Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 40
1.
Nat Commun ; 15(1): 3827, 2024 May 07.
Article En | MEDLINE | ID: mdl-38714735

The main protease (Mpro) of SARS-CoV-2 is critical for viral function and a key drug target. Mpro is only active when reduced; turnover ceases upon oxidation but is restored by re-reduction. This suggests the system has evolved to survive periods in an oxidative environment, but the mechanism of this protection has not been confirmed. Here, we report a crystal structure of oxidized Mpro showing a disulfide bond between the active site cysteine, C145, and a distal cysteine, C117. Previous work proposed this disulfide provides the mechanism of protection from irreversible oxidation. Mpro forms an obligate homodimer, and the C117-C145 structure shows disruption of interactions bridging the dimer interface, implying a correlation between oxidation and dimerization. We confirm dimer stability is weakened in solution upon oxidation. Finally, we observe the protein's crystallization behavior is linked to its redox state. Oxidized Mpro spontaneously forms a distinct, more loosely packed lattice. Seeding with crystals of this lattice yields a structure with an oxidation pattern incorporating one cysteine-lysine-cysteine (SONOS) and two lysine-cysteine (NOS) bridges. These structures further our understanding of the oxidative regulation of Mpro and the crystallization conditions necessary to study this structurally.


Catalytic Domain , Coronavirus 3C Proteases , Cysteine , Disulfides , Oxidation-Reduction , SARS-CoV-2 , Disulfides/chemistry , Disulfides/metabolism , SARS-CoV-2/metabolism , SARS-CoV-2/chemistry , Coronavirus 3C Proteases/metabolism , Coronavirus 3C Proteases/chemistry , Cysteine/chemistry , Cysteine/metabolism , Crystallography, X-Ray , Humans , Models, Molecular , Protein Multimerization , COVID-19/virology
2.
Curr Opin Struct Biol ; 80: 102601, 2023 06.
Article En | MEDLINE | ID: mdl-37182397

The past century has witnessed an exponential increase in our atomic-level understanding of molecular and cellular mechanisms from a structural perspective, with multiple landmark achievements contributing to the field. This, coupled with recent and continuing breakthroughs in artificial intelligence methods such as AlphaFold2, and enhanced computational power, is enabling our understanding of protein structure and function at unprecedented levels of accuracy and predictivity. Here, we describe some of the major recent advances across these fields, and describe, as these technologies coalesce, the potential to utilise our enhanced knowledge of intricate cellular and molecular systems to discover novel therapeutics to alleviate human suffering.


Artificial Intelligence , Biology , Humans
3.
IUCrJ ; 7(Pt 6): 965-975, 2020 Nov 01.
Article En | MEDLINE | ID: mdl-33209311

Long-wavelength pulses from the Swiss X-ray free-electron laser (XFEL) have been used for de novo protein structure determination by native single-wavelength anomalous diffraction (native-SAD) phasing of serial femtosecond crystallography (SFX) data. In this work, sensitive anomalous data-quality indicators and model proteins were used to quantify improvements in native-SAD at XFELs such as utilization of longer wavelengths, careful experimental geometry optimization, and better post-refinement and partiality correction. Compared with studies using shorter wavelengths at other XFELs and older software versions, up to one order of magnitude reduction in the required number of indexed images for native-SAD was achieved, hence lowering sample consumption and beam-time requirements significantly. Improved data quality and higher anomalous signal facilitate so-far underutilized de novo structure determination of challenging proteins at XFELs. Improvements presented in this work can be used in other types of SFX experiments that require accurate measurements of weak signals, for example time-resolved studies.

4.
J Immunother Cancer ; 8(2)2020 07.
Article En | MEDLINE | ID: mdl-32727810

Accumulation of extracellular adenosine within the microenvironment is a strategy exploited by tumors to escape detection by the immune system. Adenosine signaling through the adenosine 2A receptor (A2AR) on immune cells elicits a range of immunosuppressive effects which promote tumor growth and limit the efficacy of immune checkpoint inhibitors. Preclinical data with A2AR inhibitors have demonstrated tumor regressions in mouse models by rescuing T cell function; however, the mechanism and role on other immune cells has not been fully elucidated. METHODS: We report here the development of a small molecule A2AR inhibitor including characterization of binding and inhibition of A2AR function with varying amounts of a stable version of adenosine. Functional activity was tested in both mouse and human T cells and dendritic cells (DCs) in in vitro assays to understand the intrinsic role on each cell type. The role of adenosine and A2AR inhibition was tested in DC differentiation assays as well as co-culture assays to access the cross-priming function of DCs. Syngeneic models were used to assess tumor growth alone and in combination with alphaprogrammed death-ligand 1 (αPD-L1). Immunophenotyping by flow cytometry was performed to examine global immune cell changes upon A2AR inhibition. RESULTS: We provide the first report of AZD4635, a novel small molecule A2AR antagonist which inhibits downstream signaling and increases T cell function as well as a novel mechanism of enhancing antigen presentation by CD103+ DCs. The role of antigen presentation by DCs, particularly CD103+ DCs, is critical to drive antitumor immunity providing rational to combine a priming agent AZD4635 with check point blockade. We find adenosine impairs the maturation and antigen presentation function of CD103+ DCs. We show in multiple syngeneic mouse tumor models that treatment of AZD4635 alone and in combination with αPD-L1 led to decreased tumor volume correlating with enhanced CD103+ function and T cell response. We extend these studies into human DCs to show that adenosine promotes a tolerogenic phenotype that can be reversed with AZD4635 restoring antigen-specific T cell activation. Our results support the novel role of adenosine signaling as an intrinsic negative regulator of CD103+ DCs maturation and priming. We show that potent inhibition of A2AR with AZD4635 reduces tumor burden and enhances antitumor immunity. This unique mechanism of action in CD103+ DCs may contribute to clinical responses as AZD4635 is being evaluated in clinical trials with IMFINZI (durvalumab, αPD-L1) in patients with solid malignancies. CONCLUSION: We provide evidence implicating suppression of adaptive and innate immunity by adenosine as a mechanism for immune evasion by tumors. Inhibition of adenosine signaling through selective small molecule inhibition of A2AR using AZD4635 restores T cell function via an internal mechanism as well as tumor antigen cross-presentation by CD103+ DCs resulting in antitumor immunity.


Antigens, CD/metabolism , Antineoplastic Agents, Immunological/therapeutic use , Dendritic Cells/immunology , Integrin alpha Chains/metabolism , Neoplasms/immunology , Receptor, Adenosine A2A/metabolism , Antineoplastic Agents, Immunological/pharmacology , Cell Line, Tumor , Female , Humans , Male , Signal Transduction
5.
Angew Chem Int Ed Engl ; 59(38): 16536-16543, 2020 09 14.
Article En | MEDLINE | ID: mdl-32542862

We present a robust protocol based on iterations of free energy perturbation (FEP) calculations, chemical synthesis, biophysical mapping and X-ray crystallography to reveal the binding mode of an antagonist series to the A2A adenosine receptor (AR). Eight A2A AR binding site mutations from biophysical mapping experiments were initially analyzed with sidechain FEP simulations, performed on alternate binding modes. The results distinctively supported one binding mode, which was subsequently used to design new chromone derivatives. Their affinities for the A2A AR were experimentally determined and investigated through a cycle of ligand-FEP calculations, validating the binding orientation of the different chemical substituents proposed. Subsequent X-ray crystallography of the A2A AR with a low and a high affinity chromone derivative confirmed the predicted binding orientation. The new molecules and structures here reported were driven by free energy calculations, and provide new insights on antagonist binding to the A2A AR, an emerging target in immuno-oncology.


Purinergic P1 Receptor Antagonists/chemistry , Receptor, Adenosine A2A/chemistry , Thermodynamics , Binding Sites/drug effects , Crystallography, X-Ray , Humans , Models, Molecular , Molecular Structure , Purinergic P1 Receptor Antagonists/pharmacology , Receptor, Adenosine A2A/metabolism
6.
J Med Chem ; 63(4): 1528-1543, 2020 02 27.
Article En | MEDLINE | ID: mdl-31860301

The orexin system, which consists of the two G protein-coupled receptors OX1 and OX2, activated by the neuropeptides OX-A and OX-B, is firmly established as a key regulator of behavioral arousal, sleep, and wakefulness and has been an area of intense research effort over the past two decades. X-ray structures of the receptors in complex with 10 new antagonist ligands from diverse chemotypes are presented, which complement the existing structural information for the system and highlight the critical importance of lipophilic hotspots and water molecules for these peptidergic GPCR targets. Learnings from the structural information regarding the utility of pharmacophore models and how selectivity between OX1 and OX2 can be achieved are discussed.


Orexin Receptor Antagonists/metabolism , Orexin Receptors/metabolism , Binding Sites , Computer Simulation , Crystallography, X-Ray , HEK293 Cells , Humans , Hydrogen Bonding , Hydrophobic and Hydrophilic Interactions , Ligands , Orexin Receptor Antagonists/chemistry , Orexin Receptors/chemistry
7.
Science ; 364(6442): 775-778, 2019 05 24.
Article En | MEDLINE | ID: mdl-31072904

G protein-coupled receptors (GPCRs) in the G protein-coupled active state have higher affinity for agonists as compared with when they are in the inactive state, but the molecular basis for this is unclear. We have determined four active-state structures of the ß1-adrenoceptor (ß1AR) bound to conformation-specific nanobodies in the presence of agonists of varying efficacy. Comparison with inactive-state structures of ß1AR bound to the identical ligands showed a 24 to 42% reduction in the volume of the orthosteric binding site. Potential hydrogen bonds were also shorter, and there was up to a 30% increase in the number of atomic contacts between the receptor and ligand. This explains the increase in agonist affinity of GPCRs in the active state for a wide range of structurally distinct agonists.


Adrenergic beta-1 Receptor Agonists/chemistry , Drug Design , Receptors, G-Protein-Coupled/agonists , Adrenergic beta-1 Receptor Agonists/pharmacology , Allosteric Site/immunology , Catalytic Domain/immunology , Hydrogen Bonding , Ligands , Protein Binding , Protein Structure, Secondary , Receptors, Adrenergic, beta-1/chemistry , Receptors, Adrenergic, beta-1/immunology , Receptors, G-Protein-Coupled/chemistry , Receptors, G-Protein-Coupled/immunology , Single-Domain Antibodies/immunology
8.
Nat Commun ; 10(1): 17, 2019 01 03.
Article En | MEDLINE | ID: mdl-30604743

Neurokinins (or tachykinins) are peptides that modulate a wide variety of human physiology through the neurokinin G protein-coupled receptor family, implicated in a diverse array of pathological processes. Here we report high-resolution crystal structures of the human NK1 receptor (NK1R) bound to two small-molecule antagonist therapeutics - aprepitant and netupitant and the progenitor antagonist CP-99,994. The structures reveal the detailed interactions between clinically approved antagonists and NK1R, which induce a distinct receptor conformation resulting in an interhelical hydrogen-bond network that cross-links the extracellular ends of helices V and VI. Furthermore, the high-resolution details of NK1R bound to netupitant establish a structural rationale for the lack of basal activity in NK1R. Taken together, these co-structures provide a comprehensive structural basis of NK1R antagonism and will facilitate the design of new therapeutics targeting the neurokinin receptor family.


Neurokinin-1 Receptor Antagonists/chemistry , Receptors, Neurokinin-1/chemistry , Aprepitant/chemistry , Aprepitant/pharmacology , Binding Sites , Crystallography, X-Ray , Drug Design , HEK293 Cells , Humans , Molecular Dynamics Simulation , Neurokinin-1 Receptor Antagonists/pharmacology , Piperidines/chemistry , Piperidines/pharmacology , Protein Structure, Secondary , Pyridines/chemistry , Pyridines/pharmacology , Receptors, Neurokinin-1/isolation & purification , Receptors, Neurokinin-1/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Structure-Activity Relationship
9.
J Med Chem ; 62(1): 207-222, 2019 01 10.
Article En | MEDLINE | ID: mdl-29455526

Two interesting new X-ray structures of negative allosteric modulator (NAM) ligands for the mGlu5 receptor, M-MPEP (3) and fenobam (4), are reported. The new structures show how the binding of the ligands induces different receptor water channel conformations to previously published structures. The structure of fenobam, where a urea replaces the acetylenic linker in M-MPEP and mavoglurant, reveals a binding mode where the ligand is rotated by 180° compared to a previously proposed docking model. The need for multiple ligand structures for accurate GPCR structure-based drug design is demonstrated by the different growing vectors identified for the head groups of M-MPEP and mavoglurant and by the unexpected water-mediated receptor interactions of a new chemotype represented by fenobam. The implications of the new structures for ligand design are discussed, with extensive analysis of the energetics of the water networks of both pseudoapo and bound structures providing a new design strategy for allosteric modulators.


Receptor, Metabotropic Glutamate 5/chemistry , Allosteric Regulation , Allosteric Site , Crystallography, X-Ray , Drug Design , Humans , Imidazoles/chemistry , Imidazoles/metabolism , Indoles/chemistry , Indoles/metabolism , Ligands , Molecular Docking Simulation , Protein Structure, Tertiary , Pyridines/chemistry , Pyridines/metabolism , Receptor, Metabotropic Glutamate 5/metabolism , Thiazoles/chemistry , Thiazoles/metabolism , Water/chemistry
10.
Nat Struct Mol Biol ; 25(12): 1086-1092, 2018 12.
Article En | MEDLINE | ID: mdl-30455434

Parathyroid hormone 1 receptor (PTH1R) is a class B multidomain G-protein-coupled receptor (GPCR) that controls calcium homeostasis. Two endogenous peptide ligands, parathyroid hormone (PTH) and parathyroid hormone-related protein (PTHrP), activate the receptor, and their analogs teriparatide and abaloparatide are used in the clinic to increase bone formation as an effective yet costly treatment for osteoporosis. Activation of PTH1R involves binding of the peptide ligand to the receptor extracellular domain (ECD) and transmembrane domain (TMD), a hallmark of class B GPCRs. Here, we present the crystal structure of human PTH1R in complex with a peptide agonist at 2.5-Å resolution, allowing us to delineate the agonist binding mode for this receptor and revealing molecular details within conserved structural motifs that are critical for class B receptor function. Thus, this study provides structural insight into the function of PTH1R and extends our understanding of this therapeutically important class of GPCRs.


Receptor, Parathyroid Hormone, Type 1/chemistry , Amino Acid Sequence , Biomimetics , Crystallography, X-Ray , Humans , Models, Molecular , Parathyroid Hormone/chemistry , Peptides/metabolism , Protein Binding
11.
Nature ; 553(7686): 111-114, 2018 01 03.
Article En | MEDLINE | ID: mdl-29300009

The complement system is a crucial component of the host response to infection and tissue damage. Activation of the complement cascade generates anaphylatoxins including C5a and C3a. C5a exerts a pro-inflammatory effect via the G-protein-coupled receptor C5a anaphylatoxin chemotactic receptor 1 (C5aR1, also known as CD88) that is expressed on cells of myeloid origin. Inhibitors of the complement system have long been of interest as potential drugs for the treatment of diseases such as sepsis, rheumatoid arthritis, Crohn's disease and ischaemia-reperfusion injuries. More recently, a role of C5a in neurodegenerative conditions such as Alzheimer's disease has been identified. Peptide antagonists based on the C5a ligand have progressed to phase 2 trials in psoriasis and rheumatoid arthritis; however, these compounds exhibited problems with off-target activity, production costs, potential immunogenicity and poor oral bioavailability. Several small-molecule competitive antagonists for C5aR1, such as W-54011 and NDT9513727, have been identified by C5a radioligand-binding assays. NDT9513727 is a non-peptide inverse agonist of C5aR1, and is highly selective for the primate and gerbil receptors over those of other species. Here, to study the mechanism of action of C5a antagonists, we determine the structure of a thermostabilized C5aR1 (known as C5aR1 StaR) in complex with NDT9513727. We found that the small molecule bound between transmembrane helices 3, 4 and 5, outside the helical bundle. One key interaction between the small molecule and residue Trp2135.49 seems to determine the species selectivity of the compound. The structure demonstrates that NDT9513727 exerts its inverse-agonist activity through an extra-helical mode of action.


Benzodioxoles/chemistry , Benzodioxoles/metabolism , Imidazoles/chemistry , Imidazoles/metabolism , Receptor, Anaphylatoxin C5a/antagonists & inhibitors , Receptor, Anaphylatoxin C5a/chemistry , Animals , Benzodioxoles/pharmacology , Binding Sites , Crystallography, X-Ray , Drug Inverse Agonism , HEK293 Cells , Humans , Imidazoles/pharmacology , Models, Molecular , Mutation , Protein Stability , Protein Structure, Secondary , Receptor, Anaphylatoxin C5a/genetics , Receptor, Anaphylatoxin C5a/metabolism , Receptors, Adrenergic, beta-2/chemistry , Receptors, Adrenergic, beta-2/metabolism , Receptors, G-Protein-Coupled/chemistry , Receptors, G-Protein-Coupled/metabolism
12.
Sci Rep ; 8(1): 41, 2018 01 08.
Article En | MEDLINE | ID: mdl-29311713

Here we report an efficient method to generate multiple co-structures of the A2A G protein-coupled receptor (GPCR) with small-molecules from a single preparation of a thermostabilised receptor crystallised in Lipidic Cubic Phase (LCP). Receptor crystallisation is achieved following purification using a low affinity "carrier" ligand (theophylline) and crystals are then soaked in solutions containing the desired (higher affinity) compounds. Complete datasets to high resolution can then be collected from single crystals and seven structures are reported here of which three are novel. The method significantly improves structural throughput for ligand screening using stabilised GPCRs, thereby actively driving Structure-Based Drug Discovery (SBDD).


Receptor, Adenosine A2A/chemistry , Receptors, G-Protein-Coupled/chemistry , Crystallography, X-Ray , Humans , Ligands , Models, Molecular , Molecular Conformation , Protein Unfolding , Receptor, Adenosine A2A/metabolism , Receptors, G-Protein-Coupled/metabolism
13.
Nat Commun ; 8(1): 542, 2017 09 14.
Article En | MEDLINE | ID: mdl-28912485

Historically, room-temperature structure determination was succeeded by cryo-crystallography to mitigate radiation damage. Here, we demonstrate that serial millisecond crystallography at a synchrotron beamline equipped with high-viscosity injector and high frame-rate detector allows typical crystallographic experiments to be performed at room-temperature. Using a crystal scanning approach, we determine the high-resolution structure of the radiation sensitive molybdenum storage protein, demonstrate soaking of the drug colchicine into tubulin and native sulfur phasing of the human G protein-coupled adenosine receptor. Serial crystallographic data for molecular replacement already converges in 1,000-10,000 diffraction patterns, which we collected in 3 to maximally 82 minutes. Compared with serial data we collected at a free-electron laser, the synchrotron data are of slightly lower resolution, however fewer diffraction patterns are needed for de novo phasing. Overall, the data we collected by room-temperature serial crystallography are of comparable quality to cryo-crystallographic data and can be routinely collected at synchrotrons.Serial crystallography was developed for protein crystal data collection with X-ray free-electron lasers. Here the authors present several examples which show that serial crystallography using high-viscosity injectors can also be routinely employed for room-temperature data collection at synchrotrons.

14.
Structure ; 25(8): 1275-1285.e4, 2017 08 01.
Article En | MEDLINE | ID: mdl-28712806

The adenosine A1 and A2A receptors belong to the purinergic family of G protein-coupled receptors, and regulate diverse functions of the cardiovascular, respiratory, renal, inflammation, and CNS. Xanthines such as caffeine and theophylline are weak, non-selective antagonists of adenosine receptors. Here we report the structure of a thermostabilized human A1 receptor at 3.3 Å resolution with PSB36, an A1-selective xanthine-based antagonist. This is compared with structures of the A2A receptor with PSB36 (2.8 Å resolution), caffeine (2.1 Å), and theophylline (2.0 Å) to highlight features of ligand recognition which are common across xanthines. The structures of A1R and A2AR were analyzed to identify the differences that are important selectivity determinants for xanthine ligands, and the role of T2707.35 in A1R (M2707.35 in A2AR) in conferring selectivity was confirmed by mutagenesis. The structural differences confirmed to lead to selectivity can be utilized in the design of new subtype-selective A1R or A2AR antagonists.


Caffeine/pharmacology , Receptor, Adenosine A1/chemistry , Receptor, Adenosine A2A/chemistry , Theophylline/pharmacology , Binding Sites , Caffeine/chemistry , HEK293 Cells , Humans , Molecular Docking Simulation , Protein Binding , Receptor, Adenosine A1/metabolism , Receptor, Adenosine A2A/metabolism , Substrate Specificity , Theophylline/chemistry
16.
Nature ; 546(7657): 254-258, 2017 06 08.
Article En | MEDLINE | ID: mdl-28562585

Glucagon-like peptide 1 (GLP-1) regulates glucose homeostasis through the control of insulin release from the pancreas. GLP-1 peptide agonists are efficacious drugs for the treatment of diabetes. To gain insight into the molecular mechanism of action of GLP-1 peptides, here we report the crystal structure of the full-length GLP-1 receptor bound to a truncated peptide agonist. The peptide agonist retains an α-helical conformation as it sits deep within the receptor-binding pocket. The arrangement of the transmembrane helices reveals hallmarks of an active conformation similar to that observed in class A receptors. Guided by this structural information, we design peptide agonists with potent in vivo activity in a mouse model of diabetes.


Glucagon-Like Peptide-1 Receptor/agonists , Glucagon-Like Peptide-1 Receptor/chemistry , Peptides/chemistry , Peptides/pharmacology , Animals , Binding Sites , Crystallography, X-Ray , Dose-Response Relationship, Drug , Glucagon-Like Peptide-1 Receptor/metabolism , Humans , Male , Mice , Models, Molecular , Peptides/metabolism , Protein Conformation , Rats , Receptors, Corticotropin-Releasing Hormone/chemistry , Receptors, Glucagon/chemistry
17.
Nature ; 545(7652): 112-115, 2017 05 04.
Article En | MEDLINE | ID: mdl-28445455

Protease-activated receptors (PARs) are a family of G-protein-coupled receptors (GPCRs) that are irreversibly activated by proteolytic cleavage of the N terminus, which unmasks a tethered peptide ligand that binds and activates the transmembrane receptor domain, eliciting a cellular cascade in response to inflammatory signals and other stimuli. PARs are implicated in a wide range of diseases, such as cancer and inflammation. PARs have been the subject of major pharmaceutical research efforts but the discovery of small-molecule antagonists that effectively bind them has proved challenging. The only marketed drug targeting a PAR is vorapaxar, a selective antagonist of PAR1 used to prevent thrombosis. The structure of PAR1 in complex with vorapaxar has been reported previously. Despite sequence homology across the PAR isoforms, discovery of PAR2 antagonists has been less successful, although GB88 has been described as a weak antagonist. Here we report crystal structures of PAR2 in complex with two distinct antagonists and a blocking antibody. The antagonist AZ8838 binds in a fully occluded pocket near the extracellular surface. Functional and binding studies reveal that AZ8838 exhibits slow binding kinetics, which is an attractive feature for a PAR2 antagonist competing against a tethered ligand. Antagonist AZ3451 binds to a remote allosteric site outside the helical bundle. We propose that antagonist binding prevents structural rearrangements required for receptor activation and signalling. We also show that a blocking antibody antigen-binding fragment binds to the extracellular surface of PAR2, preventing access of the tethered ligand to the peptide-binding site. These structures provide a basis for the development of selective PAR2 antagonists for a range of therapeutic uses.


Receptor, PAR-2/chemistry , Receptor, PAR-2/metabolism , Allosteric Regulation/drug effects , Allosteric Site/drug effects , Antibodies, Blocking/chemistry , Antibodies, Blocking/pharmacology , Benzimidazoles/chemistry , Benzimidazoles/pharmacology , Benzodioxoles/chemistry , Benzodioxoles/pharmacology , Benzyl Alcohols/chemistry , Benzyl Alcohols/pharmacology , Crystallography, X-Ray , Humans , Imidazoles/chemistry , Imidazoles/pharmacology , Immunoglobulin Fab Fragments/chemistry , Immunoglobulin Fab Fragments/pharmacology , Kinetics , Ligands , Models, Molecular , Receptor, PAR-2/antagonists & inhibitors , Signal Transduction/drug effects
18.
Curr Mol Pharmacol ; 10(4): 334-344, 2017.
Article En | MEDLINE | ID: mdl-28183242

The structural analysis of class B G protein-coupled receptors (GPCR), cell surface proteins responding to peptide hormones, has until recently been restricted to the extracellular domain (ECD). Corticotropin-releasing factor receptor type 1 (CRF1R) is a class B receptor mediating stress response and also considered a drug target for depression and anxiety. Here we report the crystal structure of the transmembrane domain of human CRF1R in complex with the small-molecule antagonist CP-376395 in a hexagonal setting with translational non-crystallographic symmetry. Molecular dynamics and metadynamics simulations on this novel structure and the existing TMD structure for CRF1R provides insight as to how the small molecule ligand gains access to the induced-fit allosteric binding site with implications for the observed selectivity against CRF2R. Furthermore, molecular dynamics simulations performed using a full-length receptor model point to key interactions between the ECD and extracellular loop 3 of the TMD providing insight into the full inactive state of multidomain class B GPCRs.


Receptors, Corticotropin-Releasing Hormone/chemistry , Allosteric Site , Aminopyridines/pharmacology , Binding Sites , Crystallography, X-Ray/methods , Humans , Molecular Dynamics Simulation , Protein Conformation , Receptors, Corticotropin-Releasing Hormone/antagonists & inhibitors , Receptors, Corticotropin-Releasing Hormone/metabolism
19.
Curr Top Med Chem ; 17(1): 71-78, 2017.
Article En | MEDLINE | ID: mdl-27448651

Class C G protein-coupled receptors encompass a range of promising therapeutic targets for a variety of diseases, yet to date only two members of this sub-family of GPCRs have been drugged. Recent advances in structural biology have revealed the X-ray crystallographic structures of allosteric ligands bound to two Class C metabotropic glutamate (mGlu) receptors, mGlu1 and mGlu5. Herein, we review how this information can be leveraged to help understand some of the historical challenges of mGlu receptor allosteric modulator drug discovery, and discuss how the structural enablement can be prospectively used for structurebased drug discovery approaches across Class C GPCR targets in general.


Drug Design , Receptors, G-Protein-Coupled/drug effects , Allosteric Regulation , Crystallography, X-Ray , Protein Conformation , Receptors, G-Protein-Coupled/chemistry , Receptors, G-Protein-Coupled/metabolism
20.
Nature ; 540(7633): 462-465, 2016 12 15.
Article En | MEDLINE | ID: mdl-27926729

Chemokines and their G-protein-coupled receptors play a diverse role in immune defence by controlling the migration, activation and survival of immune cells. They are also involved in viral entry, tumour growth and metastasis and hence are important drug targets in a wide range of diseases. Despite very significant efforts by the pharmaceutical industry to develop drugs, with over 50 small-molecule drugs directed at the family entering clinical development, only two compounds have reached the market: maraviroc (CCR5) for HIV infection and plerixafor (CXCR4) for stem-cell mobilization. The high failure rate may in part be due to limited understanding of the mechanism of action of chemokine antagonists and an inability to optimize compounds in the absence of structural information. CC chemokine receptor type 9 (CCR9) activation by CCL25 plays a key role in leukocyte recruitment to the gut and represents a therapeutic target in inflammatory bowel disease. The selective CCR9 antagonist vercirnon progressed to phase 3 clinical trials in Crohn's disease but efficacy was limited, with the need for very high doses to block receptor activation. Here we report the crystal structure of the CCR9 receptor in complex with vercirnon at 2.8 Å resolution. Remarkably, vercirnon binds to the intracellular side of the receptor, exerting allosteric antagonism and preventing G-protein coupling. This binding site explains the need for relatively lipophilic ligands and describes another example of an allosteric site on G-protein-coupled receptors that can be targeted for drug design, not only at CCR9, but potentially extending to other chemokine receptors.


Receptors, CCR/antagonists & inhibitors , Receptors, CCR/chemistry , Sulfonamides/chemistry , Sulfonamides/pharmacology , Allosteric Regulation/drug effects , Allosteric Site/drug effects , Allosteric Site/genetics , Conserved Sequence , Crystallography, X-Ray , Cytoplasm/metabolism , Drug Design , Heterotrimeric GTP-Binding Proteins/antagonists & inhibitors , Heterotrimeric GTP-Binding Proteins/metabolism , Humans , Ligands , Models, Molecular , Mutagenesis , Receptors, CCR/genetics , Receptors, CCR5/chemistry , Receptors, CXCR4/chemistry
...