Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.930
Filter
1.
Cell ; 182(2): 357-371.e13, 2020 07 23.
Article in English | MEDLINE | ID: mdl-32610085

ABSTRACT

Excitatory neurotransmission meditated by glutamate receptors including N-methyl-D-aspartate receptors (NMDARs) is pivotal to brain development and function. NMDARs are heterotetramers composed of GluN1 and GluN2 subunits, which bind glycine and glutamate, respectively, to activate their ion channels. Despite importance in brain physiology, the precise mechanisms by which activation and inhibition occur via subunit-specific binding of agonists and antagonists remain largely unknown. Here, we show the detailed patterns of conformational changes and inter-subunit and -domain reorientation leading to agonist-gating and subunit-dependent competitive inhibition by providing multiple structures in distinct ligand states at 4 Å or better. The structures reveal that activation and competitive inhibition by both GluN1 and GluN2 antagonists occur by controlling the tension of the linker between the ligand-binding domain and the transmembrane ion channel of the GluN2 subunit. Our results provide detailed mechanistic insights into NMDAR pharmacology, activation, and inhibition, which are fundamental to the brain physiology.


Subject(s)
Receptors, N-Methyl-D-Aspartate/metabolism , Binding Sites , Binding, Competitive , Cryoelectron Microscopy , Crystallography, X-Ray , Dimerization , Glutamic Acid/chemistry , Glutamic Acid/metabolism , Glycine/chemistry , Glycine/metabolism , Humans , Ligands , Molecular Dynamics Simulation , Protein Structure, Quaternary , Protein Subunits/agonists , Protein Subunits/antagonists & inhibitors , Protein Subunits/genetics , Protein Subunits/metabolism , Receptors, N-Methyl-D-Aspartate/agonists , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/genetics , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification
2.
Nature ; 630(8017): 762-768, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38778115

ABSTRACT

Kainate receptors, a subclass of ionotropic glutamate receptors, are tetrameric ligand-gated ion channels that mediate excitatory neurotransmission1-4. Kainate receptors modulate neuronal circuits and synaptic plasticity during the development and function of the central nervous system and are implicated in various neurological and psychiatric diseases, including epilepsy, depression, schizophrenia, anxiety and autism5-11. Although structures of kainate receptor domains and subunit assemblies are available12-18, the mechanism of kainate receptor gating remains poorly understood. Here we present cryo-electron microscopy structures of the kainate receptor GluK2 in the presence of the agonist glutamate and the positive allosteric modulators lectin concanavalin A and BPAM344. Concanavalin A and BPAM344 inhibit kainate receptor desensitization and prolong activation by acting as a spacer between the amino-terminal and ligand-binding domains and a stabilizer of the ligand-binding domain dimer interface, respectively. Channel opening involves the kinking of all four pore-forming M3 helices. Our structures reveal the molecular basis of kainate receptor gating, which could guide the development of drugs for treatment of neurological disorders.


Subject(s)
Concanavalin A , Cryoelectron Microscopy , GluK2 Kainate Receptor , Glutamic Acid , Ion Channel Gating , Models, Molecular , Protein Domains , Receptors, Kainic Acid , Receptors, Kainic Acid/chemistry , Receptors, Kainic Acid/metabolism , Receptors, Kainic Acid/ultrastructure , Humans , Glutamic Acid/metabolism , Glutamic Acid/chemistry , Animals , Concanavalin A/chemistry , Concanavalin A/metabolism , Concanavalin A/pharmacology , Ligands , Allosteric Regulation , Binding Sites
3.
Nature ; 596(7871): 301-305, 2021 08.
Article in English | MEDLINE | ID: mdl-34321660

ABSTRACT

Ketamine is a non-competitive channel blocker of N-methyl-D-aspartate (NMDA) receptors1. A single sub-anaesthetic dose of ketamine produces rapid (within hours) and long-lasting antidepressant effects in patients who are resistant to other antidepressants2,3. Ketamine is a racemic mixture of S- and R-ketamine enantiomers, with S-ketamine isomer being the more active antidepressant4. Here we describe the cryo-electron microscope structures of human GluN1-GluN2A and GluN1-GluN2B NMDA receptors in complex with S-ketamine, glycine and glutamate. Both electron density maps uncovered the binding pocket for S-ketamine in the central vestibule between the channel gate and selectivity filter. Molecular dynamics simulation showed that S-ketamine moves between two distinct locations within the binding pocket. Two amino acids-leucine 642 on GluN2A (homologous to leucine 643 on GluN2B) and asparagine 616 on GluN1-were identified as key residues that form hydrophobic and hydrogen-bond interactions with ketamine, and mutations at these residues reduced the potency of ketamine in blocking NMDA receptor channel activity. These findings show structurally how ketamine binds to and acts on human NMDA receptors, and pave the way for the future development of ketamine-based antidepressants.


Subject(s)
Cryoelectron Microscopy , Ketamine/chemistry , Ketamine/pharmacology , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/ultrastructure , Antidepressive Agents/chemistry , Antidepressive Agents/metabolism , Antidepressive Agents/pharmacology , Asparagine/chemistry , Asparagine/metabolism , Binding Sites , Glutamic Acid/chemistry , Glutamic Acid/metabolism , Glutamic Acid/pharmacology , Glycine/chemistry , Glycine/metabolism , Glycine/pharmacology , Humans , Hydrogen Bonding , Hydrophobic and Hydrophilic Interactions , Ketamine/metabolism , Leucine/chemistry , Leucine/metabolism , Molecular Dynamics Simulation , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/ultrastructure , Receptors, N-Methyl-D-Aspartate/chemistry , Receptors, N-Methyl-D-Aspartate/metabolism
4.
J Am Chem Soc ; 146(9): 5823-5833, 2024 03 06.
Article in English | MEDLINE | ID: mdl-38174701

ABSTRACT

The biological significance of self-assembled protein filament networks and their unique mechanical properties have sparked interest in the development of synthetic filament networks that mimic these attributes. Building on the recent advancement of autoaccelerated ring-opening polymerization of amino acid N-carboxyanhydrides (NCAs), this study strategically explores a series of random copolymers comprising multiple amino acids, aiming to elucidate the core principles governing gelation pathways of these purpose-designed copolypeptides. Utilizing glutamate (Glu) as the primary component of copolypeptides, two targeted pathways were pursued: first, achieving a fast fibrillation rate with lower interaction potential using serine (Ser) as a comonomer, facilitating the creation of homogeneous fibril networks; and second, creating more rigid networks of fibril clusters by incorporating alanine (Ala) and valine (Val) as comonomers. The selection of amino acids played a pivotal role in steering both the morphology of fibril superstructures and their assembly kinetics, subsequently determining their potential to form sample-spanning networks. Importantly, the viscoelastic properties of the resulting supramolecular hydrogels can be tailored according to the specific copolypeptide composition through modulations in filament densities and lengths. The findings enhance our understanding of directed self-assembly in high molecular weight synthetic copolypeptides, offering valuable insights for the development of synthetic fibrous networks and biomimetic supramolecular materials with custom-designed properties.


Subject(s)
Hydrogels , Peptides , Hydrogels/chemistry , Peptides/chemistry , Amino Acids , Glutamic Acid/chemistry , Alanine/chemistry
5.
Chembiochem ; 25(9): e202300872, 2024 May 02.
Article in English | MEDLINE | ID: mdl-38376941

ABSTRACT

Dye-decolorizing peroxidases (DyPs) are recently identified microbial enzymes that have been used in several Biotechnology applications from wastewater treatment to lignin valorization. However, their properties and mechanism of action still have many open questions. Their heme-containing active site is buried by three conserved flexible loops with a putative role in modulating substrate access and enzyme catalysis. Here, we investigated the role of a conserved glutamate residue in stabilizing interactions in loop 2 of A-type DyPs. First, we did site saturation mutagenesis of this residue, replacing it with all possible amino acids in bacterial DyPs from Bacillus subtilis (BsDyP) and from Kitasatospora aureofaciens (KaDyP1), the latter being characterized here for the first time. We screened the resulting libraries of variants for activity towards ABTS and identified variants with increased catalytic efficiency. The selected variants were purified and characterized for activity and stability. We furthermore used Molecular Dynamics simulations to rationalize the increased catalytic efficiency and found that the main reason is the electron channeling becoming easier from surface-exposed tryptophans. Based on our findings, we also propose that this glutamate could work as a pH switch in the wild-type enzyme, preventing intracellular damage.


Subject(s)
Bacillus subtilis , Coloring Agents , Glutamic Acid , Peroxidases , Glutamic Acid/chemistry , Glutamic Acid/metabolism , Coloring Agents/chemistry , Coloring Agents/metabolism , Bacillus subtilis/enzymology , Peroxidases/chemistry , Peroxidases/metabolism , Peroxidases/genetics , Molecular Dynamics Simulation , Protein Engineering , Mutagenesis, Site-Directed
6.
Biomacromolecules ; 25(6): 3724-3730, 2024 Jun 10.
Article in English | MEDLINE | ID: mdl-38743032

ABSTRACT

A small series of copoly(α,l-glutamic acid/dl-allylglycine)s with the same chain length and allylglycine content (∼10 mol %) but different spatial distribution of allylglycine units was synthesized and subsequently glycosylated via thiol-ene chemistry. Dilute aqueous copolypeptide solutions (0.1 wt %, physiological saline) were analyzed by circular dichroism spectroscopy, dynamic light scattering, and cryogenic transmission electron microscopy. The copolypeptides adopted a random coil or α-helix conformation, depending on solution pH, and the glycosylated residues either distorted or enhanced the folding into an α-helix depending on their location and spatial distribution along the chain. However, regardless of their secondary structure and degree of charging, all partially glycosylated copolypeptides self-assembled into 3D spherical structures, supposedly driven by a hydrophilic effect promoting microphase separation into glucose-rich and glutamate-rich domains.


Subject(s)
Saline Solution , Saline Solution/chemistry , Glutamic Acid/chemistry , Glycosylation , Circular Dichroism , Solutions , Glycine/chemistry , Hydrogen-Ion Concentration
7.
Eur Biophys J ; 53(4): 193-203, 2024 May.
Article in English | MEDLINE | ID: mdl-38647543

ABSTRACT

Na+/H+ antiporters facilitate the exchange of Na+ for H+ across the cytoplasmic membrane in prokaryotic and eukaryotic cells. These transporters are crucial to maintain the homeostasis of sodium ions, consequently pH, and volume of the cells. Therefore, sodium/proton antiporters are considered promising therapeutic targets in humans. The Na+/H+ antiporter in Escherichia coli (Ec-NhaA), a prototype of cation-proton antiporter (CPA) family, transports two protons and one sodium (or Li+) in opposite direction. Previous mutagenesis experiments on Ec-NhaA have proposed Asp164, Asp163, and Asp133 amino acids with the significant implication in functional and structural integrity and create site for ion-binding. However, the mechanism and the sites for the binding of the two protons remain unknown and controversial which could be critical for pH regulation. In this study, we have explored the role of Glu78 in the regulation of pH by Ec-NhaA. Although we have created various mutants, E78C has shown a considerable effect on the stoichiometry of NhaA and presented comparable phenotypes. The ITC experiment has shown the binding of ~ 5 protons in response to the transport of one lithium ion. The phenotype analysis on selective medium showed a significant expression compared to WT Ec-NhaA. This represents the importance of Glu78 in transporting the H+ across the membrane where a single mutation with Cys amino acid alters the number of H+ significantly maintaining the activity of the protein.


Subject(s)
Escherichia coli Proteins , Escherichia coli , Glutamic Acid , Mutagenesis, Site-Directed , Sodium-Hydrogen Exchangers , Escherichia coli Proteins/genetics , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , Glutamic Acid/metabolism , Glutamic Acid/chemistry , Sodium-Hydrogen Exchangers/genetics , Sodium-Hydrogen Exchangers/chemistry , Sodium-Hydrogen Exchangers/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Hydrogen-Ion Concentration , Ion Exchange , Models, Molecular
8.
J Org Chem ; 89(16): 11261-11271, 2024 Aug 16.
Article in English | MEDLINE | ID: mdl-39104055

ABSTRACT

The negligible cytotoxicity of anion surface-linked dendrons makes glutamic acid-based dendrons a potential candidate for materials and biological applications. Despite the inherent drawbacks of the conventional solution phase synthesis of glutamic acid-based dendrons, there have been no advancements in these protocols. Herein, we demonstrate the first-ever convergent solid phase synthesis of dendrons, up to fourth generation, having glutamic acid branching points produced by preactivation of dicarboxylic acid groups with N-hydroxysuccinimide and simultaneous coupling with amine groups of two growing peptide chains, with excellent yields (30-70%). In addition to the general advantages, such as the easy workup, a final single purification step, and an overall short synthesis duration, the convergent solid phase synthesis allowed us to chemically synthesize glutamic acid branching-based dendrons that cannot be accessed by standard divergent solid phase synthesis. This method has also been validated for its application in synthesizing hard-to-achieve Janus peptide dendrimers in a single stretch on a solid support. Our work corroborates the efficacy of controlled -COOH activation to accomplish an atypical solid phase synthesis of diverse glutamic acid dendrons in a convergent fashion. This is the first example of a Janus peptide dendrimer being synthesized on a solid support, utilizing both convergent and divergent approaches simultaneously.


Subject(s)
Dendrimers , Glutamic Acid , Peptides , Solid-Phase Synthesis Techniques , Dendrimers/chemistry , Dendrimers/chemical synthesis , Solid-Phase Synthesis Techniques/methods , Peptides/chemistry , Peptides/chemical synthesis , Glutamic Acid/chemistry , Molecular Structure
9.
J Chem Inf Model ; 64(13): 5140-5150, 2024 Jul 08.
Article in English | MEDLINE | ID: mdl-38973304

ABSTRACT

Beta-N-methylamino-l-alanine (BMAA) is a potential neurotoxic nonprotein amino acid, which can reach the human body through the food chain. When BMAA interacts with bicarbonate in the human body, carbamate adducts are produced, which share a high structural similarity with the neurotransmitter glutamate. It is believed that BMAA and its l-carbamate adducts bind in the glutamate binding site of ionotropic glutamate receptor 2 (GluR2). Chronic exposure to BMAA and its adducts could cause neurological illness such as neurodegenerative diseases. However, the mechanism of BMAA action and its carbamate adducts bound to GluR2 has not yet been elucidated. Here, we investigate the binding modes and the affinity of BMAA and its carbamate adducts to GluR2 in comparison to the natural agonist, glutamate, to understand whether these can act as GluR2 modulators. Initially, we perform molecular dynamics simulations of BMAA and its carbamate adducts bound to GluR2 to examine the stability of the ligands in the S1/S2 ligand-binding core of the receptor. In addition, we utilize alchemical free energy calculations to compute the difference in the free energy of binding of the beta-carbamate adduct of BMAA to GluR2 compared to that of glutamate. Our findings indicate that carbamate adducts of BMAA and glutamate remain stable in the binding site of the GluR2 compared to BMAA. Additionally, alchemical free energy results reveal that glutamate and the beta-carbamate adduct of BMAA have comparable binding affinity to the GluR2. These results provide a rationale that BMAA carbamate adducts may be, in fact, the modulators of GluR2 and not BMAA itself.


Subject(s)
Amino Acids, Diamino , Carbamates , Cyanobacteria Toxins , Receptors, AMPA , Receptors, AMPA/metabolism , Receptors, AMPA/chemistry , Amino Acids, Diamino/chemistry , Amino Acids, Diamino/metabolism , Carbamates/chemistry , Carbamates/metabolism , Molecular Dynamics Simulation , Humans , Binding Sites , Protein Binding , Glutamic Acid/metabolism , Glutamic Acid/chemistry , Ligands
10.
J Chem Inf Model ; 64(10): 4168-4179, 2024 May 27.
Article in English | MEDLINE | ID: mdl-38745447

ABSTRACT

Hydroxyprolines are abundant in nature and widely utilized by many living organisms. Isomerization of trans-4-hydroxy-d-proline (t4D-HP) to generate 2-amino-4-ketopentanoate has been found to need a glycyl radical enzyme HplG, which catalyzes the cleavage of the C-N bond, while dehydration of trans-4-hydroxy-l-proline involves a homologous enzyme of HplG. Herein, molecular dynamics simulations and quantum mechanics/molecular mechanics (QM/MM) calculations are employed to understand the reaction mechanism of HplG. Two possible reaction pathways of HplG have been explored to decipher the origin of its chemoselectivity. The QM/MM calculations reveal that the isomerization proceeds via an initial hydrogen shift from the Cγ site of t4D-HP to a catalytic cysteine radical, followed by cleavage of the Cδ-N bond in t4D-HP to form a radical intermediate that captures a hydrogen atom from the cysteine. Activation of the Cδ-H bond in t4D-HP to bring about dehydration of t4D-HP possesses an extremely high energy barrier, thus rendering the dehydration pathway implausible in HplG. On the basis of the current calculations, conserved residue Glu429 plays a pivotal role in the isomerization pathway: the hydrogen bonding between it and t4D-HP weakens the hydroxyalkyl Cγ-Hγ bond, and it acts as a proton acceptor to trigger the cleavage of the C-N bond in t4D-HP. Our current QM/MM calculations rationalize the origin of the experimentally observed chemoselectivity of HplG and propose an H-bond-assisted bond activation strategy in radical-containing enzymes. These findings have general implications on radical-mediated enzymatic catalysis and expand our understanding of how nature wisely and selectively activates the C-H bond to modulate catalytic selectivity.


Subject(s)
Cysteine , Glutamic Acid , Molecular Dynamics Simulation , Quantum Theory , Cysteine/chemistry , Cysteine/metabolism , Glutamic Acid/chemistry , Glutamic Acid/metabolism , Free Radicals/chemistry , Free Radicals/metabolism , Hydrogen Bonding
11.
Proc Natl Acad Sci U S A ; 118(41)2021 10 12.
Article in English | MEDLINE | ID: mdl-34607959

ABSTRACT

EmrE is an Escherichia coli multidrug efflux pump and member of the small multidrug resistance (SMR) family that transports drugs as a homodimer by harnessing energy from the proton motive force. SMR family transporters contain a conserved glutamate residue in transmembrane 1 (Glu14 in EmrE) that is required for binding protons and drugs. Yet the mechanism underlying proton-coupled transport by the two glutamate residues in the dimer remains unresolved. Here, we used NMR spectroscopy to determine acid dissociation constants (pKa ) for wild-type EmrE and heterodimers containing one or two Glu14 residues in the dimer. For wild-type EmrE, we measured chemical shifts of the carboxyl side chain of Glu14 using solid-state NMR in lipid bilayers and obtained unambiguous evidence on the existence of asymmetric protonation states. Subsequent measurements of pKa values for heterodimers with a single Glu14 residue showed no significant differences from heterodimers with two Glu14 residues, supporting a model where the two Glu14 residues have independent pKa values and are not electrostatically coupled. These insights support a transport pathway with well-defined protonation states in each monomer of the dimer, including a preferred cytoplasmic-facing state where Glu14 is deprotonated in monomer A and protonated in monomer B under pH conditions in the cytoplasm of E. coli Our findings also lead to a model, hop-free exchange, which proposes how exchangers with conformation-dependent pKa values reduce proton leakage. This model is relevant to the SMR family and transporters comprised of inverted repeat domains.


Subject(s)
Antiporters/metabolism , Escherichia coli Proteins/metabolism , Escherichia coli/metabolism , Glutamic Acid/chemistry , Protein Domains/physiology , Anti-Bacterial Agents/metabolism , Antiporters/genetics , Drug Resistance, Multiple, Bacterial/genetics , Escherichia coli Proteins/genetics , Magnetic Resonance Spectroscopy , Protein Transport/physiology , Static Electricity
12.
Mikrochim Acta ; 191(9): 528, 2024 08 09.
Article in English | MEDLINE | ID: mdl-39120734

ABSTRACT

A dual-template molecularly imprinted electrochemical sensor was developed for the simultaneous detection of serotonin (5-HT) and glutamate (Glu). First, amino-functionalized reduced graphene oxide (NRGO) was used as the modification material of a GCE to increase its electrical conductivity and specific surface area, using Glu and 5-HT as dual-template molecules and o-phenylenediamine (OPD) with self-polymerization ability as functional monomers. Through self-assembly and electropolymerization, dual-template molecularly imprinted polymers were formed on the electrode. After removing the templates, the specific recognition binding sites were exposed. The amount of NRGO, polymerization parameters, and elution parameters were further optimized to construct a dual-template molecularly imprinted electrochemical sensor, which can specifically recognize double-target molecules Glu and 5-HT. The differential pulse voltammetry (DPV) technique was used to achieve simultaneous detection of Glu and 5-HT based on their distinct electrochemical activities under specific conditions. The sensor showed a good linear relationship for Glu and 5-HT in the range 1 ~ 100 µM, and the detection limits were 0.067 µM and 0.047 µM (S/N = 3), respectively. The sensor has good reproducibility, repeatability, and selectivity. It was successfully utilized to simultaneously detect Glu and 5-HT in mouse serum, offering a more dependable foundation for objectively diagnosing and early warning of depression. Additionally, the double signal sensing strategy also provides a new approach for the simultaneous detection of both electroactive and non-electroactive substances.


Subject(s)
Electrochemical Techniques , Glutamic Acid , Graphite , Limit of Detection , Molecular Imprinting , Phenylenediamines , Serotonin , Serotonin/blood , Serotonin/analysis , Electrochemical Techniques/methods , Electrochemical Techniques/instrumentation , Animals , Glutamic Acid/analysis , Glutamic Acid/blood , Glutamic Acid/chemistry , Graphite/chemistry , Mice , Phenylenediamines/chemistry , Depression/diagnosis , Depression/blood , Electrodes , Biomarkers/blood , Biomarkers/analysis , Reproducibility of Results
13.
Biophys J ; 122(6): 1068-1085, 2023 03 21.
Article in English | MEDLINE | ID: mdl-36698313

ABSTRACT

The triple glutamine (Q) mutant (QQQ) structure of a Cl-/H+ antiporter from Escherichia coli (ClC-ec1) displaying a novel backbone arrangement has been used to challenge the long-held notion that Cl-/H+ antiporters do not operate through large conformational motions. The QQQ mutant substitutes the glutamine residue for an external glutamate E148, an internal glutamate E203, and a third glutamate E113 that hydrogen-bonds with E203. However, it is unknown if QQQ represents a physiologically relevant state, as well as how the protonation of the wild-type glutamates relates to the global dynamics. We herein apply continuous constant-pH molecular dynamics to investigate the H+-coupled dynamics of ClC-ec1. Although any large-scale conformational rearrangement upon acidification would be due to the accumulation of excess charge within the protein, protonation of the glutamates significantly impacts mainly the local structure and dynamics. Despite the fact that the extracellular pore enlarges at acidic pHs, an occluded ClC-ec1 within the active pH range of 3.5-7.5 requires a protonated E148 to facilitate extracellular Cl- release. E203 is also involved in the intracellular H+ transfer as an H+ acceptor. The water wire connection of E148 with the intracellular solution is regulated by the charge states of the E113/E203 dyad with coupled proton titration. However, the dynamics extracted from our simulations are not QQQ-like, indicating that the QQQ mutant does not represent the behavior of the wild-type ClC-ec1. These findings reinforce the necessity of having a protonatable residue at the E203 position in ClC-ec1 and suggest that a higher level of complexity exists for the intracellular H+ transfer in Cl-/H+ antiporters.


Subject(s)
Antiporters , Escherichia coli Proteins , Antiporters/genetics , Antiporters/metabolism , Glutamic Acid/chemistry , Glutamine , Chlorides/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Protons , Chloride Channels/metabolism , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism
14.
J Biol Chem ; 298(10): 102441, 2022 10.
Article in English | MEDLINE | ID: mdl-36055404

ABSTRACT

Vibrio cholerae cytolysin (VCC) is a potent membrane-damaging ß-barrel pore-forming toxin. Upon binding to the target membranes, VCC monomers first assemble into oligomeric prepore intermediates and subsequently transform into transmembrane ß-barrel pores. VCC harbors a designated pore-forming motif, which, during oligomeric pore formation, inserts into the membrane and generates a transmembrane ß-barrel scaffold. It remains an enigma how the molecular architecture of the pore-forming motif regulates the VCC pore-formation mechanism. Here, we show that a specific pore-forming motif residue, E289, plays crucial regulatory roles in the pore-formation mechanism of VCC. We find that the mutation of E289A drastically compromises pore-forming activity, without affecting the structural integrity and membrane-binding potential of the toxin monomers. Although our single-particle cryo-EM analysis reveals WT-like oligomeric ß-barrel pore formation by E289A-VCC in the membrane, we demonstrate that the mutant shows severely delayed kinetics in terms of pore-forming ability that can be rescued with elevated temperature conditions. We find that the pore-formation efficacy of E289A-VCC appears to be more profoundly dependent on temperature than that of the WT toxin. Our results suggest that the E289A mutation traps membrane-bound toxin molecules in the prepore-like intermediate state that is hindered from converting into the functional ß-barrel pores by a large energy barrier, thus highlighting the importance of this residue for the pore-formation mechanism of VCC.


Subject(s)
Bacterial Proteins , Cytotoxins , Pore Forming Cytotoxic Proteins , Vibrio cholerae , Virulence Factors , Cell Membrane/metabolism , Cytotoxins/chemistry , Cytotoxins/genetics , Vibrio cholerae/pathogenicity , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Virulence Factors/chemistry , Virulence Factors/genetics , Pore Forming Cytotoxic Proteins/chemistry , Pore Forming Cytotoxic Proteins/genetics , Amino Acid Motifs , Mutation , Glutamic Acid/chemistry , Glutamic Acid/genetics
15.
Nat Chem Biol ; 17(3): 291-297, 2021 03.
Article in English | MEDLINE | ID: mdl-33398167

ABSTRACT

G protein-coupled receptors (GPCRs) relay information across cell membranes through conformational coupling between the ligand-binding domain and cytoplasmic signaling domain. In dimeric class C GPCRs, the mechanism of this process, which involves propagation of local ligand-induced conformational changes over 12 nm through three distinct structural domains, is unknown. Here, we used single-molecule FRET and live-cell imaging and found that metabotropic glutamate receptor 2 (mGluR2) interconverts between four conformational states, two of which were previously unknown, and activation proceeds through the conformational selection mechanism. Furthermore, the conformation of the ligand-binding domains and downstream domains are weakly coupled. We show that the intermediate states act as conformational checkpoints for activation and control allosteric modulation of signaling. Our results demonstrate a mechanism for activation of mGluRs where ligand binding controls the proximity of signaling domains, analogous to some receptor kinases. This design principle may be generalizable to other biological allosteric sensors.


Subject(s)
Glutamic Acid/chemistry , Receptors, Metabotropic Glutamate/chemistry , Allosteric Regulation , Amino Acids/pharmacology , Binding Sites , Biosensing Techniques , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Cyclopropanes/pharmacology , Fluorescence Resonance Energy Transfer , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Glutamic Acid/pharmacology , Glycine/analogs & derivatives , Glycine/pharmacology , HEK293 Cells , Humans , Ligands , Models, Molecular , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Protein Multimerization/drug effects , Receptors, Metabotropic Glutamate/agonists , Receptors, Metabotropic Glutamate/genetics , Receptors, Metabotropic Glutamate/metabolism , Single Molecule Imaging
16.
Nature ; 542(7639): 60-65, 2017 02 02.
Article in English | MEDLINE | ID: mdl-28099415

ABSTRACT

Cyclic-nucleotide-gated channels are essential for vision and olfaction. They belong to the voltage-gated ion channel superfamily but their activities are controlled by intracellular cyclic nucleotides instead of transmembrane voltage. Here we report a 3.5-Å-resolution single-particle electron cryo-microscopy structure of a cyclic-nucleotide-gated channel from Caenorhabditis elegans in the cyclic guanosine monophosphate (cGMP)-bound open state. The channel has an unusual voltage-sensor-like domain, accounting for its deficient voltage dependence. A carboxy-terminal linker connecting S6 and the cyclic-nucleotide-binding domain interacts directly with both the voltage-sensor-like domain and the pore domain, forming a gating ring that couples conformational changes triggered by cyclic nucleotide binding to the gate. The selectivity filter is lined by the carboxylate side chains of a functionally important glutamate and three rings of backbone carbonyls. This structure provides a new framework for understanding mechanisms of ion permeation, gating and channelopathy of cyclic-nucleotide-gated channels and cyclic nucleotide modulation of related channels.


Subject(s)
Caenorhabditis elegans Proteins/chemistry , Caenorhabditis elegans Proteins/ultrastructure , Caenorhabditis elegans , Cryoelectron Microscopy , Cyclic Nucleotide-Gated Cation Channels/chemistry , Cyclic Nucleotide-Gated Cation Channels/ultrastructure , Ion Channels/chemistry , Ion Channels/ultrastructure , Amino Acid Sequence , Animals , Caenorhabditis elegans/chemistry , Caenorhabditis elegans/ultrastructure , Caenorhabditis elegans Proteins/metabolism , Cyclic GMP/metabolism , Cyclic Nucleotide-Gated Cation Channels/metabolism , Electric Conductivity , Glutamic Acid/chemistry , Glutamic Acid/metabolism , Ion Channel Gating , Ion Channels/metabolism , Models, Biological , Models, Molecular , Protein Domains
17.
Biochem J ; 479(2): 207-223, 2022 01 28.
Article in English | MEDLINE | ID: mdl-34935873

ABSTRACT

Bacteriophage endolysins degrade peptidoglycan and have been identified as antibacterial candidates to combat antimicrobial resistance. Considering the catalytic and structural diversity of endolysins, there is a paucity of structural data to inform how these enzymes work at the molecular level - key data that is needed to realize the potential of endolysin-based antibacterial agents. Here, we determine the atomic structure and define the enzymatic function of Escherichia coli O157:H7 phage FTEBc1 endolysin, LysT84. Bioinformatic analysis reveals that LysT84 is a modular endolysin, which is unusual for Gram-negative endolysins, comprising a peptidoglycan binding domain and an enzymatic domain. The crystal structure of LysT84 (2.99 Å) revealed a mostly α-helical protein with two domains connected by a linker region but packed together. LysT84 was determined to be a monomer in solution using analytical ultracentrifugation. Small-angle X-ray scattering data revealed that LysT84 is a flexible protein but does not have the expected bimodal P(r) function of a multidomain protein, suggesting that the domains of LysT84 pack closely creating a globular protein as seen in the crystal structure. Structural analysis reveals two key glutamate residues positioned on either side of the active site cavity; mutagenesis demonstrating these residues are critical for peptidoglycan degradation. Molecular dynamic simulations suggest that the enzymatically active domain is dynamic, allowing the appropriate positioning of these catalytic residues for hydrolysis of the ß(1-4) bond. Overall, our study defines the structural basis for peptidoglycan degradation by LysT84 which supports rational engineering of related endolysins into effective antibacterial agents.


Subject(s)
Anti-Bacterial Agents/chemistry , Bacteriophages/enzymology , Endopeptidases/chemistry , Escherichia coli O157/virology , Viral Proteins/chemistry , Anti-Bacterial Agents/metabolism , Biocatalysis , Catalytic Domain , Cell Wall/metabolism , Computational Biology/methods , Crystallization , Endopeptidases/metabolism , Glutamic Acid/chemistry , Hydrolysis , Molecular Dynamics Simulation , Peptidoglycan/metabolism , Protein Conformation, alpha-Helical , Protein Domains , Viral Proteins/metabolism
18.
Proc Natl Acad Sci U S A ; 117(7): 3839-3847, 2020 02 18.
Article in English | MEDLINE | ID: mdl-32015122

ABSTRACT

Allostery can be manifested as a combination of repression and activation in multidomain proteins allowing for fine tuning of regulatory mechanisms. Here we have used single molecule fluorescence resonance energy transfer (smFRET) and molecular dynamics simulations to study the mechanism of allostery underlying negative cooperativity between the two agonists glutamate and glycine in the NMDA receptor. These data show that binding of one agonist leads to conformational flexibility and an increase in conformational spread at the second agonist site. Mutational and cross-linking studies show that the dimer-dimer interface at the agonist-binding domain mediates the allostery underlying the negative cooperativity. smFRET on the transmembrane segments shows that they are tightly coupled in the unliganded and single agonist-bound form and only upon binding both agonists the transmembrane domain explores looser packing which would facilitate activation.


Subject(s)
Receptors, N-Methyl-D-Aspartate/chemistry , Receptors, N-Methyl-D-Aspartate/metabolism , Allosteric Regulation , Animals , Binding Sites , Dimerization , Glutamic Acid/chemistry , Glutamic Acid/metabolism , Glycine/chemistry , Glycine/metabolism , Molecular Dynamics Simulation , Protein Binding , Protein Conformation , Protein Domains , Rats , Receptors, N-Methyl-D-Aspartate/genetics
19.
J Biol Chem ; 296: 100284, 2021.
Article in English | MEDLINE | ID: mdl-33450226

ABSTRACT

ETV6 is an E26 transformation specific family transcriptional repressor that self-associates by its PNT domain to facilitate cooperative DNA binding. Chromosomal translocations frequently generate constitutively active oncoproteins with the ETV6 PNT domain fused to the kinase domain of one of many protein tyrosine kinases. Although an attractive target for therapeutic intervention, the propensity of the ETV6 PNT domain to polymerize via the tight head-to-tail association of two relatively flat interfaces makes it challenging to identify suitable small molecule inhibitors of this protein-protein interaction. Herein, we provide a comprehensive biophysical characterization of the ETV6 PNT domain interaction interfaces to aid future drug discovery efforts and help define the mechanisms by which its self-association mediates transcriptional repression. Using NMR spectroscopy, X-ray crystallography, and molecular dynamics simulations, along with amide hydrogen exchange measurements, we demonstrate that monomeric PNT domain variants adopt very stable helical bundle folds that do not change in conformation upon self-association into heterodimer models of the ETV6 polymer. Surface plasmon resonance-monitored alanine scanning mutagenesis studies identified hot spot regions within the self-association interfaces. These regions include both central hydrophobic residues and flanking salt-bridging residues. Collectively, these studies indicate that small molecules targeted to these hydrophobic or charged regions within the relatively rigid interfaces could potentially serve as orthosteric inhibitors of ETV6 PNT domain polymerization.


Subject(s)
Alanine/chemistry , Aspartic Acid/chemistry , Glutamic Acid/chemistry , Proto-Oncogene Proteins c-ets/chemistry , Repressor Proteins/chemistry , Transcription, Genetic , Valine/chemistry , Alanine/metabolism , Amino Acid Substitution , Aspartic Acid/metabolism , Binding Sites , Cloning, Molecular , Crystallography, X-Ray , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Glutamic Acid/metabolism , Humans , Models, Molecular , Mutagenesis, Site-Directed , Mutation , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Protein Multimerization , Proto-Oncogene Proteins c-ets/genetics , Proto-Oncogene Proteins c-ets/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Thermodynamics , Valine/metabolism , ETS Translocation Variant 6 Protein
20.
J Am Chem Soc ; 144(16): 7208-7214, 2022 04 27.
Article in English | MEDLINE | ID: mdl-35426309

ABSTRACT

Ribonucleotide reductase (RNR) catalyzes the reduction of ribonucleotides to deoxyribonucleotides and is critical for DNA synthesis and repair in all organisms. Its mechanism requires radical transfer along a ∼32 Špathway through a series of proton-coupled electron transfer (PCET) steps. Previous simulations suggested that a glutamate residue (E623) mediates the PCET reaction between two stacked tyrosine residues (Y730 and Y731) through a proton relay mechanism. This work focuses on the adjacent PCET reaction between Y730 and a cysteine residue (C439). Quantum mechanical/molecular mechanical free energy simulations illustrate that when Y730 and Y731 are stacked, E623 stabilizes the radical on C439 through hydrogen bonding with the Y730 hydroxyl group. When Y731 is flipped away from Y730, a water molecule stabilizes the radical on C439 through hydrogen bonding with Y730 and lowers the free energy barrier for radical transfer from Y730 to C439 through electrostatic interactions with the transferring hydrogen but does not directly accept the proton. These simulations indicate that the conformational motions and electrostatic interactions of the tyrosines, cysteine, glutamate, and water strongly impact the thermodynamics and kinetics of these two coupled PCET reactions. Such insights are important for protein engineering efforts aimed at altering radical transfer in RNR.


Subject(s)
Ribonucleotide Reductases , Cysteine/metabolism , Electron Transport , Electrons , Escherichia coli/metabolism , Free Radicals/chemistry , Glutamic Acid/chemistry , Models, Molecular , Protons , Ribonucleotide Reductases/chemistry , Tyrosine/chemistry , Water
SELECTION OF CITATIONS
SEARCH DETAIL