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










Publication year range
1.
Front Cell Neurosci ; 14: 580583, 2020.
Article in English | MEDLINE | ID: mdl-33192326

ABSTRACT

Chemokines such as chemokine (C-C motif) ligand 2 (CCL2) play a role in several behaviors, including anxiety-like behavior, but whether neurons are an important source of CCL2 for behavior and how neuronal CCL2 may work to affect behavior are still debated. When a herpes simplex virus (HSV) vector was used to knockdown CCL2 mRNA in neurons of the central nucleus of the amygdala (CeA) in rats experiencing multiple withdrawals from low dose ethanol, anxiety-like behavior appeared in the social interaction task. To examine this finding further Fractalkine (CX3CL1), a chemokine that is often found to have an opposing function to CCL2 was measured in these rats. Both alcohol withdrawal and CCL2 knockdown increased the levels of the anti-inflammatory protein CX3CL1. The combination of alcohol withdrawal and CCL2 knockdown decreased CX3CL1 and may alter pro-inflammatory/anti-inflammatory balance, and thus highlights the potential importance of CCL2 and CCL2/CX3CL1 balance in anxiety. To find a mechanism by which neuronal chemokines like CCL2 could affect behavior, retrograde tracing with fluorescent nanobeads was done in two brain regions associated with anxiety the bed nucleus of the stria terminalis (BNST) and the ventral periaqueductal gray (VPAG). These studies identified CeA projection neurons to these brain regions that contain CCL2. To demonstrate that CCL2 can be transported via axons to downstream brain regions, the axonal transport blocker, colchicine, was given and 24 h later, the accumulation of CCL2 in CeA neuronal cell bodies was found. Finally, CCL2 in CeA neurons was localized to the synapse using confocal microscopy with enhanced resolution following deconvolution and electron microscopy, which along with the other evidence suggests that CCL2 may be transported down axons in CeA neurons and released from nerve terminals perhaps into brain regions like the BNST and VPAG to affect behaviors such as anxiety. These results suggest that neurons are an important target for chemokine research related to behavior.

2.
Alcohol Clin Exp Res ; 43(10): 2134-2143, 2019 10.
Article in English | MEDLINE | ID: mdl-31386210

ABSTRACT

BACKGROUND: Chronic ethanol (EtOH) exposure induces neurobehavioral maladaptations in the brain though the precise changes have not been fully explored. The central nucleus of the amygdala (CEA) regulates anxiety-like behavior induced by withdrawal from chronic intermittent EtOH (CIE) exposure, and the arginine vasopressin (AVP) system within the CEA regulates many anxiety-like behaviors. Thus, adaptations occur in the CEA AVP system due to chronic EtOH exposure, which lead to anxiety-like behaviors in rats. METHODS: Chronic exposure to a low-dose EtOH (4.5% wt/vol) induces anxiety-like behavior in rats. Wistar or Sprague Dawley rats were exposed to a modified CIE or CIE, while intra-CEA microinjections of AVP or a V1b receptor antagonist were used to elicit or block withdrawal-induced anxiety. Additionally, AVP microinjections into the CEA were given 24 hours following 15 days of continuous high-dose EtOH (7% wt/vol), a time period when rats no longer express anxiety. Chemogenetics was also used to activate the basolateral amygdala (BLA) or deactivate the dorsal periaqueductal gray=(dm/dlPAG) therefore PAG=periaqueductal gray to elicit or block withdrawal-induced anxiety. RESULTS: AVP microinjected into the CEA in lieu of exposure to the first 2 cycles of CIE was sufficient to induce anxiety-like behavior in these commonly used rat strains. The V1b receptor antagonist, but not an oxytocin receptor agonist, into the CEA during the first 2 withdrawal cycles suppressed anxiety. However, activation of the BLA in lieu of exposure to the first 2 cycles of CIE was insufficient to induce anxiety-like behavior. AVP microinjection into the CEA 24 hours into withdrawal reelicited anxiety-like behavior, and deactivation of the dm/dlPAG reduced this effect of CEA AVP. CONCLUSIONS: Taken together, this study demonstrates a role of CEA AVP and a CEA-dm/dlPAG circuit in the development of anxiety induced by CIE. Such information is valuable for identifying novel therapeutic targets for alcohol- and anxiety-associated disorders.


Subject(s)
Amygdala/drug effects , Anxiety/psychology , Arginine Vasopressin/pharmacology , Central Nervous System Depressants , Ethanol , Interpersonal Relations , Substance Withdrawal Syndrome/psychology , Animals , Anxiety/etiology , Anxiety/physiopathology , Arginine Vasopressin/administration & dosage , Behavior, Animal , Male , Microinjections , Periaqueductal Gray/drug effects , Rats , Rats, Sprague-Dawley , Rats, Wistar , Receptors, Vasopressin/drug effects , Substance Withdrawal Syndrome/complications , Substance Withdrawal Syndrome/physiopathology
3.
Psychopharmacology (Berl) ; 235(12): 3363-3379, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30392132

ABSTRACT

BACKGROUND: Arginine vasopressin (VP) has been implicated in a number of neuropsychiatric disorders with an emphasis on situations where stress increased the severity of the disorder. Based on this hypothesized role for VP in neuropsychiatric disorders, much research is currently being undertaken in humans and animals to test VP as a target for treatment of a number of these disorders including alcohol abuse. OBJECTIVES: To provide a summary of the literature regarding the role of VP in alcohol- and stress-related behaviors including the use of drugs that target VP in clinical trials. RESULTS: Changes in various components of the VP system occur with alcohol and stress. Manipulating VP or its receptors can alter alcohol- and stress-related behaviors including tolerance to alcohol, alcohol drinking, and anxiety-like behavior. Finally, the hypothalamic-pituitary-adrenal axis response to alcohol is also altered by manipulating the VP system. However, clinical trials of VP antagonists have had mixed results. CONCLUSIONS: A review of VP's involvement in alcohol's actions demonstrates that there is much to be learned about brain regions involved in VP-mediated effects on behavior. Thus, future work should focus on elucidating relevant brain regions. By using previous knowledge of the actions of VP and determining the brain regions and/or systems involved in its different behavioral effects, it may be possible to identify a specific receptor subtype target, drug treatment combination, or specific clinical contexts that may point toward a more successful treatment.


Subject(s)
Alcohol Drinking/metabolism , Alcoholism/metabolism , Ethanol/administration & dosage , Neurophysins/metabolism , Protein Precursors/metabolism , Vasopressins/metabolism , Alcohol Drinking/drug therapy , Alcohol Drinking/psychology , Alcoholism/drug therapy , Alcoholism/psychology , Animals , Antidiuretic Hormone Receptor Antagonists/pharmacology , Anxiety/drug therapy , Anxiety/metabolism , Anxiety/psychology , Arginine Vasopressin/antagonists & inhibitors , Arginine Vasopressin/metabolism , Ethanol/toxicity , Humans , Hypothalamo-Hypophyseal System/drug effects , Hypothalamo-Hypophyseal System/metabolism , Neurophysins/antagonists & inhibitors , Pituitary-Adrenal System/drug effects , Pituitary-Adrenal System/metabolism , Protein Precursors/antagonists & inhibitors , Stress, Psychological/drug therapy , Stress, Psychological/metabolism , Stress, Psychological/psychology , Vasopressins/antagonists & inhibitors
4.
Mol Ther ; 25(4): 928-934, 2017 04 05.
Article in English | MEDLINE | ID: mdl-28202388

ABSTRACT

Recent advances suggest that in vivo reprogramming of endogenous cell populations provides a viable alternative for neuron replacement. Astrocytes and oligodendrocyte precursor cells can be induced to transdifferentiate into neurons in the CNS, but, in these instances, reprogramming requires either transgenic mice or retroviral-mediated gene expression. We developed a microRNA (miRNA)-GFP construct that in vitro significantly reduced the expression of polypyrimidine tract-binding protein, and, subsequently, we packaged this construct in a novel oligodendrocyte preferring adeno-associated virus vector. Ten days after rat striatal transduction, the vast majority of the GFP-positive cells were oligodendrocytes, but 6 weeks to 6 months later, the majority of GFP-positive cells exhibited neuronal morphology and co-localized with the neuronal marker NeuN. Patch-clamp studies on striatal slices established that the GFP-positive cells exhibited electrophysiological properties indicative of mature neurons, such as spontaneous action potentials and spontaneous inhibitory postsynaptic currents. Also, 3 months after striatal vector administration, GFP-positive terminals in the ipsilateral globus pallidus or substantia nigra retrogradely transported fluorescent beads back to GFP-positive striatal cell bodies, indicating the presence of functional presynaptic terminals. Thus, this viral vector approach provides a potential means to harness resident oligodendrocytes as an endogenous source for in vivo neuronal replacement.


Subject(s)
Cell Transdifferentiation/genetics , Cellular Reprogramming/genetics , Corpus Striatum/cytology , Genetic Vectors/genetics , Neurons/cytology , Oligodendroglia/cytology , Animals , Cell Line , Dependovirus/genetics , Humans , Neurons/metabolism , Oligodendroglia/metabolism , RNA Interference , RNA, Small Interfering , Rats
5.
Brain Behav Immun ; 33: 102-11, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23770090

ABSTRACT

Anxiety-like responses to stress are accompanied by elevation of brain cytokine-mRNAs. Because cytokines microinjected into central-amygdala (CeA) substitute for stress in a behavioral paradigm, the possibility was raised that cytokines increased by stress influence behavior by affecting CeA-neural activity. Previously, cytokines increased firing-rate of CeA-neurons comparable to that induced by corticotropin-releasing factor (CRF). In this investigation, tumor-necrosis-factor-α (TNFα) increased amplitude, but not frequency of mEPSCs from CeA-neurons. Additionally, TNFα decreased the threshold for triggering action potentials from CeA-neurons without altering membrane-properties during current-clamp recording. Glutamate-receptor-antagonist blockade of mEPSCs and the TNFα-induced reduction in firing threshold implicated glutamate in these changes. A phosphatidyl-inositol-3-kinase-antagonist prevented the TNFα-induced increased in amplitude of mEPSCs, documenting a TNFα intracellular influence. Additionally, TNFα increased frequency, but not amplitude of mIPSCs. CRF-receptor-antagonists were found to prevent the TNFα-induced increase in mIPSC-frequency, without altering the TNFα-induced amplitude increase in mEPSCs or the reduced threshold for action-potentials by TNFα. To clarify how TNFα was increasing CRF-release in the presence of tetrodotoxin, the possibility tested was whether preventing glial-activation would prevent this elevated mIPSC-frequency blocked by CRF-receptor antagonists. Minocycline, which blocks glial activation, prevented the TNFα-induced increase in mIPSC-frequency - a finding consistent with glia contributing to the CRF-involvement in this TNFα action. To fully understand the means by which a CRF1-receptor-antagonist and minocycline prevent TNFα from increasing mIPSC-frequency will require further clarification. Nonetheless, these data provide convincing evidence that release of TNFα by stress could alter neural activity of CeA-neurons by influencing GABA-and glutamate function.


Subject(s)
Amygdala/immunology , Excitatory Postsynaptic Potentials/immunology , Induced Pluripotent Stem Cells/immunology , Neural Inhibition/immunology , Neurons/immunology , Stress, Physiological/immunology , Synaptic Transmission/immunology , Tumor Necrosis Factor-alpha/physiology , Amygdala/cytology , Amygdala/metabolism , Animals , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Neurons/cytology , Neurons/metabolism , Patch-Clamp Techniques , Rats , Rats, Sprague-Dawley , Tumor Necrosis Factor-alpha/metabolism
6.
Brain Behav Immun ; 25 Suppl 1: S146-54, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21377524

ABSTRACT

Stress has been shown to facilitate ethanol withdrawal-induced anxiety. Defining neurobiological mechanisms through which stress has such actions is important given the associated risk of relapse. While CRF has long been implicated in the action of stress, current results show that stress elevates the cytokine TNFα in the rat brain and thereby implicates cytokines in stress effects. In support of this view, prior TNFα microinjection into the central amygdala (CeA) of rats facilitated ethanol withdrawal-induced anxiety-a response that could not be attributed to an increase in plasma corticosterone. To test for a possible interaction between cytokines and CRF, a CRF1-receptor antagonist (SSR125543) administered prior to the repeated administration of TNFα or MCP-1/CCL2 reduced the magnitude of the withdrawal-induced anxiety. This finding provided evidence for cytokine action being dependent upon CRF. Additionally, the sensitizing effect of stress on withdrawal-induced anxiety was reduced by treating the repeated stress exposure prior to ethanol with the MEK inhibitor SL327. Consistent with cytokines having a neuromediator function distinct from a neuroimmune action, TNFα increased firing rate and GABA release from CeA neurons. Thus, an interaction of glial and neuronal function is proposed to contribute to the interaction of stress and chronic ethanol. Interrupting this potential glial-neuronal interaction could provide a novel means by which to alter the development of emotional states induced by stress that predict relapse in the alcoholic.


Subject(s)
Amygdala/physiology , Anxiety/metabolism , Corticotropin-Releasing Hormone/metabolism , Cytokines/metabolism , Ethanol/administration & dosage , Stress, Physiological/physiology , Substance Withdrawal Syndrome/metabolism , Adaptation, Psychological/drug effects , Adaptation, Psychological/physiology , Alcohols/administration & dosage , Amygdala/drug effects , Analysis of Variance , Animals , Corticosterone/blood , Electrophysiology , Hydrocarbons, Halogenated/pharmacology , Male , Microinjections , Rats , Rats, Sprague-Dawley , Receptors, Corticotropin-Releasing Hormone/antagonists & inhibitors , Receptors, Corticotropin-Releasing Hormone/metabolism , Restraint, Physical , Substance Withdrawal Syndrome/physiopathology , Thiazines/pharmacology , Tumor Necrosis Factor-alpha/administration & dosage , Tumor Necrosis Factor-alpha/metabolism
7.
Brain Res Rev ; 65(2): 113-23, 2011 Jan 01.
Article in English | MEDLINE | ID: mdl-20837058

ABSTRACT

While research on the actions of ethanol at the GABAergic synapse has focused on postsynaptic mechanisms, recent data have demonstrated that ethanol also facilitates GABA release from presynaptic terminals in many, but not all, brain regions. The ability of ethanol to increase GABA release can be regulated by different G protein-coupled receptors (GPCRs), such as the cannabinoid-1 receptor, corticotropin-releasing factor 1 receptor, GABA(B) receptor, and the 5-hydroxytryptamine 2C receptor. The intracellular messengers linked to these GPCRs, including the calcium that is released from internal stores, also play a role in ethanol-enhanced GABA release. Hypotheses are proposed to explain how ethanol interacts with the GPCR pathways to increase GABA release and how this interaction contributes to the brain region specificity of ethanol-enhanced GABA release. Defining the mechanism of ethanol-facilitated GABA release will further our understanding of the GABAergic profile of ethanol and increase our knowledge of how GABAergic neurotransmission may contribute to the intoxicating effects of alcohol and to alcohol dependence.


Subject(s)
Brain/drug effects , Ethanol/pharmacology , Receptors, G-Protein-Coupled/physiology , Receptors, GABA/physiology , gamma-Aminobutyric Acid/metabolism , Animals , Brain/metabolism , Central Nervous System Depressants/pharmacology , Humans , Synapses/drug effects , Synapses/metabolism
8.
J Neurochem ; 116(4): 554-63, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21155805

ABSTRACT

Ethanol exposure produces alterations in GABA(A) receptor function and expression associated with CNS hyperexcitability, but the mechanisms of these effects are unknown. Ethanol is known to increase both GABA(A) receptor α4 subunits and protein kinase C (PKC) isozymes in vivo and in vitro. Here, we investigated ethanol regulation of GABA(A) receptor α4 subunit expression in cultured cortical neurons to delineate the role of PKC. Cultured neurons were prepared from rat pups on postnatal day 0-1 and tested after 18 days. GABA(A) receptor α4 subunit surface expression was assessed using P2 fractionation and surface biotinylation following ethanol exposure for 4 h. Miniature inhibitory post-synaptic currents were measured using whole cell patch clamp recordings. Ethanol increased GABA(A) receptor α4 subunit expression in both the P2 and biotinylated fractions, while reducing the decay time constant in miniature inhibitory post-synaptic currents, with no effect on γ2 or δ subunits. PKC activation mimicked ethanol effects, while the PKC inhibitor calphostin C prevented ethanol-induced increases in GABA(A) receptor α4 subunit expression. PKCγ siRNA knockdown prevented ethanol-induced increases in GABA(A) receptor α4 subunit expression, but inhibition of the PKCß isoform with PKCß pseudosubstrate had no effect. We conclude that PKCγ regulates ethanol-induced alterations in α4-containing GABA(A) receptors.


Subject(s)
Cerebral Cortex/metabolism , Ethanol/pharmacology , Gene Expression Regulation , Neurons/metabolism , Protein Kinase C/metabolism , Receptors, GABA-A/biosynthesis , Animals , Animals, Newborn , Cells, Cultured , Cerebral Cortex/drug effects , Cerebral Cortex/enzymology , Gene Expression Regulation/drug effects , Neurons/drug effects , Neurons/enzymology , Rats , Rats, Sprague-Dawley
9.
Neuropharmacology ; 58(7): 1179-86, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20206640

ABSTRACT

Research on the actions of ethanol at the GABAergic synapse has traditionally focused on postsynaptic mechanisms, but recent data demonstrate that ethanol also increases both evoked and spontaneous GABA release in many brain regions. Using whole-cell voltage-clamp recordings, we previously showed that ethanol increases spontaneous GABA release at the rat interneuron-Purkinje cell synapse. This presynaptic ethanol effect is dependent on calcium release from internal stores, possibly through activation of inositol 1,4,5-trisphosphate receptors (IP(3)Rs). After confirming that ethanol targets vesicular GABA release, in the present study we used electron microscopic immunohistochemistry to demonstrate that IP(3)Rs are located in presynaptic terminals of cerebellar interneurons. Activation of IP(3)Rs requires binding of IP(3), generated through activation of phospholipase C (PLC). We find that the PLC antagonist edelfosine prevents ethanol from increasing spontaneous GABA release. Diacylglycerol generated by PLC and calcium released by activation of the IP(3)R activate protein kinase C (PKC). Ethanol-enhanced GABA release was blocked by two PKC antagonists, chelerythrine and calphostin C. When a membrane impermeable PKC antagonist, PKC (19-36), was delivered intracellularly to the postsynaptic neuron, ethanol continued to increase spontaneous GABA release. Overall, these results suggest that activation of the PLC/IP(3)R/PKC pathway is necessary for ethanol to increase spontaneous GABA release from presynaptic terminals onto Purkinje cells.


Subject(s)
Central Nervous System Depressants/pharmacology , Ethanol/pharmacology , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Protein Kinase C/metabolism , Type C Phospholipases/metabolism , gamma-Aminobutyric Acid/metabolism , Animals , Calcium/metabolism , Cerebellum/drug effects , Cerebellum/metabolism , Cerebellum/ultrastructure , Diglycerides/metabolism , In Vitro Techniques , Interneurons/drug effects , Interneurons/metabolism , Interneurons/ultrastructure , Male , Presynaptic Terminals/drug effects , Presynaptic Terminals/metabolism , Presynaptic Terminals/ultrastructure , Protein Kinase C/antagonists & inhibitors , Purkinje Cells/drug effects , Purkinje Cells/metabolism , Purkinje Cells/ultrastructure , Rats , Rats, Sprague-Dawley , Signal Transduction , Type C Phospholipases/antagonists & inhibitors
10.
Mol Ther ; 18(3): 570-8, 2010 03.
Article in English | MEDLINE | ID: mdl-20040913

ABSTRACT

DNA shuffling and directed evolution were employed to develop a novel adeno-associated virus (AAV) vector capable of crossing the seizure-compromised blood-brain barrier (BBB) and transducing cells in the brain. Capsid DNA from AAV serotypes 1-6, 8, and 9 were shuffled and recombined to create a library of chimeric AAVs. One day after kainic acid-induced limbic seizure activity in rats, the virus library was infused intravenously (i.v.), and 3 days later, neuron-rich cells were mechanically dissociated from seizure-sensitive brain sites, collected and viral DNA extracted. After three cycles of selection, green fluorescent protein (GFP)-packaged clones were administered directly into brain or i.v. 1 day after kainic acid-induced seizures. Several clones that were effective after intracranial administration did not transduce brain cells after the i.v. administration. However, two clones (32 and 83) transduced the cells after direct brain infusion and after i.v. administration transduced the cells that were localized to the piriform cortex and ventral hippocampus, areas exhibiting a seizure-compromised BBB. No transduction occurred in areas devoid of BBB compromise. Only one parental serotype (AAV8) exhibited a similar expression profile, but the biodistribution of 32 and 83 diverged dramatically from this parental serotype. Thus, novel AAV vectors have been created that can selectively cross the seizure-compromised BBB and transduce cells.


Subject(s)
Blood-Brain Barrier , Dependovirus/metabolism , Directed Molecular Evolution , Genetic Therapy/methods , Animals , Blood-Brain Barrier/chemistry , Capsid/chemistry , Cell Line , Cell Survival , DNA/metabolism , Female , Green Fluorescent Proteins/chemistry , Humans , Immunohistochemistry/methods , Kainic Acid/chemistry , Mice , Mice, Inbred BALB C , Microscopy, Confocal/methods , Neurons/metabolism , Rats , Seizures/metabolism
11.
J Neurophysiol ; 100(6): 3417-28, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18945815

ABSTRACT

Ethanol increases miniature inhibitory postsynaptic current frequency and decreases the paired-pulse ratio, which suggests that ethanol increases both spontaneous and evoked GABA release, respectively. We have shown previously that ethanol increases GABA release at the rat interneuron-Purkinje cell synapse and that this ethanol effect involves calcium release from internal stores; however, further exploration of the mechanism responsible for ethanol-enhanced GABA release was needed. We found that a cannabinoid receptor 1 (CB1) agonist, WIN-55212, and a GABA(B) receptor agonist, baclofen, decreased baseline spontaneous GABA release and prevented ethanol from increasing spontaneous GABA release. The CB1 receptor and GABA(B) receptor are Galpha i-linked G protein-coupled receptors with common downstream messengers that include adenylate cyclase and protein kinase A (PKA). Adenylate cyclase and PKA antagonists blocked ethanol from increasing spontaneous GABA release, whereas a PKA antagonist limited to the postsynaptic neuron did not block ethanol from increasing spontaneous GABA release. These results suggest that presynaptic PKA plays an essential role in ethanol-enhanced spontaneous GABA release. Similar to ethanol, we found that the mechanism of the cannabinoid-mediated decrease in spontaneous GABA release involves internal calcium stores and PKA. A PKA antagonist decreased baseline spontaneous GABA release. This effect was reduced after incubating the slice with a calcium chelator, BAPTA-AM, but was unaffected when BAPTA was limited to the postsynaptic neuron. This suggests that the PKA antagonist is acting through a presynaptic, calcium-dependent mechanism to decrease spontaneous GABA release. Overall, these results suggest that PKA activation is necessary for ethanol to increase spontaneous GABA release.


Subject(s)
Central Nervous System Depressants/pharmacology , Cerebellum/cytology , Ethanol/pharmacology , Protein Kinase C/physiology , Purkinje Cells/drug effects , gamma-Aminobutyric Acid/metabolism , Animals , Animals, Newborn , Baclofen/pharmacology , Benzoxazines/pharmacology , Benzylamines/pharmacology , Calcium/metabolism , Dose-Response Relationship, Drug , Electric Stimulation/methods , Enzyme Inhibitors/pharmacology , GABA Agents/pharmacology , In Vitro Techniques , Inhibitory Postsynaptic Potentials/drug effects , Inhibitory Postsynaptic Potentials/physiology , Membrane Potentials/drug effects , Membrane Potentials/physiology , Morpholines/pharmacology , Naphthalenes/pharmacology , Patch-Clamp Techniques , Phosphinic Acids/pharmacology , Rats , Rats, Sprague-Dawley , Sodium Channel Blockers/pharmacology , Tetrodotoxin/pharmacology
12.
J Pharmacol Exp Ther ; 326(2): 596-603, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18502983

ABSTRACT

Whereas ethanol has behavioral actions consistent with increased GABAergic function, attempts to demonstrate a direct enhancement of GABA-gated currents by ethanol have produced mixed results. Recent work has suggested that a part of the GABAergic profile of ethanol may result from enhanced GABA release from presynaptic terminals. The present study examines the effect of ethanol on GABA release in several brain regions to assess the regional nature of ethanol-induced GABA release. Whole-cell voltage clamp recording of spontaneous inhibitory postsynaptic currents (sIPSCs) from mechanically dissociated neurons and miniature inhibitory postsynaptic currents (mIPSCs) and paired-pulse ratio (PPR) from a slice preparation were used to quantify GABA release. Ethanol produced a concentration-dependent increase in the frequency of sIPSCs recorded from mechanically dissociated cerebellar Purkinje neurons and mIPSCs from substantia nigra neurons without having an effect on sIPSCs recorded from lateral septal or cerebrocortical neurons. This regional difference in the effect of ethanol on GABA release was confirmed with PPR recording from brain slices. These data indicate that ethanol can act on presynaptic terminals to increase GABA release in some brain regions while having little or no effect on GABA release in others. This regional difference is consistent with earlier in vivo studies in which ethanol affected neural activity and sensitivity to GABA in some, but not all, brain sites.


Subject(s)
Brain/drug effects , Central Nervous System Depressants/pharmacology , Ethanol/pharmacology , Presynaptic Terminals/drug effects , gamma-Aminobutyric Acid/metabolism , Animals , Brain/metabolism , Brain/physiology , Dose-Response Relationship, Drug , Electrodes , In Vitro Techniques , Membrane Potentials/drug effects , Membrane Potentials/physiology , Neurons/drug effects , Neurons/metabolism , Neurons/physiology , Patch-Clamp Techniques , Presynaptic Terminals/metabolism , Presynaptic Terminals/physiology , Purkinje Cells/drug effects , Purkinje Cells/metabolism , Purkinje Cells/physiology , Rats , Rats, Sprague-Dawley
13.
J Pharmacol Exp Ther ; 323(1): 356-64, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17652632

ABSTRACT

Recent data have demonstrated that ethanol increases gamma-aminobutyric acid (GABA) release in many brain regions, but little is known about the mechanism responsible for this action. Consistent with previous results, ethanol increased miniature inhibitory postsynaptic current (mIPSC) frequency at the interneuron-Purkinje cell synapse in the slice and in mechanically dissociated neurons. These data suggest that ethanol is increasing spontaneous GABA release at this synapse. It is generally accepted that ethanol increases levels of intracellular calcium and that changes in intracellular calcium can alter neurotransmitter release. Therefore, we examined the contribution of calcium-dependent pathways to the effect of ethanol on spontaneous GABA release at the interneuron-Purkinje cell synapse. Ethanol continued to increase mIPSC frequency in a nominally calcium-free extracellular solution and in the presence of a voltage-dependent calcium channel inhibitor, cadmium chloride. These data suggest that influx of extracellular calcium does not play a critical role in the mechanism of ethanol-enhanced spontaneous GABA release. However, a sarco/endoplasmic-reticulum calcium ATPase pump inhibitor (thapsigargin), an inositol 1,4,5-trisphosphate receptor antagonist (2-aminoethoxydiphenylborate) and a ryanodine receptor antagonist (ryanodine) significantly reduced the ability of ethanol to increase mIPSC frequency. In addition, ethanol was still able to increase mIPSC frequency in the presence of intracellular 1,2-bis(2-aminophenoxy)ethane-N,N,N',N'-tetraacetic acid (BAPTA) and a cannabinoid receptor antagonist N-(piperidin-1-yl)-5-(4-iodophenyl)-1-(2,4-dichlorophenyl)-4-methyl-1H-pyrazole-3-carboxamide (AM-251); thus, retrograde messengers are not involved in ethanol-enhanced spontaneous GABA release. Overall, these data suggest that calcium release from presynaptic internal stores plays a vital role in the mechanism of ethanol-enhanced spontaneous GABA release at the interneuron-Purkinje cell synapse.


Subject(s)
Calcium/metabolism , Cerebellum/drug effects , Ethanol/pharmacology , Interneurons/drug effects , Presynaptic Terminals/metabolism , gamma-Aminobutyric Acid/metabolism , Animals , Cerebellum/cytology , Cerebellum/metabolism , In Vitro Techniques , Inhibitory Postsynaptic Potentials/drug effects , Interneurons/metabolism , Purkinje Cells/drug effects , Purkinje Cells/metabolism , Rats , Rats, Sprague-Dawley
14.
Alcohol Clin Exp Res ; 30(8): 1400-7, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16899043

ABSTRACT

BACKGROUND: Ethanol has actions on cerebellar Purkinje neurons that can result either in a net excitation or in inhibition of neuronal activity. The present study examines the interplay of presynaptic and postsynaptic mechanisms to determine the net effect of ethanol on the neuronal firing rate of cerebellar Purkinje neurons. METHODS: Whole-cell voltage-clamp recording of miniature inhibitory postsynaptic currents (mIPSCs) from Purkinje neurons in cerebellar slices was used to examine the effect of ethanol on presynapticsynaptic release of gamma-aminobutyric acid (GABA) and glutamate. Extracellular recording was used to examine the net action of both presynaptic and postsynaptic effects of ethanol on the firing rate of Purkinje neurons. RESULTS: Under whole-cell voltage clamp, the frequency of bicuculline-sensitive miniature postsynaptic currents (mIPSCs) was increased dose-dependently by 25, 50, and 100 mM ethanol without any change in amplitude or decay time. Despite this evidence of increased release of GABA by ethanol, application of 50 mM ethanol caused an increase in firing in some neurons and a decrease in firing in others with a nonrandom distribution. When both glutamatergic and GABAergic influences were removed by simultaneous application of 6-cyano-7-nitroquinoxaline-2,3-dione and picrotoxin, respectively, ethanol caused only an increase in firing rate. CONCLUSIONS: These data are consistent with a dual action of ethanol on cerebellar Purkinje neuron activity. Specifically, ethanol acts presynaptically to increase inhibition by release of GABA, while simultaneously acting postsynaptically to increase intrinsic excitatory drive.


Subject(s)
Ethanol/pharmacology , Neural Inhibition/drug effects , Neurons/drug effects , Presynaptic Terminals/drug effects , Purkinje Cells/drug effects , Action Potentials/drug effects , Action Potentials/physiology , Animals , Cerebellum/drug effects , Cerebellum/physiology , In Vitro Techniques , Neural Inhibition/physiology , Neurons/physiology , Presynaptic Terminals/physiology , Purkinje Cells/physiology , Rats , Rats, Sprague-Dawley
15.
Neuropsychopharmacology ; 30(8): 1407-25, 2005 Aug.
Article in English | MEDLINE | ID: mdl-15856077

ABSTRACT

Early behavioral investigations supported the contention that systemic ethanol displays a GABAmimetic profile. Microinjection of GABA agonists into brain and in vivo electrophysiological studies implicated a regionally specific action of ethanol on GABA function. While selectivity of ethanol to enhance the effect of GABA was initially attributed an effect on type-I-benzodiazepine (BZD)-GABA(A) receptors, a lack of ethanol's effect on GABA responsiveness from isolated neurons with this receptor subtype discounted this contention. Nonetheless, subsequent work identified GABA(A) receptor subtypes, with limited distribution in brain, sensitive to enhancement of GABA at relevant ethanol concentrations. In view of these data, it is hypothesized that the GABAmimetic profile for ethanol is due to activation of mechanisms associated with GABA function, distinct from a direct action on the majority of postsynaptic GABA(A) receptors. The primary action proposed to account for ethanol's regional specificity on GABA transmission is its ability to release GABA from some, but not all, presynaptic GABAergic terminals. As systemic administration of ethanol increases neuroactive steroids, which can enhance GABA responsiveness, this elevated level of neurosteroids is proposed to magnify the effect of GABA released by ethanol. Additional factors contributing to the degree to which ethanol interacts with GABA function include an involvement of GABA(B) and other receptors that influence ethanol-induced GABA release, an effect of phosphorylation on GABA responsiveness, and a regional reduction of glutamatergic tone. Thus, an integration of these consequences induced by ethanol is proposed to provide a logical basis for its in vivo GABAmimetic profile.


Subject(s)
Brain/drug effects , Central Nervous System Depressants/pharmacology , Ethanol/pharmacology , Receptors, GABA/physiology , gamma-Aminobutyric Acid/metabolism , Animals , Brain/metabolism , Drug Interactions , GABA Agonists/pharmacology , Humans , Models, Biological , Receptors, GABA/classification , gamma-Aminobutyric Acid/pharmacology
16.
J Clin Invest ; 115(3): 774-9, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15765150

ABSTRACT

Essential tremor is the most common movement disorder and has an unknown etiology. Here we report that gamma-aminobutyric acidA (GABA(A)) receptor alpha1-/- mice exhibit postural and kinetic tremor and motor incoordination that is characteristic of essential tremor disease. We tested mice with essential-like tremor using current drug therapies that alleviate symptoms in essential tremor patients (primidone, propranolol, and gabapentin) and several candidates hypothesized to reduce tremor, including ethanol; the noncompetitive N-methyl-D-aspartate receptor antagonist MK-801; the adenosine A1 receptor agonist 2-chloro-N6-cyclopentyladenosine (CCPA); the GABA(A) receptor modulators diazepam, allopregnanolone, and Ro15-4513; and the L-type Ca2+ channel antagonist nitrendipine. Primidone, propranolol, and gabapentin reduced the amplitude (power) of the pathologic tremor. Nonsedative doses of ethanol eliminated tremor in mice. Diazepam, allopregnanolone, Ro15-4513, and nitrendipine had no effect or enhanced tremor, whereas MK-801 and CCPA reduced tremor. To understand the etiology of tremor in these mice, we studied the electrophysiological properties of cerebellar Purkinje cells. Cerebellar Purkinje cells in GABA(A) receptor alpha1-/- mice exhibited a profound loss of all responses to synaptic or exogenous GABA, but no differences in abundance, gross morphology, or spontaneous synaptic activity were observed. This genetic animal model elucidates a mechanism of GABAergic dysfunction in the major motor pathway and potential targets for pharmacotherapy of essential tremor.


Subject(s)
Essential Tremor/genetics , Essential Tremor/metabolism , Protein Subunits/metabolism , Receptors, GABA-A/metabolism , Adrenergic beta-Antagonists/pharmacology , Animals , Anticonvulsants/pharmacology , Disease Models, Animal , Dizocilpine Maleate/pharmacology , Essential Tremor/drug therapy , Ethanol/therapeutic use , Excitatory Amino Acid Antagonists/pharmacology , Humans , Mice , Mice, Knockout , Motor Activity/physiology , Patch-Clamp Techniques , Primidone/pharmacology , Propranolol/pharmacology , Protein Subunits/genetics , Receptors, GABA-A/genetics
17.
Brain Res Brain Res Rev ; 48(1): 57-73, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15708628

ABSTRACT

In 1973, a technique of administering 6-hydroxydopamine (2,4,5-trihydroxyphenylethylamine) intracisternally to neonate rats was introduced to selectively reduce brain dopamine (neonate-lesioned rat). This neonate treatment proved unique when compared to rats lesioned as adults with 6-hydroxydopamine--prompting the discovery of differing functional characteristics resulting from the age at which brain dopamine is reduced. A realization was that neonate-lesioned rats modeled the loss of central dopamine and the increased susceptibility for self-injury in Lesch-Nyhan disease, which allowed identification of drugs useful in treating self-injury in mentally retarded patients. The neonate-lesioned rat has also been proposed to model the hyperactivity observed in attention-deficit hyperactivity disorder. Because the neonate-lesioned rat exhibits enhanced sensitization to repeated NMDA receptor antagonist administration and has functional changes characteristic of schizophrenia, the neonate lesioning is believed to emulate the hypothesized NMDA hypofunction in this psychiatric disorder. Besides modeling features of neurological and psychiatric disorders, important neurobiological concepts emerged from pharmacological studies in the neonate-lesioned rats. One was the discovery of coupling of D1/D2-dopamine receptor function. Another was the progressive increase in responsiveness to repeated D1-dopamine agonist administration referred to as "priming" of D1-dopamine receptor function. Additionally, a unique profile of signaling protein expression related to neonate reduction of dopamine has been identified. Thus, from modeling characteristics of disease to defining adaptive mechanisms related to neonatal loss of dopamine, the neonate-lesioned rat has had a persisting influence on neuroscience. Despite an extraordinary legacy from studies of the neurobiology of this treatment, a host of unknowns remain that will inspire future investigations.


Subject(s)
Brain Chemistry/drug effects , Disease Models, Animal , Mental Disorders/metabolism , Oxidopamine/pharmacology , Age Factors , Animals , Animals, Newborn , Brain/metabolism , Brain/physiopathology , Brain Chemistry/physiology , Dopamine/metabolism , Excitatory Amino Acid Agonists/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Mental Disorders/physiopathology , Neurology/methods , Neurology/trends , Rats , Receptors, Dopamine/drug effects , Receptors, Dopamine/metabolism , Sympatholytics/pharmacology
18.
Brain Res ; 1015(1-2): 107-13, 2004 Jul 23.
Article in English | MEDLINE | ID: mdl-15223373

ABSTRACT

Volatile hydrocarbon based CNS depressants including short chain alcohols and anesthetics act, in part, by inhibition of the excitatory effect of glutamate at the NMDA receptor. While effects of several of these volatile agents on NMDA-gated currents have been demonstrated, there has been no direct comparison of different chemical classes of CNS depressant drugs on NMDA-gated currents. Here, whole-cell voltage clamp measurements of currents gated by 100 microM NMDA from cultured cerebrocortical neurons were examined in the presence of varying concentrations of the alcohols ethanol and hexanol, the halogenated alcohol trichloroethanol, the halogenated alkane halothane and the halogenated ethers isoflurane and sevoflurane. All drugs tested showed concentration-dependent inhibition of NMDA-gated currents with anesthetic concentrations of each agent producing approximately 30% inhibition of the NMDA-gated current. A rapid-translation perfusion system was used to study the onset and offset kinetics of each of the volatile agents. Onset kinetics for the CNS depressants was similar with tau values near 100 ms. Offset kinetics was more variable with tau ranging from 88.2 ms for ethanol to 221.4 ms for trichloroethanol. These data indicate that a wide variety of volatile hydrocarbon based CNS depressants produce a similar inhibition of NMDA-gated currents and that the kinetics for these agents are inconsistent with an open channel block.


Subject(s)
Anesthetics, Inhalation/pharmacology , Central Nervous System Depressants/pharmacology , Ethylene Chlorohydrin/analogs & derivatives , Membrane Potentials/drug effects , Neurons/drug effects , Receptors, N-Methyl-D-Aspartate/drug effects , Alcohols/pharmacology , Animals , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/drug effects , Dose-Response Relationship, Drug , Ethanol/pharmacology , Ethylene Chlorohydrin/pharmacology , Halothane/pharmacology , Hexanols/pharmacology , Ion Channel Gating/drug effects , Isoflurane/pharmacology , Methyl Ethers/pharmacology , Models, Biological , Patch-Clamp Techniques , Rats , Sevoflurane
19.
Life Sci ; 74(24): 3043-52, 2004 Apr 30.
Article in English | MEDLINE | ID: mdl-15051427

ABSTRACT

The effects of propyl paraben, an antimicrobial preservative, on voltage-dependent sodium current and myocardial ischemia-reperfusion injury were investigated in isolated adult rat cardiomyocytes. Whole cell voltage-clamp recording showed that propyl paraben reversibly blocked the voltage-gated sodium channel both in concentration- and voltage-dependent manners. Propyl paraben (500 microM but not 100 microM) significantly shifted the steady-state inactivation of the sodium channel toward the hyperpolarizing direction at the V(1/2) point. Consistent with the above result, the propidium iodide (PI) uptake test revealed that pretreatment with 500 microM but not 100 microM of propyl paraben significantly reduced cell death induced by 45 min of sustained ischemia followed by 15 h of reperfusion (42.37 +/- 7.01% of cell viability in control and 71.05 +/- 7.06% in the propyl paraben group), suggesting that propyl paraben can protect myocytes from ischemia-reperfusion injury. These results indicate a possible correlation between the inhibition of sodium current and cardioprotection against ischemia-reperfusion injury.


Subject(s)
Myocardial Reperfusion Injury/prevention & control , Myocytes, Cardiac/drug effects , Parabens/pharmacology , Sodium Channel Blockers/pharmacology , Sodium Channels/drug effects , Animals , Cell Survival/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Guanidines/pharmacology , Ion Channel Gating/drug effects , Ion Channel Gating/physiology , Male , Myocardial Reperfusion Injury/physiopathology , Myocytes, Cardiac/metabolism , Patch-Clamp Techniques , Propidium/metabolism , Rats , Rats, Wistar , Sodium Channels/physiology , Sulfones/pharmacology
20.
J Pharmacol Exp Ther ; 304(1): 192-9, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12490591

ABSTRACT

In vivo, ethanol alters the effect of N-methyl-D-aspartate (NMDA) and GABA in some brain regions but is without effect in others. To determine whether these regional differences were due to differences in the effect of ethanol on postsynaptic NMDA or GABAA receptors, we examined the effect of ethanol on NMDA- and GABA-gated currents from neurons acutely dissociated from the lateral septal nucleus, substantia nigra, thalamus, hippocampus, and cerebellum. Ethanol decreased the effect of NMDA similarly in all brain areas tested and had similar effects on Chinese hamster ovary cells expressing NR2A or NR2B subunits with an NR1-1a subunit. However, ifenprodil reduced the inhibition by ethanol of NMDA-gated currents from neurons isolated from the lateral septum without affecting neurons from the substantia nigra. In contrast to the robust effect of ethanol on NMDA-gated currents, ethanol (25-300 mM) was without effect on GABA-gated currents at all brain sites tested or on Ltk- cells stably expressing the alpha1, beta2, and gamma2L or gamma2S subunits. The neuroactive steroid alphaxalone profoundly enhanced GABA-gated currents in all brain areas and cell types tested, indicating a similar sensitivity to allosteric modulation; however, there was no interaction of alphaxalone with ethanol at any site tested. These data suggest that the regional differences in the effect of ethanol observed in vivo are not due to a differential action of ethanol at the postsynaptic NMDA or GABAA receptor subtypes.


Subject(s)
Brain Chemistry/drug effects , Central Nervous System Depressants/pharmacology , Ethanol/pharmacology , Excitatory Amino Acid Agonists/pharmacology , Ion Channel Gating/drug effects , N-Methylaspartate/pharmacology , Neurons/drug effects , gamma-Aminobutyric Acid/pharmacology , Anesthetics/pharmacology , Animals , Brain/cytology , CHO Cells , Cell Line , Cricetinae , Electrophysiology , Excitatory Amino Acid Antagonists/pharmacology , Excitatory Postsynaptic Potentials/drug effects , Neurons/metabolism , Patch-Clamp Techniques , Piperidines/pharmacology , Pregnanediones/pharmacology , Rats , Receptors, GABA-A/drug effects , Receptors, N-Methyl-D-Aspartate/drug effects , Receptors, Neurotransmitter/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...