Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Science ; 383(6678): 101-108, 2024 01 05.
Artigo em Inglês | MEDLINE | ID: mdl-38175886

RESUMO

ß-arrestins (ßarrs) are multifunctional proteins involved in signaling and regulation of seven transmembrane receptors (7TMRs), and their interaction is driven primarily by agonist-induced receptor activation and phosphorylation. Here, we present seven cryo-electron microscopy structures of ßarrs either in the basal state, activated by the muscarinic receptor subtype 2 (M2R) through its third intracellular loop, or activated by the ßarr-biased decoy D6 receptor (D6R). Combined with biochemical, cellular, and biophysical experiments, these structural snapshots allow the visualization of atypical engagement of ßarrs with 7TMRs and also reveal a structural transition in the carboxyl terminus of ßarr2 from a ß strand to an α helix upon activation by D6R. Our study provides previously unanticipated molecular insights into the structural and functional diversity encoded in 7TMR-ßarr complexes with direct implications for exploring novel therapeutic avenues.


Assuntos
Domínios e Motivos de Interação entre Proteínas , Receptores Acoplados a Proteínas G , beta-Arrestinas , beta-Arrestinas/química , Microscopia Crioeletrônica , Receptores Acoplados a Proteínas G/química , Transdução de Sinais , Conformação Proteica em Folha beta , Conformação Proteica em alfa-Hélice , Humanos
2.
Trends Pharmacol Sci ; 44(1): 1-3, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36057461

RESUMO

Atypical chemokine receptor subtype 3 (ACKR3), a chemokine receptor, couples selectively to ß-arrestins (ßarrs) but not to G proteins despite having seven transmembrane (7TM) helix architecture. Yen et al. present cryogenic-electron microscopy (cryo-EM) structures of agonist-bound ACKR3, elucidating a distinct chemokine-binding mechanism, and offering a structural template to probe the transducer-coupling bias at this receptor.


Assuntos
Receptores CXCR , Transdução de Sinais , beta-Arrestinas , Humanos , beta-Arrestinas/química , Ligação Proteica , Receptores CXCR/química
3.
Science ; 377(6602): 222-228, 2022 07 08.
Artigo em Inglês | MEDLINE | ID: mdl-35857540

RESUMO

G protein-coupled receptors (GPCRs) recruit ß-arrestins to coordinate diverse cellular processes, but the structural dynamics driving this process are poorly understood. Atypical chemokine receptors (ACKRs) are intrinsically biased GPCRs that engage ß-arrestins but not G proteins, making them a model system for investigating the structural basis of ß-arrestin recruitment. Here, we performed nuclear magnetic resonance (NMR) experiments on 13CH3-ε-methionine-labeled ACKR3, revealing that ß-arrestin recruitment is associated with conformational exchange at key regions of the extracellular ligand-binding pocket and intracellular ß-arrestin-coupling region. NMR studies of ACKR3 mutants defective in ß-arrestin recruitment identified an allosteric hub in the receptor core that coordinates transitions among heterogeneously populated and selected conformational states. Our data suggest that conformational selection guides ß-arrestin recruitment by tuning receptor dynamics at intracellular and extracellular regions.


Assuntos
Receptores CXCR , beta-Arrestinas , Regulação Alostérica , Ligantes , Espectroscopia de Ressonância Magnética , Mutação , Ligação Proteica , Conformação Proteica , Receptores CXCR/química , Receptores CXCR/genética , beta-Arrestinas/química
4.
Curr Opin Struct Biol ; 75: 102406, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35738165

RESUMO

Agonist-induced recruitment of ß-arrestins (ßarrs) to G protein-coupled receptors (GPCRs) plays a central role in regulating the spatio-temporal aspects of GPCR signaling. Several recent studies have provided novel structural and functional insights into our understanding of GPCR-ßarr interaction, subsequent ßarr activation and resulting functional outcomes. In this review, we discuss these recent advances with a particular emphasis on recognition of receptor-bound phosphates by ßarrs, the emerging concept of spatial positioning of key phosphorylation sites, the conformational transition in ßarrs during partial to full-engagement, and structural differences driving functional outcomes of ßarr isoforms. We also highlight the key directions that require further investigation going forward to fully understand the structural mechanisms driving ßarr activation and functional responses.


Assuntos
Receptores Acoplados a Proteínas G , Transdução de Sinais , Fosforilação , Isoformas de Proteínas , Receptores Acoplados a Proteínas G/química , beta-Arrestinas/química , beta-Arrestinas/metabolismo
5.
Nucleic Acids Res ; 50(W1): W598-W610, 2022 07 05.
Artigo em Inglês | MEDLINE | ID: mdl-35639758

RESUMO

In this study we show that protein language models can encode structural and functional information of GPCR sequences that can be used to predict their signaling and functional repertoire. We used the ESM1b protein embeddings as features and the binding information known from publicly available studies to develop PRECOGx, a machine learning predictor to explore GPCR interactions with G protein and ß-arrestin, which we made available through a new webserver (https://precogx.bioinfolab.sns.it/). PRECOGx outperformed its predecessor (e.g. PRECOG) in predicting GPCR-transducer couplings, being also able to consider all GPCR classes. The webserver also provides new functionalities, such as the projection of input sequences on a low-dimensional space describing essential features of the human GPCRome, which is used as a reference to track GPCR variants. Additionally, it allows inspection of the sequence and structural determinants responsible for coupling via the analysis of the most important attention maps used by the models as well as through predicted intramolecular contacts. We demonstrate applications of PRECOGx by predicting the impact of disease variants (ClinVar) and alternative splice forms from healthy tissues (GTEX) of human GPCRs, revealing the power to dissect system biasing mechanisms in both health and disease.


Assuntos
Aprendizado de Máquina , Receptores Acoplados a Proteínas G , Transdução de Sinais , Software , Humanos , Receptores Acoplados a Proteínas G/química , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Internet , beta-Arrestinas/química , beta-Arrestinas/metabolismo , Proteínas Heterotriméricas de Ligação ao GTP/química , Proteínas Heterotriméricas de Ligação ao GTP/metabolismo , Computadores , Predisposição Genética para Doença/genética , Processamento Alternativo/genética
6.
Int J Mol Sci ; 23(2)2022 Jan 17.
Artigo em Inglês | MEDLINE | ID: mdl-35055186

RESUMO

ß-arrestins were initially identified to desensitize and internalize G-protein-coupled receptors (GPCRs). Receptor-bound ß-arrestins also initiate a second wave of signaling by scaffolding mitogen-activated protein kinase (MAPK) signaling components, MAPK kinase kinase, MAPK kinase, and MAPK. In particular, ß-arrestins facilitate ERK1/2 or JNK3 activation by scaffolding signal cascade components such as ERK1/2-MEK1-cRaf or JNK3-MKK4/7-ASK1. Understanding the precise molecular and structural mechanisms of ß-arrestin-mediated MAPK scaffolding assembly would deepen our understanding of GPCR-mediated MAPK activation and provide clues for the selective regulation of the MAPK signaling cascade for therapeutic purposes. Over the last decade, numerous research groups have attempted to understand the molecular and structural mechanisms of ß-arrestin-mediated MAPK scaffolding assembly. Although not providing the complete mechanism, these efforts suggest potential binding interfaces between ß-arrestins and MAPK signaling components and the mechanism for MAPK signal amplification by ß-arrestin-mediated scaffolding. This review summarizes recent developments of cellular and molecular works on the scaffolding mechanism of ß-arrestin for MAPK signaling cascade.


Assuntos
Sistema de Sinalização das MAP Quinases , Receptores Acoplados a Proteínas G/metabolismo , beta-Arrestinas/metabolismo , Animais , Humanos , Modelos Moleculares , Ligação Proteica , Conformação Proteica , Receptores Acoplados a Proteínas G/química , beta-Arrestinas/química
7.
Mol Pharmacol ; 101(2): 87-94, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34853152

RESUMO

G protein-coupled receptors (GPCRs) transduce a diverse variety of extracellular stimuli into intracellular signaling. These receptors are the most clinically productive drug targets at present. Despite decades of research on the signaling consequences of molecule-receptor interactions, conformational components of receptor-effector interactions remain incompletely described. The ß 2-adrenergic receptor (ß 2AR) is a prototypical and extensively studied GPCR that can provide insight into this aspect of GPCR signaling thanks to robust structural data and rich pharmacopeia. Using bioluminescence resonance energy transfer -based biosensors, second messenger assays, and biochemical techniques, we characterize the properties of ß 2AR-F193A. This single point mutation in extracellular loop 2 of the ß 2AR is sufficient to intrinsically bias the ß 2AR away from ß-arrestin interaction and demonstrates altered regulatory outcomes downstream of this functional selectivity. This study highlights the importance of extracellular control of intracellular response to stimuli and suggests a previously undescribed role for the extracellular loops of the receptor and the extracellular pocket formed by transmembrane domains 2, 3, and 7 in GPCR regulation that may contribute to biased signaling at GPCRs. SIGNIFICANCE STATEMENT: The role of extracellular G protein-coupled receptor (GPCR) domains in mediating intracellular interactions is poorly understood. We characterized the effects of extracellular loop mutations on agonist-promoted interactions of GPCRs with G protein and ß-arrestin. Our studies reveal that F193 in extracellular loop 2 in the ß2-adrenergic receptor mediates interactions with G protein and ß-arrestin with a biased loss of ß-arrestin binding. These results provide new insights on the role of the extracellular domain in differentially modulating intracellular interactions with GPCRs.


Assuntos
Líquido Extracelular/metabolismo , Fenilalanina/metabolismo , Receptores Adrenérgicos beta 2/metabolismo , beta-Arrestinas/metabolismo , Sequência de Aminoácidos , Relação Dose-Resposta a Droga , Técnicas de Inativação de Genes/métodos , Células HEK293 , Humanos , Fenilalanina/química , Fenilalanina/genética , Estrutura Secundária de Proteína , Receptores Adrenérgicos beta 2/química , Receptores Adrenérgicos beta 2/genética , beta-Arrestinas/química , beta-Arrestinas/genética
8.
Sci Rep ; 11(1): 23424, 2021 12 06.
Artigo em Inglês | MEDLINE | ID: mdl-34873237

RESUMO

Intrinsically-photosensitive retinal ganglion cells (ipRGCs) are non-rod/non-cone retinal photoreceptors expressing the visual pigment, melanopsin, to detect ambient irradiance for various non-image-forming visual functions. The M1-subtype, amongst the best studied, mediates primarily circadian photoentrainment and pupillary light reflex. Their intrinsic light responses are more prolonged than those of rods and cones even at the single-photon level, in accordance with the typically slower time course of non-image-forming vision. The short (OPN4S) and long (OPN4L) alternatively-spliced forms of melanopsin proteins are both present in M1-ipRGCs, but their functional difference is unclear. We have examined this point by genetically removing the Opn4 gene (Opn4-/-) in mouse and re-expressing either OPN4S or OPN4L singly in Opn4-/- mice by using adeno-associated virus, but found no obvious difference in their intrinsic dim-flash responses. Previous studies have indicated that two dominant slow steps in M1-ipRGC phototransduction dictate these cells' intrinsic dim-flash-response kinetics, with time constants (τ1 and τ2) at room temperature of ~ 2 s and ~ 20 s, respectively. Here we found that melanopsin inactivation by phosphorylation or by ß-arrestins may not be one of these two steps, because their genetic disruptions did not prolong the two time constants or affect the response waveform. Disruption of GAP (GTPase-Activating-Protein) activity on the effector enzyme, PLCß4, in M1-ipRGC phototransduction to slow down G-protein deactivation also did not prolong the response decay, but caused its rising phase to become slightly sigmoidal by giving rise to a third time constant, τ3, of ~ 2 s (room temperature). This last observation suggests that GAP-mediated G-protein deactivation does partake in the flash-response termination, although normally with a time constant too short to be visible in the response waveform.


Assuntos
Células Fotorreceptoras Retinianas Cones/metabolismo , Células Ganglionares da Retina/metabolismo , Animais , Ritmo Circadiano/fisiologia , Dependovirus , Injeções Intravítreas , Cinética , Luz , Transdução de Sinal Luminoso , Camundongos , Camundongos Transgênicos , Mutação , Neurociências , Fosforilação , Opsinas de Bastonetes/química , Transdução de Sinais , Visão Ocular , beta-Arrestinas/química
9.
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
10.
Mol Cell ; 81(22): 4605-4621.e11, 2021 11 18.
Artigo em Inglês | MEDLINE | ID: mdl-34582793

RESUMO

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.


Assuntos
Membrana Celular/metabolismo , Conformação Proteica , Transdução de Sinais , beta-Arrestinas/química , Animais , Proteínas de Ligação ao GTP/química , Células HEK293 , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fosforilação , Ligação Proteica , Domínios Proteicos , Estrutura Secundária de Proteína , Transporte Proteico , Receptor da Anafilatoxina C5a/metabolismo
11.
J Biol Chem ; 296: 100503, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33684444

RESUMO

G protein-coupled receptors (GPCRs) signal through activation of G proteins and subsequent modulation of downstream effectors. More recently, signaling mediated by ß-arrestin has also been implicated in important physiological functions. This has led to great interest in the identification of biased ligands that favor either G protein or ß-arrestin-signaling pathways. However, nearly all screening techniques for measuring ß-arrestin recruitment have required C-terminal receptor modifications that can in principle alter protein interactions and thus signaling. Here, we have developed a novel luminescence-based assay to measure ß-arrestin recruitment to the membrane or early endosomes by unmodified receptors. Our strategy uses NanoLuc, an engineered luciferase from Oplophorus gracilirostris (deep-sea shrimp) that is smaller and brighter than other well-established luciferases. Recently, several publications have explored functional NanoLuc split sites for use in complementation assays. We have identified a unique split site within NanoLuc and fused the corresponding N-terminal fragment to either a plasma membrane or early endosome tether and the C-terminal fragment to ß-arrestins, which form the basis for the MeNArC and EeNArC assays, respectively. Upon receptor activation, ß-arrestin is recruited to the membrane and subsequently internalized in an agonist concentration-dependent manner. This recruitment promotes complementation of the two NanoLuc fragments, thereby reconstituting functional NanoLuc, allowing for quantification of ß-arrestin recruitment with a single luminescence signal. Our assay avoids potential artifacts related to C-terminal receptor modification and has promise as a new generic assay for measuring ß-arrestin recruitment to diverse GPCR types in heterologous or native cells.


Assuntos
Membrana Celular/metabolismo , Luciferases/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , beta-Arrestinas/metabolismo , Bioensaio/métodos , Células Cultivadas , Humanos , Ligantes , Ligação Proteica , Transdução de Sinais , beta-Arrestinas/química
12.
Biochim Biophys Acta Proteins Proteom ; 1869(4): 140603, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33421644

RESUMO

ß-arrestins bind active G protein-coupled receptors (GPCRs) and play a crucial role in receptor desensitization and internalization. The classical paradigm of arrestin function has been expanded with the identification of many non-receptor-binding partners, which indicated the multifunctional role of ß-arrestins in cellular functions. To elucidate the molecular mechanism of ß-arrestin-mediated signaling, the structural features of ß-arrestins were investigated using X-ray crystallography and cryogenic electron microscopy (cryo-EM). However, the intrinsic conformational flexibility of ß-arrestins hampers the elucidation of structural interactions between ß-arrestins and their binding partners using conventional structure determination tools. Therefore, structural information obtained using complementary structure analysis techniques would be necessary in combination with X-ray crystallography and cryo-EM data. In this review, we describe how ß-arrestins interact with their binding partners from a structural point of view, as elucidated by both traditional methods (X-ray crystallography and cryo-EM) and complementary structure analysis techniques.


Assuntos
beta-Arrestinas/química , Microscopia Crioeletrônica , Cristalografia por Raios X , Ligação Proteica , Conformação Proteica , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais , beta-Arrestinas/metabolismo
13.
Int J Mol Sci ; 21(21)2020 Oct 30.
Artigo em Inglês | MEDLINE | ID: mdl-33143110

RESUMO

Activation of the cannabinoid CB1 receptor induces different cellular signaling cascades through coupling to different effector proteins (G-proteins and ß-arrestins), triggering numerous therapeutic effects. Conformational changes and rearrangements at the intracellular domain of this GPCR receptor that accompany ligand binding dictate the signaling pathways. The GPCR-binding interface for G proteins has been extensively studied, whereas ß-arrestin/GPCR complexes are still poorly understood. To gain knowledge in this direction, we designed peptides that mimic the motifs involved in the putative interacting region: ß-arrestin1 finger loop and the transmembrane helix 7-helix 8 (TMH7-H8) elbow located at the intracellular side of the CB1 receptor. According to circular dichroism and NMR data, these peptides form a native-like, helical conformation and interact with each other in aqueous solution, in the presence of trifluoroethanol, and using zwitterionic detergent micelles as membrane mimics. These results increase our understanding of the binding mode of ß-arrestin and CB1 receptor and validate minimalist approaches to structurally comprehend complex protein systems.


Assuntos
Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/metabolismo , Receptor CB1 de Canabinoide/química , Receptor CB1 de Canabinoide/metabolismo , beta-Arrestinas/química , beta-Arrestinas/metabolismo , Dicroísmo Circular/métodos , Humanos , Espectroscopia de Ressonância Magnética/métodos , Modelos Moleculares , Estrutura Secundária de Proteína , Transdução de Sinais
14.
Adv Cancer Res ; 145: 139-156, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32089163

RESUMO

ß-Arrestins (ßarrs) are multifunctional intracellular proteins with an ability to directly interact with a large number of cellular partners including the G protein-coupled receptors (GPCRs). ßarrs contribute to multiple aspects of GPCR signaling, trafficking and downregulation. Considering the central involvement of GPCR signaling in the onset and progression of diverse types of cancers, ßarrs have also emerged as key players in the context of investigating cancer phenotypes, and as potential therapeutic targets. In this chapter, we first provide a brief account of structure and function of ßarrs and then highlight recent discoveries unfolding novel functional attributes of ßarrs in breast cancer. We also underscore the recent paradigms of modulating ßarr functions in cellular context and potential therapeutic opportunities going forward.


Assuntos
Antineoplásicos/uso terapêutico , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/metabolismo , Terapia de Alvo Molecular , beta-Arrestinas/química , beta-Arrestinas/metabolismo , Animais , Neoplasias da Mama/patologia , Feminino , Humanos , Transdução de Sinais
15.
Sci Signal ; 13(617)2020 02 04.
Artigo em Inglês | MEDLINE | ID: mdl-32019899

RESUMO

Signaling bias is the propensity for some agonists to preferentially stimulate G protein-coupled receptor (GPCR) signaling through one intracellular pathway versus another. We previously identified a G protein-biased agonist of the D2 dopamine receptor (D2R) that results in impaired ß-arrestin recruitment. This signaling bias was predicted to arise from unique interactions of the ligand with a hydrophobic pocket at the interface of the second extracellular loop and fifth transmembrane segment of the D2R. Here, we showed that residue Phe189 within this pocket (position 5.38 using Ballesteros-Weinstein numbering) functions as a microswitch for regulating receptor interactions with ß-arrestin. This residue is relatively conserved among class A GPCRs, and analogous mutations within other GPCRs similarly impaired ß-arrestin recruitment while maintaining G protein signaling. To investigate the mechanism of this signaling bias, we used an active-state structure of the ß2-adrenergic receptor (ß2R) to build ß2R-WT and ß2R-Y1995.38A models in complex with the full ß2R agonist BI-167107 for molecular dynamics simulations. These analyses identified conformational rearrangements in ß2R-Y1995.38A that propagated from the extracellular ligand binding site to the intracellular surface, resulting in a modified orientation of the second intracellular loop in ß2R-Y1995.38A, which is predicted to affect its interactions with ß-arrestin. Our findings provide a structural basis for how ligand binding site alterations can allosterically affect GPCR-transducer interactions and result in biased signaling.


Assuntos
Simulação de Dinâmica Molecular , Receptores Adrenérgicos beta 2/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais , beta-Arrestinas/metabolismo , Sequência de Aminoácidos , Animais , Sítios de Ligação/genética , Células CHO , Cricetinae , Cricetulus , Proteínas de Ligação ao GTP/química , Proteínas de Ligação ao GTP/genética , Proteínas de Ligação ao GTP/metabolismo , Células HEK293 , Humanos , Ligantes , Modelos Moleculares , Mutação , Ligação Proteica , Domínios Proteicos , Receptores Adrenérgicos beta 2/química , Receptores Adrenérgicos beta 2/genética , Receptores Acoplados a Proteínas G/química , Receptores Acoplados a Proteínas G/genética , beta-Arrestinas/química , beta-Arrestinas/genética
16.
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
17.
Commun Biol ; 3(1): 27, 2020 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-31941999

RESUMO

The structural plasticity of G-protein coupled receptors (GPCRs) enables the long-range transmission of conformational changes induced by specific orthosteric site ligands and other pleiotropic factors. Here, we demonstrate that the ligand binding cavity in the sphingosine 1-phosphate receptor S1PR1, a class A GPCR, is in allosteric communication with both the ß-arrestin-binding C-terminal tail, and a receptor surface involved in oligomerization. We show that S1PR1 oligomers are required for full response to different agonists and ligand-specific association with arrestins, dictating the downstream signalling kinetics. We reveal that the active form of the immunomodulatory drug fingolimod, FTY720-P, selectively harnesses both these intramolecular networks to efficiently recruit ß-arrestins in a stable interaction with the receptor, promoting deep S1PR1 internalization and simultaneously abrogating ERK1/2 phosphorylation. Our results define a molecular basis for the efficacy of fingolimod for people with multiple sclerosis, and attest that GPCR signalling can be further fine-tuned by the oligomeric state.


Assuntos
Regulação Alostérica , Modelos Moleculares , Conformação Proteica , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/química , Linhagem Celular , Membrana Celular/metabolismo , Cloridrato de Fingolimode/química , Cloridrato de Fingolimode/farmacologia , Humanos , Cinética , Fosforilação , Pró-Proteína Convertases/química , Pró-Proteína Convertases/metabolismo , Ligação Proteica , Multimerização Proteica , Transporte Proteico , Receptores Acoplados a Proteínas G/metabolismo , Serina Endopeptidases/química , Serina Endopeptidases/metabolismo , Transdução de Sinais , Relação Estrutura-Atividade , beta-Arrestinas/química , beta-Arrestinas/metabolismo
18.
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
19.
Int J Mol Sci ; 21(24)2020 Dec 20.
Artigo em Inglês | MEDLINE | ID: mdl-33419260

RESUMO

G protein-coupled receptors are linked to various intracellular transducers, each pathway associated with different physiological effects. Biased ligands, capable of activating one pathway over another, are gaining attention for their therapeutic potential, as they could selectively activate beneficial pathways whilst avoiding those responsible for adverse effects. We performed molecular dynamics simulations with known ß-arrestin-biased ligands like lysergic acid diethylamide and ergotamine in complex with the 5-HT2B receptor and discovered that the extent of ligand bias is directly connected with the degree of closure of the extracellular loop region. Given a loose allosteric coupling of extracellular and intracellular receptor regions, we delineate a concept for biased signaling at serotonin receptors, by which conformational interference with binding pocket closure restricts the signaling repertoire of the receptor. Molecular docking studies of biased ligands gathered from the BiasDB demonstrate that larger ligands only show plausible docking poses in the ergotamine-bound structure, highlighting the conformational constraints associated with bias. This emphasizes the importance of selecting the appropriate receptor conformation on which to base virtual screening workflows in structure-based drug design of biased ligands. As this mechanism of ligand bias has also been observed for muscarinic receptors, our studies provide a general mechanism of signaling bias transferable between aminergic receptors.


Assuntos
Ergotamina/química , Dietilamida do Ácido Lisérgico/química , Conformação Proteica , Receptor 5-HT2B de Serotonina/química , Sítios de Ligação/genética , Humanos , Ligantes , Simulação de Acoplamento Molecular , Simulação de Dinâmica Molecular , Receptor 5-HT2B de Serotonina/genética , Receptor 5-HT2B de Serotonina/ultraestrutura , Receptores Acoplados a Proteínas G/química , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/ultraestrutura , Serotonina/química , Transdução de Sinais/genética , beta-Arrestinas/química , beta-Arrestinas/genética , beta-Arrestinas/ultraestrutura
20.
Nat Struct Mol Biol ; 26(12): 1123-1131, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31740855

RESUMO

Classically, G-protein-coupled receptors (GPCRs) are thought to activate G protein from the plasma membrane and are subsequently desensitized by ß-arrestin (ß-arr). However, some GPCRs continue to signal through G protein from internalized compartments, mediated by a GPCR-G protein-ß-arr 'megaplex'. Nevertheless, the molecular architecture of the megaplex remains unknown. Here, we present its cryo-electron microscopy structure, which shows simultaneous engagement of human G protein and bovine ß-arr to the core and phosphorylated tail, respectively, of a single active human chimeric ß2-adrenergic receptor with the C-terminal tail of the arginine vasopressin type 2 receptor (ß2V2R). All three components adopt their canonical active conformations, suggesting that a single megaplex GPCR is capable of simultaneously activating G protein and ß-arr. Our findings provide a structural basis for GPCR-mediated sustained internalized G protein signaling.


Assuntos
Proteínas de Ligação ao GTP/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais , beta-Arrestinas/metabolismo , Animais , Bovinos , Microscopia Crioeletrônica , Endossomos/metabolismo , Proteínas de Ligação ao GTP/química , Proteínas de Ligação ao GTP/ultraestrutura , Humanos , Modelos Moleculares , Conformação Proteica , Receptores Adrenérgicos beta 2/química , Receptores Adrenérgicos beta 2/metabolismo , Receptores Adrenérgicos beta 2/ultraestrutura , Receptores Acoplados a Proteínas G/química , Receptores Acoplados a Proteínas G/ultraestrutura , Receptores de Vasopressinas/química , Receptores de Vasopressinas/metabolismo , Receptores de Vasopressinas/ultraestrutura , beta-Arrestinas/química , beta-Arrestinas/ultraestrutura
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA