Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
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
2.
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
3.
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
4.
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
5.
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
6.
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
7.
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
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.
Proc Natl Acad Sci U S A ; 101(46): 16339-44, 2004 Nov 16.
Artigo em Inglês | MEDLINE | ID: mdl-15534226

RESUMO

Regulator of G protein signaling (RGS) proteins negatively regulate receptor-mediated second messenger responses by enhancing the GTPase activity of Galpha subunits. We describe a receptor-specific role for an RGS protein at the level of an individual brain neuron. RGS9-2 and Gbeta(5) mRNA and protein complexes were detected in striatal cholinergic and gamma-aminobutyric acidergic neurons. Dialysis of cholinergic neurons with RGS9 constructs enhanced basal Ca(2+) channel currents and reduced D(2) dopamine receptor modulation of Cav2.2 channels. These constructs did not alter M(2) muscarinic receptor modulation of Cav2.2 currents in the same neuron. The noncatalytic DEP-GGL domain of RGS9 antagonized endogenous RGS9-2 activity, enhancing D(2) receptor modulation of Ca(2+) currents. In vitro, RGS9 constructs accelerated GTPase activity, in agreement with electrophysiological measurements, and did so more effectively at Go than Gi. These results implicate RGS9-2 as a specific regulator of dopamine receptor-mediated signaling in the striatum and identify a role for GAP activity modulation by the DEP-GGL domain.


Assuntos
Canais de Cálcio/efeitos dos fármacos , Corpo Estriado/efeitos dos fármacos , Proteínas RGS/farmacologia , Receptores de Dopamina D2/efeitos dos fármacos , Animais , Sequência de Bases , Canais de Cálcio/metabolismo , Fibras Colinérgicas/efeitos dos fármacos , Fibras Colinérgicas/metabolismo , Corpo Estriado/metabolismo , DNA/genética , Subunidades beta da Proteína de Ligação ao GTP/genética , Subunidades beta da Proteína de Ligação ao GTP/metabolismo , Técnicas In Vitro , Interneurônios/efeitos dos fármacos , Interneurônios/metabolismo , Estrutura Terciária de Proteína , Proteínas RGS/química , Proteínas RGS/genética , Proteínas RGS/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Receptores de Dopamina D2/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/farmacologia , Transdução de Sinais/efeitos dos fármacos
10.
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
11.
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
12.
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
13.
J Pharmacol Exp Ther ; 307(2): 661-9, 2003 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-12975484

RESUMO

We have shown previously that activation of endogenously expressed, Galphaq/11-coupled P2Y2 nucleotide receptors with UTP reveals an intracellular Ca2+ response to activation of recombinant, Galphai-coupled CXC chemokine receptor 2 (CXCR2) in human embryonic kidney cells. Here, we characterize further this cross talk and demonstrate that phospholipase C (PLC) and inositol 1,4,5-trisphosphate [Ins(1,4,5)P3]-dependent Ca2+ release underlies this potentiation. The putative Ins(1,4,5)P3 receptor antagonist 2-aminoethoxydiphenyl borane reduced the response to CXCR2 activation by interleukin-8, as did sustained inhibition of phosphatidylinositol 4-kinase with wortmannin, suggesting the involvement of phosphoinositides in the potentiation. Against a Li+ block of inositol monophosphatase activity, costimulation of P2Y2 nucleotide receptors and CXCR2 caused phosphoinositide accumulation that was significantly greater than that after activation of P2Y2 nucleotide receptors or CXCR2 alone, and was more than additive. Thus, PLC activity, as well as Ca2+ release, was enhanced. In these cells, agonist-mediated Ca2+ release was incremental in nature, suggesting that a potentiation of Ins(1,4,5)P3 generation in the presence of coactivation of P2Y2 nucleotide receptors and CXCR2 would be sufficient for additional Ca2+ release. Potentiated Ca2+ signaling by CXCR2 was markedly attenuated by expression of either regulator of G protein signaling 2 or the Gbetagamma-scavenger Galphat1 (transducin alpha subunit), indicating the involvement of Galphaq and Gbetagamma subunits, respectively.


Assuntos
Cálcio/metabolismo , Receptores de Interleucina-8B/metabolismo , Receptores Purinérgicos P2/metabolismo , Transdução de Sinais/fisiologia , Fosfolipases Tipo C/metabolismo , 1-Fosfatidilinositol 4-Quinase/antagonistas & inibidores , Compostos de Boro/farmacologia , Sinalização do Cálcio , Células Cultivadas , Proteínas de Ligação ao GTP/metabolismo , Humanos , Fosfatos de Inositol/metabolismo , Interleucina-8/farmacologia , Rim/citologia , Rim/embriologia , Monoéster Fosfórico Hidrolases/metabolismo , Proteína Quinase C/metabolismo , Proteínas Tirosina Quinases/metabolismo , Proteínas RGS/genética , Proteínas RGS/farmacologia , Receptor Cross-Talk , Receptores Purinérgicos P2Y2 , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo , Transdução de Sinais/efeitos dos fármacos , Tapsigargina/farmacologia , Uridina Trifosfato/farmacologia
14.
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
15.
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
16.
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
17.
J Biol Chem ; 278(11): 9418-25, 2003 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-12524446

RESUMO

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


Assuntos
Adenilil Ciclases/metabolismo , Cálcio/metabolismo , Proteínas Heterotriméricas de Ligação ao GTP/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Proteínas RGS/química , Receptores Opioides mu/metabolismo , Transdução de Sinais , Animais , Linhagem Celular , Membrana Celular/metabolismo , AMP Cíclico/metabolismo , Relação Dose-Resposta a Droga , Ala(2)-MePhe(4)-Gly(5)-Encefalina/farmacologia , Ativação Enzimática , GTP Fosfo-Hidrolases/metabolismo , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Proteínas Heterotriméricas de Ligação ao GTP/química , Ligantes , Sistema de Sinalização das MAP Quinases , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno , Morfina/farmacologia , Toxina Pertussis/farmacologia , Fosforilação , Ligação Proteica , Proteínas RGS/metabolismo , Proteínas RGS/farmacologia , Ratos , Fatores de Tempo , Transfecção , Células Tumorais Cultivadas
18.
J Physiol ; 545(2): 355-73, 2002 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-12456817

RESUMO

'Regulators of G protein Signalling' (RGSs) accelerate the activation and deactivation kinetics of G protein-gated inwardly rectifying K(+) (GIRK) channels. In an apparent paradox, RGSs do not reduce steady-state GIRK current amplitudes as expected from the accelerated rate of deactivation when reconstituted in Xenopus oocytes. We present evidence here that this kinetic anomaly is dependent on the degree of G protein-coupled receptor (GPCR) precoupling, which varies with different Galpha(i/o)-RGS complexes. The gating properties of GIRK channels (Kir3.1/Kir3.2a) activated by muscarinic m2 receptors at varying levels of G protein expression were examined with or without the co-expression of either RGS4 or RGS7 in Xenopus oocytes. Different levels of specific m2 receptor-Galpha coupling were established by uncoupling endogenous pertussis toxin (PTX)-sensitive Galpha(i/o) subunits with PTX, while expressing varying amounts of a single PTX-insensitive subunit (Galpha(i1(C351G)), Galpha(i2(C352G)), Galpha(i3(C351G)), Galpha(oA(C351G)), or Galpha(oB(C351G))). Co-expression of each of the PTX-insensitive Galpha(i/o) subunits rescued acetylcholine (ACh)-elicited GIRK currents (I(K,ACh)) in a concentration-dependent manner, with Galpha(o) isoforms being more effective than Galpha(i) isoforms. Receptor-independent 'basal' GIRK currents (I(K,basal)) were reduced with increasing expression of PTX-insensitive Galpha subunits and were accompanied by a parallel rise in I(K,ACh). These effects together are indicative of increased Gbetagamma scavenging by the expressed Galpha subunit and the subsequent formation of functionally coupled m2 receptor-G protein heterotrimers (Galpha((GDP))betagamma). Co-expression of RGS4 accelerated all the PTX-insensitive Galpha(i/o)-coupled GIRK currents to a similar extent, yet reduced I(K,ACh) amplitudes 60-90 % under conditions of low Galpha(i/o) coupling. Kinetic analysis indicated the RGS4-dependent reduction in steady-state GIRK current was fully explained by the accelerated deactivation rate. Thus kinetic inconsistencies associated with RGS4-accelerated GIRK currents occur at a critical threshold of G protein coupling. In contrast to RGS4, RGS7 selectively accelerated Galpha(o)-coupled GIRK currents. Co-expression of Gbeta5, in addition to enhancing the kinetic effects of RGS7, caused a significant reduction (70-85 %) in steady-state GIRK currents indicating RGS7-Gbeta5 complexes disrupt Galpha(o) coupling. Altogether these results provide further evidence for a GPCR-Galphabetagamma-GIRK signalling complex that is revealed by the modulatory affects of RGS proteins on GIRK channel gating. Our functional experiments demonstrate that the formation of this signalling complex is markedly dependent on the concentration and composition of G protein-RGS complexes.


Assuntos
Proteínas de Ligação ao GTP/metabolismo , Ativação do Canal Iônico/fisiologia , Oócitos/metabolismo , Canais de Potássio Corretores do Fluxo de Internalização/fisiologia , Proteínas RGS/farmacologia , Receptores Muscarínicos/fisiologia , Acetilcolina/farmacologia , Algoritmos , Animais , Western Blotting , Bovinos , DNA Complementar/biossíntese , DNA Complementar/genética , Eletrofisiologia , Proteínas de Ligação ao GTP/farmacologia , Humanos , Ativação do Canal Iônico/efeitos dos fármacos , Cinética , Potenciais da Membrana/fisiologia , Camundongos , Técnicas de Patch-Clamp , Toxina Pertussis/farmacologia , Canais de Potássio Corretores do Fluxo de Internalização/biossíntese , Canais de Potássio Corretores do Fluxo de Internalização/efeitos dos fármacos , Testes de Precipitina , Ratos , Receptor Muscarínico M2 , Receptores Muscarínicos/efeitos dos fármacos , Xenopus
19.
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
20.
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
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA