Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
Arch Biochem Biophys ; 750: 109744, 2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-37696381

RESUMO

Renal interstitial fibrosis (RIF) is considered as a common pathway for all patients with chronic kidney disease (CKD) to progress to end-stage kidney disease (ESRD). The basic pathological manifestation is the increase of matrix component in the tubular interstitium, while the injury of tubular epithelial cells in the renal interstitium and the excessive accumulation of matrix will eventually lead to tubular atrophy and obstruction, loss of effective renal units, and finally impaired renal filtration function. The relevant mechanism of RIF remains unclear. The present study will investigate the function and relevant mechanism of RGS1 in RIF. The RIF-related microarrays GSE22459 and GSE76882 were downloaded and analyzed. Renal parenchymal atrophic calyx tissues were collected from clinical RIF patients. Cellular inflammation, fibrosis and animal RIF models were constructed using Lipopolysaccharide (LPS), TGF-ß1 and unilateral ureteral occlusion (UUO). HE and Masson staining were performed to detect morphological alterations of renal tissue samples. qRT-PCR, Western blot and ELISA were carried out to detect the expression of relevant genes/proteins. RGS1 is a gene co-differentially expressed by GSE22459 and GSE76882. RGS1 expression was elevated in renal tissues of RIF patients, cells and animal RIF models. Knockdown of RGS1 inhibited renal cell inflammatory response, fibrosis and renal fibrosis in RIF mice. Overexpression of RGS1 plays the opposite role. Knockdown of RGS1 inhibited the inflammatory response in the RIF cell and mouse model. Targeting RGS1 might be a potential therapeutic strategy for RIF treatment.


Assuntos
Nefropatias , Proteínas RGS , Insuficiência Renal Crônica , Obstrução Ureteral , Humanos , Camundongos , Animais , Nefropatias/genética , Nefropatias/metabolismo , Rim/patologia , Fator de Crescimento Transformador beta1/metabolismo , Insuficiência Renal Crônica/patologia , Modelos Animais de Doenças , Fibrose , Proteínas RGS/genética , Proteínas RGS/metabolismo , Proteínas RGS/farmacologia
2.
Clin Exp Hypertens ; 45(1): 2271186, 2023 Dec 31.
Artigo em Inglês | MEDLINE | ID: mdl-37879890

RESUMO

Objective: Excessive proliferation and migration of pulmonary arterial smooth muscle cell (PASMC) is a core event of pulmonary hypertension (PH). Regulators of G protein signaling 10 (RGS10) can regulate cellular proliferation and cardiopulmonary diseases. We demonstrate whether RGS10 also serves as a regulator of PH.Methods: PASMC was challenged by hypoxia to induce proliferation and migration. Adenovirus carrying Rgs10 gene (Ad-Rgs10) was used for external expression of Rgs10. Hypoxia/SU5416 or MCT was used to induce PH. Right ventricular systolic pressure (RVSP) and right ventricular hypertrophy index (RVHI) were used to validate the establishment of PH model.Results: RGS10 was downregulated in hypoxia-challenged PASMC. Ad-Rgs10 significantly suppressed proliferation and migration of PASMC after hypoxia stimulus, while silencing RGS10 showed contrary effect. Mechanistically, we observed that phosphorylation of S6 and 4E-Binding Protein 1 (4EBP1), the main downstream effectors of mammalian target of rapamycin complex 1 (mTORC1) as well as phosphorylation of AKT, the canonical upstream of mTORC1 in hypoxia-induced PASMC were negatively modulated by RGS10. Both recovering mTORC1 activity and restoring AKT activity abolished these effects of RGS10 on PASMC. More importantly, AKT activation also abolished the inhibitory role of RGS10 in mTORC1 activity in hypoxia-challenged PASMC. Finally, we also observed that overexpression of RGS10 in vivo ameliorated pulmonary vascular wall thickening and reducing RVSP and RVHI in mouse PH model.Conclusion: Our findings reveal the modulatory role of RGS10 in PASMC and PH via AKT/mTORC1 axis. Therefore, targeting RGS10 may serve as a novel potent method for the prevention against PH."


Assuntos
Hipertensão Pulmonar , Proteínas RGS , Animais , Camundongos , Proliferação de Células , Células Cultivadas , Proteínas de Ligação ao GTP/metabolismo , Proteínas de Ligação ao GTP/farmacologia , Hipertensão Pulmonar/metabolismo , Hipertrofia Ventricular Direita , Hipóxia/metabolismo , Mamíferos/metabolismo , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Alvo Mecanístico do Complexo 1 de Rapamicina/farmacologia , Miócitos de Músculo Liso/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Artéria Pulmonar , Proteínas RGS/genética , Proteínas RGS/metabolismo , Proteínas RGS/farmacologia
3.
J Cardiovasc Pharmacol ; 80(1): 110-117, 2022 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-35522176

RESUMO

ABSTRACT: MicroRNAs have been implicated in atherosclerosis (AS) progression. Here, we focused on how miR-320a affect AS progression via vascular smooth muscle cells (VSMCs). Oxidized low-density lipoproteins (ox-LDL)-stimulated VSMCs were used as an AS cell model, and qRT-PCR was performed to measure miR-320a and regulators of G protein signaling (RGS5) levels. CCK-8 and wound healing assays were used to detect the viability and migration of VSMCs. Western blotting was used to measure the protein expression levels of PCNA, Bax, and Bcl-2. The interaction of miR-320a and RGS5 was determined by dual luciferase and RNA pull-down assays. MiR-320a was highly expressed, whereas RGS5 showed low levels of expression in the arterial plaque tissues. Silencing of miR-320a blocked cell viability and migration, inhibited expression of the proliferation-specific protein PCNA in ox-LDL-treated VSMCs, promoted Bax protein expression, and inhibited Bcl-2 protein expression. Furthermore, miR-320a was found to exert these effects by inhibiting RGS5 expression. Collectively, miR-320a promoted cell viability, migration, and proliferation while reducing apoptosis of ox-LDL-stimulated VSMCs by inhibiting RGS5.


Assuntos
Aterosclerose , MicroRNAs , Proteínas RGS , RNA Longo não Codificante , Apoptose , Aterosclerose/genética , Aterosclerose/metabolismo , Movimento Celular , Proliferação de Células , Células Cultivadas , Humanos , Lipoproteínas LDL/metabolismo , MicroRNAs/metabolismo , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , Antígeno Nuclear de Célula em Proliferação/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Proteínas RGS/genética , Proteínas RGS/metabolismo , Proteínas RGS/farmacologia , RNA Longo não Codificante/genética
4.
FASEB J ; 33(11): 11804-11820, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31365833

RESUMO

Memory deficits affect a large proportion of the human population and are associated with aging and many neurologic, neurodegenerative, and psychiatric diseases. Treatment of this mental disorder has been disappointing because all potential candidates studied thus far have failed to produce consistent effects across various types of memory and have shown limited to no effects on memory deficits. Here, we show that the promotion of neuronal arborization through the expression of the regulator of G-protein signaling 14 of 414 amino acids (RGS14414) not only induced robust enhancement of multiple types of memory but was also sufficient for the recovery of recognition, spatial, and temporal memory, which are kinds of episodic memory that are primarily affected in patients or individuals with memory dysfunction. We observed that a surge in neuronal arborization was mediated by up-regulation of brain-derived neurotrophic factor (BDNF) signaling and that the deletion of BDNF abrogated both neuronal arborization activation and memory enhancement. The activation of BDNF-dependent neuronal arborization generated almost 2-fold increases in synapse numbers in dendrites of pyramidal neurons and in neurites of nonpyramidal neurons. This increase in synaptic connections might have evoked reorganization within neuronal circuits and eventually supported an increase in the activity of such circuits. Thus, in addition to showing the potential of RGS14414 for rescuing memory deficits, our results suggest that a boost in circuit activity could facilitate memory enhancement and the reversal of memory deficits.-Masmudi-Martín, M., Navarro-Lobato, I., López-Aranda, M. F., Delgado, G., Martín-Montañez, E., Quiros-Ortega, M. E., Carretero-Rey, M., Narváez, L., Garcia-Garrido, M. F., Posadas, S., López-Téllez, J. F., Blanco, E., Jiménez-Recuerda, I., Granados-Durán, P., Paez-Rueda, J., López, J. C., Khan, Z. U. RGS14414 treatment induces memory enhancement and rescues episodic memory deficits.


Assuntos
Encéfalo/efeitos dos fármacos , Transtornos da Memória/tratamento farmacológico , Plasticidade Neuronal/efeitos dos fármacos , Fragmentos de Peptídeos/farmacologia , Proteínas RGS/farmacologia , Animais , Encéfalo/fisiopatologia , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Transtornos da Memória/metabolismo , Memória Episódica , Camundongos , Neuritos/metabolismo , Plasticidade Neuronal/fisiologia , Neurônios/metabolismo , Ratos , Transdução de Sinais/efeitos dos fármacos , Sinapses/efeitos dos fármacos , Sinapses/metabolismo
5.
Ann Clin Lab Sci ; 53(2): 238-247, 2023 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-37094862

RESUMO

OBJECTIVE: Abdominal aortic aneurysm (AAA) is characterized by vascular smooth muscle cell (VSMC) injury. Circ_0000285 has been declared to drive cancer development, but its role in AAA remains unclear. We thus intended to disclose circ_0000285's role and molecular mechanism in AAA. METHODS: VSMCs were exposed to hydrogen peroxide (H2O2) to induce cell injury. Circ_0000285, miR-599, and regulator of G protein signaling 17 (RGS17) mRNA expressions were ascertained by conducting RT-qPCR assay while the levels of RGS17 protein was ascertained via western blotting. MiR-599's predicted binding with circ_0000285 and RGS17 were validated by means of the dual-luciferase reporter experiment. Cell proliferation was evaluated through the CCK-8 and EdU assays. Cell apoptosis was assessed via the caspase-3 activity assay. RESULTS: The AAA samples and H2O2-treated VSMCs manifested high expressions of circ_0000285 and RGS17 as well as a poor miR-599 expression. H2O2 treatment impaired the proliferation of VSMCs while stimulating their apoptosis. Circ_0000285 overexpression further repressed cell proliferation and enhanced apoptosis in H2O2-treated VSMCs while miR-599 enrichment partly reversed these effects. Circ_0000285 directly bound to miR-599, and miR-599 interacted with RGS17 3'UTR. RGS17 overexpression also suppressed cell proliferation and stimulated apoptosis in H2O2-treated VSMCs. Nevertheless, these effects were offset by miR-599 enrichment. CONCLUSION: Circ_0000285 governed the miR-599/RGS17 network to regulate H2O2-induced VSMC injuries, thereby promoting the development of AAA.


Assuntos
Aneurisma da Aorta Abdominal , MicroRNAs , Proteínas RGS , Humanos , MicroRNAs/genética , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patologia , Peróxido de Hidrogênio/farmacologia , Transdução de Sinais , Aneurisma da Aorta Abdominal/genética , Aneurisma da Aorta Abdominal/metabolismo , Aneurisma da Aorta Abdominal/patologia , Proliferação de Células/genética , Apoptose , Proteínas RGS/metabolismo , Proteínas RGS/farmacologia
6.
Autoimmunity ; 56(1): 2194584, 2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-36999276

RESUMO

Impaired insulin secretion due to pancreatic ß-cell injury is an important cause of type 2 diabetes (T2D). Regulators of guanine nucleotide binding protein (G protein) signaling proteins played a key role in regulating insulin sensitivity in vivo. To explore the role of RGS7 on palmitic acid-induced pancreatic ß-cell injury, pancreatic ß-cells Beta-TC-6 and Min6 were treated with palmitic acid (PA) to similar type 2 diabetes (T2D) injury in vitro. The 3-(4,5)-dimethylthiahiazo (-z-y1)-3,5-di-phenytetrazoliumromide (MTT), 5-ethynyl-2'-deoxyuridine (EdU), and flow cytometry were used to analyze cell viability, proliferation, and apoptosis, respectively. Enzyme-linked immunosorbent assay (ELISA) kits were used to analyze the changes of inflammation-related cytokines. The expression of gene and protein was measured by quantitative real-time PCR (qRT-PCR) and western blot. PA modeling induced apoptosis, increased levels of inflammation-related cytokines, and suppressed cell viability and proliferation of pancreatic ß-cells. RGS7 silence markedly alleviated the cell injury induced by PA. RGS7 overexpression further aggravated apoptosis and inflammatory response in PA-induced pancreatic ß-cells and inhibited cell viability and proliferation. It is worth noting that RGS7 activated the chemokine signaling pathway. Silence of the key gene of the chemokine signaling pathway could eliminate the negative effect of RGS7 on PA-induced pancreatic ß-cells. RGS7 silence protects pancreatic ß-cells from PA-induced injury by inactivating the chemokine signaling pathway.


Assuntos
Diabetes Mellitus Tipo 2 , Células Secretoras de Insulina , Proteínas RGS , Humanos , Ácido Palmítico/farmacologia , Ácido Palmítico/metabolismo , Transdução de Sinais , Diabetes Mellitus Tipo 2/metabolismo , Células Secretoras de Insulina/metabolismo , Apoptose/genética , Citocinas/metabolismo , Inflamação/metabolismo , Quimiocinas , Proteínas RGS/genética , Proteínas RGS/metabolismo , Proteínas RGS/farmacologia
7.
Anticancer Res ; 41(12): 6135-6145, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34848468

RESUMO

BACKGROUND/AIM: This study aimed to explore RGS2 as a regulator of melanoma cell growth. MATERIALS AND METHODS: Effect of RGS2 over-expression was analyzed in three melanoma cell lines, and Rgs2 knockdown was performed in zebrafish. RESULTS: RGS2 was differentially expressed among the cell lines. In B16F10 cells, RGS2 over-expression inhibited MAPK and AKT activation, and prevented cell growth. A similar outcome was observed in A375 cells, but the MAPK signals were not suppressed. In A2058 cells, RGS2 repressed AKT activation, but without affecting cell growth. Moreover, MAPK and AKT constitutive activation abolished the RGS2 inhibitory effect on B16F10 cell growth. Rgs2 knockdown caused ectopic melanocyte differentiation, and promoted MAPK and AKT activation in zebrafish embryos. CONCLUSION: RGS2 prevents melanoma cell growth by inhibiting MAPK and AKT, but this effect depends on the overall cell genetic landscape. Further studies are warranted to investigate the anticancer therapeutic potential of RGS2 for melanoma.


Assuntos
Sequências Hélice-Alça-Hélice/fisiologia , Melanoma/tratamento farmacológico , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas RGS/uso terapêutico , Animais , Humanos , Melanoma/fisiopatologia , Proteínas RGS/farmacologia , Transdução de Sinais , Peixe-Zebra
8.
Eur J Pharmacol ; 587(1-3): 16-24, 2008 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-18457830

RESUMO

Termination of signalling by G-protein-coupled receptors requires inactivation of the G alpha-subunits of heterotrimeric G-proteins and the re-association of G alpha- and G betagamma-subunits. Inactivation of G alpha-subunits is achieved by the hydrolysis of bound GTP by an intrinsic GTPase activity, which is considerably enhanced by GTPase activating proteins. Regulators of G-protein signalling (RGS) proteins are a large family of GTPase activating proteins, many of which have structures indicating roles beyond GTPase activating protein activity and suggesting that the identity of the RGS protein recruited may also be critical to other aspects of signalling. There is some evidence of selective effects of RGS proteins against different G-protein-coupled receptors coupling to the same signalling pathways and growing evidence of physical interactions between RGS proteins and G-protein-coupled receptors. However, it is unclear as to how common such interactions are and the circumstances under which they are functionally relevant. Here we have examined potential selectivity of RGS2, 3 and 4 against signalling mediated by G alpha q/11-coupled muscarinic M3 receptors and gonadotropin-releasing hormone in an immortalised mouse pituitary cell line. Despite major structural differences between these two receptor types and agonist-dependent phosphorylation of the muscarinic M3- but not gonadotropin-releasing hormone receptor, signalling by both receptors was similarly inhibited by expression of either RGS2 or RGS3, whereas RGS4 has little effect. Thus, at least in these circumstances, RGS protein-dependent inhibition of signalling is not influenced by the nature of the G-protein-coupled receptor through which the signalling is mediated.


Assuntos
Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/fisiologia , Proteínas de Ligação ao GTP/farmacologia , Proteínas Ativadoras de GTPase/farmacologia , Proteínas RGS/farmacologia , Receptor Muscarínico M3/efeitos dos fármacos , Receptores LHRH/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Animais , Técnicas Biossensoriais , Sinalização do Cálcio/efeitos dos fármacos , Células Cultivadas , DNA/biossíntese , DNA/genética , Interpretação Estatística de Dados , Humanos , Imuno-Histoquímica , Receptores de Inositol 1,4,5-Trifosfato/efeitos dos fármacos , Cloreto de Metacolina/farmacologia , Agonistas Muscarínicos/farmacologia , Fosforilação , Proteínas Proto-Oncogênicas c-myc/metabolismo , Ratos , Receptores Acoplados a Proteínas G/metabolismo
9.
J Neurosci ; 20(19): 7167-73, 2000 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-11007872

RESUMO

Neuronal alpha1E Ca channel subunits are widely expressed in mammalian brain, where they are thought to form R-type Ca channels. Recent studies have demonstrated that R-type channels contribute to neurosecretion and dendritic Ca influx, but little is known concerning their modulation. Here we show that alpha1E channels are strongly stimulated, and only weakly inhibited, through M1 muscarinic acetylcholine receptors. Both forms of channel modulation are mediated by pertussis toxin-insensitive G-proteins. Channel stimulation is blocked by regulator of G-protein signaling 2 (RGS2) or the C-terminal region of phospholipase C-beta1 (PLCbeta1ct), which have been previously shown to function as GTPase-activating proteins for Galphaq. In contrast, RGS2 and PLCbeta1ct do not block inhibition of alpha1E through M1 receptors. Inhibition is prevented, however, by the C-terminal region of beta-adrenergic receptor kinase 1, which sequesters Gbetagamma dimers. Thus, stimulation of alpha1E is mediated by a pertussis toxin-insensitive Galpha subunit (e.g., Galphaq), whereas inhibition is mediated by Gbetagamma. The ability of RGS2 and PLCbeta1ct to selectively block stimulation indicates these proteins functioned primarily as effector antagonists. In support of this interpretation, RGS2 prevented stimulation of alpha1E with non-hydrolyzable guanosine 5'-0-(3-thiotriphosphate). We also report strong muscarinic stimulation of rbE-II, a variant alpha1E Ca channel that is insensitive to voltage-dependent inhibition. Our results predict that Galphaq-coupled receptors predominantly stimulate native R-type Ca channels. Receptor-mediated enhancement of R-type Ca currents may have important consequences for neurosecretion, dendritic excitability, gene expression, or other neuronal functions.


Assuntos
Bloqueadores dos Canais de Cálcio/farmacologia , Canais de Cálcio/metabolismo , Isoenzimas/metabolismo , Agonistas Muscarínicos/farmacologia , Neurônios/metabolismo , Proteínas RGS/metabolismo , Fosfolipases Tipo C/metabolismo , Animais , Encéfalo/metabolismo , Agonistas dos Canais de Cálcio/farmacologia , Canais de Cálcio/genética , Canais de Cálcio Tipo R , Carbacol/farmacologia , Proteínas de Transporte de Cátions , Linhagem Celular , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/farmacologia , Guanosina 5'-O-(3-Tiotrifosfato)/antagonistas & inibidores , Guanosina 5'-O-(3-Tiotrifosfato)/farmacologia , Humanos , Transporte de Íons/efeitos dos fármacos , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Técnicas de Patch-Clamp , Toxina Pertussis , Fosfolipase C beta , Proteínas RGS/genética , Proteínas RGS/farmacologia , Coelhos , Ratos , Receptor Muscarínico M1 , Receptores Muscarínicos/genética , Receptores Muscarínicos/metabolismo , Transdução de Sinais/efeitos dos fármacos , Estaurosporina/farmacologia , Transfecção , Fatores de Virulência de Bordetella/farmacologia
10.
J Neurosci ; 20(8): 2792-9, 2000 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-10751430

RESUMO

Mammalian rod cyclic nucleotide gated (CNG) channels (i.e., alpha plus beta subunits) are strongly inhibited by phosphatidylinositol 4, 5-bisphosphate (PIP(2)) when they are expressed in Xenopus oocytes and studied in giant membrane patches. Cytoplasmic Mg-ATP inhibits CNG currents similarly, and monoclonal antibodies to PIP(2) reverse the effect and hyperactivate currents. When alpha subunits are expressed alone, PIP(2) inhibition is less strong; olfactory CNG channels are not inhibited. In giant patches from rod outer segments, inhibition by PIP(2) is intermediate. Other anionic lipids (e.g., phosphatidyl serine and phosphatidic acid), a phosphatidylinositol-specific phospholipase C, and full-length diacylglycerol have stimulatory effects. Although ATP also potently inhibits cGMP-activated currents in rod patches, the following findings indicate that ATP is used to transphosphorylate GMP, generated from cGMP, to GTP. First, a phosphodiesterase (PDE) inhibitor, Zaprinast, blocks inhibition by ATP. Second, inhibition can be rapidly reversed by exogenous regulator of G-protein signaling 9, suggesting G-protein activation by ATP. Third, the reversal of ATP effects is greatly slowed when cyclic inosine 5'-monophosphate is used to activate currents, as expected for slow inosine 5' triphosphate hydrolysis by G-proteins. Still, other results remain suggestive of regulatory roles for PIP(2). First, the cGMP concentration producing half-maximal CNG channel activity (K(1/2)) is decreased by PIP(2) antibody in the presence of PDE inhibitors. Second, the activation of PDE activity by several nucleotides, monitored electrophysiologically and biochemically, is reversed by PIP(2) antibody. Third, exogenous PIP(2) can enhance PDE activation by nucleotides.


Assuntos
Trifosfato de Adenosina/farmacologia , Guanosina Trifosfato/farmacologia , Fosfatidilinositol 4,5-Difosfato/farmacologia , Proteínas RGS/farmacologia , Células Fotorreceptoras Retinianas Bastonetes/efeitos dos fármacos , Visão Ocular/efeitos dos fármacos , Trifosfato de Adenosina/fisiologia , Animais , Bovinos , GMP Cíclico/metabolismo , IMP Cíclico/metabolismo , Canais de Cátion Regulados por Nucleotídeos Cíclicos , Diacilglicerol Quinase/farmacologia , Diacilglicerol Quinase/fisiologia , Guanosina Trifosfato/fisiologia , Canais Iônicos/efeitos dos fármacos , Canais Iônicos/fisiologia , Técnicas de Patch-Clamp , Fosfatidilinositol 4,5-Difosfato/fisiologia , Fosfotransferases/farmacologia , Fosfotransferases/fisiologia , Proteínas RGS/fisiologia , Células Fotorreceptoras Retinianas Bastonetes/fisiologia , Visão Ocular/fisiologia , Xenopus
11.
Biochim Biophys Acta ; 1542(1-3): 95-105, 2002 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-11853883

RESUMO

Regulators of G-protein signaling (RGSs) are negative regulators of G-protein coupled receptor (GPCR)-mediated signaling that function to limit the lifetime of receptor-activated G(alpha)-proteins. Here we show that four mammalian RGSs differentially inhibit the activation of a FUS1--LacZ reporter gene by the STE2 encoded GPCR in yeast. In order to examine the role of the GPCR in modulating RGS function, we functionally expressed the human somatostatin receptor 5 (SST(5)) in yeast. In the absence of RGSs, FUS1--LacZ activation in response to somatostatin increased in a dose-dependent manner in cells expressing SST(5). In contrast to the results obtained with Ste2p, all RGSs completely inhibited SST(5)-mediated signaling even at concentrations of agonist as high as 10(minus sign5) M. The ability of RGSs to inhibit SST(5) signaling was further assessed in cells expressing modified Gpa1 proteins. Even though SST(5)-mediated FUS1--LacZ activation was 5-fold more efficient with a Gpa1p/G(i3alpha) chimera, response to somatostatin was completely abolished by all four RGSs. Furthermore, we demonstrate that RGS1, RGS2 and RGS5 have reduced ability to inhibit SST(5)-mediated activation of the RGS-resistant Gpa1p(Gly302Ser) mutant suggesting that the ability to interact with the G(alpha)-protein is required for the inhibition of signaling. Taken together, our results indicate that RGSs serve as better GAPs for Gpa1p when activated by SST(5) than when this G-protein is activated by Ste2p.


Assuntos
Subunidades alfa de Proteínas de Ligação ao GTP , Proteínas Ativadoras de GTPase , Proteínas RGS/metabolismo , Receptores de Somatostatina/antagonistas & inibidores , Proteínas de Saccharomyces cerevisiae , Saccharomyces cerevisiae/fisiologia , Fatores de Transcrição , Proteínas Fúngicas/genética , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP , Genes Reporter , Proteínas Heterotriméricas de Ligação ao GTP/biossíntese , Proteínas Heterotriméricas de Ligação ao GTP/genética , Mutação , Plasmídeos , Proteínas RGS/farmacologia , Receptores de Fator de Acasalamento , Receptores de Peptídeos/biossíntese , Receptores de Peptídeos/genética , Saccharomyces cerevisiae/genética , Transdução de Sinais/efeitos dos fármacos , Somatostatina/farmacologia
12.
Endocrinology ; 142(3): 1188-94, 2001 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11181534

RESUMO

We cloned the mouse TRH receptor type 2 (mTRH-R2) gene, which is 92% identical with rat TRH-R2 and 50% identical with mTRH-R1 at the amino acid level, and identified an intron within the coding sequence that is not present in the TRH-R1 gene structure. Similar to its rat homolog, mTRH-R2 binds TRH with an affinity indistinguishable from mTRH-R1, signals via the phosphoinositide pathway like mTRH-R1, but exhibits a higher basal signaling activity than mTRH-R1. We found that regulator of G protein signaling 4 (RGS4), which differentially inhibits signaling by other receptors that couple to Gq, inhibits TRH-stimulated signaling via mTRH-R1 and mTRH-R2 to similar extents. In contrast, other RGS proteins including RGS7, RGS9, and GAIP had no effect on signaling by mTRH-R1 or mTRH-R2 demonstrating the specificity of RGS4 action. Interestingly, RGS4 markedly inhibited basal signaling by mTRH-R2. Inhibition of basal signaling of mTRH-R2 by RGS4 suggests that modulation of agonist-independent signaling may be an important mechanism of regulation of G protein-coupled receptor activity under normal physiologic circumstances.


Assuntos
Proteínas RGS/farmacologia , Receptores do Hormônio Liberador da Tireotropina/fisiologia , Transdução de Sinais/efeitos dos fármacos , Hormônio Liberador de Tireotropina/farmacologia , Sequência de Aminoácidos/genética , Animais , Northern Blotting , Linhagem Celular , Clonagem Molecular , Humanos , Camundongos , Dados de Sequência Molecular , Isoformas de Proteínas/genética , Isoformas de Proteínas/fisiologia , Receptores do Hormônio Liberador da Tireotropina/genética
13.
Invest Ophthalmol Vis Sci ; 44(3): 1016-22, 2003 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-12601023

RESUMO

PURPOSE: A reconstituted system was used to establish a strategy to determine the rate-limiting chemistry responsible for recovery of the dim-flash response in rod photoreceptors. METHODS: A general approach for identifying the rate-limiting step in a series of reactions is to evaluate the consequences of accelerating each step separately, while monitoring the rate of formation of the end product of the series. This strategy was applied to the reactions involved in quenching phototransduction in bovine rod outer segment (bROS) homogenates. The decay of photoactivated rhodopsin (R*) and inactivation of transducin by guanosine triphosphate (GTP) hydrolysis are the leading candidates for limiting the rate of phototransduction turn-off. These reactions were accelerated separately and together by adding hydroxylamine and/or the regulator of G-protein signaling-9 catalytic domain (RGS9d) while monitoring phosphodiesterase (PDE) activity triggered by a pulse of light in bROS homogenates. RESULTS: PDE activity in bROS homogenates triggered by a flash of light returned to its dark value with a rate constant of 0.087 +/- 0.002 seconds in this system. The rate of PDE recovery increased to 0.11 +/- 0.004 seconds when R* decay was accelerated with 10 to 50 mM hydroxylamine, suggesting that R* inactivation limits the rate of phototransduction turn-off under these conditions. Adding both hydroxylamine and RGS9d, a factor that accelerates transducin inactivation, increased the rate of PDE decay even further. RGS9d had no effect on PDE recovery kinetics unless quenching of R* was also accelerated. CONCLUSIONS: Under in vitro conditions in bROS homogenates, the quenching of R* normally limits the rate of phototransduction shut-off. If R* decay is accelerated, inactivation of transducin by GTP hydrolysis becomes rate limiting. This study offers a general approach that could be used to investigate the rate-limiting chemistry of phototransduction turn-off in vivo.


Assuntos
3',5'-GMP Cíclico Fosfodiesterases/metabolismo , Proteínas do Olho , Guanosina Trifosfato/metabolismo , Lipoproteínas , Proteínas do Tecido Nervoso , Rodopsina/metabolismo , Segmento Externo da Célula Bastonete/fisiologia , Transducina/metabolismo , Visão Ocular/fisiologia , Animais , Proteínas de Ligação ao Cálcio/metabolismo , Bovinos , GMP Cíclico/metabolismo , Ativação Enzimática , GTP Fosfo-Hidrolases/metabolismo , Hipocalcina , Hidrólise , Hidroxilamina/farmacologia , Luz , Proteínas RGS/farmacologia , Recoverina , Segmento Externo da Célula Bastonete/enzimologia , Segmento Externo da Célula Bastonete/efeitos da radiação
14.
Mol Cell Endocrinol ; 191(2): 149-56, 2002 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-12062898

RESUMO

The cytoplasmic carboxyl-terminus of G-protein coupled receptors (GPCRs), absent in the mammalian gonadotropin-releasing hormone receptor (GnRHR), plays an important role in receptor expression, desensitization, internalization and efficiency of coupling to G proteins. Regulators of G protein signaling (RGS) likewise are involved in regulating GPCR-G protein mediated responses and can regulate transcription of other genes. In this study, we evaluate differential expression, ligand binding and effector coupling of the rat GnRHR (rGnRHR) and a chimera of rGnRHR with the pre-mammalian carboxyl domain (rGnRHR-C-tail). Membrane expression of the chimeric receptor and G(q)alpha and G(s)alpha-mediated signaling was increased 2- and 1.5-fold, respectively by RGS10, while RGS3 did not interfere with rGnRHR and rGnRHR-C-tail cell surface expression in spite of negatively regulating GnRH-stimulated G(q)alpha-mediated signaling by both receptors. The rGnRHR and rGnRHR-C-tail showed similar internalization rates in the presence of either RGS protein, indicating that the modification of rGnRHR expression and regulation in the presence of a carboxyl-terminus by RGS10 was not caused by alteration of the internalization rate. The observations in this study implicate the carboxyl domain of the receptor as a site of interaction for RGS10, but not RGS3. This is the first evidence of an altered cell surface expression and regulation of the GnRHR bearing a carboxyl-terminus by RGS proteins.


Assuntos
Regulação da Expressão Gênica/efeitos dos fármacos , Proteínas RGS/farmacologia , Receptores LHRH/genética , Sequência de Aminoácidos , Animais , Sítios de Ligação , Linhagem Celular , Ligantes , Ligação Proteica , Estrutura Terciária de Proteína , Transporte Proteico , Proteínas RGS/genética , Ratos , Receptores LHRH/química , Receptores LHRH/metabolismo , Proteínas Recombinantes de Fusão , Transdução de Sinais/efeitos dos fármacos , Transfecção
15.
Brain Res Dev Brain Res ; 151(1-2): 67-73, 2004 Jul 19.
Artigo em Inglês | MEDLINE | ID: mdl-15246693

RESUMO

The number and affinity of GABA(B) receptors (assayed by the specific antagonist [(3)H]CGP54626A) was unchanged when compared in carefully washed cerebrocortical membranes from young (12-day-old) and adult (90-day-old) rats. In contrast, high-affinity GTPase activity, both basal and baclofen-stimulated was significantly higher (by 45% and 56%, respectively) in adult than in young rats. Similar results were obtained by concomitant determination of agonist (baclofen)-stimulated GTP gamma S binding. Under standard conditions, baclofen-stimulated GTPase activity was further considerably enhanced by exogenously added regulator of G protein function, RGS1, but not by RGS16. RGS16 was able to affect agonist-stimulated GTPase activity only in the presence of markedly increase substrate (GTP) concentrations. RGS1 alone slightly increased GTPase activity in adult rats, but neither RGS1 nor RGS16 influenced GTPase activity in membrane preparations isolated from young animals. These findings indicate increasing functional activity of trimeric G protein(s) involved in GABAergic transmission in the developing rat brain cortex and suggest a high potential of RGS1 in regulation of high-affinity GTPase activity.


Assuntos
Baclofeno/farmacologia , Córtex Cerebral/efeitos dos fármacos , Proteínas de Ligação ao GTP/fisiologia , Envelhecimento/fisiologia , Análise de Variância , Animais , Animais Recém-Nascidos , Córtex Cerebral/crescimento & desenvolvimento , Relação Dose-Resposta a Droga , Interações Medicamentosas , Agonistas GABAérgicos/farmacologia , GTP Fosfo-Hidrolases/metabolismo , Guanosina 5'-O-(3-Tiotrifosfato)/farmacologia , Masculino , Compostos Organofosforados/farmacocinética , Ligação Proteica/efeitos dos fármacos , Ligação Proteica/fisiologia , Proteínas/farmacologia , Proteínas RGS/farmacologia , Ensaio Radioligante/métodos , Ratos , Trítio/farmacocinética
16.
Am J Physiol Cell Physiol ; 286(1): C22-30, 2004 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-12954603

RESUMO

The Ca2+-sensing receptor (CaR) couples to multiple G proteins involved in distinct signaling pathways: Galphai to inhibit the activity of adenylyl cyclase and activate ERK, Galphaq to stimulate phospholipase C and phospholipase A2, and Gbetagamma to stimulate phosphatidylinositol 3-kinase. To determine whether the receptor also couples to Galpha12/13, we investigated the signaling pathway by which the CaR regulates phospholipase D (PLD), a known Galpha12/13 target. We established Madin-Darby canine kidney (MDCK) cell lines that stably overexpress the wild-type CaR (CaRWT) or the nonfunctional mutant CaRR796W as a negative control, prelabeled these cells with [3H]palmitic acid, and measured CaR-stimulated PLD activity as the formation of [3H]phosphatidylethanol (PEt). The formation of [3H]PEt increased in a time-dependent manner in the cells that overexpress the CaRWT but not the CaRR796W. Treatment of the cells with C3 exoenzyme inhibited PLD activity, which indicates that the CaR activates the Rho family of small G proteins, targets of Galpha12/13. To determine which G protein(s) the CaR couples to in order to activate Rho and PLD, we pretreated the cells with pertussis toxin to inactivate Galphai or coexpressed regulators of G protein-signaling (RGS) proteins to attenuate G protein signaling (RGS4 for Galphai and Galphaq, and a p115RhoGEF construct containing the RGS domain for Galpha12/13). Overexpression of p115RhoGEF-RGS in the MDCK cells that overexpress CaRWT inhibited extracellular Ca2+-stimulated PLD activity, but pretreatment of cells with pertussis toxin and overexpression of RGS4 were without effect. The involvement of other signaling components such as protein kinase C, ADP-ribosylation factor, and phosphatidylinositol biphosphate was excluded. These findings demonstrate that the CaR couples to Galpha12/13 to regulate PLD via a Rho-dependent mechanism and does so independently of Galphai and Galphaq. This suggests that the CaR may regulate cytoskeleton via Galpha12/13, Rho, and PLD.


Assuntos
Subunidades alfa G12-G13 de Proteínas de Ligação ao GTP/metabolismo , Rim/metabolismo , Fosfolipase D/metabolismo , Receptores de Detecção de Cálcio/metabolismo , Proteínas de Fase Aguda/fisiologia , Animais , Linhagem Celular , Cães , Ativação Enzimática/fisiologia , Fatores de Troca do Nucleotídeo Guanina/farmacologia , Rim/citologia , Proteínas RGS/farmacologia , Fatores de Troca de Nucleotídeo Guanina Rho
17.
Endocrine ; 25(1): 49-54, 2004 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15545706

RESUMO

Follicle-stimulating hormone receptor (FSHR) and luteinizing hormone receptor (LHR) belong to the super-family of G protein-coupled receptors (GPCR); GPCRs are negatively regulated by RGS ("regulators of G protein signaling") proteins. In this study we evaluated the effects of RGS3 and RGS10 on FSHR and LHR ligand binding and effector coupling. FSHR and LHR ligand binding were unchanged in the presence of RGS3 or RGS10. However, signaling by FSHR and LHR was diminished by RGS3 but not by RGS10. This constitutes the first demonstration of an interaction between RGS proteins and LH and FSH signaling pathways and identifies a mechanism for negative regulation of RGS3 on FSHR and LHR signaling.


Assuntos
Proteínas RGS/farmacologia , Receptores do FSH/metabolismo , Receptores do LH/metabolismo , Proteínas Repressoras/farmacologia , Transdução de Sinais/fisiologia , Animais , Linhagem Celular , Feminino , Humanos , Ligantes , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos
18.
J Physiol ; 535(Pt 1): 133-43, 2001 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-11507164

RESUMO

1. In native cardiac myocytes, there is a time dependence to the G protein-gated inwardly rectifying K(+) (K(G)) channel current during voltage steps that accelerates as the concentration of acetylcholine is increased. This phenomenon has been called 'relaxation' and is not reproduced in the reconstituted Kir3.1/Kir3.4 channel in Xenopus oocytes. We have shown that RGS4, a regulator of G protein signalling, restores relaxation to the reconstituted Kir3.1/Kir3.4 channel. In this study, we examined the mechanism of this phenomenon by expressing various combinations of membrane receptors, G proteins, Kir3.0 subunits and mutants of RGS4 in Xenopus oocytes. 2. RGS4 restored relaxation to K(G) channels activated by the pertussis toxin (PTX)-sensitive G protein-coupled m(2)-muscarinic receptor but not to those activated by the G(s) protein-coupled beta(2)-adrenergic receptor. 3. RGS4 induced relaxation not only in heteromeric K(G) channels composed of Kir3.1 and Kir3.4 but also in homomeric assemblies of either an active mutant of Kir3.1 (Kir3.1/F137S) or an isoform of Kir3.2 (Kir3.2d). 4. Truncation mutants of RGS4 showed that the RGS domain itself was essential to reproduce the effect of wild-type RGS4 on the K(G) channel. 5. The mutation of residues in the RGS domain which interact with the alpha subunit of the G protein (G(alpha)) impaired the effect of RGS4. 6. This study therefore shows that interaction between the RGS domain and PTX-sensitive G(alpha) subunits mediates the effect of RGS4 on the agonist concentration-dependent relaxation of K(G) channels.


Assuntos
Proteínas de Ligação ao GTP/fisiologia , Ativação do Canal Iônico/fisiologia , Canais de Potássio/fisiologia , Proteínas RGS/fisiologia , Animais , Eletrofisiologia , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G , Ativação do Canal Iônico/efeitos dos fármacos , Camundongos , Mutação Puntual/fisiologia , Canais de Potássio/efeitos dos fármacos , Estrutura Terciária de Proteína , Proteínas RGS/química , Proteínas RGS/genética , Proteínas RGS/farmacologia , Ratos , Receptores Adrenérgicos beta/metabolismo , Receptores Muscarínicos/metabolismo , Suínos , Xenopus laevis
19.
J Neurochem ; 78(4): 797-806, 2001 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-11520900

RESUMO

Recombinant RGS1, RGS16 and RGS-GAIP, but not RGS2, were able to substantially further stimulate the maximal GTPase activity of G(o1)alpha promoted by agonists at the alpha2A-adrenoreceptor in a concentration-dependent manner. Kinetic analysis of the regulation of an alpha2A-adrenoreceptor-G(o1)alpha fusion protein by all three RGS proteins revealed that they had similar affinities for the receptor-G protein fusion. However, their maximal effects on GTP hydrolysis varied over threefold with RGS16 > RGS1 > RGS-GAIP. Both RGS1 and RGS16 reduced the potency of the alpha2A-adrenoreceptor agonist adrenaline by some 10-fold. A lower potency shift was observed for the partial agonist UK14304 and the effect was absent for the weak partial agonist oxymetazoline. Each of these RGS proteins altered the intrinsic activity of both UK14304 and oxymetazoline relative to adrenaline. Such results require the RGS interaction with G(o1)alpha to alter the conformation of the alpha2A-adrenoreceptor and are thus consistent with models invoking direct interactions between RGS proteins and receptors. These studies demonstrate that RGS1, RGS16 and RGS-GAIP show a high degree of selectivity to regulate alpha2A-adrenoreceptor-activated G(o1)alpha rather than G(i1)alpha, G(i2)alpha or G(i3)alpha and different capacities to inactivate this G protein.


Assuntos
Proteínas Heterotriméricas de Ligação ao GTP/metabolismo , Proteínas RGS/metabolismo , Receptores Adrenérgicos alfa 2/metabolismo , Transdução de Sinais/fisiologia , Agonistas alfa-Adrenérgicos/farmacologia , Animais , Células COS , Fracionamento Celular , Membrana Celular/fisiologia , Relação Dose-Resposta a Droga , Ativadores de GTP Fosfo-Hidrolase/metabolismo , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP , Cinética , Proteínas RGS/farmacologia , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Transdução de Sinais/efeitos dos fármacos , Fatores de Virulência de Bordetella/farmacologia
20.
J Pharmacol Exp Ther ; 305(2): 460-6, 2003 May.
Artigo em Inglês | MEDLINE | ID: mdl-12606627

RESUMO

We used fluorescence resonance energy transfer imaging of enhanced cyan fluorescent protein (CFP)-tagged and enhanced yellow fluorescent protein (YFP)-tagged protein pairs to examine the hypothesis that G protein gamma subunit-like (GGL) domain-containing regulators of G protein signaling (RGS) can directly bind to the Gbeta5 subunit of heterotrimeric G proteins in vivo. We observed that Gbeta5 could interact with Ggamma2 and Ggamma13, after their expression in human embryonic kidney 293 cells. Interestingly, although untagged Ggamma3 did not interact with Gbeta5, CFP-tagged Ggamma3 strongly interacted with YFP-tagged Gbeta5 in FRET studies. Moreover, CFP-Ggamma3 supported Ca(2+) channel inhibition when paired with Gbeta5 or YFP-Gbeta5, indicating a "gain of function" for CFP-Ggamma3. Gbeta5 could also interact with RGS11 and its N-terminal, but not its C-terminal domain. On the other hand, RGS11 did not interact with Gbeta1. These studies demonstrate that the GGL domain-containing N terminus of RGS 11 can directly interact with Gbeta5 in vivo and supports the hypothesis that this interaction may contribute to the specificity of Gbeta5 interactions with cellular effector molecules.


Assuntos
Subunidades beta da Proteína de Ligação ao GTP , Subunidades gama da Proteína de Ligação ao GTP , Proteínas Heterotriméricas de Ligação ao GTP/química , Proteínas RGS/química , Proteínas RGS/genética , Bloqueadores dos Canais de Cálcio/farmacologia , Canais de Cálcio Tipo N/efeitos dos fármacos , Células Cultivadas , DNA Complementar/biossíntese , DNA Complementar/genética , Eletrofisiologia , Transferência de Energia , Corantes Fluorescentes , Proteínas Heterotriméricas de Ligação ao GTP/genética , Proteínas Heterotriméricas de Ligação ao GTP/farmacologia , Humanos , Técnicas de Patch-Clamp , Proteínas RGS/farmacologia , Transfecção
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa