Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
ACS Med Chem Lett ; 13(7): 1172-1181, 2022 Jul 14.
Article in English | MEDLINE | ID: mdl-35859869

ABSTRACT

Surface plasmon resonance (SPR) is a widely used method to study ligand-protein interactions. The throughput and sensitivity of SPR has made it an important technology for measuring low-affinity, ultralow weight fragments (<200 Da) in the early stages of drug discovery. However, the biochemistry of membrane proteins, such as G-protein-coupled receptors (GPCRs), makes their SPR fragment screening particularly challenging, especially for native/wild-type, nonthermostabilized mutant receptors. In this study, we demonstrate the use of SPR-based biosensors to study the entire human family of adenosine receptors and present biologically active novel binders with a range of selectivity to human adenosine 2a receptor (hA2AR) from an ultralow weight fragment library and the public GlaxoSmithKline (GSK) kinase library. Thus, we demonstrate the ability of SPR to screen ultra-low-affinity fragments and identify biologically meaningful chemical equity and that SPR campaigns are highly effective "chemical filters" for screening small building block fragments that can be used to enable drug discovery programs.

2.
Nat Commun ; 10(1): 2208, 2019 05 17.
Article in English | MEDLINE | ID: mdl-31101817

ABSTRACT

Cortical force generators connect epithelial polarity sites with astral microtubules, allowing dynein movement to orient the mitotic spindle as astral microtubules depolymerize. Complexes of the LGN and NuMA proteins, fundamental components of force generators, are recruited to the cortex by Gαi-subunits of heterotrimeric G-proteins. They associate with dynein/dynactin and activate the motor activity pulling on astral microtubules. The architecture of cortical force generators is unknown. Here we report the crystal structure of NuMA:LGN hetero-hexamers, and unveil their role in promoting the assembly of active cortical dynein/dynactin motors that are required in orchestrating oriented divisions in polarized cells. Our work elucidates the basis for the structural organization of essential spindle orientation motors.


Subject(s)
Antigens, Nuclear/metabolism , Cell Polarity , Intracellular Signaling Peptides and Proteins/metabolism , Nuclear Matrix-Associated Proteins/metabolism , Spindle Apparatus/metabolism , Antigens, Nuclear/chemistry , Antigens, Nuclear/genetics , Antigens, Nuclear/isolation & purification , Caco-2 Cells , Cell Cycle Proteins , Crystallography, X-Ray , Dynactin Complex/metabolism , Dyneins/metabolism , Gene Knockdown Techniques , HEK293 Cells , HeLa Cells , Humans , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/isolation & purification , Microtubules/metabolism , Nuclear Matrix-Associated Proteins/chemistry , Nuclear Matrix-Associated Proteins/genetics , Nuclear Matrix-Associated Proteins/isolation & purification , Protein Binding/physiology , Protein Multimerization/physiology , RNA, Small Interfering/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism
3.
Nat Commun ; 9(1): 1025, 2018 03 09.
Article in English | MEDLINE | ID: mdl-29523789

ABSTRACT

Asymmetric cell divisions balance stem cell proliferation and differentiation to sustain tissue morphogenesis and homeostasis. During asymmetric divisions, fate determinants and niche contacts segregate unequally between daughters, but little is known on how this is achieved mechanistically. In Drosophila neuroblasts and murine mammary stem cells, the association of the spindle orientation protein LGN with the stem cell adaptor Inscuteable has been connected to asymmetry. Here we report the crystal structure of Drosophila LGN in complex with the asymmetric domain of Inscuteable, which reveals a tetrameric arrangement of intertwined molecules. We show that Insc:LGN tetramers constitute stable cores of Par3-Insc-LGN-GαiGDP complexes, which cannot be dissociated by NuMA. In mammary stem cells, the asymmetric domain of Insc bound to LGN:GαiGDP suffices to drive asymmetric fate, and reverts aberrant symmetric divisions induced by p53 loss. We suggest a novel role for the Insc-bound pool of LGN acting independently of microtubule motors to promote asymmetric fate specification.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Asymmetric Cell Division , Cytoskeletal Proteins/metabolism , Drosophila Proteins/metabolism , Drosophila/cytology , Drosophila/metabolism , Guanine Nucleotide Dissociation Inhibitors/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Stem Cells/cytology , Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/genetics , Animals , Cell Cycle Proteins , Cytoskeletal Proteins/chemistry , Cytoskeletal Proteins/genetics , Drosophila/chemistry , Drosophila/genetics , Drosophila Proteins/chemistry , Drosophila Proteins/genetics , Guanine Nucleotide Dissociation Inhibitors/chemistry , Guanine Nucleotide Dissociation Inhibitors/genetics , Humans , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/genetics , Membrane Proteins/chemistry , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Models, Molecular , Protein Binding , Stem Cells/chemistry , Stem Cells/metabolism
4.
J Vis Exp ; (128)2017 10 02.
Article in English | MEDLINE | ID: mdl-28994793

ABSTRACT

Development of new antimicrobials and vaccines for Streptococcus pneumoniae (pneumococcus) are necessary to halt the rapid rise in multiple resistant strains. Carbohydrate substrate binding proteins (SBPs) represent viable targets for the development of protein-based vaccines and new antimicrobials because of their extracellular localization and the centrality of carbohydrate import for pneumococcal metabolism, respectively. Described here is a rationalized integrated protocol to carry out a comprehensive characterization of SP0092, which can be extended to other carbohydrate SBPs from the pneumococcus and other bacteria. This procedure can aid the structure-based design of inhibitors for this class of proteins. Presented in the first part of this manuscript are protocols for biochemical analysis by thermal shift assay, multi angle light scattering (MALS), and size exclusion chromatography (SEC), which optimize the stability and homogeneity of the sample directed to crystallization trials and so enhance the probability of success. The second part of this procedure describes the characterization of the SBP crystals using a tunable wavelength anomalous diffraction synchrotron beamline, and data collection protocols for measuring data that can be used to resolve the crystallized protein structure.


Subject(s)
Bacterial Proteins/chemistry , Crystallography, X-Ray/methods , Receptors, Cell Surface/chemistry , Streptococcus pneumoniae/chemistry , Bacterial Proteins/metabolism , Molecular Structure , Protein Conformation , Receptors, Cell Surface/metabolism , Streptococcus pneumoniae/metabolism
5.
Acta Crystallogr F Struct Biol Commun ; 73(Pt 1): 54-61, 2017 01 01.
Article in English | MEDLINE | ID: mdl-28045395

ABSTRACT

Streptococcus pneumoniae is an opportunistic respiratory pathogen that remains a major cause of morbidity and mortality globally, with infants and the elderly at the highest risk. S. pneumoniae relies entirely on carbohydrates as a source of carbon and dedicates a third of all uptake systems to carbohydrate import. The structure of the carbohydrate-free substrate-binding protein SP0092 at 1.61 Šresolution reveals it to belong to the newly proposed subclass G of substrate-binding proteins, with a ligand-binding pocket that is large enough to accommodate complex oligosaccharides. SP0092 is a dimer in solution and the crystal structure reveals a domain-swapped dimer with the monomer subunits in a closed conformation but in the absence of carbohydrate ligand. This closed conformation may be induced by dimer formation and could be used as a mechanism to regulate carbohydrate uptake.


Subject(s)
Bacterial Proteins/chemistry , Oligosaccharides/chemistry , Receptors, Cell Surface/chemistry , Streptococcus pneumoniae/chemistry , Amino Acid Sequence , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Binding Sites , Cloning, Molecular , Crystallography, X-Ray , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Models, Molecular , Oligosaccharides/metabolism , Plasmids/chemistry , Plasmids/metabolism , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Protein Multimerization , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Streptococcus pneumoniae/metabolism , Substrate Specificity
6.
Structure ; 25(1): 79-93, 2017 01 03.
Article in English | MEDLINE | ID: mdl-27939783

ABSTRACT

Streptococcus pneumoniae is dependent on carbohydrate uptake for colonization and pathogenesis, and dedicates over a third of its transport systems to their uptake. The ability of the pneumococcus to utilize fructooligosaccharides (FOSs) is attributed to the presence of one of two types of FOS ATP-binding cassette (ABC) transporters. Strains encoding SfuABC are only able to utilize short-chain FOSs, while strains encoding FusABC can utilize both short- and long-chain FOSs. The crystal structures of the substrate-binding protein FusA in its open and closed conformations bound to FOSs, and solution scattering data of SfuA, delineate the structural basis for import of short- and long-chain FOSs. The structure of FusA identifies an EF hand-like calcium-binding motif. This is shown to be essential for translocation of FOSs in FusABC and forms the basis for the definition of a new class of substrate-binding proteins that regulate substrate translocation by calcium.


Subject(s)
ATP-Binding Cassette Transporters/chemistry , ATP-Binding Cassette Transporters/metabolism , Calcium/metabolism , Oligosaccharides/metabolism , Streptococcus pneumoniae/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Binding Sites , Crystallography, X-Ray , EF Hand Motifs , Gene Regulatory Networks , Models, Molecular , Protein Binding , Protein Conformation , Streptococcus pneumoniae/chemistry , Substrate Specificity
7.
Methods Mol Biol ; 1261: 233-53, 2015.
Article in English | MEDLINE | ID: mdl-25502203

ABSTRACT

Macromolecular crystallography (MX) is the most powerful technique available to structural biologists to visualize in atomic detail the macromolecular machinery of the cell. Since the emergence of structural genomics initiatives, significant advances have been made in all key steps of the structure determination process. In particular, third-generation synchrotron sources and the application of highly automated approaches to data acquisition and analysis at these facilities have been the major factors in the rate of increase of macromolecular structures determined annually. A plethora of tools are now available to users of synchrotron beamlines to enable rapid and efficient evaluation of samples, collection of the best data, and in favorable cases structure solution in near real time. Here, we provide a short overview of the emerging use of collecting X-ray diffraction data directly from the crystallization experiment. These in situ experiments are now routinely available to users at a number of synchrotron MX beamlines. A practical guide to the use of the method on the MX suite of beamlines at Diamond Light Source is given.


Subject(s)
Crystallography, X-Ray/instrumentation , Crystallography, X-Ray/methods , Macromolecular Substances/chemistry , Automation, Laboratory , Proteomics/instrumentation , Proteomics/methods , Software , Synchrotrons/instrumentation
8.
Cell Mol Life Sci ; 70(21): 4039-46, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23516018

ABSTRACT

Coordinating mitotic spindle dynamics with cortical polarity is essential for stem cell asymmetric divisions. Over the years, the protein Inscuteable (Insc) has emerged as a key element determining the spindle orientation in asymmetric mitoses. Its overexpression increases differentiative divisions in systems as diverse as mouse keratinocytes and radial glial cells. To date, the molecular explanation to account for this phenotype envisioned Insc as an adaptor molecule bridging between the polarity proteins Par3:Par6:aPKC and the spindle pulling machines assembled on NuMA:LGN:Gαi. However, recent biochemical and structural data revealed that Insc and NuMA are competitive interactors of LGN, challenging the simplistic idea of a single apical macromolecular complex, and demanding a revision of the actual working principles of Insc.


Subject(s)
Asymmetric Cell Division , Cell Cycle Proteins/metabolism , Animals , Chickens , Cytoskeletal Proteins/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster , Humans , Keratinocytes/cytology , Mice , Neuroglia/cytology , Phenotype , Protein Structure, Tertiary , Rats , Signal Transduction , Spindle Apparatus/metabolism , Stem Cells
9.
J Biol Chem ; 287(14): 10876-84, 2012 Mar 30.
Article in English | MEDLINE | ID: mdl-22334692

ABSTRACT

The protein ING4 binds to histone H3 trimethylated at Lys-4 (H3K4me3) through its C-terminal plant homeodomain, thus recruiting the HBO1 histone acetyltransferase complex to target promoters. The structure of the plant homeodomain finger bound to an H3K4me3 peptide has been described, as well as the disorder and flexibility in the ING4 central region. We report the crystal structure of the ING4 N-terminal domain, which shows an antiparallel coiled-coil homodimer with each protomer folded into a helix-loop-helix structure. This arrangement suggests that ING4 can bind simultaneously two histone tails on the same or different nucleosomes. Dimerization has a direct impact on ING4 tumor suppressor activity because monomeric mutants lose the ability to induce apoptosis after genotoxic stress. Homology modeling based on the ING4 structure suggests that other ING dimers may also exist.


Subject(s)
Cell Cycle Proteins/chemistry , Cell Cycle Proteins/metabolism , Chromatin/metabolism , Homeodomain Proteins/chemistry , Homeodomain Proteins/metabolism , Protein Multimerization , Tumor Suppressor Proteins/chemistry , Tumor Suppressor Proteins/metabolism , Amino Acid Sequence , Animals , Apoptosis , Cell Cycle Proteins/genetics , Cell Line , Cell Proliferation , Crystallography, X-Ray , Histone Acetyltransferases/metabolism , Homeodomain Proteins/genetics , Humans , Mice , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Mutation , Protein Binding , Protein Structure, Tertiary , Tumor Suppressor Proteins/genetics
10.
Proc Natl Acad Sci U S A ; 108(52): 20998-1003, 2011 Dec 27.
Article in English | MEDLINE | ID: mdl-22171003

ABSTRACT

Coupling of spindle orientation to cellular polarity is a prerequisite for epithelial asymmetric cell divisions. The current view posits that the adaptor Inscuteable (Insc) bridges between Par3 and the spindle tethering machinery assembled on NuMALGNGαi(GDP), thus triggering apico-basal spindle orientation. The crystal structure of the Drosophila ortholog of LGN (known as Pins) in complex with Insc reveals a modular interface contributed by evolutionary conserved residues. The structure also identifies a positively charged patch of LGN binding to an invariant EPE-motif present on both Insc and NuMA. In vitro competition assays indicate that Insc competes with NuMA for LGN binding, displaying a higher affinity, and that it is capable of opening the LGN conformational switch. The finding that Insc and NuMA are mutually exclusive interactors of LGN challenges the established model of force generators assembly, which we revise on the basis of the newly discovered biochemical properties of the intervening components.


Subject(s)
Asymmetric Cell Division/physiology , Cytoskeletal Proteins/genetics , Drosophila Proteins/genetics , Epithelial Cells/physiology , Guanine Nucleotide Dissociation Inhibitors/genetics , Membrane Proteins/genetics , Models, Molecular , Nerve Tissue Proteins/genetics , Amino Acid Sequence , Animals , Antigens, Nuclear/genetics , Antigens, Nuclear/metabolism , Binding, Competitive/physiology , Cell Cycle Proteins , Cell Polarity/physiology , Chickens , Chromatography, Affinity , Chromatography, Ion Exchange , Crystallization , Cytoskeletal Proteins/metabolism , Drosophila , Drosophila Proteins/metabolism , Electrophoresis, Polyacrylamide Gel , Fluorescence Polarization , Guanine Nucleotide Dissociation Inhibitors/metabolism , Humans , Membrane Proteins/metabolism , Mice , Molecular Sequence Data , Nerve Tissue Proteins/metabolism , Nuclear Matrix-Associated Proteins/genetics , Nuclear Matrix-Associated Proteins/metabolism , Oryzias , Protein Conformation , Sequence Alignment , Spindle Apparatus/physiology , Static Electricity , Xenopus laevis , Zebrafish
11.
Article in English | MEDLINE | ID: mdl-20445261

ABSTRACT

Inhibitor of growth protein 4 (ING4) belongs to the ING family of tumour suppressors and is involved in chromatin remodelling, in growth arrest and, in cooperation with p53, in senescence and apoptosis. Whereas the structure and histone H3-binding properties of the C-terminal PHD domains of the ING proteins are known, no structural information is available for the N-terminal domains. This domain contains a putative oligomerization site rich in helical structure in the ING2-5 members of the family. The N-terminal domain of ING4 was overexpressed in Escherichia coli and purified to homogeneity. Crystallization experiments yielded crystals that were suitable for high-resolution X-ray diffraction analysis. The crystals belonged to the orthorhombic space group C222, with unit-cell parameters a = 129.7, b = 188.3, c = 62.7 A. The self-rotation function and the Matthews coefficient suggested the presence of three protein dimers per asymmetric unit. The crystals diffracted to a resolution of 2.3 A using synchrotron radiation at the Swiss Light Source (SLS) and the European Synchrotron Radiation Facility (ESRF).


Subject(s)
Protein Multimerization , Tumor Suppressor Proteins/chemistry , Crystallization , Crystallography, X-Ray , Humans , Protein Interaction Domains and Motifs , Tumor Suppressor Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...