Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
PLoS One ; 9(8): e105791, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25162404

RESUMO

G beta 5 (Gbeta5, Gß5) is a unique G protein ß subunit that is thought to be expressed as an obligate heterodimer with R7 regulator of G protein signaling (RGS) proteins instead of with G gamma (Gγ) subunits. We found that D2-dopamine receptor (D2R) coexpression enhances the expression of Gß5, but not that of the G beta 1 (Gß1) subunit, in HEK293 cells, and that the enhancement of expression occurs through a stabilization of Gß5 protein. We had previously demonstrated that the vast majority of D2R either expressed endogenously in the brain or exogenously in cell lines segregates into detergent-resistant biochemical fractions. We report that when expressed alone in HEK293 cells, Gß5 is highly soluble, but is retargeted to the detergent-resistant fraction after D2R coexpression. Furthermore, an in-cell biotin transfer proximity assay indicated that D2R and Gß5 segregating into the detergent-resistant fraction specifically interacted in intact living cell membranes. Dopamine-induced D2R internalization was blocked by coexpression of Gß5, but not Gß1. However, the same Gß5 coexpression levels had no effect on agonist-induced internalization of the mu opioid receptor (MOR), cell surface D2R levels, dopamine-mediated recruitment of ß-arrestin to D2R, the amplitude of D2R-G protein coupling, or the deactivation kinetics of D2R-activated G protein signals. The latter data suggest that the interactions between D2R and Gß5 are not mediated by endogenously expressed R7 RGS proteins.


Assuntos
Dopamina/metabolismo , Subunidades beta da Proteína de Ligação ao GTP/química , Subunidades beta da Proteína de Ligação ao GTP/metabolismo , Receptores de Dopamina D2/metabolismo , Arrestinas/metabolismo , Proteínas de Transporte/metabolismo , Detergentes/farmacologia , Células HEK293 , Humanos , Técnicas In Vitro , Peptídeos e Proteínas de Sinalização Intracelular , Octoxinol/farmacologia , Estabilidade Proteica , Proteínas RGS , Receptores Opioides mu/metabolismo , beta-Arrestinas
2.
J Neurochem ; 127(1): 57-65, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23815307

RESUMO

We reconstituted D2 like dopamine receptor (D2R) and the delta opioid receptor (DOR) coupling to G-protein gated inwardly rectifying potassium channels (K(ir)3) and directly compared the effects of co-expression of G-protein coupled receptor kinase (GRK) and arrestin on agonist-dependent desensitization of the receptor response. We found, as described previously, that co-expression of a GRK and an arrestin synergistically increased the rate of agonist-dependent desensitization of DOR. In contrast, only arrestin expression was required to produce desensitization of D2R responses. Furthermore, arrestin-dependent GRK-independent desensitization of D2R-K(ir)3 coupling could be transferred to DOR by substituting the third cytoplasmic loop of DOR with that of D2R. The arrestin-dependent GRK-independent desensitization of D2R desensitization was inhibited by staurosporine treatment, and blocked by alanine substitution of putative protein kinase C phosphorylation sites in the third cytoplasmic loop of D2R. Finally, the D2R construct in which putative protein kinase C phosphorylation sites were mutated did not undergo significant agonist-dependent desensitization even after GRK co-expression, suggesting that GRK phosphorylation of D2R does not play an important role in uncoupling of the receptor.


Assuntos
Arrestina/fisiologia , Quinases de Receptores Acoplados a Proteína G/metabolismo , Receptores de Dopamina D2/metabolismo , Animais , Arrestinas/fisiologia , Clonagem Molecular , Citoplasma/metabolismo , DNA Complementar/biossíntese , DNA Complementar/genética , Fenômenos Eletrofisiológicos , Inibidores Enzimáticos/farmacologia , Feminino , Humanos , Oócitos/metabolismo , RNA Complementar/biossíntese , RNA Complementar/genética , Estaurosporina/farmacologia , Xenopus , beta-Arrestinas
3.
J Biol Chem ; 288(18): 12554-68, 2013 May 03.
Artigo em Inglês | MEDLINE | ID: mdl-23493394

RESUMO

Plasma membrane microcompartments could allow different signaling pathways to operate more efficiently and prevent cross-talk. We utilized a novel in-cell biotin transfer assay to demonstrate that the majority of plasma membrane-expressed D2 dopamine receptor (D2R) is microcompartmentalized within detergent-resistant structures. Conversely, a minority of D2R existed in a detergent-soluble form and interacted in a relatively unrestricted manner with other cellular proteins. The microcompartmentalization of D2R had functional consequences because dopamine-induced internalization of D2R was largely restricted to the compartmentalized receptor. The D2R-containing microcompartments did not correspond to putative detergent-resistant lipid raft structures. First, the detergent-insoluble D2R structures were significantly denser than detergent-resistant membrane fragments containing flotillin, a widely utilized lipid raft marker protein. Second, the detergent solubility of D2R was unaffected by treatment of cells with the cholesterol chelating agent, methyl-ß-cyclodextrin, that is thought to disrupt lipid rafts. Finally, the in-cell biotinylation assay did not provide any evidence for the membrane compartmentalization of peptide motifs thought to target to lipid rafts. Thus, our observations form one of the first demonstrations, in living cells, of plasma membrane microcompartments defined by the ability of the compartment structure to broadly restrict the interaction of resident molecules with other cellular proteins.


Assuntos
Microdomínios da Membrana/metabolismo , Receptores de Dopamina D2/metabolismo , Motivos de Aminoácidos , Animais , Células HEK293 , Humanos , Microdomínios da Membrana/genética , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Receptores de Dopamina D2/genética , beta-Ciclodextrinas/farmacologia
4.
J Neurochem ; 120(1): 56-69, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22035199

RESUMO

Detergent-resistant membranes (DRM) are thought to contain structures such as lipid rafts that are involved in compartmentalizing cell membranes. We report that the majority of D(2)-dopamine receptors (D(2)R) expressed endogenously in mouse striatum or expressed in immortalized cell-lines is found in DRM. In addition, exogenous co-expression of D(2)R in a cell line shifted the expression of regulator of G protein signaling 9-2 (RGS9-2) into DRM. RGS9-2 is a protein that is highly enriched in the striatum and specifically regulates striatal D(2)R. In the striatum, RGS9-2 is mostly associated with DRMs but when expressed in cell lines, RGS9-2 is present in the soluble cytoplasmic fraction. In contrast, the majority of mu opioid receptors and delta opioid receptors are found in detergent-soluble membrane and there was no shift of RGS9-2 into DRM after co-expression of mu opioid receptor. These data suggest that the targeting of RGS9-2 to DRM in the striatum is mediated by D(2)R and that DRM is involved in the formation of a D(2)R signaling complex. D(2)R-mediated targeting of RGS9-2 to DRM was blocked by the deletion of the RGS9-2 DEP domain or by a point mutation that abolishes the GTPase accelerating protein function of RGS9-2.


Assuntos
Membrana Celular/efeitos dos fármacos , Detergentes/farmacologia , Proteínas RGS/fisiologia , Receptores de Dopamina D2/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Western Blotting , Química Encefálica , Linhagem Celular , Corpo Estriado/metabolismo , DNA Complementar/genética , Subunidades beta da Proteína de Ligação ao GTP/genética , Humanos , Imuno-Histoquímica , Proteínas de Membrana/química , Mutação/genética , Octoxinol , Proteínas RGS/genética , Receptores de Dopamina D2/genética , Receptores Opioides delta/genética , Receptores Opioides mu/genética , Receptores Opioides mu/fisiologia , Solubilidade , Transfecção
5.
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
6.
Neurosci Lett ; 502(3): 123-8, 2011 Sep 20.
Artigo em Inglês | MEDLINE | ID: mdl-21616123

RESUMO

In this study we demonstrate up-regulation of mRNA for Regulator of G protein Signaling (RGS) 6, 7, 9 and 11, R7 family RGS binding protein (R7BP) and RGS9 anchor protein (R9AP) during neuronal differentiation of mouse embryonic stem cells (mESCs). This expression pattern was most robust for RGS9 whose transcript level was low in undifferentiated mESCs but increased over 125 fold when differentiating mESCs began to exhibit a neuronal precursor cell (NPC) phenotype. In addition, we demonstrate that RGS9 mRNA is expressed in neuronal stem cells isolated from embryonic mouse cortex. The expression of RGS9 in two distinct populations of NPCs suggests that RGS9 and its accessory proteins may play an important role in neuron development.


Assuntos
Diferenciação Celular/genética , Células-Tronco Embrionárias/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Células-Tronco Neurais/metabolismo , Proteínas RGS/genética , RNA Mensageiro/biossíntese , Animais , Células Cultivadas , Córtex Cerebral/citologia , Córtex Cerebral/embriologia , Córtex Cerebral/metabolismo , Células-Tronco Embrionárias/citologia , Camundongos , Células-Tronco Neurais/citologia , Proteínas RGS/biossíntese , Regulação para Cima/genética
7.
Biochem Biophys Res Commun ; 405(1): 85-90, 2011 Feb 04.
Artigo em Inglês | MEDLINE | ID: mdl-21215258

RESUMO

There are no pharmacological treatments to rescue botulinum neurotoxin (BoNT)-mediated paralysis of neuromuscular signaling. In part, this failure can be attributed to the lack of a cell culture model system that is neuron-based, allowing detailed elucidation of the mechanisms underlying BoNT pathogenesis, yet still compatible with modern cellular and molecular approaches. We have developed a method to derive highly enriched, glutamatergic neurons from suspension-cultured murine embryonic stem (ES) cells. Hypothesizing that ES cell-derived neurons (ESNs) might comprise a novel platform to investigate the neurotoxicology of BoNTs, we evaluated the susceptibility of ESNs to BoNT/A and BoNT/E using molecular and functional assays. ESNs express neuron-specific proteins, develop synapses and release glutamate in a calcium-dependent manner under depolarizing conditions. They express the BoNT substrate SNARE proteins SNAP25, VAMP2 and syntaxin, and treatment with BoNT/A and BoNT/E holotoxin results in proteolysis of SNAP25 within 24 h with EC50s of 0.81 and 68.6 pM, respectively. Intoxication with BoNT/A results in the functional inhibition of potassium-induced, calcium-dependent glutamate release. ESNs remain viable and susceptible to intoxication for up to 90 days after plating, enabling longitudinal screens exploring toxin-specific mechanisms underlying persistence of synaptic blockade. The evidence suggests that derived neurons are a novel, biologically relevant model system that combines the verisimilitude of primary neurons with the genetic tractability and scalable expansion of a continuous cell line, and thus should significantly accelerate BoNT research and drug discovery while dramatically decreasing animal use.


Assuntos
Toxinas Botulínicas Tipo A/toxicidade , Toxinas Botulínicas/toxicidade , Células-Tronco Embrionárias/citologia , Neurônios/citologia , Neurônios/efeitos dos fármacos , Animais , Cálcio/metabolismo , Exocitose/efeitos dos fármacos , Ácido Glutâmico/metabolismo , Camundongos , Modelos Biológicos , Neurogênese , Biossíntese de Proteínas , Sinapses/efeitos dos fármacos , Sinapses/fisiologia , Técnicas de Cultura de Tecidos
8.
J Neurochem ; 114(3): 739-49, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20477943

RESUMO

Regulator of G protein signaling 9-2 (RGS9-2), a member of the RGS family of GTPase accelerating proteins, is expressed specifically in the striatum, a brain region involved in controlling movement, motivation, mood and addiction. RGS9-2 can be found co-localized with D(2)-class dopamine receptors in medium spiny striatal neurons and altered functioning of both RGS9-2 and D(2)-like dopamine receptors have been implicated in schizophrenia, movement disorders and reward responses. Previously we showed that RGS9-2 can specifically co-localize with D(2)-dopamine receptors (D2R). Here we provide further evidence of the specificity of RGS9-2 for regulating D2R cellular functions: the expression of RGS9-2 inhibits dopamine-mediated cellular internalization of D2R, while the expression of another RGS protein, RGS4, had no effect. In addition, the agonist-mediated internalization of the G protein coupled delta opioid receptor was unaffected by RGS9-2 expression. We utilized mutant constructs of RGS9-2 to show that the RGS9-2 DEP (for Disheveled, EGL-10, Pleckstrin homology) domain and the GTPase accelerating activity of RGS9-2 were necessary for mediating specific inhibition of D2R internalization.


Assuntos
Agonistas de Dopamina/farmacologia , Antagonistas dos Receptores de Dopamina D2 , Endocitose/fisiologia , Neurônios/metabolismo , Proteínas RGS/fisiologia , Receptores de Dopamina D2/metabolismo , Animais , Linhagem Celular , Dopamina/fisiologia , Endocitose/efeitos dos fármacos , Feminino , Humanos , Neurônios/efeitos dos fármacos , Oócitos , Estrutura Terciária de Proteína/efeitos dos fármacos , Estrutura Terciária de Proteína/fisiologia , Transporte Proteico/efeitos dos fármacos , Transporte Proteico/fisiologia , Proteínas RGS/química , Proteínas RGS/metabolismo , Xenopus laevis
9.
J Agric Food Chem ; 58(7): 3965-9, 2010 Apr 14.
Artigo em Inglês | MEDLINE | ID: mdl-20014760

RESUMO

Recent data suggest that ellagitannins (ETs), a class of hydrolyzable tannins found in some fruits and nuts, may have beneficial effects against colon cancer. In the stomach and gut, ETs hydrolyze to release ellagic acid (EA) and are converted by gut microbiota to urolithin A (UA; 3,8-dihydroxy-6H-dibenzopyran-6-one) type metabolites, which may persist in the colon through enterohepatic circulation. However, little is known about the mechanisms of action of either the native compounds or their metabolites on colon carcinogenesis. Components of Wnt signaling pathways are known to play a pivotal role in human colon carcinogenesis, and inappropriate activation of the signaling cascade is observed in 90% of colorectal cancers. This study investigated the effects of UA, EA, and ET-rich fruit extracts on Wnt signaling in a human 293T cell line using a luciferase reporter of canonical Wnt pathway-mediated transcriptional activation. The ET extracts were obtained from strawberry (Fragaria annassa), Jamun berry (Eugenia jambolana), and pomegranate (Punica granatum) fruit and were all standardized to phenolic content (as gallic acid equivalents, GAEs, by the Folin-Ciocalteu method) and to EA content (by high-performance liquid chromatography methods): strawberry = 20.5% GAE, 5.0% EA; Jamun berry = 20.5% GAE, 4.2% EA; pomegranate = 55% GAE, 3.5% EA. The ET extracts (IC(50) = 28.0-30.0 microg/mL), EA (IC(50) = 19.0 microg/mL; 63 microM), and UA (IC(50) = 9.0 microg/mL; 39 microM) inhibited Wnt signaling, suggesting that ET-rich foods have potential against colon carcinogenesis and that urolithins are relevant bioactive constituents in the colon.


Assuntos
Colo/metabolismo , Cumarínicos/farmacologia , Ácido Elágico/farmacologia , Frutas/química , Taninos Hidrolisáveis/metabolismo , Extratos Vegetais/metabolismo , Transdução de Sinais/efeitos dos fármacos , Proteínas Wnt/metabolismo , Linhagem Celular Tumoral , Colo/efeitos dos fármacos , Cumarínicos/metabolismo , Ácido Elágico/metabolismo , Fragaria/química , Humanos , Taninos Hidrolisáveis/farmacologia , Lythraceae/química , Extratos Vegetais/farmacologia
10.
J Biol Chem ; 285(7): 4781-7, 2010 Feb 12.
Artigo em Inglês | MEDLINE | ID: mdl-20007977

RESUMO

The R7 subfamily of RGS proteins critically regulates neuronal G protein-signaling pathways that are essential for vision, nociception, motor coordination, and reward processing. A member of the R7 RGS family, RGS11, is a GTPase-accelerating protein specifically expressed in retinal ON-bipolar cells where it forms complexes with the atypical G protein beta subunit, Gbeta(5), and transmembrane protein R9AP. Association with R9AP has been shown to be critical for the proteolytic stability of the complex in the retina. In this study we report that R9AP can in addition stimulate the GTPase-accelerating protein activity of the RGS11 x Gbeta(5) complex at Galpha(o). Single turnover GTPase assays reveal that R9AP co-localizes RGS11 x Gbeta(5) and Galpha(o) on the membrane and allosterically potentiates the GTPase-accelerating function of RGS11 x Gbeta(5). Reconstitution of mGluR6-Galpha(o) signaling in Xenopus oocytes indicates that RGS11 x Gbeta(5)-mediated GTPase acceleration in this system requires co-expression of R9AP. The results provide new insight into the regulation of mGluR6-Galpha(o) signaling by the RGS11 x Gbeta(5) x R9AP complex and establish R9AP as a general GTPase-accelerating protein activity regulator of R7 RGS complexes.


Assuntos
Subunidades beta da Proteína de Ligação ao GTP/imunologia , Proteínas de Membrana/metabolismo , Proteínas RGS/metabolismo , Receptores de Glutamato Metabotrópico/metabolismo , Transdução de Sinais/fisiologia , Animais , Bovinos , Linhagem Celular , Subunidades beta da Proteína de Ligação ao GTP/genética , Proteínas de Membrana/genética , Camundongos , Ligação Proteica , Proteínas RGS/genética , Receptores de Glutamato Metabotrópico/genética , Transdução de Sinais/genética , Spodoptera , Xenopus
11.
Am J Obstet Gynecol ; 195(4): 1015-9, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17000234

RESUMO

OBJECTIVE: The purpose of this study was to characterize effect of progesterone (P4) on interleukin-6 (IL-6) production by fetoplacental artery explants, fetal granulocytes, and fetal and maternal mononuclear cells. STUDY DESIGN: Arteries and cord blood were obtained from 5 term pregnancies undergoing repeat cesarean section. Maternal blood was obtained from another 6 women at 16 to 20 weeks' gestation. Tissues were fractionated by dissection or Histopaque gradient. Specimens were incubated in physiologic media then exposed to lipopolysaccharide (LPS) or P4 alone, or pretreated with P4 and then exposed to LPS. Samples were evaluated for IL-6 by enzyme-linked immunosorbent assay (ELISA). RESULTS: Arteries and fetal and maternal mononuclear cells exposed to LPS increased IL-6 secretion by 9-, 27-, and 29-fold, respectively. P4 pretreatment blocked LPS induction of IL-6. Fetal granulocytes did not increase IL-6 production in response to LPS exposure. CONCLUSION: LPS induces IL-6 in arteries and fetal and maternal mononuclear cells. P4 pretreatment significantly blocks this effect in these cell populations, suggesting possible targets for anti-inflammatory actions of P4 in prevention of preterm birth.


Assuntos
Córion/irrigação sanguínea , Sangue Fetal/efeitos dos fármacos , Interleucina-6/biossíntese , Leucócitos Mononucleares/efeitos dos fármacos , Lipopolissacarídeos/farmacologia , Progesterona/farmacologia , Artérias/efeitos dos fármacos , Artérias/metabolismo , Feminino , Sangue Fetal/metabolismo , Humanos , Leucócitos Mononucleares/metabolismo , Gravidez , Fator de Necrose Tumoral alfa/biossíntese
12.
Mol Pharmacol ; 65(3): 528-37, 2004 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-14978231

RESUMO

Homologous desensitization of the micro opioid receptor (muOR) can be resolved into distinct processes that include the uncoupling of the muOR from its G-protein effectors and internalization of cell surface receptors. Using electrophysiological recordings of muOR activation of G-protein-coupled K+ channels (Kir3) in Xenopus laevis oocytes and AtT20 cells, confocal microscopy of receptor localization, and radioligand binding of cell surface receptors, we resolved these desensitization mechanisms to determine the domain of muOR important for receptor uncoupling. Activation of muOR by saturating concentrations of [D-Ala2,N-Me-Phe4,Gly5-ol]-enkephalin (DAMGO), methadone, or fentanyl, but not morphine, produced robust internalization of a green fluorescent protein-tagged muOR. A subsaturating concentration of DAMGO (100 nM) did not cause receptor internalization but markedly reduced the subsequent responsiveness of Kir3 by uncoupling muOR. muOR desensitization in AtT20 cells was confirmed to be homologous, because desensitization by 100 nM DAMGO was blocked by dominant-negative forms of either G protein-coupled receptor kinase (GRK) or arrestin, and pretreatment with DAMGO did not affect the Kir3 response to somatostatin receptor activation. Alanine substitution of a single threonine in the second cytoplasmic loop of the muOR (Threonine 180) blocked agonist-dependent receptor uncoupling without affecting receptor internalization. These results suggest that GRK-dependent phosphorylation of muOR required threonine 180 for uncoupling but that a different GRK and arrestin-dependent mechanism controlled muOR internalization in AtT20 cells.


Assuntos
Canais de Potássio Corretores do Fluxo de Internalização , Proteínas Serina-Treonina Quinases/metabolismo , Receptores Opioides mu/metabolismo , Alanina/genética , Substituição de Aminoácidos , Animais , Arrestina/metabolismo , Células Cultivadas , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G , Quinase 3 de Receptor Acoplado a Proteína G , Proteínas de Fluorescência Verde , Humanos , Proteínas Luminescentes/metabolismo , Morfina/farmacologia , Oócitos , Fagocitose/efeitos dos fármacos , Fosforilação , Canais de Potássio/metabolismo , Estrutura Terciária de Proteína , Receptores Opioides mu/química , Treonina/genética , Xenopus laevis
13.
Neuron ; 41(1): 153-63, 2004 Jan 08.
Artigo em Inglês | MEDLINE | ID: mdl-14715142

RESUMO

Stimulation of adenylyl cyclase in the hippocampus is critical for memory formation. However, generation of cAMP signals within an optimal range for memory may require a balance between stimulatory and inhibitory mechanisms. The role of adenylyl cyclase inhibitory mechanisms for memory has not been addressed. One of the mechanisms for inhibition of adenylyl cyclase is through activation of G(i)-coupled receptors, a mechanism that could serve as a constraint on memory formation. Here we report that ablation of G(ialpha1) by gene disruption increases hippocampal adenylyl cyclase activity and enhances LTP in area CA1. Furthermore, gene ablation of G(ialpha1) or antisense oligonucleotide-mediated depletion of G(ialpha1) disrupted hippocampus-dependent memory. We conclude that G(ialpha1) provides a critical mechanism for tonic inhibition of adenylyl cyclase activity in the hippocampus. We hypothesize that loss of G(ialpha1) amplifies the responsiveness of CA1 postsynaptic neurons to stimuli that strengthen synaptic efficacy, thereby diminishing synapse-specific plasticity required for new memory formation.


Assuntos
Adenilil Ciclases/metabolismo , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/fisiologia , Hipocampo/enzimologia , Potenciação de Longa Duração/fisiologia , Memória/fisiologia , Animais , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/antagonistas & inibidores , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/genética , Deleção de Genes , Hipocampo/fisiologia , Memória/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Toxina Pertussis/farmacologia
14.
J Biol Chem ; 277(18): 15729-35, 2002 May 03.
Artigo em Inglês | MEDLINE | ID: mdl-11861651

RESUMO

Receptor desensitization by G-protein receptor kinases (GRK) and arrestins is likely to be an important component underlying the development of tolerance to opioid drugs. Reconstitution of this process in Xenopus oocytes revealed distinct differences in the kinetics of GRK and arrestin regulation of the closely related opioid receptors mu (MOR), delta (DOR), and kappa (KOR). We demonstrated that under identical conditions, GRK and arrestin-dependent desensitization of MOR proceeds dramatically slower than that of DOR. Furthermore, GRK3 phosphorylation sites required for opioid receptor desensitization also greatly differ. The determinants for DOR and KOR desensitization reside in the carboxyl-terminal tail, whereas MOR depends on Thr-180 in the second intracellular loop. Although this later finding might indicate an inefficient phosphorylation of MOR Thr-180, increasing the amount of arrestin expressed greatly increased the rate of MOR desensitization to a rate comparable with that of DOR. Similarly, coexpression of a constitutively active arrestin 2(R169E) with MOR and DOR desensitized both receptors in an agonist-dependent, GRK-independent manner at rates that were indistinguishable. Together, these data suggest that it is the activation of arrestin, rather than its binding, that is the rate-limiting step in MOR desensitization. In addition, mutation of Thr-161 in DOR, homologous to MOR Thr-180, significantly inhibited the faster desensitization of DOR. These results suggest that DOR desensitization involves phosphorylation of both the carboxyl-terminal tail and the second intracellular loop that together leads to a more efficient activation of arrestin and thus faster desensitization.


Assuntos
Arrestinas/metabolismo , Receptores Opioides delta/fisiologia , Receptores Opioides mu/fisiologia , Sequência de Aminoácidos , Substituição de Aminoácidos , Animais , Arrestina , Arrestinas/genética , Clonagem Molecular , Ala(2)-MePhe(4)-Gly(5)-Encefalina/farmacologia , D-Penicilina (2,5)-Encefalina/farmacologia , Quinase 3 de Receptor Acoplado a Proteína G , Proteínas de Ligação ao GTP/metabolismo , Cinética , Camundongos , Modelos Moleculares , Mutagênese Sítio-Dirigida , Oócitos/fisiologia , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Fosforilação , Conformação Proteica , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Receptores Opioides delta/efeitos dos fármacos , Receptores Opioides delta/genética , Receptores Opioides mu/efeitos dos fármacos , Receptores Opioides mu/genética , Proteínas Recombinantes/efeitos dos fármacos , Proteínas Recombinantes/metabolismo , Treonina , Xenopus laevis
15.
J Biol Chem ; 277(11): 9043-8, 2002 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-11782458

RESUMO

Arrestins play a key role in the homologous desensitization of G protein-coupled receptors (GPCRs). These cytosolic proteins selectively bind to the agonist-activated and GPCR kinase-phosphorylated forms of the GPCR, precluding its further interaction with the G protein. Certain mutations in visual arrestin yield "constitutively active" proteins that bind with high affinity to the light-activated form of rhodopsin without requiring phosphorylation. The crystal structure of visual arrestin shows that these activating mutations perturb two groups of intramolecular interactions that keep arrestin in its basal (inactive) state. Here we introduced homologous mutations into arrestin2 and arrestin3 and found that the resulting mutants bind to the beta(2)-adrenoreceptor in vitro in a phosphorylation-independent fashion. The same mutants effectively desensitize both the beta(2)-adrenergic and delta-opioid receptors in the absence of receptor phosphorylation in Xenopus oocytes. Moreover, the arrestin mutants also desensitize the truncated delta-opioid receptor from which the C terminus, containing critical phosphorylation sites, has been removed. Conservation of the phosphate-sensitive hot spots in non-visual arrestins suggests that the overall fold is similar to that of visual arrestin and that the mechanisms whereby receptor-attached phosphates drive arrestin transition into the active binding competent state are conserved throughout the arrestin family of proteins.


Assuntos
Arrestina/química , Animais , Arrestina/farmacologia , Mutagênese Sítio-Dirigida , Fosforilação , Conformação Proteica , Receptores Adrenérgicos beta 2/efeitos dos fármacos , Relação Estrutura-Atividade , Xenopus
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...