Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 69
Filtrar
1.
Mol Cell ; 81(22): 4605-4621.e11, 2021 11 18.
Artículo en Inglés | MEDLINE | ID: mdl-34582793

RESUMEN

G-protein-coupled receptors (GPCRs), also known as seven transmembrane receptors (7TMRs), typically interact with two distinct signal-transducers, i.e., G proteins and ß-arrestins (ßarrs). Interestingly, there are some non-canonical 7TMRs that lack G protein coupling but interact with ßarrs, although an understanding of their transducer coupling preference, downstream signaling, and structural mechanism remains elusive. Here, we characterize two such non-canonical 7TMRs, namely, the decoy D6 receptor (D6R) and the complement C5a receptor subtype 2 (C5aR2), in parallel with their canonical GPCR counterparts. We discover that D6R and C5aR2 efficiently couple to ßarrs, exhibit distinct engagement of GPCR kinases (GRKs), and activate non-canonical downstream signaling pathways. We also observe that ßarrs adopt distinct conformations for D6R and C5aR2, compared to their canonical GPCR counterparts, in response to common natural agonists. Our study establishes D6R and C5aR2 as ßarr-coupled 7TMRs and provides key insights into their regulation and signaling with direct implication for biased agonism.


Asunto(s)
Membrana Celular/metabolismo , Conformación Proteica , Transducción de Señal , beta-Arrestinas/química , Animales , Proteínas de Unión al GTP/química , Células HEK293 , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fosforilación , Unión Proteica , Dominios Proteicos , Estructura Secundaria de Proteína , Transporte de Proteínas , Receptor de Anafilatoxina C5a/metabolismo
2.
J Biol Chem ; 298(9): 102294, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35872018

RESUMEN

Promiscuous G protein-coupled receptors (GPCRs) engage multiple Gα subtypes with different efficacies to propagate signals in cells. A mechanistic understanding of Gα selectivity by GPCRs is critical for therapeutic design, since signaling can be restrained by ligand-receptor complexes to preferentially engage specific G proteins. However, details of GPCR selectivity are unresolved. Here, we investigated cognate G protein selectivity using the prototypical promiscuous Gαq/11 and Gα12/13 coupling receptors, angiotensin II type I receptor (AT1R) and prostaglandin F2α receptor (FP), bioluminescence resonance energy transfer-based G protein and pathway-selective sensors, and G protein knockout cells. We determined that competition between G proteins for receptor binding occurred in a receptor- and G protein-specific manner for AT1R and FP but not for other receptors tested. In addition, we show that while Gα12/13 competes with Gαq/11 for AT1R coupling, the opposite occurs for FP, and Gαq-mediated signaling regulated G protein coupling only at AT1R. In cells, the functional modulation of biased ligands at FP and AT1R was contingent upon cognate Gα availability. The efficacy of AT1R-biased ligands, which poorly signal through Gαq/11, increased in the absence of Gα12/13. Finally, we show that a positive allosteric modulator of Gαq/11 signaling that also allosterically decreases FP-Gα12/13 coupling, lost its negative modulation in the absence of Gαq/11 coupling to FP. Together, our findings suggest that despite preferential binding of similar subsets of G proteins, GPCRs follow distinct selectivity rules, which may contribute to the regulation of ligand-mediated G protein bias of AT1R and FP.


Asunto(s)
Subunidades alfa de la Proteína de Unión al GTP G12-G13 , Subunidades alfa de la Proteína de Unión al GTP Gq-G11 , Receptor de Angiotensina Tipo 1 , Receptores de Prostaglandina , Subunidades alfa de la Proteína de Unión al GTP G12-G13/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/metabolismo , Células HEK293 , Humanos , Ligandos , Receptor de Angiotensina Tipo 1/metabolismo , Receptores de Prostaglandina/metabolismo
3.
EMBO Rep ; 21(9): e49886, 2020 09 03.
Artículo en Inglés | MEDLINE | ID: mdl-32715625

RESUMEN

ß-arrestins (ßarrs) are key regulators of G protein-coupled receptor (GPCR) signaling and trafficking, and their knockdown typically leads to a decrease in agonist-induced ERK1/2 MAP kinase activation. Interestingly, for some GPCRs, knockdown of ßarr1 augments agonist-induced ERK1/2 phosphorylation although a mechanistic basis for this intriguing phenomenon is unclear. Here, we use selected GPCRs to explore a possible correlation between the spatial positioning of receptor phosphorylation sites and the contribution of ßarr1 in ERK1/2 activation. We discover that engineering a spatially positioned double-phosphorylation-site cluster in the bradykinin receptor (B2 R), analogous to that present in the vasopressin receptor (V2 R), reverses the contribution of ßarr1 in ERK1/2 activation from inhibitory to promotive. An intrabody sensor suggests a conformational mechanism for this role reversal of ßarr1, and molecular dynamics simulation reveals a bifurcated salt bridge between this double-phosphorylation site cluster and Lys294 in the lariat loop of ßarr1, which directs the orientation of the lariat loop. Our findings provide novel insights into the opposite roles of ßarr1 in ERK1/2 activation for different GPCRs with a direct relevance to biased agonism and novel therapeutics.


Asunto(s)
Sistema de Señalización de MAP Quinasas , Receptores Acoplados a Proteínas G , Células HEK293 , Humanos , Fosforilación , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , beta-Arrestina 1/metabolismo , beta-Arrestinas/metabolismo
4.
Nature ; 531(7596): 665-8, 2016 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-27007854

RESUMEN

Arrestins are cytosolic proteins that regulate G-protein-coupled receptor (GPCR) desensitization, internalization, trafficking and signalling. Arrestin recruitment uncouples GPCRs from heterotrimeric G proteins, and targets the proteins for internalization via clathrin-coated pits. Arrestins also function as ligand-regulated scaffolds that recruit multiple non-G-protein effectors into GPCR-based 'signalsomes'. Although the dominant function(s) of arrestins vary between receptors, the mechanism whereby different GPCRs specify these divergent functions is unclear. Using a panel of intramolecular fluorescein arsenical hairpin (FlAsH) bioluminescence resonance energy transfer (BRET) reporters to monitor conformational changes in ß-arrestin2, here we show that GPCRs impose distinctive arrestin 'conformational signatures' that reflect the stability of the receptor-arrestin complex and role of ß-arrestin2 in activating or dampening downstream signalling events. The predictive value of these signatures extends to structurally distinct ligands activating the same GPCR, such that the innate properties of the ligand are reflected as changes in ß-arrestin2 conformation. Our findings demonstrate that information about ligand-receptor conformation is encoded within the population average ß-arrestin2 conformation, and provide insight into how different GPCRs can use a common effector for different purposes. This approach may have application in the characterization and development of functionally selective GPCR ligands and in identifying factors that dictate arrestin conformation and function.


Asunto(s)
Arrestinas/química , Arrestinas/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal , Animales , Activación Enzimática , Células HEK293 , Humanos , Ligandos , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Conformación Proteica , Transporte de Proteínas , Ratas , Receptores Acoplados a Proteínas G/química , beta-Arrestinas
5.
Mol Pharmacol ; 100(4): 372-387, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34353882

RESUMEN

ONC201 is a first-in-class imipridone compound that is in clinical trials for the treatment of high-grade gliomas and other advanced cancers. Recent studies identified that ONC201 antagonizes D2-like dopamine receptors at therapeutically relevant concentrations. In the current study, characterization of ONC201 using radioligand binding and multiple functional assays revealed that it was a full antagonist of the D2 and D3 receptors (D2R and D3R) with low micromolar potencies, similar to its potency for antiproliferative effects. Curve-shift experiments using D2R-mediated ß-arrestin recruitment and cAMP assays revealed that ONC201 exhibited a mixed form of antagonism. An operational model of allostery was used to analyze these data, which suggested that the predominant modulatory effect of ONC201 was on dopamine efficacy with little to no effect on dopamine affinity. To investigate how ONC201 binds to the D2R, we employed scanning mutagenesis coupled with a D2R-mediated calcium efflux assay. Eight residues were identified as being important for ONC201's functional antagonism of the D2R. Mutation of these residues followed by assessing ONC201 antagonism in multiple signaling assays highlighted specific residues involved in ONC201 binding. Together with computational modeling and simulation studies, our results suggest that ONC201 interacts with the D2R in a bitopic manner where the imipridone core of the molecule protrudes into the orthosteric binding site, but does not compete with dopamine, whereas a secondary phenyl ring engages an allosteric binding pocket that may be associated with negative modulation of receptor activity. SIGNIFICANCE STATEMENT: ONC201 is a novel antagonist of the D2 dopamine receptor with demonstrated efficacy in the treatment of various cancers, especially high-grade glioma. This study demonstrates that ONC201 antagonizes the D2 receptor with novel bitopic and negative allosteric mechanisms of action, which may explain its high selectivity and some of its clinical anticancer properties that are distinct from other D2 receptor antagonists widely used for the treatment of schizophrenia and other neuropsychiatric disorders.


Asunto(s)
Antineoplásicos/metabolismo , Antagonistas de los Receptores de Dopamina D2/metabolismo , Imidazoles/metabolismo , Piridinas/metabolismo , Pirimidinas/metabolismo , Receptores de Dopamina D2/metabolismo , Regulación Alostérica/efectos de los fármacos , Regulación Alostérica/fisiología , Animales , Antineoplásicos/química , Antineoplásicos/farmacología , Células CHO , Cricetinae , Cricetulus , Antagonistas de los Receptores de Dopamina D2/química , Antagonistas de los Receptores de Dopamina D2/farmacología , Relación Dosis-Respuesta a Droga , Células HEK293 , Humanos , Imidazoles/química , Imidazoles/farmacología , Unión Proteica/efectos de los fármacos , Unión Proteica/fisiología , Estructura Secundaria de Proteína , Piridinas/química , Piridinas/farmacología , Pirimidinas/química , Pirimidinas/farmacología , Receptores de Dopamina D2/química
6.
J Biol Chem ; 295(38): 13169-13180, 2020 09 18.
Artículo en Inglés | MEDLINE | ID: mdl-32703898

RESUMEN

The angiotensin II (AngII) type 1 receptor (AT1R), a member of the G protein-coupled receptor (GPCR) family, signals through G proteins and ß-arrestins, which act as adaptors to regulate AT1R internalization and mitogen-activated protein kinase (MAPK) ERK1/2 activation. ß-arrestin-dependent ERK1/2 regulation is the subject of important studies because its spatiotemporal control remains poorly understood for many GPCRs, including AT1R. To study the link between ß-arrestin-dependent trafficking and ERK1/2 signaling, we investigated three naturally occurring AT1R variants that show distinct receptor-ß-arrestin interactions: A163T, T282M, and C289W. Using bioluminescence resonance energy transfer (BRET)-based and conformational fluorescein arsenical hairpin-BRET sensors coupled with high-resolution fluorescence microscopy, we show that all AT1R variants form complexes with ß-arrestin2 at the plasma membrane and efficiently internalize into endosomes upon AngII stimulation. However, mutant receptors imposed distinct conformations in ß-arrestin2 and differentially impacted endosomal trafficking and MAPK signaling. Notably, T282M accumulated in endosomes, but its ability to form stable complexes following internalization was reduced, markedly impairing its ability to co-traffic with ß-arrestin2. We also found that despite ß-arrestin2 overexpression, T282M's and C289W's residency with ß-arrestin2 in endosomes was greatly reduced, leading to decreased ß-arrestin-dependent ERK1/2 activation, faster recycling of receptors to the plasma membrane, and impaired AngII-mediated proliferation. Our findings reveal that naturally occurring AT1R variants alter the patterns of receptor/ß-arrestin2 trafficking and suggest conformationally dependent ß-arrestin-mediated MAPK activation as well as endosomal receptor-ß-arrestin complex stabilization in the mitogenic response of AT1R.


Asunto(s)
Endosomas/metabolismo , Sistema de Señalización de MAP Quinasas , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Receptor de Angiotensina Tipo 1/metabolismo , beta-Arrestinas/metabolismo , Sustitución de Aminoácidos , Angiotensina II/farmacología , Endosomas/genética , Activación Enzimática , Células HEK293 , Humanos , Proteína Quinasa 1 Activada por Mitógenos/genética , Proteína Quinasa 3 Activada por Mitógenos/genética , Mutación Missense , Receptor de Angiotensina Tipo 1/genética , beta-Arrestinas/genética
7.
J Biol Chem ; 294(46): 17409-17420, 2019 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-31530642

RESUMEN

The angiotensin II (AngII) type 1 receptor (AT1R) is a member of the G protein-coupled receptor (GPCR) family and binds ß-arrestins (ß-arrs), which regulate AT1R signaling and trafficking. These processes can be biased by different ligands or mutations in the AGTR1 gene. As for many GPCRs, the exact details for AT1R-ß-arr interactions driven by AngII or ß-arr-biased ligands remain largely unknown. Here, we used the amber-suppression technology to site-specifically introduce the unnatural amino acid (UAA) p-azido-l-phenylalanine (azF) into the intracellular loops (ICLs) and the C-tail of AT1R. Our goal was to generate competent photoreactive receptors that can be cross-linked to ß-arrs in cells. We performed UV-mediated photolysis of 25 different azF-labeled AT1Rs to cross-link ß-arr1 to AngII-bound receptors, enabling us to map important contact sites in the C-tail and in the ICL2 and ICL3 of the receptor. The extent of AT1R-ß-arr1 cross-linking among azF-labeled receptors differed, revealing variability in ß-arr's contact mode with the different AT1R domains. Moreover, the signature of ligated AT1R-ß-arr complexes from a subset of azF-labeled receptors also differed between AngII and ß-arr-biased ligand stimulation of receptors and between azF-labeled AT1R bearing and that lacking a bias signaling mutation. These observations further implied distinct interaction modalities of the AT1R-ß-arr1 complex in biased signaling conditions. Our findings demonstrate that this photocross-linking approach is useful for understanding GPCR-ß-arr complexes in different activation states and could be extended to study other protein-protein interactions in cells.


Asunto(s)
Receptor de Angiotensina Tipo 1/metabolismo , beta-Arrestina 1/metabolismo , Código Genético , Células HEK293 , Humanos , Luz , Unión Proteica , Mapas de Interacción de Proteínas , Receptor de Angiotensina Tipo 1/genética
8.
Proc Natl Acad Sci U S A ; 113(16): 4524-9, 2016 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-27071102

RESUMEN

G protein-coupled receptors (GPCRs) are known to initiate a plethora of signaling pathways in vitro. However, it is unclear which of these pathways are engaged to mediate physiological responses. Here, we examine the distinct roles of Gq/11-dependent signaling and receptor phosphorylation-dependent signaling in bronchial airway contraction and lung function regulated through the M3-muscarinic acetylcholine receptor (M3-mAChR). By using a genetically engineered mouse expressing a G protein-biased M3-mAChR mutant, we reveal the first evidence, to our knowledge, of a role for M3-mAChR phosphorylation in bronchial smooth muscle contraction in health and in a disease state with relevance to human asthma. Furthermore, this mouse model can be used to distinguish the physiological responses that are regulated by M3-mAChR phosphorylation (which include control of lung function) from those responses that are downstream of G protein signaling. In this way, we present an approach by which to predict the physiological/therapeutic outcome of M3-mAChR-biased ligands with important implications for drug discovery.


Asunto(s)
Bronquios/metabolismo , Músculo Liso/metabolismo , Receptor Muscarínico M3/metabolismo , Transducción de Señal/fisiología , Animales , Bronquios/citología , Humanos , Ratones , Ratones Noqueados , Músculo Liso/citología , Fosforilación/fisiología , Receptor Muscarínico M3/genética
9.
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
10.
Methods ; 92: 5-10, 2016 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-25890247

RESUMEN

There has been a growing appreciation that G protein-coupled receptor (GPCR) functional selectivity (viz. biased signaling), in particular between G protein- and ß-arrestin-dependent signaling, can be achieved with specific ligands, and that such directed signaling represents a promising avenue for improving drug efficacy and therapy. Thus, for any given GPCRs it is important to define means to pharmacologically characterize and classify drugs for their propensity to bias signaling. Here we describe an experimental protocol and step-by-step approach to assess functional selectivity between Gαq and ß-arrestin-dependent responses using the prototypical angiotensin II (AngII) type 1 receptor (AT1R) expressed in HEK 293 cells. The protocol describes the expression of Bioluminescence Resonance Energy Transfer (BRET) sensors for either Gαq or ß-arrestin with AT1R, and the use of the operational model of pharmacological agonism to quantify ligand bias. Such methods are equally applicable to other GPCRs and their downstream signaling effectors.


Asunto(s)
Transferencia de Energía por Resonancia de Bioluminiscencia/métodos , Técnicas Biosensibles/métodos , Receptor de Angiotensina Tipo 1/análisis , Receptor de Angiotensina Tipo 1/metabolismo , Transducción de Señal/fisiología , Angiotensina II/metabolismo , Relación Dosis-Respuesta a Droga , Células HEK293 , Humanos , Ligandos
11.
J Biol Chem ; 290(5): 3137-48, 2015 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-25512374

RESUMEN

The angiotensin II type I (AT1R) and the prostaglandin F2α (PGF2α) F prostanoid (FP) receptors are both potent regulators of blood pressure. Physiological interplay between AT1R and FP has been described. Abdominal aortic ring contraction experiments revealed that PGF2α-dependent activation of FP potentiated angiotensin II-induced contraction, whereas FP antagonists had the opposite effect. Similarly, PGF2α-mediated vasoconstriction was symmetrically regulated by co-treatment with AT1R agonist and antagonist. The underlying canonical Gαq signaling via production of inositol phosphates mediated by each receptor was also regulated by antagonists for the other receptor. However, binding to their respective agonists, regulation of receptor-mediated MAPK activation and vascular smooth muscle cell growth were differentially or asymmetrically regulated depending on how each of the two receptors were occupied by either agonist or antagonist. Physical interactions between these receptors have never been reported, and here we show that AT1R and FP form heterodimeric complexes in both HEK 293 and vascular smooth muscle cells. These findings imply that formation of the AT1R/FP dimer creates a novel allosteric signaling unit that shows symmetrical and asymmetrical signaling behavior, depending on the outcome measured. AT1R/FP dimers may thus be important in the regulation of blood pressure.


Asunto(s)
Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Receptor de Angiotensina Tipo 1/metabolismo , Receptores de Prostaglandina/metabolismo , Regulación Alostérica/fisiología , Animales , Células Cultivadas , Células HEK293 , Humanos , Ratas , Transducción de Señal/fisiología
12.
J Biol Chem ; 289(34): 23302-17, 2014 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-25016018

RESUMEN

ß-Arrestins are signaling adaptors that bind to agonist-occupied G protein-coupled receptors (GPCRs) and target them for endocytosis; however, the mechanisms regulating receptor/ß-arrestin complexes and trafficking in endosomes, remain ill defined. Here we show, in live cells, differential dynamic regulation of endosomal bradykinin B2 receptor (B2R) complexes with either ß-arrestin-1 or -2. We find a novel role for MAPK in the B2R/ß-arrestin-2 complex formation, receptor trafficking and signaling mediated by an ERK1/2 regulatory motif in the hinge domain of the rat ß-arrestin-2 (PET(178)P), but not rat ß-arrestin-1 (PER(177)P). While the ERK1/2 regulatory motif is conserved between rat and mouse ß-arrestin-2, it is surprisingly not conserved in human ß-arrestin-2 (PEK(178)P). However, mutation of lysine 178 to threonine is sufficient to confer MAPK sensitivity to the human ß-arrestin-2. Furthermore, substitution for a phosphomimetic residue in both the rat and the human ß-arrestin-2 (T/K178D) significantly stabilizes B2R/ß-arrestin complexes in endosomes, delays receptor recycling to the plasma membrane and maintains intracellular MAPK signaling. Similarly, the endosomal trafficking of ß2-adrenergic, angiotensin II type 1 and vasopressin V2 receptors was altered by the ß-arrestin-2 T178D mutant. Our findings unveil a novel subtype specific mode of MAPK-dependent regulation of ß-arrestins in intracellular trafficking and signaling of GPCRs, and suggest differential endosomal receptor/ß-arrestin-2 signaling roles among species.


Asunto(s)
Arrestinas/metabolismo , Endosomas/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Secuencia de Aminoácidos , Animales , Arrestinas/química , Células COS , Chlorocebus aethiops , Endocitosis , Células HEK293 , Humanos , Datos de Secuencia Molecular , Transporte de Proteínas , Ratas , Homología de Secuencia de Aminoácido , Transducción de Señal , beta-Arrestina 1 , Arrestina beta 2 , beta-Arrestinas
13.
Org Biomol Chem ; 13(28): 7750-61, 2015 Jul 28.
Artículo en Inglés | MEDLINE | ID: mdl-26090777

RESUMEN

On pursuing molecules that delay labour, so-called tocolytics, the prostaglandin F2α receptor (FP) was targeted, because of its role in the stimulation of uterine contractions leading to birth and preterm birth. Previously, both the indolizidinone PDC-113.824 (5) and the aza-glycinyl-proline analog 6 were shown to delay labour in mice by modulating the FP function, likely by an allosteric mechanism, which features biased signalling. The crystal structure and computational analyses of the indolizidin-2-one amino acid and aza-glycinyl-proline components of 5 and 6 in model peptides have shown them to adopt a geometry that mimics ideal type I and II'ß-turns. To elucidate the precise turn geometry for receptor recognition, analogs 1-4 have now been synthesized: macrocycle and pyrroloazepinone mimics 1 and 2 to mimic type I, and glycinyl-proline and d-alaninyl-proline analogs 3 and 4 to favour type II'ß-turn geometry. Notably, transannular cyclization of peptide macrocycle 13 has provided diastereoselectively pyrroloazepinone 15 by a novel route that provides effective access to mimics 1 and 2 by way of a common intermediate. Among the four analogs, none exhibited efficacy nor potency on par with 5 and 6; however, d-alaninyl-proline analog 4 proved superior to the other analogs in reducing PGF2α-induced myometrial contractions and inhibiting FP modulation of cell ruffling, a response dependent on the Gα12/RhoA/ROCK signaling pathway. Furthermore Gly-Pro analog 3 potentiated the effect of PGF2α on Gαq mediated ERK1/2 activation. Evidence that 4 adopted turn geometry was obtained by conformational analysis using NMR spectroscopy to characterize respectively the influence of solvent and temperature on the chemical shifts of the amide NH protons. Although mimicry of the type II' geometry by 3, 4, 5 and 6 may favour activity, distortion from ideal geometry by the indolizidinone and aza-glycinyl residues of the latter appears to enhance their biological effects.


Asunto(s)
Compuestos Aza/farmacología , Indolicidinas/farmacología , Oligopéptidos/farmacología , Receptores de Prostaglandina/antagonistas & inhibidores , Animales , Compuestos Aza/química , Indolicidinas/química , Ratones , Conformación Molecular , Oligopéptidos/química , Relación Estructura-Actividad
14.
Am J Respir Cell Mol Biol ; 49(4): 563-70, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23656597

RESUMEN

Allergic asthma is a heterogeneous disease with no curative therapies. T cells infiltrate the airway smooth muscle (ASM) layer and may be implicated in airway remodeling and the increase of ASM mass, a cardinal feature of asthma. The mechanism by which CD4(+) T cells drive airway remodeling remains unknown. This study sought to determine the T cell-mediated mechanism of ASM cell proliferation. We hypothesized that CD4(+) T cells adhere to ASM cells via CD44, and induce ASM cell proliferation through the activation of the epidermal growth factor receptor (EGFR). A coculture model showed that the contact of antigen-stimulated CD4(+) T cells with ASM cells induced high levels of EGFR ligand expression in CD4(+) T cells and the activation of matrix metalloproteinase (MMP)-9, required for the shedding of EGFR ligands. The inhibition of EGFR and MMP-9 prevented the increase of ASM cell proliferation after coculture. The hyaluronan receptor CD44 is the dominant mediator of the tight adherence of T cells to ASM and is colocalized with MMP-9 on the cell surface. Moreover, the neutralization of CD44 prevents ASM cell hyperplasia. These data provide a novel mechanism by which antigen-stimulated CD4(+) T cells induce the remodeling indicative of a direct trophic role for CD4(+) T cells.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Receptores ErbB/metabolismo , Músculo Liso/metabolismo , Miocitos del Músculo Liso/metabolismo , Sistema Respiratorio/inmunología , Sistema Respiratorio/metabolismo , Animales , Linfocitos T CD4-Positivos/metabolismo , Adhesión Celular/inmunología , Células Cultivadas , Técnicas de Cocultivo , Receptores ErbB/inmunología , Receptores de Hialuranos/inmunología , Activación de Linfocitos , Metaloproteinasa 9 de la Matriz/inmunología , Metaloproteinasa 9 de la Matriz/metabolismo , Músculo Liso/inmunología , Miocitos del Músculo Liso/inmunología , Ratas
15.
Sci Signal ; 16(797): eadf2173, 2023 08 08.
Artículo en Inglés | MEDLINE | ID: mdl-37552769

RESUMEN

G protein-coupled receptors engage both G proteins and ß-arrestins, and their coupling can be biased by ligands and mutations. Here, to resolve structural elements and mechanisms underlying effector coupling to the angiotensin II (AngII) type 1 receptor (AT1R), we combined alanine scanning mutagenesis of the entire sequence of the receptor with pharmacological profiling of Gαq and ß-arrestin engagement to mutant receptors and molecular dynamics simulations. We showed that Gαq coupling to AT1R involved a large number of residues spread across the receptor, whereas fewer structural regions of the receptor contributed to ß-arrestin coupling regulation. Residue stretches in transmembrane domain 4 conferred ß-arrestin bias and represented an important structural element in AT1R for functional selectivity. Furthermore, we identified allosteric small-molecule binding sites that were enclosed by communities of residues that produced biased signaling when mutated. Last, we showed that allosteric communication within AT1R emanating from the Gαq coupling site spread beyond the orthosteric AngII-binding site and across different regions of the receptor, including currently unresolved structural regions. Our findings reveal structural elements and mechanisms within AT1R that bias Gαq and ß-arrestin coupling and that could be harnessed to design biased receptors for research purposes and to develop allosteric modulators.


Asunto(s)
Receptor de Angiotensina Tipo 1 , Transducción de Señal , beta-Arrestinas/genética , beta-Arrestinas/metabolismo , Receptor de Angiotensina Tipo 1/genética , Receptor de Angiotensina Tipo 1/metabolismo , beta-Arrestina 1/metabolismo , Proteínas de Unión al GTP/metabolismo , Angiotensina II/metabolismo
16.
Nat Commun ; 14(1): 4808, 2023 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-37558722

RESUMEN

Chemokine receptors constitute an important subfamily of G protein-coupled receptors (GPCRs), and they are critically involved in a broad range of immune response mechanisms. Ligand promiscuity among these receptors makes them an interesting target to explore multiple aspects of biased agonism. Here, we comprehensively characterize two chemokine receptors namely, CXCR4 and CXCR7, in terms of their transducer-coupling and downstream signaling upon their stimulation by a common chemokine agonist, CXCL12, and a small molecule agonist, VUF11207. We observe that CXCR7 lacks G-protein-coupling while maintaining robust ßarr recruitment with a major contribution of GRK5/6. On the other hand, CXCR4 displays robust G-protein activation as expected but exhibits significantly reduced ßarr-coupling compared to CXCR7. These two receptors induce distinct ßarr conformations even when activated by the same agonist, and CXCR7, unlike CXCR4, fails to activate ERK1/2 MAP kinase. We also identify a key contribution of a single phosphorylation site in CXCR7 for ßarr recruitment and endosomal localization. Our study provides molecular insights into intrinsic-bias encoded in the CXCR4-CXCR7 system with broad implications for drug discovery.


Asunto(s)
Receptores CXCR , Receptores CXCR/genética , Receptores CXCR/metabolismo , Receptores CXCR4/metabolismo , Transducción de Señal , Proteínas de Unión al GTP , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Quimiocina CXCL12/metabolismo
17.
J Biol Chem ; 286(33): 29035-29043, 2011 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-21724843

RESUMEN

The type II transmembrane serine protease TMPRSS6 (also known as matriptase-2) controls iron homeostasis through its negative regulation of expression of hepcidin, a key hormone involved in iron metabolism. Upstream of the hepcidin-regulated signaling pathway, TMPRSS6 cleaves its target substrate hemojuvelin (HJV) at the plasma membrane, but the dynamics of the cell-surface expression of the protease have not been addressed. Here, we report that TMPRSS6 undergoes constitutive internalization in transfected HEK293 cells and in two human hepatic cell lines, HepG2 and primary hepatocytes, both of which express TMPRSS6 endogenously. Cell surface-labeled TMPRSS6 was internalized and was detected in clathrin- and AP-2-positive vesicles via a dynamin-dependent pathway. The endocytosed TMPRSS6 next transited in early endosomes and then to lysosomes. Internalization of TMPRSS6 is dependent on specific residues within its N-terminal cytoplasmic domain, as site-directed mutagenesis of these residues abrogated internalization and maintained the enzyme at the cell surface. Cells coexpressing these mutants and HJV produced significantly decreased levels of hepcidin compared with wild-type TMPRSS6 due to the sustained cleavage of HJV at the cell surface by TMPRSS6 mutants. Our results underscore for the first time the importance of TMPRSS6 trafficking at the plasma membrane in the regulation of hepcidin expression, an event that is essential for iron homeostasis.


Asunto(s)
Membrana Celular/enzimología , Endocitosis/fisiología , Proteínas de la Membrana/metabolismo , Serina Endopeptidasas/metabolismo , Péptidos Catiónicos Antimicrobianos/biosíntesis , Péptidos Catiónicos Antimicrobianos/genética , Membrana Celular/genética , Vesículas Cubiertas por Clatrina/enzimología , Vesículas Cubiertas por Clatrina/genética , Proteínas Ligadas a GPI/genética , Proteínas Ligadas a GPI/metabolismo , Regulación de la Expresión Génica/fisiología , Células HEK293 , Proteína de la Hemocromatosis , Células Hep G2 , Hepcidinas , Homeostasis/fisiología , Humanos , Hierro/metabolismo , Proteínas de la Membrana/genética , Transporte de Proteínas/fisiología , Serina Endopeptidasas/genética
18.
J Biol Chem ; 286(4): 2696-706, 2011 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-21112970

RESUMEN

L-type prostaglandin synthase (L-PGDS) produces PGD(2), a lipid mediator involved in neuromodulation and inflammation. Here, we show that L-PGDS and arrestin-3 (Arr3) interact directly and can be co-immunoprecipitated endogenously from MG-63 osteoblasts. Perinuclear L-PGDS/Arr3 co-localization is observed in PGD(2)-producing MG-63 cells and is induced by the addition of the L-PGDS substrate or co-expression of COX-2 in HEK293 cells. Inhibition of L-PGDS activity in MG-63 cells triggers redistribution of Arr3 and L-PGDS to the cytoplasm. Perinuclear localization of L-PGDS is detected in wild-type mouse embryonic fibroblasts (MEFs) but is more diffused in MEFs-arr-2(-/-)-arr-3(-/-). Arrestin-3 promotes PGD(2) production by L-PGDS in vitro. IL-1ß-induced PGD(2) production is significantly lower in MEFs-arr-2(-/-)-arr-3(-/-) than in wild-type MEFs but can be rescued by expressing Arr2 or Arr3. A peptide corresponding to amino acids 86-100 of arrestin-3 derived from its L-PGDS binding domain stimulates L-PGDS-mediated PGD(2) production in vitro and in MG-63 cells. We report the first characterization of an interactor/modulator of a PGD(2) synthase and the identification of a new function for arrestin, which may open new opportunities for improving therapies for the treatment of inflammatory diseases.


Asunto(s)
Arrestina/metabolismo , Oxidorreductasas Intramoleculares/metabolismo , Lipocalinas/metabolismo , Prostaglandina D2/biosíntesis , Animales , Arrestina/genética , Ciclooxigenasa 2/genética , Ciclooxigenasa 2/metabolismo , Células HEK293 , Humanos , Inflamación/genética , Inflamación/metabolismo , Oxidorreductasas Intramoleculares/genética , Lipocalinas/genética , Ratones , Ratones Noqueados , Estructura Terciaria de Proteína
19.
Front Pharmacol ; 13: 852029, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35418857

RESUMEN

Cannabis (marijuana) is the most commonly used illicit product in the world and is the second most smoked plant after tobacco. There has been a rapid increase in the number of countries legalizing cannabis for both recreational and medicinal purposes. Smoking cannabis in the form of a joint is the most common mode of cannabis consumption. Combustion of cannabis smoke generates many of the same chemicals as tobacco smoke. Although the impact of tobacco smoke on respiratory health is well-known, the consequence of cannabis smoke on the respiratory system and, in particular, the inflammatory response is unclear. Besides the combustion products present in cannabis smoke, cannabis also contains cannabinoids including Δ9-tetrahydrocannabinol (Δ9-THC) and cannabidiol (CBD). These compounds are hydrophobic and not present in aqueous solutions. In order to understand the impact of cannabis smoke on pathological mechanisms associated with adverse respiratory outcomes, the development of in vitro surrogates of cannabis smoke exposure is needed. Therefore, we developed a standardized protocol for the generation of cannabis smoke extract (CaSE) to investigate its effect on cellular mechanisms in vitro. First, we determined the concentration of Δ9-THC, one of the major cannabinoids, by ELISA and found that addition of methanol to the cell culture media during generation of the aqueous smoke extract significantly increased the amount of Δ9-THC. We also observed by LC-MS/MS that CaSE preparation with methanol contains CBD. Using a functional assay in cells for CB1 receptors, the major target of cannabinoids, we found that this CaSE contains Δ9-THC which activates CB1 receptors. Finally, this standardized preparation of CaSE induces an inflammatory response in human lung fibroblasts. This study provides an optimized protocol for aqueous CaSE preparation containing biologically active cannabinoids that can be used for in vitro experimentation of cannabis smoke and its potential impact on various indices of pulmonary health.

20.
Nat Commun ; 13(1): 7428, 2022 12 02.
Artículo en Inglés | MEDLINE | ID: mdl-36460632

RESUMEN

Recent studies have shown that G protein coupled receptors (GPCRs) show selective and promiscuous coupling to different Gα protein subfamilies and yet the mechanisms of the range of coupling preferences remain unclear. Here, we use Molecular Dynamics (MD) simulations on ten GPCR:G protein complexes and show that the location (spatial) and duration (temporal) of intermolecular contacts at the GPCR:Gα protein interface play a critical role in how GPCRs selectively interact with G proteins. We identify that some GPCR:G protein interface contacts are common across Gα subfamilies and others specific to Gα subfamilies. Using large scale data analysis techniques on the MD simulation snapshots we derive a spatio-temporal code for contacts that confer G protein selective coupling and validated these contacts using G protein activation BRET assays. Our results demonstrate that promiscuous GPCRs show persistent sampling of the common contacts more than G protein specific contacts. These findings suggest that GPCRs maintain contact with G proteins through a common central interface, while the selectivity comes from G protein specific contacts at the periphery of the interface.


Asunto(s)
Bioensayo , Simulación de Dinámica Molecular , Proyectos de Investigación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA