Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Phys Chem Chem Phys ; 22(41): 24086-24096, 2020 Oct 28.
Article in English | MEDLINE | ID: mdl-33079118

ABSTRACT

G protein-coupled receptors (GPCRs) are a large and ubiquitous family of membrane receptors of great pharmacological interest. Cell-based assays are the primary tool for assessing GPCR interactions and activation but their design and intrinsic complexity limit their application. Biosensor-based assays that directly and specifically report GPCR-protein binding (e.g. arrestin or G protein) could provide a good alternative. We present an approach based on the stable immobilization of different arrestin-3 proteins (wild type, and two mutants, mutant X (arrestin-3 I386A) and mutant Y (arrestin-3 R393E)) via histidine tags on NTA(Ni2+)-coated sensors in a defined orientation. Using biolayer interferometry (BLI), surface plasmon resonance (SPR), and quartz crystal microbalance with dissipation (QCM-D), we were able to follow the interaction between the different arrestin-3 proteins and a representative GPCR, jumping spider rhodopsin-1 (JSR1), in a label-free manner in real-time. The interactions were quantified as binding affinity, association and dissociation rate constants. The combination of surface-based biosensing methods indicated that JSR1 showed the strongest binding to arrestin mutant Y. Taken together, this work introduces direct label-free, biosensor-based screening approaches that can be easily adapted for testing interactions of proteins and other compounds with different GPCRs.


Subject(s)
Immobilized Proteins/metabolism , Rhodopsin/metabolism , beta-Arrestin 2/metabolism , Animals , Arthropod Proteins/metabolism , Biosensing Techniques , Immobilized Proteins/genetics , Lipid Bilayers/chemistry , Mutation , Phosphatidylcholines/chemistry , Protein Binding , Quartz Crystal Microbalance Techniques , Spiders/chemistry , Surface Plasmon Resonance , beta-Arrestin 2/genetics
2.
Sci Rep ; 9(1): 439, 2019 01 24.
Article in English | MEDLINE | ID: mdl-30679635

ABSTRACT

Arrestin-1 desensitizes the activated and phosphorylated photoreceptor rhodopsin by forming transient rhodopsin-arrestin-1 complexes that eventually decay to opsin, retinal and arrestin-1. Via a multi-dimensional screening setup, we identified and combined arrestin-1 mutants that form lasting complexes with light-activated and phosphorylated rhodopsin in harsh conditions, such as high ionic salt concentration. Two quadruple mutants, D303A + T304A + E341A + F375A and R171A + T304A + E341A + F375A share similar heterologous expression and thermo-stability levels with wild type (WT) arrestin-1, but are able to stabilize complexes with rhodopsin with more than seven times higher half-maximal inhibitory concentration (IC50) values for NaCl compared to the WT arrestin-1 protein. These quadruple mutants are also characterized by higher binding affinities to phosphorylated rhodopsin, light-activated rhodopsin and phosphorylated opsin, as compared with WT arrestin-1. Furthermore, the assessed arrestin-1 mutants are still specifically associating with phosphorylated or light-activated receptor states only, while binding to the inactive ground state of the receptor is not significantly altered. Additionally, we propose a novel functionality for R171 in stabilizing the inactive arrestin-1 conformation as well as the rhodopsin-arrestin-1 complex. The achieved stabilization of the active rhodopsin-arrestin-1 complex might be of great interest for future structure determination, antibody development studies as well as drug-screening efforts targeting G protein-coupled receptors (GPCRs).


Subject(s)
Arrestins/metabolism , Multiprotein Complexes/metabolism , Opsins/metabolism , Protein Engineering/methods , Rhodopsin/metabolism , Animals , Arrestins/chemistry , Arrestins/genetics , Cattle , HEK293 Cells , Humans , Models, Molecular , Multiprotein Complexes/chemistry , Multiprotein Complexes/genetics , Mutation , Opsins/chemistry , Phosphorylation , Protein Binding , Protein Conformation , Protein Stability , Rhodopsin/chemistry
4.
Sci Rep ; 6: 28686, 2016 06 28.
Article in English | MEDLINE | ID: mdl-27350090

ABSTRACT

Arrestins desensitize G protein-coupled receptors (GPCRs) and act as mediators of signalling. Here we investigated the interactions of arrestin-1 with two functionally distinct forms of the dim-light photoreceptor rhodopsin. Using unbiased scanning mutagenesis we probed the individual contribution of each arrestin residue to the interaction with the phosphorylated apo-receptor (Ops-P) and the agonist-bound form (Meta II-P). Disruption of the polar core or displacement of the C-tail strengthened binding to both receptor forms. In contrast, mutations of phosphate-binding residues (phosphosensors) suggest the phosphorylated receptor C-terminus binds arrestin differently for Meta II-P and Ops-P. Likewise, mutations within the inter-domain interface, variations in the receptor-binding loops and the C-edge of arrestin reveal different binding modes. In summary, our results indicate that arrestin-1 binding to Meta II-P and Ops-P is similarly dependent on arrestin activation, although the complexes formed with these two receptor forms are structurally distinct.


Subject(s)
Arrestin/chemistry , Models, Molecular , Opsins/chemistry , Animals , Arrestin/genetics , Arrestin/metabolism , Cattle , Mutation , Opsins/genetics , Opsins/metabolism , Protein Binding , Protein Domains
5.
Curr Opin Struct Biol ; 29: 143-51, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25484000

ABSTRACT

The past years have seen tremendous progress towards understanding how arrestins recognize phosphorylated G protein-coupled receptors (GPCRs). Two arrestin crystal structures, one of a pre-activated splice variant and one bound to a GPCR phosphopeptide, provided insights into the conformational changes upon phosphate recognition. Scanning mutagenesis and spectroscopic studies complete the picture of arrestin activation and receptor binding. Most perspicuous is the C-tail exchange mechanism, by which the C-tail of arrestin is released from its basal conformation and replaced by the phosphorylated GPCR C-terminus. Three positively charged clusters could act as conserved arrestin phosphosensors. Variations in the pattern of phosphorylation in a GPCR and variations within the C-terminus of different GPCRs may encode specificity to arrestin subtypes and particular physiological responses.


Subject(s)
Arrestins/chemistry , Receptors, G-Protein-Coupled/chemistry , Amino Acid Sequence , Molecular Sequence Data , Mutagenesis , Phosphopeptides/chemistry , Phosphorylation , Protein Binding , Protein Conformation
6.
Proc Natl Acad Sci U S A ; 111(5): 1825-30, 2014 Feb 04.
Article in English | MEDLINE | ID: mdl-24449856

ABSTRACT

Arrestins function as adapter proteins that mediate G protein-coupled receptor (GPCR) desensitization, internalization, and additional rounds of signaling. Here we have compared binding of the GPCR rhodopsin to 403 mutants of arrestin-1 covering its complete sequence. This comprehensive and unbiased mutagenesis approach provides a functional dimension to the crystal structures of inactive, preactivated p44 and phosphopeptide-bound arrestins and will guide our understanding of arrestin-GPCR complexes. The presented functional map quantitatively connects critical interactions in the polar core and along the C tail of arrestin. A series of amino acids (Phe375, Phe377, Phe380, and Arg382) anchor the C tail in a position that blocks binding of the receptor. Interaction of phosphates in the rhodopsin C terminus with Arg29 controls a C-tail exchange mechanism in which the C tail of arrestin is released and exposes several charged amino acids (Lys14, Lys15, Arg18, Lys20, Lys110, and Lys300) for binding of the phosphorylated receptor C terminus. In addition to this arrestin phosphosensor, our data reveal several patches of amino acids in the finger (Gln69 and Asp73-Met75) and the lariat loops (L249-S252 and Y254) that can act as direct binding interfaces. A stretch of amino acids at the edge of the C domain (Trp194-Ser199, Gly337-Gly340, Thr343, and Thr345) could act as membrane anchor, binding interface for a second rhodopsin, or rearrange closer to the central loops upon complex formation. We discuss these interfaces in the context of experimentally guided docking between the crystal structures of arrestin and light-activated rhodopsin.


Subject(s)
Amino Acids/metabolism , Arrestin/metabolism , Animals , Arrestin/chemistry , Cattle , Humans , Inhibitory Concentration 50 , Luminescent Proteins/metabolism , Models, Molecular , Mutagenesis , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Phosphorylation , Rats , Recombinant Fusion Proteins/metabolism , Rhodopsin/chemistry , Rhodopsin/metabolism , Rod Cell Outer Segment/metabolism , Red Fluorescent Protein
7.
PLoS One ; 8(10): e78878, 2013.
Article in English | MEDLINE | ID: mdl-24205336

ABSTRACT

Scanning mutagenesis is a powerful protein engineering technique used to study protein structure-function relationship, map binding sites and design more stable proteins or proteins with altered properties. One of the time-consuming tasks encountered in application of this technique is the design of primers for site-directed mutagenesis. Here we present an open-source multi-platform software AAscan developed to design primers for this task according to a set of empirical rules such as melting temperature, overall length, length of overlap regions, and presence of GC clamps at the 3' end, for any desired substitution. We also describe additional software tools which are used to analyse a large number of sequencing results for the presence of desired mutations, as well as related software to design primers for ligation independent cloning. We have used AAscan software to design primers to make over 700 mutants, with a success rate of over 80%. We hope that the open-source nature of our software and ready availability of freeware tools used for its development will facilitate its adaptation and further development. The software is distributed under GPLv3 licence and is available at http://www.psi.ch/lbr/aascan.


Subject(s)
DNA Primers/genetics , Mutagenesis , Protein Engineering/methods , Software , Arrestin/genetics , GTP-Binding Protein alpha Subunits, Gi-Go/genetics , Polymerase Chain Reaction , User-Computer Interface
8.
Cell Signal ; 25(11): 2155-62, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23872075

ABSTRACT

The effects of activating mutations associated with night blindness on the stoichiometry of rhodopsin interactions with G protein-coupled receptor kinase 1 (GRK1) and arrestin-1 have not been reported. Here we show that the monomeric form of WT rhodopsin and its constitutively active mutants M257Y, G90D, and T94I, reconstituted into HDL particles are effectively phosphorylated by GRK1, as well as two more ubiquitously expressed subtypes, GRK2 and GRK5. All versions of arrestin-1 tested (WT, pre-activated, and constitutively monomeric mutants) bind to monomeric rhodopsin and show the same selectivity for different functional forms of rhodopsin as in native disc membranes. Rhodopsin phosphorylation by GRK1 and GRK2 promotes arrestin-1 binding to a comparable extent, whereas similar phosphorylation by GRK5 is less effective, suggesting that not all phosphorylation sites on rhodopsin are equivalent in promoting arrestin-1 binding. The binding of WT arrestin-1 to phospho-opsin is comparable to the binding to its preferred target, P-Rh*, suggesting that in photoreceptors arrestin-1 only dissociates after opsin regeneration with 11-cis-retinal, which converts phospho-opsin into inactive phospho-rhodopsin that has lower affinity for arrestin-1. Reduced binding of arrestin-1 to the phospho-opsin form of G90D mutant likely contributes to night blindness caused by this mutation in humans.


Subject(s)
Arrestin/genetics , G-Protein-Coupled Receptor Kinase 1/genetics , Mutation , Night Blindness/genetics , Retinal Rod Photoreceptor Cells/metabolism , Rhodopsin/genetics , Animals , Arrestin/metabolism , Cattle , Cholesterol, HDL/chemistry , Cholesterol, HDL/metabolism , G-Protein-Coupled Receptor Kinase 1/metabolism , Gene Expression Regulation , Isoenzymes/genetics , Isoenzymes/metabolism , Night Blindness/metabolism , Night Blindness/pathology , Opsins/genetics , Opsins/metabolism , Phosphoproteins/genetics , Phosphoproteins/metabolism , Phosphorylation , Protein Binding , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Multimerization , Retinal Rod Photoreceptor Cells/pathology , Rhodopsin/metabolism , Signal Transduction
9.
EMBO Rep ; 14(6): 520-6, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23579341

ABSTRACT

We present active-state structures of the G protein-coupled receptor (GPCRs) rhodopsin carrying the disease-causing mutation G90D. Mutations of G90 cause either retinitis pigmentosa (RP) or congenital stationary night blindness (CSNB), a milder, non-progressive form of RP. Our analysis shows that the CSNB-causing G90D mutation introduces a salt bridge with K296. The mutant thus interferes with the E113Q-K296 activation switch and the covalent binding of the inverse agonist 11-cis-retinal, two interactions that are crucial for the deactivation of rhodopsin. Other mutations, including G90V causing RP, cannot promote similar interactions. We discuss our findings in context of a model in which CSNB is caused by constitutive activation of the visual signalling cascade.


Subject(s)
Eye Diseases, Hereditary/genetics , Genetic Diseases, X-Linked/genetics , Mutation, Missense , Myopia/genetics , Night Blindness/genetics , Rhodopsin/chemistry , Arrestin/chemistry , Crystallography, X-Ray , HEK293 Cells , Humans , Models, Molecular , Protein Binding , Protein Stability , Protein Structure, Secondary , Protein Structure, Tertiary , Rhodopsin/genetics , Schiff Bases , Structural Homology, Protein , Transition Temperature
10.
J Cell Biol ; 192(2): 277-93, 2011 Jan 24.
Article in English | MEDLINE | ID: mdl-21263028

ABSTRACT

RNA polymerase I (Pol I) produces large ribosomal RNAs (rRNAs). In this study, we show that the Rpa49 and Rpa34 Pol I subunits, which do not have counterparts in Pol II and Pol III complexes, are functionally conserved using heterospecific complementation of the human and Schizosaccharomyces pombe orthologues in Saccharomyces cerevisiae. Deletion of RPA49 leads to the disappearance of nucleolar structure, but nucleolar assembly can be restored by decreasing ribosomal gene copy number from 190 to 25. Statistical analysis of Miller spreads in the absence of Rpa49 demonstrates a fourfold decrease in Pol I loading rate per gene and decreased contact between adjacent Pol I complexes. Therefore, the Rpa34 and Rpa49 Pol I-specific subunits are essential for nucleolar assembly and for the high polymerase loading rate associated with frequent contact between adjacent enzymes. Together our data suggest that localized rRNA production results in spatially constrained rRNA production, which is instrumental for nucleolar assembly.


Subject(s)
Gene Expression Regulation, Fungal/genetics , Genes, rRNA/genetics , Protein Subunits/metabolism , RNA Polymerase I/metabolism , Transcription, Genetic/genetics , Cell Nucleolus , Cell Nucleus Shape , Conserved Sequence , Genetic Complementation Test , Humans , Nuclear Localization Signals , Protein Multimerization , Protein Subunits/chemistry , RNA Polymerase I/chemistry , Saccharomyces cerevisiae/enzymology , Saccharomyces cerevisiae/genetics , Schizosaccharomyces/enzymology , Schizosaccharomyces/genetics , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...