Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
1.
J Phys Chem B ; 128(9): 2124-2133, 2024 Mar 07.
Article in English | MEDLINE | ID: mdl-38391238

ABSTRACT

G-protein-coupled receptors (GPCRs) are structurally flexible membrane proteins that mediate a host of physiological responses to extracellular ligands like hormones and neurotransmitters. Fine features of their dynamic structural behavior are hypothesized to encode the functional plasticity seen in GPCR activity, where ligands with different efficacies can direct the same receptor toward different signaling phenotypes. Although the number of GPCR crystal structures is increasing, the receptors are characterized by complex and poorly understood conformational landscapes. Therefore, we employed a fluorescence microscopy assay to monitor conformational dynamics of single ß2 adrenergic receptors (ß2ARs). To increase the biological relevance of our findings, we decided not to reconstitute the receptor in detergent micelles but rather lipid membranes as proteoliposomes. The conformational dynamics were monitored by changes in the intensity of an environmentally sensitive boron-dipyrromethene (BODIPY 493/503) fluorophore conjugated to an endogenous cysteine (located at the cytoplasmic end of the sixth transmembrane helix of the receptor). Using total internal reflection fluorescence microscopy (TIRFM) and a single small unilamellar liposome assay that we previously developed, we followed the real-time dynamic properties of hundreds of single ß2ARs reconstituted in a native-like environment─lipid membranes. Our results showed that ß2AR-BODIPY fluctuates between several states of different intensity on a time scale of seconds, compared to BODIPY-lipid conjugates that show almost entirely stable fluorescence emission in the absence and presence of the full agonist BI-167107. Agonist stimulation changes the ß2AR dynamics, increasing the population of states with higher intensities and prolonging their durations, consistent with bulk experiments. The transition density plot demonstrates that ß2AR-BODIPY, in the absence of the full agonist, interconverts between states of low and moderate intensity, while the full agonist renders transitions between moderate and high-intensity states more probable. This redistribution is consistent with a mechanism of conformational selection and is a promising first step toward characterizing the conformational dynamics of GPCRs embedded in a lipid bilayer.


Subject(s)
Boron Compounds , Lipids , Receptors, G-Protein-Coupled , Receptors, G-Protein-Coupled/chemistry , Molecular Conformation , Receptors, Adrenergic , Receptors, Adrenergic, beta-2/chemistry , Ligands
2.
Sci Adv ; 8(28): eabn4331, 2022 07 15.
Article in English | MEDLINE | ID: mdl-35857505

ABSTRACT

Zinc is essential for all organisms and yet detrimental at elevated levels. Hence, homeostasis of this metal is tightly regulated. The Zrt/Irt-like proteins (ZIPs) represent the only zinc importers in metazoans. Mutations in human ZIPs cause serious disorders, but the mechanism by which ZIPs transfer zinc remains elusive. Hitherto, structural information is only available for a model member, BbZIP, and as a single, ion-bound conformation, precluding mechanistic insights. Here, we elucidate an inward-open metal-free BbZIP structure, differing substantially in the relative positions of the two separate domains of ZIPs. With accompanying coevolutional analyses, mutagenesis, and uptake assays, the data point to an elevator-type transport mechanism, likely shared within the ZIP family, unifying earlier functional data. Moreover, the structure reveals a previously unknown ninth transmembrane segment that is important for activity in vivo. Our findings outline the mechanistic principles governing ZIP-protein transport and enhance the molecular understanding of ZIP-related disorders.


Subject(s)
Cation Transport Proteins , Zinc , Biological Transport , Cation Transport Proteins/metabolism , Humans , Ion Transport , Metals/metabolism , Zinc/metabolism
3.
Nat Commun ; 10(1): 2714, 2019 06 20.
Article in English | MEDLINE | ID: mdl-31221956

ABSTRACT

The dopamine transporter is a member of the neurotransmitter:sodium symporters (NSSs), which are responsible for termination of neurotransmission through Na+-driven reuptake of neurotransmitter from the extracellular space. Experimental evidence elucidating the coordinated conformational rearrangements related to the transport mechanism has so far been limited. Here we probe the global Na+- and dopamine-induced conformational dynamics of the wild-type Drosophila melanogaster dopamine transporter using hydrogen-deuterium exchange mass spectrometry. We identify Na+- and dopamine-induced changes in specific regions of the transporter, suggesting their involvement in protein conformational transitions. Furthermore, we detect ligand-dependent slow cooperative fluctuations of helical stretches in several domains of the transporter, which could be a molecular mechanism that assists in the transporter function. Our results provide a framework for understanding the molecular mechanism underlying the function of NSSs by revealing detailed insight into the state-dependent conformational changes associated with the alternating access model of the dopamine transporter.


Subject(s)
Dopamine Plasma Membrane Transport Proteins/metabolism , Dopamine/metabolism , Drosophila Proteins/metabolism , Molecular Dynamics Simulation , Sodium/metabolism , Dopamine Plasma Membrane Transport Proteins/chemistry , Dopamine Plasma Membrane Transport Proteins/isolation & purification , Drosophila Proteins/chemistry , Drosophila Proteins/isolation & purification , HEK293 Cells , Humans , Ligands , Mass Spectrometry , Protein Conformation, alpha-Helical/physiology , Protein Domains/physiology , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism
4.
Cell ; 177(5): 1232-1242.e11, 2019 05 16.
Article in English | MEDLINE | ID: mdl-31080064

ABSTRACT

The activation of G proteins by G protein-coupled receptors (GPCRs) underlies the majority of transmembrane signaling by hormones and neurotransmitters. Recent structures of GPCR-G protein complexes obtained by crystallography and cryoelectron microscopy (cryo-EM) reveal similar interactions between GPCRs and the alpha subunit of different G protein isoforms. While some G protein subtype-specific differences are observed, there is no clear structural explanation for G protein subtype-selectivity. All of these complexes are stabilized in the nucleotide-free state, a condition that does not exist in living cells. In an effort to better understand the structural basis of coupling specificity, we used time-resolved structural mass spectrometry techniques to investigate GPCR-G protein complex formation and G-protein activation. Our results suggest that coupling specificity is determined by one or more transient intermediate states that serve as selectivity filters and precede the formation of the stable nucleotide-free GPCR-G protein complexes observed in crystal and cryo-EM structures.


Subject(s)
GTP-Binding Proteins/chemistry , Multienzyme Complexes/chemistry , Receptors, G-Protein-Coupled/chemistry , Animals , Cattle , Cryoelectron Microscopy , Crystallography, X-Ray , Humans , Multienzyme Complexes/ultrastructure , Protein Structure, Quaternary , Rats
5.
Nat Commun ; 10(1): 1687, 2019 04 11.
Article in English | MEDLINE | ID: mdl-30976000

ABSTRACT

The serotonin transporter (SERT), a member of the neurotransmitter:sodium symporter family, is responsible for termination of serotonergic signaling by re-uptake of serotonin (5-HT) into the presynaptic neuron. Its key role in synaptic transmission makes it a major drug target, e.g. for the treatment of depression, anxiety and post-traumatic stress. Here, we apply hydrogen-deuterium exchange mass spectrometry to probe the conformational dynamics of human SERT in the absence and presence of known substrates and targeted drugs. Our results reveal significant changes in dynamics in regions TM1, EL3, EL4, and TM12 upon binding co-transported ions (Na+/K+) and ligand-mediated changes in TM1, EL3 and EL4 upon binding 5-HT, the drugs S-citalopram, cocaine and ibogaine. Our results provide a comprehensive direct view of the conformational response of SERT upon binding both biologically relevant substrate/ions and ligands of pharmaceutical interest, thus advancing our understanding of the structure-function relationship in SERT.


Subject(s)
Protein Domains , Serotonin Plasma Membrane Transport Proteins/chemistry , Cations, Monovalent/metabolism , Citalopram/chemistry , Citalopram/metabolism , Cocaine/chemistry , Cocaine/metabolism , Ibogaine/chemistry , Ibogaine/metabolism , Ligands , Mass Spectrometry , Molecular Dynamics Simulation , Potassium/metabolism , Protein Binding , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Serotonin/chemistry , Serotonin/metabolism , Serotonin Plasma Membrane Transport Proteins/isolation & purification , Serotonin Plasma Membrane Transport Proteins/metabolism , Sodium/metabolism , Structure-Activity Relationship
6.
Biophys J ; 115(2): 300-312, 2018 07 17.
Article in English | MEDLINE | ID: mdl-30021106

ABSTRACT

G-protein-coupled receptors (GPCRs) control vital cellular signaling pathways. GPCR oligomerization is proposed to increase signaling diversity. However, many reports have arrived at disparate conclusions regarding the existence, stability, and stoichiometry of GPCR oligomers, partly because of cellular complexity and ensemble averaging of intrareconstitution heterogeneities that complicate the interpretation of oligomerization data. To overcome these limitations, we exploited fluorescence-microscopy-based high-content analysis of single proteoliposomes. This allowed multidimensional quantification of intrinsic monomer-monomer interactions of three class A GPCRs (ß2-adrenergic receptor, cannabinoid receptor type 1, and opsin). Using a billion-fold less protein than conventional assays, we quantified oligomer stoichiometries, association constants, and the influence of two ligands and membrane curvature on oligomerization, revealing key similarities and differences for three GPCRs with decidedly different physiological functions. The assays introduced here will assist with the quantitative experimental observation of oligomerization for transmembrane proteins in general.


Subject(s)
Protein Multimerization , Proteolipids/metabolism , Receptors, G-Protein-Coupled/chemistry , Receptors, G-Protein-Coupled/metabolism , Ligands , Protein Structure, Quaternary , Signal Transduction , Solubility
7.
Nat Struct Mol Biol ; 25(3): 289-296, 2018 03.
Article in English | MEDLINE | ID: mdl-29434346

ABSTRACT

Camelid single-domain antibody fragments ('nanobodies') provide the remarkable specificity of antibodies within a single 15-kDa immunoglobulin VHH domain. This unique feature has enabled applications ranging from use as biochemical tools to therapeutic agents. Nanobodies have emerged as especially useful tools in protein structural biology, facilitating studies of conformationally dynamic proteins such as G-protein-coupled receptors (GPCRs). Nearly all nanobodies available to date have been obtained by animal immunization, a bottleneck restricting many applications of this technology. To solve this problem, we report a fully in vitro platform for nanobody discovery based on yeast surface display. We provide a blueprint for identifying nanobodies, demonstrate the utility of the library by crystallizing a nanobody with its antigen, and most importantly, we utilize the platform to discover conformationally selective nanobodies to two distinct human GPCRs. To facilitate broad deployment of this platform, the library and associated protocols are freely available for nonprofit research.


Subject(s)
Cell Surface Display Techniques , Receptors, G-Protein-Coupled/immunology , Single-Domain Antibodies/immunology , Antibody Specificity , Antigens/chemistry , Antigens/immunology , Cell Separation , Flow Cytometry , Humans , Protein Conformation , Receptors, G-Protein-Coupled/chemistry , Single-Domain Antibodies/chemistry , Yeasts/genetics
8.
RSC Adv ; 8(4): 2219-2228, 2018 Jan 05.
Article in English | MEDLINE | ID: mdl-35542596

ABSTRACT

A series of Gs protein peptidomimetics were designed and synthesised based on the published X-ray crystal structure of the active state ß2-adrenergic receptor (ß2AR) in complex with the Gs protein (PDB 3SN6). We hypothesised that such peptidomimetics may function as allosteric modulators that target the intracellular Gs protein binding site of the ß2AR. Peptidomimetics were designed to mimic the 15 residue C-terminal α-helix of the Gs protein and were pre-organised in a helical conformation by (i, i + 4)-stapling using copper catalysed azide alkyne cycloaddition. Linear and stapled peptidomimetics were analysed by circular dichroism (CD) and characterised in a membrane-based cAMP accumulation assay and in a bimane fluorescence assay on purified ß2AR. Several peptidomimetics inhibited agonist isoproterenol (ISO) induced cAMP formation by lowering the ISO maximal efficacy up to 61%. Moreover, some peptidomimetics were found to significantly decrease the potency of ISO up to 39-fold. In the bimane fluorescence assay none of the tested peptidomimetics could stabilise an active-like conformation of ß2AR. Overall, the obtained pharmacological data suggest that some of the peptidomimetics may be able to compete with the native Gs protein for the intracellular binding site to block ISO-induced cAMP formation, but are unable to stabilise an active-like receptor conformation.

9.
Nature ; 535(7610): 182-6, 2016 07 07.
Article in English | MEDLINE | ID: mdl-27362234

ABSTRACT

G-protein-coupled receptors (GPCRs) remain the primary conduit by which cells detect environmental stimuli and communicate with each other. Upon activation by extracellular agonists, these seven-transmembrane-domain-containing receptors interact with heterotrimeric G proteins to regulate downstream second messenger and/or protein kinase cascades. Crystallographic evidence from a prototypic GPCR, the ß2-adrenergic receptor (ß2AR), in complex with its cognate G protein, Gs, has provided a model for how agonist binding promotes conformational changes that propagate through the GPCR and into the nucleotide-binding pocket of the G protein α-subunit to catalyse GDP release, the key step required for GTP binding and activation of G proteins. The structure also offers hints about how G-protein binding may, in turn, allosterically influence ligand binding. Here we provide functional evidence that G-protein coupling to the ß2AR stabilizes a 'closed' receptor conformation characterized by restricted access to and egress from the hormone-binding site. Surprisingly, the effects of G protein on the hormone-binding site can be observed in the absence of a bound agonist, where G-protein coupling driven by basal receptor activity impedes the association of agonists, partial agonists, antagonists and inverse agonists. The ability of bound ligands to dissociate from the receptor is also hindered, providing a structural explanation for the G-protein-mediated enhancement of agonist affinity, which has been observed for many GPCR­G-protein pairs. Our data also indicate that, in contrast to agonist binding alone, coupling of a G protein in the absence of an agonist stabilizes large structural changes in a GPCR. The effects of nucleotide-free G protein on ligand-binding kinetics are shared by other members of the superfamily of GPCRs, suggesting that a common mechanism may underlie G-protein-mediated enhancement of agonist affinity.


Subject(s)
Allosteric Site , GTP-Binding Protein alpha Subunits, Gs/metabolism , Receptors, Adrenergic, beta-2/chemistry , Receptors, Adrenergic, beta-2/metabolism , Adrenergic beta-2 Receptor Agonists/metabolism , Adrenergic beta-2 Receptor Antagonists/metabolism , Allosteric Regulation/drug effects , Allosteric Site/drug effects , GTP-Binding Protein alpha Subunits, Gs/pharmacology , Guanine/metabolism , Guanine/pharmacology , Humans , Kinetics , Ligands , Models, Molecular , Protein Binding/drug effects , Protein Conformation/drug effects , Receptors, Adrenergic, beta-2/immunology , Single-Chain Antibodies/immunology , Single-Chain Antibodies/pharmacology
10.
Nat Methods ; 11(9): 931-4, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25086504

ABSTRACT

Proteoliposome reconstitution is a standard method to stabilize purified transmembrane proteins in membranes for structural and functional assays. Here we quantified intrareconstitution heterogeneities in single proteoliposomes using fluorescence microscopy. Our results suggest that compositional heterogeneities can severely skew ensemble-average proteoliposome measurements but also enable ultraminiaturized high-content screens. We took advantage of this screening capability to map the oligomerization energy of the ß2-adrenergic receptor using ∼10(9)-fold less protein than conventional assays.


Subject(s)
Algorithms , Image Interpretation, Computer-Assisted/methods , Microscopy, Fluorescence/methods , Proteolipids/chemistry , Spectrometry, Fluorescence/methods , Nanotechnology/methods , Receptors, G-Protein-Coupled/analysis , Receptors, G-Protein-Coupled/chemistry
11.
Nat Protoc ; 9(3): 674-93, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24577359

ABSTRACT

There is growing interest in using antibodies as auxiliary tools to crystallize proteins. Here we describe a general protocol for the generation of Nanobodies to be used as crystallization chaperones for the structural investigation of diverse conformational states of flexible (membrane) proteins and complexes thereof. Our technology has a competitive advantage over other recombinant crystallization chaperones in that we fully exploit the natural humoral response against native antigens. Accordingly, we provide detailed protocols for the immunization with native proteins and for the selection by phage display of in vivo-matured Nanobodies that bind conformational epitopes of functional proteins. Three representative examples illustrate that the outlined procedures are robust, making it possible to solve by Nanobody-assisted X-ray crystallography in a time span of 6-12 months.


Subject(s)
Biotechnology/methods , Crystallization/methods , Models, Molecular , Protein Conformation , Single-Domain Antibodies/biosynthesis , Animals , Camelus , Cell Surface Display Techniques , Cloning, Molecular , DNA Primers/genetics
12.
Mol Pharmacol ; 85(3): 472-81, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24319111

ABSTRACT

The biologic activity induced by ligand binding to orthosteric or allosteric sites on a G protein-coupled receptor (GPCR) is mediated by stabilization of specific receptor conformations. In the case of the ß2 adrenergic receptor, these ligands are generally small-molecule agonists or antagonists. However, a monomeric single-domain antibody (nanobody) from the Camelid family was recently found to allosterically bind and stabilize an active conformation of the ß2-adrenergic receptor (ß2AR). Here, we set out to study the functional interaction of 18 related nanobodies with the ß2AR to investigate their roles as novel tools for studying GPCR biology. Our studies revealed several sequence-related nanobody families with preferences for active (agonist-occupied) or inactive (antagonist-occupied) receptors. Flow cytometry analysis indicates that all nanobodies bind to epitopes displayed on the intracellular receptor surface; therefore, we transiently expressed them intracellularly as "intrabodies" to test their effects on ß2AR-dependent signaling. Conformational specificity was preserved after intrabody conversion as demonstrated by the ability for the intracellularly expressed nanobodies to selectively bind agonist- or antagonist-occupied receptors. When expressed as intrabodies, they inhibited G protein activation (cyclic AMP accumulation), G protein-coupled receptor kinase (GRK)-mediated receptor phosphorylation, ß-arrestin recruitment, and receptor internalization to varying extents. These functional effects were likely due to either steric blockade of downstream effector (Gs, ß-arrestin, GRK) interactions or stabilization of specific receptor conformations which do not support effector coupling. Together, these findings strongly implicate nanobody-derived intrabodies as novel tools to study GPCR biology.


Subject(s)
Receptors, Adrenergic, beta-2/metabolism , Single-Domain Antibodies/metabolism , Amino Acid Sequence , Cell Line , Cyclic AMP/metabolism , G-Protein-Coupled Receptor Kinases/metabolism , HEK293 Cells , Humans , Molecular Sequence Data , Phosphorylation/physiology , Protein Binding/physiology , Receptors, G-Protein-Coupled/metabolism , Sequence Alignment
13.
Chemistry ; 19(46): 15645-51, 2013 Nov 11.
Article in English | MEDLINE | ID: mdl-24123610

ABSTRACT

Integral membrane proteins play central roles in controlling the flow of information and molecules across membranes. Our understanding of membrane protein structures and functions, however, is seriously limited, mainly due to difficulties in handling and analysing these proteins in aqueous solution. The use of a detergent or other amphipathic agents is required to overcome the intrinsic incompatibility between the large lipophilic surfaces displayed by the membrane proteins in their native forms and the polar solvent molecules. Here, we introduce new tripod amphiphiles displaying favourable behaviours toward several membrane protein systems, leading to an enhanced protein solubilisation and stabilisation compared to both conventional detergents and previously described tripod amphiphiles.


Subject(s)
Detergents/chemistry , Membrane Proteins/analysis , Solvents/chemistry , Surface-Active Agents/chemistry , Hydrophobic and Hydrophilic Interactions , Membrane Proteins/chemistry , Solubility
14.
Nature ; 495(7442): 534-8, 2013 Mar 28.
Article in English | MEDLINE | ID: mdl-23515162

ABSTRACT

A long-held tenet of molecular pharmacology is that canonical signal transduction mediated by G-protein-coupled receptor (GPCR) coupling to heterotrimeric G proteins is confined to the plasma membrane. Evidence supporting this traditional view is based on analytical methods that provide limited or no subcellular resolution. It has been subsequently proposed that signalling by internalized GPCRs is restricted to G-protein-independent mechanisms such as scaffolding by arrestins, or GPCR activation elicits a discrete form of persistent G protein signalling, or that internalized GPCRs can indeed contribute to the acute G-protein-mediated response. Evidence supporting these various latter hypotheses is indirect or subject to alternative interpretation, and it remains unknown if endosome-localized GPCRs are even present in an active form. Here we describe the application of conformation-specific single-domain antibodies (nanobodies) to directly probe activation of the ß2-adrenoceptor, a prototypical GPCR, and its cognate G protein, Gs (ref. 12), in living mammalian cells. We show that the adrenergic agonist isoprenaline promotes receptor and G protein activation in the plasma membrane as expected, but also in the early endosome membrane, and that internalized receptors contribute to the overall cellular cyclic AMP response within several minutes after agonist application. These findings provide direct support for the hypothesis that canonical GPCR signalling occurs from endosomes as well as the plasma membrane, and suggest a versatile strategy for probing dynamic conformational change in vivo.


Subject(s)
Biosensing Techniques/methods , Endosomes/metabolism , Receptors, Adrenergic, beta-2/chemistry , Receptors, Adrenergic, beta-2/metabolism , Signal Transduction , Adrenergic beta-2 Receptor Agonists/pharmacology , Cell Membrane/chemistry , Cell Membrane/metabolism , Clathrin-Coated Vesicles , Cyclic AMP/metabolism , Endocytosis , Endosomes/chemistry , GTP-Binding Protein alpha Subunits, Gs/metabolism , Green Fluorescent Proteins/analysis , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HEK293 Cells , Humans , Isoproterenol/pharmacology , Models, Biological , Protein Conformation , Receptors, Adrenergic, beta-2/immunology , Single-Domain Antibodies/genetics , Single-Domain Antibodies/immunology
15.
Chem Commun (Camb) ; 49(23): 2287-9, 2013 Mar 21.
Article in English | MEDLINE | ID: mdl-23165475

ABSTRACT

The development of a new class of surfactants for membrane protein manipulation, "GNG amphiphiles", is reported. These amphiphiles display promising behavior for membrane proteins, as demonstrated recently by the high resolution structure of a sodium-pumping pyrophosphatase reported by Kellosalo et al. (Science, 2012, 337, 473).


Subject(s)
Glucose/chemistry , Glycols/chemistry , Membrane Proteins/chemistry , Detergents/chemistry , Membrane Proteins/metabolism , Protein Stability , Rhodobacter capsulatus/metabolism
16.
Proc Natl Acad Sci U S A ; 109(50): E3463-72, 2012 Dec 11.
Article in English | MEDLINE | ID: mdl-23151510

ABSTRACT

The steroid cholesterol is an essential component of eukaryotic membranes, and it functionally modulates membrane proteins, including G protein-coupled receptors. To reveal insight into how cholesterol modulates G protein-coupled receptors, we have used dynamic single-molecule force spectroscopy to quantify the mechanical strength and flexibility, conformational variability, and kinetic and energetic stability of structural segments stabilizing the human ß(2)-adrenergic receptor (ß(2)AR) in the absence and presence of the cholesterol analog cholesteryl hemisuccinate (CHS). CHS considerably increased the kinetic, energetic, and mechanical stability of almost every structural segment at sufficient magnitude to alter the structure and functional relationship of ß(2)AR. One exception was the structural core segment of ß(2)AR, which establishes multiple ligand binding sites, and its properties were not significantly influenced by CHS.


Subject(s)
Cholesterol Esters/metabolism , Receptors, Adrenergic, beta-2/chemistry , Receptors, Adrenergic, beta-2/metabolism , Animals , Biomechanical Phenomena , Cholesterol Esters/chemistry , Energy Metabolism , Humans , Kinetics , Microscopy, Atomic Force , Models, Molecular , Protein Conformation , Protein Interaction Mapping , Protein Stability , Proteolipids/chemistry , Proteolipids/metabolism , Receptors, Adrenergic, beta-2/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sf9 Cells , Spodoptera , Unfolded Protein Response
18.
Proc Natl Acad Sci U S A ; 108(38): 16086-91, 2011 Sep 20.
Article in English | MEDLINE | ID: mdl-21914848

ABSTRACT

The active-state complex between an agonist-bound receptor and a guanine nucleotide-free G protein represents the fundamental signaling assembly for the majority of hormone and neurotransmitter signaling. We applied single-particle electron microscopy (EM) analysis to examine the architecture of agonist-occupied ß(2)-adrenoceptor (ß(2)AR) in complex with the heterotrimeric G protein Gs (Gαsßγ). EM 2D averages and 3D reconstructions of the detergent-solubilized complex reveal an overall architecture that is in very good agreement with the crystal structure of the active-state ternary complex. Strikingly however, the α-helical domain of Gαs appears highly flexible in the absence of nucleotide. In contrast, the presence of the pyrophosphate mimic foscarnet (phosphonoformate), and also the presence of GDP, favor the stabilization of the α-helical domain on the Ras-like domain of Gαs. Molecular modeling of the α-helical domain in the 3D EM maps suggests that in its stabilized form it assumes a conformation reminiscent to the one observed in the crystal structure of Gαs-GTPγS. These data argue that the α-helical domain undergoes a nucleotide-dependent transition from a flexible to a conformationally stabilized state.


Subject(s)
Crystallography, X-Ray , GTP-Binding Protein alpha Subunits, Gs/chemistry , Protein Structure, Secondary , Protein Structure, Tertiary , Receptors, Adrenergic, beta-2/chemistry , Animals , Crystallization , GTP-Binding Protein alpha Subunits, Gs/metabolism , GTP-Binding Protein alpha Subunits, Gs/ultrastructure , Guanosine 5'-O-(3-Thiotriphosphate)/chemistry , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Guanosine Diphosphate/chemistry , Guanosine Diphosphate/metabolism , Guanosine Triphosphate/chemistry , Guanosine Triphosphate/metabolism , Microscopy, Electron , Models, Molecular , Protein Binding , Protein Conformation , Receptors, Adrenergic, beta-2/metabolism , Receptors, Adrenergic, beta-2/ultrastructure
19.
Nature ; 477(7366): 611-5, 2011 Sep 28.
Article in English | MEDLINE | ID: mdl-21956331

ABSTRACT

G protein-coupled receptors represent the largest family of membrane receptors that instigate signalling through nucleotide exchange on heterotrimeric G proteins. Nucleotide exchange, or more precisely, GDP dissociation from the G protein α-subunit, is the key step towards G protein activation and initiation of downstream signalling cascades. Despite a wealth of biochemical and biophysical studies on inactive and active conformations of several heterotrimeric G proteins, the molecular underpinnings of G protein activation remain elusive. To characterize this mechanism, we applied peptide amide hydrogen-deuterium exchange mass spectrometry to probe changes in the structure of the heterotrimeric bovine G protein, Gs (the stimulatory G protein for adenylyl cyclase) on formation of a complex with agonist-bound human ß(2) adrenergic receptor (ß(2)AR). Here we report structural links between the receptor-binding surface and the nucleotide-binding pocket of Gs that undergo higher levels of hydrogen-deuterium exchange than would be predicted from the crystal structure of the ß(2)AR-Gs complex. Together with X-ray crystallographic and electron microscopic data of the ß(2)AR-Gs complex (from refs 2, 3), we provide a rationale for a mechanism of nucleotide exchange, whereby the receptor perturbs the structure of the amino-terminal region of the α-subunit of Gs and consequently alters the 'P-loop' that binds the ß-phosphate in GDP. As with the Ras family of small-molecular-weight G proteins, P-loop stabilization and ß-phosphate coordination are key determinants of GDP (and GTP) binding affinity.


Subject(s)
GTP-Binding Protein alpha Subunits, Gs/chemistry , GTP-Binding Protein alpha Subunits, Gs/metabolism , Receptors, Adrenergic, beta-2/chemistry , Receptors, Adrenergic, beta-2/metabolism , Adrenergic beta-2 Receptor Agonists/chemistry , Adrenergic beta-2 Receptor Agonists/metabolism , Animals , Biocatalysis , Catalytic Domain , Cattle , Crystallography, X-Ray , Deuterium Exchange Measurement , GTP-Binding Protein alpha Subunits, Gs/ultrastructure , Guanosine Diphosphate/metabolism , Guanosine Triphosphate/metabolism , Humans , Models, Molecular , Protein Binding , Protein Conformation , Receptors, Adrenergic, beta-2/ultrastructure
20.
Nature ; 477(7366): 549-55, 2011 Jul 19.
Article in English | MEDLINE | ID: mdl-21772288

ABSTRACT

G protein-coupled receptors (GPCRs) are responsible for the majority of cellular responses to hormones and neurotransmitters as well as the senses of sight, olfaction and taste. The paradigm of GPCR signalling is the activation of a heterotrimeric GTP binding protein (G protein) by an agonist-occupied receptor. The ß(2) adrenergic receptor (ß(2)AR) activation of Gs, the stimulatory G protein for adenylyl cyclase, has long been a model system for GPCR signalling. Here we present the crystal structure of the active state ternary complex composed of agonist-occupied monomeric ß(2)AR and nucleotide-free Gs heterotrimer. The principal interactions between the ß(2)AR and Gs involve the amino- and carboxy-terminal α-helices of Gs, with conformational changes propagating to the nucleotide-binding pocket. The largest conformational changes in the ß(2)AR include a 14 Å outward movement at the cytoplasmic end of transmembrane segment 6 (TM6) and an α-helical extension of the cytoplasmic end of TM5. The most surprising observation is a major displacement of the α-helical domain of Gαs relative to the Ras-like GTPase domain. This crystal structure represents the first high-resolution view of transmembrane signalling by a GPCR.


Subject(s)
GTP-Binding Protein alpha Subunits, Gs/chemistry , GTP-Binding Protein alpha Subunits, Gs/metabolism , Receptors, Adrenergic, beta-2/chemistry , Receptors, Adrenergic, beta-2/metabolism , Adrenergic beta-2 Receptor Agonists/chemistry , Adrenergic beta-2 Receptor Agonists/metabolism , Animals , Catalytic Domain , Cattle , Crystallization , Crystallography, X-Ray , Enzyme Activation , Models, Molecular , Multiprotein Complexes/chemistry , Multiprotein Complexes/metabolism , Protein Binding , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...