Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 120
Filter
Add more filters

Complementary Medicines
Country/Region as subject
Publication year range
1.
Biosci Rep ; 41(8)2021 08 27.
Article in English | MEDLINE | ID: mdl-34355745

ABSTRACT

Traditional Chinese medicine detoxification prescription Chaihu-jia-Longgu-Muli decoction (CLMD) relieves depressive symptoms in patients withdrawing from methamphetamine. In the present study, we assessed the effects of CLMD on methamphetamine withdrawal in rats. A methamphetamine-intoxicated rat model was established. Rats were randomly divided into the control, model, high-dosage, medium-dosage, and low-dosage groups, receiving high, medium, and low doses of CLMD, respectively. Weekly body weight measurements revealed that rats treated with methamphetamine had the lowest body weight. The conditioned place preference (CPP) experiment revealed that methamphetamine-intoxicated rats stayed significantly longer in the drug-paired chamber than the control rats. However, after administering high-dosage CLMD, the amount of time the rats spent in the drug-paired chamber was significantly less than that of the model rats. Our open-field test revealed that the model group had lower crossing and rearing scores than the control group. Additionally, rats that received CLMD treatment exhibited higher crossing and rearing scores than the model rats. Striatal dopamine (DA), 5-hydroxytryptamine (5-HT), and endorphins (ß-EP) and serum interleukin (IL)-1α and IL-2 concentrations were estimated. Rats in the model group had lower striatal DA, 5-HT, and ß-EP and higher serum IL-1α and IL-2 concentrations than those in the control group. High-dosage CLMD administration significantly changed the concentrations of these molecules, such that they approached normal concentrations. In general, CLMD could prevent the development of methamphetamine-induced withdrawal symptoms in rats by increasing the DA, 5-HT, and ß-EP and lowering the IL-1α and IL-2 concentrations.


Subject(s)
Behavior, Animal/drug effects , Central Nervous System Stimulants , Conditioning, Psychological/drug effects , Corpus Striatum/drug effects , Drugs, Chinese Herbal/pharmacology , Methamphetamine , Substance Withdrawal Syndrome/drug therapy , Animals , Corpus Striatum/metabolism , Disease Models, Animal , Dopamine/metabolism , Interleukin-1alpha/blood , Interleukin-2/blood , Male , Open Field Test/drug effects , Rats, Sprague-Dawley , Serotonin/metabolism , Substance Withdrawal Syndrome/metabolism , Substance Withdrawal Syndrome/psychology , beta-Endorphin/metabolism
2.
Neuropharmacology ; 192: 108617, 2021 07 01.
Article in English | MEDLINE | ID: mdl-34019906

ABSTRACT

The epithalamic lateral habenula (LHb) regulates monoaminergic systems and contributes to the expression of both appetitive and aversive behaviours. Over the past years, the LHb has emerged as a vulnerable brain structure in mental illnesses including addiction. Behavioural and functional evidence in humans and rodents provide substantial support for a role of LHb in the negative affective symptoms emerging during withdrawal from addictive substances. Multiple forms of cellular and synaptic adaptations that take hold during drug withdrawal within the LHb are causally linked with the emergence of negative affective symptoms. These results indicate that targeting drug withdrawal-driven adaptations in the LHb may represent a potential strategy to normalize drug-related behavioural adaptations. In the current review we describe the mechanisms leading to functional alterations in the LHb, as well as the existing interventions used to counteract addictive behaviours. Finally, closing this loop we discuss and propose new avenues to potentially target the LHb in humans in light of the mechanistic understanding stemming from pre-clinical studies. Altogether, we provide an overview on how to leverage cellular-level understanding to envision clinically-relevant approaches for the treatment of specific aspects in drug addiction.


Subject(s)
Adaptation, Physiological/physiology , Behavior, Addictive/metabolism , Habenula/metabolism , Neurons/metabolism , Substance Withdrawal Syndrome/metabolism , Substance-Related Disorders/metabolism , Animals , Behavior, Addictive/genetics , Behavior, Addictive/therapy , Drug Evaluation, Preclinical/methods , Humans , Substance Withdrawal Syndrome/genetics , Substance Withdrawal Syndrome/therapy , Substance-Related Disorders/genetics , Substance-Related Disorders/therapy
3.
Brain Res Bull ; 172: 139-150, 2021 07.
Article in English | MEDLINE | ID: mdl-33901587

ABSTRACT

Mitragyna speciosa, also known as kratom, has been used for mitigating the severity of opioid withdrawal in humans. Its main indole alkaloid, mitragynine, has been considered as a pharmacotherapy for pain conditions and opioid replacement therapy. However, at high doses, chronic mitragynine may also have an addiction potential. The effects of chronic action of mitragynine in the brain are still unknown. The present study developed a mitragynine withdrawal model in rats and used it for a proteomic analysis of mitragynine withdrawal effects. Mitragynine (30 mg/kg, i.p.) was administered daily over a period of 14 days and then withdrawn. A proteomic analysis revealed that from a total of 1524 proteins identified, 31 proteins were upregulated, and 3 proteins were downregulated in the mitragynine withdrawal model. The Rab35 protein expression increased most profoundly in the mitragynine withdrawal group as compared to vehicle group. Therefore, it is proposed that Rab35 in the brain might be considered as a potential biomarker during mitragynine withdrawal and might be valuable target protein in developing new pharmacotherapies in the future.


Subject(s)
Brain/metabolism , Mitragyna , Plant Extracts/adverse effects , Secologanin Tryptamine Alkaloids/adverse effects , Substance Withdrawal Syndrome/metabolism , rab GTP-Binding Proteins/metabolism , Animals , Behavior, Animal/drug effects , Biomarkers/metabolism , Male , Proteomics , Rats , Rats, Sprague-Dawley
4.
Biomed Pharmacother ; 137: 111306, 2021 May.
Article in English | MEDLINE | ID: mdl-33524786

ABSTRACT

Protective effects of Puerariae flos extract (PFE) on ethanol (EtOH) exposure have been previously verified. This study attempts to explore the protective effects of PEF on EtOH withdrawal models. Sixty male Kunming mice were involved which were randomly divided into five groups (intact control, EtOH group (35-day EtOH exposure), EtOH withdrawal group (28-day exposure + 7-day withdrawal), EtOH withdrawal group + positive control (Deanxit) group, and EtOH withdrawal group + PFE group). The changes of neuropsychological behaviors; hippocampal BDNF expression and CA1 neuronal density; and plasma corticotropin-releasing hormone (CRH), ACTH, and CORT levels were observed. It was found that depression-like behaviors reduced by EtOH exposure and increased by withdrawal under the 28-day EtOH exposure and 7-day withdrawal conditions. In addition, anxiety-like behaviors worsened by EtOH exposure and unchanged by withdrawal. Deanxit and PEF ameliorated such behaviors (vs. withdrawal group). Hippocampal BDNF expression was significantly downregulated by EtOH exposure and upregulated by withdrawal. Deanxit and PEF significantly upregulated the BDNF expression. The hippocampal CA1 neuronal density significantly decreased by EtOH exposure but unchanged by withdrawal and treatments. The plasma CRH, ACTH, and CORT levels show a significant enhancement by EtOH exposure and reduced by withdrawal. They were further reduced by Deanxit and PEF. The protective effects of PEF on EtOH chronic withdrawal mouse models were verified. The results of this study also indicated a complicated scenario of neuropsychological behaviors, hippocampal BDNF expression, and hypothalamic-pituitary-adrenal axis which are affected by the timing of EtOH exposure and withdrawal.


Subject(s)
Alcoholism/drug therapy , Anxiety/prevention & control , CA1 Region, Hippocampal/drug effects , Depression/prevention & control , Drugs, Chinese Herbal/pharmacology , Hypothalamo-Hypophyseal System/drug effects , Pueraria , Substance Withdrawal Syndrome/drug therapy , Adrenocorticotropic Hormone/blood , Alcoholism/metabolism , Alcoholism/pathology , Alcoholism/psychology , Animals , Anxiety/metabolism , Anxiety/pathology , Anxiety/psychology , Behavior, Animal/drug effects , Brain-Derived Neurotrophic Factor/metabolism , CA1 Region, Hippocampal/metabolism , CA1 Region, Hippocampal/pathology , Corticotropin-Releasing Hormone/blood , Depression/metabolism , Depression/pathology , Depression/psychology , Disease Models, Animal , Drugs, Chinese Herbal/isolation & purification , Hydrocortisone/blood , Hypothalamo-Hypophyseal System/metabolism , Hypothalamo-Hypophyseal System/pathology , Male , Mice , Pueraria/chemistry , Substance Withdrawal Syndrome/metabolism , Substance Withdrawal Syndrome/pathology , Substance Withdrawal Syndrome/psychology
5.
Psychopharmacology (Berl) ; 238(3): 833-844, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33410985

ABSTRACT

RATIONALE: After alcohol ingestion, the brain partly switches from consumption of glucose to consumption of the alcohol metabolite acetate. In heavy drinkers, the switch persists after abrupt abstinence, leading to the hypothesis that the resting brain may be "starved" when acetate levels suddenly drop during abstinence, despite normal blood glucose, contributing to withdrawal symptoms. We hypothesized that ketone bodies, like acetate, could act as alternative fuels in the brain and alleviate withdrawal symptoms. OBJECTIVES: We previously reported that a ketogenic diet during alcohol exposure reduced acute withdrawal symptoms in rats. Here, our goals were to test whether (1) we could reproduce our findings, in mice and with longer alcohol exposure; (2) ketone bodies alone are sufficient to reduce withdrawal symptoms (clarifying mechanism); (3) introduction of ketogenic diets at abstinence (a clinically more practical implementation) would also be effective. METHODS: Male C57BL/6NTac mice had intermittent alcohol exposure for 3 weeks using liquid diet. Somatic alcohol withdrawal symptoms were measured as handling-induced convulsions; anxiety-like behavior was measured using the light-dark transition test. We tested a ketogenic diet, and a ketone monoester supplement with a regular carbohydrate-containing diet. RESULTS: The regular diet with ketone monoester was sufficient to reduce handling-induced convulsions and anxiety-like behaviors in early withdrawal. Only the ketone monoester reduced handling-induced convulsions when given during abstinence, consistent with faster elevation of blood ketones, relative to ketogenic diet. CONCLUSIONS: These findings support the potential utility of therapeutic ketosis as an adjunctive treatment in early detoxification in alcohol-dependent patients seeking to become abstinent. TRIAL REGISTRATION: clinicaltrials.gov NCT03878225, NCT03255031.


Subject(s)
Alcoholism/metabolism , Diet, Ketogenic , Ketone Bodies/metabolism , Ketones/therapeutic use , Substance Withdrawal Syndrome/prevention & control , Alcoholism/blood , Animals , Anxiety/drug therapy , Brain/metabolism , Clinical Trials as Topic , Dietary Supplements , Ethanol/administration & dosage , Ethanol/adverse effects , Ethanol/blood , Glucose , Humans , Male , Mice , Mice, Inbred C57BL , Rats , Substance Withdrawal Syndrome/metabolism , Substance Withdrawal Syndrome/psychology
6.
Cell Mol Neurobiol ; 41(5): 1131-1143, 2021 Jul.
Article in English | MEDLINE | ID: mdl-33433723

ABSTRACT

Chronic administration of opioids produces physical dependence and opioid-induced hyperalgesia. Users claim the Thai traditional tea "kratom" and component alkaloid mitragynine ameliorate opioid withdrawal without increased sensitivity to pain. Testing these claims, we assessed the combined kratom alkaloid extract (KAE) and two individual alkaloids, mitragynine (MG) and the analog mitragynine pseudoindoxyl (MP), evaluating their ability to produce physical dependence and induce hyperalgesia after chronic administration, and as treatments for withdrawal in morphine-dependent subjects. C57BL/6J mice (n = 10/drug) were administered repeated saline, or graded, escalating doses of morphine (intraperitoneal; i.p.), kratom alkaloid extract (orally, p.o.), mitragynine (p.o.), or MP (subcutaneously, s.c.) for 5 days. Mice treated chronically with morphine, KAE, or mitragynine demonstrated significant drug-induced hyperalgesia by day 5 in a 48 °C warm-water tail-withdrawal test. Mice were then administered naloxone (10 mg/kg, s.c.) and tested for opioid withdrawal signs. Kratom alkaloid extract and the two individual alkaloids demonstrated significantly fewer naloxone-precipitated withdrawal signs than morphine-treated mice. Additional C57BL/6J mice made physically dependent on morphine were then used to test the therapeutic potential of combined KAE, mitragynine, or MP given twice daily over the next 3 days at either a fixed dose or in graded, tapering descending doses. When administered naloxone, mice treated with KAE, mitragynine, or MP under either regimen demonstrated significantly fewer signs of precipitated withdrawal than control mice that continued to receive morphine. In conclusion, while retaining some liabilities, kratom, mitragynine, and mitragynine pseudoindoxyl produced significantly less physical dependence and ameliorated precipitated withdrawal in morphine-dependent animals, suggesting some clinical value.


Subject(s)
Analgesics, Opioid/adverse effects , Mitragyna , Morphine Dependence/prevention & control , Secologanin Tryptamine Alkaloids/administration & dosage , Secologanin Tryptamine Alkaloids/chemical synthesis , Substance Withdrawal Syndrome/prevention & control , Analgesics, Opioid/administration & dosage , Animals , Male , Mice , Mice, Inbred C57BL , Morphine Dependence/metabolism , Morphine Dependence/psychology , Pain Measurement/drug effects , Pain Measurement/methods , Receptors, Opioid, delta/agonists , Receptors, Opioid, delta/metabolism , Receptors, Opioid, mu/agonists , Receptors, Opioid, mu/metabolism , Secologanin Tryptamine Alkaloids/adverse effects , Secologanin Tryptamine Alkaloids/isolation & purification , Substance Withdrawal Syndrome/metabolism , Substance Withdrawal Syndrome/psychology
7.
Pharmacol Biochem Behav ; 200: 173088, 2021 01.
Article in English | MEDLINE | ID: mdl-33333134

ABSTRACT

Drugs of abuse and highly palatable foods (e.g. high fat or sweet foods) have powerful reinforcing effects, which can lead to compulsive and addictive drives to ingest these substances to the point of psychopathology and self-harm--specifically the development of Substance Use Disorder (SUD) and obesity. Both SUD and binge-like overeating can be defined as disorders in which the salience of the reward (food or drug) becomes exaggerated relative to, and at the expense of, other rewards that promote well-being. A major roadblock in the treatment of these disorders is high rates of relapse after periods of abstinence. It is common, although not universal, for cue-induced craving to increase over time with abstinence, often triggered by cues previously paired with the reinforcing substance. Accumulating evidence suggests that similar neural circuits and cellular mechanisms contribute to abstinence-induced and cue-triggered seeking of drugs and palatable food. Although much research has focused on the important role of corticolimbic circuitry in drug-seeking, our goal is to expand focus to the more recently explored hypothalamic-thalamic-striatal circuitry. Specifically, we review how connections, and neurotransmitters therein, among the lateral hypothalamus, paraventricular nucleus of the thalamus, and the nucleus accumbens contribute to abstinence-induced opioid- and (high fat or sweet) food-seeking. Given that biological sex and gonadal hormones have been implicated in addictive behavior across species, another layer to this review is to compare behaviors and neural circuit-based mechanisms of abstinence-induced opioid- or food-seeking between males and females when such data is available.


Subject(s)
Analgesics, Opioid/adverse effects , Behavior, Addictive/metabolism , Food/adverse effects , Neurobiology/methods , Substance Withdrawal Syndrome/metabolism , Animals , Craving/drug effects , Cues , Drug-Seeking Behavior/drug effects , Feeding Behavior , Female , Humans , Hypothalamus/metabolism , Male , Midline Thalamic Nuclei/metabolism , Nucleus Accumbens/drug effects , Recurrence , Reward , Sex Characteristics , Substance-Related Disorders/metabolism , Sucrose/adverse effects
8.
Neurosci Lett ; 739: 135347, 2020 11 20.
Article in English | MEDLINE | ID: mdl-33011195

ABSTRACT

Alcohol use disorders (AUD) are chronic relapsing brain disorder characterized by compulsive and heavy alcohol consumption. During acute withdrawal, patients with AUD display excessive daytime sleepiness, a condition linked to serious life-threatening complications, however, the mechanism is not known. Orexin and melanin-concentrating hormone (MCH) are the two hypothalamic neuropeptides that regulate many behaviors including sleep-wakefulness, and alcohol consumption, reinforcement, and reinstatement. Importantly, loss of orexin neurons causes narcolepsy, a severe sleep disorder with excessive daytime sleepiness. Does acute alcohol withdrawal reduce orexin gene expression? To investigate this, male Sprague-Dawley rats were divided in two groups: Rats were either administered with alcohol, mixed with infant formula (alcohol group) or control mixture containing water and infant formula (Controls) by gastric intubation every 8 h for 4 days using Majchrowicz's chronic binge drinking protocol. The doses of alcohol were adjusted depending on degree of intoxication, exhibited by animals, prior to each dose. The animals were euthanized after 12 h of last alcohol/water administration. During withdrawal, the hypothalamus was rapidly dissected out, and the expressions of orexin and MCH genes were examined by Real-time PCR. There was a significant reduction in orexin gene expression in rats subjected to alcohol withdrawal as compared to controls. No such change was observed in the MCH gene expression. These results suggest that downregulation of orexin gene expression may be a possible mechanism responsible for excessive daytime sleepiness associated with alcohol withdrawal in patients with AUD.


Subject(s)
Ethanol/administration & dosage , Gene Expression , Hypothalamus/metabolism , Orexins/metabolism , Substance Withdrawal Syndrome/metabolism , Animals , Down-Regulation , Hypothalamic Hormones/metabolism , Male , Protein Precursors/metabolism , Rats, Sprague-Dawley
9.
Neuropsychopharmacology ; 45(5): 866-876, 2020 04.
Article in English | MEDLINE | ID: mdl-31752015

ABSTRACT

Cognitive flexibility is the ability to switch strategic responses adaptively in changing environments. Cognitive rigidity imposed by neural circuit adaptations during nicotine abstinence may foster maladaptive nicotine taking in addicts. We systematically examined the effects of spontaneous withdrawal in mice exposed to either nicotine (6.3 or 18 mg/kg/day) or saline for 14 days on cognitive flexibility using an operant strategy set-shifting task. Because frontostriatal circuits are critical for cognitive flexibility and brain-derived neurotrophic factor (BDNF) modulates glutamate plasticity in these circuits, we also explored the effects of nicotine withdrawal on these neurochemical substrates. Mice undergoing nicotine withdrawal required more trials to attain strategy-switching criterion. Error analysis show that animals withdrawn from both nicotine doses committed higher perseverative errors, which correlated with measures of anxiety. However, animals treated with the higher nicotine dose also displayed more strategy maintenance errors that remained independent of negative affect. BDNF mRNA expression increased in the medial prefrontal cortex (mPFC) following nicotine withdrawal. Surprisingly, BDNF protein declined in mPFC but was elevated in dorsal striatum (DS). DS BDNF protein positively correlated with perseverative and maintenance errors, suggesting mPFC-DS overflow of BDNF during withdrawal. BDNF-evoked glutamate release and synapsin phosphorylation was attenuated within DS synapses, but enhanced in the nucleus accumbens, suggesting a dichotomous role of BDNF signaling in striatal regions. Taken together, these data suggest that spontaneous nicotine withdrawal impairs distinct components of cognitive set-shifting and these deficits may be linked to BDNF-mediated alterations in glutamate signaling dynamics in discrete frontostriatal circuits.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Corpus Striatum/drug effects , Frontal Lobe/drug effects , Glutamic Acid/metabolism , Nicotine/administration & dosage , Nicotinic Agonists/administration & dosage , Substance Withdrawal Syndrome/metabolism , Substance Withdrawal Syndrome/psychology , Adaptation, Physiological/drug effects , Animals , Behavior, Animal , Cognition/drug effects , Cognition/physiology , Conditioning, Operant/drug effects , Conditioning, Operant/physiology , Corpus Striatum/metabolism , Discrimination, Psychological/drug effects , Discrimination, Psychological/physiology , Frontal Lobe/metabolism , Male , Mice, Inbred C57BL , Synaptic Transmission/drug effects
10.
Horm Behav ; 119: 104650, 2020 03.
Article in English | MEDLINE | ID: mdl-31805280

ABSTRACT

Male Syrian hamsters (Mesocricetus auratus) administered anabolic/androgenic steroids during adolescent development display increased aggression and decreased anxious behavior during the adolescent exposure period. Upon withdrawal from anabolic/androgenic steroids, this neurobehavioral relationship shifts and hamsters exhibit decreased aggression and increased anxious behavior. This study investigated the hypothesis that alterations in anterior hypothalamic signaling through serotonin type-3 receptors modulate the behavioral shift between adolescent anabolic/androgenic steroid-induced aggressive and anxious behaviors during the withdrawal period. To test this, hamsters were administered anabolic/androgenic steroids during adolescence then withdrawn from drug exposure for 21 days and tested for aggressive and anxious behaviors following direct pharmacological manipulation of serotonin type-3 receptor signaling within the latero-anterior hypothalamus. Blockade of latero-anterior hypothalamic serotonin type-3 receptors both increased aggression and decreased anxious behavior in steroid-treated hamsters, effectively reversing the pattern of behavioral responding normally observed during anabolic/androgenic steroid withdrawal. These findings suggest that the state of serotonin neural signaling within the latero-anterior hypothalamus plays an important role in behavioral shifting between aggressive and anxious behaviors following adolescent exposure to anabolic/androgenic steroids.


Subject(s)
Aggression/drug effects , Anabolic Agents/pharmacology , Anxiety , Receptors, Serotonin, 5-HT3/physiology , Substance Withdrawal Syndrome/psychology , Androgens/pharmacology , Animals , Anxiety/chemically induced , Anxiety/metabolism , Anxiety/pathology , Behavior, Animal/drug effects , Cricetinae , Hypothalamus/drug effects , Hypothalamus/pathology , Male , Mesocricetus , Receptors, Serotonin, 5-HT3/metabolism , Serotonin/pharmacology , Sexual Maturation/drug effects , Substance Withdrawal Syndrome/metabolism , Substance Withdrawal Syndrome/pathology , Testosterone Congeners/pharmacology
11.
Sci Adv ; 5(9): eaax1342, 2019 09.
Article in English | MEDLINE | ID: mdl-31517050

ABSTRACT

A withdrawal-associated impairment in ß-endorphin neurotransmission in the arcuate nucleus (ARC) of the hypothalamus is associated with alcohol dependence characterized by a chronic relapsing disorder. Although acupuncture activates ß-endorphin neurons in the ARC projecting to the nucleus accumbens (NAc), a role for ARC ß-endorphin neurons in alcohol dependence and acupuncture effects has not been examined. Here, we show that acupuncture at Shenmen (HT7) points attenuates behavioral manifestation of alcohol dependence by activating endorphinergic input to the NAc from the ARC. Acupuncture attenuated ethanol withdrawal tremor, anxiety-like behaviors, and ethanol self-administration in ethanol-dependent rats, which are mimicked by local injection of ß-endorphin into the NAc. Acupuncture also reversed the decreased ß-endorphin levels in the NAc and a reduction of neuronal activity in the ARC during ethanol withdrawal. These results suggest that acupuncture may provide a novel, potential treatment strategy for alcohol use disorder by direct activation of the brain pathway.


Subject(s)
Acupuncture Therapy , Alcoholism , Arcuate Nucleus of Hypothalamus , Nucleus Accumbens , Substance Withdrawal Syndrome , beta-Endorphin/metabolism , Alcoholism/metabolism , Alcoholism/pathology , Alcoholism/therapy , Animals , Arcuate Nucleus of Hypothalamus/metabolism , Arcuate Nucleus of Hypothalamus/pathology , Male , Nucleus Accumbens/metabolism , Nucleus Accumbens/pathology , Rats , Rats, Wistar , Substance Withdrawal Syndrome/metabolism , Substance Withdrawal Syndrome/pathology , Substance Withdrawal Syndrome/therapy
12.
CNS Drugs ; 33(10): 1001-1030, 2019 10.
Article in English | MEDLINE | ID: mdl-31549358

ABSTRACT

Substance use disorder (SUD) is a major public health crisis worldwide, and effective treatment options are limited. During the past 2 decades, researchers have investigated the impact of a variety of pharmacological approaches to treat SUD, one of which is the use of medical cannabis or cannabinoids. Significant progress was made with the discovery of rimonabant, a selective CB1 receptor (CB1R) antagonist (also an inverse agonist), as a promising therapeutic for SUDs and obesity. However, serious adverse effects such as depression and suicidality led to the withdrawal of rimonabant (and almost all other CB1R antagonists/inverse agonists) from clinical trials worldwide in 2008. Since then, much research interest has shifted to other cannabinoid-based strategies, such as peripheral CB1R antagonists/inverse agonists, neutral CB1R antagonists, allosteric CB1R modulators, CB2R agonists, fatty acid amide hydrolase (FAAH) inhibitors, monoacylglycerol lipase (MAGL) inhibitors, fatty acid binding protein (FABP) inhibitors, or nonaddictive phytocannabinoids with CB1R or CB2R-binding profiles, as new therapeutics for SUDs. In this article, we first review recent progress in research regarding the endocannabinoid systems, cannabis reward versus aversion, and the underlying receptor mechanisms. We then review recent progress in cannabinoid-based medication development for the treatment of SUDs. As evidence continues to accumulate, neutral CB1R antagonists (such as AM4113), CB2R agonists (JWH133, Xie2-64), and nonselective phytocannabinoids (cannabidiol, ß-caryophyllene, ∆9-tetrahydrocannabivarin) have shown great therapeutic potential for SUDs, as shown in experimental animals. Several cannabinoid-based medications (e.g., dronabinol, nabilone, PF-04457845) that entered clinical trials have shown promising results in reducing withdrawal symptoms in cannabis and opioid users.


Subject(s)
Cannabinoid Receptor Agonists/pharmacology , Cannabinoid Receptor Antagonists/pharmacology , Receptors, Cannabinoid/metabolism , Substance Withdrawal Syndrome/drug therapy , Substance-Related Disorders/drug therapy , Substance-Related Disorders/metabolism , Animals , Humans , Ligands , Substance Withdrawal Syndrome/metabolism
13.
Brain Behav ; 9(9): e01378, 2019 09.
Article in English | MEDLINE | ID: mdl-31364821

ABSTRACT

BACKGROUND: Methamphetamine (Meth) seeking progressively increases after withdrawal (incubation of Meth craving). We previously demonstrated a role of anterior intralaminar nucleus of thalamus (AIT) to dorsomedial striatum (DMS) projections in this incubation. Here, we examined molecular alterations in DMS and AIT neurons activated (identified by neuronal activity marker Fos) during "incubated" Meth-seeking relapse test after prolonged withdrawal. METHODS: We trained male rats to self-administer Meth or saline (control condition) for 10 days (6 hr/day). Using fluorescence-activated cell sorting, we examined gene expression in Fos-positive (activated during a 2-hr relapse test) and Fos-negative (nonactivated) DMS and AIT neurons. RESULTS: In DMS, we found increased mRNA expressions of immediate early genes (IEGs) (Arc, Egr1, Npas4, Fosb), Trkb, glutamate receptors subunits (Gria3, Grin1, Grin2b, Grm1), and epigenetic enzymes (Hdac3, Hdac5, Crebbp) in Fos-positive neurons, compared with Fos-negative neurons. In AIT, we found that fewer genes (Egr1, Fosb, TrkB, Grin1, and Hdac5) exhibited increased mRNA expression in Fos-positive neurons. Unexpectedly, in both brain regions, gene alterations described above also occurred in drug-naïve saline self-administration control rats. CONCLUSIONS: These results demonstrated that transcriptional regulations in Fos-positive neurons activated during the relapse tests are brain region-specific but are not uniquely associated with drug exposure during the self-administration training.


Subject(s)
Corpus Striatum/metabolism , Gene Expression Regulation/drug effects , Methamphetamine/administration & dosage , Proto-Oncogene Proteins c-fos/metabolism , Substance Withdrawal Syndrome/genetics , Substance Withdrawal Syndrome/metabolism , Thalamus/metabolism , Animals , Corpus Striatum/drug effects , Craving/physiology , Disease Models, Animal , Male , Neurons/drug effects , Neurons/metabolism , Proto-Oncogene Proteins c-fos/genetics , Rats , Rats, Sprague-Dawley , Self Administration , Thalamus/drug effects
14.
J Pharmacol Exp Ther ; 371(2): 422-452, 2019 11.
Article in English | MEDLINE | ID: mdl-31391211

ABSTRACT

Opioid misuse and abuse is a major international public health issue. Opioid use disorder (OUD) is largely maintained by a desire to suppress aversive opioid withdrawal symptoms. Opioid withdrawal in patients seeking abstinence from illicit or prescribed opioids is often managed by provision of a µ-opioid agonist/partial agonist in combination with concomitant medications. Concomitant medications are administered based on their ability to treat specific symptoms rather than a mechanistic understanding of the opioid withdrawal syndrome; however, their use has not been statistically associated with improved treatment outcomes. Understanding the central and/or peripheral mechanisms that underlie individual withdrawal symptom expression in humans will help promote medication development for opioid withdrawal management. To support focused examination of mechanistically supported concomitant medications, this review summarizes evidence from preclinical (N = 68) and human (N = 30) studies that administered drugs acting on the dopamine, serotonin, cannabinoid, orexin/hypocretin, and glutamate systems and reported outcomes related to opioid withdrawal. These studies provide evidence that each of these systems contribute to opioid withdrawal severity. The Food and Drug Administration has approved medications acting on these respective systems for other indications and research in this area could support the repurposing of these medications to enhance opioid withdrawal treatment. These data support a focused examination of mechanistically informed concomitant medications to help reduce opioid withdrawal severity and enhance the continuum of care available for persons with OUD.


Subject(s)
Analgesics, Opioid/metabolism , Narcotic Antagonists/metabolism , Neurotransmitter Agents/metabolism , Opioid-Related Disorders/metabolism , Substance Withdrawal Syndrome/metabolism , Analgesics, Opioid/therapeutic use , Clinical Trials as Topic/methods , Dopamine/metabolism , Drug Evaluation, Preclinical/methods , Glutamic Acid/metabolism , Humans , Narcotic Antagonists/therapeutic use , Opioid-Related Disorders/drug therapy , Serotonin/metabolism , Substance Withdrawal Syndrome/drug therapy
15.
Nutrients ; 11(8)2019 Aug 14.
Article in English | MEDLINE | ID: mdl-31416242

ABSTRACT

Opioids are highly addictive substances with a relapse rate of over 90%. While preclinical models of chronic opioid exposure exist for studying opioid dependence, none recapitulate the relapses observed in human opioid addiction. The mechanisms associated with opioid dependence, the accompanying withdrawal symptoms, and the relapses that are often observed months or years after opioid dependence are poorly understood. Therefore, we developed a novel model of chronic opioid exposure whereby the level of administration is self-directed with periods of behavior acquisition, maintenance, and then extinction alternating with reinstatement. This profile arguably mirrors that seen in humans, with initial opioid use followed by alternating periods of abstinence and relapse. Recent evidence suggests that dietary interventions that reduce inflammation, including omega-3 polyunsaturated fatty acids (n-3 PUFAs), may reduce substance misuse liability. Using the self-directed intake model, we characterize the observed profile of opioid use and demonstrate that an n-3-PUFA-enriched diet ameliorates oxycodone-seeking behaviors in the absence of drug availability and reduces anxiety. Guided by the major role gut microbiota have on brain function, neuropathology, and anxiety, we profile the microbiome composition and the effects of chronic opioid exposure and n-3 PUFA supplementation. We demonstrate that the withdrawal of opioids led to a significant depletion in specific microbiota genera, whereas n-3 PUFA supplementation increased microbial richness, phylogenetic diversity, and evenness. Lastly, we examined the activation state of microglia in the striatum and found that n-3 PUFA supplementation reduced the basal activation state of microglia. These preclinical data suggest that a diet enriched in n-3 PUFAs could be used as a treatment to alleviate anxiety induced opioid-seeking behavior and relapse in human opioid addiction.


Subject(s)
Analgesics, Opioid , Behavior, Animal/drug effects , Dietary Supplements , Drug-Seeking Behavior/drug effects , Fatty Acids, Omega-3/administration & dosage , Gastrointestinal Microbiome/drug effects , Opioid-Related Disorders/drug therapy , Oxycodone , Substance Withdrawal Syndrome/drug therapy , Animals , Brain/drug effects , Brain/metabolism , Disease Models, Animal , Male , Mice, Inbred C57BL , Microglia/drug effects , Microglia/metabolism , Opioid-Related Disorders/metabolism , Opioid-Related Disorders/microbiology , Opioid-Related Disorders/psychology , Recurrence , Substance Withdrawal Syndrome/metabolism , Substance Withdrawal Syndrome/microbiology , Substance Withdrawal Syndrome/psychology
16.
Alcohol ; 76: 37-45, 2019 05.
Article in English | MEDLINE | ID: mdl-30554034

ABSTRACT

Alcohol produces complex effects on the immune system. Moderate alcohol use (1-2 drinks per day) has been shown to produce anti-inflammatory responses in human blood monocytes, whereas, the post mortem brains of severe alcoholics show increased immune gene expression and activated microglial markers. The present study was conducted to evaluate the time course of alcohol effects during exposure and after withdrawal, and to determine the relationship between microglial and cytokine responses in brain and blood. Forty-eight adult, male Wistar rats were exposed to chronic ethanol vapors, or air control, for 5 weeks. Following ethanol/air exposure blood and brains were collected at three time points: 1) while intoxicated, following 35 days of air/vapor exposure; 2) following 24 h of withdrawal from exposure, and 3) 28 days after withdrawal. One hemisphere of the brain was flash-frozen for cytokine analysis, and the other was fixed for immunohistochemical analysis. The ionized calcium-binding adapter molecule 1 (Iba-1) was used to evaluate microglia activation at the three time points, and rat cytokine/chemokine Magnetic Bead Panels (Millipore) were used to analyze frontal cortex tissue lysate and serum. Ethanol induced a significant increase in Iba-1 that peaked at day 35, remained significant after 1 day of withdrawal, and was elevated at day 28 in frontal cortex, amygdala, and substantia nigra. Ethanol exposure was associated with a transient reduction of the serum level of the major pro- and anti-inflammatory cytokines and chemokines and a transient increase of effectors of sterile inflammation. Little or no changes in these molecules were seen in the frontal cortex except for HMG1 and fractalkine that were reduced and elevated, respectively, at day 28 following withdrawal. These data show that ethanol exposure produces robust microglial activation; however, measures of inflammation in the blood differ from those in the brain over a protracted time course.


Subject(s)
Cytokines/metabolism , Ethanol/pharmacology , Frontal Lobe/metabolism , Microglia/drug effects , Substance Withdrawal Syndrome/metabolism , Animals , Calcium-Binding Proteins/metabolism , Cytokines/blood , Male , Microfilament Proteins/metabolism , Rats , Substance Withdrawal Syndrome/blood , Time Factors
17.
Br J Pharmacol ; 175(9): 1504-1518, 2018 05.
Article in English | MEDLINE | ID: mdl-29406581

ABSTRACT

BACKGROUND AND PURPOSE: Poor social behaviour and vulnerability to stress are major clinical features of stimulant use disorders. The corticotropin-releasing factor (CRF) system mediates stress responses and might underlie substance use disorders; however, its involvement in social impairment induced by stimulant substances remains unknown. CRF signalling is mediated by two receptor types, CRF1 and CRF2 . In the present study we investigated the role of the CRF2 receptor in social behaviour deficits, vulnerability to stress and related brain alterations induced by cocaine administration and withdrawal. EXPERIMENTAL APPROACH: CRF2 receptor-deficient (CRF2 -/-) and littermate wild-type mice were repeatedly tested in the three-chamber task for sociability (i.e. preference for an unfamiliar conspecific vs. an object) and social novelty preference (SNP; i.e. preference for a novel vs. a familiar conspecific) before and after chronic cocaine administration. An in situ hybridization assay was used to assess gene expression of the stress-responsive arginine vasopressin (AVP) and oxytocin (OT) neuropeptides in the hypothalamus. KEY RESULTS: CRF2 receptor deficiency eliminated the sociability deficit induced by cocaine withdrawal. Moreover, CRF2 -/- mice did not show either the stress-induced sociability deficit or the increased AVP and OT expression associated with long-term cocaine withdrawal, indicating resilience to stress. Throughout, wild-type and CRF2 -/- mice displayed SNP, suggesting that cocaine withdrawal-induced sociability deficits were not due to impaired detection of social stimuli. CONCLUSIONS AND IMPLICATIONS: These findings demonstrate a central role for the CRF2 receptor in social behaviour deficits and biomarkers of vulnerability induced by cocaine withdrawal, suggesting new therapeutic strategies for stimulant use disorders.


Subject(s)
Cocaine/adverse effects , Receptors, Corticotropin-Releasing Hormone/deficiency , Social Behavior , Stress, Psychological/psychology , Substance Withdrawal Syndrome/psychology , Animals , Arginine Vasopressin/biosynthesis , Corticotropin-Releasing Hormone , Exploratory Behavior , Hypothalamus/metabolism , Male , Mice , Mice, Knockout , Oxytocin/biosynthesis , Receptors, Corticotropin-Releasing Hormone/genetics , Stress, Psychological/metabolism , Substance Withdrawal Syndrome/metabolism
18.
Pharmacol Biochem Behav ; 161: 6-12, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28882570

ABSTRACT

Alcohol use disorder is an alarming health problem, and the withdrawal symptoms increase the risk of relapse. We have hypothesized that taurine, a multitarget substance acting as a gamma-aminobutyric acid A receptor (GABAAR) positive modulator and a partial inhibitor of N-methyl-d-aspartate (NMDA) glutamate receptors, may reduce the withdrawal symptoms or modify behaviors when combined with alcohol. Therefore, we investigated the effects of taurine on behavior in the open field test (OFT), the GABAAR α2 subunit and BDNF mRNA expression in the frontal cortex of rats after chronic alcohol treatment or upon withdrawal. Rats received alcohol 2g/kg (alcohol and withdrawal groups) or water (control group) twice daily by oral gavage for 28days. On day 29, the withdrawal rats received water instead of alcohol, and all groups were reallocated to receive 100mg/kg taurine or vehicle intraperitoneally, once a day for 5days. On day 33, the rats were exposed to OFT; 18h later, they were euthanized, and the frontal cortex was dissected for GABAAR α2 subunit detection and BDNF mRNA expression determination by real-time quantitative PCR. Taurine administration restored rearing behavior to the control levels in the withdrawal rats. Taurine also showed anxiolytic-like effects in control rats and did not change the behaviors in the chronic alcohol group. Chronic alcohol treatment or withdrawal did not change the GABAAR α2 subunit or BDNF mRNA expression in the frontal cortex, but taurine decreased the α2 subunit level in control rats and to the BDNF levels in the alcohol rat group. We conclude that taurine restored exploratory behavior after alcohol withdrawal but that this effect was not related to the GABAAR α2 subunit or BDNF mRNA expression in the frontal cortex of the rats.


Subject(s)
Brain-Derived Neurotrophic Factor/biosynthesis , Ethanol/administration & dosage , Exploratory Behavior/physiology , Frontal Lobe/metabolism , RNA, Messenger/biosynthesis , Substance Withdrawal Syndrome/metabolism , Taurine/pharmacology , Alcoholism/drug therapy , Alcoholism/genetics , Alcoholism/metabolism , Animals , Brain-Derived Neurotrophic Factor/antagonists & inhibitors , Brain-Derived Neurotrophic Factor/genetics , Exploratory Behavior/drug effects , Frontal Lobe/drug effects , Gene Expression , Male , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley , Rats, Wistar , Substance Withdrawal Syndrome/drug therapy , Substance Withdrawal Syndrome/genetics , Taurine/therapeutic use
19.
Psychopharmacology (Berl) ; 234(15): 2299-2309, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28567696

ABSTRACT

RATIONALE: One of the most commonly cited reasons for chronic cannabis use is to cope with stress. Consistent with this, cannabis users have shown reduced emotional arousal and dampened stress reactivity in response to negative imagery. OBJECTIVES: To our knowledge, the present study represents the first to examine the effects of an acute stress manipulation on subjective stress and salivary cortisol in chronic cannabis users compared to non-users. METHODS: Forty cannabis users and 42 non-users were randomly assigned to complete either the stress or no stress conditions of the Maastricht Acute Stress Test (MAST). The stress condition of the MAST manipulates both physiological (placing hand in ice bath) and psychosocial stress (performing math under conditions of social evaluation). Participants gave baseline subjective stress ratings before, during, and after the stress manipulation. Cortisol was measured from saliva samples obtained before and after the stress manipulation. Further, cannabis cravings and symptoms of withdrawal were measured. RESULTS: Subjective stress ratings and cortisol levels were significantly higher in non-users in the stress condition relative to non-users in the no stress condition. In contrast, cannabis users demonstrated blunted stress reactivity; specifically, they showed no increase in cortisol and a significantly smaller increase in subjective stress ratings. The stress manipulation had no impact on cannabis users' self-reported cravings or withdrawal symptoms. CONCLUSION: Chronic cannabis use is associated with blunted stress reactivity. Future research is needed to determine whether this helps to confer resiliency or vulnerability to stress-related psychopathology as well as the mechanisms underlying this effect.


Subject(s)
Hydrocortisone/metabolism , Marijuana Smoking/metabolism , Marijuana Smoking/psychology , Saliva/metabolism , Stress, Psychological/metabolism , Stress, Psychological/psychology , Adaptation, Psychological/physiology , Adult , Cannabis , Emotions/physiology , Female , Humans , Hydrocortisone/analysis , Male , Middle Aged , Saliva/chemistry , Self Report , Substance Withdrawal Syndrome/metabolism , Substance Withdrawal Syndrome/psychology
20.
ChemMedChem ; 12(16): 1408-1416, 2017 08 22.
Article in English | MEDLINE | ID: mdl-28417566

ABSTRACT

Cannabis sativa withdrawal syndrome is characterized mainly by psychological symptoms. By using computational tools, the aim of this study was to propose drug candidates for treating withdrawal syndrome based on the natural ligands of the cannabinoid type 1 receptor (CB1). One compound in particular, 2-n-butyl-5-n-pentylbenzene-1,3-diol (ZINC1730183, also known as stemphol), showed positive predictions as a human CB1 ligand and for facile synthetic accessibility. Therefore, ZINC1730183 is a favorable candidate scaffold for further research into pharmacotherapeutic alternatives to treat C. sativa withdrawal syndrome.


Subject(s)
Cannabis/chemistry , Ligands , Receptor, Cannabinoid, CB1/metabolism , Animals , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Caco-2 Cells , Cannabis/metabolism , Cell Membrane Permeability/drug effects , Drug Design , Half-Life , Humans , Mice , Receptor, Cannabinoid, CB1/agonists , Receptor, Cannabinoid, CB2/agonists , Receptor, Cannabinoid, CB2/metabolism , Resorcinols/chemistry , Resorcinols/pharmacokinetics , Resorcinols/pharmacology , Resorcinols/therapeutic use , Substance Withdrawal Syndrome/drug therapy , Substance Withdrawal Syndrome/metabolism , Substance Withdrawal Syndrome/pathology
SELECTION OF CITATIONS
SEARCH DETAIL