Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 63
Filter
1.
Brain ; 143(1): 161-174, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31800012

ABSTRACT

Inborn errors of CACNA1A-encoded P/Q-type calcium channels impair synaptic transmission, producing early and lifelong neurological deficits, including childhood absence epilepsy, ataxia and dystonia. Whether these impairments owe their pathologies to defective channel function during the critical period for thalamic network stabilization in immature brain remains unclear. Here we show that mice with tamoxifen-induced adult-onset ablation of P/Q channel alpha subunit (iKOp/q) display identical patterns of dysfunction, replicating the inborn loss-of-function phenotypes and, therefore demonstrate that these neurological defects do not rely upon developmental abnormality. Unexpectedly, unlike the inborn model, the adult-onset pattern of excitability changes believed to be pathogenic within the thalamic network is non-canonical. Specifically, adult ablation of P/Q channels does not promote Cacna1g-mediated burst firing or T-type calcium current (IT) in the thalamocortical relay neurons; however, burst firing in thalamocortical relay neurons remains essential as iKOp/q mice generated on a Cacna1g deleted background show substantially diminished seizure generation. Moreover, in thalamic reticular nucleus neurons, burst firing is impaired accompanied by attenuated IT. Interestingly, inborn deletion of thalamic reticular nucleus-enriched, human childhood absence epilepsy-linked gene Cacna1h in iKOp/q mice reduces thalamic reticular nucleus burst firing and promotes rather than reduces seizure, indicating an epileptogenic role for loss-of-function Cacna1h gene variants reported in human childhood absence epilepsy cases. Together, our results demonstrate that P/Q channels remain critical for maintaining normal thalamocortical oscillations and motor control in the adult brain, and suggest that the developmental plasticity of membrane currents regulating pathological rhythmicity is both degenerate and age-dependent.


Subject(s)
Ataxia/genetics , Calcium Channels, N-Type/genetics , Cerebral Cortex/metabolism , Epilepsy, Absence/genetics , Neurons/metabolism , Thalamus/metabolism , Action Potentials , Age Factors , Animals , Ataxia/metabolism , Ataxia/physiopathology , Calcium Channels, T-Type/genetics , Calcium Channels, T-Type/metabolism , Cerebral Cortex/physiopathology , Disease Models, Animal , Epilepsy, Absence/metabolism , Epilepsy, Absence/physiopathology , Excitatory Postsynaptic Potentials/genetics , Inhibitory Postsynaptic Potentials/genetics , Membrane Potentials/genetics , Mice , Mice, Knockout , Patch-Clamp Techniques , Thalamic Nuclei/cytology , Thalamus/physiopathology
2.
Nat Neurosci ; 21(10): 1392-1403, 2018 10.
Article in English | MEDLINE | ID: mdl-30258239

ABSTRACT

Multiple sclerosis (MS) patients exhibit neuropsychological symptoms in early disease despite the immune attack occurring predominantly in white matter and spinal cord. It is unclear why neurodegeneration may start early in the disease and is prominent in later stages. We assessed cortical microcircuit activity by employing spiking-specific two-photon Ca2+ imaging in proteolipid protein-immunized relapsing-remitting SJL/J mice in vivo. We identified the emergence of hyperactive cortical neurons in remission only, independent of direct immune-mediated damage and paralleled by elevated anxiety. High levels of neuronal activity were accompanied by increased caspase-3 expression. Cortical TNFα expression was mainly increased by excitatory neurons in remission; blockade with intraventricular infliximab restored AMPA spontaneous excitatory postsynaptic current frequencies, completely recovered normal neuronal network activity patterns and alleviated elevated anxiety. This suggests a dysregulation of cortical networks attempting to achieve functional compensation by synaptic plasticity mechanisms, indicating a link between immune attack and early start of neurodegeneration.


Subject(s)
Cerebral Cortex/physiopathology , Encephalomyelitis, Autoimmune, Experimental/complications , Encephalomyelitis, Autoimmune, Experimental/pathology , Hyperkinesis/etiology , Recovery of Function/physiology , Animals , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Carbazoles/therapeutic use , Cells, Cultured , Cerebral Cortex/ultrastructure , Cuprizone/toxicity , Disease Models, Animal , Egtazic Acid/analogs & derivatives , Egtazic Acid/pharmacokinetics , Encephalomyelitis, Autoimmune, Experimental/chemically induced , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Excitatory Amino Acid Antagonists/pharmacology , Female , Freund's Adjuvant/toxicity , Membrane Potentials/drug effects , Membrane Potentials/genetics , Mice , Mice, Transgenic , Microglia/pathology , Myelin Proteolipid Protein/toxicity , Peptide Fragments/toxicity , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-fos/metabolism , Quinoxalines/pharmacology , Sodium Channel Blockers/pharmacology , Tetrodotoxin/pharmacology
3.
Gene ; 641: 25-34, 2018 Jan 30.
Article in English | MEDLINE | ID: mdl-29032150

ABSTRACT

Schizophrenia (SCZ) is a chronic debilitating neuropsychiatric disorder with multiple risk factors involving numerous complex genetic influences. We examined and updated a master list of clinically relevant and susceptibility genes associated with SCZ reported in the literature and genomic databases dedicated to gene discovery for characterization of SCZ genes. We used the commercially available GeneAnalytics computer-based gene analysis program and integrated genomic databases to create a molecular profile of the updated list of 608 SCZ genes to model their impact in select categories (tissues and cells, diseases, pathways, biological processes, molecular functions, phenotypes and compounds) using specialized GeneAnalytics algorithms. Genes for schizophrenia were predominantly expressed in the cerebellum, cerebral cortex, medulla oblongata, thalamus and hypothalamus. Psychiatric/behavioral disorders incorporating SCZ genes included ADHD, bipolar disorder, autism spectrum disorder and alcohol dependence as well as cancer, Alzheimer's and Parkinson's disease, sleep disturbances and inflammation. Function based analysis of major biological pathways and mechanisms associated with SCZ genes identified glutaminergic receptors (e.g., GRIA1, GRIN2, GRIK4, GRM5), serotonergic receptors (e.g., HTR2A, HTR2C), GABAergic receptors (e.g., GABRA1, GABRB2), dopaminergic receptors (e.g., DRD1, DRD2), calcium-related channels (e.g., CACNA1H, CACNA1B), solute transporters (e.g., SLC1A1, SLC6A2) and for neurodevelopment (e.g., ADCY1, MEF2C, NOTCH2, SHANK3). Biological mechanisms involving synaptic transmission, regulation of membrane potential and transmembrane ion transport were identified as leading molecular functions associated with SCZ genes. Our approach to interrogate SCZ genes and their interactions at various levels has increased our knowledge and insight into the disease process possibly opening new avenues for therapeutic intervention.


Subject(s)
Genome-Wide Association Study , Ion Transport/genetics , Membrane Potentials/genetics , Schizophrenia/genetics , Synaptic Transmission/genetics , Amino Acid Transport Systems/genetics , Calcium Channels/genetics , Cerebellum/cytology , Cerebral Cortex/cytology , Databases, Genetic , Humans , Hypothalamus/cytology , Medulla Oblongata/cytology , Receptors, Dopamine/genetics , Receptors, GABA-A/genetics , Receptors, Ionotropic Glutamate/genetics , Receptors, Serotonin/genetics , Thalamus/cytology
4.
Biol Psychiatry ; 84(1): 55-64, 2018 07 01.
Article in English | MEDLINE | ID: mdl-29174591

ABSTRACT

BACKGROUND: Long-term synaptic plasticity is a basic ability of the brain to dynamically adapt to external stimuli and regulate synaptic strength and ultimately network function. It is dysregulated by behavioral stress in animal models of depression and in humans with major depressive disorder. Antidepressants have been shown to restore disrupted synaptic plasticity in both animal models and humans; however, the underlying mechanism is unclear. METHODS: We examined modulation of synaptic plasticity by selective serotonin reuptake inhibitors (SSRIs) in hippocampal brain slices from wild-type rats and serotonin transporter (SERT) knockout mice. Recombinant voltage-gated calcium (Ca2+) channels in heterologous expression systems were used to determine the modulation of Ca2+ channels by SSRIs. We tested the behavioral effects of SSRIs in the chronic behavioral despair model of depression both in the presence and in the absence of SERT. RESULTS: SSRIs selectively inhibited hippocampal long-term depression. The inhibition of long-term depression by SSRIs was mediated by a direct block of voltage-activated L-type Ca2+ channels and was independent of SERT. Furthermore, SSRIs protected both wild-type and SERT knockout mice from behavioral despair induced by chronic stress. Finally, long-term depression was facilitated in animals subjected to the behavioral despair model, which was prevented by SSRI treatment. CONCLUSIONS: These results showed that antidepressants protected synaptic plasticity and neuronal circuitry from the effects of stress via a modulation of Ca2+ channels and synaptic plasticity independent of SERT. Thus, L-type Ca2+ channels might constitute an important signaling hub for stress response and for pathophysiology and treatment of depression.


Subject(s)
Antidepressive Agents/therapeutic use , Calcium Channels, L-Type/metabolism , RNA-Binding Proteins/metabolism , Stress, Psychological/drug therapy , Synaptic Transmission/drug effects , Age Factors , Animals , CHO Cells , Cadmium Chloride/pharmacology , Calcium Channel Blockers/pharmacology , Calcium Channels, L-Type/genetics , Cricetulus , Disease Models, Animal , Electric Stimulation , Female , Fluvoxamine/therapeutic use , HEK293 Cells , Hindlimb Suspension/psychology , Hippocampus/cytology , Humans , In Vitro Techniques , Male , Membrane Potentials/drug effects , Membrane Potentials/genetics , Nifedipine/pharmacology , Paroxetine/pharmacology , Patch-Clamp Techniques , Piperazines/pharmacology , Pyridines/pharmacology , RNA-Binding Proteins/genetics , Rats , Rats, Transgenic , Rats, Wistar , Serotonin/pharmacology , Serotonin Antagonists/pharmacology , Selective Serotonin Reuptake Inhibitors/therapeutic use , Stress, Psychological/genetics , Swimming/psychology , Synaptic Transmission/genetics , Transfection
5.
Invert Neurosci ; 18(1): 1, 2017 Nov 28.
Article in English | MEDLINE | ID: mdl-29185074

ABSTRACT

Xenopus laevis oocytes have been extensively used as a heterologous expression system for the study of ion channels. While used successfully worldwide as tool for expressing and characterizing ion channels from a wide range of species, the limited longevity of oocytes once removed from the animal can pose significant challenges. In this study, we evaluate a simple and useful method that extends the longevity of Xenopus oocytes after removal from the animal and quantitatively assessed the reliability of the electrophysiological date obtained. The receptor used for this study was the UNC-49 receptor originally isolated from the sheep parasite, Haemonchus contortus. Overall, we found that immediate storage of the ovary in supplemented ND96 storage buffer at 4 °C could extend their use for up to 17 days with almost 80% providing reliable electrophysiological data. This means that a single extraction can provide at least 3 weeks of experiments. In addition, we examined 24-day-old oocytes (week 4) extracted from a single frog and also obtained reliable data using the same approach. However, 50% of these oocytes were usable for full dose-response experiments. Overall, we did find that this method has the potential to significantly extend the use of single oocyte extractions for two-electrode voltage clamp electrophysiology.


Subject(s)
Ligand-Gated Ion Channels/metabolism , Longevity/physiology , Animals , Biophysics , Dose-Response Relationship, Drug , Electric Stimulation , Female , Helminth Proteins/genetics , Helminth Proteins/metabolism , Ligand-Gated Ion Channels/drug effects , Ligand-Gated Ion Channels/genetics , Longevity/drug effects , Longevity/genetics , Membrane Potentials/drug effects , Membrane Potentials/genetics , Microinjections , Oocytes , Patch-Clamp Techniques , Receptors, GABA-A/genetics , Receptors, GABA-A/metabolism , Time Factors , Xenopus laevis , gamma-Aminobutyric Acid/pharmacology
6.
Glia ; 65(12): 2038-2050, 2017 12.
Article in English | MEDLINE | ID: mdl-28856727

ABSTRACT

Over- and underexposure to cholesterol activates glia in neurodegenerative brain and retinal diseases but the molecular targets of cholesterol in glial cells are not known. Here, we report that disruption of unesterified membrane cholesterol content modulates the transduction of chemical, mechanical and temperature stimuli in mouse Müller cells. Activation of TRPV4 (transient receptor potential vanilloid type 4), a nonselective polymodal cation channel was studied following the removal or supplementation of cholesterol using the methyl-beta cyclodextrin (MßCD) delivery vehicle. Cholesterol extraction disrupted lipid rafts and caveolae without affecting TRPV4 trafficking or membrane localization protein. However, MßCD suppressed agonist (GSK1016790A)- and temperature-evoked elevations in [Ca2+ ]i , and suppressed transcellular propagation of Ca2+ waves. Lowering the free membrane cholesterol content markedly prolonged the time-course of the glial swelling response, whereas MßCD:cholesterol supplementation enhanced agonist- and temperature-induced Ca2+ signals and shortened the swelling response. Taken together, these data show that membrane cholesterol modulates polymodal transduction of agonists, swelling and temperature stimuli in retinal radial glia and suggest that dyslipidemic retinas might be associated with abnormal glial transduction of ambient sensory inputs.


Subject(s)
Cholesterol/metabolism , Ependymoglial Cells/metabolism , Signal Transduction/physiology , Animals , Calcium/metabolism , Caveolin 1/genetics , Caveolin 1/metabolism , Cells, Cultured , Cholesterol/pharmacology , Ependymoglial Cells/drug effects , Female , Leucine/analogs & derivatives , Leucine/pharmacology , Male , Membrane Microdomains , Membrane Potentials/drug effects , Membrane Potentials/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Patch-Clamp Techniques , Retina/cytology , Signal Transduction/genetics , Sulfonamides/pharmacology , TRPV Cation Channels/agonists , TRPV Cation Channels/genetics , TRPV Cation Channels/metabolism , Temperature
7.
J Comp Neurol ; 525(18): 3809-3820, 2017 Dec 15.
Article in English | MEDLINE | ID: mdl-28608460

ABSTRACT

The amount, quality, and diurnal pattern of sleep change greatly during development. Developmental changes of sleep/wake architecture are in a close relationship to brain development. The fragmentation of wake episodes is one of the salient features in the neonatal period, which is also observed in mature animals and human individuals lacking neuropeptide orexin/hypocretin signaling. This raises the possibility that developmental changes of lateral hypothalamic orexin neurons are relevant to the development of sleep/wake architecture. However, little information is available on morphological and physiological features of developing orexin neurons. To address the cellular basis for maturation of the sleep/wake regulatory system, we investigated the functional development of orexin neurons in the lateral hypothalamus. The anatomical development as well as the changes in the electrophysiological characteristics of orexin neurons was examined from embryonic to postnatal stages in orexin-EGFP mice. Prepro-orexin promoter activity was detectable at embryonic day (E) 12.0, followed by expression of orexin A after E14.0. The number of orexin neurons and their membrane capacitance reached similar levels to adults by postnatal day (P) 7, while their membrane potentials, firing rates, and action potential waveforms were developed by P21. The hyperpolarizing effect of serotonin, which is a major inhibitory signal for adult orexin neurons, was detected after E18.0 and matured at P1. These results suggest that the expression of orexin peptides precedes the maturation of electrophysiological activity of orexin neurons. The function of orexin neurons gradually matures by 3 weeks after birth, coinciding with maturation of sleep/wake architecture.


Subject(s)
Gene Expression Regulation, Developmental/genetics , Hypothalamus , Neurons/physiology , Orexins/metabolism , Age Factors , Analysis of Variance , Animals , Animals, Newborn , Electric Stimulation , Embryo, Mammalian , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Hypothalamus/cytology , Hypothalamus/embryology , Hypothalamus/growth & development , In Vitro Techniques , Membrane Potentials/genetics , Mice , Mice, Transgenic , Neurons/metabolism , Orexins/genetics , Patch-Clamp Techniques , Serotonin/pharmacology , Synaptic Transmission/drug effects , Synaptic Transmission/genetics , Synaptic Transmission/physiology
8.
Front Neural Circuits ; 10: 31, 2016.
Article in English | MEDLINE | ID: mdl-27199672

ABSTRACT

Mutation of the metabotropic glutamate receptor type 7 (mGlu7) induces absence-like epileptic seizures, but its precise role in the somatosensory thalamocortical network remains unknown. By combining electrophysiological recordings, optogenetics, and pharmacology, we dissected the contribution of the mGlu7 receptor at mouse thalamic synapses. We found that mGlu7 is functionally expressed at both glutamatergic and GABAergic synapses, where it can inhibit neurotransmission and regulate short-term plasticity. These effects depend on the PDZ-ligand of the receptor, as they are lost in mutant mice. Interestingly, the very low affinity of mGlu7 receptors for glutamate raises the question of how it can be activated, namely at GABAergic synapses and in basal conditions. Inactivation of the receptor activity with the mGlu7 negative allosteric modulator (NAM), ADX71743, enhances thalamic synaptic transmission. In vivo administration of the NAM induces a lethargic state with spindle and/or spike-and-wave discharges accompanied by a behavioral arrest typical of absence epileptic seizures. This provides evidence for mGlu7 receptor-mediated tonic modulation of a physiological function in vivo preventing synchronous and potentially pathological oscillations.


Subject(s)
Cerebral Cortex/cytology , Neural Pathways/physiology , Receptors, Metabotropic Glutamate/metabolism , Thalamus/physiology , Animals , Benzoxazoles/chemistry , Benzoxazoles/pharmacology , Cerebral Cortex/physiology , Channelrhodopsins , Excitatory Amino Acid Agents/pharmacology , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/genetics , GABA Agents/pharmacology , GTP-Binding Protein alpha Subunits, Gq-G11/genetics , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , HEK293 Cells , Humans , In Vitro Techniques , Membrane Potentials/drug effects , Membrane Potentials/genetics , Mice , Mice, Transgenic , Mutation/genetics , Neurons/drug effects , Neurons/physiology , Post-Synaptic Density/drug effects , Post-Synaptic Density/genetics , Receptors, GABA-A/physiology , Receptors, Metabotropic Glutamate/genetics , Synaptic Potentials/drug effects , Synaptic Potentials/genetics
9.
J Alzheimers Dis ; 53(1): 273-87, 2016 05 04.
Article in English | MEDLINE | ID: mdl-27163808

ABSTRACT

Because traditional approaches to drug development for Alzheimer's disease are becoming increasingly expensive and in many cases disappointingly unsuccessful, alternative approaches are required to shift the paradigm. Following leads from investigations of dihydropyridine calcium channel blockers, we observed unique properties from a class of functionalized naphthyridines and sought to develop these as novel therapeutics that minimize amyloid pathology without the adverse effects associated with current therapeutics. Our data show methyl 2,4-dimethyl-5-oxo-5,6-dihydrobenzo[c][2,7]naphthyridine-1-carboxylate (BNC-1) significantly decreases amyloid burden in a well-established mouse model of amyloid pathology through a unique mechanism mediated by Elk-1, a transcriptional repressor of presenilin-1. Additionally, BNC-1 treatment leads to increased levels of synaptophysin and synapsin, markers of synaptic integrity, but does not adversely impact presenilin-2 or processing of Notch-1, thus avoiding negative off target effects associated with pan-gamma secretase inhibition. Overall, our data show BNC-1 significantly decreases amyloid burden and improves markers of synaptic integrity in a well-established mouse model of amyloid deposition by promoting phosphorylation and activation of Elk-1, a transcriptional repressor of presenilin-1 but not presenilin-2. These data suggest BNC-1 might be a novel, disease-modifying therapeutic that will alter the pathogenesis of Alzheimer's disease.


Subject(s)
Alzheimer Disease/pathology , Amyloid/metabolism , Antipsychotic Agents/therapeutic use , Gene Expression Regulation/drug effects , Alzheimer Disease/drug therapy , Alzheimer Disease/genetics , Amyloid/drug effects , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Antipsychotic Agents/chemistry , Calcium Channel Blockers/therapeutic use , Cell Line, Tumor , Disease Models, Animal , Electric Stimulation , Female , Gene Expression Regulation/genetics , Humans , Male , Maze Learning/drug effects , Membrane Potentials/drug effects , Membrane Potentials/genetics , Mice , Mice, Transgenic , Mutation/genetics , Naphthyridines/pharmacology , Naphthyridines/therapeutic use , Neuroblastoma/pathology , Nifedipine/therapeutic use , Patch-Clamp Techniques , Peptide Fragments/metabolism , Presenilin-1/genetics , Presenilin-1/metabolism , Presenilin-2/metabolism , Receptor, Notch1/metabolism , Synapsins/metabolism , Synaptophysin/metabolism , Transfection
10.
J Neurosci ; 36(11): 3350-62, 2016 Mar 16.
Article in English | MEDLINE | ID: mdl-26985042

ABSTRACT

The hypothalamo-pituitary-adrenocortical (HPA) axis regulates stress physiology and behavior. To achieve an optimally tuned adaptive response, it is critical that the magnitude of the stress response matches the severity of the threat. Corticotropin-releasing hormone (CRH) released from the paraventricular nucleus of the hypothalamus is a major regulator of the HPA axis. However, how CRH-producing neurons in an intact animal respond to different stressor intensities is currently not known. Using two-photon calcium imaging on intact larval zebrafish, we recorded the activity of CRH cells, while the larvae were exposed to stressors of varying intensity. By combining behavioral and physiological measures, we first determined how sudden alterations in environmental conditions lead to different levels of stress axis activation. Then, we measured changes in the frequency and amplitude of Ca(2+) transients in individual CRH neurons in response to such stressors. The response magnitude of individual CRH cells covaried with stressor intensity. Furthermore, stressors caused the recruitment of previously inactive CRH neurons in an intensity-dependent manner, thus increasing the pool of responsive CRH cells. Strikingly, stressor-induced activity appeared highly synchronized among CRH neurons, and also across hemispheres. Thus, the stressor strength-dependent output of CRH neurons emerges by a dual mechanism that involves both the increased activity of individual cells and the recruitment of a larger pool of responsive cells. The synchronicity of CRH neurons within and across hemispheres ensures that the overall output of the HPA axis matches the severity of the threat. SIGNIFICANCE STATEMENT: Stressors trigger adaptive responses in the body that are essential for survival. How the brain responds to acute stressors of varying intensity in an intact animal, however, is not well understood. We address this question using two-photon Ca(2+) imaging in larval zebrafish with transgenically labeled corticotropin-releasing hormone (CRH) cells, which represent a major regulator of the stress axis. We show that stressor strength-dependent responses of CRH neurons emerge via an intensity-dependent increase in the activity of individual CRH cells, and by an increase in the pool of responsive CRH cells at the population level. Furthermore, we report striking synchronicity among CRH neurons even across hemispheres, which suggests tight intrahypothalamic and interhypothalamic coordination. Thus, our work reveals how CRH neurons respond to different levels of acute stress in vivo.


Subject(s)
Corticotropin-Releasing Hormone/metabolism , Gene Expression Regulation/physiology , Hypothalamus/pathology , Membrane Potentials/physiology , Neurons/physiology , Stress, Physiological/physiology , Animals , Animals, Genetically Modified , Avoidance Learning/physiology , Calcium/metabolism , Corticotropin-Releasing Hormone/genetics , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Hydrocortisone/metabolism , Larva , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Membrane Potentials/genetics , Motor Activity/genetics , Zebrafish
11.
Pain ; 156(9): 1647-1659, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25932687

ABSTRACT

Glycine transporter 1 (GlyT1) plays a crucial role in regulating extracellular glycine concentrations and might thereby constitute a new drug target for the modulation of glycinergic inhibition in pain signaling. Consistent with this view, inhibition of GlyT1 has been found to induce antinociceptive effects in various animal pain models. We have shown previously that the lidocaine metabolite N-ethylglycine (EG) reduces GlyT1-dependent glycine uptake by functioning as an artificial substrate for this transporter. Here, we show that EG is specific for GlyT1 and that in rodent models of inflammatory and neuropathic pain, systemic treatment with EG results in an efficient amelioration of hyperalgesia and allodynia without affecting acute pain. There was no effect on motor coordination or the development of inflammatory edema. No adverse neurological effects were observed after repeated high-dose application of EG. EG concentrations both in blood and spinal fluid correlated with an increase of glycine concentration in spinal fluid. The time courses of the EG and glycine concentrations corresponded well with the antinociceptive effect. Additionally, we found that EG reduced the increase in neuronal firing of wide-dynamic-range neurons caused by inflammatory pain induction. These findings suggest that systemically applied lidocaine exerts antihyperalgesic effects through its metabolite EG in vivo, by enhancing spinal inhibition of pain processing through GlyT1 modulation and subsequent increase of glycine concentrations at glycinergic inhibitory synapses. EG and other substrates of GlyT1, therefore, may be a useful therapeutic agent in chronic pain states involving spinal disinhibition.


Subject(s)
Analgesics/therapeutic use , N-substituted Glycines/therapeutic use , Neuralgia/drug therapy , Neurogenic Inflammation/drug therapy , Pain Threshold/drug effects , Analgesics/metabolism , Animals , Disease Models, Animal , Freund's Adjuvant/toxicity , Glutamic Acid/pharmacology , Glycine/cerebrospinal fluid , Glycine/pharmacology , Male , Membrane Potentials/drug effects , Membrane Potentials/genetics , Mice , Mice, Inbred C57BL , Motor Activity/drug effects , N-substituted Glycines/metabolism , N-substituted Glycines/pharmacology , Neuralgia/etiology , Neuralgia/pathology , Neurogenic Inflammation/etiology , Pain Measurement , Physical Stimulation/adverse effects , Posterior Horn Cells/drug effects , Posterior Horn Cells/physiology , Receptors, Glycine/genetics , Receptors, Glycine/metabolism , Spinal Cord/physiopathology , Xenopus laevis
12.
Neurobiol Dis ; 77: 141-54, 2015 May.
Article in English | MEDLINE | ID: mdl-25766678

ABSTRACT

Dravet Syndrome (DS) is caused by heterozygous loss-of-function mutations in voltage-gated sodium channel NaV1.1. Our mouse genetic model of DS recapitulates its severe seizures and premature death. Sleep disturbance is common in DS, but its mechanism is unknown. Electroencephalographic studies revealed abnormal sleep in DS mice, including reduced delta wave power, reduced sleep spindles, increased brief wakes, and numerous interictal spikes in Non-Rapid-Eye-Movement sleep. Theta power was reduced in Rapid-Eye-Movement sleep. Mice with NaV1.1 deleted specifically in forebrain interneurons exhibited similar sleep pathology to DS mice, but without changes in circadian rhythm. Sleep architecture depends on oscillatory activity in the thalamocortical network generated by excitatory neurons in the ventrobasal nucleus (VBN) of the thalamus and inhibitory GABAergic neurons in the reticular nucleus of the thalamus (RNT). Whole-cell NaV current was reduced in GABAergic RNT neurons but not in VBN neurons. Rebound firing of action potentials following hyperpolarization, the signature firing pattern of RNT neurons during sleep, was also reduced. These results demonstrate imbalance of excitatory vs. inhibitory neurons in this circuit. As predicted from this functional impairment, we found substantial deficit in homeostatic rebound of slow wave activity following sleep deprivation. Although sleep disorders in epilepsies have been attributed to anti-epileptic drugs, our results show that sleep disorder in DS mice arises from loss of NaV1.1 channels in forebrain GABAergic interneurons without drug treatment. Impairment of NaV currents and excitability of GABAergic RNT neurons are correlated with impaired sleep quality and homeostasis in these mice.


Subject(s)
Disease Models, Animal , Epilepsies, Myoclonic/complications , Epilepsies, Myoclonic/pathology , Interneurons/pathology , Sleep Wake Disorders/etiology , Thalamus/pathology , Age Factors , Animals , Animals, Newborn , Electric Stimulation , Epilepsies, Myoclonic/genetics , GABAergic Neurons/pathology , Glutamate Decarboxylase/metabolism , Membrane Potentials/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation/genetics , NAV1.1 Voltage-Gated Sodium Channel/genetics , Patch-Clamp Techniques , Sleep Deprivation/physiopathology , Video Recording , Wakefulness/genetics
13.
Neuron ; 83(5): 1144-58, 2014 Sep 03.
Article in English | MEDLINE | ID: mdl-25189210

ABSTRACT

T-type calcium channels are essential contributors to the transmission of nociceptive signals in the primary afferent pain pathway. Here, we show that T-type calcium channels are ubiquitinated by WWP1, a plasma-membrane-associated ubiquitin ligase that binds to the intracellular domain III-IV linker region of the Cav3.2 T-type channel and modifies specific lysine residues in this region. A proteomic screen identified the deubiquitinating enzyme USP5 as a Cav3.2 III-IV linker interacting partner. Knockdown of USP5 via shRNA increases Cav3.2 ubiquitination, decreases Cav3.2 protein levels, and reduces Cav3.2 whole-cell currents. In vivo knockdown of USP5 or uncoupling USP5 from native Cav3.2 channels via intrathecal delivery of Tat peptides mediates analgesia in both inflammatory and neuropathic mouse models of mechanical hypersensitivity. Altogether, our experiments reveal a cell signaling pathway that regulates T-type channel activity and their role in nociceptive signaling.


Subject(s)
Calcium Channels, T-Type/metabolism , Endopeptidases/metabolism , Inflammation/physiopathology , Neuralgia/enzymology , Animals , Calcium Channels, T-Type/genetics , Cells, Cultured , Disease Models, Animal , Endopeptidases/genetics , Freund's Adjuvant/toxicity , Humans , Hyperalgesia/diagnosis , Hyperalgesia/physiopathology , In Vitro Techniques , Inflammation/chemically induced , Male , Membrane Potentials/drug effects , Membrane Potentials/genetics , Mice, Inbred C57BL , Mice, Transgenic , Nerve Tissue Proteins/metabolism , Neuralgia/drug therapy , Pain Threshold/drug effects , Pain Threshold/physiology , Peptides/therapeutic use , Sensory Receptor Cells/drug effects , Sensory Receptor Cells/physiology , Spinal Cord/cytology , Transfection , Ubiquitination/genetics , Ubiquitination/physiology
14.
J Neurosci ; 34(31): 10325-38, 2014 Jul 30.
Article in English | MEDLINE | ID: mdl-25080593

ABSTRACT

α-Tectorin (TECTA), ß-tectorin (TECTB), and carcinoembryonic antigen-related cell adhesion molecule 16 (CEACAM) are secreted glycoproteins that are present in the tectorial membrane (TM), an extracellular structure overlying the hearing organ of the inner ear, the organ of Corti. Previous studies have shown that TECTA and TECTB are both required for formation of the striated-sheet matrix within which collagen fibrils of the TM are imbedded and that CEACAM16 interacts with TECTA. To learn more about the structural and functional significance of CEACAM16, we created a Ceacam16-null mutant mouse. In the absence of CEACAM16, TECTB levels are reduced, a clearly defined striated-sheet matrix does not develop, and Hensen's stripe, a prominent feature in the basal two-thirds of the TM in WT mice, is absent. CEACAM16 is also shown to interact with TECTB, indicating that it may stabilize interactions between TECTA and TECTB. Although brain-stem evoked responses and distortion product otoacoustic emissions are, for most frequencies, normal in young mice lacking CEACAM16, stimulus-frequency and transiently evoked emissions are larger. We also observed spontaneous otoacoustic emissions (SOAEs) in 70% of the homozygous mice. This incidence is remarkable considering that <3% of WT controls have SOAEs. The predominance of SOAEs >15 kHz correlates with the loss of Hensen's stripe. Results from mice lacking CEACAM16 are consistent with the idea that the organ of Corti evolved to maximize the gain of the cochlear amplifier while preventing large oscillations. Changes in TM structure appear to influence the balance between energy generation and dissipation such that the system becomes unstable.


Subject(s)
Cell Adhesion Molecules/deficiency , Extracellular Matrix Proteins/metabolism , Organ of Corti/cytology , Otoacoustic Emissions, Spontaneous/physiology , Tectorial Membrane/physiology , Acoustic Stimulation , Animals , Cell Adhesion Molecules/genetics , Evoked Potentials, Auditory, Brain Stem/genetics , Gene Expression Regulation/genetics , HEK293 Cells , Humans , Immunoprecipitation , Membrane Potentials/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Electron, Transmission , Otoacoustic Emissions, Spontaneous/genetics , Patch-Clamp Techniques , Tectorial Membrane/ultrastructure , beta-Galactosidase/metabolism
15.
ACS Chem Neurosci ; 5(11): 1131-41, 2014 Nov 19.
Article in English | MEDLINE | ID: mdl-25029033

ABSTRACT

Epilepsy is the most common neurological disorder, with over 50 million people worldwide affected. Recent evidence suggests that the transient receptor potential cation channel subfamily V member 1 (TRPV1) may contribute to the onset and progression of some forms of epilepsy. Since the two nonpsychotropic cannabinoids cannabidivarin (CBDV) and cannabidiol (CBD) exert anticonvulsant activity in vivo and produce TRPV1-mediated intracellular calcium elevation in vitro, we evaluated the effects of these two compounds on TRPV1 channel activation and desensitization and in an in vitro model of epileptiform activity. Patch clamp analysis in transfected HEK293 cells demonstrated that CBD and CBDV dose-dependently activate and rapidly desensitize TRPV1, as well as TRP channels of subfamily V type 2 (TRPV2) and subfamily A type 1 (TRPA1). TRPV1 and TRPV2 transcripts were shown to be expressed in rat hippocampal tissue. When tested on epileptiform neuronal spike activity in hippocampal brain slices exposed to a Mg(2+)-free solution using multielectrode arrays (MEAs), CBDV reduced both epileptiform burst amplitude and duration. The prototypical TRPV1 agonist, capsaicin, produced similar, although not identical effects. Capsaicin, but not CBDV, effects on burst amplitude were reversed by IRTX, a selective TRPV1 antagonist. These data suggest that CBDV antiepileptiform effects in the Mg(2+)-free model are not uniquely mediated via activation of TRPV1. However, TRPV1 was strongly phosphorylated (and hence likely sensitized) in Mg(2+)-free solution-treated hippocampal tissue, and both capsaicin and CBDV caused TRPV1 dephosphorylation, consistent with TRPV1 desensitization. We propose that CBDV effects on TRP channels should be studied further in different in vitro and in vivo models of epilepsy.


Subject(s)
Cannabinoids/pharmacology , Membrane Potentials/drug effects , TRPV Cation Channels/metabolism , Animals , Capsaicin/pharmacology , Diterpenes/pharmacology , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HEK293 Cells , Hippocampus/cytology , Humans , In Vitro Techniques , Magnesium/metabolism , Membrane Potentials/genetics , Neurons/drug effects , Patch-Clamp Techniques , Rats , TRPA1 Cation Channel , TRPC Cation Channels/genetics , TRPC Cation Channels/metabolism , TRPV Cation Channels/antagonists & inhibitors , TRPV Cation Channels/genetics , Transfection , Vesicular Glutamate Transport Protein 1/metabolism
16.
J Neurosci ; 34(29): 9656-64, 2014 Jul 16.
Article in English | MEDLINE | ID: mdl-25031405

ABSTRACT

Layer 6 corticothalamic neurons are thought to modulate incoming sensory information via their intracortical axons targeting the major thalamorecipient layer of the neocortex, layer 4, and via their long-range feedback projections to primary sensory thalamic nuclei. However, anatomical reconstructions of individual layer 6 corticothalamic (L6 CT) neurons include examples with axonal processes ramifying within layer 5, and the relative input of the overall population of L6 CT neurons to layers 4 and 5 is not well understood. We compared the synaptic impact of L6 CT cells on neurons in layers 4 and 5. We found that the axons of L6 CT neurons densely ramified within layer 5a in both visual and somatosensory cortices of the mouse. Optogenetic activation of corticothalamic neurons generated large EPSPs in pyramidal neurons in layer 5a. In contrast, excitatory neurons in layer 4 exhibited weak excitation or disynaptic inhibition. Fast-spiking parvalbumin-positive cells in both layer 5a and layer 4 were also strongly activated by L6 CT neurons. The overall effect of L6 CT activation was to suppress layer 4 while eliciting action potentials in layer 5a pyramidal neurons. Together, our data indicate that L6 CT neurons strongly activate an output layer of the cortex.


Subject(s)
Cerebral Cortex/cytology , Neural Pathways/physiology , Neurons/physiology , Synapses/physiology , Thalamus/cytology , Animals , Cerebral Cortex/metabolism , Channelrhodopsins , Cholera Toxin/metabolism , Fluorescent Dyes/metabolism , Glutamate Decarboxylase/genetics , Glutamate Decarboxylase/metabolism , In Vitro Techniques , Integrases/genetics , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Membrane Potentials/drug effects , Membrane Potentials/genetics , Mice , Mice, Transgenic , Mutation/genetics , Neurons/classification , Photic Stimulation , Receptors, Neurotensin/genetics , Synaptophysin/genetics , Synaptophysin/metabolism , Thalamus/metabolism
17.
Neuron ; 82(4): 797-808, 2014 May 21.
Article in English | MEDLINE | ID: mdl-24768300

ABSTRACT

Brain function is mediated by neural circuit connectivity, and elucidating the role of connections is aided by techniques to block their output. We developed cell-type-selective, reversible synaptic inhibition tools for mammalian neural circuits by leveraging G protein signaling pathways to suppress synaptic vesicle release. Here, we find that the pharmacologically selective designer Gi-protein-coupled receptor hM4D is a presynaptic silencer in the presence of its cognate ligand clozapine-N-oxide (CNO). Activation of hM4D signaling sharply reduced synaptic release probability and synaptic current amplitude. To demonstrate the utility of this tool for neural circuit perturbations, we developed an axon-selective hM4D-neurexin variant and used spatially targeted intracranial CNO injections to localize circuit connections from the hypothalamus to the midbrain responsible for feeding behavior. This synaptic silencing approach is broadly applicable for cell-type-specific and axon projection-selective functional analysis of diverse neural circuits.


Subject(s)
Feeding Behavior/physiology , Hypothalamus/cytology , Mesencephalon/physiology , Nerve Net/physiology , Neurons/metabolism , Agouti-Related Protein/genetics , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Channelrhodopsins , Clozapine/analogs & derivatives , Clozapine/pharmacology , Feeding Behavior/drug effects , Humans , Hypothalamus/drug effects , In Vitro Techniques , Membrane Potentials/drug effects , Membrane Potentials/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Models, Molecular , Molecular Sequence Data , Nerve Net/drug effects , Neural Inhibition/drug effects , Neural Inhibition/genetics , Neural Inhibition/physiology , Neurons/cytology , Neurons/drug effects , Receptor, Muscarinic M4/genetics , Receptor, Muscarinic M4/metabolism , Repressor Proteins/genetics , Time Factors
18.
J Neurosci ; 34(16): 5505-14, 2014 Apr 16.
Article in English | MEDLINE | ID: mdl-24741041

ABSTRACT

Tip links between adjacent stereocilia are believed to gate mechano-electrical transducer (MET) channels and mediate the electrical responses of sensory hair cells. We found that mouse auditory hair cells that lack tip links due to genetic mutations or exposure to the Ca(2+) chelator BAPTA can, however, still respond to mechanical stimuli. These MET currents have unusual properties and are predominantly of the opposite polarity relative to those measured when tip links are present. There are other striking differences, for example, the channels are usually all closed when the hair cell is not stimulated and the currents in response to strong stimuli can be substantially larger than normal. These anomalous MET currents can also be elicited early in development, before the onset of mechano-electrical transduction with normal response polarity. Current-voltage curves of the anomalous MET currents are linear and do not show the rectification characteristic of normal MET currents. The permeant MET channel blocker dihydrostreptomycin is two orders of magnitude less effective in blocking the anomalous MET currents. The findings suggest the presence of a large population of MET channels with pore properties that are distinct from those of normal MET channels. These channels are not gated by hair-bundle links and can be activated under a variety of conditions in which normal tip-link-mediated transduction is not operational.


Subject(s)
Cell Membrane Permeability/physiology , Hair Cells, Auditory/physiology , Ion Channels/physiology , Mechanotransduction, Cellular/physiology , Animals , Animals, Newborn , Cadherin Related Proteins , Cadherins/genetics , Cell Membrane Permeability/genetics , Chelating Agents/pharmacology , Dihydrostreptomycin Sulfate/pharmacology , Egtazic Acid/analogs & derivatives , Egtazic Acid/pharmacology , Embryo, Mammalian , Female , Hair Cells, Auditory/cytology , Hair Cells, Auditory/drug effects , In Vitro Techniques , Ion Channels/drug effects , Male , Mechanotransduction, Cellular/drug effects , Mechanotransduction, Cellular/genetics , Membrane Potentials/drug effects , Membrane Potentials/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myosin VIIa , Myosins/genetics , Organ of Corti/cytology , Protein Precursors/genetics
19.
Heart Rhythm ; 11(6): 1015-1023, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24582607

ABSTRACT

BACKGROUND: Mutations in the SCN5A gene, encoding the α subunit of the cardiac Na(+) channel, Nav1.5, can result in several life-threatening arrhythmias. OBJECTIVE: To characterize a distal truncating SCN5A mutation, R1860Gfs*12, identified in a family with different phenotypes including sick sinus syndrome, atrial fibrillation (AF), atrial flutter, and atrioventricular block. METHODS: Patch-clamp and biochemical analyses were performed in human embryonic kidney 293 cells transfected with wild-type (WT) and/or mutant channels. RESULTS: The mutant channel expressed alone caused a 70% reduction in inward sodium current (INa) density compared to WT currents, which was consistent with its partial proteasomal degradation. It also led to a negative shift of steady-state inactivation and to a persistent current. When mimicking the heterozygous state of the patients by coexpressing WT and R1860Gfs*12 channels, the biophysical properties of INa were still altered and the mutant channel α subunits still interacted with the WT channels. Since the proband developed paroxysmal AF at a young age, we screened 17 polymorphisms associated with AF risk in this family and showed that the proband carries at-risk polymorphisms upstream of PITX2, a gene widely associated with AF development. In addition, when mimicking the difference in resting membrane potentials between cardiac atria and ventricles in human embryonic kidney 293 cells or when using computer model simulations, R1860Gfs*12 induced a more drastic decrease in INa at the atrial potential. CONCLUSION: We have identified a distal truncated SCN5A mutant associated with gain- and loss-of-function effects, leading to sick sinus syndrome and atrial arrhythmias. A constitutively higher susceptibility to arrhythmias of atrial tissues and genetic variability could explain the complex phenotype observed in this family.


Subject(s)
Atrial Fibrillation/genetics , NAV1.5 Voltage-Gated Sodium Channel/genetics , Sick Sinus Syndrome/genetics , Adult , Arrhythmias, Cardiac/genetics , Cells, Cultured , Electrophysiologic Techniques, Cardiac , Female , Genetic Predisposition to Disease , Heart Conduction System/physiopathology , Homeodomain Proteins/genetics , Humans , Membrane Potentials/genetics , Patch-Clamp Techniques , Pedigree , Phenotype , Polymorphism, Single Nucleotide/genetics , Transcription Factors/genetics , Transfection , Homeobox Protein PITX2
20.
Neuropharmacology ; 75: 324-36, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23973313

ABSTRACT

NMDA receptors are ligand-gated ion channels that assemble into tetrameric receptor complexes composed of glycine-binding GluN1 and GluN3 subunits (GluN3A-B) and glutamate-binding GluN2 subunits (GluN2A-D). NMDA receptors can assemble as GluN1/N2 receptors and as GluN3-containing NMDA receptors, which are either glutamate/glycine-activated triheteromeric GluN1/N2/N3 receptors or glycine-activated diheteromeric GluN1/N3 receptors. The glycine-binding GluN1 and GluN3 subunits display strikingly different pharmacological selectivity profiles. However, the pharmacological characterization of GluN3-containing receptors has been hampered by the lack of methods and pharmacological tools to study GluN3 subunit pharmacology in isolation. Here, we have developed a method to study the pharmacology of GluN3 subunits in recombinant diheteromeric GluN1/N3 receptors by mutating the orthosteric ligand-binding pocket in GluN1. This method is suitable for performing compound screening and characterization of structure-activity relationship studies on GluN3 ligands. We have performed a virtual screen of the orthosteric binding site of GluN3A in the search for antagonists with selectivity for GluN3 subunits. In the subsequent pharmacological evaluation of 99 selected compounds, we identified 6-hydroxy-[1,2,5]oxadiazolo[3,4-b]pyrazin-5(4H)-one (TK80) a novel competitive antagonist with preference for the GluN3B subunit. Serendipitously, we also identified [2-hydroxy-5-((4-(pyridin-3-yl)thiazol-2-yl)amino]benzoic acid (TK13) and 4-(2,4-dichlorobenzoyl)-1H-pyrrole-2-carboxylic acid (TK30), two novel non-competitive GluN3 antagonists. These findings demonstrate that structural differences between the orthosteric binding site of GluN3 and GluN1 can be exploited to generate selective ligands.


Subject(s)
Excitatory Amino Acid Antagonists/chemistry , Excitatory Amino Acid Antagonists/pharmacology , Membrane Potentials/drug effects , Receptors, N-Methyl-D-Aspartate/metabolism , Animals , Binding Sites/drug effects , Binding Sites/genetics , Computer Simulation , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Glycine/pharmacology , Inhibitory Concentration 50 , Membrane Potentials/genetics , Models, Molecular , Oocytes , Protein Binding/drug effects , Protein Structure, Tertiary , Protein Subunits/chemistry , Protein Subunits/genetics , Receptors, N-Methyl-D-Aspartate/chemistry , Receptors, N-Methyl-D-Aspartate/genetics , Small Molecule Libraries , Structure-Activity Relationship , Xenopus laevis
SELECTION OF CITATIONS
SEARCH DETAIL