blotting and in situ hybridization. In prefrontal cortex, RGS2 mRNA levels were increased several-fold 2 h following an acute ECS. Increases in RGS8 mRNA were of lesser magnitude (30%), and no changes were evident for the other RGS subtypes. At 24 h following a chronic ECS regimen, RGS4, -7, and -10 mRNA levels were reduced by 20-30%; only RGS10 was significantly reduced 24 h after acute ECS. Levels of RGS2 mRNA were unchanged 24 h following either acute or chronic ECS. In hippocampus, RGS2 mRNA levels were markedly increased 2 h following acute ECS. More modest increases were seen for RGS4 mRNA expression, whereas levels of the other RGS subtypes were unaltered. At 24 h following chronic ECS, RGS7, -8 and -10 mRNA levels were decreased in the granule cell layer, and RGS7 and -8 mRNA levels were decreased in the pyramidal cell layers. Only RGS8 and -10 mRNA levels were significantly reduced in hippocampus 24 h following an acute ECS. Paralleling neocortex, RGS2 mRNA content was unchanged in hippocampus 24 h following either acute or chronic ECS. In ventromedial hypothalamus, RGS4 mRNA content was increased 24 h following chronic ECS, whereas RGS7 mRNA levels were only increased 24 h following an acute ECS. The increased RGS4 mRNA levels in hypothalamus were significant by 2 h following an acute ECS. These studies demonstrate subtype-, time-, and region-specific regulation of RGS proteins by ECS, adaptations that may contribute to the antidepressant effects of this treatment.


Assuntos
Encéfalo/metabolismo , Regulação da Expressão Gênica , Proteínas RGS/metabolismo , RNA Mensageiro/metabolismo , Convulsões/metabolismo , Doença Aguda , Animais , Northern Blotting , Encéfalo/patologia , Doença Crônica , Densitometria , Eletrochoque , Hibridização In Situ , Masculino , Proteínas RGS/genética , Ratos , Ratos Sprague-Dawley , Transdução de Sinais
19.
J Neurosci ; 22(10): 4153-62, 2002 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-12019333

RESUMO

Chronic opiate exposure induces numerous neurochemical adaptations in the noradrenergic system, including upregulation of the cAMP-signaling pathway and increased expression of tyrosine hydroxylase (TH), the rate-limiting enzyme in catecholamine biosynthesis. These adaptations are thought to compensate for opiate-mediated neuronal inhibition but also contribute to physical dependence, including withdrawal after abrupt cessation of drug exposure. Little is known about molecules that regulate the noradrenergic response to opiates. Here we report that noradrenergic locus ceruleus (LC) neurons of mice with a conditional deletion of BDNF in postnatal brain respond to chronic morphine treatment with a paradoxical downregulation of cAMP-mediated excitation and lack of dynamic regulation of TH expression. This was accompanied by a threefold reduction in opiate withdrawal symptoms despite normal antinociceptive tolerance in the BDNF-deficient mice. Although expression of TrkB, the receptor for BDNF, was high in the LC, endogenous BDNF expression was absent there and in the large majority of other noradrenergic neurons. Therefore, a BDNF-signaling pathway originating from non-noradrenergic sources is essential for opiate-induced molecular adaptations of the noradrenergic system.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/metabolismo , Entorpecentes/farmacologia , Plasticidade Neuronal/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Neurônios/fisiologia , Animais , Comportamento Animal/fisiologia , Fator Neurotrófico Derivado do Encéfalo/deficiência , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina , Proteínas Quinases Dependentes de Cálcio-Calmodulina/genética , Contagem de Células , Colforsina/farmacologia , AMP Cíclico/metabolismo , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/genética , Deleção de Genes , Genes Reporter , Genótipo , Hibridização In Situ , Técnicas In Vitro , Integrases/genética , Locus Cerúleo/citologia , Locus Cerúleo/efeitos dos fármacos , Locus Cerúleo/fisiologia , Camundongos , Camundongos Mutantes , Camundongos Transgênicos , Entorpecentes/efeitos adversos , Plasticidade Neuronal/fisiologia , Neurônios/metabolismo , Norepinefrina/metabolismo , Receptores Opioides mu/agonistas , Recombinação Genética , Índice de Gravidade de Doença , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Síndrome de Abstinência a Substâncias , Transgenes , Tirosina 3-Mono-Oxigenase/metabolismo , Proteínas Virais/genética
20.
Neuron ; 34(1): 13-25, 2002 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-11931738

RESUMO

Current treatments for depression are inadequate for many individuals, and progress in understanding the neurobiology of depression is slow. Several promising hypotheses of depression and antidepressant action have been formulated recently. These hypotheses are based largely on dysregulation of the hypothalamic-pituitary-adrenal axis and hippocampus and implicate corticotropin-releasing factor, glucocorticoids, brain-derived neurotrophic factor, and CREB. Recent work has looked beyond hippocampus to other brain areas that are also likely involved. For example, nucleus accumbens, amygdala, and certain hypothalamic nuclei are critical in regulating motivation, eating, sleeping, energy level, circadian rhythm, and responses to rewarding and aversive stimuli, which are all abnormal in depressed patients. A neurobiologic understanding of depression also requires identification of the genes that make individuals vulnerable or resistant to the syndrome. These advances will fundamentally improve the treatment and prevention of depression.


Assuntos
Depressão , Neurobiologia/métodos , Animais , Depressão/diagnóstico , Depressão/tratamento farmacológico , Depressão/genética , Depressão/metabolismo , Modelos Animais de Doenças , Humanos , Neurobiologia/tendências
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...