Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
Science ; 382(6677): eadh1859, 2023 12 22.
Article in English | MEDLINE | ID: mdl-38127743

ABSTRACT

Heterotrimeric guanine nucleotide-binding protein (G protein)-coupled receptors (GPCRs) bind to extracellular ligands and drugs and modulate intracellular responses through conformational changes. Despite their importance as drug targets, the molecular origins of pharmacological properties such as efficacy (maximum signaling response) and potency (the ligand concentration at half-maximal response) remain poorly understood for any ligand-receptor-signaling system. We used the prototypical adrenaline-ß2 adrenergic receptor-G protein system to reveal how specific receptor residues decode and translate the information encoded in a ligand to mediate a signaling response. We present a data science framework to integrate pharmacological and structural data to uncover structural changes and allosteric networks relevant for ligand pharmacology. These methods can be tailored to study any ligand-receptor-signaling system, and the principles open possibilities for designing orthosteric and allosteric compounds with defined signaling properties.


Subject(s)
Adrenergic beta-2 Receptor Agonists , Receptors, Adrenergic, beta-2 , Humans , Adrenergic beta-2 Receptor Agonists/chemistry , Adrenergic beta-2 Receptor Agonists/pharmacology , Allosteric Regulation , Biosensing Techniques , Ligands , Protein Conformation , Receptors, Adrenergic, beta-2/chemistry , Receptors, Adrenergic, beta-2/genetics , Signal Transduction , Bioluminescence Resonance Energy Transfer Techniques
2.
Biochem Soc Trans ; 51(1): 13-20, 2023 02 27.
Article in English | MEDLINE | ID: mdl-36688421

ABSTRACT

G protein-coupled receptor (GPCR) family members can sense an extraordinary variety of biomolecules to activate intracellular signalling cascades that modulate key aspects of cell physiology. Apart from their crucial role in maintaining cell homeostasis, these critical sensory and modulatory properties have made GPCRs the most successful drug target class to date. However, establishing direct links between receptor activation of specific intracellular partners and individual physiological outcomes is still an ongoing challenge. By studying this receptor signalling complexity at increasing resolution through the development of novel biosensors and high-throughput techniques, a growing number of studies are revealing how receptor function can be diversified in a spatial, temporal or cell-specific manner. This mini-review will introduce recent examples of this context-dependent receptor signalling and discuss how it can impact our understanding of receptor function in health and disease, and contribute to the search of more selective, efficacious and safer GPCR drug candidates.


Subject(s)
Receptors, G-Protein-Coupled , Signal Transduction , Receptors, G-Protein-Coupled/physiology , Cell Membrane , Drug Delivery Systems , Ligands
3.
Cell Rep ; 34(12): 108862, 2021 03 23.
Article in English | MEDLINE | ID: mdl-33761344

ABSTRACT

The Melanocortin-4 Receptor (MC4R) plays a pivotal role in energy homeostasis. We used human MC4R mutations associated with an increased or decreased risk of obesity to dissect mechanisms that regulate MC4R function. Most obesity-associated mutations impair trafficking to the plasma membrane (PM), whereas obesity-protecting mutations either accelerate recycling to the PM or decrease internalization, resulting in enhanced signaling. MC4R mutations that do not affect canonical Gαs protein-mediated signaling, previously considered to be non-pathogenic, nonetheless disrupt agonist-induced internalization, ß-arrestin recruitment, and/or coupling to Gαs, establishing their causal role in severe obesity. Structural mapping reveals ligand-accessible sites by which MC4R couples to effectors and residues involved in the homodimerization of MC4R, which is disrupted by multiple obesity-associated mutations. Human genetic studies reveal that endocytosis, intracellular trafficking, and homodimerization regulate MC4R function to a level that is physiologically relevant, supporting the development of chaperones, agonists, and allosteric modulators of MC4R for weight loss therapy.


Subject(s)
Body Weight/genetics , Endocytosis , Genetic Variation , Protein Multimerization , Receptor, Melanocortin, Type 4/genetics , Animals , COS Cells , Cell Membrane/metabolism , Chlorocebus aethiops , Cyclic AMP/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , GTP-Binding Protein alpha Subunits, Gs , HEK293 Cells , Humans , Models, Biological , Mutant Proteins/metabolism , Mutation/genetics , Phosphorylation , Receptor, Melanocortin, Type 4/chemistry , Signal Transduction , beta-Arrestins/metabolism
4.
Molecules ; 26(3)2021 Jan 26.
Article in English | MEDLINE | ID: mdl-33530327

ABSTRACT

While selective inhibition is one of the key assets for a small molecule drug, many diseases can only be tackled by simultaneous inhibition of several proteins. An example where achieving selectivity is especially challenging are ligands targeting human kinases. This difficulty arises from the high structural conservation of the kinase ATP binding sites, the area targeted by most inhibitors. We investigated the possibility to identify novel small molecule ligands with pre-defined binding profiles for a series of kinase targets and anti-targets by in silico docking. The candidate ligands originating from these calculations were assayed to determine their experimental binding profiles. Compared to previous studies, the acquired hit rates were low in this specific setup, which aimed at not only selecting multi-target kinase ligands, but also designing out binding to anti-targets. Specifically, only a single profiled substance could be verified as a sub-micromolar, dual-specific EGFR/ErbB2 ligand that indeed avoided its selected anti-target BRAF. We subsequently re-analyzed our target choice and in silico strategy based on these findings, with a particular emphasis on the hit rates that can be expected from a given target combination. To that end, we supplemented the structure-based docking calculations with bioinformatic considerations of binding pocket sequence and structure similarity as well as ligand-centric comparisons of kinases. Taken together, our results provide a multi-faceted picture of how pocket space can determine the success of docking in multi-target drug discovery efforts.


Subject(s)
Molecular Docking Simulation/methods , Protein Kinases/chemistry , Protein Kinases/metabolism , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Adenosine Triphosphate/metabolism , Binding Sites , Computer Simulation , Drug Discovery , ErbB Receptors/chemistry , ErbB Receptors/metabolism , Humans , Ligands , Models, Molecular , Molecular Conformation , Proto-Oncogene Proteins B-raf/chemistry , Proto-Oncogene Proteins B-raf/metabolism , Structure-Activity Relationship
6.
Nature ; 587(7835): 650-656, 2020 11.
Article in English | MEDLINE | ID: mdl-33149304

ABSTRACT

G-protein-coupled receptors (GPCRs) are membrane proteins that modulate physiology across human tissues in response to extracellular signals. GPCR-mediated signalling can differ because of changes in the sequence1,2 or expression3 of the receptors, leading to signalling bias when comparing diverse physiological systems4. An underexplored source of such bias is the generation of functionally diverse GPCR isoforms with different patterns of expression across different tissues. Here we integrate data from human tissue-level transcriptomes, GPCR sequences and structures, proteomics, single-cell transcriptomics, population-wide genetic association studies and pharmacological experiments. We show how a single GPCR gene can diversify into several isoforms with distinct signalling properties, and how unique isoform combinations expressed in different tissues can generate distinct signalling states. Depending on their structural changes and expression patterns, some of the detected isoforms may influence cellular responses to drugs and represent new targets for developing drugs with improved tissue selectivity. Our findings highlight the need to move from a canonical to a context-specific view of GPCR signalling that considers how combinatorial expression of isoforms in a particular cell type, tissue or organism collectively influences receptor signalling and drug responses.


Subject(s)
Protein Isoforms/chemistry , Protein Isoforms/metabolism , Receptors, G-Protein-Coupled/chemistry , Receptors, G-Protein-Coupled/metabolism , Signal Transduction/drug effects , Transcriptome , Databases, Factual , Gene Expression Profiling , HEK293 Cells , Humans , Molecular Targeted Therapy , Organ Specificity/drug effects , Protein Isoforms/genetics , Proteomics , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, G-Protein-Coupled/genetics , Signal Transduction/genetics , Single-Cell Analysis
7.
Neuron ; 108(6): 1146-1162.e10, 2020 12 23.
Article in English | MEDLINE | ID: mdl-33086039

ABSTRACT

Astrocytes tile the central nervous system and are widely implicated in brain diseases, but the molecular mechanisms by which astrocytes contribute to brain disorders remain incompletely explored. By performing astrocyte gene expression analyses following 14 experimental perturbations of relevance to the striatum, we discovered that striatal astrocytes mount context-specific molecular responses at the level of genes, pathways, and upstream regulators. Through data mining, we also identified astrocyte pathways in Huntington's disease (HD) that were reciprocally altered with respect to the activation of striatal astrocyte G protein-coupled receptor (GPCR) signaling. Furthermore, selective striatal astrocyte stimulation of the Gi-GPCR pathway in vivo corrected several HD-associated astrocytic, synaptic, and behavioral phenotypes, with accompanying improvement of HD-associated astrocyte signaling pathways, including those related to synaptogenesis and neuroimmune functions. Overall, our data show that astrocytes are malleable, using context-specific responses that can be dissected molecularly and used for phenotypic benefit in brain disorders.


Subject(s)
Astrocytes/metabolism , Corpus Striatum/metabolism , Huntington Disease/metabolism , Animals , Data Mining , Humans , Huntington Disease/genetics , Mice , Neurons/metabolism , Signal Transduction/physiology , Synapses/metabolism
8.
Nat Methods ; 17(8): 777-787, 2020 08.
Article in English | MEDLINE | ID: mdl-32661425

ABSTRACT

G-protein-coupled receptors (GPCRs) are involved in numerous physiological processes and are the most frequent targets of approved drugs. The explosion in the number of new three-dimensional (3D) molecular structures of GPCRs (3D-GPCRome) over the last decade has greatly advanced the mechanistic understanding and drug design opportunities for this protein family. Molecular dynamics (MD) simulations have become a widely established technique for exploring the conformational landscape of proteins at an atomic level. However, the analysis and visualization of MD simulations require efficient storage resources and specialized software. Here we present GPCRmd (http://gpcrmd.org/), an online platform that incorporates web-based visualization capabilities as well as a comprehensive and user-friendly analysis toolbox that allows scientists from different disciplines to visualize, analyze and share GPCR MD data. GPCRmd originates from a community-driven effort to create an open, interactive and standardized database of GPCR MD simulations.


Subject(s)
Molecular Dynamics Simulation , Receptors, G-Protein-Coupled/chemistry , Software , Metabolome , Models, Molecular , Protein Conformation
10.
FEBS J ; 287(4): 736-748, 2020 02.
Article in English | MEDLINE | ID: mdl-31386272

ABSTRACT

The δ-opioid receptor (δ-OR) couples to Gi/Go proteins to modulate a variety of responses in the nervous system. Τhe regulator of G protein signalling 4 (RGS4) was previously shown to directly interact within the C-terminal region of δ-OR using its N-terminal domain to negatively modulate opioid receptor signalling. Herein, using molecular dynamics simulations and in vitro pull-down experiments we delimit this interaction to 12 helix 8 residues of δ-ΟR and to the first 17 N-terminal residues (NT) of RGS4. Monitoring the complex arrangement and stabilization between RGS4 and δ-OR by molecular dynamics simulations combined with mutagenesis studies, we defined that two critical interactions are formed: one between Phe329 of helix8 of δ-ΟR and Pro9 of the NT of RGS4 and the other a salt bridge between Glu323 of δ-ΟR and Lys17 of RGS4. Our observations allow drafting for the first time a structural model of a ternary complex including the δ-opioid receptor, a G protein and a RGS protein. Furthermore, the high degree of conservation among opioid receptors of the RGS4-binding region, points to a conserved interaction mode between opioid receptors and this important regulatory protein.


Subject(s)
GTP-Binding Protein alpha Subunits, Gi-Go/chemistry , RGS Proteins/chemistry , Receptors, Opioid, delta/chemistry , Amino Acid Sequence , Animals , Binding Sites , Cattle , Cloning, Molecular , Conserved Sequence , Crystallography, X-Ray , Escherichia coli/genetics , Escherichia coli/metabolism , GTP-Binding Protein alpha Subunits, Gi-Go/genetics , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Humans , Molecular Dynamics Simulation , Mutation , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , RGS Proteins/genetics , RGS Proteins/metabolism , Receptors, Opioid, delta/genetics , Receptors, Opioid, delta/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid
12.
Nat Commun ; 10(1): 667, 2019 02 08.
Article in English | MEDLINE | ID: mdl-30737406

ABSTRACT

Class F receptors are considered valuable therapeutic targets due to their role in human disease, but structural changes accompanying receptor activation remain unexplored. Employing population and cancer genomics data, structural analyses, molecular dynamics simulations, resonance energy transfer-based approaches and mutagenesis, we identify a conserved basic amino acid in TM6 in Class F receptors that acts as a molecular switch to mediate receptor activation. Across all tested Class F receptors (FZD4,5,6,7, SMO), mutation of the molecular switch confers an increased potency of agonists by stabilizing an active conformation as assessed by engineered mini G proteins as conformational sensors. Disruption of the switch abrogates the functional interaction between FZDs and the phosphoprotein Dishevelled, supporting conformational selection as a prerequisite for functional selectivity. Our studies reveal the molecular basis of a common activation mechanism conserved in all Class F receptors, which facilitates assay development and future discovery of Class F receptor-targeting drugs.


Subject(s)
Models, Theoretical , Molecular Dynamics Simulation , Receptors, G-Protein-Coupled/metabolism , Flow Cytometry , HEK293 Cells , Humans , Immunoblotting , Mitogen-Activated Protein Kinase 1 , Mitogen-Activated Protein Kinase 3 , Models, Molecular , Phylogeny , Protein Binding , Protein Conformation , Receptors, G-Protein-Coupled/genetics
13.
Nat Biotechnol ; 37(2): 169-178, 2019 02.
Article in English | MEDLINE | ID: mdl-30607034

ABSTRACT

Existing high-throughput methods to identify RNA-binding proteins (RBPs) are based on capture of polyadenylated RNAs and cannot recover proteins that interact with nonadenylated RNAs, including long noncoding RNA, pre-mRNAs and bacterial RNAs. We present orthogonal organic phase separation (OOPS), which does not require molecular tagging or capture of polyadenylated RNA, and apply it to recover cross-linked protein-RNA and free protein, or protein-bound RNA and free RNA, in an unbiased way. We validated OOPS in HEK293, U2OS and MCF10A human cell lines, and show that 96% of proteins recovered were bound to RNA. We show that all long RNAs can be cross-linked to proteins, and recovered 1,838 RBPs, including 926 putative novel RBPs. OOPS is approximately 100-fold more efficient than existing methods and can enable analyses of dynamic RNA-protein interactions. We also characterize dynamic changes in RNA-protein interactions in mammalian cells following nocodazole arrest, and present a bacterial RNA-interactome for Escherichia coli. OOPS is compatible with downstream proteomics and RNA sequencing, and can be applied in any organism.


Subject(s)
RNA, Messenger/chemistry , RNA-Binding Proteins/isolation & purification , RNA/isolation & purification , Cell Line, Tumor , Cluster Analysis , Cross-Linking Reagents/chemistry , Escherichia coli , Glycoproteins/chemistry , HEK293 Cells , Humans , Nocodazole/chemistry , Protein Binding , Proteome , Proteomics , RNA/chemistry , RNA, Bacterial/chemistry , RNA, Long Noncoding/chemistry , RNA-Binding Proteins/chemistry , Sequence Analysis, RNA , Thymidine/chemistry , Transcriptome
14.
Nat Rev Mol Cell Biol ; 19(10): 638-653, 2018 10.
Article in English | MEDLINE | ID: mdl-30104700

ABSTRACT

G protein-coupled receptors (GPCRs) are the largest group of cell surface receptors in humans that signal in response to diverse inputs and regulate a plethora of cellular processes. Hence, they constitute one of the primary drug target classes. Progress in our understanding of GPCR dynamics, activation and signalling has opened new possibilities for selective drug development. A key advancement has been provided by the concept of biased agonism, which describes the ability of ligands acting at the same GPCR to elicit distinct cellular signalling profiles by preferentially stabilizing different active conformational states of the receptor. Application of this concept raises the prospect of 'designer' biased agonists as optimized therapeutics with improved efficacy and/or reduced side-effect profiles. However, this application will require a detailed understanding of the spectrum of drug actions and a structural understanding of the drug-receptor interactions that drive distinct pharmacologies. The recent revolution in GPCR structural biology provides unprecedented insights into ligand binding, conformational dynamics and the control of signalling outcomes. These insights, together with new approaches to multi-dimensional analysis of drug action, are allowing refined classification of drugs according to their pharmacodynamic profiles, which can be linked to receptor structure and predictions of preclinical drug efficacy.


Subject(s)
Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/metabolism , Receptors, G-Protein-Coupled/physiology , Animals , Humans , Ligands , Signal Transduction/drug effects , Signal Transduction/physiology
15.
Cell Signal ; 42: 114-126, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29056499

ABSTRACT

Muscarinic receptors (M-Rs) for acetylcholine (ACh) belong to the class A of G protein-coupled receptors. M-Rs are activated by orthosteric agonists that bind to a specific site buried in the M-R transmembrane helix bundle. In the active conformation, receptor function can be modulated either by allosteric modulators, which bind to the extracellular receptor surface or by the membrane potential via an unknown mechanism. Here, we compared the modulation of M1-Rs and M3-Rs induced by changes in voltage to their allosteric modulation by chemical compounds. We quantified changes in receptor signaling in single HEK 293 cells with a FRET biosensor for the Gq protein cycle. In the presence of ACh, M1-R signaling was potentiated by voltage, similarly to positive allosteric modulation by benzyl quinolone carboxylic acid. Conversely, signaling of M3-R was attenuated by voltage or the negative allosteric modulator gallamine. Because the orthosteric site is highly conserved among M-Rs, but allosteric sites vary, we constructed "allosteric site" M3/M1-R chimeras and analyzed their voltage dependencies. Exchanging the entire allosteric sites eliminated the voltage sensitivity of ACh responses for both receptors, but did not affect their modulation by allosteric compounds. Furthermore, a point mutation in M3-Rs caused functional uncoupling of the allosteric and orthosteric sites and abolished voltage dependence. Molecular dynamics simulations of the receptor variants indicated a subtype-specific crosstalk between both sites, involving the conserved tyrosine lid structure of the orthosteric site. This molecular crosstalk leads to receptor subtype-specific voltage effects.


Subject(s)
Acetylcholine/chemistry , GTP-Binding Protein alpha Subunits, Gq-G11/chemistry , Gallamine Triethiodide/chemistry , Hydroxyquinolines/chemistry , Receptors, Muscarinic/chemistry , Recombinant Fusion Proteins/chemistry , Acetylcholine/pharmacology , Allosteric Site , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Binding Sites , Biosensing Techniques , Chimera/genetics , Chimera/metabolism , Fluorescence Resonance Energy Transfer , GTP-Binding Protein alpha Subunits, Gq-G11/genetics , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Gallamine Triethiodide/pharmacology , Gene Expression , HEK293 Cells , Humans , Hydroxyquinolines/pharmacology , Kinetics , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Membrane Potentials/drug effects , Molecular Dynamics Simulation , Patch-Clamp Techniques , Protein Binding , Protein Interaction Domains and Motifs , Protein Structure, Secondary , Receptors, Muscarinic/genetics , Receptors, Muscarinic/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
16.
Eur J Pharmacol ; 815: 138-146, 2017 Nov 15.
Article in English | MEDLINE | ID: mdl-28899696

ABSTRACT

The serotonin 2A (5-HT2A) receptor is a G-protein coupled receptor (GPCR) with a conserved disulfide bridge formed by Cys148 (transmembrane helix 3, TM3) and Cys227 (extracellular loop 2, ECL-2). We hypothesized that disulfide bridges may determine serotonin 5-HT2A receptor functions such as receptor activation, functional selectivity and ligand recognition. We used the reducing agent dithiothreitol (DTT) to determine how the reduction of disulfide bridges affects radioligand binding, second messenger mobilization and receptor dimerization. A DTT-induced decrease in the number of binding sites (1190 ± 63.55 fmol/mg protein for control cells compared with 921.2 ± 60.84 fmol/mg protein for DTT-treated cells) as well as in the efficacy of both signalling pathways characterized was observed, although the affinity and potency were unchanged. Bioluminiscence resonance energy transfer (BRET) assays revealed the DTT treatment did not modify the homodimeric nature of serotonin 5-HT2A receptors. In molecular dynamic simulations, the ECL-2 of the receptor with a broken cysteine bond adopts a wider variety of conformations, some of which protrude deeper into the receptor orthosteric binding pocket leading to collapse of the pocket. A shrunken binding pocket would be incapable of accommodating lysergic acid diethylamide (LSD). Our findings suggest that the decrease of efficacy may be due to disruption of disulfide bridge between TM3 and ECL-2. This reveals the integrity of the ECL-2 epitope, which should be explored in the development of novel ligands acting as allosteric modulators of serotonin 5-HT2A receptors.


Subject(s)
Disulfides/chemistry , Protein Multimerization , Receptor, Serotonin, 5-HT2A/chemistry , Receptor, Serotonin, 5-HT2A/metabolism , Signal Transduction , Amino Acid Sequence , Animals , Binding Sites , CHO Cells , Cricetinae , Cricetulus , Dithiothreitol/pharmacology , Humans , Ligands , Models, Molecular , Nuclear Proteins/metabolism , Protein Binding , Protein Structure, Quaternary
17.
Oncotarget ; 8(16): 26732-26743, 2017 Apr 18.
Article in English | MEDLINE | ID: mdl-28423643

ABSTRACT

Germline mutations in POLE and POLD1 have been shown to cause predisposition to colorectal multiple polyposis and a wide range of neoplasms, early-onset colorectal cancer being the most prevalent. In order to find additional mutations affecting the proofreading activity of these polymerases, we sequenced its exonuclease domain in 155 patients with multiple polyps or an early-onset colorectal cancer phenotype without alterations in the known hereditary colorectal cancer genes. Interestingly, none of the previously reported mutations in POLE and POLD1 were found. On the other hand, among the genetic variants detected, only two of them stood out as putative pathogenic in the POLE gene, c.1359 + 46del71 and c.1420G > A (p.Val474Ile). The first variant, detected in two families, was not proven to alter correct RNA splicing. Contrarily, c.1420G > A (p.Val474Ile) was detected in one early-onset colorectal cancer patient and located right next to the exonuclease domain. The pathogenicity of this change was suggested by its rarity and bioinformatics predictions, and it was further indicated by functional assays in Schizosaccharomyces pombe. This is the first study to functionally analyze a POLE genetic variant outside the exonuclease domain and widens the spectrum of genetic changes in this DNA polymerase that could lead to colorectal cancer predisposition.


Subject(s)
Adenomatous Polyposis Coli/diagnosis , Adenomatous Polyposis Coli/genetics , Colorectal Neoplasms/diagnosis , Colorectal Neoplasms/genetics , DNA Polymerase III/genetics , DNA Polymerase II/genetics , Poly-ADP-Ribose Binding Proteins/genetics , Adolescent , Adult , Age of Onset , Aged , Aged, 80 and over , Alleles , Amino Acid Substitution , Child , Child, Preschool , Colorectal Neoplasms/prevention & control , DNA Polymerase II/chemistry , DNA Polymerase III/chemistry , Female , Genetic Association Studies , Genetic Predisposition to Disease , Genetic Testing , Humans , Infant , Infant, Newborn , Male , Middle Aged , Models, Molecular , Mutation , Pedigree , Poly-ADP-Ribose Binding Proteins/chemistry , Protein Conformation , Protein Domains/genetics , Young Adult
18.
Nat Commun ; 8: 14505, 2017 02 21.
Article in English | MEDLINE | ID: mdl-28220900

ABSTRACT

Cholesterol is a key component of cell membranes with a proven modulatory role on the function and ligand-binding properties of G-protein-coupled receptors (GPCRs). Crystal structures of prototypical GPCRs such as the adenosine A2A receptor (A2AR) have confirmed that cholesterol finds stable binding sites at the receptor surface suggesting an allosteric role of this lipid. Here we combine experimental and computational approaches to show that cholesterol can spontaneously enter the A2AR-binding pocket from the membrane milieu using the same portal gate previously suggested for opsin ligands. We confirm the presence of cholesterol inside the receptor by chemical modification of the A2AR interior in a biotinylation assay. Overall, we show that cholesterol's impact on A2AR-binding affinity goes beyond pure allosteric modulation and unveils a new interaction mode between cholesterol and the A2AR that could potentially apply to other GPCRs.


Subject(s)
Cell Membrane/chemistry , Cholesterol/chemistry , Protein Domains , Receptors, G-Protein-Coupled/chemistry , Animals , Binding Sites , Binding, Competitive , Cell Line, Tumor , Cell Membrane/metabolism , Cholesterol/metabolism , Molecular Dynamics Simulation , Protein Binding , Rats , Receptor, Adenosine A2A/chemistry , Receptor, Adenosine A2A/metabolism , Receptors, G-Protein-Coupled/metabolism
19.
Sci Rep ; 6: 27546, 2016 06 09.
Article in English | MEDLINE | ID: mdl-27278133

ABSTRACT

Codon usage adaptation of lytic viruses to their hosts is determinant for viral fitness. In this work, we analyzed the codon usage of adenoviral proteins by principal component analysis and assessed their codon adaptation to the host. We observed a general clustering of adenoviral proteins according to their function. However, there was a significant variation in the codon preference between the host-interacting fiber protein and the rest of structural late phase proteins, with a non-optimal codon usage of the fiber. To understand the impact of codon bias in the fiber, we optimized the Adenovirus-5 fiber to the codon usage of the hexon structural protein. The optimized fiber displayed increased expression in a non-viral context. However, infection with adenoviruses containing the optimized fiber resulted in decreased expression of the fiber and of wild-type structural proteins. Consequently, this led to a drastic reduction in viral release. The insertion of an exogenous optimized protein as a late gene in the adenovirus with the optimized fiber further interfered with viral fitness. These results highlight the importance of balancing codon usage in viral proteins to adequately exploit cellular resources for efficient infection and open new opportunities to regulate viral fitness for virotherapy and vaccine development.


Subject(s)
Adenoviridae/genetics , Codon , Viral Proteins/genetics , A549 Cells , Capsid Proteins/genetics , Gene Expression Regulation, Viral , Genetic Engineering , Genome, Viral , Green Fluorescent Proteins/metabolism , HEK293 Cells , HeLa Cells , Humans , Neoplasms/therapy , Oncolytic Virotherapy , Plasmids/genetics , Principal Component Analysis , Protein Folding
20.
Sci Rep ; 6: 23754, 2016 Mar 31.
Article in English | MEDLINE | ID: mdl-27029337

ABSTRACT

Adaptation is defined as an evolutionary process allowing organisms to succeed in certain habitats or conditions. Chromosomal inversions have the potential to be key in the adaptation processes, since they can contribute to the maintenance of favoured combinations of adaptive alleles through reduced recombination between individuals carrying different inversions. We have analysed six genes (Pif1A, Abi, Sqd, Yrt, Atpα and Fmr1), located inside and outside three inversions of the O chromosome in European populations of Drosophila subobscura. Genetic differentiation was significant between inversions despite extensive recombination inside inverted regions, irrespective of gene distance to the inversion breakpoints. Surprisingly, the highest level of genetic differentiation between arrangements was found for the Atpα gene, which is located outside the O1 and O7 inversions. Two derived unrelated arrangements (O3+4+1 and O3+4+7) are nearly fixed for several amino acid substitutions at the Atpα gene that have been described to confer resistance in other species to the cardenolide ouabain, a plant toxin capable of blocking ATPases. Similarities in the Atpα variants, conferring ouabain resistance in both arrangements, may be the result of convergent substitution and be favoured in response to selective pressures presumably related to the presence of plants containing ouabain in the geographic locations where both inversions are present.


Subject(s)
Adaptation, Physiological/genetics , Chromosomes, Insect/drug effects , Drosophila/drug effects , Ouabain/toxicity , Toxins, Biological/toxicity , Amino Acid Sequence , Amino Acid Substitution , Animals , Carrier Proteins/genetics , Carrier Proteins/metabolism , Chromosome Inversion/drug effects , Chromosomes, Insect/chemistry , Drosophila/genetics , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Fragile X Mental Retardation Protein/genetics , Fragile X Mental Retardation Protein/metabolism , Models, Molecular , Ouabain/metabolism , Plants/chemistry , Plants/metabolism , Polymorphism, Genetic , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Sodium-Potassium-Exchanging ATPase/genetics , Sodium-Potassium-Exchanging ATPase/metabolism , Toxins, Biological/biosynthesis , Toxins, Biological/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...