Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
1.
J Biol Chem ; 293(25): 9759-9769, 2018 06 22.
Artículo en Inglés | MEDLINE | ID: mdl-29752411

RESUMEN

The four R-spondins (RSPO1-4) strongly potentiate Wnt signaling and play critical roles in normal development, adult stem cell survival, and cancer development and aggressiveness. All four RSPOs have been suggested to potentiate Wnt signaling by binding to three related receptors, i.e. leucine-rich repeat-containing, G protein-coupled receptors 4, 5, and 6 (LGR4/5/6), and then inducing the clearance of two E3 ubiquitin ligases (RNF43 and ZNRF3) that otherwise would ubiquitinate Wnt receptors for degradation. Here, we show that RSPO1-4 have differential dependence on LGRs in potentiating Wnt/ß-catenin signaling and that RSPO2 can enhance this pathway without any LGR. LGR4 knockout (LGR4KO) in HEK293 cells completely abrogated the Wnt/ß-catenin signaling response to RSPO1 and RSPO4 and strongly impaired the response to RSPO3. RSPO2, however, retained robust activity albeit with decreased potency. Complete rescue of RSPO1-4 activity in LGR4KO cells required the seven-transmembrane domain of LGR4. Furthermore, an RSPO2 mutant with normal binding affinity to ZNRF3 but no or little binding to LGR4 or LGR5 still potentiated Wnt/ß-catenin signaling in vitro, supported the growth of intestinal organoids ex vivo, and stimulated intestinal crypt growth in vivo Mechanistically, RSPO2 could increase Wnt receptor levels in the absence of any LGR without affecting ZNRF3 endocytosis and stability. These findings suggest that RSPO1-4 use distinct mechanisms in regulating Wnt and other signaling pathways, which have important implications for understanding the pleiotropic functions of RSPOs and LGRs in both normal and cancer development.


Asunto(s)
Transducción de Señal , Trombospondinas/metabolismo , Proteínas Wnt/metabolismo , beta Catenina/metabolismo , Animales , Células HEK293 , Humanos , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Mucosa Intestinal/citología , Mucosa Intestinal/metabolismo , Ratones , Ratones Endogámicos C57BL , Organoides/citología , Organoides/metabolismo , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Trombospondinas/genética , Proteínas Wnt/genética , beta Catenina/genética
2.
J Biol Chem ; 292(27): 11508-11530, 2017 07 07.
Artículo en Inglés | MEDLINE | ID: mdl-28522608

RESUMEN

The type 1 taste receptor member 3 (T1R3) is a G protein-coupled receptor involved in sweet-taste perception. Besides the tongue, the T1R3 receptor is highly expressed in brain areas implicated in cognition, including the hippocampus and cortex. As cognitive decline is often preceded by significant metabolic or endocrinological dysfunctions regulated by the sweet-taste perception system, we hypothesized that a disruption of the sweet-taste perception in the brain could have a key role in the development of cognitive dysfunction. To assess the importance of the sweet-taste receptors in the brain, we conducted transcriptomic and proteomic analyses of cortical and hippocampal tissues isolated from T1R3 knock-out (T1R3KO) mice. The effect of an impaired sweet-taste perception system on cognition functions were examined by analyzing synaptic integrity and performing animal behavior on T1R3KO mice. Although T1R3KO mice did not present a metabolically disrupted phenotype, bioinformatic interpretation of the high-dimensionality data indicated a strong neurodegenerative signature associated with significant alterations in pathways involved in neuritogenesis, dendritic growth, and synaptogenesis. Furthermore, a significantly reduced dendritic spine density was observed in T1R3KO mice together with alterations in learning and memory functions as well as sociability deficits. Taken together our data suggest that the sweet-taste receptor system plays an important neurotrophic role in the extralingual central nervous tissue that underpins synaptic function, memory acquisition, and social behavior.


Asunto(s)
Conducta Animal , Espinas Dendríticas/metabolismo , Aprendizaje , Memoria , Neuritas/metabolismo , Receptores Acoplados a Proteínas G/deficiencia , Conducta Social , Animales , Espinas Dendríticas/patología , Ratones , Ratones Noqueados , Neuritas/patología
3.
J Biol Chem ; 292(24): 9865-9881, 2017 06 16.
Artículo en Inglés | MEDLINE | ID: mdl-28356352

RESUMEN

The glucagon receptor (GCGR) belongs to the secretin-like (class B) family of G protein-coupled receptors (GPCRs) and is activated by the peptide hormone glucagon. The structures of an activated class B GPCR have remained unsolved, preventing a mechanistic understanding of how these receptors are activated. Using a combination of structural modeling and mutagenesis studies, we present here two modes of ligand-independent activation of GCGR. First, we identified a GCGR-specific hydrophobic lock comprising Met-338 and Phe-345 within the IC3 loop and transmembrane helix 6 (TM6) and found that this lock stabilizes the TM6 helix in the inactive conformation. Disruption of this hydrophobic lock led to constitutive G protein and arrestin signaling. Second, we discovered a polar core comprising conserved residues in TM2, TM3, TM6, and TM7, and mutations that disrupt this polar core led to constitutive GCGR activity. On the basis of these results, we propose a mechanistic model of GCGR activation in which TM6 is held in an inactive conformation by the conserved polar core and the hydrophobic lock. Mutations that disrupt these inhibitory elements allow TM6 to swing outward to adopt an active TM6 conformation similar to that of the canonical ß2-adrenergic receptor complexed with G protein and to that of rhodopsin complexed with arrestin. Importantly, mutations in the corresponding polar core of several other members of class B GPCRs, including PTH1R, PAC1R, VIP1R, and CRFR1, also induce constitutive G protein signaling, suggesting that the rearrangement of the polar core is a conserved mechanism for class B GPCR activation.


Asunto(s)
Modelos Moleculares , Receptor de Hormona Paratiroídea Tipo 1/agonistas , Receptores de Hormona Liberadora de Corticotropina/agonistas , Receptores de Glucagón/agonistas , Receptores del Polipéptido Activador de la Adenilato-Ciclasa Hipofisaria/agonistas , Receptores de Tipo I del Polipéptido Intestinal Vasoactivo/agonistas , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Sitios de Unión , Línea Celular , Secuencia Conservada , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Ligandos , Mutagénesis Sitio-Dirigida , Mutación , Fragmentos de Péptidos/agonistas , Fragmentos de Péptidos/química , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Conformación Proteica , Dominios y Motivos de Interacción de Proteínas , Estabilidad Proteica , Receptor de Hormona Paratiroídea Tipo 1/química , Receptor de Hormona Paratiroídea Tipo 1/genética , Receptor de Hormona Paratiroídea Tipo 1/metabolismo , Receptores de Hormona Liberadora de Corticotropina/química , Receptores de Hormona Liberadora de Corticotropina/genética , Receptores de Hormona Liberadora de Corticotropina/metabolismo , Receptores de Glucagón/química , Receptores de Glucagón/genética , Receptores de Glucagón/metabolismo , Receptores del Polipéptido Activador de la Adenilato-Ciclasa Hipofisaria/química , Receptores del Polipéptido Activador de la Adenilato-Ciclasa Hipofisaria/genética , Receptores del Polipéptido Activador de la Adenilato-Ciclasa Hipofisaria/metabolismo , Receptores de Tipo I del Polipéptido Intestinal Vasoactivo/química , Receptores de Tipo I del Polipéptido Intestinal Vasoactivo/genética , Receptores de Tipo I del Polipéptido Intestinal Vasoactivo/metabolismo , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Sistemas de Mensajero Secundario , Homología Estructural de Proteína
4.
J Biol Chem ; 292(36): 14989-15001, 2017 09 08.
Artículo en Inglés | MEDLINE | ID: mdl-28739799

RESUMEN

Leucine-rich repeat-containing G protein-coupled receptor 5 (LGR5) is a bona fide marker of adult stem cells in several epithelial tissues, most notably in the intestinal crypts, and is highly up-regulated in many colorectal, hepatocellular, and ovarian cancers. LGR5 activation by R-spondin (RSPO) ligands potentiates Wnt/ß-catenin signaling in vitro; however, deletion of LGR5 in stem cells has little or no effect on Wnt/ß-catenin signaling or cell proliferation in vivo Remarkably, modulation of LGR5 expression has a major impact on the actin cytoskeletal structure and cell adhesion in the absence of RSPO stimulation, but the molecular mechanism is unclear. Here, we show that LGR5 interacts with IQ motif-containing GTPase-activating protein 1 (IQGAP1), an effector of Rac1/CDC42 GTPases, in the regulation of actin cytoskeleton dynamics and cell-cell adhesion. Specifically, LGR5 decreased levels of IQGAP1 phosphorylation at Ser-1441/1443, leading to increased binding of Rac1 to IQGAP1 and thus higher levels of cortical F-actin and enhanced cell-cell adhesion. LGR5 ablation in colon cancer cells and crypt stem cells resulted in loss of cortical F-actin, reduced cell-cell adhesion, and disrupted localization of adhesion-associated proteins. No evidence of LGR5 coupling to any of the four major subtypes of heterotrimeric G proteins was found. These findings suggest that LGR5 primarily functions via the IQGAP1-Rac1 pathway to strengthen cell-cell adhesion in normal adult crypt stem cells and colon cancer cells.


Asunto(s)
Adhesión Celular , Neoplasias del Colon/patología , Receptores Acoplados a Proteínas G/metabolismo , Células Madre/citología , Proteína de Unión al GTP rac1/metabolismo , Proteínas Activadoras de ras GTPasa/metabolismo , Animales , Células CHO , Células Cultivadas , Neoplasias del Colon/metabolismo , Cricetulus , Células HEK293 , Humanos , Células Madre/metabolismo
5.
J Biol Chem ; 292(34): 14280-14289, 2017 08 25.
Artículo en Inglés | MEDLINE | ID: mdl-28655769

RESUMEN

The visual photo-transduction cascade is a prototypical G protein-coupled receptor (GPCR) signaling system, in which light-activated rhodopsin (Rho*) is the GPCR catalyzing the exchange of GDP for GTP on the heterotrimeric G protein transducin (GT). This results in the dissociation of GT into its component αT-GTP and ß1γ1 subunit complex. Structural information for the Rho*-GT complex will be essential for understanding the molecular mechanism of visual photo-transduction. Moreover, it will shed light on how GPCRs selectively couple to and activate their G protein signaling partners. Here, we report on the preparation of a stable detergent-solubilized complex between Rho* and a heterotrimer (GT*) comprising a GαT/Gαi1 chimera (αT*) and ß1γ1 The complex was formed on native rod outer segment membranes upon light activation, solubilized in lauryl maltose neopentyl glycol, and purified with a combination of affinity and size-exclusion chromatography. We found that the complex is fully functional and that the stoichiometry of Rho* to GαT* is 1:1. The molecular weight of the complex was calculated from small-angle X-ray scattering data and was in good agreement with a model consisting of one Rho* and one GT*. The complex was visualized by negative-stain electron microscopy, which revealed an architecture similar to that of the ß2-adrenergic receptor-GS complex, including a flexible αT* helical domain. The stability and high yield of the purified complex should allow for further efforts toward obtaining a high-resolution structure of this important signaling complex.


Asunto(s)
Proteínas del Ojo/metabolismo , Subunidades beta de la Proteína de Unión al GTP/metabolismo , Subunidades gamma de la Proteína de Unión al GTP/metabolismo , Modelos Moleculares , Rodopsina/metabolismo , Transducina/metabolismo , Animales , Bovinos , Cristalografía por Rayos X , Detergentes/química , Proteínas del Ojo/química , Proteínas del Ojo/genética , Proteínas del Ojo/aislamiento & purificación , Subunidades beta de la Proteína de Unión al GTP/química , Subunidades beta de la Proteína de Unión al GTP/aislamiento & purificación , Subunidades gamma de la Proteína de Unión al GTP/química , Subunidades gamma de la Proteína de Unión al GTP/aislamiento & purificación , Luz , Microscopía Electrónica , Fragmentos de Péptidos/química , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/aislamiento & purificación , Fragmentos de Péptidos/metabolismo , Conformación Proteica/efectos de la radiación , Multimerización de Proteína/efectos de la radiación , Estabilidad Proteica/efectos de la radiación , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/aislamiento & purificación , Proteínas Recombinantes de Fusión/metabolismo , Retina/enzimología , Retina/metabolismo , Retina/efectos de la radiación , Rodopsina/química , Rodopsina/aislamiento & purificación , Segmento Externo de la Célula en Bastón/enzimología , Segmento Externo de la Célula en Bastón/metabolismo , Segmento Externo de la Célula en Bastón/efectos de la radiación , Dispersión del Ángulo Pequeño , Solubilidad , Transducina/química , Transducina/genética , Transducina/aislamiento & purificación , Difracción de Rayos X
6.
J Biol Chem ; 292(4): 1524-1534, 2017 01 27.
Artículo en Inglés | MEDLINE | ID: mdl-27986810

RESUMEN

GPRC6A is a G protein-coupled receptor activated by l-amino acids, which, based on analyses of knock-out mice, has been suggested to have physiological functions in metabolism and testicular function. The human ortholog is, however, mostly retained intracellularly in contrast to the cell surface-expressed murine and goldfish orthologs. The latter orthologs are Gq-coupled and lead to intracellular accumulation of inositol phosphates and calcium release. In the present study we cloned the bonobo chimpanzee GPRC6A receptor, which is 99% identical to the human receptor, and show that it is cell surface-expressed and functional. By analyses of chimeric human/mouse and human/bonobo receptors, bonobo receptor mutants, and the single nucleotide polymorphism database at NCBI, we identify an insertion/deletion variation in the third intracellular loop responsible for the intracellular retention and lack of function of the human ortholog. Genetic analyses of the 1000 genome database and the Inter99 cohort of 6,000 Danes establish the distribution of genotypes among ethnic groups, showing that the cell surface-expressed and functional variant is much more prevalent in the African population than in European and Asian populations and that this variant is partly linked with a stop codon early in the receptor sequence (rs6907580, amino acid position 57). In conclusion, our data solve a more than decade-old question of why the cloned human GPRC6A receptor is not cell surface-expressed and functional and provide a genetic framework to study human phenotypic traits in large genome sequencing projects linked with physiological measurement and biomarkers.


Asunto(s)
Regulación de la Expresión Génica , Mutación INDEL , Receptores Acoplados a Proteínas G , Animales , Línea Celular , Humanos , Ratones , Estructura Secundaria de Proteína , Receptores Acoplados a Proteínas G/biosíntesis , Receptores Acoplados a Proteínas G/genética
7.
J Biol Chem ; 291(17): 8969-77, 2016 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-26984408

RESUMEN

The ß-arrestins (ßarrs) are versatile, multifunctional adapter proteins that are best known for their ability to desensitize G protein-coupled receptors (GPCRs), but also regulate a diverse array of cellular functions. To signal in such a complex fashion, ßarrs adopt multiple conformations and are regulated at multiple levels to differentially activate downstream pathways. Recent structural studies have demonstrated that ßarrs have a conserved structure and activation mechanism, with plasticity of their structural fold, allowing them to adopt a wide array of conformations. Novel roles for ßarrs continue to be identified, demonstrating the importance of these dynamic regulators of cellular signaling.


Asunto(s)
Arrestina/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal/fisiología , Animales , Arrestina/genética , Humanos , Conformación Proteica , Receptores Acoplados a Proteínas G/genética
8.
J Biol Chem ; 291(21): 11285-99, 2016 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-27030010

RESUMEN

Agonist-evoked endocytosis of G protein-coupled receptors has been extensively studied. The mechanisms by which agonists stimulate mobilization and plasma membrane translocation of G protein-coupled receptors from intracellular stores are unexplored. Protease-activated receptor-2 (PAR2) traffics to lysosomes, and sustained protease signaling requires mobilization and plasma membrane trafficking of PAR2 from Golgi stores. We evaluated the contribution of protein kinase D (PKD) and Gßγ to this process. In HEK293 and KNRK cells, the PAR2 agonists trypsin and 2-furoyl-LIGRLO-NH2 activated PKD in the Golgi apparatus, where PKD regulates protein trafficking. PAR2 activation induced translocation of Gßγ, a PKD activator, to the Golgi apparatus, determined by bioluminescence resonance energy transfer between Gγ-Venus and giantin-Rluc8. Inhibitors of PKD (CRT0066101) and Gßγ (gallein) prevented PAR2-stimulated activation of PKD. CRT0066101, PKD1 siRNA, and gallein all inhibited recovery of PAR2-evoked Ca(2+) signaling. PAR2 with a photoconvertible Kaede tag was expressed in KNRK cells to examine receptor translocation from the Golgi apparatus to the plasma membrane. Irradiation of the Golgi region (405 nm) induced green-red photo-conversion of PAR2-Kaede. Trypsin depleted PAR2-Kaede from the Golgi apparatus and repleted PAR2-Kaede at the plasma membrane. CRT0066101 inhibited PAR2-Kaede translocation to the plasma membrane. CRT0066101 also inhibited sustained protease signaling to colonocytes and nociceptive neurons that naturally express PAR2 and mediate protease-evoked inflammation and nociception. Our results reveal a major role for PKD and Gßγ in agonist-evoked mobilization of intracellular PAR2 stores that is required for sustained signaling by extracellular proteases.


Asunto(s)
Subunidades beta de la Proteína de Unión al GTP/metabolismo , Subunidades gamma de la Proteína de Unión al GTP/metabolismo , Proteína Quinasa C/metabolismo , Receptor PAR-2/agonistas , Receptor PAR-2/metabolismo , Animales , Transporte Biológico Activo/efectos de los fármacos , Señalización del Calcio , Línea Celular , Membrana Celular/metabolismo , Endosomas/metabolismo , Subunidades beta de la Proteína de Unión al GTP/antagonistas & inhibidores , Subunidades gamma de la Proteína de Unión al GTP/antagonistas & inhibidores , Aparato de Golgi/metabolismo , Células HEK293 , Humanos , Ratones , Ratones Endogámicos C57BL , Proteína Quinasa C/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Pirimidinas/farmacología , Ratas , Xantenos/farmacología
9.
J Biol Chem ; 291(15): 7809-20, 2016 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-26851281

RESUMEN

Designerreceptorsexclusivelyactivated by adesignerdrug (DREADDs) are clozapine-N-oxide-sensitive designer G protein-coupled receptors (GPCRs) that have emerged as powerful novel chemogenetic tools to study the physiological relevance of GPCR signaling pathways in specific cell types or tissues. Like endogenous GPCRs, clozapine-N-oxide-activated DREADDs do not only activate heterotrimeric G proteins but can also trigger ß-arrestin-dependent (G protein-independent) signaling. To dissect the relative physiological relevance of G protein-mediatedversusß-arrestin-mediated signaling in different cell types or physiological processes, the availability of G protein- and ß-arrestin-biased DREADDs would be highly desirable. In this study, we report the development of a mutationally modified version of a non-biased DREADD derived from the M3muscarinic receptor that can activate Gq/11with high efficacy but lacks the ability to interact with ß-arrestins. We also demonstrate that this novel DREADD is activein vivoand that cell type-selective expression of this new designer receptor can provide novel insights into the physiological roles of G protein (Gq/11)-dependentversusß-arrestin-dependent signaling in hepatocytes. Thus, this novel Gq/11-biased DREADD represents a powerful new tool to study the physiological relevance of Gq/11-dependent signaling in distinct tissues and cell types, in the absence of ß-arrestin-mediated cellular effects. Such studies should guide the development of novel classes of functionally biased ligands that show high efficacy in various pathophysiological conditions but display a reduced incidence of side effects.


Asunto(s)
Subunidades alfa de la Proteína de Unión al GTP Gq-G11/metabolismo , Hepatocitos/metabolismo , Mapeo de Interacción de Proteínas , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal , Animales , Arrestinas/metabolismo , Células COS , Calcio/metabolismo , Células Cultivadas , Chlorocebus aethiops , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/genética , Técnicas de Silenciamiento del Gen , Glucosa/metabolismo , Células HEK293 , Humanos , Ratones Endogámicos C57BL , Mapeo de Interacción de Proteínas/métodos , Mapas de Interacción de Proteínas , beta-Arrestinas
10.
J Biol Chem ; 291(25): 13132-46, 2016 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-27080256

RESUMEN

Although rhodopsin-like G protein-coupled receptors can exist as both monomers and non-covalently associated dimers/oligomers, the steady-state proportion of each form and whether this is regulated by receptor ligands are unknown. Herein we address these topics for the M1 muscarinic acetylcholine receptor, a key molecular target for novel cognition enhancers, by using spatial intensity distribution analysis. This method can measure fluorescent particle concentration and assess oligomerization states of proteins within defined regions of living cells. Imaging and analysis of the basolateral surface of cells expressing some 50 molecules·µm(-2) human muscarinic M1 receptor identified a ∼75:25 mixture of receptor monomers and dimers/oligomers. Both sustained and shorter term treatment with the selective M1 antagonist pirenzepine resulted in a large shift in the distribution of receptor species to favor the dimeric/oligomeric state. Although sustained treatment with pirenzepine also resulted in marked up-regulation of the receptor, simple mass action effects were not the basis for ligand-induced stabilization of receptor dimers/oligomers. The related antagonist telenzepine also produced stabilization and enrichment of the M1 receptor dimer population, but the receptor subtype non-selective antagonists atropine and N-methylscopolamine did not. In contrast, neither pirenzepine nor telenzepine altered the quaternary organization of the related M3 muscarinic receptor. These data provide unique insights into the selective capacity of receptor ligands to promote and/or stabilize receptor dimers/oligomers and demonstrate that the dynamics of ligand regulation of the quaternary organization of G protein-coupled receptors is markedly more complex than previously appreciated. This may have major implications for receptor function and behavior.


Asunto(s)
Atropina/farmacología , Antagonistas Muscarínicos/farmacología , Pirenzepina/análogos & derivados , Pirenzepina/farmacología , Multimerización de Proteína/efectos de los fármacos , Receptor Muscarínico M1/antagonistas & inhibidores , Línea Celular , Humanos , Receptor Muscarínico M1/química , Receptor Muscarínico M1/metabolismo
11.
J Biol Chem ; 291(8): 3967-81, 2016 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-26703465

RESUMEN

Angiotensin II (Ang II) is a vasopressive hormone but is also a potent activator of cellular migration. We have previously shown that it can promote the activation of the GTPase ARF6 in a heterologous overexpressing system. The molecular mechanisms by which receptors control the activation of this small G protein remain, however, largely unknown. Furthermore, how ARF6 coordinates the activation of complex cellular responses needs to be further elucidated. In this study, we demonstrate that Ang II receptors engage ß-arrestin, but not Gq, to mediate ARF6 activation in HEK 293 cells. To further confirm the key role of ß-arrestin proteins, we overexpressed ß-arrestin2-(1-320), a dominant negative mutant known to block receptor endocytosis. We show that expression of this truncated construct does not support the activation of the GTPase nor cell migration. Interestingly, ß-arrestin2 can interact with the ARF guanine nucleotide exchange factor ARNO, although the C-terminally lacking mutant does not. We finally examined whether receptor endocytosis controlled ARF6 activation and cell migration. Although the clathrin inhibitor PitStop2 did not impact the ability of Ang II to activate ARF6, cell migration was markedly impaired. To further show that ARF activation regulates key signaling events leading to migration, we also examined MAPK activation. We demonstrate that this signaling axis is relevant in smooth muscle cells of the vasculature. Altogether, our findings show for the first time that Ang II receptor signaling to ß-arrestin regulates ARF6 activation. These proteins together control receptor endocytosis and ultimately cell migration.


Asunto(s)
Factores de Ribosilacion-ADP/metabolismo , Angiotensina II/metabolismo , Arrestinas/metabolismo , Movimiento Celular/fisiología , Endocitosis/fisiología , Sistema de Señalización de MAP Quinasas/fisiología , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Factor 6 de Ribosilación del ADP , Factores de Ribosilacion-ADP/genética , Angiotensina II/genética , Animales , Arrestinas/genética , Movimiento Celular/efectos de los fármacos , Endocitosis/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Activación Enzimática/fisiología , Quinasas MAP Reguladas por Señal Extracelular/genética , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Células HEK293 , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Músculo Liso Vascular/citología , Miocitos del Músculo Liso/citología , Ratas , Ratas Wistar , Receptores de Angiotensina/genética , Receptores de Angiotensina/metabolismo , Sulfonamidas/farmacología , Tiazolidinas/farmacología , beta-Arrestinas
12.
J Biol Chem ; 290(12): 7685-92, 2015 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-25605722

RESUMEN

The ACTH receptor, known as the melanocortin-2 receptor (MC2R), plays an important role in regulating and maintaining adrenocortical function. MC2R is a subtype of the melanocortin receptor (MCR) family and has unique characteristics among MCRs. Endogenous ACTH is the only endogenous agonist for MC2R, whereas the melanocortin peptides α-, ß-, and γ-melanocyte-stimulating hormone and ACTH are full agonists for all other MCRs. In this study, we examined the molecular basis of MC2R responsible for ligand selectivity using ACTH analogs and MC2R mutagenesis. Our results indicate that substitution of Phe(7) with D-Phe or D-naphthylalanine (D-Nal(2')) in ACTH(1-24) caused a significant decrease in ligand binding affinity and potency. Substitution of Phe(7) with D-Nal(2') in ACTH(1-24) did not switch the ligand from agonist to antagonist at MC2R, which was observed in MC3R and MC4R. Substitution of Phe(7) with D-Phe(7) in ACTH(1-17) resulted in the loss of ligand binding and activity. Molecular analysis of MC2R indicated that only mutation of the third transmembrane domain of MC2R resulted in a decrease in D-Phe ACTH binding affinity and potency. Our results suggest that Phe(7) in ACTH plays an important role in ligand selectivity and that the third transmembrane domain of MC2R is crucial for ACTH selectivity and potency.


Asunto(s)
Receptores de Corticotropina/metabolismo , Secuencia de Aminoácidos , Sitios de Unión , Citometría de Flujo , Células HEK293 , Humanos , Ligandos , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Receptores de Corticotropina/química , Receptores de Corticotropina/genética
13.
J Biol Chem ; 289(18): 12330-42, 2014 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-24644289

RESUMEN

The atypical chemokine receptor, ACKR2 is a pivotal regulator of chemokine-driven inflammatory responses and works by binding, internalizing, and degrading inflammatory CC-chemokines. ACKR2 displays promiscuity of ligand binding and is capable of interacting with up to 14 different inflammatory CC-chemokines. Despite its prominent biological role, little is known about the structure/function relationship within ACKR2, which regulates ligand binding. Here we demonstrate that a conserved tyrosine motif at the N terminus of ACKR2 is essential for ligand binding, internalization, and scavenging. In addition we demonstrate that sulfation of this motif contributes to ligand internalization. Furthermore, a peptide derived from this region is capable of binding inflammatory chemokines and inhibits their interaction with their cognate signaling receptors. Importantly, the peptide is only active in the sulfated form, further confirming the importance of the sulfated tyrosines for function. Finally, we demonstrate that the bacterial protease, staphopain A, can cleave the N terminus of ACKR2 and suppress its ligand internalization activity. Overall, these results shed new light on the nature of the structural motifs in ACKR2 that are responsible for ligand binding. The study also highlights ACKR2-derived N-terminal peptides as being of potential therapeutic significance.


Asunto(s)
Secuencias de Aminoácidos , Quimiocinas/metabolismo , Receptores de Quimiocina/metabolismo , Tirosina/metabolismo , Secuencia de Aminoácidos , Animales , Sitios de Unión/genética , Western Blotting , Células CHO , Línea Celular Tumoral , Células Cultivadas , Cricetinae , Cricetulus , Cisteína Endopeptidasas/metabolismo , Endocitosis/genética , Células HEK293 , Humanos , Ligandos , Datos de Secuencia Molecular , Mutación , Péptidos/metabolismo , Unión Proteica , Interferencia de ARN , Receptores de Quimiocina/química , Receptores de Quimiocina/genética , Homología de Secuencia de Aminoácido , Sulfatos/metabolismo , Tirosina/genética
14.
J Biol Chem ; 289(38): 26492-26504, 2014 Sep 19.
Artículo en Inglés | MEDLINE | ID: mdl-25063812

RESUMEN

Cerebellar dysfunction causes ataxia characterized by loss of balance and coordination. Until now, the molecular and neuronal mechanisms of several types of inherited cerebellar ataxia have not been completely clarified. Here, we report that leucine-rich G protein-coupled receptor 4 (Lgr4/Gpr48) is highly expressed in Purkinje cells (PCs) in the cerebellum. Deficiency of Lgr4 leads to an ataxia-like phenotype in mice. Histologically, no obvious morphological changes were observed in the cerebellum of Lgr4 mutant mice. However, the number of PCs was slightly but significantly reduced in Lgr4(-/-) mice. In addition, in vitro electrophysiological analysis showed an impaired long term depression (LTD) at parallel fiber-PC (PF-PC) synapses in Lgr4(-/-) mice. Consistently, immunostaining experiments showed that the level of phosphorylated cAMP-responsive element-binding protein (Creb) was significantly decreased in Lgr4(-/-) PCs. Furthermore, treatment with forskolin, an adenylyl cyclase agonist, rescued phospho-Creb in PCs and reversed the impairment in PF-PC LTD in Lgr4(-/-) cerebellar slices, indicating that Lgr4 is an upstream regulator of Creb signaling, which is underlying PF-PC LTD. Together, our findings demonstrate for first time an important role for Lgr4 in motor coordination and cerebellar synaptic plasticity and provide a potential therapeutic target for certain types of inherited cerebellar ataxia.


Asunto(s)
Depresión Sináptica a Largo Plazo , Células de Purkinje/fisiología , Receptores Acoplados a Proteínas G/genética , Sinapsis/fisiología , Animales , Proliferación Celular , Ataxia Cerebelosa/genética , Ataxia Cerebelosa/fisiopatología , Corteza Cerebelosa/fisiopatología , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Técnicas de Placa-Clamp , Reconocimiento Visual de Modelos , Fenotipo , Desempeño Psicomotor , Receptores Acoplados a Proteínas G/deficiencia , Prueba de Desempeño de Rotación con Aceleración Constante , Transmisión Sináptica
15.
J Biol Chem ; 289(6): 3689-702, 2014 Feb 07.
Artículo en Inglés | MEDLINE | ID: mdl-24338481

RESUMEN

The C terminus of G protein-coupled receptors (GPCRs) is important for G protein-coupling and activation; in addition, sorting motifs have been identified in the C termini of several GPCRs that facilitate correct trafficking from the endoplasmic reticulum to the plasma membrane. The C terminus of the GPCR TGR5 lacks any known sorting motif such that other factors must determine its trafficking. Here, we investigate deletion and substitution variants of the membrane-proximal C terminus of TGR5 with respect to plasma membrane localization and function using immunofluorescence staining, flow cytometry, and luciferase assays. Peptides of the membrane-proximal C-terminal variants are subjected to molecular dynamics simulations and analyzed with respect to their secondary structure. Our results reveal that TGR5 plasma membrane localization and responsiveness to extracellular ligands is fostered by a long (≥ 9 residues) α-helical stretch at the C terminus, whereas the presence of ß-strands or only a short α-helical stretch leads to retention in the endoplasmic reticulum and a loss of function. As a proof-of-principle, chimeras of TGR5 containing the membrane-proximal amino acids of the ß2 adrenergic receptor (ß2AR), the sphingosine 1-phosphate receptor-1 (S1P1), or the κ-type opioid receptor (κOR) were generated. These TGR5ß2AR, TGR5S1P1, or TGR5κOR chimeras were correctly sorted to the plasma membrane. As the exchanged amino acids of the ß2AR, the S1P1, or the κOR form α-helices in crystal structures but lack significant sequence identity to the respective TGR5 sequence, we conclude that the secondary structure of the TGR5 membrane-proximal C terminus is the determining factor for plasma membrane localization and responsiveness towards extracellular ligands.


Asunto(s)
Membrana Celular/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Membrana Celular/genética , Retículo Endoplásmico/genética , Retículo Endoplásmico/metabolismo , Células HEK293 , Humanos , Péptidos/genética , Péptidos/metabolismo , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Transporte de Proteínas/fisiología , Receptores Adrenérgicos beta 2/genética , Receptores Adrenérgicos beta 2/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores de Lisoesfingolípidos/genética , Receptores de Lisoesfingolípidos/metabolismo , Receptores Opioides kappa/genética , Receptores Opioides kappa/metabolismo
16.
J Biol Chem ; 289(29): 20345-58, 2014 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-24860101

RESUMEN

The long-chain fatty acid receptor FFA4 (previously GPR120) is receiving substantial interest as a novel target for the treatment of metabolic and inflammatory disease. This study examines for the first time the detailed mode of binding of both long-chain fatty acid and synthetic agonist ligands at FFA4 by integrating molecular modeling, receptor mutagenesis, and ligand structure-activity relationship approaches in an iterative format. In doing so, residues required for binding of fatty acid and synthetic agonists to FFA4 have been identified. This has allowed for the refinement of a well validated model of the mode of ligand-FFA4 interaction that will be invaluable in the identification of novel ligands and the future development of this receptor as a therapeutic target. The model reliably predicted the effects of substituent variations on agonist potency, and it was also able to predict the qualitative effect of binding site mutations in the majority of cases.


Asunto(s)
Ácidos Grasos no Esterificados/metabolismo , Receptores Acoplados a Proteínas G/química , Receptores Acoplados a Proteínas G/metabolismo , Sustitución de Aminoácidos , Arrestinas/metabolismo , Sitios de Unión , Ácidos Grasos no Esterificados/química , Células HEK293 , Humanos , Enlace de Hidrógeno , Ligandos , Metilaminas/metabolismo , Modelos Moleculares , Mutagénesis Sitio-Dirigida , Resonancia Magnética Nuclear Biomolecular , Propionatos/metabolismo , Conformación Proteica , Receptores Acoplados a Proteínas G/genética , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Relación Estructura-Actividad , Termodinámica , Ácido alfa-Linolénico/metabolismo , beta-Arrestinas
17.
J Biol Chem ; 289(13): 9263-74, 2014 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-24509856

RESUMEN

The A2A receptor is a class A/rhodopsin-like G protein-coupled receptor. Coupling to its cognate protein, Gs, occurs via restricted collision coupling and is contingent on the presence of cholesterol. Agonist activation slows diffusion of the A2A adenosine receptor in the lipid bilayer. We explored the contribution of the hydrophobic core and of the extended C terminus by examining diffusion of quantum dot-labeled receptor variants in dissociated hippocampal neurons. Single particle tracking of the A2A receptor(1-311), which lacks the last 101 residues, revealed that agonist-induced confinement was abolished and that the agonist-induced decrease in diffusivity was reduced substantially. A fragment comprising the SH3 domain and the guanylate kinase domain of synapse-associated protein 102 (SAP102) was identified as a candidate interactor that bound to the A2A receptor C terminus. Complex formation between the A2A receptor and SAP102 was verified by coimmunoprecipitation and by tracking its impact on receptor diffusion. An analysis of all trajectories by a hidden Markov model was consistent with two diffusion states where agonist activation reduced the transition between the two states and, thus, promoted the accumulation of the A2A receptor in the compartment with slow mobility. Overexpression of SAP102 precluded the access of the A2A receptor to a compartment with restricted mobility. In contrast, a mutated A2A receptor (with (383)DVELL(387) replaced by RVRAA) was insensitive to the action of SAP102. These observations show that the hydrophobic core per se does not fully account for the agonist-promoted change in mobility of the A2A receptor. The extended carboxyl terminus allows for regulatory input by scaffolding molecules such as SAP102.


Asunto(s)
Agonistas del Receptor de Adenosina A2/farmacología , Hipocampo/citología , Modelos Neurológicos , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Proteínas Nucleares/metabolismo , Receptor de Adenosina A2A/metabolismo , Factores de Transcripción/metabolismo , Animales , Difusión/efectos de los fármacos , Células HEK293 , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Cinética , Cadenas de Markov , Mutación , Ratas , Receptor de Adenosina A2A/química , Receptor de Adenosina A2A/genética
18.
J Biol Chem ; 289(45): 31173-87, 2014 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-25271156

RESUMEN

Phosphorylation plays vital roles in the regulation of G protein-coupled receptor (GPCR) functions. The apelin and apelin receptor (APJ) system is involved in the regulation of cardiovascular function and central control of body homeostasis. Here, using tandem mass spectrometry, we first identified phosphorylated serine residues in the C terminus of APJ. To determine the role of phosphorylation sites in APJ-mediated G protein-dependent and -independent signaling and function, we induced a mutation in the C-terminal serine residues and examined their effects on the interaction between APJ with G protein or GRK/ß-arrestin and their downstream signaling. Mutation of serine 348 led to an elimination of both GRK and ß-arrestin recruitment to APJ induced by apelin-13. Moreover, APJ internalization and G protein-independent ERK signaling were also abolished by point mutation at serine 348. In contrast, this mutant at serine residues had no demonstrable impact on apelin-13-induced G protein activation and its intracellular signaling. These findings suggest that mutation of serine 348 resulted in inactive GRK/ß-arrestin. However, there was no change in the active G protein thus, APJ conformation was biased. These results provide important information on the molecular interplay and impact of the APJ function, which may be extrapolated to design novel drugs for cardiac hypertrophy based on this biased signal pathway.


Asunto(s)
Regulación de la Expresión Génica , Péptidos y Proteínas de Señalización Intercelular/química , Receptores Acoplados a Proteínas G/metabolismo , Serina/química , Secuencia de Aminoácidos , Receptores de Apelina , Calcio/química , Membrana Celular/metabolismo , Células HEK293 , Humanos , Hipertrofia , Espectrometría de Masas , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Mutación , Fosforilación , Homología de Secuencia de Aminoácido , Transducción de Señal
19.
J Biol Chem ; 289(9): 5846-59, 2014 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-24375409

RESUMEN

Structural characterization of the human Y4 receptor (hY4R) interaction with human pancreatic polypeptide (hPP) is crucial, not only for understanding its biological function but also for testing treatment strategies for obesity that target this interaction. Here, the interaction of receptor mutants with pancreatic polypeptide analogs was studied through double-cycle mutagenesis. To guide mutagenesis and interpret results, a three-dimensional comparative model of the hY4R-hPP complex was constructed based on all available class A G protein-coupled receptor crystal structures and refined using experimental data. Our study reveals that residues of the hPP and the hY4R form a complex network consisting of ionic interactions, hydrophobic interactions, and hydrogen binding. Residues Tyr(2.64), Asp(2.68), Asn(6.55), Asn(7.32), and Phe(7.35) of Y4R are found to be important in receptor activation by hPP. Specifically, Tyr(2.64) interacts with Tyr(27) of hPP through hydrophobic contacts. Asn(7.32) is affected by modifications on position Arg(33) of hPP, suggesting a hydrogen bond between these two residues. Likewise, we find that Phe(7.35) is affected by modifications of hPP at positions 33 and 36, indicating interactions between these three amino acids. Taken together, we demonstrate that the top of transmembrane helix 2 (TM2) and the top of transmembrane helices 6 and 7 (TM6-TM7) form the core of the peptide binding pocket. These findings will contribute to the rational design of ligands that bind the receptor more effectively to produce an enhanced agonistic or antagonistic effect.


Asunto(s)
Polipéptido Pancreático/química , Receptores de Neuropéptido Y/química , Animales , Sitios de Unión , Células COS , Chlorocebus aethiops , Cristalografía por Rayos X , Células HEK293 , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Polipéptido Pancreático/genética , Polipéptido Pancreático/metabolismo , Estructura Cuaternaria de Proteína , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Receptores de Neuropéptido Y/genética , Receptores de Neuropéptido Y/metabolismo
20.
J Biol Chem ; 289(27): 18966-77, 2014 Jul 04.
Artículo en Inglés | MEDLINE | ID: mdl-24838244

RESUMEN

The corticotropin-releasing factor (CRF) type 1 receptor (CRF1R) for the 41-amino acid peptide CRF is a class B G protein-coupled receptor, which plays a key role in the response of our body to stressful stimuli and the maintenance of homeostasis by regulating neural and endocrine functions. CRF and related peptides, such as sauvagine, bind to the extracellular regions of CRF1R and activate the receptor. In contrast, small nonpeptide antagonists, which are effective against stress-related disorders, such as depression and anxiety, have been proposed to interact with the helical transmembrane domains (TMs) of CRF1R and allosterically antagonize peptide binding and receptor activation. Here, we aimed to elucidate the role of the third TM (TM3) in the molecular mechanisms underlying activation of CRF1R. TM3 was selected because its tilted orientation, relative to the membrane, allows its residues to establish key interactions with ligands, other TM helices, and the G protein. Using a combination of pharmacological, biochemical, and computational approaches, we found that Phe-203(3.40) and Gly-210(3.47) in TM3 play an important role in receptor activation. Our experimental findings also suggest that Phe-203(3.40) interacts with nonpeptide antagonists.


Asunto(s)
Membrana Celular/metabolismo , Receptores de Hormona Liberadora de Corticotropina/química , Receptores de Hormona Liberadora de Corticotropina/metabolismo , Regulación Alostérica/efectos de los fármacos , Secuencia de Aminoácidos , Secuencia Conservada , Células HEK293 , Humanos , Modelos Moleculares , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Estructura Terciaria de Proteína , Pirimidinas/farmacología , Pirroles/farmacología , Receptores de Hormona Liberadora de Corticotropina/antagonistas & inhibidores , Receptores de Hormona Liberadora de Corticotropina/genética , Alineación de Secuencia , Relación Estructura-Actividad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA