Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Nat Chem Biol ; 2024 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-38744986

RESUMO

G-protein-coupled receptors (GPCRs) are key regulators of human physiology and are the targets of many small-molecule research compounds and therapeutic drugs. While most of these ligands bind to their target GPCR with high affinity, selectivity is often limited at the receptor, tissue and cellular levels. Antibodies have the potential to address these limitations but their properties as GPCR ligands remain poorly characterized. Here, using protein engineering, pharmacological assays and structural studies, we develop maternally selective heavy-chain-only antibody ('nanobody') antagonists against the angiotensin II type I receptor and uncover the unusual molecular basis of their receptor antagonism. We further show that our nanobodies can simultaneously bind to angiotensin II type I receptor with specific small-molecule antagonists and demonstrate that ligand selectivity can be readily tuned. Our work illustrates that antibody fragments can exhibit rich and evolvable pharmacology, attesting to their potential as next-generation GPCR modulators.

2.
bioRxiv ; 2023 Aug 24.
Artigo em Inglês | MEDLINE | ID: mdl-37662341

RESUMO

G protein-coupled receptors (GPCRs) are key regulators of human physiology and are the targets of many small molecule research compounds and therapeutic drugs. While most of these ligands bind to their target GPCR with high affinity, selectivity is often limited at the receptor, tissue, and cellular level. Antibodies have the potential to address these limitations but their properties as GPCR ligands remain poorly characterized. Here, using protein engineering, pharmacological assays, and structural studies, we develop maternally selective heavy chain-only antibody ("nanobody") antagonists against the angiotensin II type I receptor (AT1R) and uncover the unusual molecular basis of their receptor antagonism. We further show that our nanobodies can simultaneously bind to AT1R with specific small-molecule antagonists and demonstrate that ligand selectivity can be readily tuned. Our work illustrates that antibody fragments can exhibit rich and evolvable pharmacology, attesting to their potential as next-generation GPCR modulators.

3.
Mol Pharmacol ; 100(5): 513-525, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34580163

RESUMO

Among ß-blockers that are clinically prescribed for heart failure, carvedilol is a first-choice agent with unique pharmacological properties. Carvedilol is distinct from other ß-blockers in its ability to elicit ß-arrestin-biased agonism, which has been suggested to underlie its cardioprotective effects. Augmenting the pharmacologic properties of carvedilol thus holds the promise of developing more efficacious and/or biased ß-blockers. We recently identified compound-6 (cmpd-6), the first small molecule positive allosteric modulator of the ß2-adrenergic receptor (ß2AR). Cmpd-6 is positively cooperative with orthosteric agonists at the ß2AR and enhances agonist-mediated transducer (G-protein and ß-arrestin) signaling in an unbiased manner. Here, we report that cmpd-6, quite unexpectedly, displays strong positive cooperativity only with carvedilol among a panel of structurally diverse ß-blockers. Cmpd-6 enhances the binding affinity of carvedilol for the ß2AR and augments its ability to competitively antagonize agonist-induced cAMP generation. Cmpd-6 potentiates ß-arrestin1- but not Gs-protein-mediated high-affinity binding of carvedilol at the ß2AR and ß-arrestin-mediated cellular functions in response to carvedilol including extracellular signal-regulated kinase phosphorylation, receptor endocytosis, and trafficking into lysosomes. Importantly, an analog of cmpd-6 that selectively retains positive cooperativity with carvedilol acts as a negative modulator of agonist-stimulated ß2AR signaling. These unprecedented cooperative properties of carvedilol and cmpd-6 have implications for fundamental understanding of G-protein-coupled receptor (GPCR) allosteric modulation, as well as for the development of more effective biased beta blockers and other GPCR therapeutics. SIGNIFICANCE STATEMENT: This study reports on the small molecule-mediated allosteric modulation of the ß-arrestin-biased ß-blocker, carvedilol. The small molecule, compound-6 (cmpd-6), displays an exclusive positive cooperativity with carvedilol among other ß-blockers and enhances the binding affinity of carvedilol for the ß2-adrenergic receptor. Cooperative effects of cmpd-6 augment the ß-blockade property of carvedilol while potentiating its ß-arrestin-mediated signaling functions. These findings have potential implications in advancing G-protein-coupled receptor allostery, developing biased therapeutics and remedying cardiovascular ailments.


Assuntos
Antagonistas Adrenérgicos beta/farmacologia , Carvedilol/farmacologia , Receptores Adrenérgicos beta 2 , beta-Arrestinas/farmacologia , Antagonistas Adrenérgicos beta/química , Antagonistas Adrenérgicos beta/metabolismo , Regulação Alostérica/efeitos dos fármacos , Regulação Alostérica/fisiologia , Animais , Carvedilol/química , Carvedilol/metabolismo , Relação Dose-Resposta a Droga , Células HEK293 , Humanos , Receptores Adrenérgicos beta 2/metabolismo , Células Sf9 , beta-Arrestinas/química , beta-Arrestinas/metabolismo
4.
Science ; 371(6534)2021 03 12.
Artigo em Inglês | MEDLINE | ID: mdl-33479120

RESUMO

Heterotrimeric guanine nucleotide-binding protein (G protein)-coupled receptors (GPCRs) are common drug targets and canonically couple to specific Gα protein subtypes and ß-arrestin adaptor proteins. G protein-mediated signaling and ß-arrestin-mediated signaling have been considered separable. We show here that GPCRs promote a direct interaction between Gαi protein subtype family members and ß-arrestins regardless of their canonical Gα protein subtype coupling. Gαi:ß-arrestin complexes bound extracellular signal-regulated kinase (ERK), and their disruption impaired both ERK activation and cell migration, which is consistent with ß-arrestins requiring a functional interaction with Gαi for certain signaling events. These results introduce a GPCR signaling mechanism distinct from canonical G protein activation in which GPCRs cause the formation of Gαi:ß-arrestin signaling complexes.


Assuntos
Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , beta-Arrestinas/metabolismo , Técnicas de Transferência de Energia por Ressonância de Bioluminescência , Movimento Celular , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Células HEK293 , Humanos , Transdução de Sinais
5.
J Biol Chem ; 295(49): 16773-16784, 2020 12 04.
Artigo em Inglês | MEDLINE | ID: mdl-32978252

RESUMO

G protein-coupled receptors (GPCRs) initiate signaling cascades via G-proteins and beta-arrestins (ßarr). ßarr-dependent actions begin with recruitment of ßarr to the phosphorylated receptor tail and are followed by engagement with the receptor core. ßarrs are known to act as adaptor proteins binding receptors and various effectors, but it is unclear whether in addition to the scaffolding role ßarrs can allosterically activate their downstream targets. Here we demonstrate the direct allosteric activation of proto-oncogene kinase Src by GPCR-ßarr complexes in vitro and establish the conformational basis of the activation. Whereas free ßarr1 had no effect on Src activity, ßarr1 in complex with M2 muscarinic or ß2-adrenergic receptors reconstituted in lipid nanodiscs activate Src by reducing the lag phase in Src autophosphorylation. Interestingly, receptor-ßarr1 complexes formed with a ßarr1 mutant, in which the finger-loop, required to interact with the receptor core, has been deleted, fully retain the ability to activate Src. Similarly, ßarr1 in complex with only a phosphorylated C-terminal tail of the vasopressin 2 receptor activates Src as efficiently as GPCR-ßarr complexes. In contrast, ßarr1 and chimeric M2 receptor with nonphosphorylated C-terminal tail failed to activate Src. Taken together, these data demonstrate that the phosphorylated GPCR tail interaction with ßarr1 is necessary and sufficient to empower it to allosterically activate Src. Our findings may have implications for understanding more broadly the mechanisms of allosteric activation of downstream targets by ßarrs.


Assuntos
Receptor Muscarínico M2/metabolismo , Receptores Adrenérgicos beta 2/metabolismo , Receptores de Vasopressinas/metabolismo , beta-Arrestina 1/metabolismo , Quinases da Família src/metabolismo , Regulação Alostérica , Ativação Enzimática , Humanos , Cinética , Mutagênese Sítio-Dirigida , Nanoestruturas/química , Peptídeos/síntese química , Peptídeos/química , Fosforilação , Ligação Proteica , Proto-Oncogene Mas , Receptor Muscarínico M2/química , Receptores Adrenérgicos beta 2/química , Receptores de Vasopressinas/química , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/química , Proteínas Recombinantes/isolamento & purificação , Especificidade por Substrato , beta-Arrestina 1/química , beta-Arrestina 1/genética , Domínios de Homologia de src , Quinases da Família src/química
6.
Proc Natl Acad Sci U S A ; 117(33): 20284-20291, 2020 08 18.
Artigo em Inglês | MEDLINE | ID: mdl-32753386

RESUMO

There is considerable interest in developing antibodies as functional modulators of G protein-coupled receptor (GPCR) signaling for both therapeutic and research applications. However, there are few antibody ligands targeting GPCRs outside of the chemokine receptor group. GPCRs are challenging targets for conventional antibody discovery methods, as many are highly conserved across species, are biochemically unstable upon purification, and possess deeply buried ligand-binding sites. Here, we describe a selection methodology to enrich for functionally modulatory antibodies using a yeast-displayed library of synthetic camelid antibody fragments called "nanobodies." Using this platform, we discovered multiple nanobodies that act as antagonists of the angiotensin II type 1 receptor (AT1R). Following angiotensin II infusion in mice, we found that an affinity matured nanobody antagonist has comparable antihypertensive activity to the angiotensin receptor blocker (ARB) losartan. The unique pharmacology and restricted biodistribution of nanobody antagonists may provide a path for treating hypertensive disorders when small-molecule drugs targeting the AT1R are contraindicated, for example, in pregnancy.


Assuntos
Antagonistas de Receptores de Angiotensina , Receptores de Angiotensina/imunologia , Anticorpos de Domínio Único , Animais , Afinidade de Anticorpos , Pressão Sanguínea , Linhagem Celular , Humanos , Camundongos
7.
Science ; 367(6480): 881-887, 2020 02 21.
Artigo em Inglês | MEDLINE | ID: mdl-32079767

RESUMO

Biased signaling, in which different ligands that bind to the same G protein-coupled receptor preferentially trigger distinct signaling pathways, holds great promise for the design of safer and more effective drugs. Its structural mechanism remains unclear, however, hampering efforts to design drugs with desired signaling profiles. Here, we use extensive atomic-level molecular dynamics simulations to determine how arrestin bias and G protein bias arise at the angiotensin II type 1 receptor. The receptor adopts two major signaling conformations, one of which couples almost exclusively to arrestin, whereas the other also couples effectively to a G protein. A long-range allosteric network allows ligands in the extracellular binding pocket to favor either of the two intracellular conformations. Guided by this computationally determined mechanism, we designed ligands with desired signaling profiles.


Assuntos
Arrestinas/química , Proteínas de Ligação ao GTP/química , Receptor Tipo 1 de Angiotensina/química , Transdução de Sinais , Regulação Alostérica , Humanos , Simulação de Dinâmica Molecular , Conformação Proteica
8.
Science ; 367(6480): 888-892, 2020 02 21.
Artigo em Inglês | MEDLINE | ID: mdl-32079768

RESUMO

Biased agonists of G protein-coupled receptors (GPCRs) preferentially activate a subset of downstream signaling pathways. In this work, we present crystal structures of angiotensin II type 1 receptor (AT1R) (2.7 to 2.9 angstroms) bound to three ligands with divergent bias profiles: the balanced endogenous agonist angiotensin II (AngII) and two strongly ß-arrestin-biased analogs. Compared with other ligands, AngII promotes more-substantial rearrangements not only at the bottom of the ligand-binding pocket but also in a key polar network in the receptor core, which forms a sodium-binding site in most GPCRs. Divergences from the family consensus in this region, which appears to act as a biased signaling switch, may predispose the AT1R and certain other GPCRs (such as chemokine receptors) to adopt conformations that are capable of activating ß-arrestin but not heterotrimeric Gq protein signaling.


Assuntos
Angiotensina II/química , Receptor Tipo 1 de Angiotensina/química , Humanos , Ligantes , Conformação Proteica , Transdução de Sinais , beta-Arrestinas/química
9.
Nature ; 579(7798): 297-302, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31945772

RESUMO

After activation by an agonist, G-protein-coupled receptors (GPCRs) recruit ß-arrestin, which desensitizes heterotrimeric G-protein signalling and promotes receptor endocytosis1. Additionally, ß-arrestin directly regulates many cell signalling pathways that can induce cellular responses distinct from that of G proteins2. In contrast to G proteins, for which there are many high-resolution structures in complex with GPCRs, the molecular mechanisms underlying the interaction of ß-arrestin with GPCRs are much less understood. Here we present a cryo-electron microscopy structure of ß-arrestin 1 (ßarr1) in complex with M2 muscarinic receptor (M2R) reconstituted in lipid nanodiscs. The M2R-ßarr1 complex displays a multimodal network of flexible interactions, including binding of the N domain of ßarr1 to phosphorylated receptor residues and insertion of the finger loop of ßarr1 into the M2R seven-transmembrane bundle, which adopts a conformation similar to that in the M2R-heterotrimeric Go protein complex3. Moreover, the cryo-electron microscopy map reveals that the C-edge of ßarr1 engages the lipid bilayer. Through atomistic simulations and biophysical, biochemical and cellular assays, we show that the C-edge is critical for stable complex formation, ßarr1 recruitment, receptor internalization, and desensitization of G-protein activation. Taken together, these data suggest that the cooperative interactions of ß-arrestin with both the receptor and the phospholipid bilayer contribute to its functional versatility.


Assuntos
Lipídeos/química , Modelos Moleculares , beta-Arrestinas/química , Linhagem Celular , Simulação por Computador , Microscopia Crioeletrônica , Humanos , Nanoestruturas/química , Estrutura Terciária de Proteína
10.
J Biol Chem ; 294(36): 13218-13223, 2019 09 06.
Artigo em Inglês | MEDLINE | ID: mdl-31362983

RESUMO

A hallmark of G-protein-coupled receptors (GPCRs) is the conversion of external stimuli into specific cellular responses. In this tightly-regulated process, extracellular ligand binding by GPCRs promotes specific conformational changes within the seven transmembrane helices, leading to the coupling and activation of intracellular "transducer" proteins, such as heterotrimeric G proteins. Much of our understanding of the molecular mechanisms that govern GPCR activation is derived from experiments with purified receptors reconstituted in detergent micelles. To elucidate the influence of the phospholipid bilayer on GPCR activation, here we interrogated the functional, pharmacological, and biophysical properties of a GPCR, the ß2-adrenergic receptor (ß2AR), in high-density lipoprotein (HDL) particles. Compared with detergent-reconstituted ß2AR, the ß2AR in HDL particles had greatly enhanced levels of basal (constitutive) activity and displayed increased sensitivity to agonist activation, as assessed by activation of heterotrimeric G protein and allosteric coupling between the ligand-binding and transducer-binding pockets. Using 19F NMR spectroscopy, we directly linked these functional differences in detergent- and HDL-reconstituted ß2AR to a change in the equilibrium between inactive and active receptor states. The contrast between the low levels of ß2AR constitutive activity in cells and the high constitutive activity observed in an isolated phospholipid bilayer indicates that ß2AR basal activity depends on the reconstitution system and further suggests that various cellular mechanisms suppress ß2AR basal activity physiologically. Our findings provide critical additional insights into GPCR activation and reveal how dramatically reconstitution systems can impact membrane protein function.


Assuntos
Detergentes/farmacologia , Fosfolipídeos/metabolismo , Receptores Adrenérgicos beta 2/metabolismo , Humanos
11.
Science ; 364(6447): 1283-1287, 2019 06 28.
Artigo em Inglês | MEDLINE | ID: mdl-31249059

RESUMO

Drugs targeting the orthosteric, primary binding site of G protein-coupled receptors are the most common therapeutics. Allosteric binding sites, elsewhere on the receptors, are less well-defined, and so less exploited clinically. We report the crystal structure of the prototypic ß2-adrenergic receptor in complex with an orthosteric agonist and compound-6FA, a positive allosteric modulator of this receptor. It binds on the receptor's inner surface in a pocket created by intracellular loop 2 and transmembrane segments 3 and 4, stabilizing the loop in an α-helical conformation required to engage the G protein. Structural comparison explains the selectivity of the compound for ß2- over the ß1-adrenergic receptor. Diversity in location, mechanism, and selectivity of allosteric ligands provides potential to expand the range of receptor drugs.


Assuntos
Agonistas de Receptores Adrenérgicos beta 2/química , Anidridos Ftálicos/química , Receptores Adrenérgicos beta 2/química , Agonistas de Receptores Adrenérgicos beta 2/farmacologia , Regulação Alostérica , Cristalografia por Raios X , Mutação com Ganho de Função , Humanos , Anidridos Ftálicos/farmacologia , Receptores Adrenérgicos beta 2/genética
12.
Cell ; 176(3): 468-478.e11, 2019 01 24.
Artigo em Inglês | MEDLINE | ID: mdl-30639099

RESUMO

"Biased" G protein-coupled receptor (GPCR) agonists preferentially activate pathways mediated by G proteins or ß-arrestins. Here, we use double electron-electron resonance spectroscopy to probe the changes that ligands induce in the conformational distribution of the angiotensin II type I receptor. Monitoring distances between 10 pairs of nitroxide labels distributed across the intracellular regions enabled mapping of four underlying sets of conformations. Ligands from different functional classes have distinct, characteristic effects on the conformational heterogeneity of the receptor. Compared to angiotensin II, the endogenous agonist, agonists with enhanced Gq coupling more strongly stabilize an "open" conformation with an accessible transducer-binding site. ß-arrestin-biased agonists deficient in Gq coupling do not stabilize this open conformation but instead favor two more occluded conformations. These data suggest a structural mechanism for biased ligand action at the angiotensin receptor that can be exploited to rationally design GPCR-targeting drugs with greater specificity of action.


Assuntos
Angiotensinas/metabolismo , Receptor Tipo 1 de Angiotensina/metabolismo , Bloqueadores do Receptor Tipo 1 de Angiotensina II/farmacologia , Antagonistas de Receptores de Angiotensina/metabolismo , Arrestinas/metabolismo , Linhagem Celular , Humanos , Ligantes , Conformação Proteica , Receptores de Angiotensina/metabolismo , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais , Espectroscopia de Perda de Energia de Elétrons/métodos , beta-Arrestinas/metabolismo
13.
Cell ; 176(3): 479-490.e12, 2019 01 24.
Artigo em Inglês | MEDLINE | ID: mdl-30639100

RESUMO

The angiotensin II (AngII) type 1 receptor (AT1R) is a critical regulator of cardiovascular and renal function and is an important model for studies of G-protein-coupled receptor (GPCR) signaling. By stabilizing the receptor with a single-domain antibody fragment ("nanobody") discovered using a synthetic yeast-displayed library, we determined the crystal structure of active-state human AT1R bound to an AngII analog with partial agonist activity. The nanobody binds to the receptor's intracellular transducer pocket, stabilizing the large conformational changes characteristic of activated GPCRs. The peptide engages the AT1R through an extensive interface spanning from the receptor core to its extracellular face and N terminus, remodeling the ligand-binding cavity. Remarkably, the mechanism used to propagate conformational changes through the receptor diverges from other GPCRs at several key sites, highlighting the diversity of allosteric mechanisms among GPCRs. Our structure provides insight into how AngII and its analogs stimulate full or biased signaling, respectively.


Assuntos
Receptor Tipo 1 de Angiotensina/metabolismo , Anticorpos de Domínio Único/farmacologia , Angiotensina II , Bloqueadores do Receptor Tipo 1 de Angiotensina II/metabolismo , Arrestinas/metabolismo , Células HEK293 , Humanos , Fragmentos de Imunoglobulinas/farmacologia , Conformação Proteica , Proto-Oncogene Mas , Proteínas Proto-Oncogênicas , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais/efeitos dos fármacos , Anticorpos de Domínio Único/metabolismo , beta-Arrestinas/metabolismo
14.
Sci Signal ; 11(544)2018 08 21.
Artigo em Inglês | MEDLINE | ID: mdl-30131371

RESUMO

Biased agonists of G protein-coupled receptors (GPCRs), which selectively activate either G protein- or ß-arrestin-mediated signaling pathways, are of major therapeutic interest because they have the potential to show improved efficacy and specificity as drugs. Efforts to understand the mechanistic basis of this phenomenon have focused on the hypothesis that G proteins and ß-arrestins preferentially couple to distinct GPCR conformations. However, because GPCR kinase (GRK)-dependent receptor phosphorylation is a critical prerequisite for the recruitment of ß-arrestins to most GPCRs, GRKs themselves may play an important role in establishing biased signaling. We showed that an alanine mutant of the highly conserved residue tyrosine 219 (Y219A) in transmembrane domain five of the ß2-adrenergic receptor (ß2AR) was incapable of ß-arrestin recruitment, receptor internalization, and ß-arrestin-mediated activation of extracellular signal-regulated kinase (ERK), whereas it retained the ability to signal through G protein. We found that the impaired ß-arrestin recruitment in cells was due to reduced GRK-mediated phosphorylation of the ß2AR Y219A C terminus, which was recapitulated in vitro with purified components. Furthermore, in vitro ligation of a synthetically phosphorylated peptide onto the C terminus of ß2AR Y219A rescued both the initial recruitment of ß-arrestin and its engagement with the intracellular core of the receptor. These data suggest that the Y219A mutation generates a G protein-biased state primarily by conformational selection against GRK coupling, rather than against ß-arrestin. Together, these findings highlight the importance of GRKs in modulating the biased agonism of GPCRs.


Assuntos
Quinases de Receptores Acoplados a Proteína G/metabolismo , Mutação , Receptores Adrenérgicos beta 2/metabolismo , Transdução de Sinais , Linhagem Celular Tumoral , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Proteínas de Ligação ao GTP/metabolismo , Células HEK293 , Humanos , Fosforilação , Receptores Adrenérgicos beta 2/genética , Receptores Acoplados a Proteínas G/metabolismo , beta-Arrestinas/metabolismo
15.
Mol Pharmacol ; 94(2): 850-861, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29769246

RESUMO

Conventional drug discovery efforts at the ß2-adrenoceptor (ß2AR) have led to the development of ligands that bind almost exclusively to the receptor's hormone-binding orthosteric site. However, targeting the largely unexplored and evolutionarily unique allosteric sites has potential for developing more specific drugs with fewer side effects than orthosteric ligands. Using our recently developed approach for screening G protein-coupled receptors (GPCRs) with DNA-encoded small-molecule libraries, we have discovered and characterized the first ß2AR small-molecule positive allosteric modulators (PAMs)-compound (Cmpd)-6 [(R)-N-(4-amino-1-(4-(tert-butyl)phenyl)-4-oxobutan-2-yl)-5-(N-isopropyl-N-methylsulfamoyl)-2-((4-methoxyphenyl)thio)benzamide] and its analogs. We used purified human ß2ARs, occupied by a high-affinity agonist, for the affinity-based screening of over 500 million distinct library compounds, which yielded Cmpd-6. It exhibits a low micro-molar affinity for the agonist-occupied ß2AR and displays positive cooperativity with orthosteric agonists, thereby enhancing their binding to the receptor and ability to stabilize its active state. Cmpd-6 is cooperative with G protein and ß-arrestin1 (a.k.a. arrestin2) to stabilize high-affinity, agonist-bound active states of the ß2AR and potentiates downstream cAMP production and receptor recruitment of ß-arrestin2 (a.k.a. arrestin3). Cmpd-6 is specific for the ß2AR compared with the closely related ß1AR. Structure-activity studies of select Cmpd-6 analogs defined the chemical groups that are critical for its biologic activity. We thus introduce the first small-molecule PAMs for the ß2AR, which may serve as a lead molecule for the development of novel therapeutics. The approach described in this work establishes a broadly applicable proof-of-concept strategy for affinity-based discovery of small-molecule allosteric compounds targeting unique conformational states of GPCRs.


Assuntos
Agonistas de Receptores Adrenérgicos beta 2/metabolismo , Receptores Adrenérgicos beta 2/química , Receptores Adrenérgicos beta 2/metabolismo , Bibliotecas de Moléculas Pequenas/farmacologia , Regulação Alostérica/efeitos dos fármacos , Sítio Alostérico/efeitos dos fármacos , Sinergismo Farmacológico , Proteínas de Ligação ao GTP/metabolismo , Biblioteca Gênica , Estrutura Molecular , Bibliotecas de Moléculas Pequenas/química , Relação Estrutura-Atividade , Especificidade por Substrato , beta-Arrestina 1/metabolismo
16.
Proc Natl Acad Sci U S A ; 115(15): 3834-3839, 2018 04 10.
Artigo em Inglês | MEDLINE | ID: mdl-29581292

RESUMO

The ability of G protein-coupled receptors (GPCRs) to initiate complex cascades of cellular signaling is governed by the sequential coupling of three main transducer proteins, G protein, GPCR kinase (GRK), and ß-arrestin. Mounting evidence indicates these transducers all have distinct conformational preferences and binding modes. However, interrogating each transducer's mechanism of interaction with GPCRs has been complicated by the interplay of transducer-mediated signaling events. For example, GRK-mediated receptor phosphorylation recruits and induces conformational changes in ß-arrestin, which facilitates coupling to the GPCR transmembrane core. Here we compare the allosteric interactions of G proteins and ß-arrestins with GPCRs' transmembrane cores by using the enzyme sortase to ligate a synthetic phosphorylated peptide onto the carboxyl terminus of three different receptors. Phosphopeptide ligation onto the ß2-adrenergic receptor (ß2AR) allows stabilization of a high-affinity receptor active state by ß-arrestin1, permitting us to define elements in the ß2AR and ß-arrestin1 that contribute to the receptor transmembrane core interaction. Interestingly, ligation of the identical phosphopeptide onto the ß2AR, the muscarinic acetylcholine receptor 2 and the µ-opioid receptor reveals that the ability of ß-arrestin1 to enhance agonist binding relative to G protein differs substantially among receptors. Furthermore, strong allosteric coupling of ß-arrestin1 correlates with its ability to attenuate, or "desensitize," G protein activation in vitro. Sortase ligation thus provides a versatile method to introduce complex, defined phosphorylation patterns into GPCRs, and analogous strategies could be applied to other classes of posttranslationally modified proteins. These homogeneously phosphorylated GPCRs provide an innovative means to systematically study receptor-transducer interactions.


Assuntos
Aminoaciltransferases/metabolismo , Proteínas de Bactérias/metabolismo , Cisteína Endopeptidases/metabolismo , Receptor Muscarínico M2/metabolismo , Receptores Adrenérgicos beta 2/metabolismo , Receptores Opioides mu/metabolismo , beta-Arrestina 1/metabolismo , Regulação Alostérica , Humanos , Fosforilação , Receptor Muscarínico M2/genética , Receptores Adrenérgicos beta 2/genética , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Receptores Opioides mu/genética , beta-Arrestina 1/genética
17.
Nat Commun ; 8(1): 1706, 2017 11 22.
Artigo em Inglês | MEDLINE | ID: mdl-29167435

RESUMO

The ß1 adrenergic receptor (ß1AR) is recognized as a classical Gαs-coupled receptor. Agonist binding not only initiates G protein-mediated signaling but also signaling through the multifunctional adapter protein ß-arrestin. Some ßAR ligands, such as carvedilol, stimulate ßAR signaling preferentially through ß-arrestin, a concept known as ß-arrestin-biased agonism. Here, we identify a signaling mechanism, unlike that previously known for any Gαs-coupled receptor, whereby carvedilol induces the transition of the ß1AR from a classical Gαs-coupled receptor to a Gαi-coupled receptor stabilizing a distinct receptor conformation to initiate ß-arrestin-mediated signaling. Recruitment of Gαi is not induced by any other ßAR ligand screened, nor is it required for ß-arrestin-bias activated by the ß2AR subtype of the ßAR family. Our findings demonstrate a previously unrecognized role for Gαi in ß1AR signaling and suggest that the concept of ß-arrestin-bias may need to be refined to incorporate the selective bias of receptors towards distinct G protein subtypes.


Assuntos
Carbazóis/farmacologia , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/metabolismo , Propanolaminas/farmacologia , Receptores Adrenérgicos beta 1/metabolismo , beta-Arrestinas/metabolismo , Antagonistas de Receptores Adrenérgicos alfa 1/farmacologia , Antagonistas Adrenérgicos beta/farmacologia , Animais , Carvedilol , Feminino , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/deficiência , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/genética , Técnicas de Silenciamento de Genes , Células HEK293 , Humanos , Ligantes , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Masculino , Camundongos , Camundongos Knockout , Conformação Proteica/efeitos dos fármacos , Receptores Adrenérgicos beta 1/química , beta-Arrestinas/antagonistas & inibidores , beta-Arrestinas/genética
18.
Cell ; 169(2): 338-349.e11, 2017 04 06.
Artigo em Inglês | MEDLINE | ID: mdl-28388415

RESUMO

G-protein-coupled receptors (GPCRs) play critical roles in regulating physiological processes ranging from neurotransmission to cardiovascular function. Current methods for tracking GPCR signaling suffer from low throughput, modification or overexpression of effector proteins, and low temporal resolution. Here, we show that peroxidase-catalyzed proximity labeling can be combined with isobaric tagging and mass spectrometry to enable quantitative, time-resolved measurement of GPCR agonist response in living cells. Using this technique, termed "GPCR-APEX," we track activation and internalization of the angiotensin II type 1 receptor and the ß2 adrenoceptor. These receptors co-localize with a variety of G proteins even before receptor activation, and activated receptors are largely sequestered from G proteins upon internalization. Additionally, the two receptors show differing internalization kinetics, and we identify the membrane protein LMBRD2 as a potential regulator of ß2 adrenoceptor signaling, underscoring the value of a dynamic view of receptor function.


Assuntos
Ascorbato Peroxidases/química , Receptor Tipo 1 de Angiotensina/análise , Receptor Tipo 1 de Angiotensina/metabolismo , Transdução de Sinais , Coloração e Rotulagem/métodos , Ascorbato Peroxidases/metabolismo , Biotina/química , Proteínas de Ligação ao GTP/análise , Células HEK293 , Humanos , Oligopeptídeos/farmacologia , Engenharia de Proteínas , Receptor Tipo 1 de Angiotensina/agonistas , beta-Arrestinas/química
19.
Proc Natl Acad Sci U S A ; 114(7): 1708-1713, 2017 02 14.
Artigo em Inglês | MEDLINE | ID: mdl-28130548

RESUMO

The ß2-adrenergic receptor (ß2AR) has been a model system for understanding regulatory mechanisms of G-protein-coupled receptor (GPCR) actions and plays a significant role in cardiovascular and pulmonary diseases. Because all known ß-adrenergic receptor drugs target the orthosteric binding site of the receptor, we set out to isolate allosteric ligands for this receptor by panning DNA-encoded small-molecule libraries comprising 190 million distinct compounds against purified human ß2AR. Here, we report the discovery of a small-molecule negative allosteric modulator (antagonist), compound 15 [([4-((2S)-3-(((S)-3-(3-bromophenyl)-1-(methylamino)-1-oxopropan-2-yl)amino)-2-(2-cyclohexyl-2-phenylacetamido)-3-oxopropyl)benzamide], exhibiting a unique chemotype and low micromolar affinity for the ß2AR. Binding of 15 to the receptor cooperatively enhances orthosteric inverse agonist binding while negatively modulating binding of orthosteric agonists. Studies with a specific antibody that binds to an intracellular region of the ß2AR suggest that 15 binds in proximity to the G-protein binding site on the cytosolic surface of the ß2AR. In cell-signaling studies, 15 inhibits cAMP production through the ß2AR, but not that mediated by other Gs-coupled receptors. Compound 15 also similarly inhibits ß-arrestin recruitment to the activated ß2AR. This study presents an allosteric small-molecule ligand for the ß2AR and introduces a broadly applicable method for screening DNA-encoded small-molecule libraries against purified GPCR targets. Importantly, such an approach could facilitate the discovery of GPCR drugs with tailored allosteric effects.


Assuntos
Antagonistas Adrenérgicos beta/farmacologia , Ensaios de Triagem em Larga Escala/métodos , Receptores Adrenérgicos beta 2/metabolismo , Bibliotecas de Moléculas Pequenas/farmacologia , Antagonistas Adrenérgicos beta/química , Antagonistas Adrenérgicos beta/metabolismo , Animais , Sítios de Ligação/genética , Ligação Competitiva/efeitos dos fármacos , DNA/genética , Humanos , Ligantes , Estrutura Molecular , Mutação , Receptores Adrenérgicos beta 2/genética , Células Sf9 , Bibliotecas de Moléculas Pequenas/química , Bibliotecas de Moléculas Pequenas/metabolismo , Spodoptera
20.
Nature ; 535(7612): 448-52, 2016 07 21.
Artigo em Inglês | MEDLINE | ID: mdl-27409812

RESUMO

G-protein-coupled receptors (GPCRs) modulate many physiological processes by transducing a variety of extracellular cues into intracellular responses. Ligand binding to an extracellular orthosteric pocket propagates conformational change to the receptor cytosolic region to promote binding and activation of downstream signalling effectors such as G proteins and ß-arrestins. It is well known that different agonists can share the same binding pocket but evoke unique receptor conformations leading to a wide range of downstream responses ('efficacy'). Furthermore, increasing biophysical evidence, primarily using the ß2-adrenergic receptor (ß2AR) as a model system, supports the existence of multiple active and inactive conformational states. However, how agonists with varying efficacy modulate these receptor states to initiate cellular responses is not well understood. Here we report stabilization of two distinct ß2AR conformations using single domain camelid antibodies (nanobodies)­a previously described positive allosteric nanobody (Nb80) and a newly identified negative allosteric nanobody (Nb60). We show that Nb60 stabilizes a previously unappreciated low-affinity receptor state which corresponds to one of two inactive receptor conformations as delineated by X-ray crystallography and NMR spectroscopy. We find that the agonist isoprenaline has a 15,000-fold higher affinity for ß2AR in the presence of Nb80 compared to the affinity of isoprenaline for ß2AR in the presence of Nb60, highlighting the full allosteric range of a GPCR. Assessing the binding of 17 ligands of varying efficacy to the ß2AR in the absence and presence of Nb60 or Nb80 reveals large ligand-specific effects that can only be explained using an allosteric model which assumes equilibrium amongst at least three receptor states. Agonists generally exert efficacy by stabilizing the active Nb80-stabilized receptor state (R80). In contrast, for a number of partial agonists, both stabilization of R80 and destabilization of the inactive, Nb60-bound state (R60) contribute to their ability to modulate receptor activation. These data demonstrate that ligands can initiate a wide range of cellular responses by differentially stabilizing multiple receptor states.


Assuntos
Agonistas de Receptores Adrenérgicos beta 2/farmacologia , Receptores Adrenérgicos beta 2/química , Receptores Adrenérgicos beta 2/metabolismo , Anticorpos de Domínio Único/farmacologia , Regulação Alostérica/efeitos dos fármacos , Sítio Alostérico/efeitos dos fármacos , Cristalografia por Raios X , Agonismo Parcial de Drogas , Humanos , Isoproterenol/farmacologia , Ligantes , Modelos Moleculares , Ressonância Magnética Nuclear Biomolecular , Conformação Proteica/efeitos dos fármacos , Estabilidade Proteica/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...