Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
BMC Pharmacol Toxicol ; 15: 29, 2014 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-24899231

RESUMEN

BACKGROUND: Regulator of G protein signaling (RGS) proteins suppress G protein coupled receptor signaling by catalyzing the hydrolysis of Gα-bound guanine nucleotide triphosphate. Transgenic mice in which RGS-mediated regulation of Gαi2 is lost (RGS insensitive Gαi2G184S) exhibit beneficial (protection against ischemic injury) and detrimental (enhanced fibrosis) cardiac phenotypes. This mouse model has revealed the physiological significance of RGS/Gαi2 interactions. Previous studies of the Gαi2G184S mutation used mice that express this mutant protein throughout their lives. Thus, it is unclear whether these phenotypes result from chronic or acute Gαi2G184S expression. We addressed this issue by developing mice that conditionally express Gαi2G184S. METHODS: Mice that conditionally express RGS insensitive Gαi2G184S were generated using a floxed minigene strategy. Conditional expression of Gαi2G184S was characterized by reverse transcription polymerase chain reaction and by enhancement of agonist-induced inhibition of cAMP production in isolated cardiac fibroblasts. The impact of conditional RGS insensitive Gαi2G184S expression on ischemic injury was assessed by measuring contractile recovery and infarct sizes in isolated hearts subjected to 30 min ischemia and 2 hours reperfusion. RESULTS: We demonstrate tamoxifen-dependent expression of Gαi2G184S, enhanced inhibition of cAMP production, and cardioprotection from ischemic injury in hearts conditionally expressing Gαi2G184S. Thus the cardioprotective phenotype previously reported in mice expressing Gαi2G184S does not require embryonic or chronic Gαi2G184S expression. Rather, cardioprotection occurs following acute (days rather than months) expression of Gαi2G184S. CONCLUSIONS: These data suggest that RGS proteins might provide new therapeutic targets to protect the heart from ischemic injury. We anticipate that this model will be valuable for understanding the time course (chronic versus acute) and mechanisms of other phenotypic changes that occur following disruption of interactions between Gαi2 and RGS proteins.


Asunto(s)
Subunidad alfa de la Proteína de Unión al GTP Gi2/genética , Subunidad alfa de la Proteína de Unión al GTP Gi2/metabolismo , Isquemia Miocárdica/metabolismo , Proteínas RGS/metabolismo , Animales , Células Cultivadas , AMP Cíclico/metabolismo , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Lisofosfolípidos/farmacología , Ratones , Ratones Transgénicos , Mutación , Isquemia Miocárdica/genética , Miocardio/citología , Miocardio/metabolismo , Tamoxifeno
2.
Mamm Genome ; 25(5-6): 202-10, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24700286

RESUMEN

G protein-coupled receptors strongly modulate neuronal excitability but there has been little evidence for G protein mechanisms in genetic epilepsies. Recently, four patients with epileptic encephalopathy (EIEE17) were found to have mutations in GNAO1, the most abundant G protein in brain, but the mechanism of this effect is not known. The GNAO1 gene product, Gαo, negatively regulates neurotransmitter release. Here, we report a dominant murine model of Gnao1-related seizures and sudden death. We introduced a genomic gain-of-function knock-in mutation (Gnao1 (+/G184S)) that prevents Go turnoff by Regulators of G protein signaling proteins. This results in rare seizures, strain-dependent death between 15 and 40 weeks of age, and a markedly increased frequency of interictal epileptiform discharges. Mutants on a C57BL/6J background also have faster sensitization to pentylenetetrazol (PTZ) kindling. Both premature lethality and PTZ kindling effects are suppressed in the 129SvJ mouse strain. We have mapped a 129S-derived modifier locus on Chromosome 17 (within the region 41-70 MB) as a Modifer of G protein Seizures (Mogs1). Our mouse model suggests a novel gain-of-function mechanism for the newly defined subset of epileptic encephalopathy (EIEE17). Furthermore, it reveals a new epilepsy susceptibility modifier Mogs1 with implications for the complex genetics of human epilepsy as well as sudden death in epilepsy.


Asunto(s)
Modelos Animales de Enfermedad , Epilepsia/genética , Epilepsia/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo , Mutación , Animales , Encéfalo/metabolismo , Encéfalo/patología , Epilepsia/mortalidad , Epilepsia/patología , Femenino , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/genética , Técnicas de Sustitución del Gen , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL
3.
Naunyn Schmiedebergs Arch Pharmacol ; 385(5): 443-53, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22411356

RESUMEN

Receptors coupled to G proteins have many effects on the heart. Enhanced signaling by Gα(s) and Gα(q) leads to cardiac injury and heart failure, while Gα(i2) signaling in cardiac myocytes can protect against ischemic injury and ß-adrenergic-induced heart failure. We asked whether enhanced Gα(i2) signaling in mice could protect against heart failure using a point mutation in Gα(i2) (G184S), which prevents negative regulation by regulators of G protein signaling. Contrary to our expectation, it worsened effects of a genetic dilated cardiomyopathy (DCM) and catecholamine-induced cardiac injury. Gα (i2) (G184S/+) /DCM double heterozygote mice (TG9(+)Gα (i2) (G184S/+)) had substantially decreased survival compared to DCM animals. Furthermore, heart weight/body weight ratios (HW/BW) were significantly greater in TG9(+)Gα (i2) (G184S/+) mice as was expression of natriuretic peptide genes. Catecholamine injury in Gα (i2) (G184S/G184S) mutant mice produced markedly increased isoproterenol-induced fibrosis and collagen III gene expression vs WT mice. Cardiac fibroblasts from Gα (i2) (G184S/G184S) mice also showed a serum-dependent increase in proliferation and ERK phosphorylation, which were blocked by pertussis toxin and a mitogen-activated protein/extracellular signal-regulated kinase kinase inhibitor. Gα(i2) signaling in cardiac myocytes protects against ischemic injury but enhancing Gα(i2) signaling overall may have detrimental effects in heart failure, perhaps through actions on cardiac fibroblasts.


Asunto(s)
Cardiomiopatía Dilatada/metabolismo , Subunidad alfa de la Proteína de Unión al GTP Gi2/metabolismo , Insuficiencia Cardíaca/metabolismo , Animales , Cardiomiopatía Dilatada/patología , Catecolaminas/toxicidad , Proliferación Celular , Células Cultivadas , Colágeno Tipo III/genética , Modelos Animales de Enfermedad , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Femenino , Fibroblastos/metabolismo , Fibroblastos/patología , Fibrosis , Subunidad alfa de la Proteína de Unión al GTP Gi2/genética , Insuficiencia Cardíaca/inducido químicamente , Insuficiencia Cardíaca/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Miocardio/metabolismo , Miocardio/patología
4.
Methods Mol Biol ; 756: 75-98, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21870221

RESUMEN

The Regulator of G protein Signaling (RGS) proteins were identified as a family in 1996 and humans have more than 30 such proteins. Their best known function is to suppress G Protein-Coupled Receptors (GPCR) signaling by increasing the rate of Gα turnoff through stimulation of GTPase activity (i.e., GTPase acceleration protein or GAP activity). The GAP activity of RGS proteins on the Gαi and Gαq family of G proteins can terminate signals initiated by both α and ßγ subunits. RGS proteins also serve as scaffolds, assembling signal-regulating modules. Understanding the physiological roles of RGS proteins is of great importance, as GPCRs are major targets for drug development. The traditional method of using RGS knockout mice has provided some information about the role of RGS proteins but in many cases effects are modest, perhaps because of redundancy in RGS protein function. As an alternative approach, we have utilized a glycine-to-serine mutation in the switch 1 region of Gα subunits that prevents RGS binding. The mutation has no known effects on Gα binding to receptor, Gßγ, or effectors. Alterations in function resulting from the G>S mutation imply a role for both the specific mutated Gα subunit and its regulation by RGS protein activity. Mutant rodents expressing these G>S mutant Gα subunits have strong phenotypes and provide important information about specific physiological functions of Gαi2 and Gαo and their control by RGS. The conceptual framework behind this approach and a summary of recent results is presented in this chapter.


Asunto(s)
Subunidades de Proteína/metabolismo , Proteínas RGS/metabolismo , Transducción de Señal , Animales , Marcación de Gen/métodos , Humanos , Mutación , Subunidades de Proteína/genética , Proteínas RGS/genética , Receptores Acoplados a Proteínas G/metabolismo
5.
Med Hypotheses ; 73(1): 56-9, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19282110

RESUMEN

Noonan syndrome (NS OMIM 163950) is a relatively common autosomal dominant developmental disorder characterized by short stature, specific facial features, and congenital cardiac anomalies. Approximately 50-66% of cases have defined mutations in the K-ras/Raf/MEK/ERK pathway that lead to constitutive signaling, but a significant number remain unexplained. We hypothesize that enhanced signaling through Galpha(i2) (from the GNAI2 gene) may also produce a NS-like phenotype. This is based on a recently described mouse model in which RGS-mediated inhibition of Galpha(i2) is prevented by a knock-in mutation (G184S) that blocks RGS binding [Huang et al., Mol. Cell. Biol. 2006;26:6870-9]. The mice have short body length, cardiac hypertrophy, a triangular face with wide-set eyes and ears, and hematologic alterations. There is a slight increase in ERK activation and a pronounced enhancement of PI3K/Akt phosphorylation in MEFs from these mice suggesting that abnormal increases in Galpha(i2) signaling could represent a novel upstream mechanism for NS. This suggests a novel set of candidate genes for NS (GNAI2 and RGS proteins) and if validated could have important implications for therapy as well.


Asunto(s)
Subunidad alfa de la Proteína de Unión al GTP Gi2/metabolismo , Proteínas de Unión al GTP/metabolismo , Modelos Biológicos , Síndrome de Noonan/metabolismo , Transducción de Señal , Animales , Humanos , Ratones
6.
Mol Pharmacol ; 75(5): 1222-30, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19225179

RESUMEN

Activation of G protein-coupled alpha(2) adrenergic receptors (ARs) inhibits epileptiform activity in the hippocampal CA3 region. The specific mechanism underlying this action is unclear. This study investigated which subtype(s) of alpha(2)ARs and G proteins (Galpha(o) or Galpha(i)) are involved in this response using recordings of mouse hippocampal CA3 epileptiform bursts. Application of epinephrine (EPI) or norepinephrine (NE) reduced the frequency of bursts in a concentration-dependent manner: (-)EPI > (-)NE >>> (+)NE. To identify the alpha(2)AR subtype involved, equilibrium dissociation constants (pK(b)) were determined for the selective alphaAR antagonists atipamezole (8.79), rauwolscine (7.75), 2-(2,6-dimethoxyphenoxyethyl)aminomethyl-1,4-benzodioxane hydrochloride (WB-4101; 6.87), and prazosin (5.71). Calculated pK(b) values correlated best with affinities determined previously for the mouse alpha(2A)AR subtype (r = 0.98, slope = 1.07). Furthermore, the inhibitory effects of EPI were lost in hippocampal slices from alpha(2A)AR-but not alpha(2C)AR-knockout mice. Pretreatment with pertussis toxin also reduced the EPI-mediated inhibition of epileptiform bursts. Finally, using knock-in mice with point mutations that disrupt regulator of G protein signaling (RGS) binding to Galpha subunits to enhance signaling by that G protein, the EPI-mediated inhibition of bursts was significantly more potent in slices from RGS-insensitive Galpha(o)(G184S) heterozygous (Galpha(o)+/GS) mice compared with either Galpha(i2)(G184S) heterozygous (Galpha(i2)+/GS) or control mice (EC(50) = 2.5 versus 19 and 23 nM, respectively). Together, these findings indicate that the inhibitory effect of EPI on hippocampal CA3 epileptiform activity uses an alpha(2A)AR/Galpha(o) protein-mediated pathway under strong inhibitory control by RGS proteins. This suggests a possible role for RGS inhibitors or selective alpha(2A)AR agonists as a novel antiepileptic drug therapy.


Asunto(s)
Subunidades alfa de la Proteína de Unión al GTP Gi-Go/fisiología , Hipocampo/efectos de los fármacos , Proteínas RGS/fisiología , Receptores Adrenérgicos alfa 2/fisiología , Antagonistas de Receptores Adrenérgicos alfa 2 , Animales , Epinefrina/farmacología , Femenino , Hipocampo/fisiología , Imidazoles/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Norepinefrina/farmacología , Oximetazolina/farmacología , Toxina del Pertussis/farmacología
7.
Diabetes ; 57(1): 77-85, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17928396

RESUMEN

OBJECTIVE: Guanine nucleotide binding protein (G protein)-mediated signaling plays major roles in endocrine/metabolic function. Regulators of G protein signaling (RGSs, or RGS proteins) are responsible for the subsecond turn off of G protein signaling and are inhibitors of signal transduction in vitro, but the physiological function of RGS proteins remains poorly defined in part because of functional redundancy. RESEARCH DESIGN AND METHODS: We explore the role of RGS proteins and G alpha(i2) in the physiologic regulation of body weight and glucose homeostasis by studying genomic "knock-in" mice expressing RGS-insensitive G alpha(i2) with a G184S mutation that blocks RGS protein binding and GTPase acceleration. RESULTS: Homozygous G alpha(i2)(G184S) knock-in mice show slightly reduced adiposity. On a high-fat diet, male G alpha(i2)(G184S) mice are resistant to weight gain, have decreased body fat, and are protected from insulin resistance. This appears to be a result of increased energy expenditure. Both male and female G alpha(i2)(G184S) mice on a high-fat diet also exhibit enhanced insulin sensitivity and increased glucose tolerance despite females having similar weight gain and adiposity compared with wild-type female mice. CONCLUSIONS: RGS proteins and G alpha(i2) signaling play important roles in the control of insulin sensitivity and glucose metabolism. Identification of the specific RGS proteins involved might permit their consideration as potential therapeutic targets for obesity-related insulin resistance and type 2 diabetes.


Asunto(s)
Dieta , Grasas de la Dieta , Reguladores de Proteínas de Unión al GTP/fisiología , Subunidad alfa de la Proteína de Unión al GTP Gi2/genética , Insulina/farmacología , Obesidad/genética , Tejido Adiposo/anatomía & histología , Sustitución de Aminoácidos , Animales , Glucemia/metabolismo , Peso Corporal , Cruzamientos Genéticos , Ingestión de Energía , Femenino , Prueba de Tolerancia a la Glucosa , Inmunidad Innata/genética , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Obesidad/prevención & control , Consumo de Oxígeno , Transducción de Señal/fisiología , Triglicéridos
8.
Mol Cell Biol ; 26(18): 6870-9, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16943428

RESUMEN

Signal transduction via guanine nucleotide binding proteins (G proteins) is involved in cardiovascular, neural, endocrine, and immune cell function. Regulators of G protein signaling (RGS proteins) speed the turn-off of G protein signals and inhibit signal transduction, but the in vivo roles of RGS proteins remain poorly defined. To overcome the redundancy of RGS functions and reveal the total contribution of RGS regulation at the Galpha(i2) subunit, we prepared a genomic knock-in of the RGS-insensitive G184S Gnai2 allele. The Galpha(i2)(G184S) knock-in mice show a dramatic and complex phenotype affecting multiple organ systems (heart, myeloid, skeletal, and central nervous system). Both homozygotes and heterozygotes demonstrate reduced viability and decreased body weight. Other phenotypes include shortened long bones, a markedly enlarged spleen, elevated neutrophil counts, an enlarged heart, and behavioral hyperactivity. Heterozygous Galpha(i2)(+/G184S) mice show some but not all of these abnormalities. Thus, loss of RGS actions at Galpha(i2) produces a dramatic and pleiotropic phenotype which is more evident than the phenotype seen for individual RGS protein knockouts.


Asunto(s)
Alelos , Subunidad alfa de la Proteína de Unión al GTP Gi2/genética , Genoma/genética , Fenotipo , Proteínas RGS/metabolismo , Animales , Anomalías Cardiovasculares , Embrión de Mamíferos/citología , Femenino , Fibroblastos/citología , Fibroblastos/metabolismo , Trastornos del Crecimiento , Enfermedades Hematológicas , Homocigoto , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Receptores del Ácido Lisofosfatídico/metabolismo , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA