Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 69
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.
Pharmacol Biochem Behav ; 168: 33-42, 2018 05.
Article in English | MEDLINE | ID: mdl-29572015

ABSTRACT

Neuroimmune cytokines are increased with alcohol withdrawal and may mediate clinical responses associated with alcoholism. Because minocycline regulates the level of cytokines, it has been suggested as a therapeutic for disorders associated with alcohol. Male Wistar rats were exposed to chronic intermittent alcohol (CIA) comprising three 5-day cycles of ethanol liquid diet separated by 2 days of withdrawal. Rats were tested on social interaction, a measure of anxiety-like behavior, followed immediately by collection of amygdala tissue to measure CCL2 and TNFα or collection of the blood to measure corticosterone (CORT). One group received a single minocycline injection 3 h into the final CIA withdrawal and was tested 2 h later. A second group received injections during each of the three withdrawals and was similarly tested during the final acute withdrawal. A third group received a single injection at 23 h into withdrawal (extended withdrawal) and was tested 6 h later. Results showed that CIA withdrawal increased anxiety-like behavior. A single injection of minocycline during the final acute withdrawal increased anxiety-like behavior in rats that consumed liquid diet with or without alcohol, but this effect disappeared with repeated injections of minocycline. Differences in alcohol intake, blood alcohol level, and plasma CORT levels did not explain results. Only repeated injections of minocycline decreased TNFα mRNA levels in rats that consumed liquid diet with or without alcohol. When a single injection of minocycline was given during extended withdrawal, it decreased CCL2 mRNA levels, but did not reverse the elevation of CCL2 protein. These results suggest that minocycline has actions in brain and on behavior, but minocycline does not significantly impact these actions in relation to alcohol withdrawal.


Subject(s)
Alcoholism/blood , Anti-Bacterial Agents/administration & dosage , Minocycline/administration & dosage , Amygdala/drug effects , Amygdala/metabolism , Animals , Anxiety/chemically induced , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Corticosterone/blood , Drug Administration Schedule , Drug Interactions , Male , RNA, Messenger/metabolism , Rats, Wistar , Substance Withdrawal Syndrome/metabolism , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
5.
Psychopharmacology (Berl) ; 234(1): 79-88, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27665607

ABSTRACT

RATIONALE: Behavioral and neuroimmune vulnerability to withdrawal from chronic alcohol varies with age. The relation of anxiety-like behavior to amygdalar CCL2 responses following stress after withdrawal from chronic intermittent alcohol (CIA) was investigated in adolescent and adult rats. METHODS: Adolescent and adult Wistar rats were exposed to CIA (three 5-day blocks of dietary alcohol separated by 2 days of withdrawal) at concentrations that created similar blood alcohol levels across age. Twenty-four hours into the final withdrawal, half of the rats were exposed to 1 h of restraint stress. Four hours post-stress, rats were used for behavior or tissue assays. RESULTS: Anxiety-like behavior was increased versus controls by CIA in adolescents and by CIA + stress in adults. CCL2 mRNA was increased versus controls by CIA in adolescents and by CIA and CIA + stress in adults. CCL2 co-localization with neuronal marker NeuN was decreased versus controls by CIA in adolescents and by CIA + stress in adults. CCL2 co-localization with astrocytic marker GFAP was decreased versus controls by CIA and CIA + stress in adolescents, but experimental groups did not differ from controls in adults. CCL2 co-localization with microglial marker Iba1 was decreased versus controls by stress alone in adolescents and by CIA + stress in adults. CONCLUSIONS: Changes in CCL2 protein might control behavior at either age but are particularly associated with CIA alone in adolescents and with CIA + stress in adults. That the number of CeA neurons expressing CCL2 was altered after CIA and stress is consistent with CCL2 involvement in neural function.


Subject(s)
Amygdala/metabolism , Anxiety/metabolism , Behavior, Animal/physiology , Chemokine CCL2/metabolism , Ethanol/adverse effects , Substance Withdrawal Syndrome/metabolism , Animals , Anxiety/chemically induced , Male , Rats , Rats, Sprague-Dawley , Rats, Wistar
6.
Pain ; 158(3): 457-462, 2017 03.
Article in English | MEDLINE | ID: mdl-27918314

ABSTRACT

Chronic pain conditions are often comorbid with alcohol abuse. "Self-medication" with alcohol introduces a host of problems associated with the abuse of alcohol which over time has the potential of exacerbating the painful condition. Despite the prevalence of chronic pain being associated with alcohol abuse, rodent models which mimic the comorbid conditions are lacking. In this study, we model osteoarthritis (OA) in C57BL/6J mice by surgically destabilizing the medial meniscus (DMM). Sham-operated mice served as controls. Thirteen weeks after surgery, DMM but not sham-operated mice exhibited pronounced incapacitance of the surgically manipulated hind limb compared with the nonsurgically manipulated hind limb. At this time, the mice were exposed to the 2-bottle ethanol choice, beginning with 2.5% with a gradual increasing to 20%. Compared with sham controls, DMM mice consumed more EtOH and preferred EtOH over water at the 20% EtOH concentration. Histological analysis verified that the DMM mice exhibited significant damage to the articular cartilage and osteophyte growth compared with sham controls and these measures of the severity of OA correlated with the amount of ethanol intake. Thus, the combination of the DMM model of OA with the enhanced two-bottle ethanol choice is a potential preclinical approach in mice by which the basis of the comorbid association of alcohol abuse and chronic pain conditions can be explored.


Subject(s)
Alcohol Drinking/physiopathology , Ethanol/metabolism , Osteoarthritis, Knee/physiopathology , Analysis of Variance , Animals , Choice Behavior/physiology , Disease Models, Animal , Disease Progression , Male , Mice , Mice, Inbred C57BL
7.
Brain Sci ; 6(3)2016 Jul 27.
Article in English | MEDLINE | ID: mdl-27472367

ABSTRACT

Stress is a strong risk factor in alcoholic relapse and may exert effects that mimic aspects of chronic alcohol exposure on neurobiological systems. With the neuroimmune system becoming a prominent focus in the study of the neurobiological consequences of stress, as well as chronic alcohol exposure proving to be a valuable focus in this regard, the present study sought to compare the effects of stress and chronic ethanol exposure on induction of components of the neuroimmune system. Rats were exposed to either 1 h exposure to a mild stressor (restraint) or exposure to withdrawal from 15 days of chronic alcohol exposure (i.e., withdrawal from chronic ethanol, WCE) and assessed for neuroimmune mRNAs in brain. Restraint stress alone elevated chemokine (C-C motif) ligand 2 (CCL2), interleukin-1-beta (IL-1ß), tumor necrosis factor alpha (TNFα) and toll-like receptor 4 (TLR4) mRNAs in the cerebral cortex within 4 h with a return to a control level by 24 h. These increases were not accompanied by an increase in corresponding proteins. Withdrawal from WCE also elevated cytokines, but did so to varying degrees across different cytokines and brain regions. In the cortex, stress and WCE induced CCL2, TNFα, IL-1ß, and TLR4 mRNAs. In the hypothalamus, only WCE induced cytokines (CCL2 and IL-1ß) while in the hippocampus, WCE strongly induced CCL2 while stress and WCE induced IL-1ß. In the amygdala, only WCE induced CCL2. Finally-based on the previously demonstrated role of corticotropin-releasing factor 1 (CRF1) receptor inhibition in blocking WCE-induced cytokine mRNAs-the CRF1 receptor antagonist CP154,526 was administered to a subgroup of stressed rats and found to be inactive against induction of CCL2, TNFα, or IL-1ß mRNAs. These differential results suggest that stress and WCE manifest broad neuroimmune effects in brain depending on the cytokine and brain region, and that CRF inhibition may not be a relevant mechanism in non-alcohol exposed animals. Overall, these effects are complex in terms of their neuroimmune targets and neuroanatomical specificity. Further investigation of the differential distribution of cytokine induction across neuroanatomical regions, individual cell types (e.g., neuronal phenotypes and glia), severity of chronic alcohol exposure, as well as across differing stress types may prove useful in understanding differential mechanisms of induction and for targeting select systems for pharmacotherapeutic intervention in alcoholism.

8.
Alcohol ; 52: 9-23, 2016 05.
Article in English | MEDLINE | ID: mdl-27139233

ABSTRACT

This review updates the conceptual basis for the association of alcohol abuse with an insidious adaptation that facilitates negative affect during withdrawal from chronic intermittent alcohol (CIA) exposure - a change that later supports sensitization of stress-induced anxiety following alcohol abstinence. The finding that a CRF1-receptor antagonist (CRF1RA) minimized CIA withdrawal-induced negative affect supported an association of alcohol withdrawal with a stress mechanism. The finding that repeated stresses or multiple CRF injections into selected brain sites prior to a single 5-day chronic alcohol (CA) exposure induced anxiety during withdrawal provided critical support for a linkage of CIA withdrawal with stress. The determination that CRF1RA injection into positive CRF-sensitive brain sites prevented CIA withdrawal-induced anxiety provided support that neural path integration maintains the persistent CIA adaptation. Based upon reports that stress increases neuroimmune function, an effort was undertaken to test whether cytokines would support the adaptation induced by stress/CA exposure. Twenty-four hours after withdrawal from CIA, cytokine mRNAs were found to be increased in cortex as well as other sites in brain. Further, repeated cytokine injections into previously identified brain sites substituted for stress and CRF induction of anxiety during CA withdrawal. Discovery that a CRF1RA prevented the brain cytokine mRNA increase induced by CA withdrawal provided critical evidence for CRF involvement in this neuroimmune induction after CA withdrawal. However, the CRF1RA did not block the stress increase in cytokine mRNA increases in controls. The latter data supported the hypothesis that distinct mechanisms linked to stress and CA withdrawal can support common neuroimmune functions within a brain site. As evidence evolves concerning neural involvement in brain neuroimmune function, a better understanding of the progressive adaptation associated with CIA exposure will advance new knowledge that could possibly lead to strategies to combat alcohol abuse.


Subject(s)
Alcoholism/metabolism , Corticotropin-Releasing Hormone/metabolism , Neuroimmunomodulation/physiology , Stress, Psychological/metabolism , Substance Withdrawal Syndrome/metabolism , Alcoholism/immunology , Alcoholism/psychology , Animals , Corticotropin-Releasing Hormone/immunology , Humans , Receptors, Corticotropin-Releasing Hormone/immunology , Receptors, Corticotropin-Releasing Hormone/metabolism , Stress, Psychological/immunology , Stress, Psychological/psychology , Substance Withdrawal Syndrome/immunology
9.
Curr Top Behav Neurosci ; 29: 265-277, 2016.
Article in English | MEDLINE | ID: mdl-27029809

ABSTRACT

Lesch-Nyhan disease is a neurologically, metabolically, and behaviorally devastating condition that has eluded complete characterization and adequate treatment. While it is known that the disease is intimately associated with dysfunction of the hypoxanthine phosphoribosyltransferase 1 (HPRT1) gene that codes for an enzyme of purine metabolism (hypoxanthine-guanine phosphoribosyltransferase) and is associated with neurological, behavioral, as well as metabolic dysfunction, the mechanisms of the neurobehavioral manifestations are as yet unclear. However, discoveries over the past few decades not only have created useful novel animal models (e.g., the HPRT-deficient mouse and the serendipitously discovered perinatal 6-hydroxydopamine (6-OHDA lesion model), but also have expanded into epigenetic, genomic, and proteomic approaches to better understand the mechanisms underlying this disease. The perinatal 6-OHDA model, in addition to modeling self-injury and dopamine depletion in the clinical condition, also underscores the profound importance of development in the differential course of maladaptive progression in the face of a common/single neurotoxic insult at different ages. Recent developments from clinical and basic science efforts attest to the fact that while the disease would seem to have a simple single gene defect at its core, the manifestations of this defect are profound and unexpectedly diverse. Future efforts employing the 6-OHDA model and others in the context of the novel technologies of genome editing, chemo- and opto-genetics, epigenetics, and further studies on the mechanisms of stress-induced maladaptations in brain all hold promise in taking our understanding of this disease to the next level.


Subject(s)
Adrenergic Agents/toxicity , Lesch-Nyhan Syndrome/chemically induced , Oxidopamine/toxicity , Animals , Disease Models, Animal , Neurotoxins/toxicity , Rats
10.
Alcohol Clin Exp Res ; 39(12): 2375-85, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26556523

ABSTRACT

BACKGROUND: The role of neuroimmune activation in withdrawal from chronic alcohol (ethanol) has been established in both adolescent and adult models, but direct comparisons across age are sparse. Studies need to elucidate age-dependent neuroimmune effects of alcohol and to focus research attention on age-dependent mechanisms and outcomes. METHODS: Adult and adolescent rats from 2 commonly used strains, Wistar and Sprague Dawley (SD), were maintained on continuous 7%, 5.35%, 4.5% alcohol diet (CAD) or cycled 7% w/v alcohol diet for 15 days. Cortical tissue was collected at 0, 8, 16, and 24 hours postwithdrawal followed by measurement of chemokine (C-C motif) ligand 2 (CCL2), tumor necrosis factor alpha, and interleukin 1 beta mRNA with quantitative real-time polymerase chain reaction. RESULTS: Both age groups and strains showed a strong cytokine mRNA response at 7% CAD. Further, a greater increase in CCL2 mRNA was observed in the cortex of adolescents at 7% CAD, which correlated with higher blood alcohol levels (BALs). Adolescents exposed to 5.35% CAD exhibited similar blood levels and cytokine responses as adults exposed to 7% CAD. Substantial variability in CCL2 mRNA responses was found only in adolescent rats exposed to 7% CAD. In this group, data could be segregated into high-responding and low-responding groups. Moreover, the data from the high-responding group were associated with seizures. CONCLUSIONS: Relative to other cytokine mRNAs, CCL2 exhibits a unique response profile during withdrawal from CAD. This profile is shown in adolescents, where CCL2 is uniquely influenced by the effects of seizures. Additionally, this profile is shown by the fact that only CCL2 expression correlated with BAL that transcended age groups. These data emphasize the importance of BALs and treatment regimen on developmental neuroimmune responses and suggest that select components of the neuroimmune system are more responsive to CAD withdrawal and that neurobiological mechanisms differentiating these responses should be further explored.


Subject(s)
Alcoholism/metabolism , Blood Alcohol Content , Brain/metabolism , Chemokine CCL2/biosynthesis , RNA, Messenger/biosynthesis , Substance Withdrawal Syndrome/metabolism , Age Factors , Animals , Brain/drug effects , Ethanol/administration & dosage , Ethanol/toxicity , Male , Rats , Rats, Sprague-Dawley , Rats, Wistar
11.
Alcohol Clin Exp Res ; 37(12): 2086-97, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23895427

ABSTRACT

BACKGROUND: Many neurobiological factors may initiate and sustain alcoholism. Recently, dysregulation of the neuroimmune system by chronic ethanol (CE) has implicated Toll-like receptor 4 (TLR4) activation. Even though TLR4s are linked to CE initiation of brain cytokine mRNAs, the means by which CE influences neuroimmune signaling in brain in the absence of infection remains uncertain. Therefore, the hypothesis is tested that release of an endogenous TLR4 agonist, high-mobility group box 1 (HMGB1) and/or corticotropin-releasing factor (CRF) during CE withdrawal are responsible for CE protocols increasing cytokine mRNAs. METHODS: Acute ethanol (EtOH; 2.75 g/kg) and acute lipopolysaccharide (LPS; 250 µg/kg) dosing on cytokine mRNAs are first compared. Then, the effects of chronic LPS exposure (250 µg/kg for 10 days) on cytokine mRNAs are compared with changes induced by CE protocols (15 days of continuous 7% EtOH diet [CE protocol] or 3 intermittent 5-day cycles of 7% EtOH diet [CIE protocol]). Additionally, TLR4, HMGB1, and downstream effector mRNAs are assessed after CE, CIE, and chronic LPS. To test whether HMGB1 and/or CRF support the CE withdrawal increase in cytokine mRNAs, the HMGB1 antagonists, glycyrrhizin and ethyl pyruvate, and a CRF1 receptor antagonist (CRF1RA) are administered during 24 hours of CE withdrawal. RESULTS: While cytokine mRNAs were not increased following acute EtOH, acute LPS increased all cytokine mRNAs 4 hours after injection. CE produced no change in cytokine mRNAs prior to CE removal; however, the CE and CIE protocols increased cytokine mRNAs by 24 hours after withdrawal. In contrast, chronic LPS produced no cytokine mRNA changes 24 hours after LPS dosing. TLR4 mRNA was elevated 24 hours following both CE protocols and chronic LPS exposure. While chronic LPS had no effect on HMGB1 mRNA, withdrawal from CE protocols significantly elevated HMGB1 mRNA. Systemic administration of HMGB1 antagonists or a CRF1RA significantly reduced the cytokine mRNA increase following CE withdrawal. The CRF1RA and the HMGB1 antagonist, ethyl pyruvate, also reduced the HMGB1 mRNA increase that followed CE withdrawal. CONCLUSIONS: By blocking HMGB1 or CRF action during CE withdrawal, evidence is provided that HMGB1 and CRF release are critical for the CE withdrawal induction of selected brain cytokine mRNAs. Consequently, these results clarify a means by which withdrawal from CE exposure activates neuroimmune function in the sterile milieu of brain.


Subject(s)
Brain/physiopathology , Corticotropin-Releasing Hormone/physiology , Cytokines/genetics , Ethanol/administration & dosage , HMGB1 Protein/physiology , Substance Withdrawal Syndrome/physiopathology , Animals , Corticotropin-Releasing Hormone/antagonists & inhibitors , HMGB1 Protein/antagonists & inhibitors , HMGB1 Protein/genetics , Lipopolysaccharides/administration & dosage , Male , RNA, Messenger/analysis , Rats , Rats, Sprague-Dawley , Toll-Like Receptor 4/genetics
12.
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
13.
Methods Mol Biol ; 829: 205-30, 2012.
Article in English | MEDLINE | ID: mdl-22231816

ABSTRACT

Alcoholism is a chronic treatment-resistant disorder typically presenting with recurrent/cyclic periods of abusive drinking, withdrawal, abstinence, and relapse. Various strategies that attempt to model these processes in animals have been developed to elucidate the behavioral and neural processes underlying alcoholism. Many of these have involved chronic ethanol exposure and withdrawal with the most widely employed methods involving mice or rats. Prominent features of these methods include alcohol vapor or intragastric forced exposure, cyclic or intermittent periods of alcohol availability with various lengths of forced abstinence, voluntary consumption, the use of genetically alcohol-preferring animals, and inclusion of various pharmacological or environmental challenges to worsen or mitigate symptoms. This chapter emphasizes alcohol exposure and withdrawal and discusses representative metrics used to monitor the consequences of employing these methods. These include but are not limited to intensity and pattern of alcohol exposure, seizure sensitivity during withdrawal, and emotional responding.


Subject(s)
Alcohol Drinking , Alcoholic Intoxication/pathology , Alcoholism/pathology , Alcohol Withdrawal Seizures , Alcoholism/metabolism , Animals , Disease Models, Animal , Ethanol/administration & dosage , Ethanol/blood , Mice , Rats
14.
Addict Biol ; 17(2): 338-50, 2012 Mar.
Article in English | MEDLINE | ID: mdl-21762289

ABSTRACT

Neuropeptide Y (NPY) and protein kinase A (PKA) have been implicated in neurobiological responses to ethanol. We have previously reported that mutant mice lacking normal production of the RIIß subunit of PKA (RIIß-/- mice) show enhanced sensitivity to the locomotor stimulant effects of ethanol and increased behavioral sensitization relative to littermate wild-type RIIß+/+ mice. We now report that RIIß-/- mice also show increased NPY immunoreactivity in the nucleus accumbens (NAc) core and the ventral striatum relative to RIIß+/+ mice. These observations suggest that elevated NPY signaling in the NAc and/or striatum may contribute to the increased sensitivity to ethanol-induced behavioral sensitization that is a characteristic of RIIß-/- mice. Consistently, NPY-/- mice failed to display ethanol-induced behavioral sensitization that was evident in littermate NPY+/+ mice. To examine more directly the role of NPY in the locomotor stimulant effects of ethanol, we infused a recombinant adeno-associated virus (rAAV) into the region of the NAc core of DBA/2J mice. The rAAV-fibronectin (FIB)-NPY(13-36) vector expresses and constitutively secretes the NPY fragment NPY(13-36) (a selective Y(2) receptor agonist) from infected cells in vivo. Mice treated with the rAAV-FIB-NPY(13-36) vector exhibited reduced expression of ethanol-induced behavioral sensitization compared with mice treated with a control vector. Taken together, the current data provide the first evidence that NPY signaling in the NAc core and the Y(2) receptor modulate ethanol-induced behavioral sensitization.


Subject(s)
Basal Ganglia/metabolism , Central Nervous System Depressants/pharmacology , Cyclic AMP-Dependent Protein Kinases/metabolism , Ethanol/pharmacology , Neuropeptide Y/metabolism , Nucleus Accumbens/metabolism , Alcohol Drinking/metabolism , Alcohol Drinking/psychology , Animals , Behavior, Animal/drug effects , Male , Mice , Mice, Inbred Strains , Motor Activity/drug effects , Neuropeptide Y/pharmacology , Peptide Fragments/pharmacology , Receptors, Neuropeptide Y/antagonists & inhibitors , Reinforcement, Psychology , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/physiology
15.
Psychopharmacology (Berl) ; 218(1): 179-89, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21643675

ABSTRACT

RATIONALE: Stress may elevate ethanol drinking and anxiety associated with ethanol drinking. Studies to identify relevant neurobiological substrates are needed. OBJECTIVE: To assess roles of brain regions in corticotrophin releasing factor (CRF) effects on stressor-enhanced, ethanol deprivation-induced drinking and anxiety-like behavior. METHODS: Ethanol-preferring rats (P rats) were exposed to three cycles of a two-bottle choice paradigm with two 2-day deprivation periods that included 1 h exposure to a restraint stressor. To assess the role of CRF and to identify relevant brain regions, a CRF-1 receptor antagonist (SSR125543; 10 ug) was injected into the nucleus accumbens (NAC), amygdala (Amyg), or dorsal raphe nucleus (DRN) prior to exposure to the restraint stressor. In a second study, CRF (0.5 ug) was injected into one of these regions, or the ventral tegmental area (VTA), or paraventricular nucleus of the hypothalamus (PVN). RESULTS: Applying the restraint stressor during deprivation increased voluntary intake and sensitized anxiety-like behavior. Antagonist injection into the NAC prevented increased drinking without affecting anxiety-like behavior, whereas injection into the Amyg or DRN prevented the anxiety-like behavior without affecting drinking. To confirm CRF actions in the stressor effect, CRF was injected into selected brain regions. NAC injections (but not the VTA, Amyg, DRN, or PVN) facilitated drinking but did not change anxiety-like behavior. Injections into the DRN or Amyg (but not PVN or VTA) enhanced anxiety-like behavior. CONCLUSIONS: Results emphasize that a restraint stressor elevates ethanol intake and sensitizes ethanol deprivation-induced anxiety-like behavior through CRF1 receptors in the NAC and Amyg/DRN, respectively.


Subject(s)
Anxiety/etiology , Corticotropin-Releasing Hormone/administration & dosage , Ethanol/administration & dosage , Stress, Psychological/complications , Animals , Brain/metabolism , Disease Models, Animal , Hydrocarbons, Halogenated/pharmacology , Male , Rats , Receptors, Corticotropin-Releasing Hormone/antagonists & inhibitors , Receptors, Corticotropin-Releasing Hormone/metabolism , Thiazines/pharmacology
16.
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
17.
Pharmacol Ther ; 129(2): 149-71, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20951730

ABSTRACT

Alcoholism is a chronic relapsing disorder. Major characteristics observed in alcoholics during an initial period of alcohol abstinence are altered physiological functions and a negative emotional state. Evidence suggests that a persistent, cumulative adaptation involving a kindling/allostasis-like process occurs during the course of repeated chronic alcohol exposures that is critical for the negative symptoms observed during alcohol withdrawal. Basic studies have provided evidence for specific neurotransmitters within identified brain sites being responsible for the negative emotion induced by the persistent cumulative adaptation following intermittent-alcohol exposures. After an extended period of abstinence, the cumulative alcohol adaptation increases susceptibility to stress- and alcohol cue-induced negative symptoms and alcohol seeking, both of which can facilitate excessive ingestion of alcohol. In the alcoholic, stressful imagery and alcohol cues alter physiological responses, enhance negative emotion, and induce craving. Brain fMRI imaging following stress and alcohol cues has documented neural changes in specific brain regions of alcoholics not observed in social drinkers. Such altered activity in brain of abstinent alcoholics to stress and alcohol cues is consistent with a continuing ethanol adaptation being responsible. Therapies in alcoholics found to block responses to stress and alcohol cues would presumably be potential treatments by which susceptibility for continued alcohol abuse can be reduced. By continuing to define the neurobiological basis of the sustained alcohol adaptation critical for the increased susceptibility of alcoholics to stress and alcohol cues that facilitate craving, a new era is expected to evolve in which the high rate of relapse in alcoholism is minimized.


Subject(s)
Alcoholism/physiopathology , Alcoholism/psychology , Allostasis , Behavior, Addictive , Kindling, Neurologic , Stress, Psychological , Alcohol Drinking/physiopathology , Alcoholism/genetics , Alcoholism/therapy , Animals , Cues , Female , Humans , Male , Recurrence , Substance Withdrawal Syndrome/metabolism , Substance Withdrawal Syndrome/physiopathology , Temperance
18.
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
19.
Alcohol Clin Exp Res ; 34(9): 1603-12, 2010 Sep 01.
Article in English | MEDLINE | ID: mdl-20586753

ABSTRACT

BACKGROUND: Repeated stress or administration of corticotropin-releasing factor (CRF) prior to ethanol exposure sensitizes anxiety-like behavior in adult rats. Current experiments determined whether adolescent rats were more sensitive to these challenges in sensitizing ethanol withdrawal-induced anxiety and altering CRF levels in brain during withdrawal. METHODS: Male adult and adolescent Sprague-Dawley rats were restraint stressed (1 hour) twice 1 week apart prior to a single 5-day cycle of ethanol diet (ED; stress/withdrawal paradigm). Other rats received control diet (CD) and three 1-hour restraint stress sessions. Rats were then tested 5, 24, or 48 hours after the final withdrawal for anxiety-like behavior in the social interaction (SI) test. In other experiments, adolescent rats were given two microinjections of CRF icv 1 week apart followed by 5 days of either CD or ED and tested in social interaction 5 hours into withdrawal. Finally, CRF immunoreactivity was measured in the central nucleus of the amygdala (CeA) and paraventricular nucleus (PVN) after rats experienced control diet, repeated ethanol withdrawals, or stress/withdrawal. RESULTS: Rats of both ages had reduced SI following the stress/withdrawal paradigm, and this effect recovered within 24 hours. Higher CRF doses were required to reduce SI in adolescents than previously reported in adults. CRF immunohistochemical levels were higher in the PVN and CeA of CD-exposed adolescents. In adolescent rats, repeated ethanol withdrawals decreased CRF in the CeA but was not associated with decreased CRF cell number. There was no change in CRF from adult treatments. CONCLUSIONS: In the production of anxiety-like behavior, adolescent rats have equal sensitivity with stress and lower sensitivity with CRF compared to adults. Further, adolescents had higher basal levels of CRF within the PVN and CeA and reduced CRF levels following repeated ethanol withdrawals. This reduced CRF within the CeA could indicate increased release of CRF, and future work will determine how this change relates to behavior.


Subject(s)
Anxiety/chemically induced , Corticotropin-Releasing Hormone/pharmacology , Ethanol/pharmacology , Stress, Psychological/psychology , Substance Withdrawal Syndrome/psychology , Administration, Oral , Age Factors , Amygdala/metabolism , Animals , Anxiety/complications , Corticotropin-Releasing Hormone/administration & dosage , Ethanol/administration & dosage , Injections, Intraventricular , Male , Paraventricular Hypothalamic Nucleus/metabolism , Rats , Rats, Sprague-Dawley
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...