Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 268
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 180(2): 323-339.e19, 2020 01 23.
Article in English | MEDLINE | ID: mdl-31928845

ABSTRACT

Teneurins are ancient metazoan cell adhesion receptors that control brain development and neuronal wiring in higher animals. The extracellular C terminus binds the adhesion GPCR Latrophilin, forming a trans-cellular complex with synaptogenic functions. However, Teneurins, Latrophilins, and FLRT proteins are also expressed during murine cortical cell migration at earlier developmental stages. Here, we present crystal structures of Teneurin-Latrophilin complexes that reveal how the lectin and olfactomedin domains of Latrophilin bind across a spiraling beta-barrel domain of Teneurin, the YD shell. We couple structure-based protein engineering to biophysical analysis, cell migration assays, and in utero electroporation experiments to probe the importance of the interaction in cortical neuron migration. We show that binding of Latrophilins to Teneurins and FLRTs directs the migration of neurons using a contact repulsion-dependent mechanism. The effect is observed with cell bodies and small neurites rather than their processes. The results exemplify how a structure-encoded synaptogenic protein complex is also used for repulsive cell guidance.


Subject(s)
Nerve Tissue Proteins/ultrastructure , Receptors, Peptide/metabolism , Tenascin/metabolism , Animals , Cell Adhesion/physiology , Crystallography, X-Ray/methods , HEK293 Cells , Humans , K562 Cells , Leucine-Rich Repeat Proteins , Membrane Glycoproteins/metabolism , Membrane Glycoproteins/ultrastructure , Membrane Proteins/metabolism , Membrane Proteins/ultrastructure , Mice , Mice, Inbred C57BL/embryology , Nerve Tissue Proteins/metabolism , Neurites/metabolism , Neurogenesis/physiology , Neurons/metabolism , Platelet Glycoprotein GPIb-IX Complex/metabolism , Platelet Glycoprotein GPIb-IX Complex/ultrastructure , Protein Binding/physiology , Proteins/metabolism , Proteins/ultrastructure , Receptors, Cell Surface/metabolism , Receptors, Peptide/ultrastructure , Synapses/metabolism , Tenascin/ultrastructure
2.
Nature ; 605(7909): 304-309, 2022 05.
Article in English | MEDLINE | ID: mdl-35344984

ABSTRACT

Frontotemporal lobar degeneration (FTLD) is the third most common neurodegenerative condition after Alzheimer's and Parkinson's diseases1. FTLD typically presents in 45 to 64 year olds with behavioural changes or progressive decline of language skills2. The subtype FTLD-TDP is characterized by certain clinical symptoms and pathological neuronal inclusions with TAR DNA-binding protein (TDP-43) immunoreactivity3. Here we extracted amyloid fibrils from brains of four patients representing four of the five FTLD-TDP subclasses, and determined their structures by cryo-electron microscopy. Unexpectedly, all amyloid fibrils examined were composed of a 135-residue carboxy-terminal fragment of transmembrane protein 106B (TMEM106B), a lysosomal membrane protein previously implicated as a genetic risk factor for FTLD-TDP4. In addition to TMEM106B fibrils, we detected abundant non-fibrillar aggregated TDP-43 by immunogold labelling. Our observations confirm that FTLD-TDP is associated with amyloid fibrils, and that the fibrils are formed by TMEM106B rather than TDP-43.


Subject(s)
Amyloid , DNA-Binding Proteins , Frontotemporal Lobar Degeneration , Membrane Proteins , Nerve Tissue Proteins , Amyloid/ultrastructure , Cryoelectron Microscopy , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/ultrastructure , Frontotemporal Lobar Degeneration/metabolism , Frontotemporal Lobar Degeneration/pathology , Humans , Membrane Proteins/metabolism , Membrane Proteins/ultrastructure , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/ultrastructure
3.
Nature ; 600(7887): 164-169, 2021 12.
Article in English | MEDLINE | ID: mdl-34789875

ABSTRACT

In the clades of animals that diverged from the bony fish, a group of Mas-related G-protein-coupled receptors (MRGPRs) evolved that have an active role in itch and allergic signals1,2. As an MRGPR, MRGPRX2 is known to sense basic secretagogues (agents that promote secretion) and is involved in itch signals and eliciting pseudoallergic reactions3-6. MRGPRX2 has been targeted by drug development efforts to prevent the side effects induced by certain drugs or to treat allergic diseases. Here we report a set of cryo-electron microscopy structures of the MRGPRX2-Gi1 trimer in complex with polycationic compound 48/80 or with inflammatory peptides. The structures of the MRGPRX2-Gi1 complex exhibited shallow, solvent-exposed ligand-binding pockets. We identified key common structural features of MRGPRX2 and describe a consensus motif for peptidic allergens. Beneath the ligand-binding pocket, the unusual kink formation at transmembrane domain 6 (TM6) and the replacement of the general toggle switch from Trp6.48 to Gly6.48 (superscript annotations as per Ballesteros-Weinstein nomenclature) suggest a distinct activation process. We characterized the interfaces of MRGPRX2 and the Gi trimer, and mapped the residues associated with key single-nucleotide polymorphisms on both the ligand and G-protein interfaces of MRGPRX2. Collectively, our results provide a structural basis for the sensing of cationic allergens by MRGPRX2, potentially facilitating the rational design of therapies to prevent unwanted pseudoallergic reactions.


Subject(s)
Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/metabolism , Pruritus/metabolism , Receptors, G-Protein-Coupled/chemistry , Receptors, G-Protein-Coupled/metabolism , Receptors, Neuropeptide/chemistry , Receptors, Neuropeptide/metabolism , Allergens/immunology , Amino Acid Motifs , Amino Acid Sequence , Binding Sites , Consensus Sequence , Cryoelectron Microscopy , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Humans , Models, Molecular , Nerve Tissue Proteins/immunology , Nerve Tissue Proteins/ultrastructure , Receptors, G-Protein-Coupled/immunology , Receptors, G-Protein-Coupled/ultrastructure , Receptors, Neuropeptide/immunology , Receptors, Neuropeptide/ultrastructure
4.
Nature ; 600(7887): 170-175, 2021 12.
Article in English | MEDLINE | ID: mdl-34789874

ABSTRACT

The MRGPRX family of receptors (MRGPRX1-4) is a family of mas-related G-protein-coupled receptors that have evolved relatively recently1. Of these, MRGPRX2 and MRGPRX4 are key physiological and pathological mediators of itch and related mast cell-mediated hypersensitivity reactions2-5. MRGPRX2 couples to both Gi and Gq in mast cells6. Here we describe agonist-stabilized structures of MRGPRX2 coupled to Gi1 and Gq in ternary complexes with the endogenous peptide cortistatin-14 and with a synthetic agonist probe, respectively, and the development of potent antagonist probes for MRGPRX2. We also describe a specific MRGPRX4 agonist and the structure of this agonist in a complex with MRGPRX4 and Gq. Together, these findings should accelerate the structure-guided discovery of therapeutic agents for pain, itch and mast cell-mediated hypersensitivity.


Subject(s)
Cryoelectron Microscopy , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/chemistry , Pruritus/metabolism , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, G-Protein-Coupled/chemistry , Receptors, Neuropeptide/antagonists & inhibitors , Receptors, Neuropeptide/chemistry , Drug Inverse Agonism , GTP-Binding Protein alpha Subunits, Gi-Go/chemistry , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , GTP-Binding Protein alpha Subunits, Gi-Go/ultrastructure , GTP-Binding Protein alpha Subunits, Gq-G11/chemistry , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , GTP-Binding Protein alpha Subunits, Gq-G11/ultrastructure , Humans , Models, Molecular , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/ultrastructure , Receptors, G-Protein-Coupled/metabolism , Receptors, G-Protein-Coupled/ultrastructure , Receptors, Neuropeptide/metabolism , Receptors, Neuropeptide/ultrastructure
5.
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
6.
Nature ; 584(7822): 646-651, 2020 08.
Article in English | MEDLINE | ID: mdl-32494015

ABSTRACT

Pannexin 1 (PANX1) is an ATP-permeable channel with critical roles in a variety of physiological functions such as blood pressure regulation1, apoptotic cell clearance2 and human oocyte development3. Here we present several structures of human PANX1 in a heptameric assembly at resolutions of up to 2.8 angström, including an apo state, a caspase-7-cleaved state and a carbenoxolone-bound state. We reveal a gating mechanism that involves two ion-conducting pathways. Under normal cellular conditions, the intracellular entry of the wide main pore is physically plugged by the C-terminal tail. Small anions are conducted through narrow tunnels in the intracellular domain. These tunnels connect to the main pore and are gated by a long linker between the N-terminal helix and the first transmembrane helix. During apoptosis, the C-terminal tail is cleaved by caspase, allowing the release of ATP through the main pore. We identified a carbenoxolone-binding site embraced by W74 in the extracellular entrance and a role for carbenoxolone as a channel blocker. We identified a gap-junction-like structure using a glycosylation-deficient mutant, N255A. Our studies provide a solid foundation for understanding the molecular mechanisms underlying the channel gating and inhibition of PANX1 and related large-pore channels.


Subject(s)
Connexins/chemistry , Connexins/metabolism , Cryoelectron Microscopy , Ion Channel Gating , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/metabolism , Patch-Clamp Techniques , Adenosine Triphosphate/metabolism , Animals , Apoproteins/chemistry , Apoproteins/metabolism , Apoproteins/ultrastructure , Apoptosis , Binding Sites/drug effects , Carbenoxolone/chemistry , Carbenoxolone/metabolism , Carbenoxolone/pharmacology , Caspase 7/metabolism , Cell Line , Connexins/ultrastructure , Gap Junctions , Glycosylation , Humans , Ion Channel Gating/drug effects , Models, Molecular , Mutation , Nerve Tissue Proteins/ultrastructure , Protein Subunits/chemistry , Protein Subunits/metabolism , Sf9 Cells
7.
Nature ; 567(7746): 127-131, 2019 03.
Article in English | MEDLINE | ID: mdl-30814734

ABSTRACT

The GABAB (γ-aminobutyric acid type B) receptor is one of the principal inhibitory neurotransmitter receptors in the brain, and it signals through heterotrimeric G proteins to activate a variety of effectors, including G-protein-coupled inwardly rectifying potassium channels (GIRKs)1,2. GABAB-receptor signalling is tightly regulated by auxiliary subunits called KCTDs, which control the kinetics of GIRK activation and desensitization3-5. However, the mechanistic basis for KCTD modulation of GABAB signalling remains incompletely understood. Here, using a combination of X-ray crystallography, electron microscopy, and functional and biochemical experiments, we reveal the molecular details of KCTD binding to both GABAB receptors and G-protein ßγ subunits. KCTDs associate with the receptor by forming an asymmetric pentameric ring around a region of the receptor carboxy-terminal tail, while a second KCTD domain, H1, engages in a symmetric interaction with five copies of Gßγ in which the G-protein subunits also interact directly with one another. We further show that KCTD binding to Gßγ is highly cooperative, defining a model in which KCTD proteins cooperatively strip G proteins from GIRK channels to induce rapid desensitization following receptor activation. These results provide a framework for understanding the molecular basis for the precise temporal control of GABAB signalling by KCTD proteins.


Subject(s)
Intracellular Signaling Peptides and Proteins/chemistry , Nerve Tissue Proteins/chemistry , Proteins/chemistry , Receptors, GABA-B/chemistry , Receptors, GABA-B/metabolism , Signal Transduction , Crystallography, X-Ray , G Protein-Coupled Inwardly-Rectifying Potassium Channels/chemistry , G Protein-Coupled Inwardly-Rectifying Potassium Channels/metabolism , GTP-Binding Protein beta Subunits/chemistry , GTP-Binding Protein beta Subunits/metabolism , GTP-Binding Protein beta Subunits/ultrastructure , GTP-Binding Protein gamma Subunits/chemistry , GTP-Binding Protein gamma Subunits/metabolism , GTP-Binding Protein gamma Subunits/ultrastructure , Humans , Microscopy, Electron , Models, Biological , Models, Molecular , Nerve Tissue Proteins/ultrastructure , Protein Binding , Protein Domains , Proteins/metabolism , Proteins/ultrastructure , Receptors, GABA-B/ultrastructure
8.
Proc Natl Acad Sci U S A ; 117(27): 15620-15631, 2020 07 07.
Article in English | MEDLINE | ID: mdl-32576689

ABSTRACT

Repulsive guidance molecules (RGMs) are cell surface proteins that regulate the development and homeostasis of many tissues and organs, including the nervous, skeletal, and immune systems. They control fundamental biological processes, such as migration and differentiation by direct interaction with the Neogenin (NEO1) receptor and function as coreceptors for the bone morphogenetic protein (BMP)/growth differentiation factor (GDF) family. We determined crystal structures of all three human RGM family members in complex with GDF5, as well as the ternary NEO1-RGMB-GDF5 assembly. Surprisingly, we show that all three RGMs inhibit GDF5 signaling, which is in stark contrast to RGM-mediated enhancement of signaling observed for other BMPs, like BMP2. Despite their opposite effect on GDF5 signaling, RGMs occupy the BMP type 1 receptor binding site similar to the observed interactions in RGM-BMP2 complexes. In the NEO1-RGMB-GDF5 complex, RGMB physically bridges NEO1 and GDF5, suggesting cross-talk between the GDF5 and NEO1 signaling pathways. Our crystal structures, combined with structure-guided mutagenesis of RGMs and BMP ligands, binding studies, and cellular assays suggest that RGMs inhibit GDF5 signaling by competing with GDF5 type 1 receptors. While our crystal structure analysis and in vitro binding data initially pointed towards a simple competition mechanism between RGMs and type 1 receptors as a possible basis for RGM-mediated GDF5 inhibition, further experiments utilizing BMP2-mimicking GDF5 variants clearly indicate a more complex mechanism that explains how RGMs can act as a functionality-changing switch for two structurally and biochemically similar signaling molecules.


Subject(s)
Cell Adhesion Molecules, Neuronal/metabolism , GPI-Linked Proteins/metabolism , Growth Differentiation Factor 5/metabolism , Hemochromatosis Protein/metabolism , Nerve Tissue Proteins/metabolism , Bone Morphogenetic Protein 2/metabolism , Bone Morphogenetic Protein 2/ultrastructure , Cell Adhesion Molecules, Neuronal/ultrastructure , Crystallography, X-Ray , GPI-Linked Proteins/ultrastructure , Growth Differentiation Factor 5/ultrastructure , Hemochromatosis Protein/ultrastructure , Membrane Proteins/metabolism , Membrane Proteins/ultrastructure , Nerve Tissue Proteins/ultrastructure , Protein Domains , Recombinant Proteins/metabolism , Recombinant Proteins/ultrastructure , Signal Transduction
9.
Nucleic Acids Res ; 48(18): 10329-10341, 2020 10 09.
Article in English | MEDLINE | ID: mdl-32663306

ABSTRACT

The recently characterized mammalian writer (methyltransferase) and eraser (demethylase) of the DNA N6-methyladenine (N6mA) methyl mark act on single-stranded (ss) and transiently-unpaired DNA. As YTH domain-containing proteins bind N6mA-containing RNA in mammalian cells, we investigated whether mammalian YTH domains are also methyl mark readers of N6mA DNA. Here, we show that the YTH domain of YTHDC1 (known to localize in the nucleus) binds ssDNA containing N6mA, with a 10 nM dissociation constant. This binding is stronger by a factor of 5 than in an RNA context, tested under the same conditions. However, the YTH domains of YTHDF2 and YTHDF1 (predominantly cytoplasmic) exhibited the opposite effect with ∼1.5-2נstronger binding to ssRNA containing N6mA than to the corresponding DNA. We determined two structures of the YTH domain of YTHDC1 in complex with N6mA-containing ssDNA, which illustrated that YTHDC1 binds the methylated adenine in a single-stranded region flanked by duplexed DNA. We discuss the hypothesis that the writer-reader-eraser of N6mA-containining ssDNA is associated with maintaining genome stability. Structural comparison of YTH and SRA domains (the latter a DNA 5-methylcytosine reader) revealed them to be diverse members of a larger family of DNA/RNA modification readers, apparently having originated from bacterial modification-dependent restriction enzymes.


Subject(s)
Adenine/chemistry , Multiprotein Complexes/chemistry , Nerve Tissue Proteins/chemistry , Protein Conformation , RNA Splicing Factors/chemistry , DNA/chemistry , DNA/genetics , DNA/ultrastructure , DNA, Single-Stranded/chemistry , DNA, Single-Stranded/genetics , Histone Demethylases/genetics , Humans , Methylation , Methyltransferases/genetics , Multiprotein Complexes/genetics , Multiprotein Complexes/ultrastructure , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/ultrastructure , Protein Domains/genetics , RNA Splicing Factors/genetics , RNA Splicing Factors/ultrastructure , RNA-Binding Proteins/genetics
10.
Nature ; 520(7548): 511-7, 2015 Apr 23.
Article in English | MEDLINE | ID: mdl-25855297

ABSTRACT

The TRPA1 ion channel (also known as the wasabi receptor) is a detector of noxious chemical agents encountered in our environment or produced endogenously during tissue injury or drug metabolism. These include a broad class of electrophiles that activate the channel through covalent protein modification. TRPA1 antagonists hold potential for treating neurogenic inflammatory conditions provoked or exacerbated by irritant exposure. Despite compelling reasons to understand TRPA1 function, structural mechanisms underlying channel regulation remain obscure. Here we use single-particle electron cryo- microscopy to determine the structure of full-length human TRPA1 to ∼4 Å resolution in the presence of pharmacophores, including a potent antagonist. Several unexpected features are revealed, including an extensive coiled-coil assembly domain stabilized by polyphosphate co-factors and a highly integrated nexus that converges on an unpredicted transient receptor potential (TRP)-like allosteric domain. These findings provide new insights into the mechanisms of TRPA1 regulation, and establish a blueprint for structure-based design of analgesic and anti-inflammatory agents.


Subject(s)
Calcium Channels/chemistry , Calcium Channels/ultrastructure , Cryoelectron Microscopy , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/ultrastructure , Transient Receptor Potential Channels/chemistry , Transient Receptor Potential Channels/ultrastructure , Allosteric Regulation , Analgesics , Ankyrin Repeat , Anti-Inflammatory Agents , Binding Sites , Calcium Channels/metabolism , Cytosol/metabolism , Humans , Models, Molecular , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/metabolism , Polyphosphates/metabolism , Polyphosphates/pharmacology , Protein Stability/drug effects , Protein Subunits/chemistry , Protein Subunits/metabolism , Structure-Activity Relationship , TRPA1 Cation Channel , Transient Receptor Potential Channels/antagonists & inhibitors , Transient Receptor Potential Channels/metabolism
11.
Hum Genet ; 139(12): 1513-1529, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32529326

ABSTRACT

Prader-Willi syndrome (PWS) is a neurodevelopmental disorder caused by the loss of function of a set of imprinted genes on chromosome 15q11-15q13. One of these genes, NDN, encodes necdin, a protein that is important for neuronal differentiation and survival. Loss of Ndn in mice causes defects in the formation and function of the nervous system. Necdin is a member of the melanoma-associated antigen gene (MAGE) protein family. The functions of MAGE proteins depend highly on their interactions with other proteins, and in particular MAGE proteins interact with E3 ubiquitin ligases and deubiquitinases to form MAGE-RING E3 ligase-deubiquitinase complexes. Here, we used proximity-dependent biotin identification (BioID) and mass spectrometry (MS) to determine the network of protein-protein interactions (interactome) of the necdin protein. This process yielded novel as well as known necdin-proximate proteins that cluster into a protein network. Next, we used BioID-MS to define the interactomes of necdin proteins carrying coding variants. Variant necdin proteins had interactomes that were distinct from wildtype necdin. BioID-MS is not only a useful tool to identify protein-protein interactions, but also to analyze the effects of variants of unknown significance on the interactomes of proteins involved in genetic disease.


Subject(s)
Amino Acid Substitution/genetics , Nerve Tissue Proteins/genetics , Nuclear Proteins/genetics , Protein Interaction Maps/genetics , Ubiquitin-Protein Ligases/genetics , Animals , Biotinylation/genetics , Cell Differentiation/genetics , Deubiquitinating Enzymes/genetics , Gene Expression Regulation/genetics , HEK293 Cells , Humans , Mass Spectrometry/methods , Mice , Mutation/genetics , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/ultrastructure , Nervous System Malformations/genetics , Nervous System Malformations/pathology , Neurons/metabolism , Nuclear Proteins/chemistry , Nuclear Proteins/ultrastructure , Poly(A)-Binding Proteins/chemistry , Poly(A)-Binding Proteins/genetics , Prader-Willi Syndrome/genetics , Protein Conformation , Structure-Activity Relationship , Ubiquitin-Protein Ligases/chemistry
12.
Alcohol Clin Exp Res ; 44(7): 1344-1355, 2020 07.
Article in English | MEDLINE | ID: mdl-32424866

ABSTRACT

BACKGROUND: EtOH has a significant effect on synaptic plasticity. Munc13-1 is an essential presynaptic active zone protein involved in priming the synaptic vesicle and releasing neurotransmitter in the brain. It is a peripheral membrane protein and binds to the activator, diacylglycerol (DAG)/phorbol ester at its membrane-targeting C1 domain. Our previous studies identified Glu-582 of C1 domain as the alcohol-binding residue (Das, J. et al, J. Neurochem., 126, 715-726, 2013). METHODS: Here, we describe a 250 ns molecular dynamics (MD) simulation study on the interaction of EtOH and the activator-bound Munc13-1 C1 in the presence of varying concentrations of phosphatidylserine (PS). RESULTS: In this study, Munc13-1 C1 shows higher conformational stability in EtOH than in water. It forms fewer hydrogen bonds with phorbol 13-acetate in the presence of EtOH than in water. EtOH also affected the interaction between the protein and the membrane and between the activator and the membrane. Similar studies in a E582A mutant suggest that these effects of EtOH are mostly mediated through Glu-582. CONCLUSIONS: EtOH forms hydrogen bonds with Glu-582. While occupancy of the EtOH molecules at the vicinity (4Å) of Glu-582 is 34.4%, the occupancy in the E582A mutant is 26.5% of the simulation time. In addition, the amount of PS in the membrane influences the conformational stability of the C1 domain and interactions in the ternary complex. This study is important in providing the structural basis of EtOH's effects on synaptic plasticity.


Subject(s)
Central Nervous System Depressants/metabolism , Ethanol/metabolism , Nerve Tissue Proteins/metabolism , Synaptic Membranes/metabolism , Humans , Molecular Dynamics Simulation , Mutation , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/ultrastructure , Phorbol Esters , Presynaptic Terminals/metabolism , Protein Conformation , Protein Domains/genetics
13.
Glia ; 67(4): 634-649, 2019 04.
Article in English | MEDLINE | ID: mdl-30637801

ABSTRACT

Proteolipid protein (PLP) is the most abundant integral membrane protein in central nervous system (CNS) myelin. Expression of the Plp-gene in oligodendrocytes is not essential for the biosynthesis of myelin membranes but required to prevent axonal pathology. This raises the question whether the exceptionally high level of PLP in myelin is required later in life, or whether high-level PLP expression becomes dispensable once myelin has been assembled. Both models require a better understanding of the turnover of PLP in myelin in vivo. Thus, we generated and characterized a novel line of tamoxifen-inducible Plp-mutant mice that allowed us to determine the rate of PLP turnover after developmental myelination has been completed, and to assess the possible impact of gradually decreasing amounts of PLP for myelin and axonal integrity. We found that 6 months after targeting the Plp-gene the abundance of PLP in CNS myelin was about halved, probably reflecting that myelin is slowly turned over in the adult brain. Importantly, this reduction by 50% was sufficient to cause the entire spectrum of neuropathological changes previously associated with the developmental lack of PLP, including myelin outfoldings, lamellae splittings, and axonal spheroids. In comparison to axonopathy and gliosis, the infiltration of cytotoxic T-cells was temporally delayed, suggesting a corresponding chronology also in the genetic disorders of PLP-deficiency. High-level abundance of PLP in myelin throughout adult life emerges as a requirement for the preservation of white matter integrity.


Subject(s)
Axons/metabolism , Central Nervous System/cytology , Myelin Proteolipid Protein/metabolism , Myelin Sheath/metabolism , Animals , Axons/ultrastructure , Cytokines/genetics , Cytokines/metabolism , Estrogen Antagonists/pharmacology , Gene Expression/drug effects , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Immunohistochemistry , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/ultrastructure , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Electron, Transmission , Myelin Proteins/genetics , Myelin Proteins/metabolism , Myelin Proteins/ultrastructure , Myelin Proteolipid Protein/genetics , Myelin Proteolipid Protein/ultrastructure , Myelin Sheath/ultrastructure , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/ultrastructure , RNA, Messenger/metabolism , Tamoxifen/pharmacology
14.
Proc Natl Acad Sci U S A ; 113(6): 1546-51, 2016 Feb 09.
Article in English | MEDLINE | ID: mdl-26831073

ABSTRACT

Polyglutamine expansion within the exon1 of huntingtin leads to protein misfolding, aggregation, and cytotoxicity in Huntington's disease. This incurable neurodegenerative disease is the most prevalent member of a family of CAG repeat expansion disorders. Although mature exon1 fibrils are viable candidates for the toxic species, their molecular structure and how they form have remained poorly understood. Using advanced magic angle spinning solid-state NMR, we directly probe the structure of the rigid core that is at the heart of huntingtin exon1 fibrils and other polyglutamine aggregates, via measurements of long-range intramolecular and intermolecular contacts, backbone and side-chain torsion angles, relaxation measurements, and calculations of chemical shifts. These experiments reveal the presence of ß-hairpin-containing ß-sheets that are connected through interdigitating extended side chains. Despite dramatic differences in aggregation behavior, huntingtin exon1 fibrils and other polyglutamine-based aggregates contain identical ß-strand-based cores. Prior structural models, derived from X-ray fiber diffraction and computational analyses, are shown to be inconsistent with the solid-state NMR results. Internally, the polyglutamine amyloid fibrils are coassembled from differently structured monomers, which we describe as a type of "intrinsic" polymorphism. A stochastic polyglutamine-specific aggregation mechanism is introduced to explain this phenomenon. We show that the aggregation of mutant huntingtin exon1 proceeds via an intramolecular collapse of the expanded polyglutamine domain and discuss the implications of this observation for our understanding of its misfolding and aggregation mechanisms.


Subject(s)
Exons/genetics , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/genetics , Peptides/chemistry , Amino Acid Sequence , Amyloid/chemistry , Magnetic Resonance Spectroscopy , Models, Molecular , Molecular Sequence Data , Nerve Tissue Proteins/ultrastructure , Peptides/genetics , Protein Structure, Secondary , Stochastic Processes
15.
J Neurosci ; 37(48): 11592-11604, 2017 11 29.
Article in English | MEDLINE | ID: mdl-29074576

ABSTRACT

Human genetic studies support that loss-of-function mutations in the SH3 domain and ankyrin repeat containing family proteins (SHANK1-3), the large synaptic scaffolding proteins enriched at the postsynaptic density of excitatory synapses, are causative for autism spectrum disorder and other neuropsychiatric disorders in humans. To better understand the in vivo functions of Shank and facilitate dissection of neuropathology associated with SHANK mutations in human, we generated multiple mutations in the Shank gene, the only member of the SHANK family in Drosophila melanogaster Both male and female Shank null mutants were fully viable and fertile with no apparent morphological or developmental defects. Expression analysis revealed apparent enrichment of Shank in the neuropils of the CNS. Specifically, Shank coexpressed with another PSD scaffold protein, Homer, in the calyx of mushroom bodies in the brain. Consistent with high expression in mushroom body calyces, Shank mutants show an abnormal calyx structure and reduced olfactory acuity. These morphological and functional phenotypes were fully rescued by pan-neuronal reexpression of Shank, and only partially rescued by presynaptic but no rescue by postsynaptic reexpression of Shank. Our findings thus establish a previously unappreciated presynaptic function of Shank.SIGNIFICANCE STATEMENT Mutations in SHANK family genes are causative for idiopathic autism spectrum disorder. To understand the neural function of Shank, a large scaffolding protein enriched at the postsynaptic densities, we examined the role of Drosophila Shank in synapse development at the peripheral neuromuscular junctions and the central mushroom body calyx. Our results demonstrate that, in addition to its conventional postsynaptic function, Shank also acts presynaptically in synapse development in the brain. This study offers novel insights into the synaptic role of Shank.


Subject(s)
Nerve Tissue Proteins/physiology , Nerve Tissue Proteins/ultrastructure , Presynaptic Terminals/physiology , Presynaptic Terminals/ultrastructure , Animals , Animals, Genetically Modified , Drosophila , Female , Male , Mushroom Bodies/physiology , Mushroom Bodies/ultrastructure , Neuromuscular Junction/physiology , Neuromuscular Junction/ultrastructure
16.
J Neurosci ; 37(7): 1910-1924, 2017 02 15.
Article in English | MEDLINE | ID: mdl-28115484

ABSTRACT

Target cell type-dependent differences in presynaptic release probability (Pr ) and short-term plasticity are intriguing features of cortical microcircuits that increase the computational power of neuronal networks. Here, we tested the hypothesis that different voltage-gated Ca2+ channel densities in presynaptic active zones (AZs) underlie different Pr values. Two-photon Ca2+ imaging, triple immunofluorescent labeling, and 3D electron microscopic (EM) reconstruction of rat CA3 pyramidal cell axon terminals revealed ∼1.7-1.9 times higher Ca2+ inflow per AZ area in high Pr boutons synapsing onto parvalbumin-positive interneurons (INs) than in low Pr boutons synapsing onto mGluR1α-positive INs. EM replica immunogold labeling, however, demonstrated only 1.15 times larger Cav2.1 and Cav2.2 subunit densities in high Pr AZs. Our results indicate target cell type-specific modulation of voltage-gated Ca2+ channel function or different subunit composition as possible mechanisms underlying the functional differences. In addition, high Pr synapses are also characterized by a higher density of docked vesicles, suggesting that a concerted action of these mechanisms underlies the functional differences.SIGNIFICANCE STATEMENT Target cell type-dependent variability in presynaptic properties is an intriguing feature of cortical synapses. When a single cortical pyramidal cell establishes a synapse onto a somatostatin-expressing interneuron (IN), the synapse releases glutamate with low probability, whereas the next bouton of the same axon has high release probability when its postsynaptic target is a parvalbumin-expressing IN. Here, we used combined molecular, imaging, and anatomical approaches to investigate the mechanisms underlying these differences. Our functional experiments implied an approximately twofold larger Ca2+ channel density in high release probability boutons, whereas freeze-fracture immunolocalization demonstrated only a 15% difference in Ca2+ channel subunit densities. Our results point toward a postsynaptic target cell type-dependent regulation of Ca2+ channel function or different subunit composition as the underlying mechanism.


Subject(s)
Calcium Channels/metabolism , Glutamic Acid/metabolism , Hippocampus/cytology , Neuronal Plasticity/physiology , Neurons/metabolism , Presynaptic Terminals/metabolism , Probability , Action Potentials/physiology , Animals , Animals, Newborn , Axons/metabolism , Calcium/metabolism , Calcium Channels/ultrastructure , Glutamic Acid/classification , In Vitro Techniques , Lysine/analogs & derivatives , Lysine/metabolism , Male , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/ultrastructure , Parvalbumins/metabolism , Presynaptic Terminals/ultrastructure , Rats , Rats, Wistar , Receptors, Metabotropic Glutamate/metabolism , Receptors, Metabotropic Glutamate/ultrastructure , gamma-Aminobutyric Acid/metabolism
17.
Proc Natl Acad Sci U S A ; 112(8): 2413-8, 2015 Feb 24.
Article in English | MEDLINE | ID: mdl-25675501

ABSTRACT

G-protein signaling depends on the ability of the individual subunits of the G-protein heterotrimer to assemble into a functional complex. Formation of the G-protein ßγ (Gßγ) dimer is particularly challenging because it is an obligate dimer in which the individual subunits are unstable on their own. Recent studies have revealed an intricate chaperone system that brings Gß and Gγ together. This system includes cytosolic chaperonin containing TCP-1 (CCT; also called TRiC) and its cochaperone phosducin-like protein 1 (PhLP1). Two key intermediates in the Gßγ assembly process, the Gß-CCT and the PhLP1-Gß-CCT complexes, were isolated and analyzed by a hybrid structural approach using cryo-electron microscopy, chemical cross-linking coupled with mass spectrometry, and unnatural amino acid cross-linking. The structures show that Gß interacts with CCT in a near-native state through interactions of the Gγ-binding region of Gß with the CCTγ subunit. PhLP1 binding stabilizes the Gß fold, disrupting interactions with CCT and releasing a PhLP1-Gß dimer for assembly with Gγ. This view provides unique insight into the interplay between CCT and a cochaperone to orchestrate the folding of a protein substrate.


Subject(s)
Carrier Proteins/chemistry , Chaperonin Containing TCP-1/chemistry , GTP-Binding Protein beta Subunits/chemistry , GTP-Binding Protein gamma Subunits/chemistry , Nerve Tissue Proteins/chemistry , Protein Multimerization , Amino Acids/metabolism , Animals , Benzophenones , Carrier Proteins/ultrastructure , Chaperonin Containing TCP-1/ultrastructure , Cross-Linking Reagents/metabolism , Cryoelectron Microscopy , GTP-Binding Protein beta Subunits/ultrastructure , GTP-Binding Protein gamma Subunits/ultrastructure , Humans , Mass Spectrometry , Models, Molecular , Nerve Tissue Proteins/ultrastructure , Phenylalanine/analogs & derivatives , Protein Structure, Secondary
18.
J Neurosci ; 36(7): 2261-6, 2016 Feb 17.
Article in English | MEDLINE | ID: mdl-26888935

ABSTRACT

The voltage-gated K(+) channels Kv7.2 and Kv7.3 are located at the axon initial segment (AIS) and exert strong control over action potential generation. Therefore, changes in their localization or cell surface numbers are likely to influence neuronal signaling. However, nothing is known about the cell surface dynamics of Kv7.2/7.3 at steady state or during short-term neuronal stimulation. This is primarily attributable to their membrane topology, which hampers extracellular epitope tagging. Here we circumvent this limitation by fusing an extra phluorin-tagged helix to the N terminus of human Kv7.3. This seven transmembrane chimera, named super ecliptic phluorin (SEP)-TAC-7.3, functions and traffics as a wild-type (WT) channel. We expressed SEP-TAC-7.3 in dissociated rat hippocampal neurons to examine the lateral mobility, surface numbers, and localization of AIS Kv7.2/7.3 heteromers using live imaging. We discovered that they are extraordinarily stable and exhibit a very low surface mobility both during steady state and neuronal stimulation. In the latter case, we also found that neither localization nor cell surface numbers were changed. However, at high glutamate loads, we observed a rapid irreversible endocytosis of Kv7.2/7.3, which required the activation of NR2B-containing NMDA receptors, Ca(2+) influx, and calpain activation. This excitotoxic mechanism may be specific to ankyrin G-bound AIS proteins because Nav1.2 channels, but not AIS GABAA receptors, were also endocytosed. In conclusion, we have, for the first time, characterized the cell surface dynamics of a full-length Kv7 channel using a novel chimeric strategy. This approach is likely also applicable to other Kv channels and thus of value for the additional characterization of this ion channel subfamily. SIGNIFICANCE STATEMENT: The voltage-gated K(+) channels Kv7.2 and Kv7.3 exert strong control over action potential generation, but little is known about their cell surface dynamics. Using a novel phluorin-based approach, we here show that these channels are highly stable at steady state and different types of neuronal stimulation. However, at high glutamate loads, they undergo a rapid calpain-dependent endocytosis that likely represents an early response during excitotoxic states.


Subject(s)
Axons/metabolism , Calpain/metabolism , Down-Regulation/genetics , KCNQ2 Potassium Channel/metabolism , KCNQ3 Potassium Channel/metabolism , Nerve Tissue Proteins/metabolism , Animals , Ankyrins/genetics , Axons/ultrastructure , Calcium Signaling/genetics , Chimera/genetics , Female , Humans , KCNQ2 Potassium Channel/ultrastructure , KCNQ3 Potassium Channel/ultrastructure , Male , Mice , Nerve Tissue Proteins/ultrastructure , Patch-Clamp Techniques , Pregnancy , Rats , Receptors, Cell Surface/metabolism , Receptors, GABA-A/genetics , Receptors, N-Methyl-D-Aspartate/genetics
19.
J Biol Chem ; 291(11): 5788-5802, 2016 Mar 11.
Article in English | MEDLINE | ID: mdl-26721881

ABSTRACT

Contactin-associated protein-like 2 (CNTNAP2) encodes for CASPR2, a multidomain single transmembrane protein belonging to the neurexin superfamily that has been implicated in a broad range of human phenotypes including autism and language impairment. Using a combination of biophysical techniques, including small angle x-ray scattering, single particle electron microscopy, analytical ultracentrifugation, and bio-layer interferometry, we present novel structural and functional data that relate the architecture of the extracellular domain of CASPR2 to a previously unknown ligand, Contactin1 (CNTN1). Structurally, CASPR2 is highly glycosylated and has an overall compact architecture. Functionally, we show that CASPR2 associates with micromolar affinity with CNTN1 but, under the same conditions, it does not interact with any of the other members of the contactin family. Moreover, by using dissociated hippocampal neurons we show that microbeads loaded with CASPR2, but not with a deletion mutant, co-localize with transfected CNTN1, suggesting that CNTN1 is an endogenous ligand for CASPR2. These data provide novel insights into the structure and function of CASPR2, suggesting a complex role of CASPR2 in the nervous system.


Subject(s)
Contactin 1/metabolism , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/metabolism , Animals , Cells, Cultured , HEK293 Cells , Hippocampus/cytology , Hippocampus/metabolism , Humans , Membrane Proteins/ultrastructure , Mice, Inbred C57BL , Models, Molecular , Nerve Tissue Proteins/ultrastructure , Protein Interaction Maps , Protein Structure, Tertiary , Scattering, Small Angle , X-Ray Diffraction
20.
Biochem Biophys Res Commun ; 491(3): 760-766, 2017 09 23.
Article in English | MEDLINE | ID: mdl-28739258

ABSTRACT

Human histidine triad nucleotide binding protein 1 (hHint1) is a purine nucleoside phosphoramidase and adenylate hydrolase that has emerged as a potential therapeutic target for the management of pain. However, the molecular mechanism of Hint1 in the signaling pathway has remained less clear. The role of metal ions in regulating postsynaptic transmission is well known, and the active site of hHint1 contains multiple histidines. Here we have investigated the effect of divalent metal ions (Cd2+, Cu2+, Mg2+, Mn2+, Ni2+, and Zn2+) on the structural integrity and catalytic activity of hHint1. With the exception of Mg2+, all the divalent ions inhibited hHint1, the rank of order was found to be Cu2+ >Zn2+ >Cd2+ ≥Ni2+ >Mn2+ based on their IC50 and kin/KI values. A crystal structure of hHint1 with bound Cu2+ is described to explain the competitive reversible inactivation of hHint1 by divalent cations. All the metal ions exhibited time- and concentration- dependent inhibition, with the rate of inactivation highly dependent on alterations of the C-terminus. With the exception of Cu2+; restoration of inhibition was observed for all the metal ions after treatment with EDTA. Our studies reveal a loss in secondary structure and aggregation of hHint1 upon incubation with 10-fold excess of copper. Thus, hHint1 appears to be structurally sensitive to irreversible inactivation by copper, which may be of neurotoxicological and pharmacological significance.


Subject(s)
Metals/chemistry , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/ultrastructure , Analgesics, Opioid/chemistry , Animals , Binding Sites , Catalysis , Cations, Divalent/chemistry , Copper/chemistry , Humans , Ions , Neuralgia/metabolism , Protein Binding , Protein Unfolding
SELECTION OF CITATIONS
SEARCH DETAIL