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










Base de datos
Intervalo de año de publicación
1.
Nat Chem Biol ; 16(3): 240-249, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32080630

RESUMEN

Cholinesterase inhibitors, the current frontline symptomatic treatment for Alzheimer's disease (AD), are associated with low efficacy and adverse effects. M1 muscarinic acetylcholine receptors (M1 mAChRs) represent a potential alternate therapeutic target; however, drug discovery programs focused on this G protein-coupled receptor (GPCR) have failed, largely due to cholinergic adverse responses. Employing novel chemogenetic and phosphorylation-deficient, G protein-biased, mouse models, paired with a toolbox of probe molecules, we establish previously unappreciated pharmacologically targetable M1 mAChR neurological processes, including anxiety-like behaviors and hyper-locomotion. By mapping the upstream signaling pathways regulating these responses, we determine the importance of receptor phosphorylation-dependent signaling in driving clinically relevant outcomes and in controlling adverse effects including 'epileptic-like' seizures. We conclude that M1 mAChR ligands that promote receptor phosphorylation-dependent signaling would protect against cholinergic adverse effects in addition to driving beneficial responses such as learning and memory and anxiolytic behavior relevant for the treatment of AD.


Asunto(s)
Receptor Muscarínico M1/genética , Receptor Muscarínico M1/metabolismo , Acetilcolinesterasa/metabolismo , Enfermedad de Alzheimer/tratamiento farmacológico , Animales , Colinérgicos/farmacología , Inhibidores de la Colinesterasa/metabolismo , Inhibidores de la Colinesterasa/farmacología , Modelos Animales de Enfermedad , Diseño de Fármacos , Femenino , Técnicas de Sustitución del Gen , Masculino , Ratones , Ratones Endogámicos C57BL , Fosforilación
2.
Cell Signal ; 51: 86-98, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30075183

RESUMEN

Generation of cAMP through Gs-coupled G protein-coupled receptor (GPCR) [e.g. ß2-adrenoceptor (ß2AR), adenosine A2B receptor (A2BR)] activation, induces arterial smooth muscle relaxation, counteracting the actions of vasoconstrictors. Gs-coupled GPCR signalling is regulated by G protein-coupled receptor kinases (GRK) and arrestin proteins, and dysregulation of Gs/GPCR signalling is thought play a role in the development of hypertension, which may be a consequence of enhanced GRK2 and/or arrestin expression. However, despite numerous studies indicating that ß2AR and A2BR can be substrates for GRK/arrestin proteins, currently little is known regarding GRK/arrestin regulation of these endogenous receptors in arterial smooth muscle. Here, endogenous GRK isoenzymes and arrestin proteins were selectively depleted using RNA-interference in rat arterial smooth muscle cells (RASM) and the consequences of this for ß2AR- and A2BR-mediated adenylyl cyclase (AC) signalling were determined by assessing cAMP accumulation. GRK2 or GRK5 depletion enhanced and prolonged ß2AR/AC signalling, while combined deletion of GRK2/5 has an additive effect. Conversely, activation of AC by A2BR was regulated by GRK5, but not GRK2. ß2AR desensitization was attenuated following combined GRK2/GRK5 knockdown, but not by depletion of individual GRKs, arrestins, or by inhibiting PKA. Arrestin3 (but not arrestin2) depletion enhanced A2BR-AC signalling and attenuated A2BR desensitization, while ß2AR-AC signalling was regulated by both arrestin isoforms. This study provides a first demonstration of how different complements of GRK and arrestin proteins contribute to the regulation of signalling and desensitization of these important receptors mediating vasodilator responses in arterial smooth muscle.


Asunto(s)
Aorta/metabolismo , Quinasa 2 del Receptor Acoplado a Proteína-G/fisiología , Quinasa 5 del Receptor Acoplado a Proteína-G/fisiología , Quinasas de Receptores Acoplados a Proteína-G/fisiología , Músculo Liso/metabolismo , Miocitos del Músculo Liso/metabolismo , Receptor de Adenosina A2B/metabolismo , Receptores Adrenérgicos beta 2/metabolismo , Arrestina beta 2/fisiología , Adenilil Ciclasas/metabolismo , Animales , Aorta/citología , Arrestinas/genética , Arrestinas/fisiología , Células Cultivadas , Quinasa 2 del Receptor Acoplado a Proteína-G/genética , Quinasa 5 del Receptor Acoplado a Proteína-G/genética , Músculo Liso/citología , Miocitos del Músculo Liso/citología , Ratas , Ratas Wistar , Transducción de Señal , Arrestina beta 2/genética
3.
BMC Pulm Med ; 15: 12, 2015 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-25880173

RESUMEN

BACKGROUND: Asthma is characterized by airway hyper-responsiveness and variable airflow obstruction, in part as a consequence of hyper-contractile airway smooth muscle, which persists in primary cell culture. One potential mechanism for this hyper-contractility is abnormal intracellular Ca(2+) handling. METHODS: We sought to compare intracellular Ca(2+) handling in airway smooth muscle cells from subjects with asthma compared to non-asthmatic controls by measuring: i) bradykinin-stimulated changes in inositol 1,4,5-trisphosphate (IP3) accumulation and intracellular Ca(2+) concentration, ii) sarco/endoplasmic reticulum Ca(2+)-ATPase (SERCA) expression, iii) mechanisms of cytoplasmic Ca(2+) clearance assessed following instantaneous flash photolytic release of Ca(2+) into the cytoplasm. RESULTS: We found no differences in airway smooth muscle cell basal intracellular Ca(2+) concentrations, bradykinin-stimulated IP3 accumulation or intracellular Ca(2+) responses. Quantification of SERCA2 mRNA or protein expression levels revealed no differences in ASM cells obtained from subjects with asthma compared to non-asthmatic controls. We did not identify differences in intracellular calcium kinetics assessed by flash photolysis and calcium uncaging independent of agonist-activation with or without SERCA inhibition. However, we did observe some correlations in subjects with asthma between lung function and the different cellular measurements of intracellular Ca(2+) handling, with poorer lung function related to increased rate of recovery following flash photolytic elevation of cytoplasmic Ca(2+) concentration. CONCLUSIONS: Taken together, the experimental results reported in this study do not demonstrate major fundamental differences in Ca(2+) handling between airway smooth muscle cells from non-asthmatic and asthmatic subjects. Therefore, increased contraction of airway smooth muscle cells derived from asthmatic subjects cannot be fully explained by altered Ca(2+) homeostasis.


Asunto(s)
Asma/metabolismo , Calcio/metabolismo , Miocitos del Músculo Liso/metabolismo , ARN Mensajero/metabolismo , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/genética , Adulto , Asma/genética , Bradiquinina/farmacología , Bronquios/citología , Estudios de Casos y Controles , Femenino , Humanos , Inositol 1,4,5-Trifosfato/metabolismo , Masculino , Persona de Mediana Edad , Contracción Muscular , Miocitos del Músculo Liso/efectos de los fármacos , Fotólisis , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/metabolismo , Vasodilatadores/farmacología
4.
J Biol Chem ; 288(48): 34777-90, 2013 Nov 29.
Artículo en Inglés | MEDLINE | ID: mdl-24133207

RESUMEN

Class A G protein-coupled receptors (GPCRs) are able to form homodimers and/or oligomeric arrays. We recently proposed, based on bioluminescence resonance energy transfer studies with the M3 muscarinic receptor (M3R), a prototypic class A GPCR, that the M3R is able to form multiple, structurally distinct dimers that are probably transient in nature (McMillin, S. M., Heusel, M., Liu, T., Costanzi, S., and Wess, J. (2011) J. Biol. Chem. 286, 28584-28598). To provide more direct experimental support for this concept, we employed a disulfide cross-linking strategy to trap various M3R dimeric species present in a native lipid environment (transfected COS-7 cells). Disulfide cross-linking studies were carried out with many mutant M3Rs containing single cysteine (Cys) substitutions within two distinct cytoplasmic M3R regions, the C-terminal portion of the second intracellular loop (i2) and helix H8 (H8). The pattern of cross-links that we obtained, in combination with molecular modeling studies, was consistent with the existence of two structurally distinct M3R dimer interfaces, one involving i2/i2 contacts (TM4-TM5-i2 interface) and the other one characterized by H8-H8 interactions (TM1-TM2-H8 interface). Specific H8-H8 disulfide cross-links led to significant impairments in M3R-mediated G protein activation, suggesting that changes in the structural orientation or mobility of H8 are critical for efficient receptor-G protein coupling. Our findings provide novel structural and functional insights into the mechanisms involved in M3R dimerization (oligomerization). Because the M3R shows a high degree of sequence similarity with many other class A GPCRs, our findings should be of considerable general interest.


Asunto(s)
Conformación Proteica , Multimerización de Proteína , Receptor Muscarínico M3/química , Receptor Muscarínico M3/metabolismo , Secuencia de Aminoácidos , Animales , Sitios de Unión , Células COS , Chlorocebus aethiops , Cisteína/química , Mutagénesis Sitio-Dirigida , Fosfatidilinositoles/química , Fosfatidilinositoles/metabolismo , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Receptor Muscarínico M3/genética
5.
J Neurosci ; 33(13): 5626-37, 2013 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-23536077

RESUMEN

Nitric oxide (NO) is an important signaling molecule with a variety of functions in the CNS, including a potential role in modulating neuronal growth and synapse formation. In the present study, we used tractable, identified neurons in the CNS of the pond snail Lymnaea stagnalis to study the role of endogenous NO signaling in neuronal growth and synaptic remodeling after nerve injury. Axonal damage of L. stagnalis neurons B1 and B2 induces extensive central growth of neurites that is accompanied by changes in existing electrical connections, the transient formation of novel electrical connections, and the formation of a novel excitatory chemical synapse from B2 to B1 neurons. Partial chronic inhibition of endogenous NO synthesis reduces neurite growth in NO-synthase-expressing B2, but has only minor effects on NOS-negative B1 neurons. Chronic application of an NO donor while inhibiting endogenous NO synthesis rescues neurite extension in B2 neurons and boosts growth of B1 neurons. Blocking soluble guanylate cyclase activity completely suppresses neurite extension and synaptic remodeling after nerve crush, demonstrating the importance of cGMP in these processes. Interestingly, inhibition of cGMP-dependent protein kinase only suppresses chemical synapse formation without effects on neuronal growth and electrical synapse remodeling. We conclude that NO signaling via cGMP is an important modulator of both neurite growth and synaptic remodeling after nerve crush. However, differential effects of cGMP-dependent protein kinase inhibition on neurite growth and synaptic remodeling suggest that these effects are mediated by separate signaling pathways.


Asunto(s)
Axotomía , Sistema Nervioso Central/citología , GMP Cíclico/metabolismo , Neuritas/fisiología , Neuronas/patología , Óxido Nítrico/metabolismo , Sinapsis/fisiología , Potenciales de Acción/efectos de los fármacos , Potenciales de Acción/fisiología , Análisis de Varianza , Animales , Biofisica , Biotina/análogos & derivados , Células Cultivadas , Proteínas Quinasas Dependientes de GMP Cíclico/metabolismo , Estimulación Eléctrica , Electrofisiología , Inhibidores Enzimáticos/farmacología , Hidrazinas , Iontoforesis , Neuritas/efectos de los fármacos , Neuronas/efectos de los fármacos , Óxido Nítrico Sintasa/antagonistas & inhibidores , Óxido Nítrico Sintasa/metabolismo , Técnicas de Cultivo de Órganos , Caracoles , Sinapsis/patología
6.
PLoS One ; 7(1): e29946, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22272263

RESUMEN

BACKGROUND AND OBJECTIVE: Muscarinic acetylcholine receptors (mAChRs) are 7-transmembrane, G protein-coupled receptors that regulate a variety of physiological processes and represent potentially important targets for therapeutic intervention. mAChRs can be stimulated by full and partial orthosteric and allosteric agonists, however the relative abilities of such ligands to induce conformational changes in the receptor remain unclear. To gain further insight into the actions of mAChR agonists, we have developed a fluorescently tagged M(1) mAChR that reports ligand-induced conformational changes in real-time by changes in Förster resonance energy transfer (FRET). METHODS: Variants of CFP and YFP were inserted into the third intracellular loop and at the end of the C-terminus of the mouse M(1) mAChR, respectively. The optimized FRET receptor construct (M(1)-cam5) was expressed stably in HEK293 cells. RESULTS: The variant CFP/YFP-receptor chimera expressed predominantly at the plasma membrane of HEK293 cells and displayed ligand-binding affinities comparable with those of the wild-type receptor. It also retained an ability to interact with Gα(q/11) proteins and to stimulate phosphoinositide turnover, ERK1/2 phosphorylation and undergo agonist-dependent internalization. Addition of the full agonist methacholine caused a reversible decrease in M(1) FRET (F(EYFP)/F(ECFP)) that was prevented by atropine pre-addition and showed concentration-dependent amplitude and kinetics. Partial orthosteric agonists, arecoline and pilocarpine, as well as allosteric agonists, AC-42 and 77-LH-28-1, also caused atropine-sensitive decreases in the FRET signal, which were smaller in amplitude and significantly slower in onset compared to those evoked by methacholine. CONCLUSION: The M(1) FRET-based receptor chimera reports that allosteric and orthosteric agonists induce similar conformational changes in the third intracellular loop and/or C-terminus, and should prove to be a valuable molecular reagent for pharmacological and structural investigations of M(1) mAChR activation.


Asunto(s)
Membrana Celular/metabolismo , Transferencia Resonante de Energía de Fluorescencia/métodos , Agonistas Muscarínicos/metabolismo , Receptor Muscarínico M1/metabolismo , Animales , Arecolina/metabolismo , Arecolina/farmacología , Atropina/metabolismo , Atropina/farmacología , Unión Competitiva/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Células HEK293 , Humanos , Cinética , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Masculino , Cloruro de Metacolina/metabolismo , Cloruro de Metacolina/farmacología , Ratones , Microscopía Confocal , Agonistas Muscarínicos/farmacología , Piperidinas/metabolismo , Piperidinas/farmacología , Receptor Muscarínico M1/agonistas , Receptor Muscarínico M1/genética , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Transfección
7.
Acta Diabetol ; 49(4): 277-89, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21833779

RESUMEN

Muscarinic acetylcholine receptor (mAChR) activation of pancreatic ß-cells elevates intracellular Ca(2+) and potentiates glucose-stimulated insulin secretion. In addition, it activates a number of signaling molecules, including ERK1/2, whose activation has been shown to play an important role in regulating pancreatic ß-cell function and mass. The aim of this work was to determine how mAChR activation elevates intracellular Ca(2+) concentration ([Ca(2+)]( i )) and activates ERK1/2 in the pancreatic ß-cell line MIN6. We demonstrate that agonist-stimulated ERK1/2 activation is dependent on the activation of phospholipase C and an elevation in [Ca(2+)]( i ), but is independent of the activation of diacylglycerol-dependent protein kinase C isoenzymes. Using a pharmacological approach, we provide evidence that agonist-induced increases in [Ca(2+)]( i ) and ERK activity require (1) IP(3) receptor-mediated mobilization of Ca(2+) from the endoplasmic reticulum, (2) influx of extracellular Ca(2+) through store-operated channels, (3) closure of K(ATP) channels, and (4) Ca(2+) entry via L-type voltage-operated Ca(2+) channels. Moreover, this Ca(2+)-dependent activation of ERK is mediated via both Ras-dependent and Ras-independent mechanisms. In summary, this study provides important insights into the multifactorial signaling mechanisms linking mAChR activation to increases in [Ca(2+)]( i ) and ERK activity.


Asunto(s)
Calcio/metabolismo , Células Secretoras de Insulina/metabolismo , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Receptores Muscarínicos/fisiología , Carbacol/farmacología , Línea Celular , Agonistas Colinérgicos/farmacología , Activación Enzimática/fisiología , Receptores de Inositol 1,4,5-Trifosfato/fisiología , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Fosforilación , Transducción de Señal , Transfección , Fosfolipasas de Tipo C/antagonistas & inhibidores , Fosfolipasas de Tipo C/metabolismo
8.
FASEB J ; 26(2): 604-16, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22031716

RESUMEN

To explore the structural mechanisms underlying the assembly and activation of family A GPCR dimers, we used the rat M(3) muscarinic acetylcholine receptor (M3R) as a model system. Studies with Cys-substituted mutant M3Rs expressed in COS-7 cells led to the identification of several mutant M3Rs that exclusively existed as cross-linked dimers under oxidizing conditions. The cross-linked residues were located at the bottom of transmembrane domain 5 (TM5) and within the N-terminal portion of the third intracellular loop (i3 loop). Studies with urea-stripped membranes demonstrated that M3R disulfide cross-linking did not require the presence of heterotrimeric G proteins. Molecular modeling studies indicated that the cross-linking data were in excellent agreement with the existence of a low-energy M3R dimer characterized by a TM5-TM5 interface. [(35)S]GTPγS binding/Gα(q/11) immunoprecipitation assays revealed that an M3R dimer that was cross-linked within the N-terminal portion of the i3 loop (264C) was functionally severely impaired (∼50% reduction in receptor-G-protein coupling, as compared to control M3R). These data support the novel concept that agonist-induced activation of M3R dimers requires a conformational change of the N-terminal segment of the i3 loop. Given the high degree of structural homology among family A GPCRs, these findings should be of broad significance.


Asunto(s)
Receptor Muscarínico M3/química , Receptor Muscarínico M3/metabolismo , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Animales , Sitios de Unión/genética , Células COS , Chlorocebus aethiops , Reactivos de Enlaces Cruzados , Dimerización , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/metabolismo , Modelos Moleculares , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Proteínas Mutantes/química , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Estructura Cuaternaria de Proteína , Ratas , Receptor Muscarínico M3/genética , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
9.
Methods Mol Biol ; 746: 263-75, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21607862

RESUMEN

On activation, G-protein-coupled receptors (GPCRs) exert many of their cellular actions through -promoting guanine nucleotide exchange on Gα subunits of heterotrimeric G proteins to release Gα-GTP and free ßγ-subunits. In membrane preparations, GTP can be substituted by ³5S-labeled guanosine- 5'-O-(3-thio)triphosphate ([³5S]GTPγS) and on agonist stimulation a quasi-stable [³5S]GTPγS-Gα -complex forms and accumulates. Separation of [³5S]GTPγS-Gα complexes from free [³5S]GTPγS allows differences between basal and agonist-stimulated rates of [³5S]GTPγS-Gα complex formation- to be used to obtain pharmacological information on receptor-G-protein information transfer. Further, by releasing Gα-subunits into solution following the [³5S]GTPγS binding step, Gα-subunit-specific antibodies can be used to investigate the Gα-protein subpopulations activated by receptors by immunoprecipitation of [³5S]GTPγS-Gα complexes and quantification by scintillation counting. Here, we describe a total [³5S]GTPγS binding assay and a modification of this method that incorporates a Gα-specific immunoprecipitation step.


Asunto(s)
Proteínas de Unión al GTP/metabolismo , Guanosina Trifosfato/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Radioisótopos de Azufre/metabolismo , Animales , Bioensayo , Células CHO , Membrana Celular/metabolismo , Cricetinae , Cricetulus , Subunidades alfa de la Proteína de Unión al GTP/metabolismo , Unión Proteica
10.
Mol Pharmacol ; 79(5): 844-54, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21325018

RESUMEN

An initial stage of many neurodegenerative processes is associated with compromised synaptic function and precedes synapse loss, neurite fragmentation, and neuronal death. We showed previously that deficiency of heme, regulating many proteins of pharmacological importance, causes neurodegeneration of primary cortical neurons via N-methyl-d-aspartate receptor (NMDAR)-dependent suppression of the extracellular signal-regulated kinase 1/2 pathway. Here, we asked whether the reduction of heme causes synaptic perturbation before neurite fragmentation in neuronal cultures and investigated molecular mechanisms of synaptic dysfunction in these cells. We showed the change in the NR2B subunit phosphorylation that correlates with compromised NMDAR function after the reduction of regulatory heme and a rapid rescue of NR2B phosphorylation and NMDAR function by exogenous heme. Electrophysiological recordings demonstrated diminished NMDAR currents and NMDAR-mediated calcium influx after 24 h of inhibition of heme synthesis. These effects were reversed by treatment with heme; however, inhibition of the Src family kinases abolished the rescue effect of heme on NMDA-evoked currents. Diminished NMDAR current and Ca(2+) influx resulted in suppressed cGMP production and impairment of spine formation. Exogenous heme exerted rescue effects on NR2B tyrosine phosphorylation and NMDA-evoked currents within minutes, suggesting direct interactions within the NMDAR complex. These synaptic changes after inhibition of heme synthesis occurred at this stage without apparent dysfunction of major hemoproteins. We conclude that regulatory heme is necessary in maintaining NR2B phosphorylation and NMDAR function. NMDAR failure occurs before neurite fragmentation and may be a causal factor in neurodegeneration; this could suggest a route for an early pharmacological intervention.


Asunto(s)
Hemo/metabolismo , Neuronas/metabolismo , Receptores de N-Metil-D-Aspartato/fisiología , Adenosina Trifosfato/metabolismo , Animales , Secuencia de Bases , Cartilla de ADN , Femenino , Hemo/biosíntesis , Masculino , Ratones , Ratones Endogámicos BALB C , Oxidación-Reducción , Fosforilación , Reacción en Cadena de la Polimerasa , Receptores de N-Metil-D-Aspartato/metabolismo , Tirosina/metabolismo
11.
Mol Endocrinol ; 23(8): 1272-80, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19423652

RESUMEN

Oxytocin plays an important role in the progression, timing, and modulation of uterine contraction during labor and is widely used as an uterotonic agent. We investigated the mechanisms regulating oxytocin receptor (OTR) signaling in human primary myometrial smooth muscle cells and the ULTR cell-line. Oxytocin produced concentration-dependent increases in both total [(3)H]inositol phosphate accumulation and intracellular Ca(2+) concentration ([Ca(2+)](i)); however, responses were greater and more reproducible in the ULTR cell line. Assessment of phospholipase C activity in single cells revealed that the OTR desensitizes rapidly (within 5 min) in the presence of oxytocin (100 nm). To characterize OTR desensitization further, cells were stimulated with a maximally effective concentration of oxytocin (100 nm, 30 sec) followed by a variable washout period and a second identical application of oxytocin. This brief exposure to oxytocin caused a marked decrease (>70%) in OTR responsiveness to rechallenge and was fully reversed by increasing the time period between agonist challenges. To assess involvement of G protein-coupled receptor kinases (GRKs) in OTR desensitization, cells were transfected with small interfering RNAs to cause specific > or =75% knockdown of GRKs 2, 3, 5, or 6. In both primary myometrial and ULTR cells, knockdown of GRK6 largely prevented oxytocin-induced OTR desensitization; in contrast, selective depletion of GRKs 2, 3, or 5 was without effect. These data indicate that GRK6 recruitment is a cardinal effector of OTR responsiveness and provide mechanistic insight into the likely in vivo regulation of OTR signaling in uterine smooth muscle.


Asunto(s)
Quinasas de Receptores Acoplados a Proteína-G/metabolismo , Regulación Enzimológica de la Expresión Génica , Músculo Liso/metabolismo , Miometrio/metabolismo , Receptores de Oxitocina/metabolismo , Actinas/metabolismo , Calcio/metabolismo , Proteínas de Unión al Calcio/metabolismo , Línea Celular , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Proteínas de Microfilamentos/metabolismo , Modelos Biológicos , Oxitocina/metabolismo , Transducción de Señal , Calponinas
12.
J Pharmacol Exp Ther ; 327(2): 365-74, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18664591

RESUMEN

The M(1) muscarinic acetylcholine (mACh) receptor is among a growing number of G protein-coupled receptors that are able to activate multiple signaling cascades. AC-42 (4-n-butyl-1-[4-(2-methylphenyl)-4-oxo-1-butyl] piperidine) is an allosteric agonist that can selectively activate the M(1) mACh receptor in the absence of an orthosteric ligand. Allosteric agonists have the potential to stabilize unique receptor conformations, which may in turn cause differential activation of signal transduction pathways. In the present study, we have investigated the signaling pathways activated by AC-42, its analog 77-LH-28-1 (1-[3-(4-butyl-1-piperidinyl)propyl]-3,4-dihydro-2(1H)-quinolinone), and a range of orthosteric muscarinic agonists [oxotremorine-M (oxo-M), arecoline, and pilocarpine] in Chinese hamster ovary cells recombinantly expressing the human M(1) mACh receptor. Each agonist was able to activate Galpha(q/11)-dependent signaling, as demonstrated by an increase in guanosine 5'-O-(3-thiotriphosphate) ([(35)S]GTPgammaS) binding to Galpha(q/11) proteins and total [(3)H]inositol phosphate accumulation assays in intact cells. All three orthosteric agonists caused significant enhancements in [(35)S]GTPgammaS binding to Galpha(i1/2) subunits over basal; however, neither allosteric ligand produced a significant response. In contrast, both orthosteric and allosteric agonists are able to couple to the Galpha(s)/cAMP pathway, enhancing forskolin-stimulated cAMP accumulation. These data provide support for the concept that allosteric and orthosteric mACh receptor agonists both stabilize receptor conformations associated with Galpha(q/11)- and Galpha(s)-dependent signaling; however, AC-42 and 77-LH-28-1, unlike oxo-M, arecoline, and pilocarpine, do not seem to promote M(1) mACh receptor-Galpha(i1/2) coupling, suggesting that allosteric agonists have the potential to activate distinct subsets of downstream effectors.


Asunto(s)
Subunidades alfa de la Proteína de Unión al GTP Gq-G11/fisiología , Proteínas de Unión al GTP/fisiología , Agonistas Muscarínicos/farmacología , Piperidinas/farmacología , Quinolonas/farmacología , Receptor Muscarínico M1/agonistas , Transducción de Señal , Animales , Células CHO , Cricetinae , Cricetulus , AMP Cíclico/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/fisiología , Subunidades alfa de la Proteína de Unión al GTP Gs/fisiología , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Humanos , Inositol 1,4,5-Trifosfato/metabolismo , N-Metilescopolamina/metabolismo
13.
Br J Pharmacol ; 144(4): 566-75, 2005 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15655507

RESUMEN

1. A range of muscarinic acetylcholine (mACh) receptor agonists (methacholine (MCh), oxotremorine-M (OXO-M), oxotremorine (OXO), arecoline (AREC), bethanechol (BETH), pilocarpine (PILO)) have been investigated with respect to their binding to, and activation of, M(2) and M(4) mACh receptors, recombinantly expressed in Chinese hamster ovary cells, to explore the possibility that these agonists may differentially affect mACh receptor-G(i/o) and -G(s) coupling. 2. M(2)/M(4) mACh receptor coupling to the adenylyl cyclase/cyclic AMP signalling pathway has been explored in intact cells. G(i/o)-mediated negative coupling to adenylyl cyclase was explored functionally by assessing the ability of the mACh receptor agonists to inhibit forskolin-stimulated enzymic activity. Following pertussis toxin treatment (100 ng ml(-1), 18-20 h) to inactivate G(i/o) proteins, each agonist caused a G(s)-mediated enhancement of forskolin-stimulated adenylyl cyclase activity. 3. At both M(2) and M(4) mACh receptors, all agonists tested were more potent in mediating G(i/o)- versus G(s)-coupled responses. This difference (determined as the pIC(50) (G(i/o) coupling) minus pEC(50) (G(s) coupling) value) was greatest for AREC (65-75-fold) and least for BETH and PILO (

Asunto(s)
Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gs/metabolismo , Agonistas Muscarínicos/farmacología , Receptor Muscarínico M2/agonistas , Receptor Muscarínico M4/agonistas , Transducción de Señal/efectos de los fármacos , Animales , Unión Competitiva , Células CHO , Cricetinae , Cricetulus , Proteínas Recombinantes/metabolismo
14.
Mol Pharmacol ; 64(5): 1059-68, 2003 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-14573754

RESUMEN

Previously we have shown that G protein-coupled receptor kinase (GRK) 6 plays a major role in the regulation of the human M3 muscarinic acetylcholine receptor (M3 mAChR) in the human neuroblastoma SH-SY5Y. However, 30-fold overexpression of the catalytically inactive, dominant-negative K215RGRK6 produced only a 50% suppression of M3 mAChR phosphorylation and desensitization. Here, we have attempted to determine whether other endogenous kinases play a role in the regulation of M3 mAChR signaling. In contrast to the clear attenuating effect of K215RGRK6 expression on M3 mAChR regulation, dominant-negative forms of GRKs (K220RGRK2, K220RGRK3, K215RGRK5) and casein kinase 1alpha (K46RCK1alpha) were without effect. In addition, inhibition of a variety of second-messenger-regulated kinases and the tyrosine kinase Src also had no effect upon agonist-stimulated M3 mAChR regulation. To investigate further the desensitization process we have followed changes in inositol 1,4,5-trisphosphate in single SHSY5Y cells using the pleckstrin homology domain of PLCdelta1 tagged with green fluorescent protein (eGFP-PHPLCdelta1). Stimulation of cells with approximate EC50 concentrations of agonist before and after a desensitizing period of agonist exposure resulted in a marked attenuation of the latter response. Altered GRK6 activity, through overexpression of wild-type GRK6 or K215RGRK6, enhanced or reduced the degree of M3 mAChR desensitization, respectively. Taken together, our data indicate that M3 mAChR desensitization is mediated by GRK6 in human SH-SY5Y cells, and we show that receptor desensitization of phospholipase C signaling can be monitored in 'real-time' in single, living cells.


Asunto(s)
Proteínas Serina-Treonina Quinasas/metabolismo , Receptor Muscarínico M3/metabolismo , Transducción de Señal/fisiología , Atropina/farmacología , Línea Celular , Proteínas Quinasas Dependientes de AMP Cíclico/biosíntesis , Interacciones Farmacológicas , Quinasas de Receptores Acoplados a Proteína-G , Humanos , Agonistas Muscarínicos/farmacología , Antagonistas Muscarínicos/farmacología , Fosforilación/efectos de los fármacos , Proteína Quinasa C/metabolismo , Especificidad por Sustrato , Quinasas de Receptores Adrenérgicos beta
15.
Br J Pharmacol ; 138(7): 1340-50, 2003 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-12711635

RESUMEN

1. We have examined possible mechanisms of cross-talk between the G(q/11)-linked M(3) muscarinic acetylcholine (mACh) receptor and the G(i/o)-linked M(2) mACh receptor by stable receptor coexpression in Chinese hamster ovary (CHO) cells. A number of second messenger (cyclic AMP, Ins(1,4,5)P(3)) and mitogen-activated protein kinase (ERK and JNK) responses stimulated by the mACh receptor agonist methacholine were examined in CHO-m2m3 cells and compared to those stimulated in CHO-m2 and CHO-m3 cell-lines, expressing comparable levels of M(2) or M(3) mACh receptors. 2. Based on comparisons between cell-lines and pertussis toxin (PTx) pretreatment to eliminate receptor-G(i/o) coupling, evidence was obtained for (i) an M(2) mACh receptor-mediated contribution to the predominantly M(3) mACh receptor-mediated Ins(1,4,5)P(3) response and (ii) a facilitation of the inhibitory effect of M(2) mACh receptor on forskolin-stimulated cyclic AMP accumulation by M(3) mACh receptor coactivation at low agonist concentrations (MCh 10(-9)-10(-6) M). 3. The most profound cross-talk effects were observed with respect to ERK activation. Thus, while MCh stimulated ERK activation in both CHO-m2 and CHO-m3 cells (pEC(50) values: 5.64+/-0.09 and 5.57+/-0.16, respectively), the concentration-effect relation was approx 50-fold left-shifted in CHO-m2m3 cells (pEC(50): 7.17+/-0.07). In addition, the ERK response was greater and more sustained in CHO-m2m3 cells. In contrast, only minor differences were seen in the time-courses and concentration-dependencies of JNK activation in CHO-m3 and CHO-m2m3 cells. 4. Costimulation of endogenous P2Y(2) purinoceptors also caused an approx 10-fold left-shift in the MCh-stimulated ERK response in CHO-m2 cells, suggesting that the G(q/11)/G(i/o) interaction to affect ERK activation is not specific to muscarinic receptors. 5. PTx pretreatment of cells had unexpected effects on ERK activation by MCh in both CHO-m2m3 and CHO-m3 cells. Thus, in CHO-m3 cells PTx pretreatment caused a marked left-shift in the MCh concentration-effect curve, while in PTx-treated CHO-m2m3 cells the maximal responsiveness was decreased, but the potency of MCh was only slightly affected. 6. The data presented here strongly suggest that cross-talk between M(2) and M(3) mACh receptors occurs at the level of both second messenger and ERK regulation. Further, these data provide novel insights into the involvement of G(i/o) proteins in both positive and negative modulation of ERK responses evoked by G protein-coupled receptors.


Asunto(s)
Células CHO/metabolismo , Receptor Cross-Talk/fisiología , Receptor Muscarínico M2/fisiología , Receptor Muscarínico M3/fisiología , Receptores de Superficie Celular/fisiología , Sistemas de Mensajero Secundario/fisiología , Animales , Cricetinae , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos , Proteínas Quinasas Activadas por Mitógenos/fisiología , Mutación , Receptor Muscarínico M2/genética , Receptor Muscarínico M3/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...