Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.432
Filter
1.
Article in English | MEDLINE | ID: mdl-38729234

ABSTRACT

Methamphetamine (METH) is a major health problem without effective pharmacological treatment. Cannabidiol (CBD), a component of the Cannabis sativa plant, is believed to have the potential to inhibit drug-related behavior. However, the neurobiological mechanisms responsible for the effects of CBD remain unclear. Several studies have proposed that the suppressing effects of CBD on drug-seeking behaviors could be through the modulation of the dopamine system. The hippocampus (HIP) D1-like dopamine receptor (D1R) is essential for forming and retrieving drug-associated memory. Therefore, the present study aimed to investigate the role of D1R in the hippocampal CA1 region on the effects of CBD on the extinction and reinstatement of METH-conditioned place preference (CPP). For this purpose, different groups of rats over a 10-day extinction period were administered different doses of intra-CA1 SCH23390 (0.25, 1, or 4 µg/0.5 µl, Saline) as a D1R antagonist before ICV injection of CBD (10 µg/5 µl, DMSO12%). In addition, a different set of animals received intra-CA1 SCH23390 (0.25, 1, or 4 µg/0.5 µl) before CBD injection (50 µg/5 µl) on the reinstatement day. The results revealed that the highest dose of SCH23390 (4 µg) significantly reduced the accelerating effects of CBD on the extinction of METH-CPP (P < 0.01). Furthermore, SCH23390 (1 and 4 µg) in the reinstatement phase notably reversed the preventive effects of CBD on the reinstatement of drug-seeking behavior (P < 0.05 and P < 0.001, respectively). In conclusion, the current study revealed that CBD made a shorter extinction period and suppressed METH reinstatement in part by interacting with D1-like dopamine receptors in the CA1 area of HIP.


Subject(s)
Benzazepines , Cannabidiol , Extinction, Psychological , Methamphetamine , Rats, Wistar , Receptors, Dopamine D1 , Animals , Methamphetamine/pharmacology , Cannabidiol/pharmacology , Extinction, Psychological/drug effects , Male , Receptors, Dopamine D1/antagonists & inhibitors , Benzazepines/pharmacology , Rats , Dose-Response Relationship, Drug , Drug-Seeking Behavior/drug effects , Hippocampus/drug effects , Hippocampus/metabolism , Dopamine Antagonists/pharmacology , CA1 Region, Hippocampal/drug effects
2.
CNS Neurosci Ther ; 30(5): e14737, 2024 05.
Article in English | MEDLINE | ID: mdl-38702929

ABSTRACT

AIMS: This study aims to investigate the pharmacological effects and the underlying mechanism of cannabidiol (CBD) on methamphetamine (METH)-induced relapse and behavioral sensitization in male mice. METHODS: The conditioned place preference (CPP) test with a biased paradigm and open-field test were used to assess the effects of CBD on METH-induced relapse and behavioral sensitization in male mice. RNA sequencing and bioinformatics analysis was employed to identify differential expressed (DE) circRNAs, miRNAs, and mRNAs in the nucleus accumbens (NAc) of mice, and the interaction among them was predicted using competing endogenous RNAs (ceRNAs) network analysis. RESULTS: Chronic administration of CBD (40 mg/kg) during the METH withdrawal phase alleviated METH (2 mg/kg)-induced CPP reinstatement and behavioral sensitization in mice, as well as mood and cognitive impairments following behavioral sensitization. Furthermore, 42 DEcircRNAs, 11 DEmiRNAs, and 40 DEmRNAs were identified in the NAc of mice. The circMeis2-miR-183-5p-Kcnj5 network in the NAc of mice is involved in the effects of CBD on METH-induced CPP reinstatement and behavioral sensitization. CONCLUSIONS: This study constructed the ceRNAs network for the first time, revealing the potential mechanism of CBD in treating METH-induced CPP reinstatement and behavioral sensitization, thus advancing the application of CBD in METH use disorders.


Subject(s)
Cannabidiol , Methamphetamine , Mice, Inbred C57BL , MicroRNAs , RNA, Circular , RNA, Messenger , Animals , Cannabidiol/pharmacology , Male , Methamphetamine/pharmacology , MicroRNAs/genetics , MicroRNAs/metabolism , Mice , RNA, Circular/genetics , RNA, Messenger/metabolism , Recurrence , Central Nervous System Stimulants/pharmacology , Nucleus Accumbens/drug effects , Nucleus Accumbens/metabolism , Gene Regulatory Networks/drug effects
3.
Eur J Pharmacol ; 972: 176559, 2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38588768

ABSTRACT

This study aimed to assess whether brief recall of methamphetamine (MA) memory, when combined with ketamine (KE) treatment, may prevent stress-primed MA memory reinstatement. Combining 3-min recall and KE facilitated MA memory extinction and resistance to subsequent stress-primed reinstatement. Such combination also produced glutamate metabotropic receptor 5 (mGluR5) upregulation in animals' medial prefrontal cortex (mPFC) γ-amino-butyric acid (GABA) neuron. Accordingly, chemogenetic methods were employed to bi-directionally modulate mPFC GABA activity. Following brief recall and KE-produced MA memory extinction, intra-mPFC mDlx-Gi-coupled-human-muscarinic-receptor 4 (hM4Di)-infused mice receiving compound 21 (C21) treatment showed eminent stress-primed reinstatement, while their GABA mGluR5 expression seemed to be unaltered. Intra-mPFC mDlx-Gq-coupled-human-muscarinic-receptor 3 (hM3Dq)-infused mice undergoing C21 treatment displayed MA memory extinction and resistance to stress-provoked reinstatement. These results suggest that combining a brief recall and KE treatment and exciting mPFC GABA neuron may facilitate MA memory extinction and resistance to stress-primed recall. mPFC GABA neuronal activity plays a role in mediating brief recall/KE-produced effects on curbing the stress-provoked MA seeking.


Subject(s)
Extinction, Psychological , Ketamine , Mental Recall , Methamphetamine , Prefrontal Cortex , Receptor, Metabotropic Glutamate 5 , Stress, Psychological , Animals , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Methamphetamine/pharmacology , Ketamine/pharmacology , Male , Mice , Mental Recall/drug effects , Stress, Psychological/drug therapy , Stress, Psychological/psychology , Receptor, Metabotropic Glutamate 5/metabolism , Extinction, Psychological/drug effects , Memory/drug effects , gamma-Aminobutyric Acid/metabolism , Mice, Inbred C57BL
4.
Neurochem Int ; 176: 105743, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38641026

ABSTRACT

Neonatal brain inflammation produced by intraperitoneal (i.p.) injection of lipopolysaccharide (LPS) results in long-lasting brain dopaminergic injury and motor disturbances in adult rats. The goal of the present work is to investigate the effect of neonatal systemic LPS exposure (1 or 2 mg/kg, i.p. injection in postnatal day 5, P5, male rats)-induced dopaminergic injury to examine methamphetamine (METH)-induced behavioral sensitization as an indicator of drug addiction. On P70, subjects underwent a treatment schedule of 5 once daily subcutaneous (s.c.) administrations of METH (0.5 mg/kg) (P70-P74) to induce behavioral sensitization. Ninety-six hours following the 5th treatment of METH (P78), the rats received one dose of 0.5 mg/kg METH (s.c.) to reintroduce behavioral sensitization. Hyperlocomotion is a critical index caused by drug abuse, and METH administration has been shown to produce remarkable locomotor-enhancing effects. Therefore, a random forest model was used as the detector to extract the feature interaction patterns among the collected high-dimensional locomotor data. Our approaches identified neonatal systemic LPS exposure dose and METH-treated dates as features significantly associated with METH-induced behavioral sensitization, reinstated behavioral sensitization, and perinatal inflammation in this experimental model of drug addiction. Overall, the analysis suggests that the implementation of machine learning strategies is sensitive enough to detect interaction patterns in locomotor activity. Neonatal LPS exposure also enhanced METH-induced reduction of dopamine transporter expression and [3H]dopamine uptake, reduced mitochondrial complex I activity, and elevated interleukin-1ß and cyclooxygenase-2 concentrations in the P78 rat striatum. These results indicate that neonatal systemic LPS exposure produces a persistent dopaminergic lesion leading to a long-lasting change in the brain reward system as indicated by the enhanced METH-induced behavioral sensitization and reinstated behavioral sensitization later in life. These findings indicate that early-life brain inflammation may enhance susceptibility to drug addiction development later in life, which provides new insights for developing potential therapeutic treatments for drug addiction.


Subject(s)
Animals, Newborn , Lipopolysaccharides , Machine Learning , Methamphetamine , Animals , Methamphetamine/pharmacology , Methamphetamine/toxicity , Rats , Male , Lipopolysaccharides/toxicity , Behavior, Animal/drug effects , Central Nervous System Stimulants/pharmacology , Encephalitis/chemically induced , Encephalitis/metabolism , Neuroinflammatory Diseases/drug therapy , Neuroinflammatory Diseases/chemically induced , Neuroinflammatory Diseases/metabolism , Locomotion/drug effects , Locomotion/physiology , Female , Rats, Sprague-Dawley , Motor Activity/drug effects
5.
Drug Alcohol Depend ; 259: 111285, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38636173

ABSTRACT

BACKGROUND: The orexin system has been implicated as a mechanism underlying insomnia and methamphetamine-induced sleep disruptions, with a potential role for OX2 receptors in the sleep-modulating effects of orexin. The aim of the present study was to investigate the extent to which orexin receptors mediate the effects of acute methamphetamine administration on actigraphy-based sleep in female rhesus monkeys. METHODS: Actigraphy-based sleep measures were obtained in female rhesus monkeys (n=5) under baseline and acute test conditions. First, morning (10h) i.m. injections of methamphetamine (0.03 - 0.56mg/kg) were administered to determine the effects of methamphetamine alone. Then, saline or methamphetamine (0.3mg/kg) were administered at 10h, and evening (17h30) oral treatments with vehicle, the non-selective orexin receptor antagonist suvorexant (1 - 10mg/kg, p.o.), or the OX2-selective orexin receptor antagonist MK-1064 (1 - 10mg/kg, p.o.) were given. The ability of suvorexant and MK-1064 (10mg/kg, p.o.) to improve actigraphy-based sleep was also assessed in a group of female monkeys quantitatively identified with "short-duration sleep" (n=4). RESULTS: Methamphetamine dose-dependently disrupted actigraphy-based sleep parameters. Treatment with either suvorexant or MK-1064 dose-dependently improved actigraphy-based sleep in monkeys treated with methamphetamine. Additionally, both suvorexant and MK-1064 promoted actigraphy-based sleep in a group of monkeys with baseline short actigraphy-based sleep. CONCLUSIONS: These findings suggest that orexin-mediated mechanisms play a role in the effects of methamphetamine on actigraphy-based sleep in female monkeys. Targeting the orexin system, in particular OX2 receptors, could be an effective option for treating sleep disruptions observed in individuals with methamphetamine use disorder.


Subject(s)
Actigraphy , Macaca mulatta , Methamphetamine , Orexin Receptor Antagonists , Orexin Receptors , Sleep , Animals , Female , Methamphetamine/pharmacology , Orexin Receptors/metabolism , Orexin Receptors/drug effects , Sleep/drug effects , Sleep/physiology , Orexin Receptor Antagonists/pharmacology , Triazoles/pharmacology , Azepines/pharmacology , Central Nervous System Stimulants/pharmacology , Dose-Response Relationship, Drug
6.
Neurosci Lett ; 827: 137740, 2024 Mar 28.
Article in English | MEDLINE | ID: mdl-38521402

ABSTRACT

New psychoactive substances (NPS) are typically synthesized in clandestine laboratories in an attempt to chemically modify already federally regulated drugs in an effort to circumvent the law. Drugs derived from a phenethylamine pharmacophore, such as 4-chloroamphetamine and 3,4-methylenedioxymethamphetamine (MDMA), reliably induce thermogenesis and serotonergic deficits in the striatum and hippocampus of rodents. 4-methylamphetamine (4-MA), a relative newcomer to the NPS scene, was originally investigated in the mid-1900 s as a potential anorexigenic agent. With its phenethylamine pharmacophore, 4-MA was hypothesized to produce similar toxicological alterations as its chemical analogs. In the present study, three doses (1.0, 2.5, and 5.0 mg/kg, ip.) of 4-MA were administered to rats twice daily for two days. Core temperature data were calculated and analyzed as temperature area under the curve (TAUC). On the second day of dosing, a hypothermic response to 4-MA (2.5 and 5.0 mg/kg) was noted between 0.5 and 2.0 h post-treatment. Only the highest dose of 4-MA decreased body weight on the second day of treatment and maintained this reduction in weight for seven days after treatment ceased. None of the doses of 4-MA evaluated significantly altered serotonin levels in the hippocampus or striatum seven days after final treatment. The present findings demonstrate that the 4-methyl substitution to amphetamine generates a pharmacological and toxicological profile that differs from other similar phenethylamine analogs.


Subject(s)
Amphetamines , Designer Drugs , Methamphetamine , N-Methyl-3,4-methylenedioxyamphetamine , Rats , Animals , Methamphetamine/pharmacology , Serotonin/pharmacology , Designer Drugs/pharmacology , Temperature , N-Methyl-3,4-methylenedioxyamphetamine/pharmacology , Amphetamine/pharmacology , Hippocampus , Serotonin Agents/pharmacology , Serotonin Agents/analysis
7.
Neuroimage Clin ; 41: 103579, 2024.
Article in English | MEDLINE | ID: mdl-38447413

ABSTRACT

In stimulant use and addiction, conflict control processes are crucial for regulating substance use and sustaining abstinence, which can be particularly challenging in social-affective situations. Users of methamphetamine (METH, "Ice") and 3,4-methylenedioxymethamphetamine (MDMA, "Ecstasy") both experience impulse control deficits, but display different social-affective and addictive profiles. We thus aimed to compare the effects of chronic use of the substituted amphetamines METH and MDMA on conflict control processes in different social-affective contexts (i.e., anger and happiness) and investigate their underlying neurophysiological mechanisms. For this purpose, chronic but recently abstinent users of METH (n = 38) and MDMA (n = 42), as well as amphetamine-naïve healthy controls (n = 83) performed an emotional face-word Stroop paradigm, while event-related potentials (ERPs) were recorded. Instead of substance-specific differences, both MDMA and METH users showed smaller behavioral effects of cognitive-emotional conflict processing (independently of emotional valence) and selective deficits in emotional processing of anger content. Both effects were underpinned by stronger P3 ERP modulations suggesting that users of substituted amphetamines employ altered stimulus-response mapping and decision-making. Given that these processes are modulated by noradrenaline and that both MDMA and METH use may be associated with noradrenergic dysfunctions, the noradrenaline system may underlie the observed substance-related similarities. Better understanding the functional relevance of this currently still under-researched neurotransmitter and its functional changes in chronic users of substituted amphetamines is thus an important avenue for future research.


Subject(s)
Methamphetamine , N-Methyl-3,4-methylenedioxyamphetamine , Substance-Related Disorders , Humans , N-Methyl-3,4-methylenedioxyamphetamine/pharmacology , Methamphetamine/pharmacology , Amphetamines , Norepinephrine
8.
J Psychiatr Res ; 172: 291-299, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38428165

ABSTRACT

Treatment of Methamphetamine (METH) use disorder has become a crucial public health issue. The orexin system manipulation has provided promising evidence to attenuate addictive-like behaviors. This study explored the role of the orexin 1 receptor and orexin 2 receptor (OX1R and OX2R) in the CA1 area of the hippocampal formation in the acquisition and expression of METH-induced place preference. Animals were subjected to bilateral administration of different dosages (1, 3, 10, and 30 nmol/0.5 µl DMSO per side) of a selective OX1R antagonist, SB334867, or selective OX2R antagonist, TCS OX2 29 into the CA1 area throughout the conditioning phase or once on the post-conditioning phase in separate control and experimental groups. Behavioral data revealed that both OX1R (10 nmol; P < 0.01 and 30 nmol; P < 0.001) and OX2R (10 nmol; P < 0.05 and 30 nmol; P < 0.001) antagonism during the conditioning phase could block the formation of METH place preference dose-dependently. In addition, intra-CA1 microinjection of SB334867 on the post-conditioning phase attenuated the expression of METH place preference in a dose-dependent manner (3 nmol; P < 0.05, 10 nmol; P < 0.01 and 30 nmol; P < 0.001) whereas intra-CA1 administration of TCS OX2 29 only at the highest dosage (30 nmol) declined the expression of METH place preference (P < 0.01). It was also indicated that the suppressive effects of orexin receptor blockade on the METH-seeking behavior in the CA1 area were anatomically specific to this area. These findings support the possibility of targeting the orexin system to develop novel and successful pharmacological options for the treatment of METH dependence.


Subject(s)
Hippocampus , Methamphetamine , Rats , Animals , Orexin Receptors/metabolism , Orexins/metabolism , Rats, Wistar , Hippocampus/metabolism , Methamphetamine/pharmacology
9.
Adv Pharmacol ; 99: 217-250, 2024.
Article in English | MEDLINE | ID: mdl-38467482

ABSTRACT

Methamphetamine (METH) regulation of the dopamine transporter (DAT) and central nervous system (CNS) dopamine transmission have been extensively studied. However, our understanding of how METH influences neuroimmune communication and innate and adaptive immunity is still developing. Recent studies have shed light on the bidirectional communication between the CNS and the peripheral immune system. They have established a link between CNS dopamine levels, dopamine neuronal activity, and peripheral immunity. Akin to dopamine neurons in the CNS, a majority of peripheral immune cells also express DAT, implying that in addition to their effect in the CNS, DAT ligands such as methamphetamine may have a role in modulating peripheral immunity. For example, by directly influencing DAT-expressing peripheral immune cells and thus peripheral immunity, METH can trigger a feed-forward cascade that impacts the bidirectional communication between the CNS and peripheral immune system. In this review, we aim to discuss the current understanding of how METH modulates both innate and adaptive immunity and identify areas where knowledge gaps exist. These gaps will then be considered in guiding future research directions.


Subject(s)
Immunity , Methamphetamine , Humans , Dopamine , Methamphetamine/pharmacology
10.
Behav Processes ; 217: 105023, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38522795

ABSTRACT

The interaction of voluntary exercise and housing density on a) anxiety-like behavior and b) the stimulant effects of methamphetamine in C57Bl/6 mice were evaluated. Upon arrival, mice were housed singly or in pairs, and permitted access to home-cage running wheels or not for 4 weeks. Testing for anxiety-like behavior occurred over the next 3 weeks, one test per week [Elevated-Plus Maze (EPM) → Hyponeophagia (HNP) task → Open-Field (OF) task]. The final, OF task involved an 8-hour session in which mice were permitted to explore the chamber (drug free) during Hours 1-3; given an injection (s.c.) of methamphetamine (1.0 mg/kg) after Hour 3; followed by continued behavioral sampling during Hours 4-8. Several tasks (HNP and OF, but not EPM) consistently showed voluntary exercise induced anxiety-like behavior. In addition, two measures (time in center and time resting in the perimeter) in the OF task revealed that exercise mice compared to controls were more responsive to the anxiogenic effects of methamphetamine. Although pair housing was anxiolytic, it did not ameliorate the anxiogenic effects of voluntary exercise. Taken together, these results, when viewed in tandem with previous studies that utilized a less anxious mouse strain (Swiss Webster), may suggest that voluntary exercise is anxiogenic in an anxiety-prone mouse strain such as C57Bl/6 and highlight the importance of considering mouse strain when evaluating the impact of environmental manipulations on anxiety-like behavior in animal models.


Subject(s)
Anxiety , Behavior, Animal , Housing, Animal , Methamphetamine , Mice, Inbred C57BL , Physical Conditioning, Animal , Animals , Mice , Male , Physical Conditioning, Animal/physiology , Physical Conditioning, Animal/psychology , Behavior, Animal/physiology , Behavior, Animal/drug effects , Methamphetamine/pharmacology , Central Nervous System Stimulants/pharmacology
11.
Addict Biol ; 29(3): e13382, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38488467

ABSTRACT

Methamphetamine (METH) is a highly addictive psycho-stimulant that induces addictive behaviour by stimulating increased dopamine release in the nucleus accumbens (NAc). The sarco/endoplasmic reticulum calcium ion transport ATPases (SERCA or ATP2A) is a calcium ion (Ca2+) pump in the endoplasmic reticulum (ER) membrane. SERCA2b is a SERCA subtype mainly distributed in the central nervous system. This study used conditioned place preference (CPP), a translational drug reward model, to observe the effects of SERCA and SERCA2b on METH-CPP in mice. Result suggested that the activity of SERCA was significantly decreased in NAc after METH-CPP. Intraperitoneal SERCA agonist CDN1163 injection or bilateral CDN1163 microinjection in the NAc inhibited METH-CPP formation. SERCA2b overexpression by the Adeno-associated virus can reduce the DA release of NAc and inhibit METH-CPP formation. Although microinjection of SERCA inhibitor thapsigargin in the bilateral NAc did not significantly aggravate METH-CPP, interference with SERCA2b expression in NAc by adeno-associated virus increased DA release and promoted METH-CPP formation. METH reduced the SERCA ability to transport Ca2+ into the ER in SHSY5Y cells in vitro, which was reversed by CDN1163. This study revealed that METH dysregulates intracellular calcium balance by downregulating SERCA2b function, increasing DA release in NAc and inducing METH-CPP formation. Drugs that target SERCA2b may have the potential to treat METH addiction.


Subject(s)
Benzamides , Central Nervous System Stimulants , Methamphetamine , Mice , Animals , Methamphetamine/pharmacology , Methamphetamine/metabolism , Nucleus Accumbens , Calcium/metabolism , Aminoquinolines/metabolism , Aminoquinolines/pharmacology , Central Nervous System Stimulants/pharmacology , Central Nervous System Stimulants/metabolism
12.
Pharmacol Biochem Behav ; 239: 173751, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38548247

ABSTRACT

Risky choice is associated with maladaptive behaviors, particularly substance use disorders. Current animal models of risky choice are often confounded by other constructs like behavioral flexibility and suboptimal choice. The purpose of the current experiment was to determine if the psychostimulant methamphetamine, a drug whose popularity has increased in recent years, increases risky choice in an equivalent expected value (EEV) task. In the EEV task, rats are given a choice between two reinforcer alternatives that differ in magnitude and probability of delivery, but have equivalent expected value. Forty-eight Sprague Dawley rats were tested in three versions of the EEV task. In the first version of the EEV task, both reinforcer magnitude and probability were adjusted across blocks of trials for both alternatives. In the second and the third versions of the EEV task, reinforcer magnitude was held constant across each block of trials (either 1 vs. 2 pellets or 4 vs. 5 pellets). We found that male rats preferred the "riskier" option, except when reinforcer magnitudes were held constant at 4 and 5 pellets across each block of trials. Methamphetamine (0.5 mg/kg) increased preference for the risky option in both males and females, but only when both reinforcer magnitude and probability were manipulated across blocks of trials for each alternative. The current results demonstrate that both magnitude of reinforcement and probability of reinforcement interact to influence risky choice. Overall, this study provides additional support for using reinforcers with expected value to measure risky choice.


Subject(s)
Central Nervous System Stimulants , Choice Behavior , Methamphetamine , Rats, Sprague-Dawley , Reinforcement, Psychology , Risk-Taking , Animals , Methamphetamine/pharmacology , Methamphetamine/administration & dosage , Male , Rats , Choice Behavior/drug effects , Female , Central Nervous System Stimulants/pharmacology , Central Nervous System Stimulants/administration & dosage , Conditioning, Operant/drug effects , Probability
13.
Electrophoresis ; 45(9-10): 958-969, 2024 May.
Article in English | MEDLINE | ID: mdl-38528319

ABSTRACT

Methamphetamine (MA) is a highly addictive mental stimulant, and MA abuse remains a significant public health problem worldwide, while effective treatment options are limited. Lycium barbarum polysaccharide (LBP), a major effective component extracted from Lycium barbarum, has potential health-promoting effects on the nervous system; however, its role in MA dependence remains unclear. In this study, the conditioned place preference (CPP) of MA addiction in adult male mice was established to detect changes in gut microbiota profiles after LBP treatment through 16S rRNA gene sequencing. Our results found that LBP administration could alleviate MA-induced CPP and hyperactivity. Interestingly, LBP improved MA-induced gut microbiota dysbiosis by increasing some beneficial autochthonous genus abundances, such as Allobaculum, Gordonibacter, and Ileibacterium. MA exposure induced the co-occurrence network of intestinal microbiota to become weaker and more unstable when compared with the control group, while LBP changed the above effects when compared with the MA group. Bacterial gene function prediction showed that amphetamine addiction, cocaine addiction, and short-chain fatty acid metabolism were enriched. These findings reveal that LBP might regulate MA-induced gut microbiota and behavior changes, which showed potential therapeutic applicability in treating MA addiction by regulating the gut microbiota.


Subject(s)
Amphetamine-Related Disorders , Drugs, Chinese Herbal , Dysbiosis , Gastrointestinal Microbiome , Methamphetamine , Animals , Gastrointestinal Microbiome/drug effects , Methamphetamine/pharmacology , Dysbiosis/chemically induced , Dysbiosis/microbiology , Male , Mice , Drugs, Chinese Herbal/pharmacology , Drugs, Chinese Herbal/administration & dosage , RNA, Ribosomal, 16S/analysis , Mice, Inbred C57BL , Bacteria/drug effects , Bacteria/classification , Bacteria/isolation & purification , Bacteria/genetics
14.
Addict Biol ; 29(2): e13370, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38353028

ABSTRACT

N-Isopropylbenzylamine (N-ipb), a chain isomer of methamphetamine (METH) with similar physical properties, has been used as a substitute for METH in seized drug samples. However, the abuse potential of N-ipb remains unclear. Therefore, this study aimed to evaluate the abuse potential of N-ipb in comparison to METH, by using conditioned place preference (CPP), locomotor sensitization and intravenous self-administration tests. The results showed that N-ipb at a dose of 3 mg·kg-1 significantly induced CPP in mice, which was comparable to the effect of METH at 1 mg·kg-1 . Either acute or repeated N-ipb injections (1 or 3 mg·kg-1 ) failed to raise the locomotor activity. However, acute treatment with 10 mg·kg-1 N-ipb elevated the locomotor activity compared with saline, while chronic injection of 10 mg·kg-1 N-ipb induced a delayed and attenuated sensitization compared with 1 mg·kg-1 METH. Rats could acquire N-ipb self-administration at a dose of 1 mg·kg-1 ·infusion-1 , and a typical inverted U-shaped dose-response curve was obtained for N-ipb. The mean dose of N-ipb that maintained the maximum response was greater than that of METH, indicating that N-ipb is less potent for reinforcement than METH. In the economic behavioural analysis, comparison of essential values derived from the demand elasticity revealed that N-ipb is less efficacy as a reinforcer than METH. The present data demonstrate that N-ipb functions as a reinforcer and has a potential for abuse. However, the potency of psychomotor stimulation and the reinforcing effectiveness of N-ipb are lower than those of METH.


Subject(s)
Amines , Central Nervous System Stimulants , Methamphetamine , Mice , Rats , Animals , Central Nervous System Stimulants/pharmacology , Rodentia , Motor Activity , Methamphetamine/pharmacology
15.
J Neurosci ; 44(11)2024 Mar 13.
Article in English | MEDLINE | ID: mdl-38331582

ABSTRACT

Cerebellum has been implicated in drug addiction; however, its underlying cellular populations and neuronal circuitry remain largely unknown. In the current study, we identified a neural pathway from tyrosine hydroxylase (TH)-positive Purkinje cells (PCTH+) in cerebellar lobule VI to calcium/calmodulin-dependent protein kinase II (CaMKII)-positive glutamatergic neurons in the medial cerebellar nucleus (MedCaMKII), forming the lobule VI PCTH+-MedCaMKII pathway in male mice. In naive male mice, inhibition of PCTH+ neurons activated Med neurons. During conditioned place preference (CPP) training, exposure to methamphetamine (METH) inhibited lobule VI PCTH+ neurons while excited MedCaMKII neurons in mice. Silencing MedCaMKII using a tetanus toxin light chain (tettox) suppressed the acquisition of METH CPP in mice but resulted in motor coordination deficits in naive mice. In contrast, activating lobule VI PCTH+ terminals within Med inhibited the activity of Med neurons and subsequently blocked the acquisition of METH CPP in mice without affecting motor coordination, locomotor activity, and sucrose reinforcements in naive mice. Our findings identified a novel lobule VI PCTH+-MedCaMKII pathway within the cerebellum and explored its role in mediating the acquisition of METH-preferred behaviors.


Subject(s)
Central Nervous System Stimulants , Methamphetamine , Animals , Male , Mice , Methamphetamine/pharmacology , Tyrosine 3-Monooxygenase/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Reinforcement, Psychology , Cerebellum/metabolism , Central Nervous System Stimulants/pharmacology
16.
Article in English | MEDLINE | ID: mdl-38365104

ABSTRACT

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.


Subject(s)
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
17.
Pharmacol Biochem Behav ; 238: 173735, 2024 May.
Article in English | MEDLINE | ID: mdl-38373600

ABSTRACT

RATIONALE: Fentanyl remains the primary cause of fatal overdoses, and its co-use with methamphetamine (METH) is a growing concern. We previously demonstrated that racemic METH can either enhance or mitigate opioid-induced respiratory depression (OIRD) dependent upon whether a low or high dose is administered. The optical isomers of METH, dextromethamphetamine (d-METH) and levomethamphetamine (l-METH), differ substantially in their selectivity and potency to activate various monoamine (MA) receptors, and these pharmacological differences may underlie the bidirectional effects of the racemate. Since it is unknown which of METH's MA receptor mechanisms mediate these respiratory effects, examination of METH's pharmacologically distinct enantiomers may provide insight into treatment targets for OIRD. METHODS: The two optical isomers of METH, d-METH and l-METH, were tested in adult male mice to determine their effects on basal and fentanyl-depressed respiratory frequency, tidal volume, and minute ventilation (MVb; i.e., respiratory frequency x tidal volume) using whole-body plethysmography. RESULTS: When tested at dose ranges of 1.0-10 mg/kg, d-METH elevated MVb and l-METH decreased basal MVb. A dose of 30 mg/kg l-METH increased basal MVb. Under fentanyl-depressed conditions, the bidirectional effects of racemic METH were observed with d-METH treatment while l-METH significantly exacerbated OIRD at 1.0 and 3.0 mg/kg. CONCLUSIONS: d-METH and l-METH differentially contribute to the bidirectional respiratory modulation observed by the racemate, with d-METH exhibiting predominantly stimulatory effects and l-METH exhibiting primarily depressant effects depending on dose.


Subject(s)
Fentanyl , Methamphetamine , Rats , Mice , Animals , Male , Fentanyl/pharmacology , Rats, Sprague-Dawley , Methamphetamine/pharmacology , Amphetamine/pharmacology , Respiration , Analgesics, Opioid/pharmacology
18.
J Neural Transm (Vienna) ; 131(4): 335-358, 2024 04.
Article in English | MEDLINE | ID: mdl-38367081

ABSTRACT

Methamphetamine (METH) produces a cytopathology, which is rather specific within catecholamine neurons both in vitro and ex vivo, in animal models and chronic METH abusers. This led some authors to postulate a sort of parallelism between METH cytopathology and cell damage in Parkinson's disease (PD). In fact, METH increases and aggregates alpha-syn proto-fibrils along with producing spreading of alpha-syn. Although alpha-syn is considered to be the major component of aggregates and inclusions developing within diseased catecholamine neurons including classic Lewy body (LB), at present, no study provided a quantitative assessment of this protein in situ, neither following METH nor in LB occurring in PD. Similarly, no study addressed the quantitative comparison between occurrence of alpha-syn and other key proteins and no investigation measured the protein compared with non-protein structure within catecholamine cytopathology. Therefore, the present study addresses these issues using an oversimplified model consisting of a catecholamine cell line where the novel approach of combined light and electron microscopy (CLEM) was used measuring the amount of alpha-syn, which is lower compared with p62 or poly-ubiquitin within pathological cell domains. The scenario provided by electron microscopy reveals unexpected findings, which are similar to those recently described in the pathology of PD featuring packing of autophagosome-like vesicles and key proteins shuttling autophagy substrates. Remarkably, small seed-like areas, densely packed with p62 molecules attached to poly-ubiquitin within wide vesicular domains occurred. The present data shed new light about quantitative morphometry of catecholamine cell damage in PD and within the addicted brain.


Subject(s)
Methamphetamine , Parkinson Disease , Animals , Methamphetamine/pharmacology , alpha-Synuclein/metabolism , Parkinson Disease/metabolism , Microscopy, Electron , Catecholamines , Ubiquitins
19.
Neurotherapeutics ; 21(3): e00328, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38355360

ABSTRACT

Methamphetamine (MA) use disorder poses significant challenges to both the affected individuals and society. Current non-drug therapies like transcranial direct-current stimulation and transcranial magnetic stimulation have limitations due to their invasive nature and limited reach to deeper brain areas. Transcranial focused ultrasound (FUS) is gaining attention as a noninvasive option with precise spatial targeting, able to affect deeper areas of the brain. This research focused on assessing the effectiveness of FUS in influencing the infralimbic cortex (IL) to prevent the recurrence of MA-seeking behavior, using the conditioned place preference (CPP) method in rats. The study involved twenty male Sprague-Dawley rats. Neuronal activation by FUS was first examined via electromyography (EMG). Rats received alternately with MA or saline, and confined to one of two distinctive compartments in a three compartment apparatus over a 4-day period. After CPP test, extinction, the first reinstatement, and extinction again, FUS was applied to IL prior to the second MA priming-induced reinstatement. Safety assessments were conducted through locomotor and histological function examinations. EMG data confirmed the effectiveness of FUS in activating neurons. Significant attenuation of reinstatement of MA CPP was found, along with successful targeting of the IL region, confirmed through acoustic field scanning, c-Fos immunohistochemistry, and Evans blue dye staining. No damage to brain tissue or impaired locomotor activity was observed. The results of the study indicate that applying FUS to the IL markedly reduced the recurrence of MA seeking behavior, without harming brain tissue or impairing motor skills. This suggests that FUS could be a promising method for treating MA use disorder, with the infralimbic cortex being an effective target for FUS in preventing MA relapse.


Subject(s)
Extinction, Psychological , Methamphetamine , Rats, Sprague-Dawley , Animals , Male , Methamphetamine/pharmacology , Rats , Extinction, Psychological/drug effects , Ultrasonic Therapy/methods , Central Nervous System Stimulants/pharmacology , Prefrontal Cortex/drug effects , Proto-Oncogene Proteins c-fos/metabolism
20.
Int J Mol Sci ; 25(2)2024 Jan 11.
Article in English | MEDLINE | ID: mdl-38256016

ABSTRACT

Methamphetamine (METH) abuse inflicts both physical and psychological harm. While our previous research has established the regulatory role of miR-29c-3p in behavior sensitization, the underlying mechanisms and target genes remain incompletely understood. In this study, we employed the isobaric tags for relative and absolute quantitation (iTRAQ) technique in conjunction with Ingenuity pathway analysis (IPA) to probe the putative molecular mechanisms of METH sensitization through miR-29c-3p inhibition. Through a microinjection of AAV-anti-miR-29c-3p into the nucleus accumbens (NAc) of mice, we observed the attenuation of METH-induced locomotor effects. Subsequent iTRAQ analysis identified 70 differentially expressed proteins (DEPs), with 22 up-regulated potential target proteins identified through miR-29c-3p target gene prediction and IPA analysis. Our focus extended to the number of neuronal branches, the excitatory synapse count, and locomotion-related pathways. Notably, GPR37, NPC1, and IREB2 emerged as potential target molecules for miR-29c-3p regulation, suggesting their involvement in the modulation of METH sensitization. Quantitative PCR confirmed the METH-induced aberrant expression of Gpr37, Npc1, and Ireb2 in the NAc of mice. Specifically, the over-expression of miR-29c-3p led to a significant reduction in the mRNA level of Gpr37, while the inhibition of miR-29c-3p resulted in a significant increase in the mRNA level of Gpr37, consistent with the regulatory principle of miRNAs modulating target gene expression. This suggests that miR-29c-3p potentially influences METH sensitization through its regulation of neuroplasticity. Our research indicates that miR-29c-3p plays a crucial role in regulating METH-induced sensitization, and it identified the potential molecular of miR-29c-3p in regulating METH-induced sensitization.


Subject(s)
Amphetamine-Related Disorders , Methamphetamine , MicroRNAs , Niemann-Pick Disease, Type C , Animals , Mice , Nucleus Accumbens , Methamphetamine/pharmacology , MicroRNAs/genetics , Neuronal Plasticity/genetics , RNA, Messenger , Receptors, G-Protein-Coupled
SELECTION OF CITATIONS
SEARCH DETAIL
...