Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 124
Filter
1.
Cell ; 184(4): 983-999.e24, 2021 02 18.
Article in English | MEDLINE | ID: mdl-33606986

ABSTRACT

Interleukin-12 (IL-12) and IL-23 are heterodimeric cytokines that are produced by antigen-presenting cells to regulate the activation and differentiation of lymphocytes, and they share IL-12Rß1 as a receptor signaling subunit. We present a crystal structure of the quaternary IL-23 (IL-23p19/p40)/IL-23R/IL-12Rß1 complex, together with cryoelectron microscopy (cryo-EM) maps of the complete IL-12 (IL-12p35/p40)/IL-12Rß2/IL-12Rß1 and IL-23 receptor (IL-23R) complexes, which reveal "non-canonical" topologies where IL-12Rß1 directly engages the common p40 subunit. We targeted the shared IL-12Rß1/p40 interface to design a panel of IL-12 partial agonists that preserved interferon gamma (IFNγ) induction by CD8+ T cells but impaired cytokine production from natural killer (NK) cells in vitro. These cell-biased properties were recapitulated in vivo, where IL-12 partial agonists elicited anti-tumor immunity to MC-38 murine adenocarcinoma absent the NK-cell-mediated toxicity seen with wild-type IL-12. Thus, the structural mechanism of receptor sharing used by IL-12 family cytokines provides a protein interface blueprint for tuning this cytokine axis for therapeutics.


Subject(s)
Interleukin-12/chemistry , Interleukin-12/metabolism , Killer Cells, Natural/metabolism , Receptors, Interleukin/chemistry , Receptors, Interleukin/metabolism , T-Lymphocytes/metabolism , Amino Acid Sequence , Animals , Binding Sites , Cryoelectron Microscopy , Crystallography, X-Ray , Epitopes/immunology , Female , HEK293 Cells , Humans , Immunity , Interleukin-12/agonists , Interleukin-12 Subunit p40/chemistry , Interleukin-12 Subunit p40/metabolism , Mice, Inbred C57BL , Models, Molecular , Neoplasms/immunology , Neoplasms/pathology , Protein Structure, Quaternary , Receptors, Interleukin/ultrastructure , Receptors, Interleukin-12/metabolism , Signal Transduction , Structure-Activity Relationship
2.
Cell ; 182(6): 1574-1588.e19, 2020 09 17.
Article in English | MEDLINE | ID: mdl-32946782

ABSTRACT

Hallucinogens like lysergic acid diethylamide (LSD), psilocybin, and substituted N-benzyl phenylalkylamines are widely used recreationally with psilocybin being considered as a therapeutic for many neuropsychiatric disorders including depression, anxiety, and substance abuse. How psychedelics mediate their actions-both therapeutic and hallucinogenic-are not understood, although activation of the 5-HT2A serotonin receptor (HTR2A) is key. To gain molecular insights into psychedelic actions, we determined the active-state structure of HTR2A bound to 25-CN-NBOH-a prototypical hallucinogen-in complex with an engineered Gαq heterotrimer by cryoelectron microscopy (cryo-EM). We also obtained the X-ray crystal structures of HTR2A complexed with the arrestin-biased ligand LSD or the inverse agonist methiothepin. Comparisons of these structures reveal determinants responsible for HTR2A-Gαq protein interactions as well as the conformational rearrangements involved in active-state transitions. Given the potential therapeutic actions of hallucinogens, these findings could accelerate the discovery of more selective drugs for the treatment of a variety of neuropsychiatric disorders.


Subject(s)
GTP-Binding Protein alpha Subunits, Gq-G11/chemistry , Hallucinogens/chemistry , Receptor, Serotonin, 5-HT2A/chemistry , Receptor, Serotonin, 5-HT2A/metabolism , Animals , Cryoelectron Microscopy , Crystallography, X-Ray , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Gene Expression , HEK293 Cells , Hallucinogens/pharmacology , Hallucinogens/therapeutic use , Humans , Ligands , Lysergic Acid Diethylamide/chemistry , Lysergic Acid Diethylamide/pharmacology , Methiothepin/chemistry , Methiothepin/metabolism , Models, Chemical , Mutation , Protein Conformation, alpha-Helical , Receptor, Serotonin, 5-HT2A/genetics , Recombinant Proteins , Serotonin/metabolism , Spodoptera
3.
Cell ; 176(3): 448-458.e12, 2019 01 24.
Article in English | MEDLINE | ID: mdl-30639101

ABSTRACT

Cannabis elicits its mood-enhancing and analgesic effects through the cannabinoid receptor 1 (CB1), a G protein-coupled receptor (GPCR) that signals primarily through the adenylyl cyclase-inhibiting heterotrimeric G protein Gi. Activation of CB1-Gi signaling pathways holds potential for treating a number of neurological disorders and is thus crucial to understand the mechanism of Gi activation by CB1. Here, we present the structure of the CB1-Gi signaling complex bound to the highly potent agonist MDMB-Fubinaca (FUB), a recently emerged illicit synthetic cannabinoid infused in street drugs that have been associated with numerous overdoses and fatalities. The structure illustrates how FUB stabilizes the receptor in an active state to facilitate nucleotide exchange in Gi. The results compose the structural framework to explain CB1 activation by different classes of ligands and provide insights into the G protein coupling and selectivity mechanisms adopted by the receptor.


Subject(s)
Receptor, Cannabinoid, CB1/metabolism , Receptor, Cannabinoid, CB1/ultrastructure , Animals , Cannabinoid Receptor Agonists/pharmacology , Cannabinoids/pharmacology , Cryoelectron Microscopy/methods , Heterotrimeric GTP-Binding Proteins/metabolism , Humans , Indazoles/pharmacology , Ligands , Protein Binding , Receptor, Cannabinoid, CB1/chemistry , Receptors, Cannabinoid/chemistry , Receptors, Cannabinoid/metabolism , Receptors, Cannabinoid/ultrastructure , Receptors, G-Protein-Coupled/metabolism , Sf9 Cells , Signal Transduction/drug effects
4.
Cell ; 174(5): 1117-1126.e12, 2018 08 23.
Article in English | MEDLINE | ID: mdl-30100186

ABSTRACT

The methylation of histone 3 lysine 4 (H3K4) is carried out by an evolutionarily conserved family of methyltransferases referred to as complex of proteins associated with Set1 (COMPASS). The activity of the catalytic SET domain (su(var)3-9, enhancer-of-zeste, and trithorax) is endowed through forming a complex with a set of core proteins that are widely shared from yeast to humans. We obtained cryo-electron microscopy (cryo-EM) maps of the yeast Set1/COMPASS core complex at overall 4.0- to 4.4-Å resolution, providing insights into its structural organization and conformational dynamics. The Cps50 C-terminal tail weaves within the complex to provide a central scaffold for assembly. The SET domain, snugly positioned at the junction of the Y-shaped complex, is extensively contacted by Cps60 (Bre2), Cps50 (Swd1), and Cps30 (Swd3). The mobile SET-I motif of the SET domain is engaged by Cps30, explaining its key role in COMPASS catalytic activity toward higher H3K4 methylation states.


Subject(s)
Fungal Proteins/chemistry , Histone Methyltransferases/chemistry , Histones/chemistry , Animals , Catalytic Domain , Chaetomium/chemistry , Chromatin/chemistry , Cryoelectron Microscopy , DNA-Binding Proteins/chemistry , Epigenesis, Genetic , Histone-Lysine N-Methyltransferase/chemistry , Humans , Insecta , Intracellular Signaling Peptides and Proteins , Methylation , Protein Subunits , Saccharomyces cerevisiae/chemistry , Saccharomyces cerevisiae Proteins/chemistry , Software
5.
Cell ; 173(3): 735-748.e15, 2018 04 19.
Article in English | MEDLINE | ID: mdl-29677516

ABSTRACT

Teneurins (TENs) are cell-surface adhesion proteins with critical roles in tissue development and axon guidance. Here, we report the 3.1-Å cryoelectron microscopy structure of the human TEN2 extracellular region (ECR), revealing a striking similarity to bacterial Tc-toxins. The ECR includes a large ß barrel that partially encapsulates a C-terminal domain, which emerges to the solvent through an opening in the mid-barrel region. An immunoglobulin (Ig)-like domain seals the bottom of the barrel while a ß propeller is attached in a perpendicular orientation. We further show that an alternatively spliced region within the ß propeller acts as a switch to regulate trans-cellular adhesion of TEN2 to latrophilin (LPHN), a transmembrane receptor known to mediate critical functions in the central nervous system. One splice variant activates trans-cellular signaling in a LPHN-dependent manner, whereas the other induces inhibitory postsynaptic differentiation. These results highlight the unusual structural organization of TENs giving rise to their multifarious functions.


Subject(s)
Bacterial Toxins/chemistry , Membrane Proteins/chemistry , Nerve Tissue Proteins/chemistry , Synapses/metabolism , Alternative Splicing , Amino Acid Motifs , Animals , Axons , Cell Adhesion , Cell Line , Cyclic AMP/metabolism , Female , HEK293 Cells , Hormones/chemistry , Humans , Insecta , Membrane Proteins/metabolism , Mice , Molecular Conformation , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Neuropeptides/chemistry , Protein Binding , Receptors, G-Protein-Coupled/metabolism , Receptors, Peptide/chemistry , Signal Transduction
6.
Cell ; 171(7): 1638-1648.e7, 2017 Dec 14.
Article in English | MEDLINE | ID: mdl-29224781

ABSTRACT

Cleavage of membrane-anchored proteins by ADAM (a disintegrin and metalloproteinase) endopeptidases plays a key role in a wide variety of biological signal transduction and protein turnover processes. Among ADAM family members, ADAM10 stands out as particularly important because it is both responsible for regulated proteolysis of Notch receptors and catalyzes the non-amyloidogenic α-secretase cleavage of the Alzheimer's precursor protein (APP). We present here the X-ray crystal structure of the ADAM10 ectodomain, which, together with biochemical and cellular studies, reveals how access to the enzyme active site is regulated. The enzyme adopts an unanticipated architecture in which the C-terminal cysteine-rich domain partially occludes the enzyme active site, preventing unfettered substrate access. Binding of a modulatory antibody to the cysteine-rich domain liberates the catalytic domain from autoinhibition, enhancing enzymatic activity toward a peptide substrate. Together, these studies reveal a mechanism for regulation of ADAM activity and offer a roadmap for its modulation.


Subject(s)
ADAM10 Protein/chemistry , Amyloid Precursor Protein Secretases/chemistry , Membrane Proteins/chemistry , Proteolysis , ADAM10 Protein/metabolism , Amyloid Precursor Protein Secretases/metabolism , Crystallography, X-Ray , Humans , Membrane Proteins/metabolism , Models, Molecular , Receptors, Notch/metabolism , Signal Transduction
7.
Cell ; 169(3): 407-421.e16, 2017 04 20.
Article in English | MEDLINE | ID: mdl-28431242

ABSTRACT

The phosphorylation of agonist-occupied G-protein-coupled receptors (GPCRs) by GPCR kinases (GRKs) functions to turn off G-protein signaling and turn on arrestin-mediated signaling. While a structural understanding of GPCR/G-protein and GPCR/arrestin complexes has emerged in recent years, the molecular architecture of a GPCR/GRK complex remains poorly defined. We used a comprehensive integrated approach of cross-linking, hydrogen-deuterium exchange mass spectrometry (MS), electron microscopy, mutagenesis, molecular dynamics simulations, and computational docking to analyze GRK5 interaction with the ß2-adrenergic receptor (ß2AR). These studies revealed a dynamic mechanism of complex formation that involves large conformational changes in the GRK5 RH/catalytic domain interface upon receptor binding. These changes facilitate contacts between intracellular loops 2 and 3 and the C terminus of the ß2AR with the GRK5 RH bundle subdomain, membrane-binding surface, and kinase catalytic cleft, respectively. These studies significantly contribute to our understanding of the mechanism by which GRKs regulate the function of activated GPCRs. PAPERCLIP.


Subject(s)
G-Protein-Coupled Receptor Kinase 5/chemistry , Mammals/metabolism , Receptors, Adrenergic, beta-2/chemistry , Animals , Camelids, New World , Cattle , G-Protein-Coupled Receptor Kinase 5/genetics , G-Protein-Coupled Receptor Kinase 5/metabolism , Humans , Mass Spectrometry , Microscopy, Electron , Models, Molecular , Molecular Dynamics Simulation , Protein Binding , Rats , Receptors, Adrenergic, beta-2/genetics , Receptors, Adrenergic, beta-2/metabolism
8.
Cell ; 166(4): 907-919, 2016 Aug 11.
Article in English | MEDLINE | ID: mdl-27499021

ABSTRACT

Classically, G protein-coupled receptor (GPCR) stimulation promotes G protein signaling at the plasma membrane, followed by rapid ß-arrestin-mediated desensitization and receptor internalization into endosomes. However, it has been demonstrated that some GPCRs activate G proteins from within internalized cellular compartments, resulting in sustained signaling. We have used a variety of biochemical, biophysical, and cell-based methods to demonstrate the existence, functionality, and architecture of internalized receptor complexes composed of a single GPCR, ß-arrestin, and G protein. These super-complexes or "megaplexes" more readily form at receptors that interact strongly with ß-arrestins via a C-terminal tail containing clusters of serine/threonine phosphorylation sites. Single-particle electron microscopy analysis of negative-stained purified megaplexes reveals that a single receptor simultaneously binds through its core region with G protein and through its phosphorylated C-terminal tail with ß-arrestin. The formation of such megaplexes provides a potential physical basis for the newly appreciated sustained G protein signaling from internalized GPCRs.


Subject(s)
Receptors, G-Protein-Coupled/metabolism , Signal Transduction , beta-Arrestins/metabolism , Bioluminescence Resonance Energy Transfer Techniques , Cyclic AMP/metabolism , Endosomes/metabolism , GTP-Binding Protein alpha Subunits, Gs/metabolism , HEK293 Cells , Humans , Microscopy, Confocal , Microscopy, Electron , Multiprotein Complexes , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, G-Protein-Coupled/chemistry , beta-Arrestins/chemistry
9.
Immunity ; 54(7): 1405-1416.e7, 2021 07 13.
Article in English | MEDLINE | ID: mdl-34216564

ABSTRACT

Epstein-Barr virus (EBV) encodes a G protein-coupled receptor (GPCR) termed BILF1 that is essential for EBV-mediated immunosuppression and oncogenesis. BILF1 couples with inhibitory G protein (Gi), the major intracellular signaling effector for human chemokine receptors, and exhibits constitutive signaling activity; the ligand(s) for BILF1 are unknown. We studied the origins of BILF1's constitutive activity through structure determination of BILF1 bound to the inhibitory G protein (Gi) heterotrimer. The 3.2-Å resolution cryo-electron microscopy structure revealed an extracellular loop within BILF1 that blocked the typical chemokine binding site, suggesting ligand-autonomous receptor activation. Rather, amino acid substitutions within BILF1 transmembrane regions at hallmark ligand-activated class A GPCR "microswitches" stabilized a constitutively active BILF1 conformation for Gi coupling in a ligand-independent fashion. Thus, the constitutive activity of BILF1 promotes immunosuppression and virulence independent of ligand availability, with implications for the function of GPCRs encoded by related viruses and for therapeutic targeting of EBV.


Subject(s)
Epstein-Barr Virus Infections/immunology , Herpesvirus 4, Human/immunology , Immunologic Factors/immunology , Receptors, G-Protein-Coupled/immunology , Viral Proteins/immunology , Animals , Binding Sites/immunology , Cell Line , Chemokines/immunology , Cryoelectron Microscopy/methods , Epstein-Barr Virus Infections/virology , HEK293 Cells , Humans , Protein Binding/immunology , Sf9 Cells , Signal Transduction/immunology
10.
Nature ; 626(8001): 1141-1148, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38326620

ABSTRACT

The calcium-sensing receptor (CaSR) is a family C G-protein-coupled receptor1 (GPCR) that has a central role in regulating systemic calcium homeostasis2,3. Here we use cryo-electron microscopy and functional assays to investigate the activation of human CaSR embedded in lipid nanodiscs and its coupling to functional Gi versus Gq proteins in the presence and absence of the calcimimetic drug cinacalcet. High-resolution structures show that both Gi and Gq drive additional conformational changes in the activated CaSR dimer to stabilize a more extensive asymmetric interface of the seven-transmembrane domain (7TM) that involves key protein-lipid interactions. Selective Gi and Gq coupling by the receptor is achieved through substantial rearrangements of intracellular loop 2 and the C terminus, which contribute differentially towards the binding of the two G-protein subtypes, resulting in distinct CaSR-G-protein interfaces. The structures also reveal that natural polyamines target multiple sites on CaSR to enhance receptor activation by zipping negatively charged regions between two protomers. Furthermore, we find that the amino acid L-tryptophan, a well-known ligand of CaSR extracellular domains, occupies the 7TM bundle of the G-protein-coupled protomer at the same location as cinacalcet and other allosteric modulators. Together, these results provide a framework for G-protein activation and selectivity by CaSR, as well as its allosteric modulation by endogenous and exogenous ligands.


Subject(s)
Heterotrimeric GTP-Binding Proteins , Receptors, Calcium-Sensing , Humans , Allosteric Regulation/drug effects , Cinacalcet/pharmacology , Cryoelectron Microscopy , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Heterotrimeric GTP-Binding Proteins/metabolism , Ligands , Lipids , Nanostructures/chemistry , Polyamines/metabolism , Protein Conformation/drug effects , Receptors, Calcium-Sensing/chemistry , Receptors, Calcium-Sensing/metabolism , Receptors, Calcium-Sensing/ultrastructure , Substrate Specificity , Tryptophan/metabolism , Calcium/metabolism
11.
Nature ; 629(8014): 1182-1191, 2024 May.
Article in English | MEDLINE | ID: mdl-38480881

ABSTRACT

G-protein-coupled receptors (GPCRs) activate heterotrimeric G proteins by stimulating guanine nucleotide exchange in the Gα subunit1. To visualize this mechanism, we developed a time-resolved cryo-EM approach that examines the progression of ensembles of pre-steady-state intermediates of a GPCR-G-protein complex. By monitoring the transitions of the stimulatory Gs protein in complex with the ß2-adrenergic receptor at short sequential time points after GTP addition, we identified the conformational trajectory underlying G-protein activation and functional dissociation from the receptor. Twenty structures generated from sequential overlapping particle subsets along this trajectory, compared to control structures, provide a high-resolution description of the order of main events driving G-protein activation in response to GTP binding. Structural changes propagate from the nucleotide-binding pocket and extend through the GTPase domain, enacting alterations to Gα switch regions and the α5 helix that weaken the G-protein-receptor interface. Molecular dynamics simulations with late structures in the cryo-EM trajectory support that enhanced ordering of GTP on closure of the α-helical domain against the nucleotide-bound Ras-homology domain correlates with α5 helix destabilization and eventual dissociation of the G protein from the GPCR. These findings also highlight the potential of time-resolved cryo-EM as a tool for mechanistic dissection of GPCR signalling events.


Subject(s)
Cryoelectron Microscopy , GTP-Binding Protein alpha Subunits, Gs , Receptors, Adrenergic, beta-2 , Humans , Binding Sites , GTP-Binding Protein alpha Subunits, Gs/chemistry , GTP-Binding Protein alpha Subunits, Gs/drug effects , GTP-Binding Protein alpha Subunits, Gs/metabolism , GTP-Binding Protein alpha Subunits, Gs/ultrastructure , Guanosine Triphosphate/metabolism , Guanosine Triphosphate/pharmacology , Models, Molecular , Molecular Dynamics Simulation , Protein Binding , Receptors, Adrenergic, beta-2/metabolism , Receptors, Adrenergic, beta-2/chemistry , Receptors, Adrenergic, beta-2/ultrastructure , Time Factors , Enzyme Activation/drug effects , Protein Domains , Protein Structure, Secondary , Signal Transduction/drug effects
12.
Nature ; 613(7945): 767-774, 2023 01.
Article in English | MEDLINE | ID: mdl-36450356

ABSTRACT

Mu-opioid receptor (µOR) agonists such as fentanyl have long been used for pain management, but are considered a major public health concern owing to their adverse side effects, including lethal overdose1. Here, in an effort to design safer therapeutic agents, we report an approach targeting a conserved sodium ion-binding site2 found in µOR3 and many other class A G-protein-coupled receptors with bitopic fentanyl derivatives that are functionalized via a linker with a positively charged guanidino group. Cryo-electron microscopy structures of the most potent bitopic ligands in complex with µOR highlight the key interactions between the guanidine of the ligands and the key Asp2.50 residue in the Na+ site. Two bitopics (C5 and C6 guano) maintain nanomolar potency and high efficacy at Gi subtypes and show strongly reduced arrestin recruitment-one (C6 guano) also shows the lowest Gz efficacy among the panel of µOR agonists, including partial and biased morphinan and fentanyl analogues. In mice, C6 guano displayed µOR-dependent antinociception with attenuated adverse effects, supporting the µOR sodium ion-binding site as a potential target for the design of safer analgesics. In general, our study suggests that bitopic ligands that engage the sodium ion-binding pocket in class A G-protein-coupled receptors can be designed to control their efficacy and functional selectivity profiles for Gi, Go and Gz subtypes and arrestins, thus modulating their in vivo pharmacology.


Subject(s)
Drug Design , Fentanyl , Morphinans , Receptors, Opioid, mu , Animals , Mice , Analgesics, Opioid/chemistry , Analgesics, Opioid/metabolism , Arrestins/metabolism , Cryoelectron Microscopy , Fentanyl/analogs & derivatives , Fentanyl/chemistry , Fentanyl/metabolism , Ligands , Morphinans/chemistry , Morphinans/metabolism , Receptors, Opioid, mu/agonists , Receptors, Opioid, mu/chemistry , Receptors, Opioid, mu/metabolism , Receptors, Opioid, mu/ultrastructure , Binding Sites , Nociception
13.
Nature ; 601(7892): 274-279, 2022 01.
Article in English | MEDLINE | ID: mdl-34880492

ABSTRACT

Glucose is a primary energy source in living cells. The discovery in 1960s that a sodium gradient powers the active uptake of glucose in the intestine1 heralded the concept of a secondary active transporter that can catalyse the movement of a substrate against an electrochemical gradient by harnessing energy from another coupled substrate. Subsequently, coupled Na+/glucose transport was found to be mediated by sodium-glucose cotransporters2,3 (SGLTs). SGLTs are responsible for active glucose and galactose absorption in the intestine and for glucose reabsorption in the kidney4, and are targeted by multiple drugs to treat diabetes5. Several members within the SGLT family transport key metabolites other than glucose2. Here we report cryo-electron microscopy structures of the prototypic human SGLT1 and a related monocarboxylate transporter SMCT1 from the same family. The structures, together with molecular dynamics simulations and functional studies, define the architecture of SGLTs, uncover the mechanism of substrate binding and selectivity, and shed light on water permeability of SGLT1. These results provide insights into the multifaceted functions of SGLTs.


Subject(s)
Cryoelectron Microscopy , Glucose , Glucose/metabolism , Humans , Monocarboxylic Acid Transporters/chemistry , Monocarboxylic Acid Transporters/metabolism , Monocarboxylic Acid Transporters/ultrastructure , Sodium/metabolism , Sodium-Glucose Transporter 1/chemistry , Sodium-Glucose Transporter 1/metabolism , Sodium-Glucose Transporter 1/ultrastructure , Substrate Specificity
14.
Nature ; 604(7907): 757-762, 2022 04.
Article in English | MEDLINE | ID: mdl-35418682

ABSTRACT

Adhesion G-protein-coupled receptors (aGPCRs) are characterized by the presence of auto-proteolysing extracellular regions that are involved in cell-cell and cell-extracellular matrix interactions1. Self cleavage within the aGPCR auto-proteolysis-inducing (GAIN) domain produces two protomers-N-terminal and C-terminal fragments-that remain non-covalently attached after receptors reach the cell surface1. Upon dissociation of the N-terminal fragment, the C-terminus of the GAIN domain acts as a tethered agonist (TA) peptide to activate the seven-transmembrane domain with a mechanism that has been poorly understood2-5. Here we provide cryo-electron microscopy snapshots of two distinct members of the aGPCR family, GPR56 (also known as ADGRG1) and latrophilin 3 (LPHN3 (also known as ADGRL3)). Low-resolution maps of the receptors in their N-terminal fragment-bound state indicate that the GAIN domain projects flexibly towards the extracellular space, keeping the encrypted TA peptide away from the seven-transmembrane domain. High-resolution structures of GPR56 and LPHN3 in their active, G-protein-coupled states, reveal that after dissociation of the extracellular region, the decrypted TA peptides engage the seven-transmembrane domain core with a notable conservation of interactions that also involve extracellular loop 2. TA binding stabilizes breaks in the middle of transmembrane helices 6 and 7 that facilitate aGPCR coupling and activation of heterotrimeric G proteins. Collectively, these results enable us to propose a general model for aGPCR activation.


Subject(s)
Receptors, G-Protein-Coupled , Signal Transduction , Cell Adhesion , Cell Membrane/metabolism , Cryoelectron Microscopy , Humans , Peptides/chemistry , Protein Binding , Receptors, G-Protein-Coupled/metabolism , Receptors, Peptide
15.
Nature ; 610(7932): 582-591, 2022 10.
Article in English | MEDLINE | ID: mdl-36171289

ABSTRACT

There is considerable interest in screening ultralarge chemical libraries for ligand discovery, both empirically and computationally1-4. Efforts have focused on readily synthesizable molecules, inevitably leaving many chemotypes unexplored. Here we investigate structure-based docking of a bespoke virtual library of tetrahydropyridines-a scaffold that is poorly sampled by a general billion-molecule virtual library but is well suited to many aminergic G-protein-coupled receptors. Using three inputs, each with diverse available derivatives, a one pot C-H alkenylation, electrocyclization and reduction provides the tetrahydropyridine core with up to six sites of derivatization5-7. Docking a virtual library of 75 million tetrahydropyridines against a model of the serotonin 5-HT2A receptor (5-HT2AR) led to the synthesis and testing of 17 initial molecules. Four of these molecules had low-micromolar activities against either the 5-HT2A or the 5-HT2B receptors. Structure-based optimization led to the 5-HT2AR agonists (R)-69 and (R)-70, with half-maximal effective concentration values of 41 nM and 110 nM, respectively, and unusual signalling kinetics that differ from psychedelic 5-HT2AR agonists. Cryo-electron microscopy structural analysis confirmed the predicted binding mode to 5-HT2AR. The favourable physical properties of these new agonists conferred high brain permeability, enabling mouse behavioural assays. Notably, neither had psychedelic activity, in contrast to classic 5-HT2AR agonists, whereas both had potent antidepressant activity in mouse models and had the same efficacy as antidepressants such as fluoxetine at as low as 1/40th of the dose. Prospects for using bespoke virtual libraries to sample pharmacologically relevant chemical space will be considered.


Subject(s)
Antidepressive Agents , Pyrrolidines , Receptor, Serotonin, 5-HT2A , Animals , Mice , Antidepressive Agents/pharmacology , Cryoelectron Microscopy , Fluoxetine/administration & dosage , Fluoxetine/pharmacology , Hallucinogens/administration & dosage , Hallucinogens/pharmacology , Ligands , Pyrrolidines/administration & dosage , Pyrrolidines/pharmacology , Receptor, Serotonin, 5-HT2A/metabolism , Small Molecule Libraries
16.
Mol Cell ; 80(2): 237-245.e4, 2020 10 15.
Article in English | MEDLINE | ID: mdl-33007200

ABSTRACT

Heterotrimeric G proteins communicate signals from activated G protein-coupled receptors to downstream effector proteins. In the phototransduction pathway responsible for vertebrate vision, the G protein-effector complex is composed of the GTP-bound transducin α subunit (GαT·GTP) and the cyclic GMP (cGMP) phosphodiesterase 6 (PDE6), which stimulates cGMP hydrolysis, leading to hyperpolarization of the photoreceptor cell. Here we report a cryo-electron microscopy (cryoEM) structure of PDE6 complexed to GTP-bound GαT. The structure reveals two GαT·GTP subunits engaging the PDE6 hetero-tetramer at both the PDE6 catalytic core and the PDEγ subunits, driving extensive rearrangements to relieve all inhibitory constraints on enzyme catalysis. Analysis of the conformational ensemble in the cryoEM data highlights the dynamic nature of the contacts between the two GαT·GTP subunits and PDE6 that supports an alternating-site catalytic mechanism.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 6/metabolism , Signal Transduction , Transducin/metabolism , Animals , Biocatalysis , Catalytic Domain , Cattle , Cyclic Nucleotide Phosphodiesterases, Type 6/chemistry , Cyclic Nucleotide Phosphodiesterases, Type 6/ultrastructure , Guanosine Triphosphate/chemistry , Guanosine Triphosphate/metabolism , Models, Molecular , Protein Binding , Protein Domains , Transducin/chemistry , Transducin/ultrastructure , Vardenafil Dihydrochloride/chemistry , Vardenafil Dihydrochloride/metabolism
17.
Nature ; 595(7867): 450-454, 2021 07.
Article in English | MEDLINE | ID: mdl-34194039

ABSTRACT

Family C G-protein-coupled receptors (GPCRs) operate as obligate dimers with extracellular domains that recognize small ligands, leading to G-protein activation on the transmembrane (TM) domains of these receptors by an unknown mechanism1. Here we show structures of homodimers of the family C metabotropic glutamate receptor 2 (mGlu2) in distinct functional states and in complex with heterotrimeric Gi. Upon activation of the extracellular domain, the two transmembrane domains undergo extensive rearrangement in relative orientation to establish an asymmetric TM6-TM6 interface that promotes conformational changes in the cytoplasmic domain of one protomer. Nucleotide-bound Gi can be observed pre-coupled to inactive mGlu2, but its transition to the nucleotide-free form seems to depend on establishing the active-state TM6-TM6 interface. In contrast to family A and B GPCRs, G-protein coupling does not involve the cytoplasmic opening of TM6 but is facilitated through the coordination of intracellular loops 2 and 3, as well as a critical contribution from the C terminus of the receptor. The findings highlight the synergy of global and local conformational transitions to facilitate a new mode of G-protein activation.


Subject(s)
Heterotrimeric GTP-Binding Proteins/metabolism , Receptors, Metabotropic Glutamate/metabolism , Cell Membrane/chemistry , Cell Membrane/metabolism , GTP-Binding Protein alpha Subunits, Gi-Go/chemistry , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Heterotrimeric GTP-Binding Proteins/chemistry , Humans , Models, Molecular , Protein Multimerization , Receptors, Metabotropic Glutamate/chemistry
18.
Nature ; 595(7867): 455-459, 2021 07.
Article in English | MEDLINE | ID: mdl-34194040

ABSTRACT

The calcium-sensing receptor (CaSR), a cell-surface sensor for Ca2+, is the master regulator of calcium homeostasis in humans and is the target of calcimimetic drugs for the treatment of parathyroid disorders1. CaSR is a family C G-protein-coupled receptor2 that functions as an obligate homodimer, with each protomer composed of a Ca2+-binding extracellular domain and a seven-transmembrane-helix domain (7TM) that activates heterotrimeric G proteins. Here we present cryo-electron microscopy structures of near-full-length human CaSR in inactive or active states bound to Ca2+ and various calcilytic or calcimimetic drug molecules. We show that, upon activation, the CaSR homodimer adopts an asymmetric 7TM configuration that primes one protomer for G-protein coupling. This asymmetry is stabilized by 7TM-targeting calcimimetic drugs adopting distinctly different poses in the two protomers, whereas the binding of a calcilytic drug locks CaSR 7TMs in an inactive symmetric configuration. These results provide a detailed structural framework for CaSR activation and the rational design of therapeutics targeting this receptor.


Subject(s)
Calcium/metabolism , Cryoelectron Microscopy , Protein Multimerization , Receptors, Calcium-Sensing/chemistry , Receptors, Calcium-Sensing/metabolism , Calcium/chemistry , Humans , Models, Molecular , Peptides/chemistry , Peptides/metabolism , Protein Binding , Receptors, Calcium-Sensing/ultrastructure , Substrate Specificity
19.
Mol Cell ; 75(4): 781-790.e3, 2019 08 22.
Article in English | MEDLINE | ID: mdl-31300275

ABSTRACT

Rhodopsin (Rho), a prototypical G-protein-coupled receptor (GPCR) in vertebrate vision, activates the G-protein transducin (GT) by catalyzing GDP-GTP exchange on its α subunit (GαT). To elucidate the determinants of GT coupling and activation, we obtained cryo-EM structures of a fully functional, light-activated Rho-GT complex in the presence and absence of a G-protein-stabilizing nanobody. The structures illustrate how GT overcomes its low basal activity by engaging activated Rho in a conformation distinct from other GPCR-G-protein complexes. Moreover, the nanobody-free structures reveal native conformations of G-protein components and capture three distinct conformers showing the GαT helical domain (αHD) contacting the Gßγ subunits. These findings uncover the molecular underpinnings of G-protein activation by visual rhodopsin and shed new light on the role played by Gßγ during receptor-catalyzed nucleotide exchange.


Subject(s)
Multiprotein Complexes/chemistry , Rhodopsin/chemistry , Transducin/chemistry , Animals , Cattle , Cryoelectron Microscopy , Multiprotein Complexes/metabolism , Multiprotein Complexes/ultrastructure , Protein Domains , Protein Structure, Secondary , Rhodopsin/metabolism , Transducin/metabolism
20.
Trends Biochem Sci ; 47(2): 124-135, 2022 02.
Article in English | MEDLINE | ID: mdl-34281791

ABSTRACT

Structure-based drug discovery (SBDD) is an indispensable approach for the design and optimization of new therapeutic agents. Here, we highlight the rapid progress that has turned cryo-electron microscopy (cryoEM) into an exceptional SBDD tool, and the wealth of new structural information it is providing for high-value pharmacological targets. We review key advantages of a technique that directly images vitrified biomolecules without the need for crystallization; both in terms of a broader array of systems that can be studied and the different forms of information it can provide, including heterogeneity and dynamics. We discuss near- and far-future developments, working in concert towards achieving the resolution and throughput necessary for cryoEM to make a widespread impact on the SBDD pipeline.


Subject(s)
Cryoelectron Microscopy , Drug Discovery , Cryoelectron Microscopy/methods , Crystallography, X-Ray , Drug Discovery/methods
SELECTION OF CITATIONS
SEARCH DETAIL