Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Science ; 373(6556)2021 08 13.
Article in English | MEDLINE | ID: mdl-34385370

ABSTRACT

The neurotransmitter acetylcholine (ACh) acts in part through a family of nicotinic ACh receptors (nAChRs), which mediate diverse physiological processes including muscle contraction, neurotransmission, and sensory transduction. Pharmacologically, nAChRs are responsible for tobacco addiction and are targeted by medicines for hypertension and dementia. Nicotinic AChRs were the first ion channels to be isolated. Recent studies have identified molecules that control nAChR biogenesis, trafficking, and function. These nAChR accessories include protein and chemical chaperones as well as auxiliary subunits. Whereas some factors act on many nAChRs, others are receptor specific. Discovery of these regulatory mechanisms is transforming nAChR research in cells and tissues ranging from central neurons to spinal ganglia to cochlear hair cells. Nicotinic AChR-specific accessories also enable drug discovery on high-confidence targets for psychiatric, neurological, and auditory disorders.


Subject(s)
Molecular Chaperones/metabolism , Neurons/metabolism , Proteins/metabolism , Receptors, Nicotinic/metabolism , Animals , Cell Membrane/metabolism , Drug Discovery , Endoplasmic Reticulum/metabolism , Humans , Ligands , Muscle, Skeletal/metabolism , Neuropharmacology , Nicotine/metabolism , Protein Subunits/metabolism , Receptors, Nicotinic/chemistry
2.
J Clin Invest ; 130(11): 6158-6170, 2020 11 02.
Article in English | MEDLINE | ID: mdl-33074244

ABSTRACT

The α6ß4 nicotinic acetylcholine receptor (nAChR) is enriched in dorsal root ganglia neurons and is an attractive non-opioid therapeutic target for pain. However, difficulty expressing human α6ß4 receptors in recombinant systems has precluded drug discovery. Here, genome-wide screening identified accessory proteins that enable reconstitution of human α6ß4 nAChRs. BARP, an auxiliary subunit of voltage-dependent calcium channels, promoted α6ß4 surface expression while IRE1α, an unfolded protein response sensor, enhanced α6ß4 receptor assembly. Effects on α6ß4 involve BARP's N-terminal region and IRE1α's splicing of XBP1 mRNA. Furthermore, clinical efficacy of nicotinic agents in relieving neuropathic pain best correlated with their activity on α6ß4. Finally, BARP-knockout, but not NACHO-knockout mice lacked nicotine-induced antiallodynia, highlighting the functional importance of α6ß4 in pain. These results identify roles for IRE1α and BARP in neurotransmitter receptor assembly and unlock drug discovery for the previously elusive α6ß4 receptor.


Subject(s)
Cholinergic Agonists/pharmacology , Endoribonucleases/metabolism , Gene Expression Regulation/drug effects , Protein Serine-Threonine Kinases/metabolism , Receptors, Cholinergic/biosynthesis , Animals , Endoribonucleases/genetics , HEK293 Cells , Humans , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mice , Mice, Knockout , Protein Serine-Threonine Kinases/genetics , RNA Splicing/drug effects , Rats , Receptors, Cholinergic/genetics , X-Box Binding Protein 1/genetics
3.
Proc Natl Acad Sci U S A ; 117(39): 24534-24544, 2020 09 29.
Article in English | MEDLINE | ID: mdl-32929005

ABSTRACT

Auditory hair cells receive olivocochlear efferent innervation, which refines tonotopic mapping, improves sound discrimination, and mitigates acoustic trauma. The olivocochlear synapse involves α9α10 nicotinic acetylcholine receptors (nAChRs), which assemble in hair cells only coincident with cholinergic innervation and do not express in recombinant mammalian cell lines. Here, genome-wide screening determined that assembly and surface expression of α9α10 require ligand binding. Ion channel function additionally demands an auxiliary subunit, which can be transmembrane inner ear (TMIE) or TMEM132e. Both of these single-pass transmembrane proteins are enriched in hair cells and underlie nonsyndromic human deafness. Inner hair cells from TMIE mutant mice show altered postsynaptic α9α10 function and retain α9α10-mediated transmission beyond the second postnatal week associated with abnormally persistent cholinergic innervation. Collectively, this study provides a mechanism to link cholinergic input with α9α10 assembly, identifies unexpected functions for human deafness genes TMIE/TMEM132e, and enables drug discovery for this elusive nAChR implicated in prevalent auditory disorders.


Subject(s)
Deafness/metabolism , Hair Cells, Auditory/metabolism , Membrane Proteins/metabolism , Receptors, Nicotinic/metabolism , Animals , Cochlea/metabolism , Deafness/genetics , Humans , Ligands , Membrane Proteins/genetics , Mice , Protein Binding , Receptors, Nicotinic/genetics , Synapses/metabolism
4.
Nat Commun ; 11(1): 2799, 2020 06 03.
Article in English | MEDLINE | ID: mdl-32493979

ABSTRACT

Small molecule polyamines are abundant in all life forms and participate in diverse aspects of cell growth and differentiation. Spermidine/spermine acetyltransferase (SAT1) is the rate-limiting enzyme in polyamine catabolism and a primary genetic risk factor for suicidality. Here, using genome-wide screening, we find that SAT1 selectively controls nicotinic acetylcholine receptor (nAChR) biogenesis. SAT1 specifically augments assembly of nAChRs containing α7 or α4ß2, but not α6 subunits. Polyamines are classically studied as regulators of ion channel gating that engage the nAChR channel pore. In contrast, we find polyamine effects on assembly involve the nAChR cytosolic loop. Neurological studies link brain polyamines with neurodegenerative conditions. Our pharmacological and transgenic animal studies find that reducing polyamines enhances cortical neuron nAChR expression and augments nicotine-mediated neuroprotection. Taken together, we describe a most unexpected role for polyamines in regulating ion channel assembly, which provides a new avenue for nAChR neuropharmacology.


Subject(s)
Ion Channels/metabolism , Polyamines/metabolism , Receptors, Nicotinic/metabolism , Acetyltransferases , Animals , Biocatalysis , DNA, Complementary/genetics , Enhancer Elements, Genetic/genetics , Fluorescence , Genome, Human , HEK293 Cells , Humans , Ion Channel Gating , Mice , Neurons/metabolism , Neuroprotection/drug effects , Protein Structure, Secondary , Protein Subunits/chemistry , Protein Subunits/metabolism , Rats , Receptors, Nicotinic/chemistry
5.
Nat Commun ; 10(1): 2746, 2019 06 21.
Article in English | MEDLINE | ID: mdl-31227712

ABSTRACT

Nicotinic acetylcholine receptors (nAChRs) mediate and modulate synaptic transmission throughout the brain, and contribute to learning, memory, and behavior. Dysregulation of α7-type nAChRs in neuropsychiatric as well as immunological and oncological diseases makes them attractive targets for pharmaceutical development. Recently, we identified NACHO as an essential chaperone for α7 nAChRs. Leveraging the robust recombinant expression of α7 nAChRs with NACHO, we utilized genome-wide cDNA library screening and discovered that several anti-apoptotic Bcl-2 family proteins further upregulate receptor assembly and cell surface expression. These effects are mediated by an intracellular motif on α7 that resembles the BH3 binding domain of pro-apoptotic Bcl-2 proteins, and can be blocked by BH3 mimetic Bcl-2 inhibitors. Overexpression of Bcl-2 member Mcl-1 in neurons enhanced surface expression of endogenous α7 nAChRs, while a combination of chemotherapeutic Bcl2-inhibitors suppressed neuronal α7 receptor assembly. These results demonstrate that Bcl-2 proteins link α7 nAChR assembly to cell survival pathways.


Subject(s)
Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Neurons/metabolism , alpha7 Nicotinic Acetylcholine Receptor/metabolism , Amino Acid Motifs/genetics , Animals , Benzothiazoles/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , HEK293 Cells , Humans , Isoquinolines/pharmacology , Molecular Chaperones/metabolism , Mutation , Myeloid Cell Leukemia Sequence 1 Protein/antagonists & inhibitors , Neurons/drug effects , Nicotinic Agonists/pharmacology , Primary Cell Culture , Protein Binding/drug effects , Pyridines/pharmacology , Pyrimidines/pharmacology , Rats , Synaptic Transmission/drug effects , Thiophenes/pharmacology , Up-Regulation , alpha7 Nicotinic Acetylcholine Receptor/genetics
6.
Cell Rep ; 26(4): 866-874.e3, 2019 01 22.
Article in English | MEDLINE | ID: mdl-30673609

ABSTRACT

Acetylcholine gates a large family of nicotinic receptor cation channels that control neuronal excitation and neurotransmitter release. These receptors are key targets for neuropsychiatric disorders; however, difficulties in expressing nicotinic acetylcholine (nACh) receptors hamper elaboration of their pharmacology and obscure elucidation of their biological functions. Particularly intriguing are α6-containing nACh receptors, which mediate nicotine-induced dopamine release in striatum-nucleus accumbens. Using genome-wide cDNA screening, we identify three accessory proteins, ß-anchoring and -regulatory protein (BARP), lysosomal-associated membrane protein 5 (LAMP5), and SULT2B1, that complement the nACh receptor chaperone NACHO to reconstitute α6ß2ß3 channel function. Whereas NACHO mediates α6ß2ß3 assembly, BARP primarily enhances channel gating and LAMP5 and SULT2B1 promote receptor surface trafficking. BARP knockout mice show perturbations in presynaptic striatal nACh receptors that are consistent with BARP modulation of receptor desensitization. These studies unravel the molecular complexity of α6ß2ß3 biogenesis and enable physiological studies of this crucial neuropharmacological target.


Subject(s)
Corpus Striatum , Nucleus Accumbens/metabolism , Protein Multimerization , Receptors, Nicotinic/metabolism , Synaptic Transmission , Acetylcholine/genetics , Acetylcholine/metabolism , Animals , Corpus Striatum/metabolism , Lysosomal Membrane Proteins/genetics , Lysosomal Membrane Proteins/metabolism , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mice , Mice, Knockout , Organic Chemicals , Rats , Receptors, Nicotinic/genetics
7.
Neuron ; 96(5): 989-1001, 2017 Dec 06.
Article in English | MEDLINE | ID: mdl-29216460

ABSTRACT

Targeted therapy for neuropsychiatric disorders requires selective modulation of dysfunctional neuronal pathways. Receptors relevant to CNS disorders typically have associated proteins discretely expressed in specific neuronal pathways; these accessory proteins provide a new dimension for drug discovery. Recent studies show that targeting a TARP auxiliary subunit of AMPA receptors selectively modulates neuronal excitability in specific forebrain pathways relevant to epilepsy. Other medicinally important ion channels, gated by glutamate, γ-aminobutyric acid (GABA), and acetylcholine, also have associated proteins, which may be druggable. This emerging pharmacology of receptor-associated proteins provides a new approach for improving drug efficacy while mitigating side effects.


Subject(s)
Neuropharmacology , Receptors, Neurotransmitter/drug effects , Animals , Humans , Nerve Tissue Proteins/drug effects
8.
Cell Rep ; 19(4): 688-696, 2017 04 25.
Article in English | MEDLINE | ID: mdl-28445721

ABSTRACT

Neuronal nicotinic acetylcholine receptors (nAChRs) participate in diverse aspects of brain function and mediate behavioral and addictive properties of nicotine. Neuronal nAChRs derive from combinations of α and ß subunits, whose assembly is tightly regulated. NACHO was recently identified as a chaperone for α7-type nAChRs. Here, we find NACHO mediates assembly of all major classes of presynaptic and postsynaptic nAChR tested. NACHO acts at early intracellular stages of nAChR subunit assembly and then synergizes with RIC-3 for receptor surface expression. NACHO knockout mice show profound deficits in binding sites for α-bungarotoxin, epibatidine, and conotoxin MII, illustrating essential roles for NACHO in proper assembly of α7-, α4ß2-, and α6-containing nAChRs, respectively. By contrast, GABAA receptors are unaffected consistent with NACHO specifically modulating nAChRs. NACHO knockout mice show abnormalities in locomotor and cognitive behaviors compatible with nAChR deficiency and underscore the importance of this chaperone for physiology and disease associated with nAChRs.


Subject(s)
Brain/metabolism , Molecular Chaperones/metabolism , Receptors, Nicotinic/metabolism , Animals , Binding Sites , Bridged Bicyclo Compounds, Heterocyclic/chemistry , Bridged Bicyclo Compounds, Heterocyclic/metabolism , Bungarotoxins/chemistry , Bungarotoxins/metabolism , Cell Line , Cognitive Dysfunction/pathology , Conotoxins/chemistry , Conotoxins/metabolism , Humans , Iodine Radioisotopes/chemistry , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Knockout , Molecular Chaperones/genetics , Neurons/metabolism , Nicotine/chemistry , Nicotine/metabolism , Protein Binding , Protein Subunits/genetics , Protein Subunits/metabolism , Pyridines/chemistry , Pyridines/metabolism , Radiopharmaceuticals/chemistry , Radiopharmaceuticals/metabolism , Receptors, Nicotinic/genetics
9.
J Pharmacol Exp Ther ; 357(2): 394-414, 2016 May.
Article in English | MEDLINE | ID: mdl-26989142

ABSTRACT

Members of the α-amino-3-hydroxyl-5-methyl-4-isoxazole-propionic acid (AMPA) subtype of ionotropic glutamate receptors mediate the majority of fast synaptic transmission within the mammalian brain and spinal cord, representing attractive targets for therapeutic intervention. Here, we describe novel AMPA receptor modulators that require the presence of the accessory protein CACNG8, also known as transmembrane AMPA receptor regulatory protein γ8 (TARP-γ8). Using calcium flux, radioligand binding, and electrophysiological assays of wild-type and mutant forms of TARP-γ8, we demonstrate that these compounds possess a novel mechanism of action consistent with a partial disruption of the interaction between the TARP and the pore-forming subunit of the channel. One of the molecules, 5-[2-chloro-6-(trifluoromethoxy)phenyl]-1,3-dihydrobenzimidazol-2-one (JNJ-55511118), had excellent pharmacokinetic properties and achieved high receptor occupancy following oral administration. This molecule showed strong, dose-dependent inhibition of neurotransmission within the hippocampus, and a strong anticonvulsant effect. At high levels of receptor occupancy in rodent in vivo models, JNJ-55511118 showed a strong reduction in certain bands on electroencephalogram, transient hyperlocomotion, no motor impairment on rotarod, and a mild impairment in learning and memory. JNJ-55511118 is a novel tool for reversible AMPA receptor inhibition, particularly within the hippocampus, with potential therapeutic utility as an anticonvulsant or neuroprotectant. The existence of a molecule with this mechanism of action demonstrates the possibility of pharmacological targeting of accessory proteins, increasing the potential number of druggable targets.


Subject(s)
Benzimidazoles/therapeutic use , Calcium Channels/drug effects , Excitatory Amino Acid Antagonists/pharmacology , Receptors, AMPA/drug effects , Animals , Calcium Channels/genetics , Calcium Signaling/drug effects , Drug Design , Electroencephalography/drug effects , HEK293 Cells , Humans , Learning/drug effects , Memory/drug effects , Mice, Inbred C57BL , Mice, Knockout , Motor Activity/drug effects , Mutation/genetics , Neurons/drug effects , Postural Balance/drug effects , Rats, Sprague-Dawley , Receptors, AMPA/genetics
10.
Neuron ; 89(5): 948-55, 2016 Mar 02.
Article in English | MEDLINE | ID: mdl-26875622

ABSTRACT

Nicotine exerts its behavioral and additive actions through a family of brain nicotinic acetylcholine receptors (nAChRs). Enhancing α7-type nAChR signaling improves symptoms in Alzheimer's disease and schizophrenia. The pharmaceutical study of α7 receptors is hampered because these receptors do not form their functional pentameric structure in cell lines, and mechanisms that underlie α7 receptor assembly in neurons are not understood. Here, a genomic screening strategy solves this long-standing puzzle and identifies NACHO, a transmembrane protein of neuronal endoplasmic reticulum that mediates assembly of α7 receptors. NACHO promotes α7 protein folding, maturation through the Golgi complex, and expression at the cell surface. Knockdown of NACHO in cultured hippocampal neurons or knockout of NACHO in mice selectively and completely disrupts α7 receptor assembly and abolishes α7 channel function. This work identifies NACHO as an essential, client-specific chaperone for nAChRs and has implications for physiology and disease associated with these widely distributed neurotransmitter receptors.


Subject(s)
Hippocampus/metabolism , Neurons/physiology , Protein Subunits/metabolism , alpha7 Nicotinic Acetylcholine Receptor/metabolism , Acetylcholine/pharmacology , Action Potentials/drug effects , Action Potentials/genetics , Animals , Calnexin/metabolism , Glial Fibrillary Acidic Protein/genetics , Glial Fibrillary Acidic Protein/metabolism , Glutamic Acid/pharmacology , HEK293 Cells , Hippocampus/cytology , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Isoxazoles/pharmacology , Male , Membrane Proteins/drug effects , Membrane Proteins/metabolism , Mice , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Neurons/drug effects , Phenylurea Compounds/pharmacology , Protein Subunits/genetics , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Serotonin/pharmacology , alpha7 Nicotinic Acetylcholine Receptor/genetics
11.
Nat Neurosci ; 16(8): 1032-41, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23852113

ABSTRACT

Disrupted excitatory synapse maturation in GABAergic interneurons may promote neuropsychiatric disorders such as schizophrenia. However, establishing developmental programs for nascent synapses in GABAergic cells is confounded by their sparsity, heterogeneity and late acquisition of subtype-defining characteristics. We investigated synaptic development in mouse interneurons targeting cells by lineage from medial ganglionic eminence (MGE) or caudal ganglionic eminence (CGE) progenitors. MGE-derived interneuron synapses were dominated by GluA2-lacking AMPA-type glutamate receptors (AMPARs), with little contribution from NMDA-type receptors (NMDARs) throughout development. In contrast, CGE-derived cell synapses had large NMDAR components and used GluA2-containing AMPARs. In neonates, both MGE- and CGE-derived interneurons expressed primarily GluN2B subunit-containing NMDARs, which most CGE-derived interneurons retained into adulthood. However, MGE-derived interneuron NMDARs underwent a GluN2B-to-GluN2A switch that could be triggered acutely with repetitive synaptic activity. Our findings establish ganglionic eminence-dependent rules for early synaptic integration programs of distinct interneuron cohorts, including parvalbumin- and cholecystokinin-expressing basket cells.


Subject(s)
GABAergic Neurons/cytology , Gene Expression Regulation, Developmental , Hippocampus/cytology , Interneurons/cytology , Neocortex/cytology , Neuronal Plasticity , Receptors, AMPA/chemistry , Receptors, N-Methyl-D-Aspartate/chemistry , Telencephalon/embryology , Aging/metabolism , Animals , Animals, Newborn , Biomarkers , Cell Lineage , Excitatory Postsynaptic Potentials , Female , GABAergic Neurons/metabolism , Hippocampus/embryology , Hippocampus/growth & development , Interneurons/classification , Interneurons/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neocortex/embryology , Neocortex/growth & development , Nerve Tissue Proteins/analysis , Organ Specificity , Receptors, AMPA/analysis , Receptors, AMPA/biosynthesis , Receptors, AMPA/genetics , Receptors, N-Methyl-D-Aspartate/analysis , Receptors, N-Methyl-D-Aspartate/biosynthesis , Receptors, N-Methyl-D-Aspartate/genetics , Synaptic Transmission , Telencephalon/cytology
12.
Neuron ; 70(2): 339-51, 2011 Apr 28.
Article in English | MEDLINE | ID: mdl-21521618

ABSTRACT

In cerebral cortex there is a developmental switch from NR2B- to NR2A-containing NMDA receptors (NMDARs) driven by activity and sensory experience. This subunit switch alters NMDAR function, influences synaptic plasticity, and its dysregulation is associated with neurological disorders. However, the mechanisms driving the subunit switch are not known. Here, we show in hippocampal CA1 pyramidal neurons that the NR2B to NR2A switch driven acutely by activity requires activation of NMDARs and mGluR5, involves PLC, Ca(2+) release from IP(3)R-dependent stores, and PKC activity. In mGluR5 knockout mice the developmental NR2B-NR2A switch in CA1 is deficient. Moreover, in visual cortex of mGluR5 knockout mice, the NR2B-NR2A switch evoked in vivo by visual experience is absent. Thus, we establish that mGluR5 and NMDARs are required for the activity-dependent NR2B-NR2A switch and play a critical role in experience-dependent regulation of NMDAR subunit composition in vivo.


Subject(s)
Adaptation, Physiological/physiology , Pyramidal Cells/physiology , Receptors, Metabotropic Glutamate/metabolism , Receptors, N-Methyl-D-Aspartate/physiology , Synapses/physiology , Animals , Animals, Newborn , Electric Stimulation/methods , Enzyme Inhibitors/pharmacology , Estrenes/pharmacology , Excitatory Amino Acid Agonists/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Female , Hippocampus/cytology , In Vitro Techniques , Male , Mice , Mice, Knockout , Models, Biological , N-Methylaspartate/pharmacology , Piperidines/pharmacology , Pyramidal Cells/drug effects , Pyridines/pharmacology , Pyrrolidinones/pharmacology , Quinoxalines/pharmacology , Rats , Rats, Wistar , Receptor, Metabotropic Glutamate 5 , Receptors, Metabotropic Glutamate/genetics , Thiazoles/pharmacology , Time Factors , Visual Cortex/physiology
13.
Curr Pharm Biotechnol ; 12(1): 95-101, 2011 Jan 01.
Article in English | MEDLINE | ID: mdl-20932254

ABSTRACT

Transient Receptor Potential Vanilloid Type 1 is a prominent "pain" receptor expressed in sensory afferent neurons. TRPV1 on peripheral nerve terminals detects a variety of noxious stimuli generated at sites of injury and inflammation, and in turn, drives the excitation and sensitization of C-fibers neurons. Significantly, TRPV1 is also located on the central terminals of sensory neurons projecting to the spinal cord and brainstem. These TRPV1 channels appear to stimulate the secretion of glutamate. Further, TRPV1 is expressed diffusely in the brain and there is emerging evidence for TRPV1 modulating transmission at various brain synapses. Here we discuss our current understanding of the potential roles for TRPV1 in synaptic transmission.


Subject(s)
Ion Channel Gating , Neurons, Afferent/physiology , Nociceptors/physiology , Spinal Cord/physiopathology , Synaptic Transmission , TRPV Cation Channels/physiology , Animals , Humans , Sensory Receptor Cells/physiology , TRPV Cation Channels/agonists , TRPV Cation Channels/antagonists & inhibitors
14.
Neuron ; 67(6): 984-96, 2010 Sep 23.
Article in English | MEDLINE | ID: mdl-20869595

ABSTRACT

N-methyl-D-aspartate (NMDA) receptors (NMDARs) play a central role in development, synaptic plasticity, and neurological disease. NMDAR subunit composition defines their biophysical properties and downstream signaling. Casein kinase 2 (CK2) phosphorylates the NR2B subunit within its PDZ-binding domain; however, the consequences for NMDAR localization and function are unclear. Here we show that CK2 phosphorylation of NR2B regulates synaptic NR2B and NR2A in response to activity. We find that CK2 phosphorylates NR2B, but not NR2A, to drive NR2B-endocytosis and remove NR2B from synapses resulting in an increase in synaptic NR2A expression. During development there is an activity-dependent switch from NR2B to NR2A at cortical synapses. We observe an increase in CK2 expression and NR2B phosphorylation over this same critical period and show that the acute activity-dependent switch in NR2 subunit composition at developing hippocampal synapses requires CK2 activity. Thus, CK2 plays a central role in determining the NR2 subunit content of synaptic NMDARs.


Subject(s)
Casein Kinase II/physiology , Neurons/metabolism , Protein Subunits/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Synapses/metabolism , Adenosine Triphosphate/pharmacokinetics , Amino Acid Sequence , Animals , Animals, Newborn , Benzimidazoles/pharmacology , Biotinylation/methods , Cells, Cultured , Cerebral Cortex/cytology , Disks Large Homolog 4 Protein , Embryo, Mammalian , Endocytosis/genetics , Excitatory Amino Acid Antagonists/pharmacology , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/genetics , Gene Expression Regulation/genetics , Green Fluorescent Proteins/genetics , Hippocampus/cytology , Hippocampus/metabolism , Humans , Immunoprecipitation/methods , In Vitro Techniques , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Models, Biological , Neurons/drug effects , PDZ Domains/physiology , Patch-Clamp Techniques , Phosphorus Isotopes/pharmacokinetics , Phosphorylation/drug effects , Phosphorylation/genetics , Piperidines/pharmacology , Protein Subunits/genetics , Rats , Rats, Sprague-Dawley , Rats, Wistar , Receptors, N-Methyl-D-Aspartate/genetics , Sodium Channel Blockers/pharmacology , Subcellular Fractions/drug effects , Subcellular Fractions/metabolism , Synaptophysin/metabolism , Tetrodotoxin/pharmacology , Transfection/methods , Tyrosine/metabolism
15.
Nat Neurosci ; 13(10): 1199-207, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20802490

ABSTRACT

Trafficking of AMPA receptors (AMPARs) is important for many forms of synaptic plasticity. However, the link between activity and resulting synaptic alterations is not fully understood. We identified a direct interaction between N-ethylmaleimide-sensitive fusion protein (NSF), an ATPase involved in membrane fusion events and stabilization of surface AMPARs, and Polo-like kinase- 2 (Plk2), an activity-inducible kinase that homeostatically decreases excitatory synapse number and strength. Plk2 disrupted the interaction of NSF with the GluA2 subunit of AMPARs, promoting extensive loss of surface GluA2 in rat hippocampal neurons, greater association of GluA2 with adaptor proteins PICK1 and GRIP1, and decreased synaptic AMPAR current. Plk2 engagement of NSF, but not Plk2 kinase activity, was required for this mechanism and occurred through a motif in the Plk2 protein that was independent of the canonical polo box interaction sites. These data reveal that heightened synaptic activity, acting through Plk2, leads to homeostatic decreases in surface AMPAR expression via the direct dissociation of NSF from GluA2.


Subject(s)
Homeostasis/physiology , N-Ethylmaleimide-Sensitive Proteins/metabolism , Neurons/metabolism , Protein Serine-Threonine Kinases/metabolism , Receptors, AMPA/metabolism , Adenosine Triphosphate/pharmacology , Amino Acid Motifs/genetics , Amino Acid Motifs/physiology , Animals , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cells, Cultured , Chlorocebus aethiops , Cytoskeletal Proteins , Embryo, Mammalian , Endocytosis/drug effects , Endocytosis/genetics , GABA Antagonists/pharmacology , Gene Expression Regulation/drug effects , Green Fluorescent Proteins/genetics , Hippocampus/cytology , Homeostasis/genetics , Humans , Immunoprecipitation/methods , Intracellular Signaling Peptides and Proteins , Microscopy, Confocal/methods , N-Ethylmaleimide-Sensitive Proteins/genetics , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurons/drug effects , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Picrotoxin/pharmacology , Protein Binding/drug effects , Protein Binding/physiology , Protein Serine-Threonine Kinases/genetics , Protein Transport , RNA Interference/physiology , Rats , Synapses/drug effects , Synapses/metabolism , Time Factors , Transfection/methods
16.
Curr Biol ; 19(11): 900-8, 2009 Jun 09.
Article in English | MEDLINE | ID: mdl-19481459

ABSTRACT

BACKGROUND: Ionotropic glutamate receptors (iGluRs) are glutamate-gated ion channels that mediate excitatory neurotransmission in the central nervous system. Based on both molecular and pharmacological criteria, iGluRs have been divided into two major classes, the non-NMDA class, which includes both AMPA and kainate subtypes of receptors, and the NMDA class. One evolutionarily conserved feature of iGluRs is their desensitization in the continued presence of glutamate. Thus, when in a desensitized state, iGluRs can be bound to glutamate, yet the channel remains closed. However, the relevance of desensitization to nervous system function has remained enigmatic. RESULTS: Here, we report the identification and characterization of a novel polypeptide (con-ikot-ikot) from the venom of a predatory marine snail Conus striatus that specifically disrupts the desensitization of AMPA receptors (AMPARs). The stoichiometry of con-ikot-ikot appears reminiscent of the proposed subunit organization of AMPARs, i.e., a dimer of dimers, suggesting that it acts as a molecular four-legged clamp that holds the AMPAR channel open. Application of con-ikot-ikot to hippocampal slices caused a large and rapid increase in resting AMPAR-mediated current leading to neuronal death. CONCLUSIONS: Our findings provide insight into the mechanisms that regulate receptor desensitization and demonstrate that in the arms race between prey and predators, evolution has selected for a toxin that blocks AMPAR desensitization, thus revealing the fundamental importance of desensitization for regulating neural function.


Subject(s)
Conus Snail/metabolism , Mollusk Venoms/chemistry , Neurotoxins/pharmacology , Peptides/pharmacology , Receptors, AMPA/metabolism , Animals , Benzothiadiazines/pharmacology , Binding Sites , Chemical Fractionation , Chromatography, High Pressure Liquid , Conus Snail/chemistry , Dimerization , Electric Conductivity , Hippocampus/drug effects , Neurotoxins/chemistry , Neurotoxins/isolation & purification , Patch-Clamp Techniques , Peptides/chemistry , Peptides/isolation & purification , Rats , Receptors, AMPA/chemistry , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Recombinant Proteins/pharmacology , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Xenopus
17.
Mol Pharmacol ; 74(5): 1261-8, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18689441

ABSTRACT

General anesthetics (GAs) are central nervous system depressants that render patients unresponsive to external stimuli. In contrast, many of these agents are also known to stimulate peripheral sensory nerves, raising the possibility that they may exacerbate tissue inflammation. We have found that pungent GAs excite sensory neurons by directly activating the transient receptor potential (TRP) A1 ion channel. Here, we show that GAs also sensitize the capsaicin receptor TRPV1, a key ion channel expressed in nociceptive neurons. Clinically relevant concentrations of isoflurane, sevoflurane, enflurane, and desflurane sensitize TRPV1 to capsaicin and protons and reduce the threshold for heat activation. Furthermore, isoflurane directly activates TRPV1 after stimulation of protein kinase C. Likewise, isoflurane excites TRPV1 and sensory neurons during concomitant application of bradykinin, a key inflammatory mediator formed during tissue injury. Thus, GAs can enhance the activation of TRPV1 that occurs during surgically induced tissue damage. These results support the hypothesis that some GAs, through direct actions at TRP channels, increase postsurgical pain and inflammation.


Subject(s)
Anesthetics, General/pharmacology , TRPV Cation Channels/drug effects , Animals , Bradykinin/physiology , Capsaicin/pharmacology , Cell Line , Female , Humans , Mice , Patch-Clamp Techniques , Protein Kinase C/metabolism , Xenopus laevis
18.
Proc Natl Acad Sci U S A ; 105(25): 8784-9, 2008 Jun 24.
Article in English | MEDLINE | ID: mdl-18574153

ABSTRACT

General anesthetics (GAs) have transformed surgery through their actions to depress the central nervous system and blunt the perception of surgical insults. Counterintuitively, many of these agents activate peripheral nociceptive neurons. However, the underlying mechanisms and significance of these effects have not been explored. Here, we show that clinical concentrations of noxious i.v. and inhalation GAs excite sensory neurons by selectively activating TRPA1, a key ion channel in the pain pathway. Further, these GAs induce pain-related responses in mice that are abolished in TRPA1-null animals. Significantly, TRPA1-dependent neurogenic inflammation is greater in mice anesthetized with pungent compared with nonpungent anesthetics. Thus, our results show that TRPA1 is essential for sensing noxious GAs. The pronociceptive effects of GAs combined with surgical tissue damage could lead to a paradoxical increase in postoperative pain and inflammation.


Subject(s)
Anesthetics, General/pharmacology , Calcium Channels/metabolism , Inflammation/physiopathology , Isoflurane/pharmacology , Pain/physiopathology , Transient Receptor Potential Channels/metabolism , Animals , Ankyrins , Humans , Inflammation/metabolism , Mice , Pain/metabolism , Rats , TRPA1 Cation Channel , TRPC Cation Channels , TRPM Cation Channels/metabolism , TRPV Cation Channels/metabolism
19.
J Physiol ; 585(Pt 2): 469-82, 2007 Dec 01.
Article in English | MEDLINE | ID: mdl-17932142

ABSTRACT

TRPV1 and TRPM8 are sensory nerve ion channels activated by heating and cooling, respectively. A variety of physical and chemical stimuli activate these receptors in a synergistic manner but the underlying mechanisms are unclear. Both channels are voltage sensitive, and temperature and ligands modulate this voltage dependence. Thus, a voltage-sensing mechanism has become an attractive model to explain the generalized gating of these and other thermo-sensitive TRP channels. We show here using whole-cell and single channel measurements that voltage produces only a partial activation of TRPV1 and TRPM8. At room temperature (20-25 degrees C) membrane depolarization evokes responses that saturate at approximately 50-60% of the maximum open probability. Furthermore, high concentrations of capsaicin (10 microm), resiniferatoxin (5 microm) and menthol (6 mm) reveal voltage-independent gating. Similarly, other modes of TRPV1 regulation including heat, protein kinase C-dependent phosphorylation, and protons enhance both the efficacy and sensitivity of voltage activation. In contrast, the TRPV1 antagonist capsazepine produces the opposite effects. These data can be explained by an allosteric model in which voltage, temperature, agonists and inverse agonists are independently coupled, either positively or negatively, to channel gating. Thus, voltage acts separately but in concert with other stimuli to regulate channel activation, and, therefore, a voltage-sensitive mechanism is unlikely to represent a final, gating mechanism for these channels.


Subject(s)
Hot Temperature , Ion Channel Gating/physiology , Neurons, Afferent/physiology , Protein Kinase C/metabolism , TRPM Cation Channels/physiology , TRPV Cation Channels/physiology , Allosteric Regulation , Animals , Capsaicin/analogs & derivatives , Capsaicin/pharmacology , Cell Line , Diterpenes/pharmacology , Electric Stimulation , Humans , Ion Channel Gating/drug effects , Kidney/cytology , Membrane Potentials/drug effects , Membrane Potentials/physiology , Models, Chemical , Rats , Sensory System Agents/pharmacology , TRPM Cation Channels/chemistry , TRPV Cation Channels/chemistry
20.
J Neurophysiol ; 98(5): 2693-704, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17804581

ABSTRACT

Excitatory postsynaptic currents (EPSCs) from dorsolateral medium spiny neurons (MSNs) were recorded in cortico-striatal slice preparations from postnatal day 6-8 (P6-8) and >P12 wild-type mice and mice that were lacking either the NR2A or the NR2C subunit of the N-methyl-D-aspartate (NMDA) receptor. EPSCs were elicited by stimulation of the excitatory afferents and the NMDA and non-NMDA receptor-mediated components were pharmacologically isolated. The ratio of these components decreased with development and was significantly reduced only between age-matched +/+ and NR2A -/- neurons. In many MSNs, the NMDA-EPSC decay was characterized by the presence of a slow exponential component with a time constant lasting >1 s regardless of genotype or age. In the NR2A -/-, no developmental increase in the decay time (Tw) of the NMDA-EPSCs was observed although it was almost twofold longer than in +/+ MSNs. NR1/NR2B antagonists were ineffective in reducing the slow NMDA-EPSCs at all ages. Input-output studies revealed differences in stimulation threshold sensitivity of MSNs based on stimulus location. High-threshold responders were preferentially identified with stimulation from intracortical locations that produced considerably faster NMDA-EPSCs, whereas low-threshold responders were mainly elicited with stimulation more proximal to the striatum and exhibited slower NMDA-EPSCs. A low-affinity competitive antagonist of NMDA receptors failed to alter the decay of NMDA-EPSCs elicited from either location, suggesting that glutamate spillover is not responsible for the long-lasting NMDA-EPSCs. Our data are consistent with the expression of a unique NMDA receptor complex in MSNs with very slow deactivation kinetics.


Subject(s)
Corpus Striatum/cytology , Excitatory Postsynaptic Potentials/physiology , Neurons/physiology , Receptors, N-Methyl-D-Aspartate/physiology , 6-Cyano-7-nitroquinoxaline-2,3-dione/pharmacology , Age Factors , Animals , Animals, Newborn , Dose-Response Relationship, Radiation , Electric Stimulation , Excitatory Amino Acid Agonists/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/genetics , Excitatory Postsynaptic Potentials/radiation effects , In Vitro Techniques , Mice , Mice, Inbred C57BL , Mice, Knockout , N-Methylaspartate/pharmacology , Neurons/classification , Patch-Clamp Techniques/methods , Receptors, N-Methyl-D-Aspartate/deficiency , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...