dynorphins and Substance-Withdrawal-Syndrome

dynorphins has been researched along with Substance-Withdrawal-Syndrome* in 56 studies

Reviews

5 review(s) available for dynorphins and Substance-Withdrawal-Syndrome

ArticleYear
Acupuncture for Detoxification in Treatment of Opioid Addiction.
    East Asian archives of psychiatry : official journal of the Hong Kong College of Psychiatrists = Dong Ya jing shen ke xue zhi : Xianggang jing shen ke yi xue yuan qi kan, 2016, Volume: 26, Issue:2

    Opioid is a popular drug of abuse and addiction. We evaluated acupuncture as a non-pharmacological treatment with a focus on managing withdrawal symptoms. Electrical stimulation at a low frequency (2 Hz) accelerates endorphin and encephalin production. High-frequency stimulation (100 Hz) up-regulates the dynorphin level that in turn suppresses withdrawal at the spinal level. The effect of 100-Hz electroacupuncture may be associated with brain-derived neurotrophic factor activation at the ventral tegmental area, down-regulation of cAMP response element-binding protein, and enhanced dynorphin synthesis in the spinal cord, periaqueductal grey, and hypothalamus. Clinical trials of acupuncture for the management of different withdrawal symptoms were reviewed. The potential of acupuncture to allay opioid-associated depression and anxiety, and its possible use as an adjuvant treatment were evident. A lack of effect was indicated for opioid craving. Most studies were hampered by inadequate reporting details and heterogeneity, thus future well-designed studies are needed to confirm the efficacy of acupuncture in opioid addiction treatment.

    Topics: Acupuncture Therapy; Adenosine Monophosphate; Animals; Brain-Derived Neurotrophic Factor; Dynorphins; Electroacupuncture; Humans; Opioid-Related Disorders; Substance Withdrawal Syndrome

2016
Targeting dynorphin/kappa opioid receptor systems to treat alcohol abuse and dependence.
    Alcohol (Fayetteville, N.Y.), 2012, Volume: 46, Issue:4

    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.

    Topics: Alcoholism; Amygdala; Animals; Disease Models, Animal; Dynorphins; Ethanol; Humans; Neurotransmitter Agents; Receptors, Opioid, kappa; Stress, Psychological; Substance Withdrawal Syndrome

2012
Conceptualizing withdrawal-induced escalation of alcohol self-administration as a learned, plasticity-dependent process.
    Alcohol (Fayetteville, N.Y.), 2012, Volume: 46, Issue:4

    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.

    Topics: Alcoholism; Amygdala; Animals; Dynorphins; Ethanol; Humans; Matrix Metalloproteinase Inhibitors; Neuronal Plasticity; Rats; Receptors, Opioid, kappa; Reinforcement, Psychology; Self Administration; Substance Withdrawal Syndrome

2012
kappa-Opioid receptor signaling and brain reward function.
    Brain research reviews, 2009, Dec-11, Volume: 62, Issue:1

    The dynorphin-like peptides have profound effects on the state of the brain reward system and human and animal behavior. The dynorphin-like peptides affect locomotor activity, food intake, sexual behavior, anxiety-like behavior, and drug intake. Stimulation of kappa-opioid receptors, the endogenous receptor for the dynorphin-like peptides, inhibits dopamine release in the striatum (nucleus accumbens and caudate putamen) and induces a negative mood state in humans and animals. The administration of drugs of abuse increases the release of dopamine in the striatum and mediates the concomitant release of dynorphin-like peptides in this brain region. The reviewed studies suggest that chronic drug intake leads to an upregulation of the brain dynorphin system in the striatum and in particular in the dorsal part of the striatum/caudate putamen. This might inhibit drug-induced dopamine release and provide protection against the neurotoxic effects of high dopamine levels. After the discontinuation of chronic drug intake these neuroadaptations remain unopposed which has been suggested to contribute to the negative emotional state associated with drug withdrawal and increased drug intake. kappa-Opioid receptor agonists have also been shown to inhibit calcium channels. Calcium channel inhibitors have antidepressant-like effects and inhibit the release of norepinephrine. This might explain that in some studies kappa-opioid receptor agonists attenuate nicotine and opioid withdrawal symptomatology. A better understanding of the role of dynorphins in the regulation of brain reward function might contribute to the development of novel treatments for mood disorders and other disorders that stem from a dysregulation of the brain reward system.

    Topics: Animals; Brain; Dynorphins; Humans; Receptors, Opioid, kappa; Reward; Substance Withdrawal Syndrome; Substance-Related Disorders

2009
Review. Neurobiological mechanisms for opponent motivational processes in addiction.
    Philosophical transactions of the Royal Society of London. Series B, Biological sciences, 2008, Oct-12, Volume: 363, Issue:1507

    The conceptualization of drug addiction as a compulsive disorder with excessive drug intake and loss of control over intake requires motivational mechanisms. Opponent process as a motivational theory for the negative reinforcement of drug dependence has long required a neurobiological explanation. Key neurochemical elements involved in reward and stress within basal forebrain structures involving the ventral striatum and extended amygdala are hypothesized to be dysregulated in addiction to convey the opponent motivational processes that drive dependence. Specific neurochemical elements in these structures include not only decreases in reward neurotransmission such as dopamine and opioid peptides in the ventral striatum, but also recruitment of brain stress systems such as corticotropin-releasing factor (CRF), noradrenaline and dynorphin in the extended amygdala. Acute withdrawal from all major drugs of abuse produces increases in reward thresholds, anxiety-like responses and extracellular levels of CRF in the central nucleus of the amygdala. CRF receptor antagonists block excessive drug intake produced by dependence. A brain stress response system is hypothesized to be activated by acute excessive drug intake, to be sensitized during repeated withdrawal, to persist into protracted abstinence and to contribute to stress-induced relapse. The combination of loss of reward function and recruitment of brain stress systems provides a powerful neurochemical basis for the long hypothesized opponent motivational processes responsible for the negative reinforcement driving addiction.

    Topics: Amygdala; Basal Ganglia; Corticotropin-Releasing Hormone; Dynorphins; Humans; Motivation; Neurobiology; Norepinephrine; Recurrence; Stress, Physiological; Substance Withdrawal Syndrome; Substance-Related Disorders

2008

Trials

4 trial(s) available for dynorphins and Substance-Withdrawal-Syndrome

ArticleYear
Effects of dynorphin A(1-13) on opiate withdrawal in humans.
    Psychopharmacology, 1998, Volume: 137, Issue:4

    The objectives of the current study were to determine 1) the effects of various doses of dynorphin A (1-13) on opiate withdrawal in humans and 2) the safety of dynorphin at these doses. Opiate dependent subjects who had been stabilized on morphine received a single IV dose of placebo, 150, 500 or 1000 microg/kg dynorphin after exhibiting spontaneous withdrawal using a randomized, double-blinded, between-subjects study design. Observer Withdrawal Scores were lower in the 150 and 1000 microg/kg groups as compared to placebo (P < 0.05) but no significant differences were observed on the observer-rated Wang or Sickness Scales. Significant decreases were also found for self-reported symptoms of nervousness, runny nose, sneezing, and painful joints in the 500 microg/kg group. Significant increases in serum prolactin levels were seen after all dynorphin doses; however, these were not dose-related. Dynorphin A (1-13) was well tolerated and safe, with no changes in physiologic parameters. We conclude that dynorphin A (1-13) has a modest effect in reducing mild opiate withdrawal in humans and is well tolerated at doses up to 1000 microg/kg.

    Topics: Adult; Biomarkers; Dose-Response Relationship, Drug; Double-Blind Method; Dynorphins; Evoked Potentials; Female; Humans; Male; Middle Aged; Morphine; Narcotics; Peptide Fragments; Prolactin; Substance Withdrawal Syndrome; Treatment Outcome

1998
Human pharmacology of the opioid neuropeptide dynorphin A(1-13).
    The Journal of pharmacology and experimental therapeutics, 1997, Volume: 281, Issue:3

    We evaluated the human pharmacology of dynorphin A(1-13) and determined whether this peptide can modulate naloxone-precipitated withdrawal effects. Such information could help determine its receptor mechanism of action and whether dynorphin is useful for treating opioid dependence. Six opioid-experienced subjects participated in a within-subject, placebo-controlled design. There were two phases, each with four test sessions. In phase 1, volunteers who were not physically dependent were administered 0, 0.1, 0.32 and 1 mg/kg dynorphin (15-min i.v. infusion) in ascending order, and subjective, observer-rated and physiological effects were monitored. Dynorphin produced brief, dose-related increases in drug effect ratings with both good and bad drug effects reported by different subjects. There were no significant changes in pupil size, respiratory rate, skin temperature, heart rate or blood pressure, These data are consistent with preclinical findings that dynorphin has a short duration of action and does not primarily exert its direct effects through mu-opioid receptors. In four separate sessions of phase 2, acute morphine pretreatment (45 mg/70 kg i.m.) was followed 15 or 18 hr later by dynorphin (0 vs. 1 mg/kg, 15-min i.v.) and then naloxone (1 or 3 vs. 10 mg/70 kg, 5-min i.v.). Under these conditions, dynorphin weakly potentiated naloxone-precipitated withdrawal. These data contrast with those of previous preclinical studies showing dependence-attenuating effects of dynorphin and fail to support its use as an antiwithdrawal agent in humans.

    Topics: Adolescent; Adult; Aged; Body Temperature; Dose-Response Relationship, Drug; Dynorphins; Humans; Male; Substance Withdrawal Syndrome

1997
Clinical effect of dynorphin on heroin addicts.
    Journal of the Medical Association of Thailand = Chotmaihet thangphaet, 1995, Volume: 78, Issue:10

    This study assessed the clinical effectiveness, physiological responses and also adverse effects of dynorphin in heroin dependent detoxication. The subjects were randomly assigned into three groups and received intravenous injections of dynorphin at the dose of 180 (6 patients) or 60 (7 patients) or 0 (8 patients) micrograms/kg three times a day for 6 days. There was no statistical difference in withdrawal syndrome between each group. However, the drug craving feeling in the dynorphin groups was reduced when compared to the placebo group. There was no report of any adverse effects of the drug or abnormal laboratory results from the subjects during the study period.

    Topics: Adult; Dynorphins; Heroin Dependence; Humans; Male; Prognosis; Substance Withdrawal Syndrome

1995
Suppression of withdrawal symptoms by dynorphin in heroin addicts.
    European journal of pharmacology, 1982, Aug-27, Volume: 82, Issue:3-4

    The effectiveness of dynorphin-(1-13) in suppressing heroin withdrawal was studied. At a dose of 60 micrograms/kg body weight, dynorphin-(1-13) was effective in suppressing withdrawal symptoms. When saline was administered a strong placebo effect was noticed that, however, did not influence the outcome of the results. Only three out of the twelve patients receiving dynorphin-(1-13) experienced some mild side-effects such as feeling warm, dizziness and precordial formication.

    Topics: Adult; Clinical Trials as Topic; Double-Blind Method; Dynorphins; Endorphins; Heroin Dependence; Humans; Male; Substance Withdrawal Syndrome

1982

Other Studies

47 other study(ies) available for dynorphins and Substance-Withdrawal-Syndrome

ArticleYear
Upregulation of dynorphin/kappa opioid receptor system in the dorsal hippocampus contributes to morphine withdrawal-induced place aversion.
    Acta pharmacologica Sinica, 2023, Volume: 44, Issue:3

    Aversive emotion of opioid withdrawal generates motivational state leading to compulsive drug seeking and taking. Kappa opioid receptor (KOR) and its endogenous ligand dynorphin have been shown to participate in the regulation of aversive emotion. In the present study, we investigated the role of dynorphin/KOR system in the aversive emotion following opioid withdrawal in acute morphine-dependent mice. We found that blockade of KORs before pairing by intracerebroventricular injection of KOR antagonist norBNI (20, 40 μg) attenuated the development of morphine withdrawal-induced conditioned place aversion (CPA) behavior. We further found that morphine withdrawal increased dynorphin A expression in the dorsal hippocampus, but not in the amygdala, prefrontal cortex, nucleus accumbens, and thalamus. Microinjection of norBNI (20 μg) into the dorsal hippocampus significantly decreased morphine withdrawal-induced CPA behavior. We further found that p38 MAPK was significantly activated in the dorsal hippocampus after morphine withdrawal, and the activation of p38 MAPK was blocked by pretreatment with norBNI. Accordingly, microinjection of p38 MAPK inhibitor SB203580 (5 μg) into the dorsal hippocampus significantly decreased morphine withdrawal-produced CPA behavior. This study demonstrates that upregulation of dynorphin/KOR system in the dorsal hippocampus plays a critical role in the formation of aversive emotion associated with morphine withdrawal, suggesting that KOR antagonists may have therapeutic value for the treatment of opioid withdrawal-induced mood-related disorders.

    Topics: Analgesics, Opioid; Animals; Dynorphins; Hippocampus; Mice; Morphine; Narcotic Antagonists; p38 Mitogen-Activated Protein Kinases; Receptors, Opioid, kappa; Substance Withdrawal Syndrome; Up-Regulation

2023
Amygdalar κ-opioid receptor-dependent upregulating glutamate transporter 1 mediates depressive-like behaviors of opioid abstinence.
    Cell reports, 2021, 11-02, Volume: 37, Issue:5

    Opiates produce a strong rewarding effect, but abstinence from opiate use emerges with severe negative emotions. Depression is one of the most frequent emotion disorders associated with opiate abstinence, which is thought to be a main cause for relapse. However, neurobiological bases of such an aversive emotion processing are poorly understood. Here, we find that morphine abstinence activates κ-opioid receptors (KORs) by increasing endogenous KOR ligand dynorphin expression in the amygdala, which in turn facilitates glutamate transporter 1 (GLT1) expression by activation of p38 mitogen-activated protein kinase (MAPK). Upregulation of GLT1 expression contributes to opiate-abstinence-elicited depressive-like behaviors through modulating amygdalar glutamatergic inputs to the nucleus accumbens (NAc). Intra-amygdala injection of GLT1 inhibitor DHK or knockdown of GLT1 expression in the amygdala significantly suppresses morphine-abstinence-induced depressive-like behaviors. Pharmacological and pharmacogenetic activation of amygdala-NAc projections prevents morphine-abstinence-induced behaviors. Overall, our study provides key molecular and circuit insights into the mechanisms of depression associated with opiate abstinence.

    Topics: Amygdala; Animals; Behavior, Animal; Depression; Disease Models, Animal; Dynorphins; Excitatory Postsynaptic Potentials; Glucose Transporter Type 1; Glutamic Acid; Male; Mice, Inbred C57BL; Mice, Knockout; Morphine; Neural Pathways; Nucleus Accumbens; p38 Mitogen-Activated Protein Kinases; Receptors, Opioid, kappa; Signal Transduction; Substance Withdrawal Syndrome

2021
Cocaine Dysregulates Dynorphin Modulation of Inhibitory Neurotransmission in the Ventral Pallidum in a Cell-Type-Specific Manner.
    The Journal of neuroscience : the official journal of the Society for Neuroscience, 2020, 02-05, Volume: 40, Issue:6

    Cocaine-driven changes in the modulation of neurotransmission by neuromodulators are poorly understood. The ventral pallidum (VP) is a key structure in the reward system, in which GABA neurotransmission is regulated by opioid neuropeptides, including dynorphin. However, it is not known whether dynorphin acts differently on different cell types in the VP and whether its effects are altered by withdrawal from cocaine. Here, we trained wild-type, D1-Cre, A2A-Cre, or vGluT2-Cre:Ai9 male and female mice in a cocaine conditioned place preference protocol followed by 2 weeks of abstinence, and then recorded GABAergic synaptic input evoked either electrically or optogenetically onto identified VP neurons before and after applying dynorphin. We found that after cocaine CPP and abstinence dynorphin attenuated inhibitory input to VP

    Topics: Animals; Basal Forebrain; Cocaine; Drug-Seeking Behavior; Dynorphins; Female; Male; Mice; Mice, Inbred C57BL; Neurons; Reward; Substance Withdrawal Syndrome; Synaptic Transmission

2020
Short-term withdrawal from repeated exposure to cocaine during adolescence modulates dynorphin mRNA levels and BDNF signaling in the rat nucleus accumbens.
    Drug and alcohol dependence, 2019, 04-01, Volume: 197

    Early-life stressful events affect the neurobiological maturation of cerebral circuitries including the endogenous opioid system and the effects elicited by adolescent cocaine exposure on this system have been poorly investigated. Here, we evaluated whether cocaine exposure during adolescence causes short- or long-term alterations in mRNAs codifying for selected elements belonging to the opioid system. Moreover, since brain-derived neurotrophic factor (BDNF) may undergo simultaneous alterations with the opioid peptide dynorphin, we also evaluated its signaling pathway as well.. Adolescent male rats were exposed to cocaine (20 mg/kg/day) from post-natal day (PND) 28 to PND42, approximately corresponding to human adolescence. After short- (PND45) or long-term (PND90) abstinence, prodynorphin-κ-opioid receptor (pDYN-KOP) and pronociceptin-nociceptin receptor (pN/OFQ-NOP) gene expression were evaluated in the nucleus accumbens (NAc) and hippocampus (Hip) together with the analysis of BDNF signaling pathways.. In the NAc of PND45 rats, pDYN mRNA levels were up-regulated, an effect paralled by increased BDNF signaling. Differently from NAc, pDYN mRNA levels were down-regulated in the Hip of PND45 rats without significant changes of BDNF pathway. At variance from PND45 rats, we did not find any significant alteration of the investigated parameters either in NAc and Hip of PND90 rats.. Our results indicate that the short-term withdrawal from adolescent cocaine exposure is characterized by a parallel pDYN mRNA and BDNF signaling increase in the NAc. Given the depressive-like state experienced during short abstinence in humans, we hypothesize that such changes may contribute to promote the risk of cocaine abuse escalation and relapse.

    Topics: Animals; Brain-Derived Neurotrophic Factor; Cocaine-Related Disorders; Dynorphins; Enkephalins; Gene Expression; Male; Nociceptin Receptor; Nucleus Accumbens; Opioid Peptides; Protein Precursors; Rats; Rats, Sprague-Dawley; Receptors, Opioid; Receptors, Opioid, kappa; RNA, Messenger; Signal Transduction; Substance Withdrawal Syndrome

2019
Maladaptive behavioral regulation in alcohol dependence: Role of kappa-opioid receptors in the bed nucleus of the stria terminalis.
    Neuropharmacology, 2018, 09-15, Volume: 140

    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.

    Topics: Alcoholism; Animals; Conditioning, Operant; Dynorphins; Ethanol; Gene Expression Regulation; Male; Maze Learning; Naltrexone; Rats; Receptors, Opioid, kappa; RNA, Messenger; Self Administration; Septal Nuclei; Substance Withdrawal Syndrome; Up-Regulation; Vocalization, Animal

2018
Species differences in the effects of the κ-opioid receptor antagonist zyklophin.
    Alcohol (Fayetteville, N.Y.), 2016, Volume: 51

    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.

    Topics: Animals; Dose-Response Relationship, Drug; Dynorphins; Ethanol; Male; Mice; Mice, Inbred C57BL; Narcotic Antagonists; Peptide Fragments; Rats; Rats, Wistar; Receptors, Opioid, kappa; Self Administration; Species Specificity; Substance Withdrawal Syndrome

2016
The kappa-opioid receptor antagonist, nor-binaltorphimine (nor-BNI), decreases morphine withdrawal and the consequent conditioned place aversion in rats.
    Behavioural brain research, 2015, Apr-15, Volume: 283

    Much data suggest that the binding of dynorphin-like peptides to kappa-opioid receptors (KORs) during the administration of and withdrawal from a variety of addictive drugs is aversive and serves to limit the reinforcing properties of those drugs and to enhance tolerance, withdrawal, and the probability of stress-induced relapse. In this study, we examined the role of KORs in mediating opioid withdrawal and its aversive consequences in rats. We found that selective blockade of KORs by i.p. administration of 20mg/kg nor-binaltorphimine (nor-BNI) 5h prior to naltrexone-precipitated withdrawal in morphine-dependent rats decreased feces excreted during a 30-min withdrawal session. More critically, this injection of nor-BNI decreased the subsequent conditioned place aversion (CPA) for the withdrawal chamber 2 days later. The subsequent finding that administration of nor-BNI 2h following withdrawal did not affect the CPA 2 days later suggested that nor-BNI reduced the CPA in the prior experiment because it reduced the aversive effects of withdrawal, not because it reduced the aversive/anxiogenic effects of the withdrawal chamber at the time of CPA testing. These data indicate that the binding of dynorphin-like peptides to KORs during opioid withdrawal serves to enhance withdrawal and its aversive consequences and suggest that selective KOR antagonists may be useful in reducing these aversive effects and consequent relapse.

    Topics: Animals; Conditioning, Psychological; Defecation; Dynorphins; Male; Morphine; Morphine Dependence; Motor Activity; Naltrexone; Narcotic Antagonists; Narcotics; Neurotransmitter Agents; Random Allocation; Rats, Long-Evans; Receptors, Opioid, kappa; Spatial Behavior; Substance Withdrawal Syndrome

2015
Protracted withdrawal from ethanol and enhanced responsiveness stress: regulation via the dynorphin/kappa opioid receptor system.
    Alcohol (Fayetteville, N.Y.), 2013, Volume: 47, Issue:5

    Although recent work suggests that the dynorphin/kappa opioid receptor (DYN/KOR) system may be a key mediator in the stress-related effects of alcohol, the regulation of long-term changes associated with protracted withdrawal from ethanol via the DYN/KOR system has yet to be explored. The objective of the present study was to determine the role of the DYN/KOR system in the regulation of anxiety-related behaviors during an extended period of abstinence from ethanol in animals with a history of ethanol dependence. Male Wistar rats (n = 94) were fed an ethanol or control liquid diet for 25-30 days. Six weeks after its removal, rats were exposed to 20 min of immobilization, and the ability of the KOR antagonist nor-binaltorphimine (nor-BNI) (0-20 mg/kg, intraperitoneal [i.p.]) to attenuate the enhanced responsiveness to stress observed in rats chronically exposed to ethanol was investigated using the elevated plus maze. In addition, the ability of U50,488 (0-10 mg/kg, i.p.) to prime anxiety-like behavior during protracted withdrawal was also examined. Rats with a history of ethanol dependence showed a significant decrease in open-arm exploration after exposure to restraint, indicating an anxiety-like state, compared to similarly treated controls, an effect that was blocked by nor-BNI. nor-BNI also selectively decreased center time and open-arm approaches in ethanol-exposed rats. The highest dose of U50,488 decreased open-arm exploration and the total number of arm entries in ethanol-exposed and control rats. Although lower doses of U50,488 did not affect open-arm exploration in either group, the 0.1 mg/kg dose selectively decreased motor activity in the ethanol-exposed rats when compared to similarly pretreated controls. These findings further support the hypothesis that behaviors associated with withdrawal from ethanol are in part regulated by the DYN/KOR system, and suggest that these effects may be long lasting in nature.

    Topics: 3,4-Dichloro-N-methyl-N-(2-(1-pyrrolidinyl)-cyclohexyl)-benzeneacetamide, (trans)-Isomer; Alcoholism; Analgesics, Non-Narcotic; Animals; Anxiety; Dose-Response Relationship, Drug; Dynorphins; Ethanol; Male; Maze Learning; Motor Activity; Naltrexone; Narcotic Antagonists; Rats; Rats, Wistar; Receptors, Opioid, kappa; Stress, Psychological; Substance Withdrawal Syndrome

2013
Different alcohol exposures induce selective alterations on the expression of dynorphin and nociceptin systems related genes in rat brain.
    Addiction biology, 2013, Volume: 18, Issue:3

    Molecular mechanisms of adaptive transformations caused by alcohol exposure on opioid dynorphin and nociceptin systems have been investigated in the rat brain. Alcohol was intragastrically administered to rats to resemble human drinking with several hours of exposure: water or alcohol (20% in water) at a dose of 1.5 g/kg three times daily for 1 or 5 days. The development of tolerance and dependence were recorded daily. Brains were dissected 30 minutes (1- and 5-day groups) or 1, 3 or 7 days after the last administration for the three other 5-day groups (groups under withdrawal). Specific alterations in opioid genes expression were ascertained. In the amygdala, an up-regulation of prodynorphin and pronociceptin was observed in the 1-day group; moreover, pronociceptin and the kappa opioid receptor mRNAs in the 5-day group and both peptide precursors in the 1-day withdrawal group were also up-regulated. In the prefrontal cortex, an increase in prodynorhin expression in the 1-day group was detected. These data indicate a relevant role of the dynorphinergic system in the negative hedonic states associated with multiple alcohol exposure. The pattern of alterations observed for the nociceptin system appears to be consistent with its role of functional antagonism towards the actions of ethanol associated with other opioid peptides. Our findings could help to the understanding of how alcohol differentially affects the opioid systems in the brain and also suggest the dynorphin and nociceptin systems as possible targets for the treatment and/or prevention of alcohol dependence.

    Topics: Alcohol Drinking; Alcoholic Intoxication; Amygdala; Animals; Brain; Central Nervous System Depressants; Dose-Response Relationship, Drug; Dynorphins; Enkephalins; Ethanol; Gene Expression; Male; Nociceptin; Opioid Peptides; Protein Precursors; Rats; Rats, Sprague-Dawley; Reflex, Righting; Substance Withdrawal Syndrome

2013
Chronic escalating cocaine exposure, abstinence/withdrawal, and chronic re-exposure: effects on striatal dopamine and opioid systems in C57BL/6J mice.
    Neuropharmacology, 2013, Volume: 67

    Cocaine addiction is a chronic relapsing disease with periods of chronic escalating self-exposure, separated by periods of abstinence/withdrawal of varying duration. Few studies compare such cycles in preclinical models. This study models an "addiction-like cycle" in mice to determine neurochemical/molecular alterations that underlie the chronic, relapsing nature of this disease. Groups of male C57BL/6J mice received acute cocaine exposure (14-day saline/14-day withdrawal/13-day saline + 1-day cocaine), chronic cocaine exposure (14 day cocaine) or chronic re-exposure (14-day cocaine/14-day withdrawal/14-day cocaine). Escalating-dose binge cocaine (15-30 mg/kg/injection × 3/day, i.p. at hourly intervals) or saline (14-day saline) was administered, modeling initial exposure. In "re-exposure" groups, after a 14-day injection-free period (modeling abstinence/withdrawal), mice that had received cocaine were re-injected with 14-day escalating-dose binge cocaine, whereas controls received saline. Microdialysis was conducted on the 14th day of exposure or re-exposure to determine striatal dopamine content. Messenger RNA levels of preprodynorphin (Pdyn), dopamine D1 (Drd1) and D2 (Drd2) in the caudate putamen were determined by real-time PCR. Basal striatal dopamine levels were lower in mice after 14-day escalating exposure or re-exposure than in those in the acute cocaine group and controls. Pdyn mRNA levels were higher in the cocaine groups than in controls. Long-term adaptation was observed across the stages of this addiction-like cycle, in that the effects of cocaine on dopamine levels were increased after re-exposure compared to exposure. Changes in striatal dopaminergic responses across chronic escalating cocaine exposure and re-exposure are a central feature of the neurobiology of relapsing addictive states.

    Topics: Animals; Cocaine; Cocaine-Related Disorders; Corpus Striatum; Dopamine; Dynorphins; Male; Mice; Mice, Inbred C57BL; Motor Activity; Protein Precursors; Receptors, Dopamine D1; Receptors, Dopamine D2; Substance Withdrawal Syndrome; Time Factors

2013
CRF2 receptor-deficiency eliminates opiate withdrawal distress without impairing stress coping.
    Molecular psychiatry, 2012, Volume: 17, Issue:12

    The opiate withdrawal syndrome is a severe stressor that powerfully triggers addictive drug intake. However, no treatment yet exists that effectively relieves opiate withdrawal distress and spares stress-coping abilities. The corticotropin-releasing factor (CRF) system mediates the stress response, but its role in opiate withdrawal distress and bodily strategies aimed to cope with is unknown. CRF-like signaling is transmitted by two receptor pathways, termed CRF(1) and CRF(2). Here, we report that CRF(2) receptor-deficient (CRF(2)(-/-)) mice lack the dysphoria-like and the anhedonia-like states of opiate withdrawal. Moreover, in CRF(2)(-/-) mice opiate withdrawal does not increase the activity of brain dynorphin, CRF and periaqueductal gray circuitry, which are major substrates of opiate withdrawal distress. Nevertheless, CRF(2) receptor-deficiency does not impair brain, neuroendocrine and autonomic stress-coping responses to opiate withdrawal. The present findings point to the CRF(2) receptor pathway as a unique target to relieve opiate withdrawal distress without impairing stress-coping abilities.

    Topics: Adaptation, Psychological; Animals; Behavior, Addictive; Brain; Corticosterone; Corticotropin-Releasing Hormone; Disease Models, Animal; Dynorphins; Mice; Mice, Inbred C57BL; Mice, Knockout; Opioid-Related Disorders; Receptors, Corticotropin-Releasing Hormone; Stress, Psychological; Substance Withdrawal Syndrome; Tyrosine 3-Monooxygenase

2012
The kappa opioid receptor antagonist JDTic attenuates alcohol seeking and withdrawal anxiety.
    Addiction biology, 2012, Volume: 17, Issue:3

    The role of kappa-opioid receptors (KOR) in the regulation of alcohol-related behaviors is not completely understood. For example, alcohol consumption has been reported to increase following treatment with KOR antagonists in rats, but was decreased in mice with genetic deletion of KOR. Recent studies have further suggested that KOR antagonists may selectively decrease alcohol self-administration in rats following a history of dependence. We assessed the effects of the KOR antagonist JDTic on alcohol self-administration, reinstatement of alcohol seeking induced by alcohol-associated cues or stress, and acute alcohol withdrawal-induced anxiety ('hangover anxiety'). JDTic dose-dependently reversed hangover anxiety when given 48 hours prior to testing, a time interval corresponding to the previously demonstrated anxiolytic efficacy of this drug. In contrast, JDTic decreased alcohol self-administration and cue-induced reinstatement of alcohol seeking when administered 2 hours prior to testing, but not at longer pre-treatment times. For comparison, we determined that the prototypical KOR antagonist nor-binaltorphimine can suppress self-administration of alcohol at 2 hours pre-treatment time, mimicking our observations with JDTic. The effects of JDTic were behaviorally specific, as it had no effect on stress-induced reinstatement of alcohol seeking, self-administration of sucrose, or locomotor activity. Further, we demonstrate that at a 2 hours pre-treatment time JDTic antagonized the antinociceptive effects of the KOR agonist U50,488H but had no effect on morphine-induced behaviors. Our results provide additional evidence for the involvement of KOR in regulation of alcohol-related behaviors and provide support for KOR antagonists, including JDTic, to be evaluated as medications for alcoholism.

    Topics: Alcoholism; Animals; Anxiety; Central Nervous System Depressants; Conditioning, Operant; Cues; Dynorphins; Ethanol; Male; Narcotic Antagonists; Piperidines; Rats; Rats, Wistar; Receptors, Opioid, kappa; Recurrence; Stress, Psychological; Substance Withdrawal Syndrome; Tetrahydroisoquinolines

2012
Stress produces aversion and potentiates cocaine reward by releasing endogenous dynorphins in the ventral striatum to locally stimulate serotonin reuptake.
    The Journal of neuroscience : the official journal of the Society for Neuroscience, 2012, Dec-05, Volume: 32, Issue:49

    Activation of the dynorphin/κ-opioid receptor (KOR) system by repeated stress exposure or agonist treatment produces place aversion, social avoidance, and reinstatement of extinguished cocaine place preference behaviors by stimulation of p38α MAPK, which subsequently causes the translocation of the serotonin transporter (SERT, SLC6A4) to the synaptic terminals of serotonergic neurons. In the present study we extend those findings by showing that stress-induced potentiation of cocaine conditioned place preference occurred by a similar mechanism. In addition, SERT knock-out mice did not show KOR-mediated aversion, and selective reexpression of SERT by lentiviral injection into the dorsal raphe restored the prodepressive effects of KOR activation. Kinetic analysis of several neurotransporters demonstrated that repeated swim stress exposure selectively increased the V(max) but not K(m) of SERT without affecting dopamine transport or the high-capacity, low-affinity transporters. Although the serotonergic neurons in the dorsal raphe project throughout the forebrain, a significant stress-induced increase in cell-surface SERT expression was only evident in the ventral striatum, and not in the dorsal striatum, hippocampus, prefrontal cortex, amygdala, or dorsal raphe. Stereotaxic microinjections of the long-lasting KOR antagonist norbinaltorphimine demonstrated that local KOR activation in the nucleus accumbens, but not dorsal raphe, mediated this stress-induced increase in ventral striatal surface SERT expression. Together, these results support the hypothesis that stress-induced activation of the dynorphin/KOR system produces a transient increase in serotonin transport locally in the ventral striatum that may underlie some of the adverse consequences of stress exposure, including the potentiation of the rewarding effects of cocaine.

    Topics: Animals; Avoidance Learning; Brain; Cocaine; Corpus Striatum; Dopamine; Dynorphins; G-Protein-Coupled Receptor Kinase 3; Male; Membrane Transport Proteins; Mice; Mice, Inbred C57BL; Mice, Knockout; Mice, Transgenic; Microinjections; Naltrexone; Narcotic Antagonists; Nicotine; Nucleus Accumbens; p38 Mitogen-Activated Protein Kinases; Raphe Nuclei; Receptors, Opioid, kappa; Reward; Serotonin; Serotonin Plasma Membrane Transport Proteins; Signal Transduction; Stress, Psychological; Substance Withdrawal Syndrome; Synaptosomes

2012
Systemic κ-opioid receptor antagonism by nor-binaltorphimine reduces dependence-induced excessive alcohol self-administration in rats.
    Addiction biology, 2011, Volume: 16, Issue:1

    Altered dynorphin opioid peptide systems contribute to increased ethanol self-administration during withdrawal following chronic alcohol exposure. We previously identified that the κ-opioid receptor antagonist nor-binaltorphimine (nor-BNI) selectively reduced ethanol self-administration in dependent animals. The purpose of this study was twofold: (1) determine whether peripherally administered nor-BNI could reduce dependence-induced ethanol self-administration and (2) confirm the selective κ-opioid effects of nor-BNI by administering it 24 hours prior to ethanol self-administration sessions occurring during acute withdrawal. Nor-BNI decreased ethanol self-administration in ethanol-dependent animals, with no effect in nondependent animals. Thus, the κ-opioid/dynorphin system is a viable pharmacotherapeutic target for the treatment of alcoholism.

    Topics: Administration, Inhalation; Alcohol Deterrents; Alcoholism; Animals; Dose-Response Relationship, Drug; Dynorphins; Ethanol; Injections, Subcutaneous; Male; Naltrexone; Narcotic Antagonists; Premedication; Rats; Rats, Wistar; Receptors, Opioid, kappa; Self Administration; Substance Withdrawal Syndrome

2011
Multiple 100 Hz electroacupuncture treatments produced cumulative effect on the suppression of morphine withdrawal syndrome: Central preprodynorphin mRNA and p-CREB implicated.
    Peptides, 2011, Volume: 32, Issue:4

    Alleviating opiate withdrawal syndrome in addicts is a critical precondition to break away from drug and further to prevent reuse. Electroacupuncture (EA) was claimed to be effective for alleviating withdrawal syndrome, but the optimal protocol remained unclear. In the present study we found that (1) 100 Hz EA administered 12-24h after the last morphine injection suppressed the withdrawal syndrome in rats, multiple sessions of EA were more effective than single session, with the after-effect lasting for at least 7 days. (2) A down-regulation of preprodynorphin (PPD) mRNA level was observed in spinal cord, PAG and hypothalamus 60 h after the last morphine injection, which could be reversed by multiple sessions, but not a single session of EA. (3) Accompanied with the decrease of PPD mRNA level, there was an up-regulation of p-CREB in the three CNS regions, which was abolished by 100 Hz EA treatment. The findings suggest that down-regulation of p-CREB and acceleration of dynorphin synthesis in spinal cord, PAG and hypothalamus may be implicated in the cumulative effect of multiple 100Hz EA treatment for opioid detoxification.

    Topics: Animals; Base Sequence; Cyclic AMP Response Element-Binding Protein; DNA Primers; Dynorphins; Electroacupuncture; Male; Morphine; Phosphorylation; Protein Precursors; Rats; Rats, Sprague-Dawley; Reverse Transcriptase Polymerase Chain Reaction; RNA, Messenger; Substance Withdrawal Syndrome

2011
Nicotine withdrawal and kappa-opioid receptors.
    Psychopharmacology, 2010, Volume: 210, Issue:2

    The synthesis and release of dynorphin are increased in the caudate/putamen (CPU) and nucleus accumbens (NAc) of nicotine-withdrawn mice, suggesting a role in the nicotine abstinence syndrome.. This study aims to investigate the consequences of enhanced dynorphinergic activity on kappa-opioid receptor (KOPr) expression, coupling, and function in CPU and NAc following chronic nicotine administration and withdrawal.. Mice were injected with nicotine-free base 2 mg/kg, or saline, sc, four times daily for 14 days and experiments performed at 24, 48, and 72 h after drug discontinuation. KOPr binding and mRNA were evaluated by [(3)H]-U69,593 autoradiography and in situ hybridization. KOPr coupling and function were investigated by agonist (U69-593)-stimulated [(35)S]GTPgammaS binding autoradiography and inhibition of adenylyl cyclase activity.. KOPr binding density and mRNA in CPU and NAc were unaltered during nicotine withdrawal; however, KPOr mRNA was increased in midbrain. U69,593-stimulated [(35)S]GTPgammaS binding was attenuated in both striatal regions, especially in NAc. In NAc shell and core, stimulated [(35)S]GTPgammaS binding was significantly decreased by 24 h and further declined over the 72 h observation period. In CPU, significant changes were observed only at 72 h. Basal adenylyl cyclase activity decreased early during nicotine withdrawal and recovered by 48 h. Stimulation with U69,593 failed to inhibit adenylyl cyclase activity at all times studied.. These observations suggest that KOPr coupling and function are impaired in NAc and CPU during nicotine withdrawal, and imply receptor desensitization. KOPr desensitization might be a mechanism to ameliorate aversive behavioral symptoms, as nicotine withdrawal evolves.

    Topics: Adenylyl Cyclase Inhibitors; Animals; Basal Ganglia; Benzeneacetamides; Caudate Nucleus; Dynorphins; GTP-Binding Proteins; Male; Mice; Nicotine; Nucleus Accumbens; Putamen; Pyrrolidines; Receptors, Opioid, kappa; RNA, Messenger; Substance Withdrawal Syndrome

2010
Suppressive effects by cysteine protease inhibitors on naloxone-precipitated withdrawal jumping in morphine-dependent mice.
    Neuropeptides, 2010, Volume: 44, Issue:3

    The effects of various protease inhibitors on naloxone-precipitated withdrawal jumping were examined in morphine-dependent mice. The doses of morphine were subcutaneously given twice daily for 2 days (day 1, 30 mg/kg; day 2, 60 mg/kg). On day 3, naloxone (8 mg/kg) was intraperitoneally administered 3h after final injection of morphine (60 mg/kg), and the number of jumping was immediately recorded for 20 min. Naloxone-precipitated withdrawal jumping was significantly suppressed by the intracerebroventricular administration of N-ethylmaleimide (0.5 nmol) and Boc-Tyr-Gly-NHO-Bz (0.4 nmol), inhibitors of cysteine proteases involved in dynorphin degradation, 5 min before each morphine treatment during the induction phase, with none given on the test day, as well as by dynorphin A (62.5 pmol) and dynorphin B (250 pmol). However, amastatin, an aminopeptidase inhibitor, phosphoramidon, an endopeptidase 24.11 inhibitor, and captopril, an angiotensin-converting enzyme inhibitor, caused no changes. The present results suggest that cysteine protease inhibitors suppress naloxone-precipitated withdrawal jumping in morphine-dependent mice, presumably through the inhibition of dynorphin degradation.

    Topics: Animals; Cysteine Proteinase Inhibitors; Dipeptides; Dynorphins; Ethylmaleimide; Injections, Intraventricular; Male; Mice; Morphine Dependence; Naloxone; Substance Withdrawal Syndrome

2010
Induction of FosB/DeltaFosB in the brain stress system-related structures during morphine dependence and withdrawal.
    Journal of neurochemistry, 2010, Volume: 114, Issue:2

    The transcription factor DeltaFosB is induced in the nucleus accumbens (NAc) by drugs of abuse. This study was designed to evaluate the possible modifications in FosB/DeltaFosB expression in both hypothalamic and extrahypothalamic brain stress system during morphine dependence and withdrawal. Rats were made dependent on morphine and, on day 8, were injected with saline or naloxone. Using immunohistochemistry and western blot, the expression of FosB/DeltaFosB, tyrosine hydroxylase (TH), corticotropin-releasing factor (CRF) and pro-dynorphin (DYN) was measured in different nuclei from the brain stress system in morphine-dependent rats and after morphine withdrawal. Additionally, we studied the expression of FosB/DeltaFosB in CRF-, TH- and DYN-positive neurons. FosB/DeltaFosB was induced after chronic morphine administration in the parvocellular part of the hypothalamic paraventricular nucleus (PVN), NAc-shell, bed nucleus of the stria terminalis, central amygdala and A(2) noradrenergic part of the nucleus tractus solitarius (NTS-A(2)). Morphine dependence and withdrawal evoked an increase in FosB/DeltaFosB-TH and FosB/DeltaFosB-CRF double labelling in NTS-A(2) and PVN, respectively, besides an increase in TH levels in NTS-A(2) and CRF expression in PVN. These data indicate that neuroadaptation to addictive substances, observed as accumulation of FosB/DeltaFosB, is not limited to the reward circuits but may also manifest in other brain regions, such as the brain stress system, which have been proposed to be directly related to addiction.

    Topics: Animals; Brain; Corticotropin-Releasing Hormone; Dynorphins; Enkephalins; Hypothalamo-Hypophyseal System; Male; Morphine; Morphine Dependence; Pituitary-Adrenal System; Protein Precursors; Proto-Oncogene Proteins c-fos; Rats; Rats, Wistar; Stress, Physiological; Substance Withdrawal Syndrome; Tyrosine 3-Monooxygenase

2010
Pharmacological evidence for a motivational role of kappa-opioid systems in ethanol dependence.
    Neuropsychopharmacology : official publication of the American College of Neuropsychopharmacology, 2008, Volume: 33, Issue:3

    The purpose of this study was to test the hypothesis that activation of the dynorphin/kappa (kappa)-opioid system has a role in the increased consumption of ethanol in dependent animals. The effects of three opioid receptor antagonists with different effects on opioid receptors, naltrexone, nalmefene, and nor-binaltorphimine (nor-BNI), were compared in their ability to decrease ethanol self-administration in nondependent and ethanol-dependent male Wistar rats. Nalmefene and naltrexone are both opioid receptor ligands with comparable molecular weights and pharmacokinetic profiles, but differing specificity for the three opioid receptor subtypes at low doses, while nor-BNI is a selective kappa-opioid receptor antagonist. Dependence was induced in half the animals by subjecting them to a 4-week intermittent vapor exposure period in which animals were exposed to ethanol vapor for 14 h per day. Subsequent to dependence induction, nalmefene, naltrexone, and nor-BNI were tested for their ability to modulate self-administration of ethanol in vapor-exposed and control rats. The results indicated that both nalmefene and naltrexone induced a significant dose-dependent decrease in the number of lever presses for ethanol in both groups of animals. However, in ethanol-dependent animals, nalmefene was significantly more effective in suppressing ethanol intake than naltrexone. Nor-BNI selectively attenuated ethanol-dependent self-administration while leaving nondependent ethanol self-administration intact. Because naltrexone is primarily selective for the mu-opioid receptor, and nalmefene is primarily selective for the mu- and kappa-opioid receptor subtypes, the fact that nalmefene demonstrates more suppression in dependent animals suggests that opioid systems distinct from the mu-regulated portion may be involved in the increased drinking seen during withdrawal in dependent animals. The results with nor-BNI confirm that kappa-opioid receptor antagonism selectively decreases dependence-induced ethanol self-administration, which supports the hypothesis that dynorphin/kappa-opioid systems are dysregulated in dependence and contribute to the increased drinking seen during acute withdrawal in dependent rats.

    Topics: Administration, Inhalation; Alcoholism; Animals; Central Nervous System Depressants; Conditioning, Operant; Data Interpretation, Statistical; Dynorphins; Ethanol; Injections, Intraventricular; Male; Motivation; Naltrexone; Narcotic Antagonists; Rats; Rats, Wistar; Receptors, Opioid, delta; Receptors, Opioid, kappa; Receptors, Opioid, mu; Self Administration; Substance Withdrawal Syndrome

2008
Dynorphin and prodynorphin mRNA changes in the striatum during nicotine withdrawal.
    Synapse (New York, N.Y.), 2008, Volume: 62, Issue:6

    Nicotine withdrawal causes somatic and negative affective symptoms that contribute to relapse and continued tobacco smoking. So far, the neuronal substrates involved are not fully understood, and an opioid role has been suggested. In this regard, the opioid dynorphin (Dyn) is of interest as it produces aversive states and has been speculated to play a role in the nicotine behavioral syndrome. These studies explore whether Dyn metabolism is altered during withdrawal following chronic administration of nicotine. Mice were administered nicotine, 2 mg/kg, s.c., four times daily for 14 days, and Dyn and prodynorphin (PD) mRNA estimated in selective brain regions at various times (30 min to 96 h) following drug discontinuation. The content of Dyn, estimated by RIA, was decreased in the striatum for a protracted time, from 30 min to over 72 h. In contrast, the mRNA for PD, evaluated by Northern blot, was elevated, appearing by 8 h and lasting over 96 h. Dyn was decreased in both ventral and dorsal striatum, and PD mRNA was differentially increased in the two striatal compartments as demonstrated by in situ hybridization. PD message was predominantly augmented in the nucleus accumbens, rostral pole, core, and shell, and the medial aspects of caudate/putamen. We interpret these data to indicate increased activity of striatal, particularly accumbal, dynorphinergic neurons during nicotine withdrawal resulting in enhanced peptide release and compensatory synthesis. Heightened dynorphinergic tone might be responsible, in part, for the emergence of the negative affective states observed during nicotine withdrawal.

    Topics: Analysis of Variance; Animals; Behavior, Animal; Brain Mapping; Corpus Striatum; Drug Administration Schedule; Dynorphins; Enkephalins; Gene Expression Regulation; Male; Mice; Nicotine; Protein Precursors; Radioimmunoassay; RNA, Messenger; Substance Withdrawal Syndrome; Time Factors

2008
Effects of cocaine place conditioning, chronic escalating-dose "binge" pattern cocaine administration and acute withdrawal on orexin/hypocretin and preprodynorphin gene expressions in lateral hypothalamus of Fischer and Sprague-Dawley rats.
    Neuroscience, 2008, Jun-02, Volume: 153, Issue:4

    Recent evidence suggests an important role for hypothalamic orexins/hypocretins in modulation of drug reward and addiction-like behaviors in rodents. Our recent study has shown that the aversive state of arousal during acute morphine withdrawal is associated with increased orexin gene expression in lateral hypothalamus (LH) of Fischer 344 (F344) inbred rats, with no change in the expression of preprodynorphin (ppDyn), a gene co-expressed with LH orexin. Therefore, we determined whether orexin and ppDyn mRNA levels in LH or medial hypothalamus (including perifornical and dorsomedial areas) of F344 or Sprague-Dawley (SD) outbred rats, are altered following: 1) cocaine (10 mg/kg, i.p.) conditioned place preference (CPP); 2) chronic (14 days) cocaine exposure using both "binge" pattern administration in steady-dose (45 mg/kg/day) and escalating-dose (45-90 mg/kg/day) regimens; and 3) acute (1 day) and chronic (14 days) withdrawal from cocaine with opioid receptor antagonist naloxone treatment (1 mg/kg). We found that orexin mRNA levels were decreased after cocaine place conditioning in the LH of SD rats. A decreased LH orexin mRNA level was also observed after chronic escalating-dose cocaine (but not CPP pattern regimen without conditioning, or steady-dose regimen) in both strains. In F344 rats only, acute withdrawal from chronic escalating-dose cocaine administration resulted in increases in both LH orexin and ppDyn mRNA levels, which were unaltered by naloxone or after chronic withdrawal. Our results suggest that (1) alteration of LH orexin gene expression is region-specific after cocaine place conditioning in SD rats and dose-dependent after chronic exposure in both strains; and (2) increased LH orexin and ppDyn gene expressions in F344 rats may contribute to negative affective states in cocaine withdrawal.

    Topics: Animals; Behavior, Animal; Cocaine; Conditioning, Operant; Dopamine Uptake Inhibitors; Dose-Response Relationship, Drug; Drug Administration Schedule; Dynorphins; Gene Expression Regulation; Hypothalamic Area, Lateral; Intracellular Signaling Peptides and Proteins; Male; Naloxone; Narcotic Antagonists; Neuropeptides; Orexins; Protein Precursors; Rats; Rats, Inbred F344; Rats, Sprague-Dawley; RNA, Messenger; Substance Withdrawal Syndrome; Time Factors

2008
Disruption of the CRF/CRF1 receptor stress system exacerbates the somatic signs of opiate withdrawal.
    Neuron, 2007, Feb-15, Volume: 53, Issue:4

    Escape from the extremely stressful opiate withdrawal syndrome may motivate opiate seeking and taking. The corticotropin-releasing factor receptor-1 (CRF1) pathway mediates behavioral and endocrine responses to stress. Here, we report that genetic inactivation (CRF1-/-) as well as pharmacological antagonism of the CRF/CRF1 receptor pathway increased and prolonged the somatic expression of opiate withdrawal. Opiate-withdrawn CRF1-/- mice also showed aberrant CRF and dynorphin expression in the paraventricular nucleus of the hypothalamus (PVN) and the striatum, indicating profound impairments in stress-responsive brain circuitry. Intake of nonstressful amounts of corticosterone effectively reduced the exaggerated somatic reactions of CRF1-/- mice to opiate withdrawal. Exogenous corticosterone also restored "wild-type-like" patterns of CRF and dynorphin gene expression in the PVN and the striatum of opiate-withdrawn CRF1-/- mice, respectively. The present findings unravel a key role for the hypothalamus-pituitary-adrenal (HPA) system and brain extra-hypothalamic CRF/CRF1 receptor circuitry in somatic, molecular, and endocrine alterations induced by opiate withdrawal.

    Topics: Analysis of Variance; Animals; Behavior, Animal; Corticosterone; Corticotropin-Releasing Hormone; Disease Models, Animal; Dynorphins; Female; Mice; Mice, Knockout; Morphine; Paraventricular Hypothalamic Nucleus; Receptors, Corticotropin-Releasing Hormone; Signal Transduction; Stress, Psychological; Substance Withdrawal Syndrome; Time Factors

2007
Absence of quasi-morphine withdrawal syndrome in adenosine A2A receptor knockout mice.
    Psychopharmacology, 2006, Volume: 185, Issue:2

    Caffeine and other methylxanthines induce behavioral activation and anxiety responses in mice via antagonist action at A2A adenosine receptors. When combined with the opioid antagonist naloxone, methylxanthines produce a characteristic quasi-morphine withdrawal syndrome (QMWS) in opiate-naive animals.. The aim of this study was to establish the role of A2A receptors in the quasi-morphine withdrawal syndrome induced by co-administration of caffeine and naloxone and in the behavioral effects of caffeine.. We have used A2A receptor knockout (A(2A)R(-/-)) mice in comparison with their wild-type and heterozygous littermates to measure locomotor activity in the open field and withdrawal symptoms induced by caffeine and naloxone. Naïve wild-type and knockout mice were also examined for enkephalin and dynorphin mRNA expression by in situ hybridization and for mu-opiate receptor by ligand binding autoradiography to check for possible opiate receptor changes induced by A2A receptor inactivation.. Caffeine increases locomotion and anxiety in wild-type animals, but it has no psychomotor effects in A(2A)R(-/-) mice. Co-administration of caffeine (20 mg/kg) and naloxone (2 mg/kg) resulted in a severe quasi-morphine withdrawal syndrome in wild-type mice that was almost completely abolished in A(2A)R(-/-) mice. Heterozygous animals exhibited a 40% reduction in withdrawal symptoms, suggesting that there is no genetic/developmental compensation for the inactivation of one of the A(2A)R alleles. A(2A)R(-/-) and wild-type mice have similar levels of striatal mu-opioid receptors, thus the effect is not due to altered opioid receptor expression.. Our results demonstrate that A2A receptors are required for the induction of quasi-morphine withdrawal syndrome by co-administration of caffeine and naloxone and implicate striatal A2A receptors and mu-opiate receptors in tonic inhibition of motor activity in the striatum.

    Topics: Adenosine A2 Receptor Antagonists; Animals; Caffeine; Central Nervous System Stimulants; Corpus Striatum; Dose-Response Relationship, Drug; Dynorphins; Enkephalins; Exploratory Behavior; Male; Mice; Mice, Knockout; Morphine; Motor Activity; Naloxone; Narcotic Antagonists; Receptor, Adenosine A2A; Receptors, Opioid, mu; RNA, Messenger; Substance Withdrawal Syndrome

2006
Reduced alcohol consumption in mice lacking preprodynorphin.
    Alcohol (Fayetteville, N.Y.), 2006, Volume: 40, Issue:2

    Many studies suggest a role for endogenous opioid peptides and their receptors in regulation of ethanol intake. It is commonly accepted that the kappa-opioid receptors and their endogenous ligands, dynorphins, produce a dysphoric state and therefore may be responsible for avoidance of alcohol. We used mutant mice lacking preprodynorphin in a variety of behavioral tests of alcohol actions. Null mutant female, but not male, mice showed significantly lower preference for alcohol and consumed lower amounts of alcohol in a two-bottle choice test as compared with wild-type littermates. In the same test, knockout mice of both sexes showed a strong reduction of preference for saccharin compared to control mice. In contrast, under conditions of limited (4 h) access (light phase of the light/dark cycle), null mutant mice did not show any differences in consumption of saccharin, but they showed significantly reduced intake of sucrose. To determine the possible cause for reduction of ethanol preference and intake, we studied other ethanol-related behaviors in mice lacking the preprodynorphin gene. There were no differences between null mutant and wild-type mice in ethanol-induced loss of righting reflex, acute ethanol withdrawal, ethanol-induced conditioned place preference, or conditioned taste aversion to ethanol. These results indicate that deletion of preprodynorphin leads to substantial reduction of alcohol intake in female mice, and suggest that this is caused by decreased orosensory reward of alcohol (sweet taste and/or palatability).

    Topics: Alcohol Drinking; Animals; Central Nervous System Depressants; Conditioning, Operant; Dynorphins; Ethanol; Mice; Mice, Inbred C57BL; Mice, Knockout; Phenotype; Postural Balance; Protein Precursors; Receptors, Opioid, kappa; Reflex; Reward; Saccharin; Substance Withdrawal Syndrome; Sucrose; Taste

2006
The corticotropin-releasing factor receptor-1 pathway mediates the negative affective states of opiate withdrawal.
    Proceedings of the National Academy of Sciences of the United States of America, 2005, Dec-20, Volume: 102, Issue:51

    The negative affective symptoms of opiate withdrawal powerfully motivate drug-seeking behavior and may trigger relapse to heroin abuse. To date, no medications exist that effectively relieve the negative affective symptoms of opiate withdrawal. The corticotropin-releasing factor (CRF) system has been hypothesized to mediate the motivational effects of drug dependence. The CRF signal is transmitted by two distinct receptors named CRF receptor-1 (CRF1) and CRF2. Here we report that genetic disruption of CRF1 receptor pathways in mice eliminates the negative affective states of opiate withdrawal. In particular, neither CRF1 receptor heterozygous (CRF1+/-) nor homozygous (CRF1-/-) null mutant mice avoided environmental cues repeatedly paired with the early phase of opiate withdrawal. These results were not due to altered associative learning processes because CRF1+/- and CRF1-/- mice displayed reliable, conditioned place aversions to environmental cues paired with the kappa-opioid receptor agonist U-50,488H. We also examined the impact of CRF1 receptor-deficiency upon opiate withdrawal-induced dynorphin activity in the nucleus accumbens, a brain molecular mechanism thought to underlie the negative affective states of drug withdrawal. Consistent with the behavioral indices, we found that, during the early phase of opiate withdrawal, neither CRF1+/- nor CRF1-/- showed increased dynorphin mRNA levels in the nucleus accumbens. This study reveals a cardinal role for CRF/CRF1 receptor pathways in the negative affective states of opiate withdrawal and suggests therapeutic strategies for the treatment of opiate addiction.

    Topics: 3,4-Dichloro-N-methyl-N-(2-(1-pyrrolidinyl)-cyclohexyl)-benzeneacetamide, (trans)-Isomer; Animals; Dynorphins; Female; Gene Expression Regulation; Mice; Mice, Knockout; Nucleus Accumbens; Opioid-Related Disorders; Receptors, Corticotropin-Releasing Hormone; Receptors, Opioid, kappa; RNA, Messenger; Signal Transduction; Substance Withdrawal Syndrome

2005
Conditioned opioid withdrawal decreases nociceptin/orphanin FQ levels in the frontal cortex and olfactory tubercle.
    Neuropsychopharmacology : official publication of the American College of Neuropsychopharmacology, 2002, Volume: 27, Issue:2

    Clinical evidence suggests that individuals experiencing drug withdrawal can become conditioned to environmental situations, whereby previously neutral stimuli can produce symptoms of withdrawal. It is believed that this "conditioned withdrawal" can have motivational significance, but the neurobiological basis for conditioned withdrawal is unknown. The goal of this study was to determine adaptations in endogenous opioid systems that may be responsible for expression of conditioned withdrawal. Opioid-dependent rats trained to lever press for food were exposed to tone and scent cues in the presence of naloxone or saline. Naloxone but not saline predictably suppressed responding for food. One month later and in a post-dependent state, all rats again were exposed to the cues but not naloxone. The conditioned cues alone suppressed responding for food in the rats previously paired with naloxone, but no suppression was seen in rats previously paired with saline. Radioimmunoassay (RIA) analysis for nociceptin/orphanin FQ (nociceptin), met-enkephalin-Arg-Phe (MEAP), and dynorphin A (dyn A) was performed from dissections of various brain regions of the rats undergoing conditioned withdrawal. Significant reductions in nociceptin peptide levels were seen in the frontal cortex and olfactory tubercle of these rats. Unconditioned opioid withdrawal and unconditioned footshock stress produced different patterns of opioid peptide regulation in separate groups of rats. These results shed light on adaptations of endogenous opioid systems to conditioned cues, stress, and withdrawal, all factors that play a role in motivating drug intake.

    Topics: Animals; Behavior, Animal; Conditioning, Psychological; Cues; Down-Regulation; Dynorphins; Electric Stimulation; Enkephalin, Methionine; Male; Naloxone; Narcotic Antagonists; Nociceptin; Olfactory Pathways; Opioid Peptides; Opioid-Related Disorders; Prefrontal Cortex; Rats; Rats, Wistar; Reinforcement, Psychology; Stress, Physiological; Substance Withdrawal Syndrome

2002
Reduction of opioid dependence by the CB(1) antagonist SR141716A in mice: evaluation of the interest in pharmacotherapy of opioid addiction.
    British journal of pharmacology, 2001, Volume: 132, Issue:8

    Several compounds, mainly opioid agonists such as methadone, are currently used for long term medication of heroin addicts. Nevertheless, these maintenance treatments have the disadvantage to induce a dependence to another opiate. As interactions between opioid and cannabinoid systems have been demonstrated, the ability of the CB(1) antagonist, SR141716A to reduce morphine-induced addiction was investigated. The effects of SR141716A on the rewarding responses of morphine were evaluated in the place conditioning paradigm. No significant conditioned preference or aversion were observed after repeated treatment with the CB(1) antagonist alone. However, SR141716A was able to antagonize the acquisition of morphine-induced conditioned place preference. SR141716A was co-administered with morphine for 5 days, and the withdrawal syndrome was precipitated by naloxone administration. A reduction in the incidence of two main signs of abstinence: wet dog shakes and jumping was observed while the other were not significantly modified. In contrast, an acute injection of the CB(1) antagonist just before naloxone administration was unable to modify the incidence of the behavioural manifestations of the withdrawal, suggesting that only chronic blockade of CB(1) receptors is able to reduce morphine-induced physical dependence. Several biochemical mechanisms could explain the reduction of opioid dependence by CB(1) antagonists. Whatever the hypotheses, this study supports the reported interaction between the endogenous cannabinoid and opioid systems, and suggests that SR 141716A warrants further investigations for a possible use in opioid addiction.

    Topics: Animals; Brain Chemistry; Cannabinoids; Conditioning, Operant; Dynorphins; Male; Mice; Naloxone; Narcotic Antagonists; Opioid-Related Disorders; Piperidines; Pyrazoles; Radioimmunoassay; Receptors, Cannabinoid; Receptors, Drug; Receptors, Opioid, kappa; Rimonabant; Substance Withdrawal Syndrome; Synapses

2001
Dynorphin A (2-17) attenuates the unconditioned but not the conditioned effects of opiate withdrawal in the rat.
    Psychopharmacology, 2000, Volume: 151, Issue:4

    An unbiased place preference conditioning procedure was used to examine the influence of the non-opioid peptide, dynorphin A 2-17 (DYN 2-17), upon the conditioned and unconditioned effects of opiate withdrawal in the rat.. Rats were implanted SC with two pellets containing 75 mg morphine or placebo. Single-trial place conditioning sessions with saline and the opioid receptor antagonist naloxone (0.1-1.0 mg/kg; SC) commenced 4 days later. Ten minutes before SC injections, animals received an IV infusion of saline or DYN 2-17 (0.1-5.0 mg/kg). Additional groups of placebo- and morphine-pelleted animals were conditioned with saline and DYN 2-17. During each 30-min conditioning session, somatic signs of withdrawal were quantified. Tests of place conditioning were conducted in pelleted animals 24 h later.. Naloxone produced wet-dog shakes, body weight loss, ptosis and diarrhea in morphine-pelleted animals. Morphine-pelleted animals also exhibited significant aversions for an environment previously associated with the administration of naloxone. These effects were not observed in placebo-pelleted animals. DYN 2-17 pretreatment resulted in a dose-related attenuation of somatic withdrawal signs. However, conditioned place aversions were still observed in morphine-pelleted animals that had received DYN 2-17 in combination with naloxone. Furthermore, the magnitude of this effect did not differ from control animals.. These data demonstrate that the administration of DYN 2-17 attenuates the somatic, but not the conditioned aversive effects of antagonist-precipitated withdrawal from morphine in the rat. Differential effects of this peptide in modulating the conditioned and unconditioned effects of opiate withdrawal are suggested.

    Topics: Animals; Conditioning, Psychological; Dynorphins; Male; Naloxone; Opioid-Related Disorders; Peptide Fragments; Rats; Rats, Sprague-Dawley; Receptors, N-Methyl-D-Aspartate; Substance Withdrawal Syndrome

2000
A heroin-, but not a cocaine-expecting, self-administration state preferentially alters endogenous brain peptides.
    European journal of pharmacology, 1999, Jan-22, Volume: 365, Issue:2-3

    The purpose of the current study was to assess neuropeptidergic alterations during a phase of the drug addiction cycle associated with drug craving as compared to a time period when the drug had been recently self-administered. Male Wistar rats were allowed to self-administer cocaine, heroin or saline for 6 h for 5 consecutive days. Immediately following the last self-administration session ('acute drug on board' state), and just before the next scheduled session ('drug expecting' state), the animals were decapitated and the levels of dynorphin A and B, [Met5]- and [Leu5]-enkephalin and substance P were measured in different brain areas. During the 'acute drug on board' state, peptide levels in animals that self-administered heroin or cocaine were not significantly changed. In contrast, during the 'drug expecting' state, heroin-treated animals had increased levels of dynorphin A, dynorphin B and [Met5]-enkephalin in the caudal striatum as compared to the cocaine- and saline-treated animals, and the level of [Leu5]-enkephalin was increased as compared to the cocaine-treated group. In the septum, an increase of [Met5]-enkephalin and substance P was observed in the animals expecting heroin as compared to the saline- and/or cocaine-treated animals. In the caudal striatum, substance P levels were elevated in the heroin- and cocaine-expecting animals. In conclusion, heroin, as compared to cocaine, appears to have a more pronounced effect on dynorphin, enkephalin and substance P levels in the caudal striatum and septum, especially during periods when self-administration of the drug is expected.

    Topics: Animals; Behavior, Animal; Brain; Cocaine; Dynorphins; Enkephalins; Heroin; Injections, Intravenous; Male; Neuropeptides; Radioimmunoassay; Rats; Rats, Wistar; Self Administration; Substance P; Substance Withdrawal Syndrome; Substance-Related Disorders; Time Factors

1999
Suppression of morphine withdrawal by electroacupuncture in rats: dynorphin and kappa-opioid receptor implicated.
    Brain research, 1999, Dec-18, Volume: 851, Issue:1-2

    Our previous work has demonstrated that 100-Hz electroacupuncture (EA) or 100-Hz transcutaneous electrical nerve stimulation (TENS) was very effective in ameliorating the morphine withdrawal syndrome in rats and humans. The mechanism was obscure. (1) Rats were made dependent on morphine by repeated morphine injections (5-140 mg/kg, s.c., twice a day) for eight days. They were then given 100-Hz EA for 30 min 24 h after the last injection of morphine. A marked increase in tail flick latency (TFL) was observed. This effect of 100-Hz EA could be blocked by naloxone (NX) at 20 mg/kg, but not at 1 mg/kg, suggesting that 100-Hz EA-induced analgesia observed in morphine-dependent rats is mediated by kappa-opioid receptors. (2) A significant decrease of the concentration of dynorphin A (1-17) immunoreactivity (-ir) was observed in the spinal perfusate in morphine-dependent rats, that could be brought back to normal level by 100-Hz EA. (3) 100-Hz EA was very effective in suppressing NX-precipitated morphine withdrawal syndrome. This effect of EA could be prevented by intrathecal administration of nor-BNI (2.5 micrograms/20 microliters), a kappa-opioid receptor antagonist, or dynorphin A (1-13) antibodies (25 micrograms/20 microliters) administered 10 min prior to EA. In conclusion, while the steady-state spinal dynorphin release is low in morphine-dependent rats, it can be activated by 100-Hz EA stimulation, which may be responsible for eliciting an analgesic effect and ameliorating morphine withdrawal syndrome, most probably via interacting with kappa-opioid receptor at spinal level.

    Topics: Animals; Body Weight; Dynorphins; Electroacupuncture; Male; Morphine; Naloxone; Narcotic Antagonists; Narcotics; Pain Measurement; Rats; Rats, Wistar; Receptors, Opioid, kappa; Substance Withdrawal Syndrome

1999
Levels of immunoreactive dynorphin A1-13 during development of morphine dependence in rats.
    Zhongguo yao li xue bao = Acta pharmacologica Sinica, 1998, Volume: 19, Issue:6

    To study the relationship between the levels of immunoreactive dynorphin A1-13 (ir-dynorphin A1-13) and the degree of morphine dependence.. The levels of ir-dynorphin A1-13 in discrete brain regions, spinal cord, and plasma in rats were determined by radioimmunoassay, and the degree of morphine dependence was assessed by scoring withdrawal signs on d 3, d 6, and d 12.. Morphine injection s.c. decreased the levels of ir-dynorphin A1-13 in spinal cord, pituitary, and plasma. The levels of ir-dynorphin A1-13 in hippocampus and hypothalamus were increased. No changes in cortex, midbrain, cerebellum, pons, and medulla were observed. With continuous injection of morphine, withdrawal signs scores were increased on d 6, but there was no difference between the scores of d 6 and d 12.. The changes of the levels of endogenous ir-dynorphin A1-13 in pituitary, spinal cord, and plasma were compatible with the degree of morphine dependence.

    Topics: Animals; Dynorphins; Male; Morphine Dependence; Peptide Fragments; Pituitary Gland; Rats; Rats, Sprague-Dawley; Spinal Cord; Substance Withdrawal Syndrome

1998
A comparison between microwave irradiation and decapitation: basal levels of dynorphin and enkephalin and the effect of chronic morphine treatment on dynorphin peptides.
    Neuropeptides, 1997, Volume: 31, Issue:4

    Opioid peptides were analysed in tissue extracts of various brain structures and the pituitary gland from rats sacrificed by microwave irradiation, and compared with peptide levels in tissue extracts from decapitated rats. Dynorphin A, dynorphin B and Leu-enkephalinArg6, derived from prodynorphin, and Met-enkephalinArg6Phe7 from proenkephalin, were measured. Basal immunoreactive levels of dynorphin A and B were consistently higher in extracts from microwave-irradiated rats, whereas in these extracts immunoreactive levels of Leu-enkephalinArg6, an endogenous metabolite of dynorphin peptides, were either lower than, the same as or higher than in decapitated rats. Immunoreactive levels of Met-enkephalinArg6Phe7 were higher in microwave-irradiated rats. Effects of morphine treatment on prodynorphin peptide levels were evaluated and compared with previous findings in decapitated rats. Dynorphin immunoreactive levels were higher in the nucleus accumbens and striatum of morphine-tolerant rats than in corresponding areas in saline-treated rats. These results indicate tissue-specific metabolism of prodynorphin peptides and show that metabolism of opioid peptides occurs during the dissection procedure after decapitation of the rat even though precautions are taken to minimize degradation.

    Topics: Animals; Brain Chemistry; Decerebrate State; Dynorphins; Endorphins; Enkephalins; Injections, Subcutaneous; Male; Microwaves; Morphine; Opioid Peptides; Rats; Rats, Sprague-Dawley; Substance Withdrawal Syndrome

1997
Brain dynorphin and enkephalin systems in Fischer and Lewis rats: effects of morphine tolerance and withdrawal.
    Brain research, 1995, Jun-12, Volume: 683, Issue:1

    Lewis rats are more likely to self-administer various drugs of abuse than Fischer rats. Here these two strains of rats were compared with regard to basal brain opioid peptide levels and the response to chronic morphine treatment and to naloxone-precipitated withdrawal. Lewis rats had lower basal dynorphin peptides in the substantia nigra, striatum (not Leu-enkephalinArg6) and VTA (not dynorphin B) and the pituitary gland. Leu-enkephalinArg6 levels were also lower in these structures (with the exception of striatum which had higher levels) and in the nucleus accumbens. There were also strain differences in the response to chronic morphine treatment; in the nucleus accumbens, morphine treatment increased dynorphin A levels in Fischer rats only, in the ventral tegmental area effects were opposite with increased dynorphin levels in Fischer and decreased levels in Lewis rats, in the hippocampus dynorphin levels were markedly reduced in Lewis rats only. In Fischer rats, chronic morphine strongly affected peptide levels in the substantia nigra and striatum, whereas Lewis rats responded less in these areas. Leu-enkephalin, which derives from both prodynorphin and proenkephalin, and Met-enkephalin, which derives from proenkephalin, were affected by chronic morphine mainly in Fischer rats, increasing levels in most of the brain areas examined. The results in this study show (1) strain differences in basal levels of prodynorphin-derived opioid peptides, (2) the prodynorphin system to be differently influenced by morphine in Lewis rats than in Fischer rats and 3) the proenkephalin system to be influenced by chronic morphine in brain areas related to reward processes only in Fischer rats.

    Topics: Amino Acid Sequence; Animals; Body Weight; Brain Chemistry; Drug Tolerance; Dynorphins; Enkephalins; Female; Male; Molecular Sequence Data; Morphine; Naloxone; Narcotic Antagonists; Rats; Rats, Inbred F344; Rats, Inbred Lew; Species Specificity; Substance Withdrawal Syndrome

1995
The effects of morphine treatment and morphine withdrawal on the dynorphin and enkephalin systems in Sprague-Dawley rats.
    Psychopharmacology, 1995, Volume: 118, Issue:4

    The effect of morphine tolerance and withdrawal on prodynorphin peptides was studied in relevant brain areas and in the pituitary gland of male Sprague-Dawley rats, and compared with effects on the proenkephalin-derived peptide Met-enkephalin. After 8 days of morphine injections (twice daily), dynorphin A and B levels increased in the nucleus accumbens and dynorphin A levels increased also in the striatum. Morphine treatment increased striatal Met-enkephalin. Leu-enkephalinArg6 levels were reduced in the ventral tegmental area (VTA). Morphine-treated rats had very low Leu-enkephalinArg6 levels in the hippocampus as compared to saline control rats. Comparison of the relative amounts of dynorphin peptides and the shorter prodynorphin-derived peptides, Leu-enkephalinArg6 and Leu-enkephalin, revealed a relative increase in dynorphin peptides versus shorter fragments in the nucleus accumbens, VTA and hippocampus. Morphine-tolerant rats had lower levels of dynorphin A in both lobes of the pituitary gland, whereas hypothalamic dynorphin levels were unaffected by morphine. Leu-enkephalinArg6 levels were reduced in the hypothalamus, but not changed in the pituitary gland. Naloxone-precipitated withdrawal accentuated the increase in dynorphin A and B levels in the accumbens and dynorphin A levels in the striatum, while inducing an increase in enkephalin levels in the accumbens and Met-enkephalin in the VTA. In the hippocampus, Leu-enkephalinArg6 levels remained low in the withdrawal state. The low dynorphin levels in the anterior part of the pituitary gland were reversed by naloxone, whereas the low dynorphin A levels in the neurointermediate lobe were 0ven lower in the withdrawal state.(ABSTRACT TRUNCATED AT 250 WORDS)

    Topics: Analysis of Variance; Animals; Drug Tolerance; Dynorphins; Endorphins; Enkephalin, Leucine; Male; Morphine; Naloxone; Peptides; Rats; Rats, Sprague-Dawley; Substance Withdrawal Syndrome

1995
Dynorphin A modulates acute and chronic opioid effects.
    The Journal of pharmacology and experimental therapeutics, 1995, Volume: 273, Issue:1

    A single dose of dynorphin A-(1-13) [dyn A(1-13)] is effective in suppressing the expression of opioid withdrawal and tolerance in morphine-dependent mice. In addition, this modulatory activity is retained by the corresponding non-opioid [des-Tyr1]-dynorphin A peptide [dynA(2-17)]. We have further investigated the non-opioid nature of this activity by comparing the efficacies of dyn A(1-13) and (2-17) under different experimental protocols with a variety of dosing regimens. The effect of dyn A(1-13) on withdrawal and tolerance expression was dose-dependent and could be enhanced by repeated dosing. Thus, the ED50 of naloxone to precipitate withdrawal jumping was increased 1.8-fold when morphine-dependent mice were treated with 4.2 mumol/kg dyn A(1-13) on the fourth day after pellet implantation and 2.4-fold on the sixth day with continued daily dyn A(1-13) treatment. The maximal effect was observed on day 6 when the ED50 of mice treated with 8.4 mumol/kg of dyn A(1-13) was increased nearly 6-fold over that of saline controls. Dyn A(2-17) proved to be nearly as effective as dyn A(1-13).

    Topics: Animals; Dose-Response Relationship, Drug; Dynorphins; Male; Mice; Mice, Inbred ICR; Morphine; Naloxone; Narcotics; Opioid-Related Disorders; Peptide Fragments; Substance Withdrawal Syndrome

1995
[Des-Tyr1]dynorphin A-(2-17) has naloxone-insensitive antinociceptive effect in the writhing assay.
    The Journal of pharmacology and experimental therapeutics, 1995, Volume: 273, Issue:2

    The dynorphin family of peptides stands out among the opioids in that its members are not antinociceptive after central administration in the common antinociceptive assays. In addition, reports of spinal antinociception have been conflicting. We have tested the antinociceptive activity of i.v. dynorphin A-(1-13) in the writhing assay and have found it to be very potent, with an ED50 of 1.0 (0.99-1.02) mumol/kg. Remarkably, [des-tyr1]dyn A-(2-17) was equally active with an ED50 of 1.1 (0.99-1.20). This activity was also retained by several smaller, non-opioid dynorphin A fragments and was not affected by the presence of either 50 mumol/kg naloxone or 20 mumol/kg Nor-BNI. Further, ED50 values were not different in morphine-dependent mice. The peak effect of dyn A-(1-13) and A-(2-17) was observed 5 min after administration and the effect of dyn A-(1-13) or dyn A-(2-17) was still measurable 1 hr after i.v. administration with a 5- to 6-fold increase in ED50 at this time. The ED50 values after i.c.v. and i.t. administration of dyn A-(1-13) were similar to those reported previously. Dyn A(2-17) was also effective by these routes with ED50 values not significantly different from those of dyn A-(1-13). Both dyn A-(1-13) and A-(2-17) were also active when injected i.p., whereas ED50 values increased substantially after s.c. administration.(ABSTRACT TRUNCATED AT 250 WORDS)

    Topics: Analgesics, Opioid; Animals; Behavior, Animal; Dynorphins; Kinetics; Ligands; Male; Mice; Mice, Inbred ICR; Naloxone; Narcotics; Pain Measurement; Peptide Fragments; Substance Withdrawal Syndrome

1995
Suppression by dynorphin A and [des-Tyr1]dynorphin A peptides of the expression of opiate withdrawal and tolerance in morphine-dependent mice.
    The Journal of pharmacology and experimental therapeutics, 1993, Volume: 266, Issue:1

    Previously, we demonstrated that the expression of opiate withdrawal and antinociceptive tolerance can be suppressed by dynorphin (dyn) A-(1-13) in morphine-dependent mice. In this study, it was shown that the normal, endogenous dyn, dyn A-(1-17) also possessed this suppressive property. While using the nonopioid dyn analog, [des-Tyr1]dyn A [dyn A-(2-17)] as a negative control, we discovered unexpectedly that this peptide fragment also suppressed naloxone-induced withdrawal and the expression of morphine tolerance in morphine-dependent mice. Thus, an extensive structure activity relationship was studied using 11 peptide fragments. It was determined that the amino acid sequence of dyn A was required for the suppressive activity because dyn B and alpha-neoendorphin both failed to suppress naloxone-precipitated withdrawal jumping. Of the [des-Tyr1]dyn fragments, the minimal amino acid sequence required to suppress naloxone-induced withdrawal was determined to be dyn A-(2-8), containing the sequence G-G-F-L-R-R-I.

    Topics: Amino Acid Sequence; Animals; Drug Tolerance; Dynorphins; Male; Mice; Molecular Sequence Data; Morphine; Morphine Dependence; Naloxone; Nociceptors; Peptide Fragments; Structure-Activity Relationship; Substance Withdrawal Syndrome

1993
Chronic administration of morphine decreases level of dynorphin A in the rat nucleus accumbens.
    Neuropharmacology, 1993, Volume: 32, Issue:7

    The effect of chronically administered morphine on the levels of dynorphin A in distinct regions of the brain (including medial frontal cortex, olfactory tubercule, nucleus accumbens, dorsal and medial striatum), was determined in male Sprague-Dawley rats. The drug was delivered through a subcutaneously implanted Azlet miniosmotic pump over a period of 5 days. The concentration of peptide was probed by radioimmunoassay, following pre-separation of tissue extracts by reversed phase separation on a SepPak C-18 cartridge. The result showed that the level of dynorphin A remained unaltered in all regions studied immediately before (tolerance) and 20 hr after (withdrawal) the pump was removed. A significant decrease in the level of dynorphin was found in the n. accumbens 48 hr (abstinence) after removal of the pump. It is suggested that previously observed changes in the reward system during abstinence may be connected with dynorphinergic neurones in the limbic system.

    Topics: Animals; Cerebral Cortex; Corpus Striatum; Dopamine; Dynorphins; Infusion Pumps, Implantable; Limbic System; Male; Morphine; Motor Activity; Nociceptors; Nucleus Accumbens; Radioimmunoassay; Rats; Rats, Sprague-Dawley; Substance Withdrawal Syndrome

1993
The effect of morphine tolerance dependence and abstinence on immunoreactive dynorphin (1-13) levels in discrete brain regions, spinal cord, pituitary gland and peripheral tissues of the rat.
    Brain research, 1992, Jul-03, Volume: 584, Issue:1-2

    The effect of morphine tolerance dependence and protracted abstinence on the levels of dynorphin (1-13) in discrete brain regions, spinal cord, pituitary gland and peripheral tissues was determined in male Sprague-Dawley rats. Of all the tissues examined, the highest level of dynorphin (1-13) was found to be in the pituitary gland. Among the brain regions and spinal cord examined, the levels of dynorphin (1-13) in descending order were: hypothalamus, spinal cord, midbrain, pons and medulla, hippocampus, cortex, amygdala and striatum. The descending order for the levels of dynorphin (1-13) in peripheral tissues was: adrenals, heart and kidneys. In morphine tolerant rats, the levels of dynorphin (1-13) increased in amygdala but were decreased in pons and medulla. In morphine abstinent rats, the levels of dynorphin (1-13) were increased in amygdala, hypothalamus and hippocampus. The levels of dynorphin (1-13) were increased in pituitary but decreased in spinal cord and remained so even during protracted abstinence. The levels of dynorphin (1-13) in the peripheral tissues of morphine tolerant rats were unaffected. However, in the heart and kidneys of morphine abstinent rats, the levels of dynorphin (1-13) were increased significantly. It is concluded that both morphine tolerance and abstinence modify the levels of dynorphin (1-13) in pituitary, central and peripheral tissues. Morphine abstinence differed from non-abstinence process in that there were additional changes (increases) in the levels of dynorphin (1-13) in brain regions (hypothalamus and hippocampus) and peripheral tissues (heart and kidneys) and may contribute to the symptoms of the morphine abstinence syndrome.(ABSTRACT TRUNCATED AT 250 WORDS)

    Topics: Adrenal Glands; Analgesics; Animals; Brain Chemistry; Drug Tolerance; Dynorphins; Heart; Kidney; Male; Morphine; Morphine Dependence; Myocardium; Peptide Fragments; Pituitary Gland; Rats; Rats, Inbred Strains; Spinal Cord; Substance Withdrawal Syndrome

1992
Suppression by dynorphin A-(1-13) of the expression of opiate withdrawal and tolerance in mice.
    European journal of pharmacology, 1992, Oct-20, Volume: 221, Issue:2-3

    Dynorphin A-(1-13) has been shown to suppress the expression of opiate withdrawal and tolerance dose dependently in morphine-dependent mice when administered i.v. The ED50 of naloxone to precipitate withdrawal jumping was increased by 1.5- and 7-fold when morphine-dependent mice were pretreated with 2.5 and 5.0 mumol/kg of dynorphin A-(1-13), respectively. When dynorphin A-(1-13) (5.0 mumol/kg, i.v.) was administered after the precipitation of withdrawal with naloxone, the ED50 of naloxone was still increased by over 2-fold. Also, the expression of tolerance which was estimated by noting the antinociceptive ED50 of morphine, was inhibited by over 70% with a dynorphin A-(1-13) dose of 2.5 mumol/kg and completely suppressed by pretreatment with 5.0 mumol/kg of dynorphin A-(1-13) i.v. The mechanism by which dynorphin A-(1-13) produces these effects when given i.v. remains to be elucidated.

    Topics: Animals; Drug Tolerance; Dynorphins; Male; Mice; Morphine; Naloxone; Opioid-Related Disorders; Peptide Fragments; Substance Withdrawal Syndrome

1992
Systemic administration of dynorphin A(1-13) markedly inhibits different behavioural responses induced by cocaine in the mouse.
    Neuropharmacology, 1992, Volume: 31, Issue:9

    The effects of systemic administration (i.p.) of dynorphin A(1-13) on the cocaine-induced behavioural alterations in the mouse were determined by using multi-dimensional behavioural analyses, based upon a capacitance system. A 1.0 mg/kg dose of cocaine did not influence behaviour, while increasing doses to 3-30 mg/kg produced a significant increment in the frequency of behaviour, such as linear locomotion, circling, rearing and grooming. Although a 1.0 mg/kg dose of dynorphin A(1-13) alone produced a significant decrease in grooming behaviour, larger doses (3.0 and 10.0 mg/kg) of the peptide failed to affect different behaviour. The cocaine (3.0 mg/kg)-induced increases in linear locomotion, circling and rearing behaviour were significantly inhibited by dynorphin A(1-13) (10.0 mg/kg). The inhibitory effects of dynorphin A(1-13) (10.0 mg/kg) were antagonized by the opioid antagonist Mr 2266 (5.6 mg/kg). It is thus possible that the systemic administration of dynorphin A(1-13) inhibits different behavioural responses induced by cocaine through the blood-brain barrier, although the instability of amino acid bonds or the relatively large molecular weight of dynorphin A(1-13), may result in the failure to demonstrate opioid activity by the peptide after systemic administration.

    Topics: Analgesics; Animals; Behavior, Animal; Benzomorphans; Cocaine; Dynorphins; Injections, Intraperitoneal; Male; Mice; Mice, Inbred Strains; Motor Activity; Narcotic Antagonists; Peptide Fragments; Substance Withdrawal Syndrome

1992
Previous chronic blockade of NMDA receptors intensifies morphine dependence in rats.
    Pharmacology, biochemistry, and behavior, 1991, Volume: 39, Issue:3

    Chronic exposure of receptors to antagonists generally results in upregulation and/or supersensitivity. On the other hand, the noncompetitive NMDA receptor antagonists ketamine (K) and dextromethorphan (DM) suppress opiate abstinence syndrome by blocking NMDA receptors. Therefore, 40 mg/kg ketamine (K), 5 mg/kg dextromethorphan (DM), 5 mg/kg morphine (M) and 2 mg/kg naloxone (NL) alone or in combination with NL were IP administered to the rats five times during the daytime only for five days to see whether they would intensify abstinence syndrome through upregulation and/or supersensitivity of NMDA receptors. Three days following the implantation of three M-containing pellets, abstinence syndrome was brought about by 2 mg/kg NL injection. Jumping, wet dog shake, writhing, teeth chattering, diarrhoea, defecation and ptosis were observed for ten min. All drugs used alone or in combination with NL increased the intensity of abstinence syndrome. Since K and DM are noncompetitive NMDA receptor antagonists, the intensifying effect of NL or M was considered to be related to their interactions with NMDA receptors. Furthermore, on the basis of the results of the previous and present study, NL was claimed to act on NMDA receptors, like other opioids, but with higher affinity for and weaker blocking effect on NMDA receptors.

    Topics: Amino Acids; Analgesics; Animals; Behavior, Animal; Dextromethorphan; Dynorphins; Injections, Spinal; Kainic Acid; Male; Morphine Dependence; Naloxone; Peptide Fragments; Rats; Rats, Inbred Strains; Receptors, N-Methyl-D-Aspartate; Substance Withdrawal Syndrome

1991
Physical dependence liability of dynorphin A analogs in rodents.
    European journal of pharmacology, 1991, Aug-29, Volume: 201, Issue:2-3

    To assess the physical dependence liability of dynorphin A analogs, mice were given repeated injections of various dynorphin A analogs twice daily for 5 days, and rats were given repeated administration of [N-methyl-Tyr1,N-methyl-Arg7,D-Leu8]dynorphin-A-(1-8) ethylamide (E-2078) twice daily for up to 7 weeks. Mice that had received repeated [D-Cys2,Cys5,N-methyl-Arg7,D-Leu8]dynorphin-A-(1-9) amide displayed jumping behavior after subcutaneous injection of naloxone, an opioid receptor antagonist. In contrast, the animals that had received repeated E-2078 or [N-methyl-Tyr1,Phe4(p-NO2),N-methyl-Arg7,D-Leu8]dynorphin-A-(1-8) ethylamide displayed very few jumps after naloxone administration. Rats that had received repeated E-2078 administration did not display withdrawal signs, such as weight loss, after either abrupt withdrawal or naloxone administration. These results indicate that E-2078 and [N-methyl-Tyr1,Phe4(p-NO2),N-methyl-Arg7,D-Leu8]dynorphin-A-(1-8) ethylamide may have little dependence liability and that [D-Cys2,Cys5,N-methyl-Arg7,D-Leu8]dynorphin-A-(1-9) amide can cause physical dependence.

    Topics: Analgesics; Animals; Dynorphins; Injections, Subcutaneous; Male; Mice; Mice, Inbred Strains; Naloxone; Peptide Fragments; Rats; Rats, Inbred Strains; Substance Withdrawal Syndrome; Substance-Related Disorders

1991
Dynorphin A-(1-13) attenuates withdrawal in morphine-dependent rats: effect of route of administration.
    European journal of pharmacology, 1988, Jan-19, Volume: 145, Issue:3

    Rats were made tolerant to morphine by subcutaneous implantation of morphine alkaloid pellets. Three days after pellet implantation, withdrawal was induced by pellet removal and was assessed 6 h later. Immediately prior to withdrawal assessment, rats were injected with dynorphin A-(1-13) either i.th. (via a catheter), i.c.v. (via a cannula) or i.v. (via the tail vein). When administered i.th. in the dose range 1.25-5 nmol/rat, dynorphin A-(1-13) attenuated withdrawal over the 40 min observation period. Similarly, dynorphin A-(1-13) administered i.v. (37.5-150 nmol/kg) attenuated withdrawal, though only over the first 20 min following administration. Dynorphin A-(1-13) up to 10 nmol/rat had no effect on withdrawal scores. These data indicate that dynorphin acts at spinal sites to suppress withdrawal in morphine-dependent rats and may play a role in tolerance and dependence mechanisms.

    Topics: Animals; Catheters, Indwelling; Dynorphins; Injections, Intravenous; Injections, Intraventricular; Injections, Spinal; Male; Morphine; Narcotics; Peptide Fragments; Rats; Rats, Inbred Strains; Substance Withdrawal Syndrome; Visceral Prolapse; Yawning

1988
Evaluation on the treatment of morphine addiction by acupuncture Chinese herbs and opioid peptides.
    The American journal of Chinese medicine, 1986, Volume: 14, Issue:1-2

    Experimental studies on the effects of acupuncture, combined Chinese herbs, and opioid peptides on morphine withdrawal symptoms were carried out in 119 addicted rats. Electroacupuncture was found to be the most effective method as it reduced the morphine withdrawal scores to -85%. The combined herbs, Qiang Huo, Gou Teng, Chuan Xion, Fu Zi and Yan Hu Suo suppressed the withdrawal scores of -68%. The opioid peptides, endorphin, enkephalin, and dynorphin, produced marked sedative effect and alleviated the withdrawal symptoms, reducing the scores from -28% to -74%. It is suggested that acupuncture and herbs, being non-opiate and having less side effect, might be used as alternative or supplementary treatment on morphine addiction.

    Topics: Acupuncture Therapy; Animals; beta-Endorphin; Combined Modality Therapy; Dynorphins; Endorphins; Enkephalin, Methionine; Medicine, Chinese Traditional; Medicine, East Asian Traditional; Morphine; Plants, Medicinal; Rats; Rats, Inbred Strains; Substance Withdrawal Syndrome

1986
Comparison of the effectiveness of different opioid peptides in suppressing heroin withdrawal.
    European journal of pharmacology, 1984, Apr-20, Volume: 100, Issue:2

    The effectiveness of beta-endorphin, dynorphin-(1-13), dynorphin-(1-10) amide, alpha-neoendorphin and [D-Ala2,D-Leu5]enkephalin in suppressing withdrawal in heroin addicts was compared in this study. Groups of six patients were stabilized overnight in the hospital and were treated with either saline or peptide when withdrawal symptoms began to appear the following morning. Withdrawal was scored before and after treatment by the patient himself and an independent observer. Peptides were administered in a bolus dose of 60 micrograms/kg body weight. The patient, the observer and the physician who administered the injection were all blind to the nature of the compound given. All treatments, including those with saline, produced an overall reduction of withdrawal score. However, by statistical analysis, only treatments with beta-endorphin, [D-Ala2,D-Leu5]enkephalin and dynorphin-(1-13) were effective in producing a significant decrease of withdrawal symptoms. The length of relief brought about by the different peptides varied from less than an hour to a maximum of 5 h in one case. The average period of relief brought about by beta-endorphin, dynorphin-(1-13) and [D-Ala2,D-Leu5]enkephalin was 44, 46 and 60 min, respectively. Of the five peptides administered [D-Ala2,D-Leu5]enkephalin produced the largest number of side-effects.

    Topics: Adult; Animals; beta-Endorphin; Dynorphins; Endorphins; Enkephalin, Leucine; Enkephalin, Leucine-2-Alanine; Heroin; Humans; Male; Mice; Peptide Fragments; Protein Precursors; Substance Withdrawal Syndrome

1984
Possible regulatory role of dynorphin on morphine- and beta-endorphin-induced analgesia.
    The Journal of pharmacology and experimental therapeutics, 1981, Volume: 219, Issue:2

    In previous studies, we observed that dynorphin- (1-13), but not dynorphin-(1-9), can significantly inhibit morphine- or beta-endorphin-induced analgesia despite not having any appreciable analgesic activity itself. Dynorphin-(1-13) showed no inhibitory effect on Sandoz FK33824-induced analgesia. In the present study, we examined the effect of dynorphin on morphine-, beta-endorphin-, D-ala2-D-leu5-enkephalin- or Sandoz FK33824-induced analgesia in both naive, morphine-tolerant and morphine-dependent mice. It was found that although dynorphin may inhibit morphine- or beta-endorphin-induced analgesia in naive animals, the peptide is not effective in inhibiting D-ala2-D-leu5-enkephalin- or Sandoz FK33824-induced analgesia. Dynorphin is also effective in blocking spontaneous withdrawal jumping in morphine-dependent animals. It is suggested that dynorphin-(1-13) may play a modulatory role in regulating analgesia due to morphine or beta-endorphin, but not that due to enkephalin. The action of peptides on morphine- or beta-endorphin-induced analgesia in the naive state is different from that of the tolerant state, suggesting that dynorphin may be involved in the development of morphine tolerance and physical dependence.

    Topics: Analgesics; Animals; beta-Endorphin; Drug Interactions; Drug Tolerance; Dynorphins; Endorphins; Enkephalins; Humans; Male; Mice; Mice, Inbred ICR; Morphine; Morphine Dependence; Substance Withdrawal Syndrome

1981