Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 69
Filter
1.
Cell ; 186(2): 413-427.e17, 2023 01 19.
Article in English | MEDLINE | ID: mdl-36638794

ABSTRACT

Opioids are effective analgesics, but their use is beset by serious side effects, including addiction and respiratory depression, which contribute to the ongoing opioid crisis. The human opioid system contains four opioid receptors (µOR, δOR, κOR, and NOPR) and a set of related endogenous opioid peptides (EOPs), which show distinct selectivity toward their respective opioid receptors (ORs). Despite being key to the development of safer analgesics, the mechanisms of molecular recognition and selectivity of EOPs to ORs remain unclear. Here, we systematically characterize the binding of EOPs to ORs and present five structures of EOP-OR-Gi complexes, including ß-endorphin- and endomorphin-bound µOR, deltorphin-bound δOR, dynorphin-bound κOR, and nociceptin-bound NOPR. These structures, supported by biochemical results, uncover the specific recognition and selectivity of opioid peptides and the conserved mechanism of opioid receptor activation. These results provide a structural framework to facilitate rational design of safer opioid drugs for pain relief.


Subject(s)
Receptors, Opioid , Humans , Analgesics, Opioid/pharmacology , Opioid Peptides , Receptors, Opioid, mu/metabolism , Receptors, Opioid/chemistry
2.
Cell ; 185(23): 4361-4375.e19, 2022 11 10.
Article in English | MEDLINE | ID: mdl-36368306

ABSTRACT

Morphine and fentanyl are among the most used opioid drugs that confer analgesia and unwanted side effects through both G protein and arrestin signaling pathways of µ-opioid receptor (µOR). Here, we report structures of the human µOR-G protein complexes bound to morphine and fentanyl, which uncover key differences in how they bind the receptor. We also report structures of µOR bound to TRV130, PZM21, and SR17018, which reveal preferential interactions of these agonists with TM3 side of the ligand-binding pocket rather than TM6/7 side. In contrast, morphine and fentanyl form dual interactions with both TM3 and TM6/7 regions. Mutations at the TM6/7 interface abolish arrestin recruitment of µOR promoted by morphine and fentanyl. Ligands designed to reduce TM6/7 interactions display preferential G protein signaling. Our results provide crucial insights into fentanyl recognition and signaling of µOR, which may facilitate rational design of next-generation analgesics.


Subject(s)
Fentanyl , Morphine , Humans , Analgesics, Opioid/pharmacology , Arrestin/metabolism , Fentanyl/pharmacology , GTP-Binding Proteins/metabolism , Morphine/pharmacology , Receptors, Opioid, mu
3.
Cell ; 184(4): 931-942.e18, 2021 02 18.
Article in English | MEDLINE | ID: mdl-33571431

ABSTRACT

The D1- and D2-dopamine receptors (D1R and D2R), which signal through Gs and Gi, respectively, represent the principal stimulatory and inhibitory dopamine receptors in the central nervous system. D1R and D2R also represent the main therapeutic targets for Parkinson's disease, schizophrenia, and many other neuropsychiatric disorders, and insight into their signaling is essential for understanding both therapeutic and side effects of dopaminergic drugs. Here, we report four cryoelectron microscopy (cryo-EM) structures of D1R-Gs and D2R-Gi signaling complexes with selective and non-selective dopamine agonists, including two currently used anti-Parkinson's disease drugs, apomorphine and bromocriptine. These structures, together with mutagenesis studies, reveal the conserved binding mode of dopamine agonists, the unique pocket topology underlying ligand selectivity, the conformational changes in receptor activation, and potential structural determinants for G protein-coupling selectivity. These results provide both a molecular understanding of dopamine signaling and multiple structural templates for drug design targeting the dopaminergic system.


Subject(s)
Receptors, Dopamine D1/chemistry , Receptors, Dopamine D1/metabolism , Receptors, Dopamine D2/chemistry , Receptors, Dopamine D2/metabolism , Signal Transduction , 2,3,4,5-Tetrahydro-7,8-dihydroxy-1-phenyl-1H-3-benzazepine/analogs & derivatives , 2,3,4,5-Tetrahydro-7,8-dihydroxy-1-phenyl-1H-3-benzazepine/pharmacology , Amino Acid Sequence , Conserved Sequence , Cryoelectron Microscopy , Cyclic AMP/metabolism , GTP-Binding Proteins/metabolism , HEK293 Cells , Humans , Ligands , Models, Molecular , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Receptors, Adrenergic, beta-2/metabolism , Receptors, Dopamine D1/ultrastructure , Receptors, Dopamine D2/ultrastructure , Structural Homology, Protein
4.
Cell ; 180(4): 645-654.e13, 2020 02 20.
Article in English | MEDLINE | ID: mdl-32004460

ABSTRACT

Drugs selectively targeting CB2 hold promise for treating neurodegenerative disorders, inflammation, and pain while avoiding psychotropic side effects mediated by CB1. The mechanisms underlying CB2 activation and signaling are poorly understood but critical for drug design. Here we report the cryo-EM structure of the human CB2-Gi signaling complex bound to the agonist WIN 55,212-2. The 3D structure reveals the binding mode of WIN 55,212-2 and structural determinants for distinguishing CB2 agonists from antagonists, which are supported by a pair of rationally designed agonist and antagonist. Further structural analyses with computational docking results uncover the differences between CB2 and CB1 in receptor activation, ligand recognition, and Gi coupling. These findings are expected to facilitate rational structure-based discovery of drugs targeting the cannabinoid system.


Subject(s)
GTP-Binding Protein alpha Subunits, Gi-Go/chemistry , Receptor, Cannabinoid, CB2/chemistry , Signal Transduction , Animals , Binding Sites , CHO Cells , Cannabinoid Receptor Agonists/chemical synthesis , Cannabinoid Receptor Agonists/pharmacology , Cannabinoid Receptor Antagonists/chemical synthesis , Cannabinoid Receptor Antagonists/pharmacology , Cricetinae , Cricetulus , Cryoelectron Microscopy , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Humans , Molecular Docking Simulation , Protein Binding , Receptor, Cannabinoid, CB2/agonists , Receptor, Cannabinoid, CB2/antagonists & inhibitors , Receptor, Cannabinoid, CB2/metabolism , Sf9 Cells , Spodoptera
5.
Cell ; 170(3): 457-469.e13, 2017 Jul 27.
Article in English | MEDLINE | ID: mdl-28753425

ABSTRACT

G protein-coupled receptors (GPCRs) mediate diverse signaling in part through interaction with arrestins, whose binding promotes receptor internalization and signaling through G protein-independent pathways. High-affinity arrestin binding requires receptor phosphorylation, often at the receptor's C-terminal tail. Here, we report an X-ray free electron laser (XFEL) crystal structure of the rhodopsin-arrestin complex, in which the phosphorylated C terminus of rhodopsin forms an extended intermolecular ß sheet with the N-terminal ß strands of arrestin. Phosphorylation was detected at rhodopsin C-terminal tail residues T336 and S338. These two phospho-residues, together with E341, form an extensive network of electrostatic interactions with three positively charged pockets in arrestin in a mode that resembles binding of the phosphorylated vasopressin-2 receptor tail to ß-arrestin-1. Based on these observations, we derived and validated a set of phosphorylation codes that serve as a common mechanism for phosphorylation-dependent recruitment of arrestins by GPCRs.


Subject(s)
Arrestins/chemistry , Rhodopsin/chemistry , Amino Acid Sequence , Animals , Arrestins/metabolism , Chromatography, Liquid , Humans , Mice , Models, Molecular , Phosphorylation , Rats , Rhodopsin/metabolism , Sequence Alignment , Tandem Mass Spectrometry , X-Rays
6.
Mol Cell ; 81(6): 1147-1159.e4, 2021 03 18.
Article in English | MEDLINE | ID: mdl-33548201

ABSTRACT

The dopamine system, including five dopamine receptors (D1R-D5R), plays essential roles in the central nervous system (CNS), and ligands that activate dopamine receptors have been used to treat many neuropsychiatric disorders. Here, we report two cryo-EM structures of human D3R in complex with an inhibitory G protein and bound to the D3R-selective agonists PD128907 and pramipexole, the latter of which is used to treat patients with Parkinson's disease. The structures reveal agonist binding modes distinct from the antagonist-bound D3R structure and conformational signatures for ligand-induced receptor activation. Mutagenesis and homology modeling illuminate determinants of ligand specificity across dopamine receptors and the mechanisms for Gi protein coupling. Collectively our work reveals the basis of agonist binding and ligand-induced receptor activation and provides structural templates for designing specific ligands to treat CNS diseases targeting the dopaminergic system.


Subject(s)
Cryoelectron Microscopy , GTP-Binding Protein alpha Subunits, Gi-Go/chemistry , Models, Molecular , Multiprotein Complexes/ultrastructure , Receptors, Dopamine D3/chemistry , Benzopyrans/chemistry , HEK293 Cells , Humans , Multiprotein Complexes/chemistry , Oxazines/chemistry , Pramipexole/chemistry , Protein Domains , Structure-Activity Relationship
7.
Mol Cell ; 77(3): 669-680.e4, 2020 02 06.
Article in English | MEDLINE | ID: mdl-32004470

ABSTRACT

Corticotropin-releasing factor (CRF) and the three related peptides urocortins 1-3 (UCN1-UCN3) are endocrine hormones that control the stress responses by activating CRF1R and CRF2R, two members of class B G-protein-coupled receptors (GPCRs). Here, we present two cryoelectron microscopy (cryo-EM) structures of UCN1-bound CRF1R and CRF2R with the stimulatory G protein. In both structures, UCN1 adopts a single straight helix with its N terminus dipped into the receptor transmembrane bundle. Although the peptide-binding residues in CRF1R and CRF2R are different from other members of class B GPCRs, the residues involved in receptor activation and G protein coupling are conserved. In addition, both structures reveal bound cholesterol molecules to the receptor transmembrane helices. Our structures define the basis of ligand-binding specificity in the CRF receptor-hormone system, establish a common mechanism of class B GPCR activation and G protein coupling, and provide a paradigm for studying membrane protein-lipid interactions for class B GPCRs.


Subject(s)
Receptors, Corticotropin-Releasing Hormone/ultrastructure , Amino Acid Sequence , Binding Sites , Corticotropin-Releasing Hormone , Cryoelectron Microscopy/methods , GTP-Binding Protein alpha Subunits, Gs/metabolism , GTP-Binding Proteins/metabolism , Humans , Peptides/metabolism , Receptors, Corticotropin-Releasing Hormone/metabolism , Urocortins/metabolism
8.
Nature ; 592(7854): 469-473, 2021 04.
Article in English | MEDLINE | ID: mdl-33762731

ABSTRACT

Serotonin, or 5-hydroxytryptamine (5-HT), is an important neurotransmitter1,2 that activates the largest subtype family of G-protein-coupled receptors3. Drugs that target 5-HT1A, 5-HT1D, 5-HT1E and other 5-HT receptors are used to treat numerous disorders4. 5-HT receptors have high levels of basal activity and are subject to regulation by lipids, but the structural basis for the lipid regulation and basal activation of these receptors and the pan-agonism of 5-HT remains unclear. Here we report five structures of 5-HT receptor-G-protein complexes: 5-HT1A in the apo state, bound to 5-HT or bound to the antipsychotic drug aripiprazole; 5-HT1D bound to 5-HT; and 5-HT1E in complex with a 5-HT1E- and 5-HT1F-selective agonist, BRL-54443. Notably, the phospholipid phosphatidylinositol 4-phosphate is present at the G-protein-5-HT1A interface, and is able to increase 5-HT1A-mediated G-protein activity. The receptor transmembrane domain is surrounded by cholesterol molecules-particularly in the case of 5-HT1A, in which cholesterol molecules are directly involved in shaping the ligand-binding pocket that determines the specificity for aripiprazol. Within the ligand-binding pocket of apo-5-HT1A are structured water molecules that mimic 5-HT to activate the receptor. Together, our results address a long-standing question of how lipids and water molecules regulate G-protein-coupled receptors, reveal how 5-HT acts as a pan-agonist, and identify the determinants of drug recognition in 5-HT receptors.


Subject(s)
Cryoelectron Microscopy , Ligands , Lipids , Receptors, Serotonin, 5-HT1/metabolism , Receptors, Serotonin, 5-HT1/ultrastructure , Apoproteins/chemistry , Apoproteins/metabolism , Apoproteins/ultrastructure , Aripiprazole/metabolism , Aripiprazole/pharmacology , Binding Sites , Cholesterol/pharmacology , Heterotrimeric GTP-Binding Proteins/chemistry , Heterotrimeric GTP-Binding Proteins/metabolism , Heterotrimeric GTP-Binding Proteins/ultrastructure , Humans , Models, Molecular , Phosphatidylinositol Phosphates/chemistry , Phosphatidylinositol Phosphates/metabolism , Phosphatidylinositol Phosphates/pharmacology , Receptor, Serotonin, 5-HT1A/chemistry , Receptor, Serotonin, 5-HT1A/metabolism , Receptor, Serotonin, 5-HT1A/ultrastructure , Receptors, Serotonin, 5-HT1/chemistry , Serotonin 5-HT1 Receptor Agonists/chemistry , Serotonin 5-HT1 Receptor Agonists/metabolism , Serotonin 5-HT1 Receptor Agonists/pharmacology , Water/chemistry
9.
Nature ; 586(7827): 151-155, 2020 10.
Article in English | MEDLINE | ID: mdl-32968275

ABSTRACT

CpG methylation by de novo DNA methyltransferases (DNMTs) 3A and 3B is essential for mammalian development and differentiation and is frequently dysregulated in cancer1. These two DNMTs preferentially bind to nucleosomes, yet cannot methylate the DNA wrapped around the nucleosome core2, and they favour the methylation of linker DNA at positioned nucleosomes3,4. Here we present the cryo-electron microscopy structure of a ternary complex of catalytically competent DNMT3A2, the catalytically inactive accessory subunit DNMT3B3 and a nucleosome core particle flanked by linker DNA. The catalytic-like domain of the accessory DNMT3B3 binds to the acidic patch of the nucleosome core, which orients the binding of DNMT3A2 to the linker DNA. The steric constraints of this arrangement suggest that nucleosomal DNA must be moved relative to the nucleosome core for de novo methylation to occur.


Subject(s)
Cryoelectron Microscopy , DNA (Cytosine-5-)-Methyltransferases/chemistry , DNA (Cytosine-5-)-Methyltransferases/metabolism , Nucleosomes/metabolism , Animals , Biocatalysis , Chromatin Assembly and Disassembly , DNA/chemistry , DNA/metabolism , DNA Methylation , DNA Methyltransferase 3A , Histones/chemistry , Histones/genetics , Histones/metabolism , Humans , Models, Molecular , Nucleosomes/chemistry , Protein Binding , Protein Domains , Xenopus/genetics , DNA Methyltransferase 3B
10.
Nature ; 561(7724): E44, 2018 09.
Article in English | MEDLINE | ID: mdl-29930353

ABSTRACT

In the PDF version of this Article, owing to a typesetting error, an incorrect figure was used for Extended Data Fig. 5; the correct figure was used in the HTML version. This has been corrected online.

11.
Nature ; 558(7711): 553-558, 2018 06.
Article in English | MEDLINE | ID: mdl-29899450

ABSTRACT

G-protein-coupled receptors comprise the largest family of mammalian transmembrane receptors. They mediate numerous cellular pathways by coupling with downstream signalling transducers, including the hetrotrimeric G proteins Gs (stimulatory) and Gi (inhibitory) and several arrestin proteins. The structural mechanisms that define how G-protein-coupled receptors selectively couple to a specific type of G protein or arrestin remain unknown. Here, using cryo-electron microscopy, we show that the major interactions between activated rhodopsin and Gi are mediated by the C-terminal helix of the Gi α-subunit, which is wedged into the cytoplasmic cavity of the transmembrane helix bundle and directly contacts the amino terminus of helix 8 of rhodopsin. Structural comparisons of inactive, Gi-bound and arrestin-bound forms of rhodopsin with inactive and Gs-bound forms of the ß2-adrenergic receptor provide a foundation to understand the unique structural signatures that are associated with the recognition of Gs, Gi and arrestin by activated G-protein-coupled receptors.


Subject(s)
Cryoelectron Microscopy , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , GTP-Binding Protein alpha Subunits, Gi-Go/ultrastructure , Rhodopsin/metabolism , Rhodopsin/ultrastructure , Arrestin/chemistry , Arrestin/metabolism , Binding Sites , GTP-Binding Protein alpha Subunits, Gi-Go/chemistry , GTP-Binding Protein alpha Subunits, Gs/chemistry , GTP-Binding Protein alpha Subunits, Gs/metabolism , Humans , Models, Molecular , Receptors, Adrenergic, beta-2/chemistry , Receptors, Adrenergic, beta-2/metabolism , Rhodopsin/chemistry , Signal Transduction , Substrate Specificity
12.
Genes Dev ; 29(4): 440-50, 2015 Feb 15.
Article in English | MEDLINE | ID: mdl-25691470

ABSTRACT

The orphan nuclear receptor TLX regulates neural stem cell self-renewal in the adult brain and functions primarily as a transcription repressor through recruitment of Atrophin corepressors, which bind to TLX via a conserved peptide motif termed the Atro box. Here we report crystal structures of the human and insect TLX ligand-binding domain in complex with Atro box peptides. In these structures, TLX adopts an autorepressed conformation in which its helix H12 occupies the coactivator-binding groove. Unexpectedly, H12 in this autorepressed conformation forms a novel binding pocket with residues from helix H3 that accommodates a short helix formed by the conserved ALXXLXXY motif of the Atro box. Mutations that weaken the TLX-Atrophin interaction compromise the repressive activity of TLX, demonstrating that this interaction is required for Atrophin to confer repressor activity to TLX. Moreover, the autorepressed conformation is conserved in the repressor class of orphan nuclear receptors, and mutations of corresponding residues in other members of this class of receptors diminish their repressor activities. Together, our results establish the functional conservation of the autorepressed conformation and define a key sequence motif in the Atro box that is essential for TLX-mediated repression.


Subject(s)
Drosophila melanogaster/chemistry , Models, Molecular , Orphan Nuclear Receptors/chemistry , Receptors, Cytoplasmic and Nuclear/chemistry , Animals , Crystallization , Drosophila Proteins/chemistry , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , HEK293 Cells , Humans , Orphan Nuclear Receptors/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism
13.
PLoS Pathog ; 16(3): e1008323, 2020 03.
Article in English | MEDLINE | ID: mdl-32163521

ABSTRACT

Fusarium is a genus of filamentous fungi that includes species that cause devastating diseases in major staple crops, such as wheat, maize, rice, and barley, resulting in severe yield losses and mycotoxin contamination of infected grains. Phenamacril is a novel fungicide that is considered environmentally benign due to its exceptional specificity; it inhibits the ATPase activity of the sole class I myosin of only a subset of Fusarium species including the major plant pathogens F. graminearum, F. asiaticum and F. fujikuroi. To understand the underlying mechanisms of inhibition, species specificity, and resistance mutations, we have determined the crystal structure of phenamacril-bound F. graminearum myosin I. Phenamacril binds in the actin-binding cleft in a new allosteric pocket that contains the central residue of the regulatory Switch 2 loop and that is collapsed in the structure of a myosin with closed actin-binding cleft, suggesting that pocket occupancy blocks cleft closure. We have further identified a single, transferable phenamacril-binding residue found exclusively in phenamacril-sensitive myosins to confer phenamacril selectivity.


Subject(s)
Cyanoacrylates/chemistry , Fungal Proteins/chemistry , Fungicides, Industrial/chemistry , Fusarium/enzymology , Myosin Type I/chemistry , Cyanoacrylates/pharmacology , Fungal Proteins/genetics , Fungal Proteins/metabolism , Fungicides, Industrial/pharmacology , Fusarium/chemistry , Fusarium/drug effects , Fusarium/genetics , Myosin Type I/genetics , Myosin Type I/metabolism , Plant Diseases/microbiology , Triticum/microbiology , Zea mays/microbiology
14.
Biochem J ; 478(23): 4137-4149, 2021 12 10.
Article in English | MEDLINE | ID: mdl-34796899

ABSTRACT

Ornithine decarboxylase (ODC) is the rate-limiting enzyme for the synthesis of polyamines (PAs). PAs are oncometabolites that are required for proliferation, and pharmaceutical ODC inhibition is pursued for the treatment of hyperproliferative diseases, including cancer and infectious diseases. The most potent ODC inhibitor is 1-amino-oxy-3-aminopropane (APA). A previous crystal structure of an ODC-APA complex indicated that APA non-covalently binds ODC and its cofactor pyridoxal 5-phosphate (PLP) and functions by competing with the ODC substrate ornithine for binding to the catalytic site. We have revisited the mechanism of APA binding and ODC inhibition through a new crystal structure of APA-bound ODC, which we solved at 2.49 Šresolution. The structure unambiguously shows the presence of a covalent oxime between APA and PLP in the catalytic site, which we confirmed in solution by mass spectrometry. The stable oxime makes extensive interactions with ODC but cannot be catabolized, explaining APA's high potency in ODC inhibition. In addition, we solved an ODC/PLP complex structure with citrate bound at the substrate-binding pocket. These two structures provide new structural scaffolds for developing more efficient pharmaceutical ODC inhibitors.


Subject(s)
Ornithine Decarboxylase Inhibitors/metabolism , Ornithine Decarboxylase/metabolism , Propylamines/metabolism , Humans , Protein Binding , Protein Domains
15.
Nature ; 525(7568): 269-73, 2015 Sep 10.
Article in English | MEDLINE | ID: mdl-26258305

ABSTRACT

The plant hormone jasmonate plays crucial roles in regulating plant responses to herbivorous insects and microbial pathogens and is an important regulator of plant growth and development. Key mediators of jasmonate signalling include MYC transcription factors, which are repressed by jasmonate ZIM-domain (JAZ) transcriptional repressors in the resting state. In the presence of active jasmonate, JAZ proteins function as jasmonate co-receptors by forming a hormone-dependent complex with COI1, the F-box subunit of an SCF-type ubiquitin E3 ligase. The hormone-dependent formation of the COI1-JAZ co-receptor complex leads to ubiquitination and proteasome-dependent degradation of JAZ repressors and release of MYC proteins from transcriptional repression. The mechanism by which JAZ proteins repress MYC transcription factors and how JAZ proteins switch between the repressor function in the absence of hormone and the co-receptor function in the presence of hormone remain enigmatic. Here we show that Arabidopsis MYC3 undergoes pronounced conformational changes when bound to the conserved Jas motif of the JAZ9 repressor. The Jas motif, previously shown to bind to hormone as a partly unwound helix, forms a complete α-helix that displaces the amino (N)-terminal helix of MYC3 and becomes an integral part of the MYC N-terminal fold. In this position, the Jas helix competitively inhibits MYC3 interaction with the MED25 subunit of the transcriptional Mediator complex. Our structural and functional studies elucidate a dynamic molecular switch mechanism that governs the repression and activation of a major plant hormone pathway.


Subject(s)
Arabidopsis Proteins/antagonists & inhibitors , Arabidopsis Proteins/chemistry , Arabidopsis Proteins/metabolism , Arabidopsis , Cyclopentanes/metabolism , Oxylipins/metabolism , Plant Growth Regulators/metabolism , Repressor Proteins/chemistry , Repressor Proteins/metabolism , Signal Transduction , Trans-Activators/antagonists & inhibitors , Trans-Activators/chemistry , Amino Acid Motifs , Apoproteins/chemistry , Apoproteins/metabolism , Arabidopsis/chemistry , Arabidopsis/metabolism , Arabidopsis Proteins/genetics , Binding, Competitive/genetics , Crystallography, X-Ray , DNA-Binding Proteins , Models, Molecular , Nuclear Proteins/metabolism , Proteasome Endopeptidase Complex/metabolism , Protein Binding/genetics , Protein Conformation , Repressor Proteins/genetics , Trans-Activators/genetics , Trans-Activators/metabolism , Ubiquitination
16.
Nature ; 523(7562): 561-7, 2015 Jul 30.
Article in English | MEDLINE | ID: mdl-26200343

ABSTRACT

G-protein-coupled receptors (GPCRs) signal primarily through G proteins or arrestins. Arrestin binding to GPCRs blocks G protein interaction and redirects signalling to numerous G-protein-independent pathways. Here we report the crystal structure of a constitutively active form of human rhodopsin bound to a pre-activated form of the mouse visual arrestin, determined by serial femtosecond X-ray laser crystallography. Together with extensive biochemical and mutagenesis data, the structure reveals an overall architecture of the rhodopsin-arrestin assembly in which rhodopsin uses distinct structural elements, including transmembrane helix 7 and helix 8, to recruit arrestin. Correspondingly, arrestin adopts the pre-activated conformation, with a ∼20° rotation between the amino and carboxy domains, which opens up a cleft in arrestin to accommodate a short helix formed by the second intracellular loop of rhodopsin. This structure provides a basis for understanding GPCR-mediated arrestin-biased signalling and demonstrates the power of X-ray lasers for advancing the frontiers of structural biology.


Subject(s)
Arrestin/chemistry , Arrestin/metabolism , Rhodopsin/chemistry , Rhodopsin/metabolism , Animals , Binding Sites , Crystallography, X-Ray , Disulfides/chemistry , Disulfides/metabolism , Humans , Lasers , Mice , Models, Molecular , Multiprotein Complexes/biosynthesis , Multiprotein Complexes/chemistry , Multiprotein Complexes/metabolism , Protein Binding , Reproducibility of Results , Signal Transduction , X-Rays
17.
J Biol Chem ; 294(3): 953-967, 2019 01 18.
Article in English | MEDLINE | ID: mdl-30478170

ABSTRACT

AMP-activated protein kinase (AMPK) is an attractive therapeutic target for managing metabolic diseases. A class of pharmacological activators, including Merck 991, binds the AMPK ADaM site, which forms the interaction surface between the kinase domain (KD) of the α-subunit and the carbohydrate-binding module (CBM) of the ß-subunit. Here, we report the development of two new 991-derivative compounds, R734 and R739, which potently activate AMPK in a variety of cell types, including ß2-specific skeletal muscle cells. Surprisingly, we found that they have only minor effects on direct kinase activity of the recombinant α1ß2γ1 isoform yet robustly enhance protection against activation loop dephosphorylation. This mode of activation is reminiscent of that of ADP, which activates AMPK by binding to the nucleotide-binding sites in the γ-subunit, more than 60 Å away from the ADaM site. To understand the mechanisms of full and partial AMPK activation, we determined the crystal structures of fully active phosphorylated AMPK α1ß1γ1 bound to AMP and R734/R739 as well as partially active nonphosphorylated AMPK bound to R734 and AMP and phosphorylated AMPK bound to R734 in the absence of added nucleotides at <3-Å resolution. These structures and associated analyses identified a novel conformational state of the AMPK autoinhibitory domain associated with partial kinase activity and provide new insights into phosphorylation-dependent activation loop stabilization in AMPK.


Subject(s)
AMP-Activated Protein Kinases/antagonists & inhibitors , AMP-Activated Protein Kinases/chemistry , Enzyme Activators/chemistry , AMP-Activated Protein Kinases/metabolism , Catalytic Domain , Hep G2 Cells , Humans , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism
18.
J Biol Chem ; 293(44): 16994-17007, 2018 11 02.
Article in English | MEDLINE | ID: mdl-30206123

ABSTRACT

AMP-activated protein kinase (AMPK) is a master regulator of energy homeostasis and a promising drug target for managing metabolic diseases such as type 2 diabetes. Many pharmacological AMPK activators, and possibly unidentified physiological metabolites, bind to the allosteric drug and metabolite (ADaM) site at the interface between the kinase domain (KD) in the α-subunit and the carbohydrate-binding module (CBM) in the ß-subunit. Here, using double electron-electron resonance (DEER) spectroscopy, we demonstrate that the CBM-KD interaction is partially dissociated and the interface highly disordered in the absence of pharmacological ADaM site activators as inferred from a low depth of modulation and broad DEER distance distributions. ADaM site ligands such as 991, and to a lesser degree phosphorylation, stabilize the KD-CBM association and strikingly reduce conformational heterogeneity in the ADaM site. Our findings that the ADaM site, formed by the KD-CBM interaction, can be modulated by diverse ligands and by phosphorylation suggest that it may function as a hub for integrating regulatory signals.


Subject(s)
AMP-Activated Protein Kinases/chemistry , AMP-Activated Protein Kinases/metabolism , AMP-Activated Protein Kinases/genetics , Adenosine Monophosphate/chemistry , Adenosine Monophosphate/metabolism , Allosteric Regulation , Benzimidazoles/chemistry , Benzimidazoles/metabolism , Benzoates/chemistry , Benzoates/metabolism , Binding Sites , Catalytic Domain , Crystallography, X-Ray , Humans , Ligands , Protein Conformation , Protein Domains
19.
Nature ; 500(7463): 486-9, 2013 Aug 22.
Article in English | MEDLINE | ID: mdl-23851396

ABSTRACT

Folate receptors (FRα, FRß and FRγ) are cysteine-rich cell-surface glycoproteins that bind folate with high affinity to mediate cellular uptake of folate. Although expressed at very low levels in most tissues, folate receptors, especially FRα, are expressed at high levels in numerous cancers to meet the folate demand of rapidly dividing cells under low folate conditions. The folate dependency of many tumours has been therapeutically and diagnostically exploited by administration of anti-FRα antibodies, high-affinity antifolates, folate-based imaging agents and folate-conjugated drugs and toxins. To understand how folate binds its receptors, we determined the crystal structure of human FRα in complex with folic acid at 2.8 Å resolution. FRα has a globular structure stabilized by eight disulphide bonds and contains a deep open folate-binding pocket comprised of residues that are conserved in all receptor subtypes. The folate pteroate moiety is buried inside the receptor, whereas its glutamate moiety is solvent-exposed and sticks out of the pocket entrance, allowing it to be conjugated to drugs without adversely affecting FRα binding. The extensive interactions between the receptor and ligand readily explain the high folate-binding affinity of folate receptors and provide a template for designing more specific drugs targeting the folate receptor system.


Subject(s)
Folate Receptor 1/chemistry , Folate Receptor 1/metabolism , Folic Acid/metabolism , Binding Sites/genetics , Crystallography, X-Ray , Folate Receptor 1/genetics , Folic Acid/chemistry , Humans , Ligands , Models, Molecular , Mutation , Protein Binding , Structure-Activity Relationship
20.
Acta Pharmacol Sin ; 40(10): 1364-1372, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31253939

ABSTRACT

Arthrobacter globiformis Uricase (AgUricase) is a homotetrameric uricase with the potential for therapeutic use in treating hyperuricemia-related diseases. To achieve sufficient therapeutic effects, it is essential for this enzyme to have high thermostability and long half-life in physiological condition. To improve the thermostability of this enzyme, we introduced a series of cysteine pair mutations into the AgUricase subunits based on its structural model and studied the thermostability of the mutant enzymes with introduced disulfide bridges. Two intersubunit cysteine pair mutations, K12C-E286C and S296C-S296C, were found to markedly increase the melting temperatures of the corresponding mutant enzymes compared with WT AgUricase. The crystal structure of the K12C-E286C mutant at 1.99 Å resolution confirmed the formation of a distinct disulfide bond between the two subunits in the dimer. Structural analysis and biochemical data revealed that the C-terminal loop of AgUricase was flexible, and its interaction with neighboring subunits was required for the stability of the enzyme. We introduced an additional intersubunit K244C-C302 disulfide bond based on the crystal structure of the K12C-E286C mutant and confirmed that this additional disulfide bond further stabilized the flexible C-terminal loop and improved the thermostability of the enzyme. Disulfide cross-linking also protected AgUricase from protease digestion. Our studies suggest that the introduction of disulfide bonds into proteins is a potential strategy for enhancing the thermostability of multimeric proteins for medical applications.


Subject(s)
Gout/metabolism , Hyperuricemia/metabolism , Temperature , Urate Oxidase/metabolism , Arthrobacter/enzymology , Crystallography, X-Ray , Enzyme Stability , Gout/therapy , Hyperuricemia/therapy , Models, Molecular , Protein Conformation , Urate Oxidase/chemistry , Urate Oxidase/isolation & purification
SELECTION OF CITATIONS
SEARCH DETAIL