Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
J Biol Chem ; 294(15): 5747-5758, 2019 04 12.
Artículo en Inglés | MEDLINE | ID: mdl-30745359

RESUMEN

Transmembrane signals initiated by a range of extracellular stimuli converge on members of the Gq family of heterotrimeric G proteins, which relay these signals in target cells. Gq family G proteins comprise Gq, G11, G14, and G16, which upon activation mediate their cellular effects via inositol lipid-dependent and -independent signaling to control fundamental processes in mammalian physiology. To date, highly specific inhibition of Gq/11/14 signaling can be achieved only with FR900359 (FR) and YM-254890 (YM), two naturally occurring cyclic depsipeptides. To further development of FR or YM mimics for other Gα subunits, we here set out to rationally design Gα16 proteins with artificial FR/YM sensitivity by introducing an engineered depsipeptide-binding site. Thereby we permit control of G16 function through ligands that are inactive on the WT protein. Using CRISPR/Cas9-generated Gαq/Gα11-null cells and loss- and gain-of-function mutagenesis along with label-free whole-cell biosensing, we determined the molecular coordinates for FR/YM inhibition of Gq and transplanted these to FR/YM-insensitive G16. Intriguingly, despite having close structural similarity, FR and YM yielded biologically distinct activities: it was more difficult to perturb Gq inhibition by FR and easier to install FR inhibition onto G16 than perturb or install inhibition with YM. A unique hydrophobic network utilized by FR accounted for these unexpected discrepancies. Our results suggest that non-Gq/11/14 proteins should be amenable to inhibition by FR scaffold-based inhibitors, provided that these inhibitors mimic the interaction of FR with Gα proteins harboring engineered FR-binding sites.


Asunto(s)
Depsipéptidos/farmacología , Inhibidores Enzimáticos/farmacología , Subunidades alfa de la Proteína de Unión al GTP , Péptidos Cíclicos/farmacología , Ingeniería de Proteínas , Animales , Sistemas CRISPR-Cas , Subunidades alfa de la Proteína de Unión al GTP/antagonistas & inhibidores , Subunidades alfa de la Proteína de Unión al GTP/genética , Subunidades alfa de la Proteína de Unión al GTP/metabolismo , Células HEK293 , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Ratones
2.
Mol Pharmacol ; 91(5): 518-532, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28254957

RESUMEN

Pairing orphan G protein­coupled receptors (GPCRs) with their cognate endogenous ligands is expected to have a major impact on our understanding of GPCR biology. It follows that the reproducibility of orphan receptor ligand pairs should be of fundamental importance to guide meaningful investigations into the pharmacology and function of individual receptors. GPR17 is an orphan receptor characterized by some as a dualistic uracil nucleotide/cysteinyl leukotriene receptor and by others as inactive toward these stimuli altogether. Whereas regulation of central nervous system myelination by GPR17 is well established, verification of activity of its putative endogenous ligands has proven elusive so far. Herein we report that uracil nucleotides and cysteinyl leukotrienes do not activate human, mouse, or rat GPR17 in various cellular backgrounds, including primary cells, using eight distinct functional assay platforms based on labelfree pathway-unbiased biosensor technologies, as well as canonical second-messenger or biochemical assays. Appraisal of GPR17 activity can neither be accomplished with co-application of both ligand classes, nor with exogenous transfection of partner receptors (nucleotide P2Y12, cysteinyl-leukotriene CysLT1) to reconstitute the elusive pharmacology. Moreover, our study does not support the inhibition of GPR17 by the marketed antiplatelet drugs cangrelor and ticagrelor, previously suggested to antagonize GPR17. Whereas our data do not disagree with a role of GPR17 per se as an orchestrator of central nervous system functions, they challenge the utility of the proposed (ant)agonists as tools to imply direct contribution of GPR17 in complex biologic settings.


Asunto(s)
Cisteína/farmacología , Leucotrienos/farmacología , Receptores Acoplados a Proteínas G/metabolismo , Nucleótidos de Uracilo/farmacología , Adenosina/análogos & derivados , Adenosina/farmacología , Adenosina Monofosfato/análogos & derivados , Adenosina Monofosfato/farmacología , Animales , Células CHO , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Cricetinae , Cricetulus , Células HEK293 , Humanos , Ligandos , Ratones , Proteínas del Tejido Nervioso/metabolismo , Ratas , Transducción de Señal/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas/farmacología , Ticagrelor
3.
J Biol Chem ; 291(2): 705-18, 2016 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-26620557

RESUMEN

Recent studies have recognized G protein-coupled receptors as important regulators of oligodendrocyte development. GPR17, in particular, is an orphan G protein-coupled receptor that has been identified as oligodendroglial maturation inhibitor because its stimulation arrests primary mouse oligodendrocytes at a less differentiated stage. However, the intracellular signaling effectors transducing its activation remain poorly understood. Here, we use Oli-neu cells, an immortalized cell line derived from primary murine oligodendrocytes, and primary rat oligodendrocyte cultures as model systems to identify molecular targets that link cell surface GPR17 to oligodendrocyte maturation blockade. We demonstrate that stimulation of GPR17 by the small molecule agonist MDL29,951 (2-carboxy-4,6-dichloro-1H-indole-3-propionic acid) decreases myelin basic protein expression levels mainly by triggering the Gαi/o signaling pathway, which in turn leads to reduced activity of the downstream cascade adenylyl cyclase-cAMP-PKA-cAMP response element-binding protein (CREB). In addition, we show that GPR17 activation also diminishes myelin basic protein abundance by lessening stimulation of the exchange protein directly activated by cAMP (EPAC), thus uncovering a previously unrecognized role for EPAC to regulate oligodendrocyte differentiation. Together, our data establish PKA and EPAC as key downstream effectors of GPR17 that inhibit oligodendrocyte maturation. We envisage that treatments augmenting PKA and/or EPAC activity represent a beneficial approach for therapeutic enhancement of remyelination in those demyelinating diseases where GPR17 is highly expressed, such as multiple sclerosis.


Asunto(s)
Diferenciación Celular , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Oligodendroglía/citología , Receptores Acoplados a Proteínas G/metabolismo , Animales , Diferenciación Celular/efectos de los fármacos , AMP Cíclico/análogos & derivados , AMP Cíclico/farmacología , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gq-G11 , Factores de Intercambio de Guanina Nucleótido/metabolismo , Indoles/farmacología , Ratones , Modelos Biológicos , Proteína Básica de Mielina/metabolismo , Proteínas del Tejido Nervioso/agonistas , Fosforilación/efectos de los fármacos , Propionatos/farmacología , Ratas , Ratas Wistar , Receptores Acoplados a Proteínas G/agonistas , Transducción de Señal , Tionucleótidos/farmacología
4.
Trends Pharmacol Sci ; 44(3): 131-133, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36604224

RESUMEN

Endosomes, long considered as sorting stations for downregulation and recycling of cell surface G protein-coupled receptors (GPCRs), are now well-established sites of signal transduction. Recent work from the groups of Jin Zhang and Roger Sunahara features endosomes as signaling hubs and physical platforms for noncanonical activation of ERK by GPCRs.


Asunto(s)
Endosomas , Sistema de Señalización de MAP Quinasas , Receptores Acoplados a Proteínas G , Humanos , Membrana Celular/metabolismo , Endosomas/metabolismo , Transporte de Proteínas/fisiología , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal/fisiología
5.
J Biol Chem ; 286(14): 11890-4, 2011 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-21339298

RESUMEN

Among dietary components, conjugated linoleic acids (CLAs) have attracted considerable attention as weight loss supplements in the Western world because they reduce fat stores and increase muscle mass. However, a number of adverse effects are also ascribed to the intake of CLAs such as aggravation of insulin resistance and the risk of developing diabetes. However, the mechanisms accounting for the effects of CLAs on glucose homeostasis are incompletely understood. Herein we provide evidence that CLAs specifically activate the cell surface receptor FFA1, an emerging therapeutic target to treat type 2 diabetes. Using different recombinant cellular systems engineered to stably express FFA1 and a set of diverse functional assays including the novel, label-free non-invasive dynamic mass redistribution technology (Corning® Epic® biosensor), both CLA isomers cis-9, trans-11-CLA and trans-10, cis-12-CLA were found to activate FFA1 in vitro at concentrations sufficient to also account for FFA1 activation in vivo. Each CLA isomer markedly increased glucose-stimulated insulin secretion in insulin-producing INS-1E cells that endogenously express FFA1 and in primary pancreatic ß-cells of wild type but not FFA1-/- knock-out mice. Our findings establish a clear mechanistic link between CLAs and insulin production and identify the cell surface receptor FFA1 as a molecular target for CLAs, explaining their acute stimulatory effects on insulin secretion in vivo. CLAs are also revealed as insulinotropic components in widely used nutraceuticals, a finding with significant implication for development of FFA1 modulators to treat type 2 diabetes.


Asunto(s)
Insulina/metabolismo , Ácidos Linoleicos Conjugados/farmacología , Receptores Acoplados a Proteínas G/agonistas , Animales , Calcio/metabolismo , Línea Celular , Línea Celular Tumoral , Humanos , Ratas , Receptores Acoplados a Proteínas G/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
6.
Cell Chem Biol ; 29(10): 1541-1555.e7, 2022 10 20.
Artículo en Inglés | MEDLINE | ID: mdl-36126653

RESUMEN

Therapies that promote neuroprotection and axonal survival by enhancing myelin regeneration are an unmet need to prevent disability progression in multiple sclerosis. Numerous potentially beneficial compounds have originated from phenotypic screenings but failed in clinical trials. It is apparent that current cell- and animal-based disease models are poor predictors of positive treatment options, arguing for novel experimental approaches. Here we explore the experimental power of humanized zebrafish to foster the identification of pro-remyelination compounds via specific inhibition of GPR17. Using biochemical and imaging techniques, we visualize the expression of zebrafish (zf)-gpr17 during the distinct stages of oligodendrocyte development, thereby demonstrating species-conserved expression between zebrafish and mammals. We also demonstrate species-conserved function of zf-Gpr17 using genetic loss-of-function and rescue techniques. Finally, using GPR17-humanized zebrafish, we provide proof of principle for in vivo analysis of compounds acting via targeted inhibition of human GPR17. We anticipate that GPR17-humanized zebrafish will markedly improve the search for effective pro-myelinating pharmacotherapies.


Asunto(s)
Oligodendroglía , Profármacos , Animales , Humanos , Pez Cebra/metabolismo , Profármacos/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Diferenciación Celular , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Modelos Animales de Enfermedad , Mamíferos/metabolismo
7.
Nat Commun ; 13(1): 7109, 2022 11 19.
Artículo en Inglés | MEDLINE | ID: mdl-36402762

RESUMEN

Carvedilol is among the most effective ß-blockers for improving survival after myocardial infarction. Yet the mechanisms by which carvedilol achieves this superior clinical profile are still unclear. Beyond blockade of ß1-adrenoceptors, arrestin-biased signalling via ß2-adrenoceptors is a molecular mechanism proposed to explain the survival benefits. Here, we offer an alternative mechanism to rationalize carvedilol's cellular signalling. Using primary and immortalized cells genome-edited by CRISPR/Cas9 to lack either G proteins or arrestins; and combining biological, biochemical, and signalling assays with molecular dynamics simulations, we demonstrate that G proteins drive all detectable carvedilol signalling through ß2ARs. Because a clear understanding of how drugs act is imperative to data interpretation in basic and clinical research, to the stratification of clinical trials or to the monitoring of drug effects on the target pathway, the mechanistic insight gained here provides a foundation for the rational development of signalling prototypes that target the ß-adrenoceptor system.


Asunto(s)
Antagonistas Adrenérgicos beta , Infarto del Miocardio , Humanos , Carvedilol/farmacología , Antagonistas Adrenérgicos beta/farmacología , Receptores Adrenérgicos beta 2/genética , Infarto del Miocardio/tratamiento farmacológico
8.
Mol Cell Neurosci ; 44(4): 342-52, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20447457

RESUMEN

Mice deficient for the neuronal glycine transporter subtype 2 (GlyT2) die during the second postnatal week after developing neuromotor deficiencies, which resembles severe forms of human hyperekplexia. This phenotype has been attributed to a dramatic reduction in glycinergic neurotransmission. In the present study we analyzed the development of GABAergic and glycinergic synaptic transmission in GlyT2-knockout mice during early postnatal life. Anti-glycine immunohistochemistry in spinal cord and brainstem slices and whole-cell voltage-clamp recordings of glycinergic inhibitory postsynaptic currents (IPSCs) from hypoglossal motoneurons revealed strikingly reduced levels of synaptic glycine already at birth. Since GABA and glycine use the same vesicular inhibitory amino acid transporter (VIAAT or VGAT) we also analysed GABAergic neurotransmission. No increase of GABA immunoreactivity was observed in the spinal cord and brainstem of GlyT2(-/-) mice at any stage of postnatal development. Correspondingly no up-regulation of GABAergic IPSCs was detected in GlyT2(-/-) hypoglossal motoneurons. These data suggest that in the first postnatal week, loss of the glycine transporter 2 is neither compensated by glycine de-novo synthesis nor by up-regulation of the GABAergic transmission in GlyT2(-/-) mice.


Asunto(s)
Proteínas de Transporte de Glicina en la Membrana Plasmática/fisiología , Glicina/metabolismo , Potenciales Postsinápticos Inhibidores/fisiología , Animales , Animales Recién Nacidos , Tronco Encefálico/metabolismo , Tronco Encefálico/fisiopatología , Nervio Hipogloso/metabolismo , Nervio Hipogloso/fisiopatología , Inmunohistoquímica , Ratones , Ratones Noqueados , Neuronas Motoras/metabolismo , Técnicas de Placa-Clamp/métodos , Médula Espinal/metabolismo , Médula Espinal/fisiopatología , Vesículas Sinápticas/metabolismo , Ácido gamma-Aminobutírico/fisiología
9.
Glia ; 58(9): 1066-73, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20468048

RESUMEN

The glycine transporter 1 (GlyT1) is expressed in astrocytes and selected neurons of the mammalian CNS. In newborn mice, GlyT1 is crucial for efficient termination of glycine-mediated inhibitory neurotransmission. Furthermore, GlyT1 has been implicated in the regulation of excitatory N-methyl-D-asparate (NMDA) receptors. To evaluate whether glial and neuronal GlyT1 have distinct roles at inhibitory synapses, we inactivated the GlyT1 gene cell type-specifically using mice carrying floxed GlyT1 alleles GlyT1((+)/+)). GlyT1((+)/(+)) mice expressing Cre recombinase in glial cells developed severe neuromotor deficits during the first postnatal week, which mimicked the phenotype of conventional GlyT1 knock-out mice and are consistent with glycinergic over-inhibition. In contrast, Cre-mediated inactivation of the GlyT1 gene in neuronal cells did not result in detectable motor impairment. Notably, some animals deficient for glial GlyT1 survived the first postnatal week and did not develop neuromotor deficits throughout adulthood, although GlyT1 expression was efficiently reduced. Thus, glial GlyT1 is critical for the regulation of glycine levels at inhibitory synapses only during early postnatal life.


Asunto(s)
Encéfalo/fisiología , Proteínas de Transporte de Glicina en la Membrana Plasmática/metabolismo , Neuroglía/fisiología , Neuronas/fisiología , Médula Espinal/fisiología , Envejecimiento , Animales , Animales Recién Nacidos , Encéfalo/crecimiento & desarrollo , Tronco Encefálico/crecimiento & desarrollo , Tronco Encefálico/fisiología , Supervivencia Celular/fisiología , Discinesias/metabolismo , Proteínas de Transporte de Glicina en la Membrana Plasmática/genética , Ratones , Ratones Transgénicos , Inhibición Neural/fisiología , Fenotipo , Médula Espinal/crecimiento & desarrollo , Sinapsis/fisiología
10.
Trends Biochem Sci ; 30(6): 325-33, 2005 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15950877

RESUMEN

Glycine has important neurotransmitter functions at inhibitory and excitatory synapses in the vertebrate central nervous system. The effective synaptic concentrations of glycine are regulated by glycine transporters (GlyTs), which mediate its reuptake into nerve terminals and adjacent glial cells. GlyTs are members of the Na(+)/Cl(-)-dependent transporter family, whose activities and subcellular distributions are regulated by phosphorylation and interactions with other proteins. The analysis of GlyT knockout mice has revealed distinct functions of individual GlyT subtypes in synaptic transmission and provided animal models for two hereditary human diseases, glycine encephalopathy and hyperekplexia. Selective GlyT inhibitors could be of therapeutic value in cognitive disorders, schizophrenia and pain.


Asunto(s)
Sistemas de Transporte de Aminoácidos Neutros/metabolismo , Neurotransmisores/metabolismo , Transmisión Sináptica/fisiología , Animales , Glicina/farmacocinética , Proteínas de Transporte de Glicina en la Membrana Plasmática , Humanos , Coma Hiperglucémico Hiperosmolar no Cetósico/etiología , Ratones , Ratones Noqueados , Trastornos del Movimiento/etiología , Reflejo de Sobresalto/fisiología
11.
Neuron ; 40(4): 785-96, 2003 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-14622582

RESUMEN

The glycine transporter subtype 1 (GlyT1) is widely expressed in astroglial cells throughout the mammalian central nervous system and has been implicated in the regulation of N-methyl-D-aspartate (NMDA) receptor activity. Newborn mice deficient in GlyT1 are anatomically normal but show severe motor and respiratory deficits and die during the first postnatal day. In brainstem slices from GlyT1-deficient mice, in vitro respiratory activity is strikingly reduced but normalized by the glycine receptor (GlyR) antagonist strychnine. Conversely, glycine or the GlyT1 inhibitor sarcosine suppress respiratory activity in slices from wild-type mice. Thus, during early postnatal life, GlyT1 is essential for regulating glycine concentrations at inhibitory GlyRs, and GlyT1 deletion generates symptoms found in human glycine encephalopathy.


Asunto(s)
Sistemas de Transporte de Aminoácidos Neutros/deficiencia , Encéfalo/metabolismo , Glicina/metabolismo , Inhibición Neural/genética , Neuroglía/metabolismo , Sistemas de Transporte de Aminoácidos Neutros/genética , Animales , Animales Recién Nacidos , Encéfalo/citología , Encéfalo/crecimiento & desarrollo , Modelos Animales de Enfermedad , Genes Letales/genética , Glicina/farmacocinética , Proteínas de Transporte de Glicina en la Membrana Plasmática , Homocigoto , Ratones , Ratones Noqueados , Proteínas del Tejido Nervioso/metabolismo , Fenotipo , Respiración/efectos de los fármacos , Sarcosina/farmacología , Estricnina/farmacología , Transmisión Sináptica/genética
12.
Neuron ; 40(4): 797-806, 2003 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-14622583

RESUMEN

The glycine transporter subtype 2 (GlyT2) is localized in the axon terminals of glycinergic neurons. Mice deficient in GlyT2 are normal at birth but during the second postnatal week develop a lethal neuromotor deficiency that resembles severe forms of human hyperekplexia (hereditary startle disease) and is characterized by spasticity, tremor, and an inability to right. Histological and immunological analyses failed to reveal anatomical or biochemical abnormalities, but the amplitudes of glycinergic miniature inhibitory currents (mIPSCs) were strikingly reduced in hypoglossal motoneurons and dissociated spinal neurons from GlyT2-deficient mice. Thus, postnatal GlyT2 function is crucial for efficient transmitter loading of synaptic vesicles in glycinergic nerve terminals, and the GlyT2 gene constitutes a candidate disease gene in human hyperekplexia patients.


Asunto(s)
Sistemas de Transporte de Aminoácidos Neutros/deficiencia , Tronco Encefálico/metabolismo , Genes Letales/genética , Trastornos Heredodegenerativos del Sistema Nervioso/genética , Reflejo de Sobresalto/genética , Sistemas de Transporte de Aminoácidos Neutros/genética , Animales , Animales Recién Nacidos , Tronco Encefálico/crecimiento & desarrollo , Tronco Encefálico/fisiopatología , Modelos Animales de Enfermedad , Feto , Eliminación de Gen , Glicina/metabolismo , Proteínas de Transporte de Glicina en la Membrana Plasmática , Trastornos Heredodegenerativos del Sistema Nervioso/metabolismo , Trastornos Heredodegenerativos del Sistema Nervioso/fisiopatología , Nervio Hipogloso/metabolismo , Nervio Hipogloso/fisiopatología , Ratones , Ratones Noqueados , Neuronas Motoras/metabolismo , Inhibición Neural/genética , Técnicas de Cultivo de Órganos , Fenotipo , Terminales Presinápticos/metabolismo , Transmisión Sináptica/genética , Vesículas Sinápticas/metabolismo
13.
Front Mol Neurosci ; 11: 279, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30177871

RESUMEN

The endogenous neutral amino acid L-proline exhibits a variety of physiological and behavioral actions in the nervous system, highlighting the importance of accurately regulating its extracellular abundance. The L-proline transporter PROT (Slc6A7) is believed to control the spatial and temporal distribution of L-proline at glutamatergic synapses by rapid uptake of this amino acid into presynaptic terminals. Despite the importance of members of the Slc6 transporter family regulating neurotransmitter signaling and homeostasis in brain, evidence that PROT dysfunction supports risk for mental illness is lacking. Here we report the disruption of the PROT gene by homologous recombination. Mice defective in PROT displayed altered expression of glutamate transmission-related synaptic proteins in cortex and thalamus. PROT deficiency perturbed mouse behavior, such as reduced locomotor activity, decreased approach motivation and impaired memory extinction. Thus, our study demonstrates that PROT regulates behaviors that are needed to respond to environmental changes in vivo and suggests that PROT dysfunctions might contribute to mental disorders showing altered response choice following task contingency changes.

15.
Cell Chem Biol ; 25(6): 775-786.e5, 2018 06 21.
Artículo en Inglés | MEDLINE | ID: mdl-29706593

RESUMEN

Identification of additional uses for existing drugs is a hot topic in drug discovery and a viable alternative to de novo drug development. HAMI3379 is known as an antagonist of the cysteinyl-leukotriene CysLT2 receptor, and was initially developed to treat cardiovascular and inflammatory disorders. In our study we identified HAMI3379 as an antagonist of the orphan G protein-coupled receptor GPR17. HAMI3379 inhibits signaling of recombinant human, rat, and mouse GPR17 across various cellular backgrounds, and of endogenous GPR17 in primary rodent oligodendrocytes. GPR17 blockade by HAMI3379 enhanced maturation of primary rat and mouse oligodendrocytes, but was without effect in oligodendrocytes from GPR17 knockout mice. In human oligodendrocytes prepared from inducible pluripotent stem cells, GPR17 is expressed and its activation impaired oligodendrocyte differentiation. HAMI3379, conversely, efficiently favored human oligodendrocyte differentiation. We propose that HAMI3379 holds promise for pharmacological exploitation of orphan GPR17 to enhance regenerative strategies for the promotion of remyelination in patients.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Ácidos Ciclohexanocarboxílicos/farmacología , Reposicionamiento de Medicamentos , Oligodendroglía/citología , Oligodendroglía/efectos de los fármacos , Ácidos Ftálicos/farmacología , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Animales , Ácidos Ciclohexanocarboxílicos/química , Relación Dosis-Respuesta a Droga , Humanos , Indoles/química , Indoles/farmacología , Ratones , Ratones Noqueados , Estructura Molecular , Ácidos Ftálicos/química , Propionatos/química , Propionatos/farmacología , Ratas , Receptores Acoplados a Proteínas G/deficiencia , Receptores Acoplados a Proteínas G/metabolismo , Relación Estructura-Actividad
16.
Nat Commun ; 9(1): 341, 2018 01 23.
Artículo en Inglés | MEDLINE | ID: mdl-29362459

RESUMEN

G protein-independent, arrestin-dependent signaling is a paradigm that broadens the signaling scope of G protein-coupled receptors (GPCRs) beyond G proteins for numerous biological processes. However, arrestin signaling in the collective absence of functional G proteins has never been demonstrated. Here we achieve a state of "zero functional G" at the cellular level using HEK293 cells depleted by CRISPR/Cas9 technology of the Gs/q/12 families of Gα proteins, along with pertussis toxin-mediated inactivation of Gi/o. Together with HEK293 cells lacking ß-arrestins ("zero arrestin"), we systematically dissect G protein- from arrestin-driven signaling outcomes for a broad set of GPCRs. We use biochemical, biophysical, label-free whole-cell biosensing and ERK phosphorylation to identify four salient features for all receptors at "zero functional G": arrestin recruitment and internalization, but-unexpectedly-complete failure to activate ERK and whole-cell responses. These findings change our understanding of how GPCRs function and in particular of how they activate ERK1/2.


Asunto(s)
Proteínas de Unión al GTP/genética , Sistema de Señalización de MAP Quinasas , Receptores Acoplados a Proteínas G/metabolismo , beta-Arrestina 1/metabolismo , Arrestina beta 2/metabolismo , Sistemas CRISPR-Cas , Subunidades alfa de la Proteína de Unión al GTP/genética , Subunidades alfa de la Proteína de Unión al GTP G12-G13/genética , Subunidades alfa de la Proteína de Unión al GTP Gs/genética , Proteínas de Unión al GTP/metabolismo , Técnicas de Inactivación de Genes , Células HEK293 , Humanos , Fosforilación , Transducción de Señal , beta-Arrestinas/metabolismo
17.
J Neurosci ; 22(15): 6347-52, 2002 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-12151512

RESUMEN

A proper balance between striatal muscarinic cholinergic and dopaminergic neurotransmission is required for coordinated locomotor control. Activation of striatal muscarinic acetylcholine receptors (mAChRs) is known to modulate striatal dopamine release. To identify the mAChR subtype(s) involved in this activity, we used genetically altered mice that lacked functional M1-M5 mAChRs [knock-out (KO) mice]. In superfused striatal slices from wild-type mice, the non-subtype-selective muscarinic agonist oxotremorine led to concentration-dependent increases in potassium-stimulated [3H]dopamine release (by up to 60%). The lack of M1 or M2 receptors had no significant effect on the magnitude of these responses. Strikingly, oxotremorine-mediated potentiation of stimulated striatal [3H]dopamine release was abolished in M4 receptor KO mice, significantly increased in M3 receptor-deficient mice, and significantly reduced (but not abolished) in M5 receptor KO mice. Additional release studies performed in the presence of tetrodotoxin suggested that the dopamine release-stimulating M4 receptors are probably located on neuronal cell bodies, but that the release-facilitating M5 and the release-inhibiting M3 receptors are likely to be located on nerve terminals. Studies with the GABA(A) receptor blocker bicuculline methochloride suggested that M3 and M4 receptors mediate their dopamine release-modulatory effects via facilitation or inhibition, respectively, of striatal GABA release. These results provide unambiguous evidence that multiple mAChR subtypes are involved in the regulation of striatal dopamine release. These findings should contribute to a better understanding of the important functional roles that the muscarinic cholinergic system plays in striatal function.


Asunto(s)
Cuerpo Estriado/metabolismo , Dopamina/metabolismo , Receptores Muscarínicos/metabolismo , Animales , Cuerpo Estriado/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Antagonistas del GABA/farmacología , Antagonistas de Receptores de GABA-A , Técnicas In Vitro , Ratones , Ratones Noqueados , Agonistas Muscarínicos/farmacología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Oxotremorina/farmacología , Potasio/farmacología , Receptor Muscarínico M1 , Receptor Muscarínico M2 , Receptor Muscarínico M3 , Receptor Muscarínico M4 , Receptor Muscarínico M5 , Receptores Muscarínicos/deficiencia , Receptores Muscarínicos/genética , Tetrodotoxina/farmacología
18.
J Neurosci ; 22(5): 1709-17, 2002 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-11880500

RESUMEN

Forebrain muscarinic acetylcholine (ACh) receptors (mAChRs; M1-M5) are predicted to play important roles in many fundamental central functions, including higher cognitive processes and modulation of extrapyramidal motor activity. Synaptic ACh levels are known to be regulated by the activity of presynaptic muscarinic autoreceptors mediating inhibition of ACh release. Primarily because of the use of ligands with limited receptor subtype selectivity, classical pharmacological studies have led to conflicting results regarding the identity of the mAChR subtypes mediating this activity in different areas of the brain. To investigate the molecular identity of hippocampal, cortical, and striatal inhibitory muscarinic autoreceptors in a more direct manner, we used genetically altered mice lacking functional M2 and/or M4 mAChRs [knock-out (KO) mice]. After labeling of cellular ACh pools with [3H]choline, potassium-stimulated [3H]ACh release was measured in superfused brain slices, either in the absence or the presence of muscarinic drugs. The nonsubtype-selective muscarinic agonist, oxotremorine (0.1-10 microm), inhibited potassium-stimulated [3H]ACh release in hippocampal, cortical, and striatal slices prepared from wild-type mice by up to 80%. This activity was totally abolished in tissues prepared from M2-M4 receptor double KO mice. Strikingly, release studies with brain slices from M2 and M4 receptor single KO mice indicated that autoinhibition of ACh release is mediated primarily by the M2 receptor in hippocampus and cerebral cortex, but predominantly by the M4 receptor in the striatum. These results, together with additional receptor localization studies, support the novel concept that autoinhibition of ACh release involves different mAChRs in different regions of the brain.


Asunto(s)
Autorreceptores/metabolismo , Encéfalo/metabolismo , Proteínas de Transporte de Membrana , Inhibición Neural/fisiología , Receptores Muscarínicos/deficiencia , Receptores Muscarínicos/metabolismo , Proteínas de Transporte Vesicular , Acetilcolina/metabolismo , Animales , Encéfalo/efectos de los fármacos , Proteínas Portadoras/metabolismo , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/metabolismo , Cuerpo Estriado/efectos de los fármacos , Cuerpo Estriado/metabolismo , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Técnicas In Vitro , Ratones , Ratones Noqueados , Agonistas Muscarínicos/farmacología , Antagonistas Muscarínicos , Potasio/farmacología , Receptor Muscarínico M2 , Receptor Muscarínico M4 , Receptores Muscarínicos/genética , Proteínas de Transporte Vesicular de Acetilcolina
19.
J Neurosci ; 22(12): RC229, 2002 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-12045234

RESUMEN

We recently described a novel endogenous mechanism of peripheral antinociception, possibly involving activation of muscarinic M2 acetylcholine receptors that are expressed on nociceptive nerve endings and decrease their sensitivity. In the present study, this mechanism was scrutinized in skin taken from mice with targeted deletions of the muscarinic M2 receptor gene and, for control purposes, of the M4 receptor gene. Two different approaches were taken. Electrophysiologically the effects of muscarine on nociceptive afferents were investigated using the mouse skin-saphenous nerve preparation, in vitro. Muscarine did not excite nociceptors in the wild-type littermates (WT) and M4 knock-out (M4 KO) mice, but almost all fibers exhibited marked desensitization to mechanical and heat stimuli. Surprisingly, in the M2 KO mice, muscarine was able to excite C-nociceptors and to induce a mild sensitization to heat but caused no alteration in mechanical responsiveness tested with von Frey hairs. In the second, neurochemical approach, the heat-induced cutaneous release of calcitonin gene-related peptide (CGRP) was investigated to gain comparative data on the neurosecretory (vasodilatory) functions of the primary afferent neurons. The substantial increase of CGRP release evoked by noxious heat (47 degrees C) was diminished under muscarine by >50% in the WT and M4 KO animals but remained unaltered in the M2 KO mice. Together, these data provide direct evidence that M2 receptors on cutaneous nerve endings mediate effective depression of nociceptive responsiveness. This observation should be of interest for the development of novel classes of analgesic agents.


Asunto(s)
Nociceptores/metabolismo , Nervios Periféricos/fisiología , Receptores Muscarínicos/metabolismo , Analgésicos/farmacología , Animales , Péptido Relacionado con Gen de Calcitonina/metabolismo , Técnicas de Cultivo , Sistemas de Liberación de Medicamentos , Conductividad Eléctrica , Calor , Cinética , Ratones , Ratones Noqueados , Muscarina/farmacología , Agonistas Muscarínicos/farmacología , Antagonistas Muscarínicos/farmacología , Fibras Nerviosas/efectos de los fármacos , Fibras Nerviosas/fisiología , Neuronas Aferentes/efectos de los fármacos , Neuronas Aferentes/fisiología , Nervios Periféricos/química , Nervios Periféricos/efectos de los fármacos , Receptor Muscarínico M2 , Receptor Muscarínico M4 , Receptores Muscarínicos/análisis , Receptores Muscarínicos/genética , Piel/inervación
20.
J Neurosci ; 24(45): 10117-27, 2004 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-15537882

RESUMEN

Muscarinic acetylcholine receptors are known to play key roles in facilitating cognitive processes. However, the specific roles of the individual muscarinic receptor subtypes (M1-M5) in learning and memory are not well understood at present. In the present study, we used wild-type (M2+/+) and M2 receptor-deficient (M2-/-) mice to examine the potential role of M2 receptors in learning and memory and hippocampal synaptic plasticity. M2-/- mice showed significant deficits in behavioral flexibility and working memory in the Barnes circular maze and the T-maze delayed alternation tests, respectively. The behavioral deficits of M2-/- mice were associated with profound changes in neuronal plasticity studied at the Schaffer-CA1 synapse of hippocampal slices. Strikingly, short-term potentiation (STP) was abolished, and long-term potentiation (LTP) was drastically reduced after high-frequency stimulation of M2-/- hippocampi. Treatment of M2-/- hippocampal slices with the GABA(A) receptor antagonist, bicuculline, restored STP and significantly increased LTP. Whole-cell recordings from CA1 pyramidal cells demonstrated a much stronger disinhibition of GABAergic than glutamatergic transmission in M2-/- hippocampi, which was particularly prominent during stimulus trains. Increased strength of GABAergic inhibition is thus a likely mechanism underlying the impaired synaptic plasticity observed with M2-/- hippocampi. Moreover, the persistent enhancement of excitatory synaptic transmission in CA1 pyramidal cells induced by the transient application of a low concentration of a muscarinic agonist (referred to as LTP(m)) was totally abolished in M2-/- mice. Because impaired muscarinic cholinergic neurotransmission is associated with Alzheimer's disease and normal aging processes, these findings should be of considerable therapeutic relevance.


Asunto(s)
Amnesia/fisiopatología , Conducta Animal/fisiología , Hipocampo/fisiopatología , Memoria/fisiología , Plasticidad Neuronal/fisiología , Receptor Muscarínico M2/deficiencia , Amnesia/genética , Animales , Conducta Animal/efectos de los fármacos , Bicuculina/farmacología , Carbacol/farmacología , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Potenciales Postsinápticos Excitadores/fisiología , Trietyoduro de Galamina/farmacología , Hipocampo/efectos de los fármacos , Potenciación a Largo Plazo/genética , Potenciación a Largo Plazo/fisiología , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Aprendizaje por Laberinto/fisiología , Memoria/efectos de los fármacos , Ratones , Ratones Noqueados , Agonistas Muscarínicos/farmacología , Plasticidad Neuronal/genética , Técnicas de Placa-Clamp , Células Piramidales/efectos de los fármacos , Células Piramidales/fisiología , Receptor Muscarínico M2/genética , Receptor Muscarínico M2/fisiología , Receptores de GABA-A/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA