Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 86
Filter
1.
Proc Natl Acad Sci U S A ; 119(24): e2201103119, 2022 06 14.
Article in English | MEDLINE | ID: mdl-35671422

ABSTRACT

The quaternary organization of rhodopsin-like G protein-coupled receptors in native tissues is unknown. To address this we generated mice in which the M1 muscarinic acetylcholine receptor was replaced with a C-terminally monomeric enhanced green fluorescent protein (mEGFP)-linked variant. Fluorescence imaging of brain slices demonstrated appropriate regional distribution, and using both anti-M1 and anti-green fluorescent protein antisera the expressed transgene was detected in both cortex and hippocampus only as the full-length polypeptide. M1-mEGFP was expressed at levels equal to the M1 receptor in wild-type mice and was expressed throughout cell bodies and projections in cultured neurons from these animals. Signaling and behavioral studies demonstrated M1-mEGFP was fully active. Application of fluorescence intensity fluctuation spectrometry to regions of interest within M1-mEGFP-expressing neurons quantified local levels of expression and showed the receptor was present as a mixture of monomers, dimers, and higher-order oligomeric complexes. Treatment with both an agonist and an antagonist ligand promoted monomerization of the M1-mEGFP receptor. The quaternary organization of a class A G protein-coupled receptor in situ was directly quantified in neurons in this study, which answers the much-debated question of the extent and potential ligand-induced regulation of basal quaternary organization of such a receptor in native tissue when present at endogenous expression levels.


Subject(s)
Cerebral Cortex , Hippocampus , Receptor, Muscarinic M1 , Animals , Cerebral Cortex/metabolism , Green Fluorescent Proteins , Hippocampus/metabolism , Ligands , Mice , Mice, Knockout , Neurons/metabolism , Optical Imaging , Receptor, Muscarinic M1/chemistry , Receptor, Muscarinic M1/genetics , Receptor, Muscarinic M1/metabolism
2.
J Biol Chem ; 298(5): 101932, 2022 05.
Article in English | MEDLINE | ID: mdl-35427647

ABSTRACT

GPR84 is an immune cell-expressed, proinflammatory receptor currently being assessed as a therapeutic target in conditions including fibrosis and inflammatory bowel disease. Although it was previously shown that the orthosteric GPR84 activators 2-HTP and 6-OAU promoted its interactions with arrestin-3, a G protein-biased agonist DL-175 did not. Here, we show that replacement of all 21 serine and threonine residues within i-loop 3 of GPR84, but not the two serines in the C-terminal tail, eliminated the incorporation of [32P] and greatly reduced receptor-arrestin-3 interactions promoted by 2-HTP. GPR84 was phosphorylated constitutively on residues Ser221 and Ser224, while various other amino acids are phosphorylated in response to 2-HTP. Consistent with this, an antiserum able to identify pSer221/pSer224 recognized GPR84 from cells treated with and without activators, whereas an antiserum able to identify pThr263/pThr264 only recognized GPR84 after exposure to 2-HTP and not DL-175. Two distinct GPR84 antagonists as well as inhibition of G protein-coupled receptor kinase 2/3 prevented phosphorylation of pThr263/pThr264, but neither strategy affected constitutive phosphorylation of Ser221/Ser224. Furthermore, mutation of residues Thr263 and Thr264 to alanine generated a variant of GPR84 also limited in 2-HTP-induced interactions with arrestin-2 and -3. By contrast, this mutant was unaffected in its capacity to reduce cAMP levels. Taken together, these results define a key pair of threonine residues, regulated only by subsets of GPR84 small molecule activators and by GRK2/3 that define effective interactions with arrestins and provide novel tools to monitor the phosphorylation and functional status of GPR84.


Subject(s)
Arrestins , Threonine , Arrestins/metabolism , Humans , Ligands , Mutation , Phosphorylation , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Serine/metabolism , Threonine/metabolism , beta-Arrestin 2/metabolism
3.
J Biol Chem ; 296: 100139, 2021.
Article in English | MEDLINE | ID: mdl-33268380

ABSTRACT

CXCR4, a member of the family of chemokine-activated G protein-coupled receptors, is widely expressed in immune response cells. It is involved in both cancer development and progression as well as viral infection, notably by HIV-1. A variety of methods, including structural information, have suggested that the receptor may exist as a dimer or an oligomer. However, the mechanistic details surrounding receptor oligomerization and its potential dynamic regulation remain unclear. Using both biochemical and biophysical means, we confirm that CXCR4 can exist as a mixture of monomers, dimers, and higher-order oligomers in cell membranes and show that oligomeric structure becomes more complex as receptor expression levels increase. Mutations of CXCR4 residues located at a putative dimerization interface result in monomerization of the receptor. Additionally, binding of the CXCR4 antagonist IT1t-a small drug-like isothiourea derivative-rapidly destabilizes the oligomeric structure, whereas AMD3100, another well-characterized CXCR4 antagonist, does not. Although a mutation that regulates constitutive activity of CXCR4 also results in monomerization of the receptor, binding of IT1t to this variant promotes receptor dimerization. These results provide novel insights into the basal organization of CXCR4 and how antagonist ligands of different chemotypes differentially regulate its oligomerization state.


Subject(s)
Benzylamines/pharmacology , Cyclams/pharmacology , Receptors, CXCR4/antagonists & inhibitors , Receptors, CXCR4/metabolism , Small Molecule Libraries/pharmacology , Thiourea/pharmacology , Anti-HIV Agents/pharmacology , Cells, Cultured , Green Fluorescent Proteins/metabolism , Heterocyclic Compounds/chemistry , Heterocyclic Compounds/pharmacology , Humans , Ligands , Protein Binding , Protein Conformation/drug effects , Protein Multimerization/drug effects , Receptors, CXCR4/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/isolation & purification , Recombinant Fusion Proteins/metabolism , Signal Transduction
4.
Nat Methods ; 16(6): 493-496, 2019 06.
Article in English | MEDLINE | ID: mdl-31110281

ABSTRACT

Here, we introduce fluorescence intensity fluctuation spectrometry for determining the identity, abundance and stability of protein oligomers. This approach was tested on monomers and oligomers of known sizes and was used to uncover the oligomeric states of the epidermal growth factor receptor and the secretin receptor in the presence and absence of their agonist ligands. This method is fast and is scalable for high-throughput screening of drugs targeting protein-protein interactions.


Subject(s)
Fluorescence , Image Processing, Computer-Assisted/methods , Protein Multimerization , Receptors, G-Protein-Coupled/metabolism , Receptors, Gastrointestinal Hormone/metabolism , ErbB Receptors/chemistry , ErbB Receptors/metabolism , Humans , Ligands , Microscopy, Confocal , Protein Binding , Protein Interaction Domains and Motifs , Signal Transduction , Spectrometry, Fluorescence
5.
Mol Pharmacol ; 95(2): 196-209, 2019 02.
Article in English | MEDLINE | ID: mdl-30514721

ABSTRACT

Chemoproteomic approaches to identify ligand-receptor interactions have gained popularity. However, identifying transmembrane receptors remains challenging. A new trifunctional probe to aid the nonbiased identification of such receptors was developed and synthesized using a convenient seven-step synthesis. This probe contained three functional groups: 1) an N-hydroxysuccinimide ester for ligand-coupling through free amines, 2) a diazirine moiety to capture the receptor of interest upon irradiation with UV light, and 3) a biotin group which allowed affinity purification of the final adduct using streptavidin. The interaction between the G protein-coupled tachykinin neurokinin 1 (NK1) receptor, expressed in an inducible manner, and the peptidic ligand substance P was used as a test system. Liquid chromatography-mass spectrometry analysis confirmed successful coupling of the probe to substance P, while inositol monophosphate accumulation assays demonstrated that coupling of the probe did not interfere substantially with the substance P-NK1 receptor interaction. Confocal microscopy and western blotting provided evidence of the formation of a covalent bond between the probe and the NK1 receptor upon UV activation. As proof of concept, the probe was used in full ligand-based receptor-capture experiments to identify the substance P-binding receptor via liquid chromatography-tandem mass spectrometry, resulting in the successful identification of only the NK1 receptor. This provides proof of concept toward general utilization of this probe to define interactions between ligands and previously unidentified plasma-membrane receptors.


Subject(s)
Diazomethane/metabolism , Receptors, G-Protein-Coupled/metabolism , Cell Membrane/metabolism , Chromatography, Liquid/methods , HEK293 Cells , Humans , Ligands , Receptors, Neurokinin-1/metabolism , Substance P/metabolism , Tandem Mass Spectrometry/methods
6.
Ann Bot ; 124(4): 553-566, 2019 10 29.
Article in English | MEDLINE | ID: mdl-30137291

ABSTRACT

BACKGROUND AND AIMS: The cultivation of dedicated biomass crops, including miscanthus, on marginal land provides a promising approach to the reduction of dependency on fossil fuels. However, little is known about the impact of environmental stresses often experienced on lower-grade agricultural land on cell-wall quality traits in miscanthus biomass crops. In this study, three different miscanthus genotypes were exposed to drought stress and nutrient stress, both separately and in combination, with the aim of evaluating their impact on plant growth and cell-wall properties. METHODS: Automated imaging facilities at the National Plant Phenomics Centre (NPPC-Aberystwyth) were used for dynamic phenotyping to identify plant responses to separate and combinatorial stresses. Harvested leaf and stem samples of the three miscanthus genotypes (Miscanthus sinensis, Miscanthus sacchariflorus and Miscanthus × giganteus) were separately subjected to saccharification assays, to measure sugar release, and cell-wall composition analyses. KEY RESULTS: Phenotyping showed that the M. sacchariflorus genotype Sac-5 and particularly the M. sinensis genotype Sin-11 coped better than the M. × giganteus genotype Gig-311 with drought stress when grown in nutrient-poor compost. Sugar release by enzymatic hydrolysis, used as a biomass quality measure, was significantly affected by the different environmental conditions in a stress-, genotype- and organ-dependent manner. A combination of abundant water and low nutrients resulted in the highest sugar release from leaves, while for stems this was generally associated with the combination of drought and nutrient-rich conditions. Cell-wall composition analyses suggest that changes in fine structure of cell-wall polysaccharides, including heteroxylans and pectins, possibly in association with lignin, contribute to the observed differences in cell-wall biomass sugar release. CONCLUSIONS: The results highlight the importance of the assessment of miscanthus biomass quality measures in addition to biomass yield determinations and the requirement for selecting suitable miscanthus genotypes for different environmental conditions.


Subject(s)
Droughts , Poaceae , Biomass , Lignin , Nutrients
7.
Anal Biochem ; 540-541: 45-51, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29154787

ABSTRACT

A paramagnetic nanocomposite coated with chitosan and N-(5-Amino-1-carboxy-pentyl) iminodiacetic acid (NTA) that is suitable for protein immobilization applications has been prepared and characterized. The nanoparticle core was synthesized by controlled aggregation of Fe3O4 under alkaline conditions, and Transmission Electron Microscopy revealed a size distribution of 10-50 nm. The nanoparticle core was coated with chitosan and derivatized with glutaraldehyde and NTA, as confirmed by Fourier Transform Infrared Spectroscopy. The final nanoparticles were used as a metal affinity matrix to separate a recombinant polyhistidine-tagged ß-galactosidase from Bacillus subtilis directly from E. coli cell lysates with high purity (>95%). After loading with Ni2+, nanoparticles demonstrated a binding capacity of 250 µg of a polyhistidine-tagged ß-galactosidase per milligram of support. The immobilized enzyme retained 80% activity after 9 cycles of washing, and the immobilized recombinant protein could be eluted with high purity with imidazole. The applications for these nanomagnetic composites extend beyond protein purification, and can also be used for immobilizing enzymes, where the ß-galactosidase immobilized on the nanomagnetic support was used in multiple cycles of catalytic reactions with no significant loss of catalytic activity.


Subject(s)
Chromatography, Affinity , Magnetite Nanoparticles/chemistry , Recombinant Proteins/isolation & purification , beta-Galactosidase/metabolism , Bacillus subtilis/enzymology , Chitosan/chemistry , Enzymes, Immobilized/chemistry , Enzymes, Immobilized/metabolism , Ferrosoferric Oxide/chemistry , Histidine/chemistry , Histidine/genetics , Histidine/metabolism , Imino Acids/chemistry , Microscopy, Electron, Transmission , Oligopeptides/chemistry , Oligopeptides/genetics , Oligopeptides/metabolism , Particle Size , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Spectroscopy, Fourier Transform Infrared , beta-Galactosidase/chemistry , beta-Galactosidase/genetics
8.
Biochem J ; 474(11): 1879-1895, 2017 05 24.
Article in English | MEDLINE | ID: mdl-28424368

ABSTRACT

Previous studies have indicated that the G-protein-coupled secretin receptor is present as a homodimer, organized through symmetrical contacts in transmembrane domain IV, and that receptor dimerization is critical for high-potency signalling by secretin. However, whether all of the receptor exists in the dimeric form or if this is regulated is unclear. We used measures of quantal brightness of the secretin receptor tagged with monomeric enhanced green fluorescent protein (mEGFP) and spatial intensity distribution analysis to assess this. Calibration using cells expressing plasma membrane-anchored forms of mEGFP initially allowed us to demonstrate that the epidermal growth factor receptor is predominantly monomeric in the absence of ligand and while wild-type receptor was rapidly converted into a dimeric form by ligand, a mutated form of this receptor remained monomeric. Equivalent studies showed that, at moderate expression levels, the secretin receptor exists as a mixture of monomeric and dimeric forms, with little evidence of higher-order complexity. However, sodium butyrate-induced up-regulation of the receptor resulted in a shift from monomeric towards oligomeric organization. In contrast, a form of the secretin receptor containing a pair of mutations on the lipid-facing side of transmembrane domain IV was almost entirely monomeric. Down-regulation of the secretin receptor-interacting G-protein Gαs did not alter receptor organization, indicating that dimerization is defined specifically by direct protein-protein interactions between copies of the receptor polypeptide, while short-term treatment with secretin had no effect on organization of the wild-type receptor but increased the dimeric proportion of the mutated receptor variant.


Subject(s)
Receptors, G-Protein-Coupled/chemistry , Receptors, Gastrointestinal Hormone/chemistry , Animals , CHO Cells , Cricetinae , Cricetulus , Green Fluorescent Proteins/genetics , Protein Multimerization , Receptors, G-Protein-Coupled/genetics , Receptors, Gastrointestinal Hormone/genetics
9.
J Biol Chem ; 291(25): 13132-46, 2016 Jun 17.
Article in English | MEDLINE | ID: mdl-27080256

ABSTRACT

Although rhodopsin-like G protein-coupled receptors can exist as both monomers and non-covalently associated dimers/oligomers, the steady-state proportion of each form and whether this is regulated by receptor ligands are unknown. Herein we address these topics for the M1 muscarinic acetylcholine receptor, a key molecular target for novel cognition enhancers, by using spatial intensity distribution analysis. This method can measure fluorescent particle concentration and assess oligomerization states of proteins within defined regions of living cells. Imaging and analysis of the basolateral surface of cells expressing some 50 molecules·µm(-2) human muscarinic M1 receptor identified a ∼75:25 mixture of receptor monomers and dimers/oligomers. Both sustained and shorter term treatment with the selective M1 antagonist pirenzepine resulted in a large shift in the distribution of receptor species to favor the dimeric/oligomeric state. Although sustained treatment with pirenzepine also resulted in marked up-regulation of the receptor, simple mass action effects were not the basis for ligand-induced stabilization of receptor dimers/oligomers. The related antagonist telenzepine also produced stabilization and enrichment of the M1 receptor dimer population, but the receptor subtype non-selective antagonists atropine and N-methylscopolamine did not. In contrast, neither pirenzepine nor telenzepine altered the quaternary organization of the related M3 muscarinic receptor. These data provide unique insights into the selective capacity of receptor ligands to promote and/or stabilize receptor dimers/oligomers and demonstrate that the dynamics of ligand regulation of the quaternary organization of G protein-coupled receptors is markedly more complex than previously appreciated. This may have major implications for receptor function and behavior.


Subject(s)
Atropine/pharmacology , Muscarinic Antagonists/pharmacology , Pirenzepine/analogs & derivatives , Pirenzepine/pharmacology , Protein Multimerization/drug effects , Receptor, Muscarinic M1/antagonists & inhibitors , Cell Line , Humans , Receptor, Muscarinic M1/chemistry , Receptor, Muscarinic M1/metabolism
10.
J Biol Chem ; 290(20): 12844-57, 2015 May 15.
Article in English | MEDLINE | ID: mdl-25825490

ABSTRACT

The questions of whether G protein-coupled receptors exist as monomers, dimers, and/or oligomers and if these species interconvert in a ligand-dependent manner are among the most contentious current issues in biology. When employing spatial intensity distribution analysis to laser scanning confocal microscope images of cells stably expressing either a plasma membrane-associated form of monomeric enhanced green fluorescent protein (eGFP) or a tandem version of this fluorophore, the eGFP tandem was identified as a dimer. Similar studies on cells stably expressing an eGFP-tagged form of the epidermal growth factor receptor demonstrated that, although largely a monomer in the basal state, this receptor rapidly became predominantly dimeric upon the addition of its ligand epidermal growth factor. In cells induced to express an eGFP-tagged form of the serotonin 5-hydroxytryptamine 2C (5-HT2C) receptor, global analysis of construct quantal brightness was consistent with the predominant form of the receptor being dimeric. However, detailed spatial intensity distribution analysis demonstrated the presence of multiple forms ranging from monomers to higher-order oligomers. Furthermore, treatment with chemically distinct 5-HT2C receptor antagonists resulted in a time-dependent change in the quaternary organization to one in which there was a preponderance of receptor monomers. This antagonist-mediated effect was reversible, because washout of the ligand resulted in the regeneration of many of the oligomeric forms of the receptor.


Subject(s)
Multiprotein Complexes/metabolism , Protein Multimerization/physiology , Receptor, Serotonin, 5-HT2C/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HEK293 Cells , Humans , Multiprotein Complexes/genetics , Protein Multimerization/drug effects , Protein Structure, Quaternary , Receptor, Serotonin, 5-HT2C/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Serotonin 5-HT2 Receptor Antagonists/pharmacology
11.
Mol Pharmacol ; 87(6): 936-53, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25769304

ABSTRACT

G protein-coupled receptors, including the M3 muscarinic acetylcholine receptor, can form homo-oligomers. However, the basis of these interactions and the overall organizational structure of such oligomers are poorly understood. Combinations of site-directed mutagenesis and homogenous time-resolved fluorescence resonance energy transfer studies that assessed interactions between receptor protomers at the surface of transfected cells indicated important contributions of regions of transmembrane domains I, IV, V, VI, and VII as well as intracellular helix VIII to the overall organization. Molecular modeling studies based on both these results and an X-ray structure of the inactive state of the M3 receptor bound by the antagonist/inverse agonist tiotropium were then employed. The results could be accommodated fully by models in which a proportion of the cell surface M3 receptor population is a tetramer with rhombic, but not linear, orientation. This is consistent with previous studies based on spectrally resolved, multiphoton fluorescence resonance energy transfer. Modeling studies furthermore suggest an important role for molecules of cholesterol at the dimer + dimer interface of the tetramer, which is consistent with the presence of cholesterol at key locations in many G protein-coupled receptor crystal structures. Mutants that displayed disrupted quaternary organization were often poorly expressed and showed immature N-glycosylation. Sustained treatment of cells expressing such mutants with the muscarinic receptor inverse agonist atropine increased cellular levels and restored both cell surface delivery and quaternary organization to many of the mutants. These observations suggest that organization as a tetramer may occur before plasma membrane delivery and may be a key step in cellular quality control assessment.


Subject(s)
Receptor, Muscarinic M3/metabolism , Atropine/pharmacology , Cell Membrane/metabolism , Cholesterol/chemistry , Drug Inverse Agonism , Fluorescence Resonance Energy Transfer , Glycosylation , HEK293 Cells , Humans , Models, Molecular , Muscarinic Agonists/chemistry , Muscarinic Agonists/pharmacology , Muscarinic Antagonists/chemistry , Mutagenesis, Site-Directed , Promoter Regions, Genetic , Protein Multimerization , Protein Transport , Radioligand Assay , Receptor, Muscarinic M3/chemistry , Receptor, Muscarinic M3/genetics , Scopolamine Derivatives/chemistry , Tiotropium Bromide
12.
Biochim Biophys Acta ; 1838(1 Pt A): 3-14, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23590995

ABSTRACT

The interaction between ligands and the G protein-coupled receptors (GPCRs) to which they bind has long been the focus of intensive investigation. The signalling cascades triggered by receptor activation, due in most cases to ligand binding, are of great physiological and medical importance; indeed, GPCRs are targeted by in excess of 30% of small molecule therapeutic medicines. Attempts to identify further pharmacologically useful GPCR ligands, for receptors with known and unknown endogenous ligands, continue apace. In earlier days direct assessment of such interactions was restricted largely to the use of ligands incorporating radioactive isotope labels as this allowed detection of the ligand and monitoring its interaction with the GPCR. This use of such markers has continued with the development of ligands labelled with fluorophores and their application to the study of receptor-ligand interactions using both light microscopy and resonance energy transfer techniques, including homogenous time-resolved fluorescence resonance energy transfer. Details of ligand-receptor interactions via X-ray crystallography are advancing rapidly as methods suitable for routine production of substantial amounts and stabilised forms of GPCRs have been developed and there is hope that this may become as routine as the co-crystallisation of serine/threonine kinases with ligands, an approach that has facilitated widespread use of rapid structure-based ligand design. Conformational changes involved in the activation of GPCRs, widely predicted by biochemical and biophysical means, have inspired the development of intramolecular FRET-based sensor forms of GPCRs designed to investigate the events following ligand binding and resulting in a signal propagation across the cell membrane. Finally, a number of techniques are emerging in which ligand-GPCR binding can be studied in ways that, whilst indirect, are able to monitor its results in an unbiased and integrated manner. This article is part of a Special Issue entitled: Structural and biophysical characterisation of membrane protein-ligand binding.


Subject(s)
Biophysics , Fluorescence Resonance Energy Transfer , Fluorescent Dyes/chemistry , Receptors, G-Protein-Coupled/metabolism , Crystallography, X-Ray , Ligands , Protein Binding , Protein Conformation , Receptors, G-Protein-Coupled/chemistry
13.
Appl Microbiol Biotechnol ; 99(12): 5095-107, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25605422

ABSTRACT

Xyloglucan-specific endo-ß-1,4-glucanases (Xegs, EC 3.2.1.151) exhibit high catalytic specificity for ß-1,4 linkages of xyloglucan, a branched hemicellulosic polysaccharide abundant in dicot primary cell walls and present in many monocot species. In nature, GH12 Xegs are not associated with carbohydrate-binding modules (CBMs), and here, we have investigated the effect of the fusion of the xyloglucan-specific CBM44 on the structure and function of a GH12 Xeg from Aspergillus niveus (XegA). This fusion presented enhanced catalytic properties and conferred superior thermal stability on the XegA. An increased k cat (chimera, 177.03 s(-1); XegA, 144.31 s(-1)) and reduced KM (chimera, 1.30 mg mL(-1); XegA, 1.50 mg mL(-1)) resulted in a 1.3-fold increase in catalytic efficiency of the chimera over the parental XegA. Although both parental and chimeric enzymes presented catalytic optima at pH 5.5 and 60 °C, the thermostabilitiy of the chimera at 60 °C was greater than the parental XegA. Moreover, the crystallographic structure of XegA together with small-angle X-ray scattering (SAXS) and molecular dynamics simulations revealed that the spatial arrangement of the domains in the chimeric enzyme resulted in the formation of an extended binding cleft that may explain the improved kinetic properties of the CBM44-XegA chimera.


Subject(s)
Aspergillus/enzymology , Endo-1,3(4)-beta-Glucanase/chemistry , Endo-1,3(4)-beta-Glucanase/metabolism , Fungal Proteins/chemistry , Fungal Proteins/metabolism , Glucans/metabolism , Xylans/metabolism , Amino Acid Sequence , Aspergillus/chemistry , Aspergillus/genetics , Endo-1,3(4)-beta-Glucanase/genetics , Fungal Proteins/genetics , Glucans/chemistry , Kinetics , Molecular Dynamics Simulation , Molecular Sequence Data , Protein Engineering , Protein Structure, Tertiary , Scattering, Small Angle , Substrate Specificity , X-Ray Diffraction , Xylans/chemistry
14.
Biochem J ; 461(1): 61-73, 2014 Jul 01.
Article in English | MEDLINE | ID: mdl-24673457

ABSTRACT

Robo (Roundabout) receptors and their Slit polypeptide ligands are known to play key roles in neuronal development and have been implicated in both angiogenesis and cancer. Like the other family members, Robo1 is a large single transmembrane domain polypeptide containing a series of well-defined extracellular elements. However, the intracellular domain lacks structural definition and little is known about the quaternary structure of Robo receptors or how binding of a Slit might affect this. To address these questions combinations of both autofluorescent protein-based FRET imaging and time-resolved FRET were employed. Both approaches identified oligomeric organization of Robo1 that did not require the presence of the intracellular domain. SpIDA (spatial intensity distribution analysis) of eGFP-tagged forms of Robo1 indicated that for a C-terminally deleted version approximately two-thirds of the receptor was present as a dimer and one-third as a monomer. By contrast, full-length Robo1 was present almost exclusively as a dimer. In each case this was unaffected by the addition of Slit2, although parallel studies demonstrated the biological activity of Slit2 and its interaction with Robo1. Deletion of both the immunoglobulin and fibronectin type III extracellular repeats prevented dimer formation, with the immunoglobulin repeats providing the bulk of the protein-protein interaction affinity.


Subject(s)
Intercellular Signaling Peptides and Proteins/chemistry , Intercellular Signaling Peptides and Proteins/metabolism , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/metabolism , Protein Multimerization/physiology , Receptors, Immunologic/chemistry , Receptors, Immunologic/metabolism , Cells, Cultured , HEK293 Cells , Humans , Leukocytes, Mononuclear/metabolism , Ligands , Protein Binding/physiology , Roundabout Proteins
15.
J Biol Chem ; 288(24): 17296-312, 2013 Jun 14.
Article in English | MEDLINE | ID: mdl-23589301

ABSTRACT

FFA2 is a G protein-coupled receptor that responds to short chain fatty acids and has generated interest as a therapeutic target for metabolic and inflammatory conditions. However, definition of its functions has been slowed by a dearth of selective ligands that can distinguish it from the closely related FFA3. At present, the only selective ligands described for FFA2 suffer from poor potency, altered signaling due to allosteric modes of action, or a lack of function at non-human orthologs of the receptor. To address the need for novel selective ligands, we synthesized two compounds potentially having FFA2 activity and examined the molecular basis of their function. These compounds were confirmed to be potent and selective orthosteric FFA2 agonists. A combination of ligand structure-activity relationship, pharmacological analysis, homology modeling, species ortholog comparisons, and mutagenesis studies were then employed to define the molecular basis of selectivity and function of these ligands. From this, we identified key residues within both extracellular loop 2 and the transmembrane domain regions of FFA2 critical for ligand function. One of these ligands was active with reasonable potency at rodent orthologs of FFA2 and demonstrated the role of FFA2 in inhibition of lipolysis and glucagon-like peptide-1 secretion in murine-derived 3T3-L1 and STC-1 cell lines, respectively. Together, these findings describe the first potent and selective FFA2 orthosteric agonists and demonstrate key aspects of ligand interaction within the binding site of FFA2 that will be invaluable in future ligand development at this receptor.


Subject(s)
Butyrates/pharmacology , Cyclopropanes/pharmacology , Receptors, Cell Surface/agonists , Thiazoles/pharmacology , Adipocytes/drug effects , Adipocytes/metabolism , Allosteric Regulation , Amino Acid Motifs , Amino Acid Substitution , Animals , Benzeneacetamides/pharmacology , Binding Sites , Cyclopropanes/chemistry , Enteroendocrine Cells/metabolism , Glucagon-Like Peptide 1/metabolism , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , HEK293 Cells , Humans , Lipolysis/drug effects , Mice , Models, Molecular , Mutagenesis, Site-Directed , Protein Binding , Protein Structure, Tertiary , Rats , Receptors, Cell Surface/chemistry , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Thiazoles/chemistry
16.
J Phys Chem B ; 128(18): 4354-4366, 2024 May 09.
Article in English | MEDLINE | ID: mdl-38683784

ABSTRACT

G protein-coupled receptors (GPCRs) are a major gateway to cellular signaling, which respond to ligands binding at extracellular sites through allosteric conformational changes that modulate their interactions with G proteins and arrestins at intracellular sites. High-resolution structures in different ligand states, together with spectroscopic studies and molecular dynamics simulations, have revealed a rich conformational landscape of GPCRs. However, their supramolecular structure and spatiotemporal distribution is also thought to play a significant role in receptor activation and signaling bias within the native cell membrane environment. Here, we applied single-molecule fluorescence techniques, including single-particle tracking, single-molecule photobleaching, and fluorescence correlation spectroscopy, to characterize the diffusion and oligomerization behavior of the muscarinic M1 receptor (M1R) in live cells. Control samples included the monomeric protein CD86 and fixed cells, and experiments performed in the presence of different orthosteric M1R ligands and of several compounds known to change the fluidity and organization of the lipid bilayer. M1 receptors exhibit Brownian diffusion characterized by three diffusion constants: confined/immobile (∼0.01 µm2/s), slow (∼0.04 µm2/s), and fast (∼0.14 µm2/s), whose populations were found to be modulated by both orthosteric ligands and membrane disruptors. The lipid raft disruptor C6 ceramide led to significant changes for CD86, while the diffusion of M1R remained unchanged, indicating that M1 receptors do not partition in lipid rafts. The extent of receptor oligomerization was found to be promoted by increasing the level of expression and the binding of orthosteric ligands; in particular, the agonist carbachol elicited a large increase in the fraction of M1R oligomers. This study provides new insights into the balance between conformational and environmental factors that define the movement and oligomerization states of GPCRs in live cells under close-to-native conditions.


Subject(s)
Receptor, Muscarinic M1 , Ligands , Receptor, Muscarinic M1/metabolism , Receptor, Muscarinic M1/chemistry , Diffusion , Humans , Cell Membrane/metabolism , Cell Membrane/chemistry , Protein Multimerization/drug effects , Animals , Spectrometry, Fluorescence , Molecular Dynamics Simulation , Lipid Bilayers/chemistry , Lipid Bilayers/metabolism
17.
Bioresour Bioprocess ; 11(1): 77, 2024 Jul 29.
Article in English | MEDLINE | ID: mdl-39073555

ABSTRACT

The ß-glucosidase gene from Aspergillus nidulans FGSC A4 was cloned and overexpressed in the A. nidulans A773. The resulting purified ß-glucosidase, named AnGH3, is a monomeric enzyme with a molecular weight of approximately 80 kDa, as confirmed by SDS-PAGE. Circular dichroism further validated its unique canonical barrel fold (ß/α), a feature also observed in the 3D homology model of AnGH3. The most striking aspect of this recombinant enzyme is its robustness, as it retained 100% activity after 24 h of incubation at 45 and 50 ºC and pH 6.0. Even at 55 °C, it maintained 72% of its enzymatic activity after 6 h of incubation at the same pH. The kinetic parameters Vmax, KM, and Kcat/KM for ρ-nitrophenyl-ß-D-glucopyranoside (ρNPG) and cellobiose were also determined. Using ρNPG, the enzyme demonstrated a Vmax of 212 U mg - 1, KM of 0.0607 mmol L - 1, and Kcat/KM of 4521 mmol L - 1 s - 1 when incubated at pH 6.0 and 65 °C. The KM, Vmax, and Kcat/KM using cellobiose were 2.7 mmol L - 1, 57 U mg - 1, and 27 mmol -1 s - 1, respectively. AnGH3 activity was significantly enhanced by xylose and ethanol at concentrations up to 1.5 mol L - 1 and 25%, respectively. Even in challenging conditions, at 65 °C and pH 6.0, the enzyme maintained its activity, retaining 100% and 70% of its initial activity in the presence of 200 mmol L - 1 furfural and 5-hydroxymethylfurfural (HMF), respectively. The potential of this enzyme was further demonstrated by its application in the saccharification of the forage grass Panicum maximum, where it led to a 48% increase in glucose release after 24 h. These unique characteristics, including high catalytic performance, good thermal stability in hydrolysis temperature, and tolerance to elevated concentrations of ethanol, D-xylose, furfural, and HMF, position this recombinant enzyme as a promising tool in the hydrolysis of lignocellulosic biomass as part of an efficient multi-enzyme cocktail, thereby opening new avenues in the field of biotechnology and enzymology.

18.
J Biol Chem ; 287(18): 14937-49, 2012 Apr 27.
Article in English | MEDLINE | ID: mdl-22389503

ABSTRACT

Intramolecular fluorescence resonance energy transfer (FRET) sensors able to detect changes in distance or orientation between the 3rd intracellular loop and C-terminal tail of the human orexin OX(1) and OX(2) G protein-coupled receptors following binding of agonist ligands were produced and expressed stably. These were directed to the plasma membrane and, despite the substantial sequence alterations introduced, in each case were able to elevate [Ca(2+)](i), promote phosphorylation of the ERK1/2 MAP kinases and become internalized effectively upon addition of the native orexin peptides. Detailed characterization of the OX(1) sensor demonstrated that it was activated with rank order of potency orexin A > orexin B > orexin A 16-33, that it bound antagonist ligands with affinity similar to the wild-type receptor, and that mutation of a single residue, D203A, greatly reduced the binding and function of orexin A but not antagonist ligands. Addition of orexin A to individual cells expressing an OX(1) sensor resulted in a time- and concentration-dependent reduction in FRET signal consistent with mass-action and potency/affinity estimates for the peptide. Compared with the response kinetics of a muscarinic M(3) acetylcholine receptor sensor upon addition of agonist, response of the OX(1) and OX(2) sensors to orexin A was slow, consistent with a multistep binding and activation process. Such sensors provide means to assess the kinetics of receptor activation and how this may be altered by mutation and sequence variation of the receptors.


Subject(s)
Intracellular Signaling Peptides and Proteins/pharmacology , MAP Kinase Signaling System/drug effects , Neuropeptides/pharmacology , Peptides/pharmacology , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/metabolism , Receptors, Neuropeptide/agonists , Receptors, Neuropeptide/metabolism , Amino Acid Substitution , Biosensing Techniques/methods , Fluorescence Resonance Energy Transfer , HEK293 Cells , Humans , Kinetics , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Mutation, Missense , Orexin Receptors , Orexins , Phosphorylation/drug effects , Protein Structure, Secondary , Receptor, Muscarinic M3/agonists , Receptor, Muscarinic M3/genetics , Receptor, Muscarinic M3/metabolism , Receptors, G-Protein-Coupled/genetics , Receptors, Neuropeptide/genetics
19.
Biochim Biophys Acta ; 1824(3): 461-7, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22230786

ABSTRACT

Xyloglucan is a major structural polysaccharide of the primary (growing) cell wall of higher plants. It consists of a cellulosic backbone (beta-1,4-linked glucosyl residues) that is frequently substituted with side chains. This report describes Aspergillus nidulans strain A773 recombinant secretion of a dimeric xyloglucan-specific endo-ß-1,4-glucanohydrolase (XegA) cloned from Aspergillus niveus. The ORF of the A. niveus xegA gene is comprised of 714 nucleotides, and encodes a 238 amino acid protein with a calculated molecular weight of 23.5kDa and isoelectric point of 4.38. The optimal pH and temperature were 6.0 and 60°C, respectively. XegA generated a xyloglucan-oligosaccharides (XGOs) pattern similar to that observed for cellulases from family GH12, i.e., demonstrating that its mode of action includes hydrolysis of the glycosidic linkages between glucosyl residues that are not branched with xylose. In contrast to commercial lichenase, mixed linkage beta-glucan (lichenan) was not digested by XegA, indicating that the enzyme did not cleave glucan ß-1,3 or ß-1,6 bonds. The far-UV CD spectrum of the purified enzyme indicated a protein rich in ß-sheet structures as expected for GH12 xyloglucanases. Thermal unfolding studies displayed two transitions with mid-point temperatures of 51.3°C and 81.3°C respectively, and dynamic light scattering studies indicated that the first transition involves a change in oligomeric state from a dimeric to a monomeric form. Since the enzyme is a predominantly a monomer at 60°C, the enzymatic assays demonstrated that XegA is more active in its monomeric state.


Subject(s)
Aspergillus/chemistry , Cell Wall/chemistry , Cellulase/chemistry , Fungal Proteins/chemistry , Glucans/chemistry , Xylans/chemistry , Amino Acid Sequence , Aspergillus/enzymology , Aspergillus nidulans/genetics , Cell Wall/enzymology , Cellulase/genetics , Cellulase/metabolism , Circular Dichroism , Fungal Proteins/genetics , Fungal Proteins/metabolism , Glucans/metabolism , Glycoside Hydrolases/chemistry , Glycoside Hydrolases/metabolism , Hydrogen-Ion Concentration , Isoelectric Point , Kinetics , Light , Molecular Sequence Data , Molecular Weight , Protein Multimerization , Protein Structure, Secondary , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Scattering, Radiation , Substrate Specificity , Temperature , Xylans/metabolism
20.
Biochem J ; 441(1): 95-104, 2012 Jan 01.
Article in English | MEDLINE | ID: mdl-21880019

ABSTRACT

Cellulases participate in a number of biological events, such as plant cell wall remodelling, nematode parasitism and microbial carbon uptake. Their ability to depolymerize crystalline cellulose is of great biotechnological interest for environmentally compatible production of fuels from lignocellulosic biomass. However, industrial use of cellulases is somewhat limited by both their low catalytic efficiency and stability. In the present study, we conducted a detailed functional and structural characterization of the thermostable BsCel5A (Bacillus subtilis cellulase 5A), which consists of a GH5 (glycoside hydrolase 5) catalytic domain fused to a CBM3 (family 3 carbohydrate-binding module). NMR structural analysis revealed that the Bacillus CBM3 represents a new subfamily, which lacks the classical calcium-binding motif, and variations in NMR frequencies in the presence of cellopentaose showed the importance of polar residues in the carbohydrate interaction. Together with the catalytic domain, the CBM3 forms a large planar surface for cellulose recognition, which conducts the substrate in a proper conformation to the active site and increases enzymatic efficiency. Notably, the manganese ion was demonstrated to have a hyper-stabilizing effect on BsCel5A, and by using deletion constructs and X-ray crystallography we determined that this effect maps to a negatively charged motif located at the opposite face of the catalytic site.


Subject(s)
Bacillus subtilis/enzymology , Bacterial Proteins/metabolism , Cellulases/metabolism , Bacillus subtilis/genetics , Bacillus subtilis/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Calcium/metabolism , Cellulases/chemistry , Cellulases/genetics , Cloning, Molecular , Gene Expression Regulation, Bacterial/physiology , Hot Temperature , Kinetics , Manganese/chemistry , Models, Molecular , Protein Conformation , Protein Structure, Tertiary , Substrate Specificity
SELECTION OF CITATIONS
SEARCH DETAIL