Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
Add more filters










Publication year range
1.
Neuropharmacology ; 228: 109457, 2023 05 01.
Article in English | MEDLINE | ID: mdl-36764577

ABSTRACT

The dynorphin (DYN)/kappa-opioid receptor (KOR) system is involved in dysphoria and negative emotional states. Dysregulation of KOR function promotes maladaptive behavioral regulation during withdrawal associated with alcohol dependence. Mesolimbic dopaminergic (DA) projections from the ventral tegmental area (VTA) innervate the extended amygdala circuitry and presynaptic KORs attenuate DA in these regions leading to an excessive alcohol consumption and negative affective-like behavior, whereas mesocortical KOR-regulated DA projections have been implicated in executive function and decision-making. Thus, the neuroadaptations occurring in DYN/KOR systems are important aspects to consider for the development of personalized therapeutic solutions. Herein, we study the contribution of the VTA DA neuron Oprk1 (KOR gene) in excessive alcohol consumption, negative emotional state, and executive function. To do so, Oprk1 mRNA expression and KOR function were characterized to confirm alcohol dependence-induced dysregulation in the VTA. Then, a transgenic Cre-Lox rat model (male and female TH::Cre rats) was used to allow for conditional and inducible overexpression of Oprk1 in VTA DA neurons. The effect of this overexpression was evaluated on operant alcohol self-administration, negative emotional states, and executive function. We found that VTA Oprk1 overexpression recapitulates some phenotypes of alcohol dependence including escalated alcohol self-administration and depressive-like behavior. However, working memory performance was not impacted following VTA Oprk1 overexpression in TH::Cre rats. This supports the hypothesis that dysregulated KOR signaling within the mesolimbic DA system is an important contributor to symptoms of alcohol dependence and shows that understanding Oprk1-mediated contributions to alcohol use disorder (AUD) should be an important future goal.


Subject(s)
Alcoholism , Rats , Male , Female , Animals , Alcoholism/metabolism , Ventral Tegmental Area/metabolism , Receptors, Opioid, kappa/metabolism , Dynorphins/metabolism , Ethanol , Dopamine/metabolism , Phenotype
2.
Addict Biol ; 27(2): e13138, 2022 03.
Article in English | MEDLINE | ID: mdl-35138672

ABSTRACT

Impaired working memory is one symptom contributing to compromised executive function in alcohol use disorder (AUD). Dysregulation of cortical dynorphin (DYN) and κ-opioid receptors (KORs) has been implicated in alcohol dependence-induced impairment in executive function. The present experiments test the hypothesis that dysregulated medial prefrontal cortex (mPFC) KORs contribute to impaired working memory in alcohol dependence. Alcohol dependence was induced in male Wistar rats via 4 months of intermittent ethanol vapor exposure prior to training/testing in an mPFC-dependent working memory task (delayed nonmatching-to-sample task; DNMST). mPFC KOR function in alcohol-naïve rats was compared with that of alcohol-dependent and nondependent rats using a DYN A-stimulated [35S ]GTPγS coupling assay. A functional role for mPFC KORs in the regulation of working memory was assessed via intra-mPFC infusions of a KOR agonist prior to assessment in the DNMST, and the contribution of mPFC KORs to compromised working memory in dependence was assessed via mPFC infusions of the KOR antagonist norbinaltorphimine (nor-BNI). In alcohol-dependent rats, impaired performance in the DNMST confirmed compromised working memory. Furthermore, DYN A-stimulated mPFC KOR function was pathologically increased in alcohol-dependent rats compared with nondependent and alcohol-naïve rats. Additionally, mPFC KOR involvement in working memory was functionally confirmed by intra-mPFC KOR agonist-induced deficits in DNMST performance. Importantly, alcohol dependence-induced impairment in the DNMST was ameliorated by intra-mPFC KOR antagonism. Regulation of working memory by mPFC KORs and alcohol dependence-induced dysregulation of mPFC KOR function identify a novel therapeutic target to treat AUD-related symptoms of working memory impairment.


Subject(s)
Alcoholism , Receptors, Opioid, kappa , Alcoholism/complications , Alcoholism/drug therapy , Animals , Male , Memory Disorders , Memory, Short-Term , Prefrontal Cortex/metabolism , Rats , Rats, Wistar
3.
Addict Biol ; 25(2): e12715, 2020 03.
Article in English | MEDLINE | ID: mdl-30648329

ABSTRACT

A role for matrix metalloproteinases (MMPs) in plasticity-dependent learning has been established. MMPs degrade the extracellular matrix (ECM) when synaptic reorganization is warranted. Previously, we showed that escalation of alcohol self-administration is a learned plasticity-dependent process that requires an intact MMP system. To identify the MMP subtypes within specific brain regions that are associated with plasticity underlying the negative reinforcing effects of alcohol (as measured by escalated alcohol self-administration) during acute withdrawal in alcohol dependence, male Wistar rats were trained to self-administer alcohol in an operant paradigm, subjected to one month of intermittent alcohol vapor exposure to induce alcohol dependence and then allowed to self-administer alcohol during repeated acute withdrawal self-administration sessions. Subsequently, rat brains were extracted after initial or stable escalated alcohol self-administration phases of acute withdrawal and analyzed by immunoblot to detect MMP-2, -3, and -9 levels in the anterior cingulate cortex (ACC), bed nucleus of the stria terminalis, central amygdala (CeA), hippocampus, and nucleus accumbens (NAc). The results showed that MMP-9 expression in the CeA and NAc of alcohol-dependent rats was increased, however, MMP-9 expression in the ACC was decreased during negative reinforcement learning. Subsequently, the importance of plasticity mediated by MMP-9 in escalated alcohol self-administration during acute withdrawal was functionally assessed through site-specific intra-CeA MMP-9 inhibition during repeated acute withdrawal self-administration sessions. MMP-9 inhibition prevented acute withdrawal-induced escalation of alcohol self-administration in a manner that was not confounded by locomotor effects or a permanent inability to learn about the negative reinforcing effects of alcohol.


Subject(s)
Alcoholism/physiopathology , Brain/metabolism , Brain/physiopathology , Matrix Metalloproteinase 9/metabolism , Neuronal Plasticity/drug effects , Reinforcement, Psychology , Alcoholism/metabolism , Animals , Brain/drug effects , Disease Models, Animal , Learning , Male , Rats , Rats, Wistar , Self Administration
4.
Neuron ; 102(3): 564-573.e6, 2019 05 08.
Article in English | MEDLINE | ID: mdl-30878290

ABSTRACT

Negative affective states affect quality of life for patients suffering from pain. These maladaptive emotional states can lead to involuntary opioid overdose and many neuropsychiatric comorbidities. Uncovering the mechanisms responsible for pain-induced negative affect is critical in addressing these comorbid outcomes. The nucleus accumbens (NAc) shell, which integrates the aversive and rewarding valence of stimuli, exhibits plastic adaptations in the presence of pain. In discrete regions of the NAc, activation of the kappa opioid receptor (KOR) decreases the reinforcing properties of rewards and induces aversive behaviors. Using complementary techniques, we report that in vivo recruitment of NAc shell dynorphin neurons, acting through KOR, is necessary and sufficient to drive pain-induced negative affect. Taken together, our results provide evidence that pain-induced adaptations in the kappa opioid system within the NAc shell represent a functional target for therapeutic intervention that could circumvent pain-induced affective disorders. VIDEO ABSTRACT.


Subject(s)
Affect/physiology , Dynorphins/metabolism , Inflammation/metabolism , Mood Disorders/metabolism , Neurons/metabolism , Nucleus Accumbens/metabolism , Pain/metabolism , Receptors, Opioid, kappa/metabolism , Animals , Inflammation/complications , Inflammation/psychology , Mice , Mood Disorders/etiology , Mood Disorders/psychology , Neural Inhibition , Neuronal Plasticity , Nucleus Accumbens/cytology , Pain/complications , Pain/psychology , Rats
5.
Neuropharmacology ; 140: 162-173, 2018 09 15.
Article in English | MEDLINE | ID: mdl-30075159

ABSTRACT

There is an important emerging role for the endogenous opioid dynorphin (DYN) and the kappa-opioid receptor (KOR) in the treatment of alcohol dependence. Evidence suggests that the DYN/KOR system in the bed nucleus of the stria terminalis (BNST) contributes to maladaptive behavioral regulation during withdrawal in alcohol dependence. The current experiments were designed to assess dysregulation of the BNST DYN/KOR system by evaluating alcohol dependence-induced changes in DYN/KOR gene expression (Pdyn and Oprk1, respectively), and the sensitivity of alcohol self-administration, negative affective-like behavior and physiological withdrawal to intra-BNST KOR antagonism during acute withdrawal. Wistar rats trained to self-administer alcohol, or not trained, were subjected to an alcohol dependence induction procedure (14 h alcohol vapor/10 h air) or air-exposure. BNST micropunches from air- and vapor-exposed animals were analyzed using RT-qPCR to quantify dependence-induced changes in Pdyn and Oprk1 mRNA expression. In addition, vapor- and air-exposed groups received an intra-BNST infusion of a KOR antagonist or vehicle prior to measurement of alcohol self-administration. A separate cohort of vapor-exposed rats was assessed for physiological withdrawal and negative affective-like behavior signs following intra-BNST KOR antagonism. During acute withdrawal, following alcohol dependence induction, there was an upregulation in Oprk1 mRNA expression in alcohol self-administering animals, but not non-alcohol self-administering animals, that confirmed dysregulation of the KOR/DYN system within the BNST. Furthermore, intra-BNST KOR antagonism attenuated escalated alcohol self-administration and negative affective-like behavior during acute withdrawal without reliably impacting physiological symptoms of withdrawal. The results confirm KOR system dysregulation in the BNST in alcohol dependence, illustrating the therapeutic potential of targeting the KOR to treat alcohol dependence.


Subject(s)
Alcoholism/metabolism , Alcoholism/psychology , Receptors, Opioid, kappa/biosynthesis , Septal Nuclei/metabolism , Substance Withdrawal Syndrome/prevention & control , Alcoholism/complications , Animals , Conditioning, Operant/drug effects , Dynorphins/biosynthesis , Ethanol/administration & dosage , Gene Expression Regulation/drug effects , Male , Maze Learning/drug effects , Naltrexone/analogs & derivatives , Naltrexone/pharmacology , RNA, Messenger/biosynthesis , Rats , Receptors, Opioid, kappa/antagonists & inhibitors , Self Administration , Septal Nuclei/drug effects , Up-Regulation , Vocalization, Animal/drug effects
6.
Neuropsychopharmacology ; 42(12): 2470, 2017 Nov.
Article in English | MEDLINE | ID: mdl-29026256
7.
Alcohol ; 51: 43-9, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26992699

ABSTRACT

We have shown that dysregulation of the dynorphin/kappa-opioid receptor (DYN/KOR) system contributes to escalated alcohol self-administration in alcohol dependence and that KOR antagonists with extended durations of action selectively reduce escalated alcohol consumption in alcohol-dependent animals. As KOR antagonism has gained widespread attention as a potential therapeutic target to treat alcoholism and multiple neuropsychiatric disorders, we tested the effect of zyklophin (a short-acting KOR antagonist) on escalated alcohol self-administration in rats made alcohol-dependent using intermittent alcohol vapor exposure. Following dependence induction, zyklophin was infused centrally prior to alcohol self-administration sessions and locomotor activity tests during acute withdrawal. Zyklophin did not impact alcohol self-administration or locomotor activity in either exposure condition. To investigate the neurobiological basis of this atypical effect for a KOR antagonist, we utilized a κ-, µ-, and δ-opioid receptor agonist-stimulated GTPyS coupling assay to examine the opioid receptor specificity of zyklophin in the rat brain and mouse brain. In rats, zyklophin did not affect U50488-, DAMGO-, or DADLE-stimulated GTPyS coupling, whereas the prototypical KOR antagonist nor-binaltorphimine (norBNI) attenuated U50488-induced stimulation in the rat brain tissue at concentrations that did not impact µ- and δ-receptor function. To reconcile the discrepancy between the present rat data and published mouse data, comparable GTPyS assays were conducted using mouse brain tissue; zyklophin effects were consistent with KOR antagonism in mice. Moreover, at higher concentrations, zyklophin exhibited agonist properties in rat and mouse brains. These results identify species differences in zyklophin efficacy that, given the rising interest in the development of short-duration KOR antagonists, should provide valuable information for therapeutic development efforts.


Subject(s)
Dynorphins/pharmacology , Narcotic Antagonists/pharmacology , Peptide Fragments/pharmacology , Receptors, Opioid, kappa/antagonists & inhibitors , Receptors, Opioid, kappa/genetics , Animals , Dose-Response Relationship, Drug , Dynorphins/metabolism , Ethanol/administration & dosage , Male , Mice , Mice, Inbred C57BL , Narcotic Antagonists/metabolism , Peptide Fragments/metabolism , Rats , Rats, Wistar , Receptors, Opioid, kappa/metabolism , Self Administration , Species Specificity , Substance Withdrawal Syndrome/genetics , Substance Withdrawal Syndrome/psychology
8.
Neuropsychopharmacology ; 41(2): 560-7, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26105136

ABSTRACT

Chronic intermittent alcohol vapor exposure leads to increased dynorphin (DYN) A-like peptide expression and heightened kappa-opioid receptor (KOR) signaling in the central nucleus of the amygdala (CeA) and these neuroadaptive responses differentiate alcohol-dependent from non-dependent phenotypes. Important for therapeutic development efforts is understanding the nature of the stimulus that drives dependence-like phenotypes such as escalated alcohol self-administration. Accordingly, the present study examined the impact of intra-CeA KOR antagonism on escalated operant alcohol self-administration and physiological withdrawal symptoms during acute withdrawal and protracted abstinence in rats previously exposed to chronic intermittent alcohol vapor. Following operant training, rats were implanted with intra-CeA guide cannula and exposed to long-term intermittent alcohol vapor exposure that resulted in escalated alcohol self-administration and elevated physiological withdrawal signs during acute withdrawal. Animals received intra-CeA infusions of the KOR antagonist nor-binaltorphimine (nor-BNI; 0, 2, 4, or 6 µg) prior to operant alcohol self-administration sessions and physiological withdrawal assessment during acute withdrawal and protracted abstinence. The results indicated that site-specific KOR antagonism in the CeA ameliorated escalated alcohol self-administration during both acute withdrawal and protracted abstinence test sessions, whereas KOR antagonism had no effect on physiological withdrawal scores at either time point. These results dissociate escalated alcohol self-administration from physiological withdrawal symptoms in relation to KOR signaling in the CeA and help clarify the nature of the stimulus that drives escalated alcohol self-administration during acute withdrawal and protracted abstinence.


Subject(s)
Alcoholism/metabolism , Alcoholism/psychology , Central Amygdaloid Nucleus/metabolism , Receptors, Opioid, kappa/metabolism , Substance Withdrawal Syndrome/metabolism , Substance Withdrawal Syndrome/psychology , Administration, Intranasal , Alcoholism/drug therapy , Animals , Central Nervous System Depressants/administration & dosage , Conditioning, Operant/drug effects , Conditioning, Operant/physiology , Disease Models, Animal , Dose-Response Relationship, Drug , Ethanol/administration & dosage , Male , Motivation/drug effects , Motivation/physiology , Naltrexone/analogs & derivatives , Naltrexone/pharmacology , Narcotic Antagonists/pharmacology , Rats, Wistar , Receptors, Opioid, kappa/antagonists & inhibitors , Self Administration , Substance Withdrawal Syndrome/drug therapy
9.
J Neurosci ; 35(35): 12217-31, 2015 Sep 02.
Article in English | MEDLINE | ID: mdl-26338332

ABSTRACT

Pain management in opioid abusers engenders ethical and practical difficulties for clinicians, often resulting in pain mismanagement. Although chronic opioid administration may alter pain states, the presence of pain itself may alter the propensity to self-administer opioids, and previous history of drug abuse comorbid with chronic pain promotes higher rates of opioid misuse. Here, we tested the hypothesis that inflammatory pain leads to increased heroin self-administration resulting from altered mu opioid receptor (MOR) regulation of mesolimbic dopamine (DA) transmission. To this end, the complete Freund's adjuvant (CFA) model of inflammation was used to assess the neurochemical and functional changes induced by inflammatory pain on MOR-mediated mesolimbic DA transmission and on rat intravenous heroin self-administration under fixed ratio (FR) and progressive ratio (PR) schedules of reinforcement. In the presence of inflammatory pain, heroin intake under an FR schedule was increased for high, but attenuated for low, heroin doses with concomitant alterations in mesolimbic MOR function suggested by DA microdialysis. Consistent with the reduction in low dose FR heroin self-administration, inflammatory pain reduced motivation for a low dose of heroin, as measured by responding under a PR schedule of reinforcement, an effect dissociable from high heroin dose PR responding. Together, these results identify a connection between inflammatory pain and loss of MOR function in the mesolimbic dopaminergic pathway that increases intake of high doses of heroin. These findings suggest that pain-induced loss of MOR function in the mesolimbic pathway may promote opioid dose escalation and contribute to opioid abuse-associated phenotypes. SIGNIFICANCE STATEMENT: This study provides critical new insights that show that inflammatory pain alters heroin intake through a desensitization of MORs located within the VTA. These findings expand our knowledge of the interactions between inflammatory pain and opioid abuse liability, and should help to facilitate the development of novel and safer opioid-based strategies for treating chronic pain.


Subject(s)
Analgesics, Opioid/administration & dosage , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/administration & dosage , Pain , Receptors, Opioid, mu/metabolism , Ventral Tegmental Area/metabolism , Action Potentials/drug effects , Animals , Conditioning, Operant/drug effects , Disease Models, Animal , Excitatory Amino Acid Antagonists/pharmacology , Glycine Agents/pharmacology , Heroin/administration & dosage , Hyperalgesia/drug therapy , Inflammation/chemically induced , Inflammation/complications , Inhibitory Postsynaptic Potentials/drug effects , Male , Neurons/drug effects , Pain/drug therapy , Pain/pathology , Pain/psychology , Pain Threshold/drug effects , Quinoxalines/pharmacology , Rats , Rats, Sprague-Dawley , Strychnine/pharmacology , Sucrose/administration & dosage , Ventral Tegmental Area/drug effects , Ventral Tegmental Area/pathology
10.
J Neurochem ; 135(4): 659-65, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26257334

ABSTRACT

Opioid receptors can display spontaneous agonist-independent G-protein signaling (basal signaling/constitutive activity). While constitutive κ-opioid receptor (KOR) activity has been documented in vitro, it remains unknown if KORs are constitutively active in native systems. Using [(35) S] guanosine 5'-O-[gamma-thio] triphosphate coupling assay that measures receptor functional state, we identified the presence of medial prefrontal cortex KOR constitutive activity in young rats that declined with age. Furthermore, basal signaling showed an age-related decline and was insensitive to neutral opioid antagonist challenge. Collectively, the present data are first to demonstrate age-dependent alterations in the medial prefrontal cortex KOR constitutive activity in rats and changes in the constitutive activity of KORs can differentially impact KOR ligand efficacy. These data provide novel insights into the functional properties of the KOR system and warrant further consideration of KOR constitutive activity in normal and pathophysiological behavior. Opioid receptors exhibit agonist-independent constitutive activity; however, kappa-opioid receptor (KOR) constitutive activity has not been demonstrated in native systems. Our results confirm KOR constitutive activity in the medial prefrontal cortex (mPFC) that declines with age. With the ability to presynaptically inhibit multiple neurotransmitter systems in the mPFC, maturational or patho-logical alterations in constitutive activity could disrupt corticofugal glutamatergic pyramidal projection neurons mediating executive function. Regulation of KOR constitutive activity could serve as a therapeutic target to treat compromised executive function.


Subject(s)
Prefrontal Cortex/growth & development , Prefrontal Cortex/metabolism , Receptors, Opioid, kappa/metabolism , Age Factors , Analgesics, Opioid/pharmacology , Animals , Dose-Response Relationship, Drug , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology , Guanosine 5'-O-(3-Thiotriphosphate)/pharmacokinetics , Male , Naltrexone/analogs & derivatives , Naltrexone/pharmacology , Narcotic Antagonists/pharmacology , Prefrontal Cortex/diagnostic imaging , Prefrontal Cortex/drug effects , Protein Binding/drug effects , Radionuclide Imaging , Rats , Rats, Wistar , Somatostatin/analogs & derivatives , Somatostatin/pharmacology , Sulfur Isotopes/pharmacokinetics
11.
Biol Psychiatry ; 75(10): 774-82, 2014 May 15.
Article in English | MEDLINE | ID: mdl-23611261

ABSTRACT

BACKGROUND: The dynorphin (DYN)/kappa-opioid receptor (KOR) system undergoes neuroadaptations following chronic alcohol exposure that promote excessive operant self-administration and negative affective-like states; however, the exact mechanisms are unknown. The present studies tested the hypothesis that an upregulated DYN/KOR system mediates excessive alcohol self-administration that occurs during withdrawal in alcohol-dependent rats by assessing DYN A peptide expression and KOR function, in combination with site-specific pharmacologic manipulations. METHODS: Male Wistar rats were trained to self-administer alcohol using operant behavioral strategies and subjected to intermittent alcohol vapor or air exposure. Changes in self-administration were assessed by pharmacologic challenges during acute withdrawal. In addition, 22-kHz ultrasonic vocalizations were utilized to measure negative affective-like states. Immunohistochemical techniques assessed DYN A peptide expression and [(35)S]GTPγS coupling assays were performed to assess KOR function. RESULTS: Alcohol-dependent rats displayed increased alcohol self-administration, negative affective-like behavior, DYN A-like immunoreactivity, and KOR signaling in the amygdala compared with nondependent control rats. Site-specific infusions of a KOR antagonist selectively attenuated self-administration in dependent rats, whereas a mu-opioid receptor/delta-opioid receptor antagonist cocktail selectively reduced self-administration in nondependent rats. A mu-opioid receptor antagonist/partial KOR agonist attenuated self-administration in both cohorts. CONCLUSIONS: Increased DYN A and increased KOR signaling could set the stage for a one-two punch during withdrawal that drives excessive alcohol consumption in alcohol dependence. Importantly, intracentral nucleus of the amygdala pharmacologic challenges functionally confirmed a DYN/KOR system involvement in the escalated alcohol self-administration. Together, the DYN/KOR system is heavily dysregulated in alcohol dependence and contributes to the excessive alcohol consumption during withdrawal.


Subject(s)
Alcoholism/metabolism , Central Amygdaloid Nucleus/metabolism , Central Nervous System Depressants/administration & dosage , Ethanol/administration & dosage , Receptors, Opioid, kappa/metabolism , Receptors, Opioid/metabolism , Animals , Central Amygdaloid Nucleus/drug effects , Male , Rats , Rats, Wistar , Self Administration , Substance Withdrawal Syndrome/metabolism
12.
Neuropsychopharmacology ; 38(11): 2278-85, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23689673

ABSTRACT

The kappa-opioid receptor (KOR) is the primary target for the endogenous opioid peptide dynorphin (DYN), and KORs reside within brain circuitry underlying the complex integration of information related to different behavioral domains such as motivation, negative affect, and decision-making. Alterations in extended amygdala DYNs and KOR function following chronic alcohol exposure have been shown to mediate escalated alcohol self-administration during acute withdrawal. In addition to excessive alcohol consumption and increased negative affect, other symptoms of alcohol dependence include compromised impulse control. Given that DYN and KOR expressions are dysregulated within prefrontal brain circuitry associated with decision-making and impulse control in alcohol-dependent humans and rodents, and have been shown to modify multiple neurotransmitter systems associated with impulse-control disorders, we hypothesized that KOR activation could contribute to impulsive phenotypes. To test this hypothesis, separate cohorts of male Wistar rats were trained in one of the two animal models of impulsivity: delay-discounting (DD) or stop-signal reaction time (SSRT) tasks, and once stable responding was observed, received intracerebroventricular (ICV) infusions of the KOR agonist U50,488 (0-50 µg) according to a within-subject dosing regimen. The results demonstrated a dissociable effect of U50,488 on impulsive phenotypes related to intolerance to delay or response inhibition, with selective effects in the SSRT. Furthermore, the pro-impulsive effects of KOR activation were rescued by pretreatment with the KOR antagonist nor-binaltorphimine (nor-BNI). Therefore, KOR activation was shown to induce an impulsive phenotype that was nor-BNI-sensitive. Dysregulation of impulsive behavior by increased DYN/KOR activity could serve to increase vulnerability for the initiation, or perpetuate existing patterns of excessive alcohol abuse and can enhance the probability of relapse in dependent individuals. Furthermore, KOR-mediated impulsivity has implications for numerous neuropsychiatric disorders.


Subject(s)
3,4-Dichloro-N-methyl-N-(2-(1-pyrrolidinyl)-cyclohexyl)-benzeneacetamide, (trans)-Isomer/pharmacology , Analgesics, Non-Narcotic/agonists , Impulsive Behavior/psychology , Receptors, Opioid, kappa/agonists , 3,4-Dichloro-N-methyl-N-(2-(1-pyrrolidinyl)-cyclohexyl)-benzeneacetamide, (trans)-Isomer/administration & dosage , 3,4-Dichloro-N-methyl-N-(2-(1-pyrrolidinyl)-cyclohexyl)-benzeneacetamide, (trans)-Isomer/antagonists & inhibitors , Analgesics, Non-Narcotic/antagonists & inhibitors , Animals , Conditioning, Operant/drug effects , Dose-Response Relationship, Drug , Impulsive Behavior/chemically induced , Infusions, Intraventricular , Male , Naltrexone/analogs & derivatives , Naltrexone/pharmacology , Narcotic Antagonists/pharmacology , Phenotype , Rats , Receptors, Opioid, kappa/antagonists & inhibitors
13.
Neuropsychopharmacology ; 38(4): 647-54, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23212453

ABSTRACT

Negative affect promotes dysregulated alcohol consumption in non-dependent and alcohol-dependent animals, and cues associated with negative affective states induce withdrawal-like symptoms in rats. This study was designed to test the hypotheses that: (1) the kappa-opioid receptor (KOR) system mediates phenotypes related to alcohol withdrawal and withdrawal-like negative affective states and (2) cues associated with negative affective states would result in dysregulated alcohol consumption when subsequently presented alone. To accomplish these goals, intracerebroventricular infusion of the KOR antagonist nor-binaltorphimine (nor-BNI) was assessed for the ability to attenuate the increase in 22-kHz ultrasonic vocalizations (USVs) associated with alcohol withdrawal and KOR activation in adult male wistar rats. Furthermore, cues associated with a KOR agonist-induced negative affective state were assessed for the ability to dysregulate alcohol consumption and the efficacy of intracerebroventricular KOR antagonism to reduce such dysregulation was evaluated. KOR antagonism blocked the increased number of 22-kHz USVs observed during acute alcohol withdrawal and a KOR agonist (U50,488) resulted in a nor-BNI reversible increase in 22-kHz USVs (mimicking an alcohol-dependent state). Additionally, cues associated with negative affective states resulted in escalated alcohol self-administration, an effect that was nor-BNI sensitive. Taken together, this study implicates negative affective states induced by both alcohol withdrawal and conditioned stimuli as being produced, in part, by activity of the DYN/KOR system.


Subject(s)
Cues , Ethanol/administration & dosage , Receptors, Opioid, kappa/physiology , Substance Withdrawal Syndrome/physiopathology , Ultrasonics , Vocalization, Animal/physiology , Alcoholism/physiopathology , Animals , Dose-Response Relationship, Drug , Male , Rats , Rats, Wistar , Receptors, Opioid, kappa/agonists , Receptors, Opioid, kappa/antagonists & inhibitors , Self Administration
14.
Front Mol Neurosci ; 5: 95, 2012.
Article in English | MEDLINE | ID: mdl-23060746

ABSTRACT

Alcoholism is a chronic relapsing disorder characterized by continued alcohol use despite numerous adverse consequences. Alcohol has been shown to interact with numerous neurotransmitter systems to exert its pharmacological effects. The endogenous opioid system (EOS) has been strongly implicated in the positive and negative reinforcing effects of alcohol. Traditionally recognized as dysphoric/anhedonic in nature, the dynorphin/kappa-opioid receptor (DYN/KOR) system has recently received considerable attention due to evidence suggesting that an upregulated DYN/KOR system may be a critical contributor to the complex factors that result in escalated alcohol consumption once dependent. The present review will discuss alcohol-induced plasticity in the DYN/KOR system and how these neuroadaptations could contribute to excessive alcohol seeking and consumption.

16.
Psychopharmacology (Berl) ; 223(1): 75-88, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22461104

ABSTRACT

RATIONALE: Once dependent on alcohol or opioids, negative affect may accompany withdrawal. Dependent individuals are hypothesized to "self-medicate" in order to cope with withdrawal, which promotes escalated alcohol and drug use. OBJECTIVES: The current study aimed to develop a reliable animal model to assess symptoms that occur during spontaneous alcohol and opioid withdrawal. METHODS: Dependence was induced using intermittent alcohol exposure or pulsatile heroin delivery and assessed for the presence of withdrawal symptoms during acute withdrawal by measuring somatic signs, behavior in the forced swim test (FST), and air-puff-induced 22-kHz ultrasonic vocalizations (USVs). Additional animals subjected to 8 weeks of alcohol vapor exposure were evaluated for altered somatic signs, operant alcohol self-administration, and 22-kHz USV production, as well as performance in the elevated plus maze (EPM). RESULTS: During spontaneous withdrawal from pulsatile heroin or intermittent alcohol vapor, animals displayed increased somatic withdrawal signs, FST immobility, and 22-kHz USV production but did not show any behavioral change in the EPM unless the duration of alcohol exposure was extended to 4 weeks. Following 8 weeks of alcohol vapor exposure, animals displayed somatic withdrawal signs, escalated alcohol self-administration, and increased 22-kHz USVs. CONCLUSIONS: These paradigms provide consistent methods to evaluate the behavioral ramifications, and neurobiological substrates, of alcohol and opioid dependence during spontaneous withdrawal. As immobility in the FST and percent open-arm time in the EPM were dissociable, with 22-kHz USVs paralleling immobility in the FST, assessment of air-puff-induced 22-kHz USVs could provide an ethologically valid alternative to the FST.


Subject(s)
Affect/drug effects , Ethanol/administration & dosage , Heroin/administration & dosage , Substance Withdrawal Syndrome , Alcoholism/physiopathology , Animals , Conditioning, Operant , Disease Models, Animal , Heroin Dependence/physiopathology , Male , Maze Learning/drug effects , Rats , Rats, Wistar , Self Administration , Swimming , Time Factors , Vocalization, Animal
17.
Alcohol ; 46(4): 359-70, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22459870

ABSTRACT

This review represents the focus of a symposium that was presented at the "Alcoholism and Stress: A Framework for Future Treatment Strategies" conference in Volterra, Italy on May 3-6, 2011 and organized/chaired by Dr. Brendan M. Walker. The primary goal of the symposium was to evaluate and disseminate contemporary findings regarding the emerging role of kappa-opioid receptors (KORs) and their endogenous ligands dynorphins (DYNs) in the regulation of escalated alcohol consumption, negative affect and cognitive dysfunction associated with alcohol dependence, as well as DYN/KOR mediation of the effects of chronic stress on alcohol reward and seeking behaviors. Dr. Glenn Valdez described a role for KORs in the anxiogenic effects of alcohol withdrawal. Dr. Jay McLaughlin focused on the role of KORs in repeated stress-induced potentiation of alcohol reward and increased alcohol consumption. Dr. Brendan Walker presented data characterizing the effects of KOR antagonism within the extended amygdala on withdrawal-induced escalation of alcohol self-administration in dependent animals. Dr. Georgy Bakalkin concluded with data indicative of altered DYNs and KORs in the prefrontal cortex of alcohol dependent humans that could underlie diminished cognitive performance. Collectively, the data presented within this symposium identified the multifaceted contribution of KORs to the characteristics of acute and chronic alcohol-induced behavioral dysregulation and provided a foundation for the development of pharmacotherapeutic strategies to treat certain aspects of alcohol use disorders.


Subject(s)
Alcoholism/physiopathology , Amygdala/drug effects , Ethanol/pharmacology , Receptors, Opioid, kappa/physiology , Stress, Psychological/complications , Substance Withdrawal Syndrome/drug therapy , Alcoholism/drug therapy , Alcoholism/psychology , Amygdala/physiopathology , Animals , Disease Models, Animal , Dynorphins/pharmacology , Dynorphins/physiology , Dynorphins/therapeutic use , Humans , Neurotransmitter Agents/pharmacology , Neurotransmitter Agents/physiology , Neurotransmitter Agents/therapeutic use , Receptors, Opioid, kappa/agonists , Substance Withdrawal Syndrome/physiopathology
18.
Alcohol ; 46(4): 339-48, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22459874

ABSTRACT

This article represents one of five contributions focusing on the topic "Plasticity and neuroadaptive responses within the extended amygdala in response to chronic or excessive alcohol exposure" that were developed by awardees participating in the Young Investigator Award Symposium at the "Alcoholism and Stress: A Framework for Future Treatment Strategies" conference in Volterra, Italy on May 3-6, 2011 that was organized/chaired by Drs. Antonio Noronha and Fulton Crews and sponsored by the National Institute on Alcohol Abuse and Alcoholism. This review discusses the dependence-induced neuroadaptations in affective systems that provide a basis for negative reinforcement learning and presents evidence demonstrating that escalated alcohol consumption during withdrawal is a learned, plasticity-dependent process. The review concludes by identifying changes within extended amygdala dynorphin/kappa-opioid receptor systems that could serve as the foundation for the occurrence of negative reinforcement processes. While some evidence contained herein may be specific to alcohol dependence-related learning and plasticity, much of the information will be of relevance to any addictive disorder involving negative reinforcement mechanisms. Collectively, the information presented within this review provides a framework to assess the negative reinforcing effects of alcohol in a manner that distinguishes neuroadaptations produced by chronic alcohol exposure from the actual plasticity that is associated with negative reinforcement learning in dependent organisms.


Subject(s)
Alcoholism/physiopathology , Amygdala/physiopathology , Ethanol/pharmacology , Matrix Metalloproteinase Inhibitors , Neuronal Plasticity/drug effects , Reinforcement, Psychology , Substance Withdrawal Syndrome/physiopathology , Alcoholism/psychology , Amygdala/metabolism , Animals , Dynorphins/metabolism , Humans , Rats , Receptors, Opioid, kappa/metabolism , Self Administration
19.
Neurobiol Learn Mem ; 96(2): 199-206, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21530666

ABSTRACT

Repeated cycles of ethanol intoxication and withdrawal associated with dependence induce neuroadaptations in a variety of brain systems. Withdrawal-induced negative emotional states can be ameliorated by ethanol consumption; a learned process termed negative reinforcement. Accordingly, a dependence-induced phenotype is escalated ethanol self-administration. Matrix metalloproteinases (MMPs) are proteolytic enzymes which degrade the extracellular matrix to allow for synaptic reorganization and plasticity. To test the hypothesis that an intact MMP system is required for animals to learn about the negative reinforcing effects of ethanol and display escalated self-administration during acute withdrawal when ethanol-dependent, male Wistar rats were trained to self-administer ethanol and then assigned to either acute or chronic MMP inhibition treatment groups. The chronic treatment group received intracerebroventricular (ICV) infusions of the broad spectrum MMP inhibitor FN-439 or artificial cerebrospinal fluid (aCSF) via osmotic minipumps during a 1 month ethanol dependence induction period and subsequent post-dependence induction self-administration sessions that occurred during acute withdrawal. The acute treatment group only received ICV FN-439 or aCSF on the day of self-administration sessions following dependence induction during acute withdrawal. The results showed that inhibition of MMPs attenuated escalated ethanol self-administration following chronic and acute exposure conditions. Furthermore, once learning (i.e., plasticity) had occurred, MMP inhibition had no impact on escalated response patterns and animals previously subjected to MMP inhibition that did not escalate evidenced normal escalations in operant ethanol self-administration once FN-439 treatments were terminated. Thus, the present data identified that an intact MMP system is required for the escalated responding that occurs during acute withdrawal in dependent animals and implicate such escalation as a learned response.


Subject(s)
Alcohol-Related Disorders/metabolism , Ethanol/administration & dosage , Matrix Metalloproteinases/metabolism , Neuronal Plasticity/physiology , Substance Withdrawal Syndrome/metabolism , Animals , Conditioning, Operant/physiology , Male , Rats , Rats, Wistar , Self Administration
20.
Neuropharmacology ; 61(1-2): 35-42, 2011.
Article in English | MEDLINE | ID: mdl-21338616

ABSTRACT

Previously, it was shown that ethanol-dependent animals display increased sensitivity to the general opioid receptor antagonist nalmefene compared to naltrexone. It was hypothesized that the dissociable effects of the two antagonists were attributable to a κ-opioid receptor mechanism. Nucleus accumbens dynorphin is upregulated following chronic ethanol exposure and such neuroadaptations could contribute to nalmefene's increased potency in ethanol-dependent animals. To test this hypothesis, male Wistar rats were trained to self-administer ethanol using an operant conditioning procedure. Animals were then implanted with bilateral intra-accumbens shell guide cannulae and assigned to either a chronic intermittent ethanol vapor-exposure condition (to induce dependence) or an air-exposed control group. Following a one-month exposure period, nalmefene, nor-binaltorphimine (nor-BNI; selective for κ-opioid receptors) or a combination of the selective opioid receptor antagonists CTOP and naltrindole (selective for the µ- and δ-opioid receptors, respectively) were site-specifically infused into the nucleus accumbens shell prior to ethanol self-administration sessions during acute withdrawal. Nalmefene and CTOP/naltrindole dose-dependently reduced ethanol self-administration in nondependent and dependent animals, whereas nor-BNI selectively attenuated ethanol self-administration in ethanol-dependent animals without affecting the self-administration of nondependent animals. Further analysis indentified that intra-accumbens shell nalmefene was more potent in ethanol-dependent animals and that the increased potency was attributable to a κ-opioid receptor mechanism. These data support the concept that dysregulation of DYN/κ-opioid receptor systems contributes to the excessive self-administration observed in dependent animals and suggest that pharmacotherapeutics for ethanol dependence that target κ-opioid receptors, in addition to µ- and δ-opioid receptors, are preferable to those that target µ- and δ-opioid receptor mechanisms alone.


Subject(s)
Alcoholism/drug therapy , Alcoholism/metabolism , Ethanol/administration & dosage , Naltrexone/analogs & derivatives , Nucleus Accumbens/drug effects , Receptors, Opioid, kappa/physiology , Animals , Infusions, Intraventricular , Male , Naltrexone/administration & dosage , Nucleus Accumbens/physiology , Rats , Rats, Wistar , Receptors, Opioid, kappa/antagonists & inhibitors , Self Administration
SELECTION OF CITATIONS
SEARCH DETAIL
...