Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
1.
Methods ; 203: 249-258, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-34737032

RESUMEN

G protein-coupled receptors (GPCRs) are an important receptor superfamily and common therapeutic targets. The second messenger cyclic adenosine monophosphate (cAMP) is a key mediator in many GPCR signaling pathways. Monitoring intracellular cAMP levels can help identify orthosteric agonists and antagonists, as well as allosteric modulators. In this regard, luminescence-based biosensors have revolutionized our ability to monitor GPCR signaling kinetics. The GloSensor™ cAMP assay enables real-time monitoring of signaling downstream of many GPCRs. However, it is crucial to optimize assay conditions such as temperature. As well, it has not been reported whether the effects of temperature on biosensor activity are reversible. Here, we describe the temperature sensitivity and reversibility of the GloSensor™ cAMP assay, and which GloSensor™ version is optimal for measuring cytosolic cAMP. We also present a detailed protocol for monitoring cAMP levels in live cells expressing endogenous or exogenous GPCRs. Temperature optimization studies were carried out using HEK293H cells transiently transfected with the adenosine receptor A2a and the GloSensor™ plasmid (pGloSensor-20F or -22F). We found that preincubation and luminescence reading at room temperature were optimal as compared to higher temperatures. As well, the GloSensor-22F biosensor had a superior signal-to-background ratio and the effect of temperature on biosensor activity was reversible. However, thermal instability of the biosensor may pose a problem for in vivo studies. Nevertheless, the GloSensor™ cAMP assay can be applied to analyze signaling by a wide range of GPCRs for drug discovery purposes.


Asunto(s)
AMP Cíclico , Receptores Acoplados a Proteínas G , Bioensayo , AMP Cíclico/análisis , AMP Cíclico/metabolismo , Cinética , Luciferasas/genética , Luciferasas/metabolismo , Receptores Acoplados a Proteínas G/genética , Temperatura
2.
J Biol Chem ; 295(8): 2520-2540, 2020 02 21.
Artículo en Inglés | MEDLINE | ID: mdl-31892516

RESUMEN

Proteinase-activated receptor (PAR)-4 is a member of the proteolytically-activated PAR family of G-protein-coupled receptors (GPCR) that represents an important target in the development of anti-platelet therapeutics. PARs are activated by proteolytic cleavage of their receptor N terminus by enzymes such as thrombin, trypsin, and cathepsin-G. This reveals the receptor-activating motif, termed the tethered ligand that binds intramolecularly to the receptor and triggers signaling. However, PARs are also activated by exogenous application of synthetic peptides derived from the tethered-ligand sequence. To better understand the molecular basis for PAR4-dependent signaling, we examined PAR4-signaling responses to a peptide library derived from the canonical PAR4-agonist peptide, AYPGKF-NH2, and we monitored activation of the Gαq/11-coupled calcium-signaling pathway, ß-arrestin recruitment, and mitogen-activated protein kinase (MAPK) pathway activation. We identified peptides that are poor activators of PAR4-dependent calcium signaling but were fully competent in recruiting ß-arrestin-1 and -2. Peptides that were unable to stimulate PAR4-dependent calcium signaling could not trigger MAPK activation. Using in silico docking and site-directed mutagenesis, we identified Asp230 in the extracellular loop-2 as being critical for PAR4 activation by both agonist peptide and the tethered ligand. Probing the consequence of biased signaling on platelet activation, we found that a peptide that cannot activate calcium signaling fails to cause platelet aggregation, whereas a peptide that is able to stimulate calcium signaling and is more potent for ß-arrestin recruitment triggered greater levels of platelet aggregation compared with the canonical PAR4 agonist peptide. These findings uncover molecular determinants critical for agonist binding and biased signaling through PAR4.


Asunto(s)
Receptores de Trombina/metabolismo , Transducción de Señal , Trombina/metabolismo , Alanina/genética , Sustitución de Aminoácidos , Calcio/metabolismo , Señalización del Calcio , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/metabolismo , Células HEK293 , Humanos , Isomerismo , Sistema de Señalización de MAP Quinasas , Metilación , Simulación del Acoplamiento Molecular , Proteínas Mutantes/metabolismo , Mutación/genética , Péptidos/metabolismo , Fosforilación , Agregación Plaquetaria , Receptores de Trombina/agonistas , Homología Estructural de Proteína , beta-Arrestinas/metabolismo
3.
Cell Signal ; 59: 163-170, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30826455

RESUMEN

Regulator of G protein signaling 2 (RGS2) is upregulated by multiple forms of stress and can augment translational attenuation associated with the phosphorylation of the initiation factor eIF2, a hallmark of several stress-induced coping mechanisms. Under stress-induced translational inhibition, key factors, such as ATF4, are selectively expressed via alternative translation mechanisms. These factors are known to regulate molecular switches that control cell fate by regulating pro-survival and pro-apoptotic signals. The molecular mechanisms that balance these opposing responses to stresses are unclear. The present results suggest that RGS2 may be an important regulatory component in the cellular stress response through its translational control abilities. Previously, we have shown that RGS2 can interact with the translation initiation factor, eIF2B, and inhibit de novo protein synthesis. Here, we demonstrate that the expression of either full length RGS2 or its eIF2B-interacting domain (RGS2eb) significantly increases levels of ATF4 and CHOP, both of which are linked to stress-induced apoptosis. Furthermore, we show that these effects are translationally regulated and independent of eIF2 phosphorylation. The present results thus point to a novel function of RGS2 in the stress response directly related to its ability to reduce global protein synthesis.


Asunto(s)
Factor de Transcripción Activador 4/biosíntesis , Iniciación de la Cadena Peptídica Traduccional , Proteínas RGS/fisiología , Estrés Fisiológico/fisiología , Factor de Transcripción CHOP/biosíntesis , Factor de Transcripción Activador 4/genética , Animales , Apoptosis , Factor 2B Eucariótico de Iniciación/química , Ratones , Células 3T3 NIH , Dominios Proteicos , Proteínas RGS/genética , Factor de Transcripción CHOP/genética
4.
ACS Pharmacol Transl Sci ; 2(3): 155-167, 2019 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-32259054

RESUMEN

The parathyroid hormone 1 receptor (PTH1R) is a Class B G-protein-coupled receptor that is a target for osteoporosis therapeutics. Activated PTH1R couples through Gs to the stimulation of adenylyl cyclase. As well, ß-arrestin is recruited to PTH1R leading to receptor internalization and MAPK/ERK signaling. Previously, we reported that the agonist potency of PTH1R is increased in the presence of extracellular ATP, which acts as a positive allosteric modulator of PTH signaling. Another nucleotide, cytidine 5'-monophosphate (CMP), also enhances PTH1R signaling, suggesting that ATP and CMP share a moiety responsible for positive allostery, possibly ribose-5-phosphate. Therefore, we examined the effect of extracellular sugar phosphates on PTH1R signaling. cAMP levels and ß-arrestin recruitment were monitored using luminescence-based assays. Alone, ribose-5-phosphate had no detectable effect on adenylyl cyclase activity in UMR-106 rat osteoblastic cells, which endogenously express PTH1R. However, ribose-5-phosphate markedly enhanced the activation of adenylyl cyclase induced by PTH. Other sugar phosphates, including glucose-1-phosphate, glucose-6-phosphate, fructose-6-phosphate, and fructose-1,6-bisphosphate, also potentiated PTH-induced adenylyl cyclase activation. As well, some sugar phosphates enhanced PTH-induced ß-arrestin recruitment to human PTH1R heterologously expressed in HEK293H cells. Interestingly, the effects of glucose-1-phosphate were greater than those of its isomer glucose-6-phosphate. Our results suggest that phosphorylated monosaccharides such as ribose-5-phosphate contain the pharmacophore for positive allosteric modulation of PTH1R. At least in some cases, the extent of modulation depends on the position of the phosphate group. Knowledge of the pharmacophore may permit future development of positive allosteric modulators to increase the therapeutic efficacy of PTH1R agonists.

6.
Cell Signal ; 46: 103-112, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29501726

RESUMEN

Parathyroid hormone (PTH) activates the PTH/PTH-related peptide receptor (PTH1R) on osteoblasts and other target cells. Mechanical stimulation of cells, including osteoblasts, causes release of nucleotides such as ATP into the extracellular fluid. In addition to its role as an energy source, ATP serves as an agonist at P2 receptors and an allosteric regulator of many proteins. We investigated the effects of concentrations of extracellular ATP, comparable to those that activate low affinity P2X7 receptors, on PTH1R signaling. Cyclic AMP levels were monitored in real-time using a bioluminescence reporter and ß-arrestin recruitment to PTH1R was followed using a complementation-based luminescence assay. ATP markedly enhanced cyclic AMP and ß-arrestin signaling as well as downstream activation of CREB. CMP - a nucleotide that lacks a high energy bond and does not activate P2 receptors - mimicked this effect of ATP. Moreover, potentiation was not inhibited by P2 receptor antagonists, including a specific blocker of P2X7. Thus, nucleotide-induced potentiation of signaling pathways was independent of P2 receptor signaling. ATP and CMP reduced the concentration of PTH (1-34) required to produce a half-maximal cyclic AMP or ß-arrestin response, with no evident change in maximal receptor activity. Increased potency was similarly apparent with PTH1R agonists PTH (1-14) and PTH-related peptide (1-34). These observations suggest that extracellular nucleotides increase agonist affinity, efficacy or both, and are consistent with modulation of signaling at the level of the receptor or a closely associated protein. Taken together, our findings establish that ATP enhances PTH1R signaling through a heretofore unrecognized allosteric mechanism.


Asunto(s)
Adenosina Trifosfato , Citidina Monofosfato , Osteoblastos/metabolismo , Receptor de Hormona Paratiroídea Tipo 1/metabolismo , Adenosina Trifosfato/metabolismo , Adenosina Trifosfato/fisiología , Animales , Línea Celular , AMP Cíclico/metabolismo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Citidina Monofosfato/metabolismo , Citidina Monofosfato/fisiología , Ratones , Hormona Paratiroidea/metabolismo , Antagonistas del Receptor Purinérgico P2X/farmacología , Ratas , beta-Arrestinas/metabolismo
7.
J Cell Mol Med ; 22(4): 2200-2209, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29377505

RESUMEN

Offspring of diabetic mothers are at risk of cardiovascular diseases in adulthood. However, the underlying molecular mechanisms are not clear. We hypothesize that prenatal exposure to maternal diabetes up-regulates myocardial NOX2 expression and enhances ischaemia/reperfusion (I/R) injury in the adult offspring. Maternal diabetes was induced in C57BL/6 mice by streptozotocin. Glucose-tolerant adult offspring of diabetic mothers and normal controls were subjected to myocardial I/R injury. Vascular endothelial growth factor (VEGF) expression, ROS generation, myocardial apoptosis and infarct size were assessed. The VEGF-Akt (protein kinase B)-mammalian target of rapamycin (mTOR)-NOX2 signalling pathway was also studied in cultured cardiomyocytes in response to high glucose level. In the hearts of adult offspring from diabetic mothers, increases were observed in VEGF expression, NOX2 protein levels and both Akt and mTOR phosphorylation levels as compared to the offspring of control mothers. After I/R, ROS generation, myocardial apoptosis and infarct size were all significantly higher in the offspring of diabetic mothers relative to offspring of control mothers, and these differences were diminished by in vivo treatment with the NADPH oxidase inhibitor apocynin. In cultured cardiomyocytes, high glucose increased mTOR phosphorylation, which was inhibited by the PI3 kinase inhibitor LY294002. Notably, high glucose-induced NOX2 protein expression and ROS production were inhibited by rapamycin. In conclusion, maternal diabetes promotes VEGF-Akt-mTOR-NOX2 signalling and enhances myocardial I/R injury in the adult offspring. Increased ROS production from NOX2 is a possible molecular mechanism responsible for developmental origins of cardiovascular disease in offspring of diabetic mothers.


Asunto(s)
Envejecimiento/patología , Diabetes Mellitus Experimental/patología , Diabetes Gestacional/patología , Daño por Reperfusión Miocárdica/patología , NADPH Oxidasa 2/metabolismo , Regulación hacia Arriba , Animales , Animales Recién Nacidos , Apoptosis , Células Cultivadas , Femenino , Glucosa/toxicidad , Ratones Endogámicos C57BL , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Embarazo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal , Serina-Treonina Quinasas TOR/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo
8.
J Mol Cell Cardiol ; 108: 194-202, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28641980

RESUMEN

Regulator of G protein signalling 2 (RGS2) is known to play a protective role in maladaptive cardiac hypertrophy and heart failure via its ability to inhibit Gq- and Gs- mediated GPCR signalling. We previously demonstrated that RGS2 can also inhibit protein translation and can thereby attenuate cell growth. This G protein-independent inhibitory effect has been mapped to a 37 amino acid domain (RGS2eb) within RGS2 that binds to eukaryotic initiation factor 2B (eIF2B). When expressed in neonatal rat cardiomyocytes, RGS2eb attenuates both protein synthesis and hypertrophy induced by Gq- and Gs- activating agents. In the current study, we investigated the potential cardioprotective role of RGS2eb by determining whether RGS2eb transgenic (RGS2eb TG) mice with cardiomyocyte specific overexpression of RGS2eb show resistance to the development of hypertrophy in comparison to wild-type (WT) controls. Using transverse aortic constriction (TAC) in a pressure-overload hypertrophy model, we demonstrated that cardiac hypertrophy was inhibited in RGS2eb TG mice compared to WT controls following four weeks of TAC. Expression of the hypertrophic markers atrial natriuretic peptide (ANP) and ß-myosin heavy chain (MHC-ß) was also reduced in RGS2eb TG compared to WT TAC animals. Furthermore, cardiac function in RGS2eb TG TAC mice was significantly improved compared to WT TAC mice. Notably, cardiomyocyte cell size was significantly decreased in TG compared to WT TAC mice. These results suggest that RGS2 may limit pathological cardiac hypertrophy at least in part via the function of its eIF2B-binding domain.


Asunto(s)
Cardiomegalia/genética , Cardiomegalia/metabolismo , Expresión Génica , Miocitos Cardíacos/metabolismo , Dominios y Motivos de Interacción de Proteínas/genética , Proteínas RGS/genética , Transducción de Señal , Animales , Biomarcadores , Cardiomegalia/patología , Cardiomegalia/fisiopatología , Modelos Animales de Enfermedad , Pruebas de Función Cardíaca , Hemodinámica , Ratones , Ratones Transgénicos , Especificidad de Órganos/genética , Proteínas RGS/química
9.
Sci Signal ; 9(432): ra60, 2016 06 14.
Artículo en Inglés | MEDLINE | ID: mdl-27303056

RESUMEN

Anxiety and stress increase the frequency of epileptic seizures. These behavioral states induce the secretion of corticotropin-releasing factor (CRF), a 40-amino acid neuropeptide neurotransmitter that coordinates many behavioral responses to stress in the central nervous system. In the piriform cortex, which is one of the most seizurogenic regions of the brain, CRF normally dampens excitability. By contrast, CRF increased the excitability of the piriform cortex in rats subjected to kindling, a model of temporal lobe epilepsy. In nonkindled rats, CRF activates its receptor, a G protein (heterotrimeric guanosine triphosphate-binding protein)-coupled receptor, and signals through a Gαq/11-mediated pathway. After seizure induction, CRF signaling occurred through a pathway involving Gαs This change in signaling was associated with reduced abundance of regulator of G protein signaling protein type 2 (RGS2), which has been reported to inhibit Gαs-dependent signaling. RGS2 knockout mice responded to CRF in a similar manner as epileptic rats. These observations indicate that seizures produce changes in neuronal signaling that can increase seizure occurrence by converting a beneficial stress response into an epileptic trigger.


Asunto(s)
Epilepsia/metabolismo , Corteza Piriforme/metabolismo , Receptores de Hormona Liberadora de Corticotropina/metabolismo , Transducción de Señal , Animales , Hormona Liberadora de Corticotropina/genética , Hormona Liberadora de Corticotropina/metabolismo , Epilepsia/genética , Subunidades alfa de la Proteína de Unión al GTP/genética , Subunidades alfa de la Proteína de Unión al GTP/metabolismo , Masculino , Ratones , Ratones Noqueados , Corteza Piriforme/patología , Corteza Piriforme/fisiopatología , Proteínas RGS/genética , Proteínas RGS/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores de Hormona Liberadora de Corticotropina/genética
10.
Methods ; 92: 87-93, 2016 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-26297537

RESUMEN

Many GPCRs are able to activate multiple distinct signaling pathways, and these may include biochemical cascades activated via either heterotrimeric G proteins or by ß-arrestins. The relative potencies and/or efficacies among a series of agonists that act on a common receptor can vary depending upon which signaling pathway is being activated. This phenomenon is known as biased signaling or functional selectivity, and is presumed to reflect underlying differences in ligand binding affinities for alternate conformational states of the receptor. The first part of this review discusses how various cellular GPCR interacting proteins (GIPs) can influence receptor conformation and thereby affect ligand-receptor interactions and contribute to signaling bias. Upon activation, receptors trigger biochemical cascades that lead to altered cellular function, and measuring points along the cascade (e.g., second messenger production) conveys information about receptor activity. As a signal continues along its way, the observed concentration dependence of a GPCR ligand may change due to amplification and saturation of biochemical steps. The second part of this review considers additional cellular factors that affect signal processing, focusing mainly on structural elements and deamplification mechanisms, and discusses the relevance of these to measurements of potency and functional selectivity.


Asunto(s)
Proteínas RGS/metabolismo , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal/fisiología , Animales , Arrestinas/metabolismo , Arrestinas/farmacología , Humanos , Unión Proteica/fisiología , Transducción de Señal/efectos de los fármacos , beta-Arrestinas
11.
J Recept Signal Transduct Res ; 35(3): 208-12, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-26366680

RESUMEN

The GPCR gods smiled on us last year as the 15th Annual GPCR Retreat was held last October 2nd-4th in Bromont, Québec. The fall colors were at their peak and the meeting attendees were also in fine form. The program was one of the best we have seen at any GPCR-related meeting in years and there was a great deal of excitement about new methodological approaches to understanding receptor biology, new concepts in GPCR signaling and a continued emphasis on translation of these discoveries. This year was also the first year we opened the meeting with a short course on biased agonism and how to measure and analyze it.


Asunto(s)
Perfilación de la Expresión Génica/tendencias , Modelos Biológicos , Modelos Químicos , Biología Molecular/tendencias , Receptores Acoplados a Proteínas G/química , Receptores Acoplados a Proteínas G/metabolismo , Animales , Humanos , Modelos Moleculares
12.
Development ; 142(15): 2633-40, 2015 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-26160904

RESUMEN

During oocyte maturation, capacity and sensitivity of Ca(2+) signaling machinery increases dramatically, preparing the metaphase II (MII)-arrested egg for fertilization. Upon sperm-egg fusion, Ca(2+) release from IP3-sensitive endoplasmic reticulum stores results in cytoplasmic Ca(2+) oscillations that drive egg activation and initiate early embryo development. Premature Ca(2+) release can cause parthenogenetic activation prior to fertilization; thus, preventing inappropriate Ca(2+) signaling is crucial for ensuring robust MII arrest. Here, we show that regulator of G-protein signaling 2 (RGS2) suppresses Ca(2+) release in MII eggs. Rgs2 mRNA was recruited for translation during oocyte maturation, resulting in ∼ 20-fold more RGS2 protein in MII eggs than in fully grown immature oocytes. Rgs2-siRNA-injected oocytes matured to MII; however, they had increased sensitivity to low pH and acetylcholine (ACh), which caused inappropriate Ca(2+) release and premature egg activation. When matured in vitro, RGS2-depleted eggs underwent spontaneous Ca(2+) increases that were sufficient to cause premature zona pellucida conversion. Rgs2(-/-) females had reduced litter sizes, and their eggs had increased sensitivity to low pH and ACh. Rgs2(-/-) eggs also underwent premature zona pellucida conversion in vivo. These findings indicate that RGS2 functions as a brake to suppress premature Ca(2+) release in eggs that are poised on the brink of development.


Asunto(s)
Señalización del Calcio/fisiología , Calcio/metabolismo , Óvulo/fisiología , Proteínas RGS/metabolismo , Interacciones Espermatozoide-Óvulo/fisiología , Animales , Femenino , Técnica del Anticuerpo Fluorescente , Immunoblotting , Ratones , Óvulo/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Estadísticas no Paramétricas
13.
Mol Cell Biochem ; 405(1-2): 33-40, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25842189

RESUMEN

Heterotrimeric G protein signaling is limited by intracellular proteins that impede the binding of or accelerate the hydrolysis of the activating nucleotide GTP, exemplified respectively by the G protein-signaling modifier (GPSM) and regulator of G protein-signaling (RGS) families of proteins. Little is known about how members of these groups of proteins might influence the impact of the other on G protein activity. In the present study, we have identified novel binding and functional interactions between GPSM3 (also known as activator of G protein-signaling 4 (AGS4) or G18) and RGS5, both of which were found to be expressed in primary rat aortic smooth muscle cell cultures. The binding of GPSM3 to RGS5 appears to be selective as no interactions were detected with other RGS proteins tested. In solution-based experiments, the addition of GPSM3 was found to enhance the ability of RGS5 to accelerate GTP hydrolysis by Gαi1 but not that of RGS4. In membrane-based assays utilizing M2 muscarinic receptor-activated Gαi1, GPSM3 decreased the rate of GTP hydrolysis in the presence of RGS4 but not RGS5, suggesting that the enhancement of RGS5 activity by GPSM3 is maintained under these conditions and/or that the binding of RGS5 to GPSM3 impedes its inhibitory effect on GTP turnover. Overall these findings show that it is possible for GPSM and RGS proteins to bind to one another to produce distinct regulatory effects on heterotrimeric G protein activity.


Asunto(s)
Reguladores de Proteínas de Unión al GTP/metabolismo , Proteínas de Unión al GTP/metabolismo , Inhibidores de Disociación de Guanina Nucleótido/metabolismo , Proteínas de Unión al GTP Heterotriméricas/metabolismo , Proteínas RGS/metabolismo , Animales , Células CHO , Línea Celular , Cricetulus , Guanosina Trifosfato/metabolismo , Masculino , Miocitos del Músculo Liso/metabolismo , Unión Proteica/fisiología , Ratas , Ratas Endogámicas WKY , Transducción de Señal/fisiología
14.
Methods Mol Biol ; 1175: 121-52, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25150869

RESUMEN

The identification and characterization of the genes encoding G protein-coupled receptors (GPCRs) and the proteins necessary for the processes of ligand binding, GPCR activation, inactivation, and receptor trafficking to the membrane are discussed in the context of human genetic disease. In addition to functional GPCR variants, the identification of genetic disruptions affecting proteins necessary to GPCR functions have provided insights into the function of these pathways. Gsα and Gß subunit polymorphisms have been found to result in complex phenotypes. Disruptions in accessory proteins that normally modify or organize heterotrimeric G-protein coupling may also result in disease states. These include the contribution of variants of the regulator of G protein signaling (RGS) protein to hypertension; the role variants of the activator of G protein signaling (AGS) proteins to phenotypes (such as the type III AGS8 variant to hypoxia); the contribution of G protein-coupled receptor kinase (GRK) proteins, such as GRK4, in disorders such as hypertension. The role of accessory proteins in GPCR structure and function is discussed in the context of genetic disorders associated with disruption of the genes that encode them. An understanding of the pharmacogenomics of GPCR and accessory protein signaling provides the basis for examining both GPCR pharmacogenetics and the genetics of monogenic disorders that result from disruption of given receptor systems.


Asunto(s)
Quinasas de Receptores Acoplados a Proteína-G/metabolismo , Farmacogenética , Proteínas RGS/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal , Animales , Quinasas de Receptores Acoplados a Proteína-G/genética , Humanos , Hipertensión/tratamiento farmacológico , Hipertensión/genética , Hipertensión/metabolismo , Fenotipo , Polimorfismo Genético , Proteínas RGS/genética , Receptores Acoplados a Proteínas G/química , Receptores Acoplados a Proteínas G/genética
15.
Cell Signal ; 26(6): 1226-34, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24576550

RESUMEN

The protective effect of Regulator of G protein Signaling 2 (RGS2) in cardiac hypertrophy is thought to occur through its ability to inhibit the chronic GPCR signaling that promotes pathogenic growth both in vivo and in cultured cardiomyocytes. However, RGS2 is known to have additional functions beyond its activity as a GTPase accelerating protein, such as the ability to bind to eukaryotic initiation factor, eIF2B, and inhibit protein synthesis. The RGS2 eIF2B-interacting domain (RGS2(eb)) was examined for its ability to regulate hypertrophy in neonatal ventricular myocytes. Both full-length RGS2 and RGS2(eb) were able to inhibit agonist-induced cardiomyocyte hypertrophy, but RGS2(eb) had no effect on receptor-mediated inositol phosphate production, cAMP production, or ERK 1/2 activation. These results suggest that the protective effects of RGS2 in cardiac hypertrophy may derive at least in part from its ability to govern protein synthesis.


Asunto(s)
Cardiomegalia/metabolismo , Miocitos Cardíacos/metabolismo , Proteínas RGS/fisiología , Receptores Acoplados a Proteínas G/agonistas , Animales , Animales Recién Nacidos , Tamaño de la Célula/efectos de los fármacos , Células Cultivadas , AMP Cíclico/metabolismo , Factor 2B Eucariótico de Iniciación , Expresión Génica , Fosfatos de Inositol/metabolismo , Isoproterenol/farmacología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/patología , Fenilefrina/farmacología , Biosíntesis de Proteínas , Dominios y Motivos de Interacción de Proteínas , Proteínas RGS/química , Ratas , Receptores Acoplados a Proteínas G/fisiología , Sistemas de Mensajero Secundario
16.
Prog Mol Biol Transl Sci ; 115: 421-53, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23415100

RESUMEN

Heterotrimeric G proteins convey receptor signals to intracellular effectors. Superimposed over the basic GPCR-G protein-effector scheme are three types of auxiliary proteins that also modulate Gα. Regulator of G protein signaling proteins and G protein signaling modifier proteins respectively promote GTPase activity and hinder GTP/GDP exchange to limit Gα activation. There are also diverse proteins that, like GPCRs, can promote nucleotide exchange and thus activation. Here we review the impact of these auxiliary proteins on GPCR signaling. Although their precise physiological functions are not yet clear, all of them can produce significant effects in experimental systems. These signaling changes are generally consistent with established effects on isolated Gα; however, the activation state of Gα is seldom verified and many such changes appear also to reflect the physical disruption of or indirect effects on interactions between Gα and its associated GPCR, Gßγ, and/or effector.


Asunto(s)
Proteínas de Unión al GTP/metabolismo , Espacio Intracelular/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal , Animales , Humanos , Modelos Biológicos , Unión Proteica
17.
J Recept Signal Transduct Res ; 33(3): 129-34, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23351073

RESUMEN

In London, Ontario, the 13th Annual Joint meeting of the Great Lakes GPCR Retreat and the Club des Récepteurs à Sept Domaines Transmembranaires (known simply as the GPCR Retreat) was held on 17-19 October 2012, organized by Steve Ferguson and Peter Chidiac. This meeting gathered together a core group of investigators from Michigan, Ontario and Québec and has steadily increased its attendance in both the eastern (Europe) and western (USA, Canada) directions. This year's buzz naturally centered around the Nobel Prize in Chemistry, which was won the week before by Brian Kobilka and Robert Lefkowitz for their work on receptor structure and function. Michel Bouvier provided a heartfelt tribute to one of the attendees, Marc Caron, a pioneer in the GPCR field, has made many contributions to the work that led to this year's Nobel Prize. The meeting featured interesting sessions on the physiological roles of GPCRs in the nervous system, circadian biology and cancer, dealing at the cellular and molecular level with GPCR, G protein and effector structure and function, regulation and trafficking--with an overall focus on how to move molecular pharmacology in vivo.


Asunto(s)
Proteínas de Unión al GTP , Receptores Acoplados a Proteínas G , Transducción de Señal/genética , Animales , Canadá , Proteínas de Unión al GTP/genética , Proteínas de Unión al GTP/metabolismo , Proteínas de Unión al GTP/fisiología , Humanos , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Receptores Acoplados a Proteínas G/fisiología , Estados Unidos
18.
J Cell Biochem ; 114(6): 1414-23, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23255434

RESUMEN

RGS14 is a 60 kDa protein that contains a regulator of G protein signaling (RGS) domain near its N-terminus, a central region containing a pair of tandem Ras-binding domains (RBD), and a GPSM (G protein signaling modulator) domain (a.k.a. Gi/o-Loco binding [GoLoco] motif) near its C-terminus. The RGS domain of RGS14 exhibits GTPase accelerating protein (GAP) activity toward Gαi/o proteins, while its GPSM domain acts as a guanine nucleotide dissociation inhibitor (GDI) on Gαi1 and Gαi3. In the current study, we investigate the contribution of different domains of RGS14 to its biochemical functions. Here we show that the full-length protein has a greater GTPase activating activity but a weaker inhibition of nucleotide dissociation relative to its isolated RGS and GPSM regions, respectively. Our data suggest that these differences may be attributable to an inter-domain interaction within RGS14 that promotes the activity of the RGS domain, but simultaneously inhibits the activity of the GPSM domain. The RBD region seems to play an essential role in this regulatory activity. Moreover, this region of RGS14 is also able to bind to members of the B/R4 subfamily of RGS proteins and enhance their effects on GPCR-activated Gi/o proteins. Overall, our results suggest a mechanism wherein the RBD region associates with the RGS domain region, producing an intramolecular interaction within RGS14 that enhances the GTPase activating function of its RGS domain while disfavoring the negative effect of its GPSM domain on nucleotide dissociation.


Asunto(s)
Proteínas de Unión al GTP Heterotriméricas/metabolismo , Proteínas RGS/fisiología , Carbacol/farmacología , Agonistas Colinérgicos/farmacología , Activación Enzimática , Humanos , Antagonistas Muscarínicos/farmacología , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Proteínas RGS/química , Receptor Muscarínico M2/metabolismo , Receptores Adrenérgicos alfa 2/metabolismo , Células Sf9 , Transducción de Señal , Tropicamida/farmacología
19.
Biochem Biophys Res Commun ; 426(1): 129-34, 2012 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-22922103

RESUMEN

Regulator of G protein signaling (RGS) proteins are GTPase accelerating proteins for heterotrimeric G protein α-subunits. RGS2 has recently been shown to have additional G protein-independent functions including control of ion channel currents, microtubule polymerization, and protein synthesis. Cellular levels of RGS2 mRNA and protein are upregulated in response to various forms of stress suggesting that it may be a stress-adaptive protein; however, direct evidence to support this notion has remained elusive. In this report, we show that thermal stress upregulates RGS2 expression and this serves to arrest de novo protein synthesis. The latter is an established cellular response to stress. Inhibiting the stress-induced RGS2 upregulation by way of siRNA knockdown diminished the repression of global protein synthesis. The collective results of our study implicate RGS2 upregulation as a cellular mechanism of controlling de novo protein synthesis in response to stress. This work provides greater insight into the stress proteome and the role of RGS2.


Asunto(s)
Proteínas RGS/metabolismo , Estrés Fisiológico , Animales , Apoptosis , Línea Celular , Factor 2 Eucariótico de Iniciación/genética , Factor 2 Eucariótico de Iniciación/metabolismo , Fibroblastos/fisiología , Técnicas de Silenciamiento del Gen , Respuesta al Choque Térmico , Ratones , Biosíntesis de Proteínas , Proteoma/genética , Proteoma/metabolismo , Proteínas RGS/genética , ARN Interferente Pequeño/genética
20.
Front Physiol ; 3: 239, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22754542

RESUMEN

The heart receives sympathetic and parasympathetic efferent innervation as well as the ability to process information internally via an intrinsic cardiac autonomic nervous system (ICANS). For over a century, the role of the parasympathetics via vagal acetylcholine release was related to controlling primarily heart rate. Although in the late 1800s shown to play a role in atrial arrhythmia, the myocardium took precedence from the mid-1950s until in the last decade a resurgence of interest in the autonomics along with signaling cascades, regulators, and ion channels. Originally ignored as being benign and thus untreated, recent emphasis has focused on atrial arrhythmia as atrial fibrillation (AF) is the most common arrhythmia seen by the general practitioner. It is now recognized to have significant mortality and morbidity due to resultant stroke and heart failure. With the aging population, there will be an unprecedented increased burden on health care resources. Although it has been known for more than half a century that cholinergic stimulation can initiate AF, the classical concept focused on the M2 receptor and its signaling cascade including RGS4, as these had been shown to have predominant effects on nodal function (heart rate and conduction block) as well as contractility. However, recent evidence suggests that the M3 receptor may also playa role in initiation and perpetuation of AF and thus RGS2, a putative regulator of the M3 receptor, may be a target for therapeutic intervention. Mice lacking RGS2 (RGS2(-/-)), were found to have significantly altered electrophysiological atrial responses and were more susceptible to electrically induced AF. Vagally induced or programmed stimulation-induced AF could be blocked by the selective M3R antagonist, darifenacin. These results suggest a potential surgical target (ICANS) and pharmacological targets (M3R, RGS2) for the management of AF.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA