Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Nature ; 620(7975): 904-910, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37558880

ABSTRACT

Arrestins have pivotal roles in regulating G protein-coupled receptor (GPCR) signalling by desensitizing G protein activation and mediating receptor internalization1,2. It has been proposed that the arrestin binds to the receptor in two different conformations, 'tail' and 'core', which were suggested to govern distinct processes of receptor signalling and trafficking3,4. However, little structural information is available for the tail engagement of the arrestins. Here we report two structures of the glucagon receptor (GCGR) bound to ß-arrestin 1 (ßarr1) in glucagon-bound and ligand-free states. These structures reveal a receptor tail-engaged binding mode of ßarr1 with many unique features, to our knowledge, not previously observed. Helix VIII, instead of the receptor core, has a major role in accommodating ßarr1 by forming extensive interactions with the central crest of ßarr1. The tail-binding pose is further defined by a close proximity between the ßarr1 C-edge and the receptor helical bundle, and stabilized by a phosphoinositide derivative that bridges ßarr1 with helices I and VIII of GCGR. Lacking any contact with the arrestin, the receptor core is in an inactive state and loosely binds to glucagon. Further functional studies suggest that the tail conformation of GCGR-ßarr governs ßarr recruitment at the plasma membrane and endocytosis of GCGR, and provides a molecular basis for the receptor forming a super-complex simultaneously with G protein and ßarr to promote sustained signalling within endosomes. These findings extend our knowledge about the arrestin-mediated modulation of GPCR functionalities.


Subject(s)
Receptors, Glucagon , beta-Arrestin 1 , beta-Arrestin 1/chemistry , beta-Arrestin 1/metabolism , Cell Membrane/metabolism , Endocytosis , Endosomes/metabolism , Glucagon/metabolism , Heterotrimeric GTP-Binding Proteins/metabolism , Ligands , Phosphatidylinositols/metabolism , Receptors, Glucagon/chemistry , Receptors, Glucagon/metabolism , Protein Binding
2.
Nat Commun ; 14(1): 1151, 2023 03 01.
Article in English | MEDLINE | ID: mdl-36859440

ABSTRACT

Understanding the molecular basis of arrestin-mediated regulation of GPCRs is critical for deciphering signaling mechanisms and designing functional selectivity. However, structural studies of GPCR-arrestin complexes are hampered by their highly dynamic nature. Here, we dissect the interaction of arrestin-2 (arr2) with the secretin-like parathyroid hormone 1 receptor PTH1R using genetically encoded crosslinking amino acids in live cells. We identify 136 intermolecular proximity points that guide the construction of energy-optimized molecular models for the PTH1R-arr2 complex. Our data reveal flexible receptor elements missing in existing structures, including intracellular loop 3 and the proximal C-tail, and suggest a functional role of a hitherto overlooked positively charged region at the arrestin N-edge. Unbiased MD simulations highlight the stability and dynamic nature of the complex. Our integrative approach yields structural insights into protein-protein complexes in a biologically relevant live-cell environment and provides information inaccessible to classical structural methods, while also revealing the dynamics of the system.


Subject(s)
Amino Acids , Receptor, Parathyroid Hormone, Type 1 , beta-Arrestin 1 , beta-Arrestin 1/chemistry , Models, Molecular , Receptor, Parathyroid Hormone, Type 1/chemistry
3.
J Phys Chem B ; 126(9): 1917-1932, 2022 03 10.
Article in English | MEDLINE | ID: mdl-35196859

ABSTRACT

The large conformational flexibility of G protein-coupled receptors (GPCRs) has been a puzzle in structural and pharmacological studies for the past few decades. Apart from structural rearrangements induced by ligands, enzymatic phosphorylations by GPCR kinases (GRKs) at the carboxy-terminal tail (C-tail) of a GPCR also make conformational alterations to the transmembrane helices and facilitates the binding of one of its transducer proteins named ß-arrestin. The phosphorylation-induced conformational transition of the receptor that causes specific binding to ß-arrestin but prevents the association of other transducers such as G proteins lacks atomistic understanding and is elusive to experimental studies. Using microseconds of all-atom conventional and Gaussian accelerated molecular dynamics (GaMD) simulations, we investigate the allosteric mechanism of phosphorylation induced-conformational changes in ß2-adrenergic receptor, a well-characterized GPCR model system. Free energy profiles reveal that the phosphorylated receptor samples a new conformational state in addition to the canonical active state corroborating with recent nuclear magnetic resonance experimental findings. The new state has a smaller intracellular cavity that is likely to accommodate ß-arrestin better than G protein. Using contact map and inter-residue interaction energy calculations, we found the phosphorylated C-tail adheres to the cytosolic surface of the transmembrane domain of the receptor. Transfer entropy calculations show that the C-tail residues drive the correlated motions of TM residues, and the allosteric signal is relayed via several residues at the cytosolic surface. Our results also illustrate how the redistribution of inter-residue nonbonding interaction couples with the allosteric communication from the phosphorylated C-tail to the transmembrane. Atomistic insight into phosphorylation-induced ß-arrestin specific conformation is therapeutically important to design drugs with higher efficacy and fewer side effects. Our results, therefore, open novel opportunities to fine-tune ß-arrestin bias in GPCR signaling.


Subject(s)
Receptors, G-Protein-Coupled , Signal Transduction , GTP-Binding Proteins/metabolism , Phosphorylation , Protein Binding , Receptors, G-Protein-Coupled/metabolism , beta-Arrestin 1/chemistry , beta-Arrestin 1/metabolism , beta-Arrestins/metabolism
4.
J Mol Biol ; 434(7): 167465, 2022 04 15.
Article in English | MEDLINE | ID: mdl-35077767

ABSTRACT

Arrestin binding to active phosphorylated G protein-coupled receptors terminates G protein coupling and initiates another wave of signaling. Among the effectors that bind directly to receptor-associated arrestins are extracellular signal-regulated kinases 1/2 (ERK1/2), which promote cellular proliferation and survival. Arrestins may also engage ERK1/2 in isolation in a pre- or post-signaling complex that is likely in equilibrium with the full signal initiation complex. Molecular details of these binary complexes remain unknown. Here, we investigate the molecular mechanisms whereby arrestin-2 and arrestin-3 (a.k.a. ß-arrestin1 and ß-arrestin2, respectively) engage ERK1/2 in pairwise interactions. We find that purified arrestin-3 binds ERK2 more avidly than arrestin-2. A combination of biophysical techniques and peptide array analysis demonstrates that the molecular basis in this difference of binding strength is that the two non-visual arrestins bind ERK2 via different parts of the molecule. We propose a structural model of the ERK2-arrestin-3 complex in solution using size-exclusion chromatography coupled to small angle X-ray scattering (SEC-SAXS). This binary complex exhibits conformational heterogeneity. We speculate that this drives the equilibrium either toward the full signaling complex with receptor-bound arrestin at the membrane or toward full dissociation in the cytoplasm. As ERK1/2 regulates cell migration, proliferation, and survival, understanding complexes that relate to its activation could be exploited to control cell fate.


Subject(s)
Mitogen-Activated Protein Kinase 1 , beta-Arrestin 1 , beta-Arrestin 2 , Mitogen-Activated Protein Kinase 1/chemistry , Protein Binding , Scattering, Small Angle , X-Ray Diffraction , beta-Arrestin 1/chemistry , beta-Arrestin 2/chemistry
5.
Nat Commun ; 12(1): 7158, 2021 12 09.
Article in English | MEDLINE | ID: mdl-34887409

ABSTRACT

ß-arrestins (ßarrs) play multifaceted roles in the function of G protein-coupled receptors (GPCRs). ßarrs typically interact with phosphorylated C-terminal tail (C tail) and transmembrane core (TM core) of GPCRs. However, the effects of the C tail- and TM core-mediated interactions on the conformational activation of ßarrs have remained elusive. Here, we show the conformational changes for ßarr activation upon the C tail- and TM core-mediated interactions with a prototypical GPCR by nuclear magnetic resonance (NMR) spectroscopy. Our NMR analyses demonstrated that while the C tail-mediated interaction alone induces partial activation, in which ßarr exists in equilibrium between basal and activated conformations, the TM core- and the C tail-mediated interactions together completely shift the equilibrium toward the activated conformation. The conformation-selective antibody, Fab30, promotes partially activated ßarr into the activated-like conformation. This plasticity of ßarr conformation in complex with GPCRs engaged in different binding modes may explain the multifunctionality of ßarrs.


Subject(s)
Receptors, G-Protein-Coupled/metabolism , beta-Arrestin 1/chemistry , beta-Arrestin 1/metabolism , Humans , Magnetic Resonance Spectroscopy , Protein Binding , Protein Conformation, beta-Strand , Protein Domains , Receptors, G-Protein-Coupled/chemistry , Receptors, G-Protein-Coupled/genetics , beta-Arrestin 1/genetics
6.
J Biol Chem ; 295(49): 16773-16784, 2020 12 04.
Article in English | MEDLINE | ID: mdl-32978252

ABSTRACT

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.


Subject(s)
Receptor, Muscarinic M2/metabolism , Receptors, Adrenergic, beta-2/metabolism , Receptors, Vasopressin/metabolism , beta-Arrestin 1/metabolism , src-Family Kinases/metabolism , Allosteric Regulation , Enzyme Activation , Humans , Kinetics , Mutagenesis, Site-Directed , Nanostructures/chemistry , Peptides/chemical synthesis , Peptides/chemistry , Phosphorylation , Protein Binding , Proto-Oncogene Mas , Receptor, Muscarinic M2/chemistry , Receptors, Adrenergic, beta-2/chemistry , Receptors, Vasopressin/chemistry , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Substrate Specificity , beta-Arrestin 1/chemistry , beta-Arrestin 1/genetics , src Homology Domains , src-Family Kinases/chemistry
7.
Nat Commun ; 11(1): 4857, 2020 09 25.
Article in English | MEDLINE | ID: mdl-32978402

ABSTRACT

Characterization of the dynamic conformational changes in membrane protein signaling complexes by nuclear magnetic resonance (NMR) spectroscopy remains challenging. Here we report the site-specific incorporation of 4-trimethylsilyl phenylalanine (TMSiPhe) into proteins, through genetic code expansion. Crystallographic analysis revealed structural changes that reshaped the TMSiPhe-specific amino-acyl tRNA synthetase active site to selectively accommodate the trimethylsilyl (TMSi) group. The unique up-field 1H-NMR chemical shift and the highly efficient incorporation of TMSiPhe enabled the characterization of multiple conformational states of a phospho-ß2 adrenergic receptor/ß-arrestin-1(ß-arr1) membrane protein signaling complex, using only 5 µM protein and 20 min of spectrum accumulation time. We further showed that extracellular ligands induced conformational changes located in the polar core or ERK interaction site of ß-arr1 via direct receptor transmembrane core interactions. These observations provided direct delineation and key mechanism insights that multiple receptor ligands were able to induce distinct functionally relevant conformational changes of arrestin.


Subject(s)
Arrestin/chemistry , Arrestin/genetics , Arrestin/metabolism , Ligands , Proton Magnetic Resonance Spectroscopy/methods , Binding Sites , Crystallography, X-Ray , Humans , Models, Molecular , Phenylalanine , Protein Binding , Protein Conformation , Receptors, Adrenergic, beta-2/metabolism , Signal Transduction , beta-Arrestin 1/chemistry , beta-Arrestin 1/genetics , beta-Arrestin 1/metabolism
8.
Biochim Biophys Acta Mol Cell Res ; 1867(12): 118849, 2020 12.
Article in English | MEDLINE | ID: mdl-32916203

ABSTRACT

FPR2, a member of the family of G protein-coupled receptors (GPCRs), mediates neutrophil migration, a response that has been linked to ß-arrestin recruitment. ß-Arrestin regulates GPCR endocytosis and can also elicit non-canonical receptor signaling. To determine the poorly understood role of ß-arrestin in FPR2 endocytosis and in NADPH-oxidase activation in neutrophils, Barbadin was used as a research tool in this study. Barbadin has been shown to bind the clathrin adaptor protein (AP2) and thereby prevent ß-arrestin/AP2 interaction and ß-arrestin-mediated GPCR endocytosis. In agreement with this, AP2/ß-arrestin interaction induced by an FPR2-specific agonist was inhibited by Barbadin. Unexpectedly, however, Barbadin did not inhibit FPR2 endocytosis, indicating that a mechanism independent of ß-arrestin/AP2 interaction may sustain FPR2 endocytosis. This was confirmed by the fact, that FPR2 also underwent agonist-promoted endocytosis in ß-arrestin deficient cells, albeit at a diminished level as compared to wild type cells. Dissection of the Barbadin effects on FPR2-mediated neutrophil functions including NADPH-oxidase activation mediated release of reactive oxygen species (ROS) and chemotaxis revealed that Barbadin had no effect on chemotactic migration whereas the release of ROS was potentiated/primed. The effect of Barbadin on ROS production was reversible, independent of ß-arrestin recruitment, and similar to that induced by latrunculin A. Taken together, our data demonstrate that endocytic uptake of FPR2 occurs independently of ß-arrestin, while Barbadin selectively augments FPR2-mediated ROS production independently of receptor endocytosis. Given that Barbadin binds to AP2 and prevents the AP2/ß-arrestin interaction, our results indicate a role for AP2 in FPR2-mediated ROS release from neutrophils.


Subject(s)
Endocytosis/genetics , Pyrimidines/pharmacology , Receptors, Formyl Peptide/genetics , Receptors, Lipoxin/genetics , beta-Arrestin 1/genetics , Adaptor Protein Complex 2/chemistry , Adaptor Protein Complex 2/genetics , Clathrin/chemistry , Endocytosis/drug effects , HEK293 Cells , Humans , NADPH Oxidases/genetics , Neutrophils/drug effects , Protein Binding/drug effects , Pyrimidines/chemistry , Reactive Oxygen Species/metabolism , Receptors, Formyl Peptide/chemistry , Receptors, G-Protein-Coupled/genetics , Receptors, Lipoxin/chemistry , Signal Transduction/drug effects , beta-Arrestin 1/chemistry
9.
J Biol Chem ; 295(41): 14111-14124, 2020 10 09.
Article in English | MEDLINE | ID: mdl-32753481

ABSTRACT

The multifaceted adaptor protein ß-arr1 (ß-arrestin1) promotes activation of focal adhesion kinase (FAK) by the chemokine receptor CXCR4, facilitating chemotaxis. This function of ß-arr1 requires the assistance of the adaptor protein STAM1 (signal-transducing adaptor molecule 1) because disruption of the interaction between STAM1 and ß-arr1 reduces CXCR4-mediated activation of FAK and chemotaxis. To begin to understand the mechanism by which ß-arr1 together with STAM1 activates FAK, we used site-directed spin-labeling EPR spectroscopy-based studies coupled with bioluminescence resonance energy transfer-based cellular studies to show that STAM1 is recruited to activated ß-arr1 by binding to a novel surface on ß-arr1 at the base of the finger loop, at a site that is distinct from the receptor-binding site. Expression of a STAM1-deficient binding ß-arr1 mutant that is still able to bind to CXCR4 significantly reduced CXCL12-induced activation of FAK but had no impact on ERK-1/2 activation. We provide evidence of a novel surface at the base of the finger loop that dictates non-GPCR interactions specifying ß-arrestin-dependent signaling by a GPCR. This surface might represent a previously unidentified switch region that engages with effector molecules to drive ß-arrestin signaling.


Subject(s)
Adaptor Proteins, Signal Transducing , Endosomal Sorting Complexes Required for Transport , MAP Kinase Signaling System , Phosphoproteins , Receptors, CXCR4 , beta-Arrestin 1 , Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Chemokine CXCL12/chemistry , Chemokine CXCL12/genetics , Chemokine CXCL12/metabolism , Endosomal Sorting Complexes Required for Transport/chemistry , Endosomal Sorting Complexes Required for Transport/genetics , Endosomal Sorting Complexes Required for Transport/metabolism , Focal Adhesion Kinase 1/chemistry , Focal Adhesion Kinase 1/genetics , Focal Adhesion Kinase 1/metabolism , HEK293 Cells , Humans , Phosphoproteins/chemistry , Phosphoproteins/genetics , Phosphoproteins/metabolism , Protein Structure, Secondary , Receptors, CXCR4/chemistry , Receptors, CXCR4/genetics , Receptors, CXCR4/metabolism , beta-Arrestin 1/chemistry , beta-Arrestin 1/genetics , beta-Arrestin 1/metabolism
10.
Nature ; 583(7818): 862-866, 2020 07.
Article in English | MEDLINE | ID: mdl-32555462

ABSTRACT

The ß1-adrenoceptor (ß1AR) is a G-protein-coupled receptor (GPCR) that couples1 to the heterotrimeric G protein Gs. G-protein-mediated signalling is terminated by phosphorylation of the C terminus of the receptor by GPCR kinases (GRKs) and by coupling of ß-arrestin 1 (ßarr1, also known as arrestin 2), which displaces Gs and induces signalling through the MAP kinase pathway2. The ability of synthetic agonists to induce signalling preferentially through either G proteins or arrestins-known as biased agonism3-is important in drug development, because the therapeutic effect may arise from only one signalling cascade, whereas the other pathway may mediate undesirable side effects4. To understand the molecular basis for arrestin coupling, here we determined the cryo-electron microscopy structure of the ß1AR-ßarr1 complex in lipid nanodiscs bound to the biased agonist formoterol5, and the crystal structure of formoterol-bound ß1AR coupled to the G-protein-mimetic nanobody6 Nb80. ßarr1 couples to ß1AR in a manner distinct to that7 of Gs coupling to ß2AR-the finger loop of ßarr1 occupies a narrower cleft on the intracellular surface, and is closer to transmembrane helix H7 of the receptor when compared with the C-terminal α5 helix of Gs. The conformation of the finger loop in ßarr1 is different from that adopted by the finger loop of visual arrestin when it couples to rhodopsin8. ß1AR coupled to ßarr1 shows considerable differences in structure compared with ß1AR coupled to Nb80, including an inward movement of extracellular loop 3 and the cytoplasmic ends of H5 and H6. We observe weakened interactions between formoterol and two serine residues in H5 at the orthosteric binding site of ß1AR, and find that formoterol has a lower affinity for the ß1AR-ßarr1 complex than for the ß1AR-Gs complex. The structural differences between these complexes of ß1AR provide a foundation for the design of small molecules that could bias signalling in the ß-adrenoceptors.


Subject(s)
Cryoelectron Microscopy , Formoterol Fumarate/chemistry , Formoterol Fumarate/metabolism , Receptors, Adrenergic, beta-1/chemistry , Receptors, Adrenergic, beta-1/ultrastructure , beta-Arrestin 1/chemistry , beta-Arrestin 1/ultrastructure , Amino Acid Sequence , Animals , Binding Sites , GTP-Binding Protein alpha Subunits, Gs/chemistry , GTP-Binding Protein alpha Subunits, Gs/metabolism , GTP-Binding Protein alpha Subunits, Gs/ultrastructure , HEK293 Cells , Humans , Models, Molecular , Multiprotein Complexes , Receptors, Adrenergic, beta-1/metabolism , Single-Chain Antibodies/chemistry , Single-Chain Antibodies/metabolism , Single-Chain Antibodies/ultrastructure , Zebrafish , beta-Arrestin 1/metabolism
11.
Biochim Biophys Acta Biomembr ; 1862(9): 183355, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32413442

ABSTRACT

G protein-coupled receptors (GPCRs) are critically regulated by arrestins, which not only desensitize G-protein signaling but also initiate a G protein-independent wave of signaling. The information from structure determination was herein exploited to build a structural model of the ternary complex, comprising fully phosphorylated V2 vasopressin receptor (V2R), the agonist arginine vasopressin (AVP), and ß-arrestin 1 (ß-arr1). Molecular simulations served to explore dynamics and structural communication in the ternary complex. Flexibility and mechanical profiles reflect fold of V2R and ß-arr1. Highly conserved amino acids tend to behave as hubs in the structure network and contribute the most to the mechanical rigidity of V2R seven-helix bundle and of ß-arr1. Two structurally and dynamically distinct receptor-arrestin interfaces assist the twist of the N- and C-terminal domains (ND and CD, respectively) of ß-arr1 with respect to each other, which is linked to arrestin activation. While motion of the ND is essentially assisted by the fully phosphorylated C-tail of V2R (V2RCt), that of CD is assisted by the second and third intracellular loops and the cytosolic extensions of helices 5 and 6. In the presence of the receptor, the ß-arr1 inter-domain twist angle correlates with the modes describing the essential subspace of the ternary complex. ß-arr1 motions are also influenced by the anchoring to the membrane of the C-edge-loops in the ß-arr1-CD. Overall fluctuations reveal a coupling between motions of the agonist binding site and of ß-arr1-ND, which are in allosteric communication between each other. Mechanical rigidity points, often acting as hubs in the structure network and distributed along the main axis of the receptor helix bundle, contribute to establish a preferential communication pathway between agonist ligand and the ND of arrestin. Such communication, mediated by highly conserved amino acids, involves also the first amino acid in the arrestin C-tail, which is highly dynamic and is involved in clathrin-mediated GPCR internalization.


Subject(s)
Molecular Dynamics Simulation , Receptors, Vasopressin/chemistry , Vasopressins/chemistry , beta-Arrestin 1/chemistry , Humans
12.
Trends Pharmacol Sci ; 41(6): 387-389, 2020 06.
Article in English | MEDLINE | ID: mdl-32362341

ABSTRACT

The ability to design 'biased' drugs that selectively activate G protein-coupled receptor (GPCR) signaling pathways beneficial in treating a disease, while limiting their side effects, is of broad significance. Lee et al. move us a step closer to this important goal by identifying structural differences in the ß1-adrenoceptor in complex with ß-arrestin 1 versus a G protein-mimicking nanobody.


Subject(s)
Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/chemistry , beta-Arrestin 1/chemistry , Animals , Biomimetic Materials/chemistry , Humans , Models, Molecular , Molecular Targeted Therapy , Receptors, Adrenergic, beta-1/chemistry , Receptors, Adrenergic, beta-1/metabolism , Receptors, G-Protein-Coupled/metabolism , Signal Transduction/drug effects , Single-Domain Antibodies/chemistry , beta-Arrestin 1/metabolism
13.
PLoS One ; 15(4): e0228121, 2020.
Article in English | MEDLINE | ID: mdl-32236094

ABSTRACT

Melanopsin is a visual pigment expressed in a small subset of ganglion cells in the mammalian retina known as intrinsically photosensitive retinal ganglion cells (ipRGCs) and is implicated in regulating non-image forming functions such as circadian photoentrainment and pupil constriction and contrast sensitivity in image formation. Mouse melanopsin's Carboxy-terminus (C-terminus) possesses 38 serine and threonine residues, which can potentially serve as phosphorylation sites for a G-protein Receptor Kinase (GRK) and be involved in the deactivation of signal transduction. Previous studies suggest that S388, T389, S391, S392, S394, S395 on the proximal region of the C-terminus of mouse melanopsin are necessary for melanopsin deactivation. We expressed a series of mouse melanopsin C-terminal mutants in HEK293 cells and using calcium imaging, and we found that the necessary cluster of six serine and threonine residues, while being critical, are insufficient for proper melanopsin deactivation. Interestingly, the additional six serine and threonine residues adjacent to the required six sites, in either proximal or distal direction, are capable of restoring wild-type deactivation of melanopsin. These findings suggest an element of plasticity in the molecular basis of melanopsin phosphorylation and deactivation. In addition, C-terminal chimeric mutants and molecular modeling studies support the idea that the initial steps of deactivation and ß-arrestin binding are centered around these critical phosphorylation sites (S388-S395). The degree of functional versatility described in this study, along with ipRGC biophysical heterogeneity and the possible use of multiple signal transduction cascades, might contribute to the diverse ipRGC light responses for use in non-image and image forming behaviors, even though all six sub types of ipRGCs express the same melanopsin gene OPN4.


Subject(s)
Light Signal Transduction/physiology , Recombinant Fusion Proteins/metabolism , Rod Opsins/metabolism , beta-Arrestin 1/metabolism , HEK293 Cells , Humans , Models, Molecular , Mutagenesis, Site-Directed , Mutation , Phosphorylation/physiology , Protein Binding , Receptor, Angiotensin, Type 1/genetics , Receptor, Angiotensin, Type 1/metabolism , Receptors, Adrenergic, beta-2/genetics , Receptors, Adrenergic, beta-2/metabolism , Recombinant Fusion Proteins/genetics , Rod Opsins/chemistry , Rod Opsins/genetics , Serine/genetics , Serine/metabolism , Threonine/genetics , Threonine/metabolism , beta-Arrestin 1/chemistry
14.
Structure ; 28(3): 314-323.e3, 2020 03 03.
Article in English | MEDLINE | ID: mdl-31948726

ABSTRACT

Arrestins desensitize and/or internalize G-protein-coupled receptors by interacting with phosphorylated receptors. A few studies have reported that arrestins themselves can be phosphorylated, and the phosphorylation status modulates their cellular functions. However, the effects of phosphorylation on arrestin structure have not been studied. Here, we investigated the conformational changes in ß-arrestin-1 and -2 upon incorporation of phospho-mimetic mutations into the known phosphorylation sites (i.e., S412D for ß-arrestin-1 and S14D, T276D, S14D/T276D, S361D, T383D, and S361D/T383D for ß-arrestin-2) by using hydrogen/deuterium-exchange mass spectrometry (HDX-MS). HDX-MS analysis suggested that ß-arrestin-2 S14D/T276D shows an HDX profile similar to the pre-active states, resulting in increased interaction with receptors. Phospho-mimetic mutation at corresponding residues of ß-arrestin-1 (i.e., S13D/T275D) induced similar conformational and functional consequences, and the detailed structural changes related to ß-arrestin-1 S13D/T275D were investigated further by X-ray crystallography.


Subject(s)
Mutation , beta-Arrestin 1/chemistry , beta-Arrestin 1/metabolism , beta-Arrestin 2/chemistry , beta-Arrestin 2/metabolism , Amino Acid Sequence , Animals , Crystallography, X-Ray , Hydrogen Deuterium Exchange-Mass Spectrometry , Models, Molecular , Phosphorylation , Protein Binding , Protein Conformation , Rats , beta-Arrestin 1/genetics , beta-Arrestin 2/genetics
15.
Nature ; 579(7798): 303-308, 2020 03.
Article in English | MEDLINE | ID: mdl-31945771

ABSTRACT

Arrestin proteins bind to active, phosphorylated G-protein-coupled receptors (GPCRs), thereby preventing G-protein coupling, triggering receptor internalization and affecting various downstream signalling pathways1,2. Although there is a wealth of structural information detailing the interactions between GPCRs and G proteins, less is known about how arrestins engage GPCRs. Here we report a cryo-electron microscopy structure of full-length human neurotensin receptor 1 (NTSR1) in complex with truncated human ß-arrestin 1 (ßarr1(ΔCT)). We find that phosphorylation of NTSR1 is critical for the formation of a stable complex with ßarr1(ΔCT), and identify phosphorylated sites in both the third intracellular loop and the C terminus that may promote this interaction. In addition, we observe a phosphatidylinositol-4,5-bisphosphate molecule forming a bridge between the membrane side of NTSR1 transmembrane segments 1 and 4 and the C-lobe of arrestin. Compared with a structure of a rhodopsin-arrestin-1 complex, in our structure arrestin is rotated by approximately 85° relative to the receptor. These findings highlight both conserved aspects and plasticity among arrestin-receptor interactions.


Subject(s)
Models, Molecular , Receptors, Neurotensin/chemistry , beta-Arrestin 1/chemistry , Cryoelectron Microscopy , Humans , Phosphorylation , Protein Stability , Protein Structure, Quaternary , Receptors, Neurotensin/metabolism , beta-Arrestin 1/metabolism
16.
J Vis Exp ; (148)2019 06 28.
Article in English | MEDLINE | ID: mdl-31305535

ABSTRACT

Interactions between G-protein coupled receptors (GPCRs) and ß-arrestins are vital processes with physiological implications of great importance. Currently, the characterization of novel drugs towards their interactions with ß-arrestins and other cytosolic proteins is extremely valuable in the field of GPCR drug discovery particularly during the study of GPCR biased agonism. Here, we show the application of a novel structural complementation assay to accurately monitor receptor-ß-arrestin interactions in real time living systems. This method is simple, accurate and can be easily extended to any GPCR of interest and also it has the advantage that it overcomes unspecific interactions due to the presence of a low expression promoter present in each vector system. This structural complementation assay provides key features that allow an accurate and precise monitoring of receptor-ß-arrestin interactions, making it suitable in the study of biased agonism of any GPCR system as well as GPCR c-terminus 'phosphorylation codes' written by different GPCR-kinases (GRKs) and post-translational modifications of arrestins that stabilize or destabilize the receptor-ß-arrestin complex.


Subject(s)
Drug Discovery , Receptors, G-Protein-Coupled/metabolism , beta-Arrestin 1/chemistry , beta-Arrestin 2/metabolism , Cell Membrane/metabolism , Phosphorylation
17.
Nat Commun ; 10(1): 1261, 2019 03 19.
Article in English | MEDLINE | ID: mdl-30890705

ABSTRACT

Cellular functions of arrestins are determined in part by the pattern of phosphorylation on the G protein-coupled receptors (GPCRs) to which arrestins bind. Despite high-resolution structural data of arrestins bound to phosphorylated receptor C-termini, the functional role of each phosphorylation site remains obscure. Here, we employ a library of synthetic phosphopeptide analogues of the GPCR rhodopsin C-terminus and determine the ability of these peptides to bind and activate arrestins using a variety of biochemical and biophysical methods. We further characterize how these peptides modulate the conformation of arrestin-1 by nuclear magnetic resonance (NMR). Our results indicate different functional classes of phosphorylation sites: 'key sites' required for arrestin binding and activation, an 'inhibitory site' that abrogates arrestin binding, and 'modulator sites' that influence the global conformation of arrestin. These functional motifs allow a better understanding of how different GPCR phosphorylation patterns might control how arrestin functions in the cell.


Subject(s)
Arrestin/metabolism , Phosphorylation/physiology , Rhodopsin/metabolism , beta-Arrestin 1/metabolism , beta-Arrestin 2/metabolism , Amino Acid Motifs/physiology , Animals , Arrestin/chemistry , Arrestin/genetics , Arrestin/isolation & purification , Biological Assay , Cattle , Cell Membrane/metabolism , Mutation , Nuclear Magnetic Resonance, Biomolecular , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Rhodopsin/chemistry , Rod Cell Outer Segment/metabolism , beta-Arrestin 1/chemistry , beta-Arrestin 1/isolation & purification , beta-Arrestin 2/chemistry , beta-Arrestin 2/isolation & purification
18.
Nat Commun ; 9(1): 194, 2018 01 15.
Article in English | MEDLINE | ID: mdl-29335412

ABSTRACT

The C-terminal region of G-protein-coupled receptors (GPCRs), stimulated by agonist binding, is phosphorylated by GPCR kinases, and the phosphorylated GPCRs bind to arrestin, leading to the cellular responses. To understand the mechanism underlying the formation of the phosphorylated GPCR-arrestin complex, we performed NMR analyses of the phosphorylated ß2-adrenoceptor (ß2AR) and the phosphorylated ß2AR-ß-arrestin 1 complex, in the lipid bilayers of nanodisc. Here we show that the phosphorylated C-terminal region adheres to either the intracellular side of the transmembrane region or lipids, and that the phosphorylation of the C-terminal region allosterically alters the conformation around M2155.54 and M2796.41, located on transemembrane helices 5 and 6, respectively. In addition, we found that the conformation induced by the phosphorylation is similar to that corresponding to the ß-arrestin-bound state. The phosphorylation-induced structures revealed in this study propose a conserved structural motif of GPCRs that enables ß-arrestin to recognize dozens of GPCRs.


Subject(s)
Receptors, Adrenergic, beta-2/chemistry , Receptors, Adrenergic, beta-2/metabolism , beta-Arrestin 1/chemistry , beta-Arrestin 1/metabolism , Cell Membrane/chemistry , Cell Membrane/genetics , Cell Membrane/metabolism , Humans , Magnetic Resonance Spectroscopy , Models, Molecular , Phosphorylation , Protein Binding , Protein Conformation , Protein Domains , Receptors, Adrenergic, beta-2/genetics , beta-Arrestin 1/genetics
19.
Nat Chem Biol ; 14(2): 126-134, 2018 02.
Article in English | MEDLINE | ID: mdl-29227473

ABSTRACT

Development of biased ligands targeting G protein-coupled receptors (GPCRs) is a promising approach for current drug discovery. Although structure-based drug design of biased agonists remains challenging even with an abundance of GPCR crystal structures, we present an approach for translating GPCR structural data into ß-arrestin-biased ligands for aminergic GPCRs. We identified specific amino acid-ligand contacts at transmembrane helix 5 (TM5) and extracellular loop 2 (EL2) responsible for Gi/o and ß-arrestin signaling, respectively, and targeted those residues to develop biased ligands. For these ligands, we found that bias is conserved at other aminergic GPCRs that retain similar residues at TM5 and EL2. Our approach provides a template for generating arrestin-biased ligands by modifying predicted ligand interactions that block TM5 interactions and promote EL2 interactions. This strategy may facilitate the structure-guided design of arrestin-biased ligands at other GPCRs, including polypharmacological biased ligands.


Subject(s)
Ligands , Receptors, G-Protein-Coupled/chemistry , beta-Arrestin 1/chemistry , Aripiprazole/chemistry , Crystallography, X-Ray , Cyclic AMP/chemistry , Drug Design , Drug Discovery , HEK293 Cells , Humans , Hydrogen Bonding , Indoles/chemistry , Kinetics , Molecular Dynamics Simulation , Mutation , Protein Binding , Protein Conformation , Serine/chemistry , Signal Transduction
20.
Nat Commun ; 8: 14335, 2017 02 09.
Article in English | MEDLINE | ID: mdl-28181498

ABSTRACT

Acute hormone secretion triggered by G protein-coupled receptor (GPCR) activation underlies many fundamental physiological processes. GPCR signalling is negatively regulated by ß-arrestins, adaptor molecules that also activate different intracellular signalling pathways. Here we reveal that TRV120027, a ß-arrestin-1-biased agonist of the angiotensin II receptor type 1 (AT1R), stimulates acute catecholamine secretion through coupling with the transient receptor potential cation channel subfamily C 3 (TRPC3). We show that TRV120027 promotes the recruitment of TRPC3 or phosphoinositide-specific phospholipase C (PLCγ) to the AT1R-ß-arrestin-1 signalling complex. Replacing the C-terminal region of ß-arrestin-1 with its counterpart on ß-arrestin-2 or using a specific TAT-P1 peptide to block the interaction between ß-arrestin-1 and PLCγ abolishes TRV120027-induced TRPC3 activation. Taken together, our results show that the GPCR-arrestin complex initiates non-desensitized signalling at the plasma membrane by coupling with ion channels. This fast communication pathway might be a common mechanism of several cellular processes.


Subject(s)
Catecholamines/metabolism , Receptor, Angiotensin, Type 1/agonists , TRPC Cation Channels/metabolism , beta-Arrestin 1/metabolism , beta-Arrestin 2/metabolism , Animals , Calcium/metabolism , Estrenes/pharmacology , HEK293 Cells , Humans , Ligands , Mice, Knockout , Oligopeptides/pharmacology , Phospholipase C gamma/metabolism , Pyrrolidinones/pharmacology , Receptor, Angiotensin, Type 1/metabolism , Signal Transduction/drug effects , beta-Arrestin 1/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...