Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 360
1.
Article En | MEDLINE | ID: mdl-38365104

Methamphetamine (METH) abuse is associated with a spectrum of behavioral consequences, among which heightened aggression presents a significant challenge. However, the causal role of METH's impact in aggression and its target circuit mechanisms remains largely unknown. We established an acute METH exposure-aggression mouse model to investigate the role of ventral tegmental area (VTA) dopaminergic neurons and ventral medial hypothalamus VMH glutamatergic neuron. Our findings revealed that METH-induced VTA dopamine excitability activates the ventromedial hypothalamus (VMH) glutamatergic neurons, contributing to pathological aggression. Notably, we uncovered a dopaminergic transmission within the VTA-VMH circuit that exclusively functioned under METH influence. This dopaminergic pathway emerged as a potential key player in enabling dopamine-related pathological aggression, with heightened dopaminergic excitability implicated in various psychiatric symptoms. Also, the modulatory function of this pathway opens new possibilities for targeted therapeutic strategies for intervention to improve treatment in METH abuse and may have broader implications for addressing pathological aggression syndromes.


Amphetamine-Related Disorders , Methamphetamine , Mice , Animals , Methamphetamine/pharmacology , Aggression , Dopamine/metabolism , Ventral Tegmental Area/metabolism , Dopaminergic Neurons/metabolism , Amphetamine-Related Disorders/metabolism , Hypothalamus, Middle/metabolism
2.
J Ethnopharmacol ; 314: 116635, 2023 Oct 05.
Article En | MEDLINE | ID: mdl-37182675

ETHNOPHARMACOLOGICAL RELEVANCE: Uncaria rhynchophylla (Miq.) Miq. ex Havil. is a plant species that is routinely devoted in traditional Chinese medicine to treat central nervous system disorders. Rhynchophylline (Rhy), a predominant alkaloid isolated from Uncaria rhynchophylla (Miq.) Miq. ex Havil., has been demonstrated to reverse methamphetamine-induced (METH-induced) conditioned place preference (CPP) effects in mice, rats and zebrafish. The precise mechanism is still poorly understood, thus further research is necessary. AIM OF STUDY: This study aimed to investigate the role of miRNAs in the inhibitory effect of Rhy on METH dependence. MATERIALS AND METHODS: A rat CPP paradigm and a PC12 cell addiction model were established. Microarray assays were used to screen and identify the candidate miRNA. Behavioral assessment, real-time PCR, dual-luciferase reporter assay, western blotting, stereotaxic injection of antagomir/agomir and cell transfection experiments were performed to elucidate the effect of the candidate miRNA and intervention mechanism of Rhy on METH dependence. RESULTS: Rhy successfully reversed METH-induced CPP effect and the upregulated miR-181a-5p expression in METH-dependent rat hippocampus and PC12 cells. Moreover, suppression of miR-181a-5p by antagomir 181a reversed METH-induced CPP effect. Meanwhile, overexpression of miR-181a-5p by agomir 181a in combination with low-dose METH (0.5 mg/kg) elicited a significant CPP effect, which was blocked by Rhy through inhibiting miR-181a-5p. Finally, the result demonstrated that miR-181a-5p exerted its regulatory role by targeting γ-aminobutyric acid A receptor α1 (GABRA1) both in vivo and in vitro. CONCLUSION: This finding reveals that Rhy inhibits METH dependence via modulating the miR-181a-5p/GABRA1 axis, which may be a promising target for treatment of METH dependence.


Amphetamine-Related Disorders , Methamphetamine , MicroRNAs , Rats , Mice , Animals , Receptors, GABA , Antagomirs , Zebrafish/genetics , Amphetamine-Related Disorders/genetics , Amphetamine-Related Disorders/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Methamphetamine/pharmacology
3.
Neurotox Res ; 41(4): 324-337, 2023 Aug.
Article En | MEDLINE | ID: mdl-37014368

Methamphetamine (Meth), a commonly used central nervous system stimulant, is highly addictive. Currently, there is no effective treatment for Meth dependence and abuse, although cell adhesion molecules (CAMs) have been shown to play an important role in the formation and remodeling of synapses in the nervous system while also being involved in addictive behavior. Contactin 1 (CNTN1) is a CAM that is widely expressed in the brain; nevertheless, its role in Meth addiction remains unclear. Therefore, in the present study, we established mouse models of single and repeated Meth exposure and subsequently determined that CNTN1 expression in the nucleus accumbens (NAc) was upregulated in mice following single or repeated Meth exposure, whereas CNTN1 expression in the hippocampus was not significantly altered. Intraperitoneal injection of the dopamine receptor 2 antagonist haloperidol reversed Meth-induced hyperlocomotion and upregulation of CNTN1 expression in the NAc. Additionally, repeated Meth exposure also induced conditioned place preference (CPP) in mice and upregulated the expression levels of CNTN1, NR2A, NR2B, and PSD95 in the NAc. Using an AAV-shRNA-based approach to specifically silence CNTN1 expression in the NAc via brain stereotaxis reversed Meth-induced CPP and decreased the expression levels of NR2A, NR2B, and PSD95 in the NAc. These findings suggest that CNTN1 expression in the NAc plays an important role in Meth-induced addiction, and the underlying mechanism may be related to the expression of synapse-associated proteins in the NAc. The results of this study improved our understanding of the role of cell adhesion molecules in Meth addiction.


Amphetamine-Related Disorders , Central Nervous System Stimulants , Methamphetamine , Mice , Animals , Methamphetamine/pharmacology , Nucleus Accumbens , Contactin 1/metabolism , Central Nervous System Stimulants/pharmacology , Brain/metabolism , Amphetamine-Related Disorders/metabolism
4.
Neurosci Lett ; 792: 136952, 2023 01 01.
Article En | MEDLINE | ID: mdl-36336087

Drug addiction, including methamphetamine (METH) addiction, is a significant public health and social issue. Perturbations in intracellular Ca2+ homeostasis are associated with drug addiction. K+-dependent Na+/Ca2+ exchanger 2 (NCKX2) is located on neuronal cell membranes and constitutes a Ca2+ clearance mechanism, with key roles in synaptic plasticity. NCKX2 is associated with motor learning, memory, and cognitive functions. However, the role of NCKX2 in METH addiction remains unclear. In this study, we investigated the expression levels of NCKX2 in four addiction-related brain regions: the prefrontal cortex (PFc), nucleus accumbens (NAc), dorsal striatum (DS), and hippocampus (Hip) in a C57/BL6 mouse model of METH-induced conditioned place preference (CPP) and behavioral sensitization. Levels of NCKX2 were unchanged in these brain regions in mice with METH-induced CPP but were decreased in the PFc and NAc of mice with METH-induced behavioral sensitization. Adeno-associated virus (AAV)-mediated overexpression of NCKX2 in the PFc attenuated the expression phase of METH-induced behavioral sensitization in mice, whereas AAV-mediated knockdown of NCKX2 enhanced the effects of METH. Collectively, our results suggest that NCKX2 is involved in METH-induced behavioral sensitization but does not affect conditioned reward-related memory, highlighting the potential of NCKX2 as a molecular target for studying the mechanisms underscoring METH addiction.


Amphetamine-Related Disorders , Central Nervous System Stimulants , Methamphetamine , Animals , Mice , Methamphetamine/pharmacology , Sodium-Calcium Exchanger/metabolism , Nucleus Accumbens/metabolism , Amphetamine-Related Disorders/metabolism , Reward , Central Nervous System Stimulants/pharmacology
5.
World J Biol Psychiatry ; 24(3): 209-222, 2023 03.
Article En | MEDLINE | ID: mdl-35673936

OBJECTIVES: Methamphetamine (METH) as a potent psychostimulant drug with a high potency of dependence rate that results in neurotoxicity has become a major drug of abuse in many parts of the world. Unfortunately, there is limited evidence regarding treatment of METH withdrawal syndrome. Therefore, we aimed to investigate whether metformin mitigate the methamphetamine (METH) withdrawal syndrome in male mice. Based on the literature, depression and anxiety are the major METH withdrawal symptoms. METHODS: Here, METH (2 mg/kg) was administered to mice twice a day for 14 constitutive days to induce animal model of METH-induced withdrawal syndrome. To do this, mice in control group and those with METH withdrawal syndrome were divided into treatment (receiving metformin in 3 doses of 50, 100 and 200 mg/kg for 10 days) and non-treatment sub-groups. Following the behavioural test, the animals were sacrificed; their hippocampus was dissected to measure oxidative stress parameters and expression of cellular energy homeostasis and immune-inflammatory genes. RESULTS: Our data revealed that metformin provoked antidepressant effects in behavioural tests through AMPK overexpression as an important mitochondrial energetic sensor and inhibition of Tlr4 overexpression in the immune system gene expression. In addition, metformin was able to improve oxidative stress biomarkers and neuronal damage in the hippocampus and restore cellular energy homeostasis and immune system gene expression. CONCLUSIONS: The data suggested that metformin can influence the hippocampus through targeting mitochondria and their performance, and consequently, neuroinflammation responses and brain metabolic changes. It is supposed to be a new therapeutic option in clinical trials of depression and anxiety following METH withdrawal treatment.


Amphetamine-Related Disorders , Central Nervous System Stimulants , Methamphetamine , Substance Withdrawal Syndrome , Mice , Male , Animals , Central Nervous System Stimulants/adverse effects , Methamphetamine/adverse effects , Amphetamine-Related Disorders/drug therapy , Amphetamine-Related Disorders/metabolism , Substance Withdrawal Syndrome/drug therapy , Brain/metabolism
6.
J Neurosci ; 43(5): 803-811, 2023 02 01.
Article En | MEDLINE | ID: mdl-36564185

Anxiety is one of the most common withdrawal symptoms of methamphetamine (METH) abuse, which further drives relapse to drugs. Interpeduncular nucleus (IPN) has been implicated in anxiety-like behaviors and addiction, yet its role in METH-abstinence-induced anxiety remains unknown. Here, we found that prolonged abstinence from METH enhanced anxiety-like behaviors in male mice, accompanied by more excited IPN GABAergic neurons, as indicated by the increased c-fos expression and the enhanced neuronal excitability by electrophysiological recording in the GABAergic neurons. Using the designer receptors exclusively activated by designer drugs method, specific inhibition of IPN GABAergic neurons rescued the aberrant neuronal excitation of IPN GABAergic neurons and efficiently reduced anxiety-like behaviors, whereas it did not induce depression-like behaviors in male mice after prolonged abstinence from METH. These findings reveal that IPN GABAergic neurons should be a promising brain target to alleviate late withdrawal symptoms from METH with few side effects.SIGNIFICANCE STATEMENT Prolonged abstinence from METH triggers IPN GABAergic neurons and ultimately increases anxiety in male mice. Suppressing IPN GABAergic neurons rescues METH abstinence-induced aberrant neuronal excitation of IPN GABAergic neurons and efficiently reduces anxiety in mice.


Amphetamine-Related Disorders , Interpeduncular Nucleus , Methamphetamine , Substance Withdrawal Syndrome , Mice , Male , Animals , Methamphetamine/pharmacology , Interpeduncular Nucleus/metabolism , Anxiety/metabolism , GABAergic Neurons/metabolism , Substance Withdrawal Syndrome/metabolism , Amphetamine-Related Disorders/metabolism
7.
Behav Brain Res ; 437: 114109, 2023 02 02.
Article En | MEDLINE | ID: mdl-36108778

Human immunodeficiency virus (HIV) continues to infect millions worldwide, negatively impacting neurobehavioral function. Further understanding of the combined effects of HIV and methamphetamine use is crucial, as methamphetamine use is prevalent in people with HIV. The HIV-associated protein Tat may contribute to cognitive dysfunction, modeled preclinically in mice using doxycycline (DOX)-inducible Tat expression (iTat). Tat may exert its effects on cognitive function via disruption of the dopamine transporter, similar to the action of methamphetamine. Additionally, Tat and methamphetamine both decrease interneuron populations, including those expressing calbindin. It is important to understand the combined effects of Tat and methamphetamine in preclinical models of HIV infection. Here, we used iTat transgenic mice and a chronic binge regimen of methamphetamine exposure to determine their combined impact on reward learning and motivation. We also measured calbindin expression in behavior-relevant brain regions. Before induction with DOX, iTat mice exhibited no differences in behavior. Chronic methamphetamine exposure before Tat induction impaired initial reward learning but did not affect motivation. Furthermore, DOX-induced Tat expression did not alter behavior, but slowed latencies to retrieve rewards. This effect of Tat, however, was not observed in methamphetamine-treated mice, indicative of a potential protective effect. Finally, Tat expression was associated with an increase in calbindin-expressing cells in the VTA, while methamphetamine exposure did not alter calbindin numbers. These findings may indicate a protective role of methamphetamine in HIV neuropathology, which in turn may help in our understanding of why people with HIV use methamphetamine at disproportionately higher rates.


Amphetamine-Related Disorders , HIV Infections , Methamphetamine , tat Gene Products, Human Immunodeficiency Virus , Animals , Humans , Mice , Calbindins/metabolism , Disease Models, Animal , HIV Infections/complications , HIV Infections/psychology , Methamphetamine/adverse effects , Methamphetamine/pharmacology , Mice, Transgenic , Reward , tat Gene Products, Human Immunodeficiency Virus/metabolism , Amphetamine-Related Disorders/complications , Amphetamine-Related Disorders/metabolism
8.
Article En | MEDLINE | ID: mdl-38707245

Methamphetamine use disorder (MUD) is a neuropsychiatric disorder characterized by binge drug taking episodes, intervals of abstinence, and relapses to drug use even during treatment. MUD has been modeled in rodents and investigators are attempting to identify its molecular bases. Preclinical experiments have shown that different schedules of methamphetamine self-administration can cause diverse transcriptional changes in the dorsal striatum of Sprague-Dawley rats. In the present review, we present data on differentially expressed genes (DEGs) identified in the rat striatum following methamphetamine intake. These include genes involved in transcription regulation, potassium channel function, and neuroinflammation. We then use the striatal data to discuss the potential significance of the molecular changes induced by methamphetamine by reviewing concordant or discordant data from the literature. This review identified potential molecular targets for pharmacological interventions. Nevertheless, there is a need for more research on methamphetamine-induced transcriptional consequences in various brain regions. These data should provide a more detailed neuroanatomical map of methamphetamine-induced changes and should better inform therapeutic interventions against MUD.


Amphetamine-Related Disorders , Central Nervous System Stimulants , Disease Models, Animal , Methamphetamine , Animals , Methamphetamine/pharmacology , Methamphetamine/administration & dosage , Amphetamine-Related Disorders/metabolism , Amphetamine-Related Disorders/physiopathology , Amphetamine-Related Disorders/genetics , Rats , Central Nervous System Stimulants/pharmacology , Epigenesis, Genetic/drug effects , Recurrence , Brain/metabolism , Brain/drug effects
9.
Int J Mol Sci ; 23(19)2022 Sep 23.
Article En | MEDLINE | ID: mdl-36232524

Methamphetamine (METH) is a highly addictive drug abused by millions of users worldwide, thus becoming a global health concern with limited management options. The inefficiency of existing treatment methods has driven research into understanding the mechanisms underlying METH-induced disorders and finding effective treatments. This study aims to understand the complex interactions of the gastrointestinal-immune-nervous systems following an acute METH dose administration as one of the potential underlying molecular mechanisms concentrating on the impact of METH abuse on gut permeability. Findings showed a decreased expression of tight junction proteins ZO-1 and EpCAm in intestinal tissue and the presence of FABP-1 in sera of METH treated mice suggests intestinal wall disruption. The increased presence of CD45+ immune cells in the intestinal wall further confirms gut wall inflammation/disruption. In the brain, the expression of inflammatory markers Ccl2, Cxcl1, IL-1ß, TMEM119, and the presence of albumin were higher in METH mice compared to shams, suggesting METH-induced blood-brain barrier disruption. In the spleen, cellular and gene changes are also noted. In addition, mice treated with an acute dose of METH showed anxious behavior in dark and light, open field, and elevated maze tests compared to sham controls. The findings on METH-induced inflammation and anxiety may provide opportunities to develop effective treatments for METH addiction in the future.


Amphetamine-Related Disorders , Central Nervous System Stimulants , Methamphetamine , Albumins/metabolism , Amphetamine-Related Disorders/metabolism , Animals , Anxiety , Blood-Brain Barrier/metabolism , Brain/metabolism , Central Nervous System Stimulants/pharmacology , Epithelial Cell Adhesion Molecule/metabolism , Inflammation/metabolism , Methamphetamine/metabolism , Methamphetamine/toxicity , Mice
10.
Psychopharmacology (Berl) ; 239(9): 2997-3008, 2022 Sep.
Article En | MEDLINE | ID: mdl-35881147

RATIONALE: MicroRNA (miRNA) control of post-transcription gene expression in the nucleus accumbens (NAc) has been implicated in methamphetamine (METH) dependence. Conditioned place preference (CPP) is a classical animal procedure that reflects the rewarding effects of addictive drugs. miR-222-3p has been reported to play a key role in various neurological diseases and is strongly associated with alcohol dependence. Nevertheless, the role of miR-222-3p in METH dependence remains unclear. OBJECTIVE: To explore the molecular mechanisms underlying the role of miR-222-3p in the NAc in METH-induced CPP. METHODS: miR-222-3p expression in the NAc of METH-induced CPP mice was detected by quantitative real-time (qPCR). Following adeno-associated virus (AAV)-mediated overexpression or knockdown of miR-222-3p in the NAc, mice were subjected to CPP to investigate the effects of miR-222-3p on METH-induced CPP. Target genes of mir-222-3p were predicted using bioinformatics analysis. Candidate target genes for METH-induced CPP were validated by qPCR. RESULTS: miR-222-3p expression in the NAc was decreased in CPP mice. Overexpression of miR-222-3p in the NAc blunted METH-induced CPP. Ppp3r1, Cdkn1c, Fmr1, and PPARGC1A were identified as target gene transcripts potentially mediating the effects of miR-222-3p on METH-induced CPP. CONCLUSION: Our results highlight miR-222-3p as a key epigenetic regulator in METH-induced CPP and suggest a potential role for miR-222-3p in the regulation of METH-induced reward-related changes in the brain.


Amphetamine-Related Disorders , Central Nervous System Stimulants , Methamphetamine , MicroRNAs , Amphetamine-Related Disorders/metabolism , Animals , Central Nervous System Stimulants/metabolism , Central Nervous System Stimulants/pharmacology , Fragile X Mental Retardation Protein , Methamphetamine/metabolism , Methamphetamine/pharmacology , Mice , MicroRNAs/genetics , MicroRNAs/metabolism , Nucleus Accumbens
11.
Int J Mol Sci ; 23(9)2022 Apr 28.
Article En | MEDLINE | ID: mdl-35563262

Methamphetamine (MA) is a highly addictive psychostimulant drug, and the number of MA-related overdose deaths has reached epidemic proportions. Repeated MA exposure induces a robust and persistent neuroinflammatory response, and the evidence supports the potential utility of targeting neuroimmune function using non-selective phosphodiesterase 4 (PDE4) inhibitors as a therapeutic strategy for attenuating addiction-related behavior. Off-target, emetic effects associated with non-selective PDE4 blockade led to the development of isozyme-selective inhibitors, of which the PDE4B-selective inhibitor A33 was demonstrated recently to reduce binge drinking in two genetically related C57BL/6 (B6) substrains (C57BL/6NJ (B6NJ) and C57BL/6J (B6J)) that differ in their innate neuroimmune response. Herein, we determined the efficacy of A33 for reducing MA self-administration and MA-seeking behavior in these two B6 substrains. Female and male mice of both substrains were first trained to nose poke for a 100 mg/L MA solution followed by a characterization of the dose-response function for oral MA reinforcement (20 mg/L-3.2 g/L), the demand-response function for 400 mg/L MA, and cue-elicited MA seeking following a period of forced abstinence. During this substrain comparison of MA self-administration, we also determined the dose-response function for A33 pretreatment (0-1 mg/kg) on the maintenance of MA self-administration and cue-elicited MA seeking. Relative to B6NJ mice, B6J mice earned fewer reinforcers, consumed less MA, and took longer to reach acquisition criterion with males of both substrains exhibiting some signs of lower MA reinforcement than their female counterparts during the acquisition phase of the study. A33 pretreatment reduced MA reinforcement at all doses tested. These findings provide the first evidence that pretreatment with a selective PDE4B inhibitor effectively reduces MA self-administration in both male and female mice of two genetically distinct substrains but does not impact cue-elicited MA seeking following abstinence. If relevant to humans, these results posit the potential clinical utility of A33 or other selective PDE4B inhibitors for curbing active drug-taking in MA use disorder.


Amphetamine-Related Disorders , Central Nervous System Stimulants , Methamphetamine , Phosphodiesterase 4 Inhibitors , Amphetamine-Related Disorders/drug therapy , Amphetamine-Related Disorders/metabolism , Animals , Central Nervous System Stimulants/pharmacology , Cyclic Nucleotide Phosphodiesterases, Type 4 , Female , Male , Methamphetamine/adverse effects , Mice , Mice, Inbred C57BL , Phosphodiesterase 4 Inhibitors/pharmacology , Reinforcement, Psychology , Self Administration
12.
Physiol Behav ; 251: 113804, 2022 07 01.
Article En | MEDLINE | ID: mdl-35398334

INTRODUCTION: Amphetamine (AMPH) abuse results in neurobehavioral alterations related to the reward circuit. The hippocampus plays a role in cognition, reward, and drug addiction. There are no pharmacological approaches to prevent AMPH relapse. Physical exercise has been studied as a non-pharmacological promising influence to attenuate reward symptoms related to addictive drugs. OBJECTIVE: This study aimed to compare the effects of non-weight-loaded and weight-loaded physical exercise on behavioral (relapse, memory and anxiety) and hippocampal molecular parameters associated with AMPH addiction in Wistar rats. METHODS: Male rats were subjected to the AMPH-Conditioned Place Preference (CPP) paradigm. After 8-conditioning days, they were subjected to swimming physical exercise protocol (without or with weight-load). Behavioral evaluations were performed to assess the influence of both exercise protocols in addiction parameters, including relapse after AMPH reconditioning, working memory, locomotor activity, and anxiety-like symptoms. Subsequently, protein levels of Brain-Derived Neurotrophic Factor (BDNF) and pro-BDNF ex-vivo assays were carried out in samples of the hippocampus of the animals. RESULTS: AMPH relapse and anxiety-like behaviors were reduced only in rats subjected to non-weight-loaded exercise. Hippocampal BDNF and pro-BDNF immunoreactivity were increased in non-weight-loaded exercise rats. Behavioral and molecular analyses were not modified in rats subjected to weight-loaded exercise. CONCLUSION: These findings demonstrate that non-weight-loaded exercise was more effective against relapse and anxiety-like behavior induced by AMPH. Non-weight-loaded exercise upregulated the hippocampal immunocontent levels in rats.


Amphetamine-Related Disorders , Brain-Derived Neurotrophic Factor , Amphetamine/pharmacology , Amphetamine-Related Disorders/metabolism , Animals , Brain-Derived Neurotrophic Factor/metabolism , Hippocampus/metabolism , Male , Rats , Rats, Wistar , Recurrence
13.
Behav Brain Res ; 423: 113770, 2022 04 09.
Article En | MEDLINE | ID: mdl-35085702

Methamphetamine-induced behavioral effects are mediated by several neurotransmitters that act via the G-protein coupled receptors (GPCRs). The functioning of GPCRs are negatively regulated by regulators of G-protein signaling (RGS) proteins. The goal of this study was to assess the role of two specific RGS proteins namely the RGS2 and the RGS4 proteins in methamphetamine-induced behaviors. The effects of methamphetamine (1 mg/kg; i.p.) on conditioned place preference (CPP) and locomotor activity were assessed in genetically modified male and female mice lacking either RGS2 or RGS4 and their wildtype littermates to achieve the above goal. Locomotor activity after methamphetamine administration was assessed in both methamphetamine-naïve and -experienced mice. Methamphetamine-induced CPP at the tested dose was blocked in male, but not female, mice lacking RGS4 compared to respective controls. Interestingly, methamphetamine-induced increase in locomotor activity at the tested dose was observed in methamphetamine-experienced, but not in the methamphetamine-naïve, male mice lacking RGS4. However, methamphetamine-induced increase in locomotor activity at the tested dose was blocked in both methamphetamine-naïve and -experienced female mice lacking RGS4. Interestingly, methamphetamine-induced rewarding effects and methamphetamine-induced increase in locomotor activity at the tested dose were observed in mice lacking RGS2, irrespective of sex and/or history of methamphetamine exposure. Together, the data suggest that RGS4 plays a role in methamphetamine-induced behaviors and could serve as a potential target for medications intended to treat the acute effects of methamphetamine.


Amphetamine-Related Disorders/metabolism , Amphetamine-Related Disorders/physiopathology , Behavior, Animal/drug effects , Central Nervous System Stimulants/pharmacology , Locomotion/drug effects , Methamphetamine/pharmacology , RGS Proteins/deficiency , Animals , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
14.
Arch Pharm Res ; 44(9-10): 890-901, 2021 Oct.
Article En | MEDLINE | ID: mdl-34741727

Drug use disorder, a chronic and relapsing mental disorder, is primarily diagnosed via self-reports of drug-seeking behavioral and psychological conditions, accompanied by psychiatric assessment. Therefore, the identification of peripheral biomarkers that reflect pathological changes caused by such disorders is essential for improving treatment monitoring. Hair possesses great potential as a metabolomic sample for monitoring chronic diseases. This study aimed to investigate metabolic alterations in hair to elucidate a suitable treatment modality for methamphetamine (MA) use disorder. Consequently, both targeted and untargeted metabolomics analyses were performed via mass spectrometry on hair samples obtained from current and former patients with MA use disorder. Healthy subjects (HS), current (CP), and former (FP) patients with this disorder were selected based on psychiatric diagnosis and screening the concentrations of MA in hair. The drug abuse screening questionnaire scores did not differentiate between CP and FP. Moreover, according to both targeted and untargeted metabolomics, clustering was not observed among all three groups. Nevertheless, a model of partial least squares-discriminant analysis was established between HS and CP based on seven metabolites derived from the targeted metabolomics results. Thus, this study demonstrates the promising potential of hair metabolomes for monitoring recovery from drug use disorders in clinical practice.


Amphetamine-Related Disorders/diagnosis , Hair/metabolism , Metabolome , Metabolomics , Methamphetamine , Spectrometry, Mass, Electrospray Ionization , Substance Abuse Detection , Tandem Mass Spectrometry , Adult , Amphetamine-Related Disorders/metabolism , Amphetamine-Related Disorders/rehabilitation , Humans , Male , Middle Aged , Predictive Value of Tests
15.
Int J Mol Sci ; 22(15)2021 Jul 30.
Article En | MEDLINE | ID: mdl-34360984

Dopamine D1 receptor (D1R) function is regulated by membrane/lipid raft-resident protein caveolin-1 (Cav1). We examined whether altered expression of Cav1 in the dorsal striatum would affect self-administration of methamphetamine, an indirect agonist at the D1Rs. A lentiviral construct expressing Cav1 (LV-Cav1) or containing a short hairpin RNA against Cav1 (LV-shCav1) was used to overexpress or knock down Cav1 expression respectively, in the dorsal striatum. Under a fixed-ratio schedule, LV-Cav1 enhanced and LV-shCav1 reduced responding for methamphetamine in an extended access paradigm compared to LV-GFP controls. LV-Cav1 and LV-shCav1 also produced an upward and downward shift in a dose-response paradigm, generating a drug vulnerable/resistant phenotype. LV-Cav1 and LV-shCav1 did not alter responding for sucrose. Under a progressive-ratio schedule, LV-shCav1 generally reduced positive-reinforcing effects of methamphetamine and sucrose as seen by reduced breakpoints. Western blotting confirmed enhanced Cav1 expression in LV-Cav1 rats and reduced Cav1 expression in LV-shCav1 rats. Electrophysiological findings in LV-GFP rats demonstrated an absence of high-frequency stimulation (HFS)-induced long-term potentiation (LTP) in the dorsal striatum after extended access methamphetamine self-administration, indicating methamphetamine-induced occlusion of plasticity. LV-Cav1 prevented methamphetamine-induced plasticity via increasing phosphorylation of calcium calmodulin kinase II, suggesting a mechanism for addiction vulnerability. LV-shCav1 produced a marked deficit in the ability of HFS to produce LTP and, therefore, extended access methamphetamine was unable to alter striatal plasticity, indicating a mechanism for resistance to addiction-like behavior. Our results demonstrate that Cav1 expression and knockdown driven striatal plasticity assist with modulating addiction to drug and nondrug rewards, and inspire new strategies to reduce psychostimulant addiction.


Amphetamine-Related Disorders/metabolism , Caveolin 1/metabolism , Corpus Striatum/metabolism , Long-Term Potentiation , Amphetamine-Related Disorders/genetics , Amphetamine-Related Disorders/physiopathology , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Caveolin 1/genetics , Corpus Striatum/drug effects , Male , Methamphetamine/toxicity , Rats , Rats, Long-Evans , Reward
16.
Neuropharmacology ; 196: 108692, 2021 09 15.
Article En | MEDLINE | ID: mdl-34217776

Group II metabotropic glutamate receptors (mGlu2 and mGlu3 receptors) shape mechanisms of methamphetamine addiction, but the individual role played by the two subtypes is unclear. We measured methamphetamine-induced conditioned place preference (CPP) and motor responses to single or repeated injections of methamphetamine in wild-type, mGlu2-/-, and mGlu3-/-mice. Only mGlu3-/-mice showed methamphetamine preference in the CPP test. Motor response to the first methamphetamine injection was dramatically reduced in mGlu2-/-mice, unless these mice were treated with the mGlu5 receptor antagonist, MTEP. In contrast, methamphetamine-induced sensitization was increased in mGlu3-/-mice compared to wild-type mice. Only mGlu3-/-mice sensitized to methamphetamine showed increases in phospho-ERK1/2 levels in the nucleus accumbens (NAc) and free radical formation in the NAc and medial prefrontal cortex. These changes were not detected in mGlu2-/-mice. We also measured a series of biochemical parameters related to the mechanism of action of methamphetamine in naïve mice to disclose the nature of the differential behavioural responses of the three genotypes. We found a reduced expression and activity of dopamine transporter (DAT) and vesicular monoamine transporter-2 in the NAc and striatum of mGlu2-/-and mGlu3-/-mice, whereas expression of the DAT adaptor, syntaxin 1A, was selectively increased in the striatum of mGlu3-/-mice. Methamphetamine-stimulated dopamine release in striatal slices was largely reduced in mGlu2-/-, but not in mGlu3-/-, mice. These findings suggest that drugs that selectively enhance mGlu3 receptor activity or negatively modulate mGlu2 receptors might be beneficial in the treatment of methamphetamine addiction and associated brain damage.


Amphetamine-Related Disorders/metabolism , Behavior, Animal/drug effects , Central Nervous System Stimulants/pharmacology , Conditioning, Classical/drug effects , Methamphetamine/pharmacology , Receptors, Metabotropic Glutamate/genetics , Amphetamine-Related Disorders/physiopathology , Animals , Behavior, Animal/physiology , Disease Models, Animal , Dopamine/metabolism , Dopamine Plasma Membrane Transport Proteins/metabolism , Male , Mice , Mice, Knockout , Mitogen-Activated Protein Kinase 1/drug effects , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/drug effects , Mitogen-Activated Protein Kinase 3/metabolism , Neostriatum/drug effects , Neostriatum/metabolism , Phosphorylation , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Pyridines/pharmacology , Receptor, Metabotropic Glutamate 5/antagonists & inhibitors , Receptors, Metabotropic Glutamate/metabolism , Syntaxin 1/drug effects , Syntaxin 1/metabolism , Thiazoles/pharmacology , Vesicular Monoamine Transport Proteins/metabolism
17.
Exp Neurol ; 343: 113793, 2021 09.
Article En | MEDLINE | ID: mdl-34166684

INTRODUCTION: Methamphetamine users are typically young adults, placing them at risk for significant drug-related harms. Neurological harms include stroke and Parkinson's disease, both of which may develop prematurely in the context of methamphetamine use. MATERIAL AND METHODS: We conducted a narrative review examining the evidence first, for stroke under 45 years and second, early onset of Parkinson's disease (PD) and parkinsonism related to methamphetamine use. We summarise epidemiological factors and common clinical features, before examining in detail the underlying pathology and causal mechanisms. RESULTS AND DISCUSSION: Methamphetamine use among young people (<45 years) is associated with heightened risk for haemorrhagic stroke. Compared to age-matched all-cause fatal stroke, haemorrhage secondary to aneurysmal rupture is more common among young people with methamphetamine-related stroke and is associated with significantly poorer prognosis. Aetiology is related primarily to both acute and chronic hypertension associated with methamphetamine's sympathomimetic action. Evidence from a variety of sources supports a link between methamphetamine use and increased risk for the development of PD and parkinsonism, and with their early onset in a subset of individuals. Despite this, direct evidence of degeneration of dopaminergic neurons in methamphetamine users has not been demonstrated to date. CONCLUSIONS: Stroke and Parkinson's Disease/parkinsonism are neurological harms observed prematurely in methamphetamine users.


Amphetamine-Related Disorders , Central Nervous System Stimulants/adverse effects , Methamphetamine/adverse effects , Parkinson Disease, Secondary/chemically induced , Stroke/chemically induced , Amphetamine-Related Disorders/metabolism , Amphetamine-Related Disorders/pathology , Humans , Parkinson Disease, Secondary/metabolism , Parkinson Disease, Secondary/pathology , Risk Factors , Stroke/metabolism , Stroke/pathology
18.
Eur J Pharmacol ; 897: 173935, 2021 Apr 15.
Article En | MEDLINE | ID: mdl-33577836

Stimulant-induced neurochemical changes may occur at different times for different brain regions or neurotransmitter systems. This study sought to examine the behavioral and neurochemical effects of extended access to α-pyrrolidinopentiophenone (α-PVP) and 4-methylmethcathinone (4MMC). Male and female Sprague-Dawley rats were trained to self-administer α-PVP (0.1 mg/kg/infusion) or 4MMC (0.5 mg/kg/infusion) through autoshaping, and then self-administered for 21 days during 1 h (short access; ShA) or 6 h (long access; LgA) sessions. Separate rats were assigned to a naïve control group. Amygdala, hippocampus, hypothalamus, prefrontal cortex (PFC), striatum, and thalamus were extracted, and tissue was analyzed with electrochemical detection and liquid chromatography mass spectrometry. Rats acquired self-administration of α-PVP and 4MMC, and LgA rats showed more escalation of self-administration than ShA rats. Synthetic cathinone administration produced several effects on neurotransmitters. LgA self-administration of α-PVP increased 5-HIAA levels in all brain regions, compared to control. In contrast, both LgA and ShA 4MMC self-administration decreased 5-HT and 5-HIAA levels in most brain regions. LgA exposure to both synthetic cathinones increased DOPAC levels in hypothalamus and striatum, and increased HVA levels in striatum compared to control. LgA self-administration of either synthetic cathinone produced region-specific increases in NE levels, whereas ShA self-administration lowered NE levels in select locations compared to control. These alterations in neurotransmitter levels indicate that synthetic cathinone use may produce differential neurochemical changes during the transition from use to abuse, and that 21 days of self-administration only models the beginning stages of dysregulated drug intake.


Amphetamine-Related Disorders/metabolism , Brain/drug effects , Central Nervous System Stimulants/administration & dosage , Methamphetamine/analogs & derivatives , Neurotransmitter Agents/metabolism , Pentanones/administration & dosage , Pyrrolidines/administration & dosage , 3,4-Dihydroxyphenylacetic Acid/metabolism , Amphetamine-Related Disorders/physiopathology , Animals , Brain/metabolism , Brain/physiopathology , Female , Hydroxyindoleacetic Acid/metabolism , Male , Methamphetamine/administration & dosage , Norepinephrine/metabolism , Rats, Sprague-Dawley , Self Administration , Serotonin/metabolism , Sex Factors , Time Factors
19.
Sci Rep ; 11(1): 1422, 2021 01 14.
Article En | MEDLINE | ID: mdl-33446840

Methamphetamine (METH) is a highly addictive psychostimulant that causes long-lasting effects in the brain and increases the risk of developing neurodegenerative diseases. The cellular and molecular effects of METH in the brain are functionally linked to alterations in glutamate levels. Despite the well-documented effects of METH on glutamate neurotransmission, the underlying mechanism by which METH alters glutamate levels is not clearly understood. In this study, we report an essential role of proline biosynthesis in maintaining METH-induced glutamate homeostasis. We observed that acute METH exposure resulted in the induction of proline biosynthetic enzymes in both undifferentiated and differentiated neuronal cells. Proline level was also increased in these cells after METH exposure. Surprisingly, METH treatment did not increase glutamate levels nor caused neuronal excitotoxicity. However, METH exposure resulted in a significant upregulation of pyrroline-5-carboxylate synthase (P5CS), the key enzyme that catalyzes synthesis of proline from glutamate. Interestingly, depletion of P5CS by CRISPR/Cas9 resulted in a significant increase in glutamate levels upon METH exposure. METH exposure also increased glutamate levels in P5CS-deficient proline-auxotropic cells. Conversely, restoration of P5CS expression in P5CS-deficient cells abrogated the effect of METH on glutamate levels. Consistent with these findings, P5CS expression was significantly enhanced in the cortical brain region of mice administered with METH and in the slices of cortical brain tissues treated with METH. Collectively, these results uncover a key role of P5CS for the molecular effects of METH and highlight that excess glutamate can be sequestered for proline biosynthesis as a protective mechanism to maintain glutamate homeostasis during drug exposure.


Amphetamine-Related Disorders/metabolism , Cerebral Cortex/metabolism , Glutamic Acid/metabolism , Homeostasis/drug effects , Methamphetamine/toxicity , Proline/biosynthesis , Acute Disease , Aldehyde Dehydrogenase/metabolism , Animals , CHO Cells , Cricetulus , Humans , Male , Mice , Neurons/metabolism
20.
Pharmacol Biochem Behav ; 201: 173089, 2021 02.
Article En | MEDLINE | ID: mdl-33422599

Methamphetamine (meth) dependence is often characterized by persistent and chronic relapse (i.e., return to drug use). Previous work suggests females may be at greater risk to relapse. In this study, we extended this limited evidence and identified sex-dependent neural substrates related to meth-triggered reinstatement. Male and female Sprague-Dawley rats were implanted with indwelling jugular catheters. Half of the rats were then trained to self-administer meth (0.05 mg/kg/inf); the other half self-administered saline during 21 daily sessions (2 h). Rats were then given 12 extinction sessions. Twenty-four hours after the last extinction session, rats received reinstatement testing. Half of the rats received a meth-prime (0.3 mg/kg, IP) injection and the remaining rats received a saline injection. This design resulted in 4 separate groups for each sex, allowing for careful investigation of brain regions related to meth-triggered reinstatement. Brains were harvested following the reinstatement session and c-Fos immunoreactivity was measured in multiple brain regions. Meth triggered reinstatement in both sexes and this effect was more robust in females compared to males. Significant sex differences were detected. Females showed greater c-Fos immunoreactivity in the cingulate cortex area 1, lateral orbitofrontal cortex, prelimbic cortex, caudate-putamen, nucleus accumbens core and shell, and central nucleus of the amygdala following meth-primed reinstatement.


Amphetamine-Related Disorders/metabolism , Amphetamine-Related Disorders/psychology , Central Nervous System Stimulants/administration & dosage , Drug-Seeking Behavior/drug effects , Methamphetamine/administration & dosage , Proto-Oncogene Proteins c-fos/metabolism , Animals , Central Amygdaloid Nucleus/metabolism , Disease Models, Animal , Extinction, Psychological , Female , Gyrus Cinguli/metabolism , Immunohistochemistry/methods , Male , Nucleus Accumbens/metabolism , Prefrontal Cortex/metabolism , Rats , Rats, Sprague-Dawley , Recurrence , Self Administration , Sex Factors
...