Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
PLoS One ; 19(3): e0300981, 2024.
Article in English | MEDLINE | ID: mdl-38517842

ABSTRACT

Taste receptor type 1 (T1r) proteins are responsible for recognizing nutrient chemicals in foods. In humans, T1r2/T1r3 and T1r1/T1r3 heterodimers serve as the sweet and umami receptors that recognize sugars or amino acids and nucleotides, respectively. T1rs are conserved among vertebrates, and T1r2a/T1r3 from medaka fish is currently the only member for which the structure of the ligand-binding domain (LBD) has been solved. T1r2a/T1r3 is an amino acid receptor that recognizes various l-amino acids in its LBD as observed with other T1rs exhibiting broad substrate specificities. Nevertheless, the range of chemicals that are recognized by T1r2a/T1r3LBD has not been extensively explored. In the present study, the binding of various chemicals to medaka T1r2a/T1r3LBD was analyzed. A binding assay for amino acid derivatives verified the specificity of this protein to l-α-amino acids and the importance of α-amino and carboxy groups for receptor recognition. The results further indicated the significance of the α-hydrogen for recognition as replacing it with a methyl group resulted in a substantially decreased affinity. The binding ability to the protein was not limited to proteinogenic amino acids, but also to non-proteinogenic amino acids, such as metabolic intermediates. Besides l-α-amino acids, no other chemicals showed significant binding to the protein. These results indicate that all of the common structural groups of α-amino acids and their geometry in the l-configuration are recognized by the protein, whereas a wide variety of α-substituents can be accommodated in the ligand binding sites of the LBDs.


Subject(s)
Oryzias , Taste , Animals , Humans , Receptors, G-Protein-Coupled/metabolism , Oryzias/metabolism , Receptors, Amino Acid , Ligands , Amino Acids
2.
Protein J ; 42(6): 698-708, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37737932

ABSTRACT

Single-chain monellin (SCM) is an engineered protein that links the two chains of monellin, a naturally sweet-tasting protein. This protein is an attractive candidate for use as a sugar replacement in food and beverages and has numerous other applications. Therefore, generating SCM mutants with improved stability is an active area of research to broaden the range of its potential applications. In this study, we focused on the Cys41 residue of SCM, which is a single cysteine residue present at a structurally important position. This residue is often substituted with Ser. However, this substitution may destabilize SCM because Cys41 is buried in the hydrophobic core of the protein. Therefore, we designed mutants that substituted Ala, Val, and Leu for this residue, namely C41A, C41V, and C41L. We characterized these three mutants, SCM C41S, and wild type (WT). Differential scanning fluorimetric analysis revealed that substituting Cys41 with Ala or Val increased the thermal stability of SCM, while substitution with Ser or Leu decreased its stability. Determination of the crystal structures of SCM C41A and C41V mutants revealed that the overall structures and main chain structures around the 41st residue of both mutants were almost identical to the WT. On the other hand, the orientations of the amino acid side chains near the 41st residue differed among the SCM variants. Taken together, our results indicate that substituting Cys41 with Ala or Val increases the stability of SCM and provide insight into the structural basis of this improvement.

3.
J Biochem ; 174(5): 451-459, 2023 Oct 31.
Article in English | MEDLINE | ID: mdl-37527916

ABSTRACT

Sweet and umami taste receptors recognize chemicals such as sugars and amino acids on their extracellular side and transmit signals into the cytosol of the taste cell. In contrast to ligands that act on the extracellular side of these receptors, little is known regarding the molecules that regulate receptor functions within the cytosol. In this study, we analysed the interaction between sweet and umami taste receptors and calmodulin, a representative Ca2+-dependent cytosolic regulatory protein. High prediction scores for calmodulin binding were observed on the C-terminal cytosolic side of mouse taste receptor type 1 subunit 3 (T1r3), a subunit that is common to both sweet and umami taste receptors. Pull-down assay and surface plasmon resonance analyses showed different affinities of calmodulin to the C-terminal tails of distinct T1r subtypes. Furthermore, we found that T1r3 and T1r2 showed the highest and considerable binding to calmodulin, whereas T1r1 showed weaker binding affinity. Finally, the binding of calmodulin to T1rs was consistently higher in the presence of Ca2+ than in its absence. The results suggested a possibility of the Ca2+-dependent feedback regulation process of sweet and umami taste receptor signaling by calmodulin.


Subject(s)
Calmodulin , Taste , Mice , Animals , Taste/physiology , Calmodulin/metabolism , Receptors, G-Protein-Coupled/metabolism , Calcium/metabolism , Cytosol/metabolism
4.
Elife ; 122023 02 28.
Article in English | MEDLINE | ID: mdl-36852482

ABSTRACT

Salt taste sensation is multifaceted: NaCl at low or high concentrations is preferably or aversively perceived through distinct pathways. Cl- is thought to participate in taste sensation through an unknown mechanism. Here, we describe Cl- ion binding and the response of taste receptor type 1 (T1r), a receptor family composing sweet/umami receptors. The T1r2a/T1r3 heterodimer from the medaka fish, currently the sole T1r amenable to structural analyses, exhibited a specific Cl- binding in the vicinity of the amino-acid-binding site in the ligand-binding domain (LBD) of T1r3, which is likely conserved across species, including human T1r3. The Cl- binding induced a conformational change in T1r2a/T1r3LBD at sub- to low-mM concentrations, similar to canonical taste substances. Furthermore, oral Cl- application to mice increased impulse frequencies of taste nerves connected to T1r-expressing taste cells and promoted their behavioral preferences attenuated by a T1r-specific blocker or T1r3 knock-out. These results suggest that the Cl- evokes taste sensations by binding to T1r, thereby serving as another preferred salt taste pathway at a low concentration.


Humans perceive taste when proteins called taste receptors on the surface of the tongue are activated by molecules of food. These receptors turn on nerve cells that send signals the brain can read as sweet, sour, salty, bitter, or umami, depending on which receptor was activated. Most animals with backbones share the same five types of taste receptors. In food, salty flavors are usually the result of adding table salt, which has two components: a sodium ion and chloride ion. The main taste receptors that signal to the brain that a food is salty become activated when they bind to the sodium ion. However, some studies have shown that salt is also perceived as sweet when eaten in minuscule amounts. It is poorly understood why this happens, but it is possible that the chloride half of salt drives the sweet taste. In 2017, scientists worked out the structure of a taste receptor from a fish, that is equivalent to the sweet receptor in humans. Curiously, one part of this receptor, known as T1r2a/T1r3LBD, was bound to a chloride ion. This prompted Atsumi, Yasumatsu et al. to think about the 'sweet' taste of salt, leading them to take a closer look at T1r2a/T1r3LBD and whether chloride could indeed activate it. Atsumi, Yasumatsu et al. used structural biology techniques to examine T1r2a/T1r3LBD and found evidence that the receptor might be binding chloride. Further biophysical experiments confirmed that chloride does indeed bind to the receptor, and that it also causes it to change shape. Usually, changes in shape are hallmarks of receptor activation, suggesting that chloride may activate T1r2a/T1r3LBD. Next, Atsumi, Yasumatsu et al. checked whether chloride could stimulate the neurons that signal when food tastes sweet, by using an approach known as electrophysiology to measure the activity of these neurons in mice. The results showed that the neurons became active when a solution containing small amounts of chloride was placed on the mouse's tongue. This activity went away when a compound that can block the receptor's activity was delivered alongside the chloride. Additionally, when mice were given a choice of plain water or water containing chloride, they seemed to prefer the latter. This confirmed that mice recognized the sweetness of chloride via the activation of sweet taste receptors and neurons. Based on these findings, Atsumi, Yasumatsu et al. propose that small amounts of salt may taste sweet because the chloride ions in the salt activate sweet taste receptors and their linked neurons. Their results also suggest that animals sense salt in many ways, likely because balanced salt levels are essential for the body to work properly. Future experiments on human taste receptors may reveal how these pathways help assess salt levels in humans.


Subject(s)
Taste Buds , Taste , Animals , Humans , Mice , Chlorides , Ligands , Sodium Chloride , Sodium Chloride, Dietary , Extracellular Space/metabolism
5.
Nat Commun ; 14(1): 621, 2023 02 04.
Article in English | MEDLINE | ID: mdl-36739439

ABSTRACT

DNA methyltransferases (DNMTs) catalyze methylation at the C5 position of cytosine with S-adenosyl-L-methionine. Methylation regulates gene expression, serving a variety of physiological and pathophysiological roles. The chemical mechanisms regulating DNMT enzymatic activity, however, are not fully elucidated. Here, we show that protein S-nitrosylation of a cysteine residue in DNMT3B attenuates DNMT3B enzymatic activity and consequent aberrant upregulation of gene expression. These genes include Cyclin D2 (Ccnd2), which is required for neoplastic cell proliferation in some tumor types. In cell-based and in vivo cancer models, only DNMT3B enzymatic activity, and not DNMT1 or DNMT3A, affects Ccnd2 expression. Using structure-based virtual screening, we discovered chemical compounds that specifically inhibit S-nitrosylation without directly affecting DNMT3B enzymatic activity. The lead compound, designated DBIC, inhibits S-nitrosylation of DNMT3B at low concentrations (IC50 ≤ 100 nM). Treatment with DBIC prevents nitric oxide (NO)-induced conversion of human colonic adenoma to adenocarcinoma in vitro. Additionally, in vivo treatment with DBIC strongly attenuates tumor development in a mouse model of carcinogenesis triggered by inflammation-induced generation of NO. Our results demonstrate that de novo DNA methylation mediated by DNMT3B is regulated by NO, and DBIC protects against tumor formation by preventing aberrant S-nitrosylation of DNMT3B.


Subject(s)
DNA (Cytosine-5-)-Methyltransferases , Epigenesis, Genetic , Animals , Humans , Mice , Cell Transformation, Neoplastic/genetics , DNA (Cytosine-5-)-Methyltransferase 1/genetics , DNA (Cytosine-5-)-Methyltransferase 1/metabolism , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methylation , DNA Modification Methylases/metabolism , DNA Methyltransferase 3B
6.
J Biochem ; 169(5): 585-599, 2021 Jul 03.
Article in English | MEDLINE | ID: mdl-33386843

ABSTRACT

Synthetic binding proteins that have the ability to bind with molecules can be generated using various protein domains as non-antibody scaffolds. These designer proteins have been used widely in research studies, as their properties overcome the disadvantages of using antibodies. Here, we describe the first application of a phage display to generate synthetic binding proteins using a sweet protein, monellin, as a non-antibody scaffold. Single-chain monellin (scMonellin), in which two polypeptide chains of natural monellin are connected by a short linker, has two loops on one side of the molecule. We constructed phage display libraries of scMonellin, in which the amino acid sequence of the two loops is diversified. To validate the performance of these libraries, we sorted them against the folding mutant of the green fluorescent protein variant (GFPuv) and yeast small ubiquitin-related modifier. We successfully obtained scMonellin variants exhibiting moderate but significant affinities for these target proteins. Crystal structures of one of the GFPuv-binding variants in complex with GFPuv revealed that the two diversified loops were involved in target recognition. scMonellin, therefore, represents a promising non-antibody scaffold in the design and generation of synthetic binding proteins. We termed the scMonellin-derived synthetic binding proteins 'SWEEPins'.


Subject(s)
Carrier Proteins/chemistry , Peptide Library , Plant Proteins/chemistry , Ranunculales/chemistry , Carrier Proteins/genetics , Plant Proteins/genetics , Ranunculales/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics
7.
J Biochem ; 169(5): 555-564, 2021 Jul 03.
Article in English | MEDLINE | ID: mdl-33377147

ABSTRACT

The large, secreted glycoprotein reelin regulates embryonic brain development as well as adult brain functions. Although reelin binds to its receptors via its central part, the N-terminal region directs multimer formation and is critical for efficient signal transduction. In fact, the inhibitory antibody CR-50 interacts with the N-terminal region and prevents higher-order multimerization and signalling. Reelin is a multidomain protein in which the central part is composed of eight characteristic repeats, named reelin repeats, each of which is further divided by insertion of a epidermal growth factor (EGF) module into two subrepeats. In contrast, the N-terminal region shows unique 'irregular' domain architecture since it comprises three consecutive subrepeats without the intervening EGF module. Here, we determined the crystal structure of the murine reelin fragment named RX-R1 including the irregular region and the first reelin repeat at 2.0-Å resolution. The overall structure of RX-R1 has a branched Y-shaped form. Interestingly, two incomplete subrepeats cooperatively form one entire subrepeat structure, though an additional subrepeat is inserted between them. We further reveal that Arg335 of RX-R1 is crucial for binding CR-50. A possible self-association mechanism via the N-terminal region is proposed based on our results.


Subject(s)
Cell Adhesion Molecules, Neuronal/chemistry , Extracellular Matrix Proteins/chemistry , Nerve Tissue Proteins/chemistry , Protein Multimerization , Serine Endopeptidases/chemistry , Animals , Antibodies, Monoclonal/chemistry , Cell Adhesion Molecules, Neuronal/genetics , Crystallography, X-Ray , Extracellular Matrix Proteins/genetics , Mice , Nerve Tissue Proteins/genetics , Protein Domains , Reelin Protein , Repetitive Sequences, Amino Acid , Serine Endopeptidases/genetics
8.
PLoS One ; 14(10): e0218909, 2019.
Article in English | MEDLINE | ID: mdl-31584955

ABSTRACT

Taste receptor type 1 (T1r) is responsible for the perception of essential nutrients, such as sugars and amino acids, and evoking sweet and umami (savory) taste sensations. T1r receptors recognize many of the taste substances at their extracellular ligand-binding domains (LBDs). In order to detect a wide array of taste substances in the environment, T1r receptors often possess broad ligand specificities. However, the entire ranges of chemical spaces and their binding characteristics to any T1rLBDs have not been extensively analyzed. In this study, we exploited the differential scanning fluorimetry (DSF) to medaka T1r2a/T1r3LBD, a current sole T1rLBD heterodimer amenable for recombinant preparation, and analyzed their thermal stabilization by adding various amino acids. The assay showed that the agonist amino acids induced thermal stabilization and shifted the melting temperatures (Tm) of the protein. An agreement between the DSF results and the previous biophysical assay was observed, suggesting that DSF can detect ligand binding at the orthosteric-binding site in T1r2a/T1r3LBD. The assay further demonstrated that most of the tested l-amino acids, but no d-amino acid, induced Tm shifts of T1r2a/T1r3LBD, indicating the broad l-amino acid specificities of the proteins probably with several different manners of recognition. The Tm shifts by each amino acid also showed a fair correlation with the responses exhibited by the full-length receptor, verifying the broad amino-acid binding profiles at the orthosteric site in LBD observed by DSF.


Subject(s)
Amino Acids/chemistry , Fish Proteins/chemistry , Oryzias , Protein Multimerization , Receptors, G-Protein-Coupled/chemistry , Animals , Binding Sites
9.
Sci Rep ; 9(1): 4722, 2019 03 18.
Article in English | MEDLINE | ID: mdl-30886277

ABSTRACT

Green fluorescent protein (GFP) is amenable to recombinant expression in various kinds of cells and is widely used in life science research. We found that the recombinant expression of GFPuv, a commonly-used mutant of GFP, in E. coli produced two distinct molecular species as judged by in-gel fluorescence SDS-PAGE. These molecular species, namely form I and II, could be separately purified by anion-exchange chromatography without any remarkable differences in the fluorescence spectra. Mass spectrometric analyses revealed that the molecular mass of form I is almost the same as the calculated value, while that of form II is approximately 1 Da larger than that of form I. Further mass spectrometric top-down sequencing pinpointed the modification in GFPuv form II, where the ε-amino group of the C-terminal Lys238 residue is converted into the hydroxyl group. No equivalent modification was observed in the native GFP in jellyfish Aequorea victoria, suggesting that this modification is not physiologically relevant. Crystal structure analysis of the two species verified the structural identity of the backbone and the vicinity of the chromophore. The modification found in this study may also be generated in other GFP variants as well as in other recombinant expression systems.


Subject(s)
Green Fluorescent Proteins/chemistry , Luminescent Agents/chemistry , Lysine/chemistry , Animals , Escherichia coli , Fluorescence , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/isolation & purification , Green Fluorescent Proteins/ultrastructure , Mass Spectrometry , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/isolation & purification , Protein Isoforms/ultrastructure , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/ultrastructure , Scyphozoa/genetics
10.
Nat Commun ; 8: 15530, 2017 05 23.
Article in English | MEDLINE | ID: mdl-28534491

ABSTRACT

The taste receptor type 1 (T1r) family perceives 'palatable' tastes. These receptors function as T1r2-T1r3 and T1r1-T1r3 heterodimers to recognize a wide array of sweet and umami (savory) tastes in sugars and amino acids. Nonetheless, it is unclear how diverse tastes are recognized by so few receptors. Here we present crystal structures of the extracellular ligand-binding domains (LBDs), the taste recognition regions of the fish T1r2-T1r3 heterodimer, bound to different amino acids. The ligand-binding pocket in T1r2LBD is rich in aromatic residues, spacious and accommodates hydrated percepts. Biophysical studies show that this binding site is characterized by a broad yet discriminating chemical recognition, contributing for the particular trait of taste perception. In contrast, the analogous pocket in T1r3LBD is occupied by a rather loosely bound amino acid, suggesting that the T1r3 has an auxiliary role. Overall, we provide a structural basis for understanding the chemical perception of taste receptors.


Subject(s)
Oryzias/physiology , Receptors, G-Protein-Coupled/chemistry , Taste Buds/physiology , Taste/physiology , Animals , Binding Sites , Crystallography, X-Ray , Ligands , Mutagenesis , Protein Structure, Quaternary , Receptors, G-Protein-Coupled/isolation & purification , Receptors, G-Protein-Coupled/physiology , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Structure-Activity Relationship , Taste Buds/chemistry
11.
EMBO Rep ; 18(6): 982-999, 2017 06.
Article in English | MEDLINE | ID: mdl-28446613

ABSTRACT

Apolipoprotein E receptor 2 (ApoER2) is a close homologue of low-density lipoprotein receptor (LDLR) that mediates the endocytosis of ligands, including LDL particles. LDLR family members have been presumed to explore a large conformational space to capture ligands in the extended conformation at the cell surface. Ligands are subsequently released through a pH-titrated structural transition to a self-docked, contracted-closed conformation. In addition to lipoprotein uptake, ApoER2 is implicated in signal transduction during brain development through capture of the extracellular protein reelin. From crystallographic analysis, we determine that the full-length ApoER2 ectodomain adopts an intermediate contracted-open conformation when complexed with the signaling-competent reelin fragment, and we identify a previously unappreciated auxiliary low-affinity binding interface. Based on mutational analyses, we propose that the pH shift during endocytosis weakens the affinity of the auxiliary interface and destabilizes the ligand-receptor complex. Furthermore, this study elucidates that the contracted-open conformation of ligand-bound ApoER2 at neutral pH resembles the contracted-closed conformation of ligand-unbound LDLR at acidic pH in a manner suggestive of being primed for ligand release even prior to internalization.


Subject(s)
Cell Adhesion Molecules, Neuronal/physiology , Extracellular Matrix Proteins/physiology , LDL-Receptor Related Proteins/chemistry , LDL-Receptor Related Proteins/metabolism , Nerve Tissue Proteins/physiology , Serine Endopeptidases/physiology , Animals , CHO Cells , Cell Adhesion Molecules, Neuronal/chemistry , Cricetulus , Crystallography , Endocytosis , Endosomes/physiology , Extracellular Matrix Proteins/chemistry , Humans , Hydrogen-Ion Concentration , LDL-Receptor Related Proteins/genetics , Ligands , Lipoproteins, LDL/metabolism , Nerve Tissue Proteins/chemistry , Neurons/physiology , Protein Conformation , Receptors, LDL/metabolism , Reelin Protein , Serine Endopeptidases/chemistry , Signal Transduction , Surface Plasmon Resonance
13.
Methods Mol Biol ; 1493: 57-72, 2017.
Article in English | MEDLINE | ID: mdl-27787842

ABSTRACT

Plexins are type I membrane proteins that function as receptors for semaphorins. All of the known plexins contain a large globular domain, termed the sema domain, in the N-terminal extracellular region, which interacts with semaphorins during signal transduction. Here, we describe procedures for protein production and purification that we utilized in the crystallographic study of the mouse Plexin A2 (mPlxnA2) extracellular fragment, including the sema domain. A mutant mammalian cell line, HEK293S GnTI-, was used as an expression host for the production of a crystallizable-quality mPlxnA2 fragment, which contains several N-glycosylation sites and disulfide bonds.


Subject(s)
Chromatography, Affinity/methods , Nerve Tissue Proteins/isolation & purification , Receptors, Cell Surface/isolation & purification , Animals , Crystallography, X-Ray , Electrophoresis, Polyacrylamide Gel , Glycosylation , HEK293 Cells , Humans , Mice , Mutation , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/genetics , Receptors, Cell Surface/chemistry , Receptors, Cell Surface/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification
14.
Sci Rep ; 6: 25745, 2016 05 10.
Article in English | MEDLINE | ID: mdl-27160511

ABSTRACT

Sweet and umami tastes are perceived by T1r taste receptors in oral cavity. T1rs are class C G-protein coupled receptors (GPCRs), and the extracellular ligand binding domains (LBDs) of T1r1/T1r3 and T1r2/T1r3 heterodimers are responsible for binding of chemical substances eliciting umami or sweet taste. However, molecular analyses of T1r have been hampered due to the difficulties in recombinant expression and protein purification, and thus little is known about mechanisms for taste perception. Here we show the first molecular view of reception of a taste substance by a taste receptor, where the binding of the taste substance elicits a different conformational state of T1r2/T1r3 LBD heterodimer. Electron microscopy has showed a characteristic dimeric structure. Förster resonance energy transfer and X-ray solution scattering have revealed the transition of the dimerization manner of the ligand binding domains, from a widely spread to compactly organized state upon taste substance binding, which may correspond to distinct receptor functional states.


Subject(s)
Extracellular Space/chemistry , Protein Multimerization , Receptors, G-Protein-Coupled/chemistry , Receptors, G-Protein-Coupled/metabolism , Taste , Animals , Glutamine/metabolism , Ligands , Oryzias , Protein Domains , Receptors, G-Protein-Coupled/ultrastructure , Recombinant Proteins/metabolism , Scattering, Small Angle , X-Ray Diffraction
15.
Mol Cell ; 54(6): 1034-1041, 2014 Jun 19.
Article in English | MEDLINE | ID: mdl-24910098

ABSTRACT

Cell signaling depends on dynamic protein-protein interaction (PPI) networks, often assembled through modular domains each interacting with multiple peptide motifs. This complexity raises a conceptual challenge, namely to define whether a particular cellular response requires assembly of the complete PPI network of interest or can be driven by a specific interaction. To address this issue, we designed variants of the Grb2 SH2 domain ("pY-clamps") whose specificity is highly biased toward a single phosphotyrosine (pY) motif among many potential pYXNX Grb2-binding sites. Surprisingly, directing Grb2 predominantly to a single pY site of the Ptpn11/Shp2 phosphatase, but not other sites tested, was sufficient for differentiation of the essential primitive endoderm lineage from embryonic stem cells. Our data suggest that discrete connections within complex PPI networks can underpin regulation of particular biological events. We propose that this directed wiring approach will be of general utility in functionally annotating specific PPIs.


Subject(s)
Cell Differentiation , Embryonic Stem Cells/cytology , GRB2 Adaptor Protein/metabolism , Protein Interaction Maps/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism , Animals , Binding Sites/genetics , Cell Differentiation/genetics , Cell Line , Crystallography, X-Ray , Embryonic Stem Cells/metabolism , Fibroblast Growth Factor 4/metabolism , GRB2 Adaptor Protein/genetics , Mice , Models, Molecular , Protein Binding/genetics , Protein Structure, Tertiary , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 11/ultrastructure , Signal Transduction/genetics
16.
Proc Natl Acad Sci U S A ; 110(37): 14924-9, 2013 Sep 10.
Article in English | MEDLINE | ID: mdl-23980151

ABSTRACT

The dysregulated tyrosine kinase BCR-ABL causes chronic myelogenous leukemia in humans and forms a large multiprotein complex that includes the Src-homology 2 (SH2) domain-containing phosphatase 2 (SHP2). The expression of SHP2 is necessary for BCR-ABL-dependent oncogenic transformation, but the precise signaling mechanisms of SHP2 are not well understood. We have developed binding proteins, termed monobodies, for the N- and C-terminal SH2 domains of SHP2. Intracellular expression followed by interactome analysis showed that the monobodies are essentially monospecific to SHP2. Two crystal structures revealed that the monobodies occupy the phosphopeptide-binding sites of the SH2 domains and thus can serve as competitors of SH2-phosphotyrosine interactions. Surprisingly, the segments of both monobodies that bind to the peptide-binding grooves run in the opposite direction to that of canonical phosphotyrosine peptides, which may contribute to their exquisite specificity. When expressed in cells, monobodies targeting the N-SH2 domain disrupted the interaction of SHP2 with its upstream activator, the Grb2-associated binder 2 adaptor protein, suggesting decoupling of SHP2 from the BCR-ABL protein complex. Inhibition of either N-SH2 or C-SH2 was sufficient to inhibit two tyrosine phosphorylation events that are critical for SHP2 catalytic activity and to block ERK activation. In contrast, targeting the N-SH2 or C-SH2 revealed distinct roles of the two SH2 domains in downstream signaling, such as the phosphorylation of paxillin and signal transducer and activator of transcription 5. Our results delineate a hierarchy of function for the SH2 domains of SHP2 and validate monobodies as potent and specific antagonists of protein-protein interactions in cancer cells.


Subject(s)
Fusion Proteins, bcr-abl/antagonists & inhibitors , Protein Tyrosine Phosphatase, Non-Receptor Type 11/antagonists & inhibitors , Adaptor Proteins, Signal Transducing/metabolism , Amino Acid Sequence , Cell Transformation, Neoplastic , Crystallography, X-Ray , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Fusion Proteins, bcr-abl/chemistry , Fusion Proteins, bcr-abl/genetics , HEK293 Cells , Humans , K562 Cells , Models, Molecular , Peptide Library , Peptides/chemistry , Peptides/genetics , Peptides/pharmacology , Protein Binding , Protein Interaction Domains and Motifs , Protein Tyrosine Phosphatase, Non-Receptor Type 11/chemistry , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , Signal Transduction , src Homology Domains
17.
J Mol Biol ; 424(5): 391-9, 2012 Dec 14.
Article in English | MEDLINE | ID: mdl-23041298

ABSTRACT

Antibodies directed against histone posttranslational modifications (PTMs) are critical tools in epigenetics research, particularly in the widely used chromatin immunoprecipitation (ChIP) experiments. However, a lack of quantitative methods for characterizing such antibodies has been a major bottleneck in accurate and reproducible analysis of histone modifications. Here, we report a simple and sensitive method for quantitatively characterizing polyclonal and monoclonal antibodies for histone PTMs in a ChIP-like format. Importantly, it determines the apparent dissociation constants for the interactions of an antibody with peptides harboring cognate or off-target PTMs. Analyses of commercial antibodies revealed large ranges of affinity, specificity and binding capacity as well as substantial lot-to-lot variations, suggesting the importance of quantitatively characterizing each antibody intended to be used in ChIP experiments and optimizing experimental conditions accordingly. Furthermore, using this method, we identified additional factors potentially affecting the interpretation of ChIP experiments.


Subject(s)
Antibodies/immunology , Antibody Affinity , Chromatin Immunoprecipitation/methods , Histones/immunology , Antibodies/classification , Sensitivity and Specificity
18.
J Biol Chem ; 286(40): 35247-56, 2011 Oct 07.
Article in English | MEDLINE | ID: mdl-21844191

ABSTRACT

Reelin is a 3461-residue secreted glycoprotein that plays a critical role in brain development through its action on target neurons. Although it is known that functional reelin protein exists as multimer formed by interchain disulfide bond(s) as well as through non-covalent interactions, the chemical nature of the multimer assembly has been elusive. In the present study, we identified, among 122 cysteines present in full-length reelin, the single critical cysteine residue (Cys(2101)) responsible for the covalent multimerization. C2101A mutant reelin failed to assemble into disulfide-bonded multimers, whereas it still exhibited non-covalently associated high molecular weight oligomeric states in solution. Detailed analysis of tryptic fragments produced from the purified reelin proteins revealed that the minimum unit of the multimer is a homodimeric reelin linked via Cys(2101) present in the central region and that this cysteine does not connect to the N-terminal region of reelin, which had been postulated as the primary oligomerization domain. A surface plasmon resonance binding assay confirmed that C2101A mutant reelin retained binding capability toward two neuronal receptors apolipoprotein E receptor 2 and very low density lipoprotein receptor. However, it failed to show signaling activity in the assay using the cultured neurons. These results indicate that an intact higher order architecture of reelin multimer maintained by both Cys(2101)-mediated homodimerization and other non-covalent association present elsewhere in the reelin primary structure are essential for exerting its full biological activity.


Subject(s)
Cell Adhesion Molecules, Neuronal/chemistry , Extracellular Matrix Proteins/chemistry , Nerve Tissue Proteins/chemistry , Serine Endopeptidases/chemistry , Amino Acid Sequence , Animals , Brain/metabolism , CHO Cells , Cell Adhesion Molecules, Neuronal/metabolism , Cell Line , Cricetinae , Cricetulus , Dimerization , Disulfides/chemistry , Extracellular Matrix Proteins/metabolism , Humans , Mice , Molecular Sequence Data , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Phosphorylation , Protein Binding , Protein Structure, Tertiary , Reelin Protein , Sequence Homology, Amino Acid , Serine Endopeptidases/metabolism , Signal Transduction
19.
PLoS One ; 6(4): e19411, 2011 Apr 28.
Article in English | MEDLINE | ID: mdl-21552542

ABSTRACT

Neurexins (Nrxs) are presynaptic membrane proteins with a single membrane-spanning domain that mediate asymmetric trans-synaptic cell adhesion by binding to their postsynaptic receptor neuroligins. α-Nrx has a large extracellular region comprised of multiple copies of laminin, neurexin, sex-hormone-binding globulin (LNS) domains and epidermal growth factor (EGF) modules, while that of ß-Nrx has but a single LNS domain. It has long been known that the larger α-Nrx and the shorter ß-Nrx show distinct binding behaviors toward different isoforms/variants of neuroligins, although the underlying mechanism has yet to be elucidated. Here, we describe the crystal structure of a fragment corresponding to the C-terminal one-third of the Nrx1α ectodomain, consisting of LNS5-EGF3-LNS6. The 2.3 Å-resolution structure revealed the presence of a domain configuration that was rigidified by inter-domain contacts, as opposed to the more common flexible "beads-on-a-string" arrangement. Although the neuroligin-binding site on the LNS6 domain was completely exposed, the location of the α-Nrx specific LNS5-EGF3 segment proved incompatible with the loop segment inserted in the B+ neuroligin variant, which explains the variant-specific neuroligin recognition capability observed in α-Nrx. This, combined with a low-resolution molecular envelope obtained by a single particle reconstruction performed on negatively stained full-length Nrx1α sample, allowed us to derive a structural model of the α-Nrx ectodomain. This model will help us understand not only how the large α-Nrx ectodomain is accommodated in the synaptic cleft, but also how the trans-synaptic adhesion mediated by α- and ß-Nrxs could differentially affect synaptic structure and function.


Subject(s)
Cell Adhesion Molecules, Neuronal/metabolism , Glycoproteins/chemistry , Glycoproteins/metabolism , Neuropeptides/chemistry , Neuropeptides/metabolism , Amino Acid Sequence , Animals , Cattle , Extracellular Matrix/metabolism , Humans , Models, Molecular , Molecular Sequence Data , Protein Binding , Protein Isoforms/metabolism , Protein Structure, Tertiary , Substrate Specificity , Synapses/metabolism
20.
Article in English | MEDLINE | ID: mdl-21206043

ABSTRACT

Low-density lipoprotein receptor (LDLR) relative with 11 binding repeats (LR11; also known as sorLA) is genetically associated with late-onset Alzheimer's disease and is thought to be involved in neurodegenerative processes. LR11 contains a vacuolar protein-sorting 10 protein (Vps10p) domain. As this domain has been implicated in protein-protein interaction in other receptors, its structure and function are of great biological interest. Human LR11 Vps10p domain was expressed in mammalian cells and the purified protein was crystallized using the hanging-drop vapour-diffusion method. Enzymatic deglycosylation of the sample was critical to obtaining diffraction-quality crystals. Deglycosylated LR11 Vps10p-domain crystals belonged to the hexagonal space group P6(1)22. A diffraction data set was collected to 2.4 Šresolution and a clear molecular-replacement solution was obtained.


Subject(s)
LDL-Receptor Related Proteins/chemistry , Membrane Transport Proteins/chemistry , Protein Structure, Tertiary , Crystallization , Crystallography, X-Ray , Humans , LDL-Receptor Related Proteins/genetics , Membrane Transport Proteins/genetics , Molecular Sequence Data
SELECTION OF CITATIONS
SEARCH DETAIL
...