Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Cell ; 165(3): 704-14, 2016 Apr 21.
Article in English | MEDLINE | ID: mdl-27062927

ABSTRACT

N-methyl-D-aspartate receptors (NMDARs) are glutamate-gated, calcium-permeable ion channels that mediate synaptic transmission and underpin learning and memory. NMDAR dysfunction is directly implicated in diseases ranging from seizure to ischemia. Despite its fundamental importance, little is known about how the NMDAR transitions between inactive and active states and how small molecules inhibit or activate ion channel gating. Here, we report electron cryo-microscopy structures of the GluN1-GluN2B NMDA receptor in an ensemble of competitive antagonist-bound states, an agonist-bound form, and a state bound with agonists and the allosteric inhibitor Ro25-6981. Together with double electron-electron resonance experiments, we show how competitive antagonists rupture the ligand binding domain (LBD) gating "ring," how agonists retain the ring in a dimer-of-dimers configuration, and how allosteric inhibitors, acting within the amino terminal domain, further stabilize the LBD layer. These studies illuminate how the LBD gating ring is fundamental to signal transduction and gating in NMDARs.


Subject(s)
Receptors, N-Methyl-D-Aspartate/chemistry , Xenopus Proteins/chemistry , Animals , Cryoelectron Microscopy , Electron Spin Resonance Spectroscopy , Models, Molecular , Protein Domains , Protein Subunits/chemistry , Receptors, N-Methyl-D-Aspartate/agonists , Xenopus laevis
2.
Cell ; 156(4): 717-29, 2014 Feb 13.
Article in English | MEDLINE | ID: mdl-24507937

ABSTRACT

Acid-sensing ion channels (ASICs) detect extracellular protons produced during inflammation or ischemic injury and belong to the superfamily of degenerin/epithelial sodium channels. Here, we determine the cocrystal structure of chicken ASIC1a with MitTx, a pain-inducing toxin from the Texas coral snake, to define the structure of the open state of ASIC1a. In the MitTx-bound open state and in the previously determined low-pH desensitized state, TM2 is a discontinuous α helix in which the Gly-Ala-Ser selectivity filter adopts an extended, belt-like conformation, swapping the cytoplasmic one-third of TM2 with an adjacent subunit. Gly 443 residues of the selectivity filter provide a ring of three carbonyl oxygen atoms with a radius of ∼3.6 Å, presenting an energetic barrier for hydrated ions. The ASIC1a-MitTx complex illuminates the mechanism of MitTx action, defines the structure of the selectivity filter of voltage-independent, sodium-selective ion channels, and captures the open state of an ASIC.


Subject(s)
Acid Sensing Ion Channels/chemistry , Avian Proteins/chemistry , Chickens , Elapid Venoms/chemistry , Elapidae , Acid Sensing Ion Channels/metabolism , Amino Acid Sequence , Animals , Avian Proteins/metabolism , Crystallography, X-Ray , Elapid Venoms/metabolism , Models, Molecular , Molecular Sequence Data , Sequence Alignment , Sodium Channels/chemistry
3.
Nature ; 610(7933): 796-803, 2022 10.
Article in English | MEDLINE | ID: mdl-36224384

ABSTRACT

The initial step in the sensory transduction pathway underpinning hearing and balance in mammals involves the conversion of force into the gating of a mechanosensory transduction channel1. Despite the profound socioeconomic impacts of hearing disorders and the fundamental biological significance of understanding mechanosensory transduction, the composition, structure and mechanism of the mechanosensory transduction complex have remained poorly characterized. Here we report the single-particle cryo-electron microscopy structure of the native transmembrane channel-like protein 1 (TMC-1) mechanosensory transduction complex isolated from Caenorhabditis elegans. The two-fold symmetric complex is composed of two copies each of the pore-forming TMC-1 subunit, the calcium-binding protein CALM-1 and the transmembrane inner ear protein TMIE. CALM-1 makes extensive contacts with the cytoplasmic face of the TMC-1 subunits, whereas the single-pass TMIE subunits reside on the periphery of the complex, poised like the handles of an accordion. A subset of complexes additionally includes a single arrestin-like protein, arrestin domain protein (ARRD-6), bound to a CALM-1 subunit. Single-particle reconstructions and molecular dynamics simulations show how the mechanosensory transduction complex deforms the membrane bilayer and suggest crucial roles for lipid-protein interactions in the mechanism by which mechanical force is transduced to ion channel gating.


Subject(s)
Caenorhabditis elegans , Cryoelectron Microscopy , Ion Channels , Mechanotransduction, Cellular , Animals , Arrestins/chemistry , Arrestins/metabolism , Arrestins/ultrastructure , Caenorhabditis elegans/chemistry , Caenorhabditis elegans/ultrastructure , Caenorhabditis elegans Proteins/chemistry , Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans Proteins/ultrastructure , Calcium-Binding Proteins/chemistry , Calcium-Binding Proteins/metabolism , Calcium-Binding Proteins/ultrastructure , Ion Channel Gating , Ion Channels/chemistry , Ion Channels/metabolism , Ion Channels/ultrastructure , Lipids
4.
Proc Natl Acad Sci U S A ; 121(8): e2314096121, 2024 Feb 20.
Article in English | MEDLINE | ID: mdl-38354260

ABSTRACT

Mechanotransduction is the process by which a mechanical force, such as touch, is converted into an electrical signal. Transmembrane channel-like (TMC) proteins are an evolutionarily conserved family of membrane proteins whose function has been linked to a variety of mechanosensory processes, including hearing and balance sensation in vertebrates and locomotion in Drosophila. TMC1 and TMC2 are components of ion channel complexes, but the molecular features that tune these complexes to diverse mechanical stimuli are unknown. Caenorhabditis elegans express two TMC homologs, TMC-1 and TMC-2, both of which are the likely pore-forming subunits of mechanosensitive ion channels but differ in their expression pattern and functional role in the worm. Here, we present the single-particle cryo-electron microscopy structure of the native TMC-2 complex isolated from C. elegans. The complex is composed of two copies of the pore-forming TMC-2 subunit, the calcium and integrin binding protein CALM-1 and the transmembrane inner ear protein TMIE. Comparison of the TMC-2 complex to the recently published cryo-EM structure of the C. elegans TMC-1 complex highlights conserved protein-lipid interactions, as well as a π-helical structural motif in the pore-forming helices, that together suggest a mechanism for TMC-mediated mechanosensory transduction.


Subject(s)
Caenorhabditis elegans Proteins , Mechanotransduction, Cellular , Animals , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Cryoelectron Microscopy , Ion Channels/metabolism , Lipids , Mechanotransduction, Cellular/physiology , Membrane Proteins/metabolism
5.
Nature ; 511(7508): 191-7, 2014 Jul 10.
Article in English | MEDLINE | ID: mdl-25008524

ABSTRACT

N-methyl-d-aspartate (NMDA) receptors are Hebbian-like coincidence detectors, requiring binding of glycine and glutamate in combination with the relief of voltage-dependent magnesium block to open an ion conductive pore across the membrane bilayer. Despite the importance of the NMDA receptor in the development and function of the brain, a molecular structure of an intact receptor has remained elusive. Here we present X-ray crystal structures of the Xenopus laevis GluN1-GluN2B NMDA receptor with the allosteric inhibitor, Ro25-6981, partial agonists and the ion channel blocker, MK-801. Receptor subunits are arranged in a 1-2-1-2 fashion, demonstrating extensive interactions between the amino-terminal and ligand-binding domains. The transmembrane domains harbour a closed-blocked ion channel, a pyramidal central vestibule lined by residues implicated in binding ion channel blockers and magnesium, and a ∼twofold symmetric arrangement of ion channel pore loops. These structures provide new insights into the architecture, allosteric coupling and ion channel function of NMDA receptors.


Subject(s)
Models, Molecular , Receptors, N-Methyl-D-Aspartate/chemistry , Xenopus laevis/physiology , Animals , Dizocilpine Maleate/chemistry , Ion Channels/chemistry , Ligands , Phenols , Piperidines/chemistry , Protein Binding , Protein Structure, Tertiary , Protein Subunits/chemistry
6.
Nature ; 503(7474): 141-5, 2013 Nov 07.
Article in English | MEDLINE | ID: mdl-24121440

ABSTRACT

The biogenic amine transporters (BATs) regulate endogenous neurotransmitter concentrations and are targets for a broad range of therapeutic agents including selective serotonin reuptake inhibitors (SSRIs), serotonin-noradrenaline reuptake inhibitors (SNRIs) and tricyclic antidepressants (TCAs). Because eukaryotic BATs are recalcitrant to crystallographic analysis, our understanding of the mechanism of these inhibitors and antidepressants is limited. LeuT is a bacterial homologue of BATs and has proven to be a valuable paradigm for understanding relationships between their structure and function. However, because only approximately 25% of the amino acid sequence of LeuT is in common with that of BATs, and as LeuT is a promiscuous amino acid transporter, it does not recapitulate the pharmacological properties of BATs. Indeed, SSRIs and TCAs bind in the extracellular vestibule of LeuT and act as non-competitive inhibitors of transport. By contrast, multiple studies demonstrate that both TCAs and SSRIs are competitive inhibitors for eukaryotic BATs and bind to the primary binding pocket. Here we engineered LeuT to harbour human BAT-like pharmacology by mutating key residues around the primary binding pocket. The final LeuBAT mutant binds the SSRI sertraline with a binding constant of 18 nM and displays high-affinity binding to a range of SSRIs, SNRIs and a TCA. We determined 12 crystal structures of LeuBAT in complex with four classes of antidepressants. The chemically diverse inhibitors have a remarkably similar mode of binding in which they straddle transmembrane helix (TM) 3, wedge between TM3/TM8 and TM1/TM6, and lock the transporter in a sodium- and chloride-bound outward-facing open conformation. Together, these studies define common and simple principles for the action of SSRIs, SNRIs and TCAs on BATs.


Subject(s)
Antidepressive Agents, Second-Generation/pharmacology , Antidepressive Agents, Tricyclic/pharmacology , Biogenic Amines/metabolism , Plasma Membrane Neurotransmitter Transport Proteins , Recombinant Fusion Proteins/chemistry , Selective Serotonin Reuptake Inhibitors/pharmacology , Serotonin Plasma Membrane Transport Proteins/chemistry , Serotonin Plasma Membrane Transport Proteins/metabolism , Antidepressive Agents, Second-Generation/metabolism , Antidepressive Agents, Tricyclic/metabolism , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Binding, Competitive/drug effects , Chlorides/metabolism , Crystallography, X-Ray , Humans , Mazindol/metabolism , Mazindol/pharmacology , Models, Molecular , Mutation , Norepinephrine/metabolism , Plasma Membrane Neurotransmitter Transport Proteins/antagonists & inhibitors , Plasma Membrane Neurotransmitter Transport Proteins/chemistry , Plasma Membrane Neurotransmitter Transport Proteins/genetics , Plasma Membrane Neurotransmitter Transport Proteins/metabolism , Protein Conformation/drug effects , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Reproducibility of Results , Serotonin Plasma Membrane Transport Proteins/genetics , Selective Serotonin Reuptake Inhibitors/metabolism , Sertraline/metabolism , Sertraline/pharmacology , Sodium/metabolism , Structure-Activity Relationship
8.
bioRxiv ; 2023 Aug 17.
Article in English | MEDLINE | ID: mdl-37645790

ABSTRACT

Mechanotransduction is the process by which a mechanical force, such as touch, is converted into an electrical signal. Transmembrane channel-like (TMC) proteins are an evolutionarily-conserved family of ion channels whose function has been linked to a variety of mechanosensory processes, including hearing and balance sensation in vertebrates and locomotion in Drosophila. The molecular features that tune homologous TMC ion channel complexes to diverse mechanical stimuli are unknown. Caenorhabditis elegans express two TMC homologs, TMC-1 and TMC-2, both of which are the likely pore-forming subunits of mechanosensitive ion channels but differ in their expression pattern and functional role in the worm. Here we present the single particle cryo-electron microscopy structure of the native TMC-2 complex isolated from C. elegans. The complex is composed of two copies each of the pore-forming TMC-2 subunit, the calcium and integrin binding protein CALM-1 and the transmembrane inner ear protein TMIE. Comparison of the TMC-2 complex to the recently published cryo-EM structure of the C. elegans TMC-1 complex reveals differences in subunit composition and highlights conserved protein-lipid interactions, as well as other structural features, that together suggest a mechanism for TMC-mediated mechanosensory transduction.

9.
Nat Protoc ; 18(9): 2699-2716, 2023 09.
Article in English | MEDLINE | ID: mdl-37495753

ABSTRACT

Purification of membrane proteins for biochemical and structural studies is commonly achieved by recombinant overexpression in heterologous cell lines. However, many membrane proteins do not form a functional complex in a heterologous system, and few methods exist to purify sufficient protein from a native source for use in biochemical, biophysical and structural studies. Here, we provide a detailed protocol for the isolation of membrane protein complexes from transgenic Caenorhabditis elegans. We describe how to grow a genetically modified C. elegans line in abundance using standard laboratory equipment, and how to optimize purification conditions on a small scale using fluorescence-detection size-exclusion chromatography. Optimized conditions can then be applied to a large-scale preparation, enabling the purification of adequate quantities of a target protein for structural, biochemical and biophysical studies. Large-scale worm growth can be accomplished in ~9 d, and each optimization experiment can be completed in less than 1 d. We have used these methods to isolate the transmembrane channel-like protein 1 complex, as well as three additional protein complexes (transmembrane-like channel 2, lipid transfer protein and 'Protein S'), from transgenic C. elegans, demonstrating the utility of this approach in purifying challenging, low-abundance membrane protein complexes.


Subject(s)
Caenorhabditis elegans Proteins , Caenorhabditis elegans , Animals , Animals, Genetically Modified , Membrane Proteins/genetics , Membrane Proteins/metabolism , Chromatography, Gel , Cell Line , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism
10.
bioRxiv ; 2023 Dec 12.
Article in English | MEDLINE | ID: mdl-38168376

ABSTRACT

Hearing and balance rely on the conversion of a mechanical stimulus into an electrical signal, a process known as mechanosensory transduction (MT). In vertebrates, this process is accomplished by an MT complex that is located in hair cells of the inner ear. While the past three decades of research have identified many subunits that are important for MT and revealed interactions between these subunits, the composition and organization of a functional complex remains unknown. The major challenge associated with studying the MT complex is its extremely low abundance in hair cells; current estimates of MT complex quantity range from 3-60 attomoles per cochlea or utricle, well below the detection limit of most biochemical assays that are used to characterize macromolecular complexes. Here we describe the optimization of two single molecule assays, single molecule pull-down (SiMPull) and single molecule array (SiMoA), to study the composition and quantity of native mouse MT complexes. We demonstrate that these assays are capable of detecting and quantifying low attomoles of the native MT subunits protocadherin-15 (PCDH15) and lipoma HMGIC fusion partner-like protein 5 (LHFPL5). Our results illuminate the stoichiometry of PCDH15- and LHFPL5-containing complexes and establish SiMPull and SiMoA as productive methods for probing the abundance, composition, and arrangement of subunits in the native MT complex.

11.
Elife ; 102021 12 29.
Article in English | MEDLINE | ID: mdl-34964715

ABSTRACT

Mechanosensory transduction (MT), the conversion of mechanical stimuli into electrical signals, underpins hearing and balance and is carried out within hair cells in the inner ear. Hair cells harbor actin-filled stereocilia, arranged in rows of descending heights, where the tips of stereocilia are connected to their taller neighbors by a filament composed of protocadherin 15 (PCDH15) and cadherin 23 (CDH23), deemed the 'tip link.' Tension exerted on the tip link opens an ion channel at the tip of the shorter stereocilia, thus converting mechanical force into an electrical signal. While biochemical and structural studies have provided insights into the molecular composition and structure of isolated portions of the tip link, the architecture, location, and conformational states of intact tip links, on stereocilia, remains unknown. Here, we report in situ cryo-electron microscopy imaging of the tip link in mouse stereocilia. We observe individual PCDH15 molecules at the tip and shaft of stereocilia and determine their stoichiometry, conformational heterogeneity, and their complexes with other filamentous proteins, perhaps including CDH23. The PCDH15 complexes occur in clusters, frequently with more than one copy of PCDH15 at the tip of stereocilia, suggesting that tip links might consist of more than one copy of PCDH15 complexes and, by extension, might include multiple MT complexes.


Subject(s)
Cadherin Related Proteins/chemistry , Protein Precursors/chemistry , Stereocilia/ultrastructure , Animals , Cadherin Related Proteins/ultrastructure , Cryoelectron Microscopy , Mice , Molecular Conformation , Molecular Structure , Protein Precursors/ultrastructure
12.
Sci Transl Med ; 11(500)2019 07 10.
Article in English | MEDLINE | ID: mdl-31292262

ABSTRACT

Autoimmunity to membrane proteins in the central nervous system has been increasingly recognized as a cause of neuropsychiatric disease. A key recent development was the discovery of autoantibodies to N-methyl-d-aspartate (NMDA) receptors in some cases of encephalitis, characterized by cognitive changes, memory loss, and seizures that could lead to long-term morbidity or mortality. Treatment approaches and experimental studies have largely focused on the pathogenic role of these autoantibodies. Passive antibody transfer to mice has provided useful insights but does not produce the full spectrum of the human disease. Here, we describe a de novo autoimmune mouse model of anti-NMDA receptor encephalitis. Active immunization of immunocompetent mice with conformationally stabilized, native-like NMDA receptors induced a fulminant encephalitis, consistent with the behavioral and pathologic characteristics of human cases. Our results provide evidence for neuroinflammation and immune cell infiltration as components of the autoimmune response in mice. Use of transgenic mice indicated that mature T cells and antibody-producing cells were required for disease induction. This active immunization model may provide insights into disease induction and a platform for testing therapeutic approaches.


Subject(s)
Encephalitis/immunology , Hashimoto Disease/immunology , Receptors, N-Methyl-D-Aspartate/chemistry , Receptors, N-Methyl-D-Aspartate/immunology , Vaccination/adverse effects , Animals , Autoantibodies/blood , Autoantibodies/immunology , B-Lymphocytes/immunology , Behavior, Animal , Brain/pathology , Encephalitis/blood , Encephalitis/pathology , HEK293 Cells , Hashimoto Disease/blood , Hashimoto Disease/pathology , Humans , Immunoglobulin G/blood , Inflammation/pathology , Leukocytes/pathology , Mice , Neuroglia/metabolism , Neurons/metabolism , Protein Conformation , Proteolipids/metabolism , Rats , T-Lymphocytes/immunology
13.
Elife ; 72018 08 02.
Article in English | MEDLINE | ID: mdl-30070639

ABSTRACT

Hearing and balance involve the transduction of mechanical stimuli into electrical signals by deflection of bundles of stereocilia linked together by protocadherin 15 (PCDH15) and cadherin 23 'tip links'. PCDH15 transduces tip link tension into opening of a mechano-electrical transduction (MET) ion channel. PCDH15 also interacts with LHFPL5, a candidate subunit of the MET channel. Here we illuminate the PCDH15-LHFPL5 structure, showing how the complex is composed of PCDH15 and LHFPL5 subunit pairs related by a 2-fold axis. The extracellular cadherin domains define a mobile tether coupled to a rigid, 2-fold symmetric 'collar' proximal to the membrane bilayer. LHFPL5 forms extensive interactions with the PCDH15 transmembrane helices and stabilizes the overall PCDH15-LHFPL5 assembly. Our studies illuminate the architecture of the PCDH15-LHFPL5 complex, localize mutations associated with deafness, and shed new light on how forces in the PCDH15 tether may be transduced into the stereocilia membrane.


Subject(s)
Cadherins/chemistry , Cadherins/metabolism , Membrane Proteins/metabolism , Protein Precursors/chemistry , Protein Precursors/metabolism , Stereocilia/metabolism , Amino Acid Sequence , Animals , Cadherin Related Proteins , Cadherins/ultrastructure , HEK293 Cells , Humans , Imaging, Three-Dimensional , Membrane Proteins/chemistry , Membrane Proteins/ultrastructure , Mice , Models, Molecular , Protein Multimerization , Protein Precursors/ultrastructure , Sf9 Cells
14.
Mol Biol Cell ; 14(11): 4329-41, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14551258

ABSTRACT

Ubiquitin is a small modifier protein that is conjugated to substrates to target them for degradation. Recently, a surprising number of ubiquitin-like proteins have been identified that also can be attached to proteins. Herein, we identify two molecular functions for the posttranslational protein modifier from Saccharomyces cerevisiae, Urm1p. Simultaneous loss of Urm1p and Cla4p, a p21-activated kinase that functions in budding, is lethal. This result suggests a role for the urmylation pathway in budding. Furthermore, loss of the urmylation pathway causes defects in invasive growth and confers sensitivity to rapamycin. Our results indicate that the sensitivity to rapamycin is due to a genetic interaction with the TOR pathway, which is important for regulation of cell growth in response to nutrients. We have found that Urm1p can be attached to a number of proteins. Loss of five genes that are also essential in a cla4Delta strain, NCS2, NCS6, ELP2, ELP6, and URE2, affect the level of at least one Urm1p conjugate. Moreover, these five genes have a role in invasive growth and display genetic interactions with the TOR pathway. In summary, our results suggest the urmylation pathway is involved in nutrient sensing and budding.


Subject(s)
Prions/metabolism , Protein Serine-Threonine Kinases/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Antibiotics, Antineoplastic/pharmacology , Cell Cycle Proteins , Cloning, Molecular , Glutathione Peroxidase , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Protein Binding , Protein Serine-Threonine Kinases/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/antagonists & inhibitors , Saccharomyces cerevisiae Proteins/genetics , Signal Transduction , Sirolimus/pharmacology
15.
Mol Biol Cell ; 14(4): 1501-16, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12686605

ABSTRACT

The p21-activated kinases Ste20p and Cla4p carry out undefined functions that are essential for viability during budding in Saccharomyces cerevisiae. To gain insight into the roles of Ste20p, we have used a synthetic lethal mutant screen to identify additional genes that are required in the absence of Cla4p. Altogether, we identified 65 genes, including genes with roles in cell polarity, mitosis, and cell wall maintenance. Herein, we focus on a set that defines a function carried out by Bni1p and several of its interacting proteins. We found that Bni1p and a group of proteins that complex with Bni1p (Bud6p, Spa2p, and Pea2p) are essential in a cla4delta mutant background. Bni1p, Bud6p, Spa2, and Pea2p are members of a group of polarity determining proteins referred to as the polarisome. Loss of polarisome proteins from a cla4delta strain causes cells to form elongated buds that have mislocalized septin rings. In contrast, other proteins that interact with or functionally associate with Bni1p and have roles in nuclear migration and cytokinesis, including Num1p and Hof1p, are not essential in the absence of Cla4p. Finally, we have found that Bni1p is phosphorylated in vivo, and a substantial portion of this phosphorylation is dependent on STE20. Together, these results suggest that one function of Ste20p may be to activate the polarisome complex by phosphorylation of Bni1p.


Subject(s)
Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Base Sequence , Cell Division , Cell Nucleus/metabolism , Cell Polarity , Cytoskeletal Proteins , DNA, Fungal/genetics , Fungal Proteins/genetics , Fungal Proteins/metabolism , Genes, Fungal , Intracellular Signaling Peptides and Proteins , MAP Kinase Kinase Kinases , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Mutation , Phosphorylation , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/growth & development , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Spindle Apparatus/metabolism
16.
Science ; 355(6331)2017 03 24.
Article in English | MEDLINE | ID: mdl-28232581

ABSTRACT

N-methyl-d-aspartate receptors (NMDARs) are heterotetrameric ion channels assembled as diheteromeric or triheteromeric complexes. Here, we report structures of the triheteromeric GluN1/GluN2A/GluN2B receptor in the absence or presence of the GluN2B-specific allosteric modulator Ro 25-6981 (Ro), determined by cryogenic electron microscopy (cryo-EM). In the absence of Ro, the GluN2A and GluN2B amino-terminal domains (ATDs) adopt "closed" and "open" clefts, respectively. Upon binding Ro, the GluN2B ATD clamshell transitions from an open to a closed conformation. Consistent with a predominance of the GluN2A subunit in ion channel gating, the GluN2A subunit interacts more extensively with GluN1 subunits throughout the receptor, in comparison with the GluN2B subunit. Differences in the conformation of the pseudo-2-fold-related GluN1 subunits further reflect receptor asymmetry. The triheteromeric NMDAR structures provide the first view of the most common NMDA receptor assembly and show how incorporation of two different GluN2 subunits modifies receptor symmetry and subunit interactions, allowing each subunit to uniquely influence receptor structure and function, thus increasing receptor complexity.


Subject(s)
Protein Multimerization , Receptors, Glutamate/chemistry , Receptors, N-Methyl-D-Aspartate/chemistry , Xenopus Proteins/chemistry , Allosteric Regulation , Animals , Antibodies, Monoclonal , Cryoelectron Microscopy , Models, Molecular , Neuronal Plasticity , Protein Domains , Receptors, Glutamate/immunology , Receptors, Glutamate/ultrastructure , Receptors, N-Methyl-D-Aspartate/immunology , Receptors, N-Methyl-D-Aspartate/ultrastructure , Xenopus Proteins/immunology , Xenopus Proteins/ultrastructure , Xenopus laevis
17.
Neuropharmacology ; 46(3): 299-310, 2004 Mar.
Article in English | MEDLINE | ID: mdl-14975685

ABSTRACT

Significant progress has been made toward understanding the mechanisms by which organisms learn from experiences and how those experiences are translated into memories. Advances in molecular, electrophysiological and genetic technologies have permitted great strides in identifying biochemical and structural changes that occur at synapses during processes that are thought to underlie learning and memory. Cellular events that generate the second messenger cyclic AMP (cAMP) and activate protein kinase A (PKA) have been linked to synaptic plasticity and long-term memory. In this review we will focus on the role of PKA in synaptic plasticity and discuss how the compartmentalization of PKA through its association with A-Kinase Anchoring Proteins (AKAPs) affect PKA function in this process.


Subject(s)
Adaptor Proteins, Signal Transducing , Carrier Proteins/physiology , Cyclic AMP-Dependent Protein Kinases/physiology , Neuronal Plasticity/physiology , Signal Transduction/physiology , Synapses/enzymology , Animals , Carrier Proteins/chemistry , Cyclic AMP-Dependent Protein Kinases/chemistry , Humans
18.
Adv Exp Med Biol ; 547: 91-105, 2004.
Article in English | MEDLINE | ID: mdl-15230095

ABSTRACT

We examined the morphogenetic transitions that yeast cells undergo in response to extracellular cues, and determined that multiple mechanisms control specificity of signal transduction pathway signaling and the attendant physiological response that ensues. This article describes the approaches that we used to determine these mechanisms. Our findings indicate that scaffolding proteins, which organize signal transduction pathways, are an especially powerful means to achieve specificity. We do not yet know how general this mechanism is. Our studies have also started to reveal ways in which a protein, Ste20, first identified as a participant in signal transduction pathways, may also connect to the basic cell biology machinery. Synthetic lethal genetic analysis has suggested that the polarisome and a new ubiquitin-like system may be targets of Ste20.


Subject(s)
Adaptor Proteins, Signal Transducing , Carrier Proteins/metabolism , Gene Expression Regulation, Fungal/physiology , Models, Biological , Pheromones/pharmacology , Saccharomyces cerevisiae Proteins/metabolism , Signal Transduction/physiology , Yeasts/cytology , Yeasts/physiology , Adaptation, Physiological/drug effects , Adaptation, Physiological/physiology , Cell Division/drug effects , Cell Size/drug effects , Gene Expression Regulation, Fungal/drug effects , Yeasts/drug effects
19.
Nat Protoc ; 9(11): 2574-85, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25299155

ABSTRACT

Structural, biochemical and biophysical studies of eukaryotic membrane proteins are often hampered by difficulties in overexpression of the candidate molecule. Baculovirus transduction of mammalian cells (BacMam), although a powerful method to heterologously express membrane proteins, can be cumbersome for screening and expression of multiple constructs. We therefore developed plasmid Eric Gouaux (pEG) BacMam, a vector optimized for use in screening assays, as well as for efficient production of baculovirus and robust expression of the target protein. In this protocol, we show how to use small-scale transient transfection and fluorescence-detection size-exclusion chromatography (FSEC) experiments using a GFP-His8-tagged candidate protein to screen for monodispersity and expression level. Once promising candidates are identified, we describe how to generate baculovirus, transduce HEK293S GnTI(-) (N-acetylglucosaminyltransferase I-negative) cells in suspension culture and overexpress the candidate protein. We have used these methods to prepare pure samples of chicken acid-sensing ion channel 1a (cASIC1) and Caenorhabditis elegans glutamate-gated chloride channel (GluCl) for X-ray crystallography, demonstrating how to rapidly and efficiently screen hundreds of constructs and accomplish large-scale expression in 4-6 weeks.


Subject(s)
Membrane Proteins/genetics , Protein Engineering/methods , Acid Sensing Ion Channels/genetics , Acid Sensing Ion Channels/metabolism , Animals , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Chickens , Chloride Channels/genetics , Chloride Channels/metabolism , Chromatography, Gel , Genetic Vectors , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HEK293 Cells , Histidine/genetics , Humans , Mammals , Membrane Proteins/metabolism , N-Acetylglucosaminyltransferases/metabolism , Plasmids/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Transfection/methods
20.
Science ; 325(5943): 1010-4, 2009 Aug 21.
Article in English | MEDLINE | ID: mdl-19608859

ABSTRACT

Amino acid, polyamine, and organocation (APC) transporters are secondary transporters that play essential roles in nutrient uptake, neurotransmitter recycling, ionic homeostasis, and regulation of cell volume. Here, we present the crystal structure of apo-ApcT, a proton-coupled broad-specificity amino acid transporter, at 2.35 angstrom resolution. The structure contains 12 transmembrane helices, with the first 10 consisting of an inverted structural repeat of 5 transmembrane helices like the leucine transporter LeuT. The ApcT structure reveals an inward-facing, apo state and an amine moiety of lysine-158 located in a position equivalent to the sodium ion site Na2 of LeuT. We propose that lysine-158 is central to proton-coupled transport and that the amine group serves the same functional role as the Na2 ion in LeuT, thus demonstrating common principles among proton- and sodium-coupled transporters.


Subject(s)
Amino Acid Transport Systems/chemistry , Amino Acid Transport Systems/metabolism , Archaeal Proteins/chemistry , Archaeal Proteins/metabolism , Methanococcus/chemistry , Amino Acid Sequence , Amino Acids/metabolism , Antiporters/chemistry , Apoproteins/chemistry , Apoproteins/metabolism , Crystallization , Crystallography, X-Ray , Escherichia coli Proteins/chemistry , Models, Molecular , Molecular Sequence Data , Protein Conformation , Protein Folding , Protein Structure, Secondary , Protons , Sodium/metabolism , Substrate Specificity
SELECTION OF CITATIONS
SEARCH DETAIL