Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 7.456
Filter
1.
Neurosci Biobehav Rev ; 164: 105841, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39098738

ABSTRACT

Stimulants represent the first line pharmacological treatment for attention-deficit/hyperactivity disorder (ADHD) and are among the most prescribed psychopharmacological treatments. Their mechanism of action at synaptic level has been extensively studied. However, it is less clear how their mechanism of action determines clinically observed benefits. To help bridge this gap, we provide a comprehensive review of stimulant effects, with an emphasis on nuclear medicine and magnetic resonance imaging (MRI) findings. There is evidence that stimulant-induced modulation of dopamine and norepinephrine neurotransmission optimizes engagement of task-related brain networks, increases perceived saliency, and reduces interference from the default mode network. An acute administration of stimulants may reduce brain alterations observed in untreated individuals in fronto-striato-parieto-cerebellar networks during tasks or at rest. Potential effects of prolonged treatment remain controversial. Overall, neuroimaging has fostered understanding on stimulant mechanism of action. However, studies are often limited by small samples, short or no follow-up, and methodological heterogeneity. Future studies should address age-related and longer-term effects, potential differences among stimulants, and predictors of treatment response.


Subject(s)
Attention Deficit Disorder with Hyperactivity , Brain , Central Nervous System Stimulants , Nerve Net , Humans , Attention Deficit Disorder with Hyperactivity/drug therapy , Attention Deficit Disorder with Hyperactivity/physiopathology , Attention Deficit Disorder with Hyperactivity/diagnostic imaging , Central Nervous System Stimulants/pharmacology , Brain/drug effects , Brain/diagnostic imaging , Brain/metabolism , Brain/physiopathology , Nerve Net/drug effects , Nerve Net/diagnostic imaging , Nerve Net/physiopathology , Neurons/drug effects
2.
Cereb Cortex ; 34(8)2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39098820

ABSTRACT

Drug addiction is a chronic and relapse brain disorder. Psychostimulants such as cocaine and amphetamine are highly addictive drugs. Abuse drugs target various brain areas in the nervous system. Recent studies have shown that the prefrontal cortex (PFC) plays a key role in regulating addictive behaviors. The PFC is made up of excitatory glutamatergic cells and gamma-aminobutyric acid (GABAergic) interneurons. Recently, studies showed that GABA level was related with psychostimulant addiction. In this review, we will introduce the role and mechanism of GABA and γ-aminobutyric acid receptors (GABARs) of the PFC in regulating drug addiction, especially in psychostimulant addiction.


Subject(s)
Central Nervous System Stimulants , Prefrontal Cortex , Substance-Related Disorders , gamma-Aminobutyric Acid , Prefrontal Cortex/metabolism , Prefrontal Cortex/drug effects , Humans , gamma-Aminobutyric Acid/metabolism , Animals , Substance-Related Disorders/metabolism , Central Nervous System Stimulants/pharmacology , Receptors, GABA/metabolism
3.
Prog Brain Res ; 288: 115-132, 2024.
Article in English | MEDLINE | ID: mdl-39168554

ABSTRACT

Caffeine, the main psychoactive component in coffee, has garnered significant attention for its potential impact on the most prevalent mental health conditions like anxiety and depression. This chapter comprehensively examines the neurobiological effects of caffeine, its influence on anxiety and depression, and relevant clinical studies. Caffeine exerts its psychostimulant effects primarily through antagonizing adenosine receptors, modulating neurotransmitter systems, and influencing intracellular calcium signaling in the brain. Caffeine exhibits dose-dependent effects. While moderate caffeine consumption is safe in healthy adults and may offer benefits for mental health, excessive intake is linked to adverse effects on neurological and psychiatric health and can aggravate symptoms, highlighting the importance of adjusting consumption patterns. High caffeine intake correlates with elevated anxiety levels, especially in individuals predisposed to anxiety disorders. However, the relationship between caffeine consumption and the risk of depression is intricate, with some studies suggesting a potential protective effect of moderate intake, while others find no significant association. Individual variations in caffeine metabolism, sensitivity, and genetic factors considerably impact responses to caffeine. The chapter also explores the therapeutic potential of caffeine as an adjunct treatment and outlines challenges and future research directions in elucidating caffeine's multifaceted role in mental health.


Subject(s)
Anxiety , Caffeine , Central Nervous System Stimulants , Coffee , Depression , Caffeine/pharmacology , Caffeine/administration & dosage , Humans , Depression/drug therapy , Anxiety/drug therapy , Central Nervous System Stimulants/pharmacology , Central Nervous System Stimulants/administration & dosage , Animals
4.
Prog Brain Res ; 288: 81-114, 2024.
Article in English | MEDLINE | ID: mdl-39168560

ABSTRACT

Consuming coffee, a widely enjoyed beverage with caffeine, can impact the central nervous system and disturb sleep if taken too close to bedtime. Caffeine impacts sleep by slowing the onset, blocking adenosine receptors, lowering deep sleep levels, disrupting sleep patterns, and lessening rapid eye movement sleep. Although coffee can help with alertness in the morning, it may disturb sleep in the evening, particularly for individuals who are sensitive to caffeine. To enhance the quality of sleep, reduce the consumption of caffeine in the afternoon and evening, refrain from drinking caffeine before going to bed, and choose decaffeinated drinks instead. Variables such as personal reactions, ability to handle caffeine, and engagement with other compounds also influence the impact of coffee on sleep. Keeping track of how much caffeine you consume and your sleeping habits can assist in recognizing any disturbances and making needed changes. Furthermore, taking into account variables such as metabolism, age, and the timing of coffee consumption can assist in lessening the effects of coffee on sleep. In general, paying attention to the amount of caffeine consumed from different sources and consuming it at the right times can assist in preserving healthy sleep patterns even while enjoying coffee.


Subject(s)
Caffeine , Coffee , Sleep , Humans , Caffeine/pharmacology , Caffeine/administration & dosage , Sleep/drug effects , Sleep/physiology , Central Nervous System Stimulants/pharmacology
5.
Prog Brain Res ; 289: 181-191, 2024.
Article in English | MEDLINE | ID: mdl-39168580

ABSTRACT

The physiological structure and functioning of the brain are determined by activity-dependent processes and affected by "synapse plasticity." Because chemical transmitters target and regulate synapses, exogenous chemical stimulants and transmitters can alter their physiological functions by interacting with synaptic surface receptors or chemical modulators. Caffeine, a commonly used pharmacologic substance, can target and alter synapses. It impact various biological, chemical, and metabolic processes related to synaptic function. This chapter investigates how caffeine affects fluctuations in structure and function in the hippocampus formation and neocortical structure, regions known for their high synaptic plasticity profile. Specifically, caffeine modulates various synaptic receptors and channel activities by mobilizing intracellular calcium, inhibiting phosphodiesterase, and blocking adenosine and GABA cellular receptors. These caffeine-induced pathways and functions allow neurons to generate plastic modulations in synaptic actions such as efficient and morphological transmission. Moreover, at a network level, caffeine can stimulate neural oscillators in the cortex, resulting in repetitive signals that strengthen long-range communication between cortical areas reliant on N-methyl-d-aspartate receptors. This suggests that caffeine could facilitate the reorganization of cortical network functions through its effects on synaptic mobilization.


Subject(s)
Caffeine , Neuronal Plasticity , Synapses , Neuronal Plasticity/physiology , Neuronal Plasticity/drug effects , Caffeine/pharmacology , Animals , Humans , Synapses/drug effects , Synapses/physiology , Coffee , Central Nervous System Stimulants/pharmacology , Synaptic Transmission/drug effects , Synaptic Transmission/physiology
6.
J Psychiatry Neurosci ; 49(4): E242-E251, 2024.
Article in English | MEDLINE | ID: mdl-39122408

ABSTRACT

BACKGROUND: Emotional dysregulation affects up to two-thirds of adult patients with attention-deficit/hyperactivity disorder (ADHD) and is increasingly seen as a core ADHD symptom that is clinically associated with greater functional impairment and psychiatric comorbidity. We sought to investigate emotional dysregulation in ADHD and explored its neural underpinnings. METHODS: We studied emotion induction and regulation in a clinical cohort of adult patients with ADHD before and after a stimulant challenge. We compared patients with age- and gender-matched healthy controls using behavioural, structural, and functional measures. We hypothesized that patients would demonstrate aberrant emotion processing compared with healthy controls, and sought to find whether this could be normalized by stimulant medication. RESULTS: Behaviourally, the ADHD group showed reduced emotion induction and regulation capacity. Brain imaging revealed abberant activation and deactivation patterns during emotion regulation, lower grey-matter volume in limbic and paralimbic areas, and greater grey-matter volume in visual and cerebellar areas, compared with healthy controls. The behavioural and functional deficits seen in emotion induction and regulation in the ADHD group were not normalized by stimulant medication. CONCLUSION: Patients with ADHD may have impaired emotion induction and emotion regulation capacity, but these deficits are not reversed by stimulant medication. These results have important clinical implications when assessing which aspects of emotional dysregulation are relevant for patients and if and how traditional ADHD pharmacotherapy affects emotion induction and emotion regulation.


Subject(s)
Attention Deficit Disorder with Hyperactivity , Brain , Central Nervous System Stimulants , Magnetic Resonance Imaging , Humans , Attention Deficit Disorder with Hyperactivity/drug therapy , Attention Deficit Disorder with Hyperactivity/physiopathology , Central Nervous System Stimulants/pharmacology , Male , Female , Adult , Brain/diagnostic imaging , Brain/physiopathology , Brain/drug effects , Emotional Regulation/physiology , Young Adult , Gray Matter/diagnostic imaging , Gray Matter/pathology , Gray Matter/drug effects , Affective Symptoms/drug therapy , Affective Symptoms/physiopathology , Emotions/physiology , Emotions/drug effects , Case-Control Studies , Middle Aged
7.
Behav Brain Funct ; 20(1): 19, 2024 Aug 05.
Article in English | MEDLINE | ID: mdl-39103929

ABSTRACT

Caffeine is a widely used drug that broadly affects human cognition and brain function. Caffeine acts as an antagonist to the adenosine receptors in the brain. Previous anecdotal reports have also linked caffeine intake with changes in pupil diameter. By modifying the retinal irradiance, pupil diameter modulates all ocular light exposure relevant for visual (i.e., perception, detection and discrimination of visual stimuli) and non-visual (i.e., circadian) functions. To date, the extent of the influence of caffeine on pupillary outcomes, including pupil diameter, has not been examined in a systematic review. We implemented a systematic review laid out in a pre-registered protocol following PRISMA-P guidelines. We only included original research articles written in English reporting studies with human participants, in which caffeine was administered, and pupil diameter was measured using objective methods. Using broad search strategies, we consulted various databases (PsycINFO, Medline, Embase, Cochrane Library, bioRxiv and medRxiv) and used the Covidence platform to screen, review and extract data from studies. After importing studies identified through database search (n = 517 imported, n = 46 duplicates), we screened the title and abstracts (n = 471), finding 14 studies meeting our eligibility criteria. After full-text review, we excluded seven studies, leaving only a very modest number of included studies (n = 7). Extraction of information revealed that the existing literature on the effect of caffeine on pupil parameters is very heterogeneous, differing in pupil assessment methods, time of day of caffeine administration, dose, and protocol timing and design. The evidence available in the literature does not provide consistent results but studies rated as valid by quality assessment suggest a small effect of caffeine on pupil parameters. We summarize the numeric results as both differences in absolute pupil diameter and in terms of effect sizes. More studies are needed using modern pupil assessment methods, robust study design, and caffeine dose-response methodology.


Subject(s)
Caffeine , Pupil , Humans , Caffeine/pharmacology , Caffeine/administration & dosage , Pupil/drug effects , Pupil/physiology , Central Nervous System Stimulants/pharmacology , Central Nervous System Stimulants/administration & dosage
8.
Prog Brain Res ; 288: 35-58, 2024.
Article in English | MEDLINE | ID: mdl-39168558

ABSTRACT

Caffeine is an alkaloid obtained from plants and is one of the most consumptive drug in the form of chocolate, coffee and beverages. The potential impact of caffeine within CNS can be easily understood by mechanism of action-antagonism of adenosine receptor, calcium influx, inhibits phosphodiesterases. Adenosine a neuromodulator for adenosine receptors, which are abundantly expressed within the central nervous system. Caffeine antagonized the adenosine receptor, hence stimulate expression of dopamine. It plays pivotal role in many metabolic pathways within the brain and nervous system, it reduced the amyloid-ß-peptide (Aß) accumulation, downregulation of tau protein phosphorylation, stimulate cholinergic neurons and inhibits the acetylcholinestrase (AChE). It also possess antioxidant and antiapoptotic activity. Caffeine act as nutraceutical product, improves mental health. It contains antioxidants, vitamins, minerals and dietary supplements, by reducing the risk factor of several neurodegenerations including Alzheimer's disease, migraine, gallstone, cancer, Huntington's disease and sclerosis. This act as a stimulant and have capability to increase the effectiveness of certain pain killer. Beside positive affects, over-consumption of caffeine leads to negative impact: change in sleep pattern, hallucinations, high blood pressure, mineral loss and even heartburn. This chapter highlights pros and cons of caffeine consumption.


Subject(s)
Caffeine , Caffeine/pharmacology , Humans , Animals , Central Nervous System/drug effects , Central Nervous System/metabolism , Central Nervous System Stimulants/pharmacology , Receptors, Purinergic P1/metabolism
9.
Article in English | MEDLINE | ID: mdl-39002928

ABSTRACT

Zebrafish are a dynamic research model in the domains of neuropsychopharmacology, biological psychiatry and behaviour. Working with larvae ≤4 days post-fertilisation (dpf) offers an avenue for high-throughput investigation whilst aligning with the 3Rs principles of animal research. The light/dark assay, which is the most widely used behavioural assay for larval neuropharmacology research, lacks experimental reliability and standardisation. This study aimed to formulate a robust, reproducible and standardised light/dark behavioural assay using 4 dpf zebrafish larvae. Considerable between-batch and inter-individual variability was found, which we rectified with a normalisation approach to ensure a reliable foundation for analysis. We then identified that 5-min light/dark transition periods are optimal for locomotor activity. We also found that a 30-min acclimation in the light was found to produce significantly increased dark phase larval locomotion. Next, we confirmed the pharmacological predictivity of the standardised assay using ethanol which, as predicted, caused hyperlocomotion at low concentrations and hypolocomotion at high concentrations. Finally, the assay was validated by assessing the behavioural phenotype of hyperactive transgenic (adgrl3.1-/-) larvae, which was rescued with psychostimulant medications. Our standardised assay not only provides a clear experimental and analytical framework to work with 4 dpf larvae, but also facilitates between-laboratory collaboration using our normalisation approach.


Subject(s)
Behavior, Animal , Larva , Locomotion , Zebrafish , Animals , Zebrafish/physiology , Behavior, Animal/drug effects , Behavior, Animal/physiology , Locomotion/drug effects , Locomotion/physiology , Animals, Genetically Modified , Ethanol/pharmacology , Reproducibility of Results , Motor Activity/drug effects , Motor Activity/physiology , Photoperiod , Light , Central Nervous System Stimulants/pharmacology
10.
Nutrients ; 16(14)2024 Jul 11.
Article in English | MEDLINE | ID: mdl-39064667

ABSTRACT

While previous studies have explored a range of factors governing the optimal use of caffeine (CAF) in athletes, limited research has explored how time of day (TOD) affects the ergogenic effects of various CAF dosages on physical performance. This study aimed to increase knowledge about how different recommended CAF doses (3 mg/kg vs. 6 mg/kg) ingested at different TODs affected maximal high-intensity physical performance and the perception of potential side effects in female athletes. In this double-blind, randomized, and counterbalanced study, 15 low CAF consumer athletes (aged 18.3 ± 0.5 y) underwent six trials, including three testing conditions assessed across two TODs: one in the morning (08:00 a.m.) and one in the evening (06:00 p.m.). During each condition, the participants ingested either a placebo, 3 mg/kg CAF (CAF (3 mg)), or 6 mg/kg CAF (CAF (6 mg)) capsules 60 min before each test with an in-between washout period of at least 72 h. In each trial, the participants performed a countermovement jumps test (CMJ), a modified agility t test (MATT), a repeated sprint ability (RSA), a rating of perceived exertion (RPE), and finally, a CAF side effects questionnaire. Our findings indicate the absence of an ergogenic effect on CMJ, MAT, and RSA performance in the evening after administering CAF (3 mg) or CAF (6 mg) compared to a placebo. Likewise, when CAF was ingested in the morning, there was an improvement in these performances with both CAF (3 mg) and CAF (6 mg), with greater improvement observed after CAF (6 mg). Additionally, neither the CAF dosage nor the TOD had a significant effect on the RPE. The occurrence of side effects increased significantly after the evening ingestion of CAF, particularly with a moderate dose of CAF (6 mg). Our findings indicate that the effectiveness of CAF depends on the TOD and CAF dosage. When ingested in the morning, a moderate dose of CAF (6 mg), rather than CAF (3 mg), is more effective in improving short-term physical performance without affecting CAF side effects in female athletes. Nevertheless, when ingested in the evening, neither dose was sufficient to enhance short-term physical performance, and both dosages increased the incidence of CAF side effects, particularly at a moderate dose.


Subject(s)
Athletes , Athletic Performance , Caffeine , Humans , Caffeine/administration & dosage , Caffeine/pharmacology , Caffeine/adverse effects , Female , Double-Blind Method , Athletic Performance/physiology , Adolescent , Young Adult , Dose-Response Relationship, Drug , Performance-Enhancing Substances/administration & dosage , Performance-Enhancing Substances/adverse effects , Drug Administration Schedule , Time Factors , Central Nervous System Stimulants/administration & dosage , Central Nervous System Stimulants/pharmacology
11.
J Psychopharmacol ; 38(7): 636-646, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39068640

ABSTRACT

BACKGROUND: Methamphetamine is frequently co-consumed with alcohol, yet combined effects on visually guided behaviours have not been experimentally assessed. This study examined whether methamphetamine and alcohol-induced changes in gaze behaviour can be accurately detected and indexed during a simulated driving task to establish characteristic patterns relevant to traffic safety. METHODS: In a randomised, placebo-controlled, cross-over study design, the effects of acute oral methamphetamine (0.42 mg/kg) were assessed with and without low doses of alcohol (target 0.04% blood alcohol content) on gaze behaviour during driving. Twenty healthy adults (mean age 29.5 years (SD ± 4.9), 40% female) completed four, 1-h simulated drives with simultaneous eye monitoring using the SensoMotoric Instruments cap-mounted eye tracker over a 4-week experimental paradigm. Gaze entropy measures were used to quantify visual scanning efficiency, expressed as gaze transition entropy and stationary gaze entropy. Fixations, recorded as duration (milliseconds, ms) and rate (count) per minute, were examined in 10-min bins over the duration of the drive. Driving performance was assessed by the standard deviation of lateral position, standard deviation of speed and steering variability. RESULTS: Methamphetamine increased the rate and duration of fixations and produced a less dispersed but more disorganised pattern of gaze during highway driving while preserving performance. Alcohol alone impaired both oculomotor control and driving performance, even when consumed at levels well below the legal limit stipulated in many international jurisdictions. CONCLUSIONS: Methamphetamine-affected drivers display inefficient exploration in a limited visual range during driving. Eye-tracking metrics thus show potential for indexing intoxication due to psychoactive substance usage.


Subject(s)
Alcohol Drinking , Automobile Driving , Cross-Over Studies , Methamphetamine , Humans , Female , Male , Double-Blind Method , Adult , Methamphetamine/administration & dosage , Young Adult , Eye-Tracking Technology , Eye Movements/drug effects , Fixation, Ocular/drug effects , Fixation, Ocular/physiology , Ethanol/pharmacology , Ethanol/administration & dosage , Psychomotor Performance/drug effects , Central Nervous System Stimulants/pharmacology , Central Nervous System Stimulants/administration & dosage
12.
eNeuro ; 11(8)2024 Aug.
Article in English | MEDLINE | ID: mdl-38969501

ABSTRACT

Cognitive dysfunction is associated with methamphetamine use disorder (MUD). Here, we used genetic and pharmacological approaches to examine the involvement of either Group 2 metabotropic glutamate (mGlu2) or mGlu3 receptors in memory deficit induced by methamphetamine in mice. Methamphetamine treatment (1 mg/kg, i.p., once a day for 5 d followed by 7 d of withdrawal) caused an impaired performance in the novel object recognition test in wild-type mice, but not in mGlu2-/- or mGlu3-/- mice. Memory deficit in wild-type mice challenged with methamphetamine was corrected by systemic treatment with selectively negative allosteric modulators of mGlu2 or mGlu3 receptors (compounds VU6001966 and VU0650786, respectively). Methamphetamine treatment in wild-type mice caused large increases in levels of mGlu2/3 receptors, the Type 3 activator of G-protein signaling (AGS3), Rab3A, and the vesicular glutamate transporter, vGlut1, in the prefrontal cortex (PFC). Methamphetamine did not alter mGlu2/3-mediated inhibition of cAMP formation but abolished the ability of postsynaptic mGlu3 receptors to boost mGlu5 receptor-mediated inositol phospholipid hydrolysis in PFC slices. Remarkably, activation of presynaptic mGlu2/3 receptors did not inhibit but rather amplified depolarization-induced [3H]-D-aspartate release in synaptosomes prepared from the PFC of methamphetamine-treated mice. These findings demonstrate that exposure to methamphetamine causes changes in the expression and function of mGlu2 and mGlu3 receptors, which might alter excitatory synaptic transmission in the PFC and raise the attractive possibility that selective inhibitors of mGlu2 or mGlu3 receptors (or both) may be used to improve cognitive dysfunction in individuals affected by MUD.


Subject(s)
Central Nervous System Stimulants , Methamphetamine , Mice, Inbred C57BL , Mice, Knockout , Receptors, Metabotropic Glutamate , Recognition, Psychology , Animals , Methamphetamine/pharmacology , Receptors, Metabotropic Glutamate/metabolism , Recognition, Psychology/drug effects , Recognition, Psychology/physiology , Male , Central Nervous System Stimulants/pharmacology , Memory Disorders/metabolism , Mice , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism
13.
Behav Brain Res ; 472: 115151, 2024 Aug 24.
Article in English | MEDLINE | ID: mdl-39019091

ABSTRACT

Caffeine exerts a biphasic effect on zebrafish behavior. High doses of caffeine have been associated with increased stress and anxiety, whereas low doses have been found to enhance performance on tasks requiring focus and attention. However, the sex-specific nature of these biphasic effects on behavior and physiology remains unclear. This study assessed the behavioral responses and hormone levels in male and female zebrafish after acute exposure to caffeine ranging from 0.3 to 600 mg/L. The results showed no significant difference in caffeine intake between males and females after acute exposure at each concentration. Caffeine-induced behavioral and physiological responses indicated a threshold dosage existed between 30 and 300 mg/L. Female fish displayed increased anxiety-like behavioral phenotypes, i.e., latency to upper and freezing, whereas males exhibited more erratic movement following acute exposure to a high-dose treatment. In addition, females exhibited a significant increase in whole-body cortisol levels, while males experienced a testosterone elevation at 300 mg/L of caffeine acute exposure. There was a significant decrease in the duration of erratic movements in males treated with the androgen receptor antagonist flutamide compared to the control group. The transcriptome analysis uncovered 511 and 592 up-regulated and 761 and 922 down-regulated differential expression genes in males and females, respectively, compared to the control. The Kyoto Encyclopedia of Genes and Genomes (KEGG) and Gene Ontology (GO) pathway analysis revealed that caffeine has the potential to impact various pathways in zebrafish, including phototransduction and steroid hormone biosynthesis. Our findings demonstrate that testosterone and cortisol play a combined role in regulating stress responses in both behavior and physiology. Furthermore, our study highlights the significance of encompassing both male and female zebrafish as a model system.


Subject(s)
Anxiety , Behavior, Animal , Caffeine , Hydrocortisone , Sex Characteristics , Testosterone , Zebrafish , Animals , Caffeine/pharmacology , Caffeine/administration & dosage , Female , Male , Anxiety/chemically induced , Anxiety/metabolism , Hydrocortisone/metabolism , Hydrocortisone/blood , Testosterone/metabolism , Testosterone/blood , Behavior, Animal/drug effects , Central Nervous System Stimulants/pharmacology , Central Nervous System Stimulants/administration & dosage , Flutamide/pharmacology , Sex Factors , Dose-Response Relationship, Drug
14.
Behav Brain Res ; 472: 115152, 2024 Aug 24.
Article in English | MEDLINE | ID: mdl-39032868

ABSTRACT

The high rate of relapse to compulsive methamphetamine (MA)-taking and seeking behaviors after abstinence constitutes a major obstacle to the treatment of MA addiction. Perineuronal nets (PNNs), essential components of the extracellular matrix, play a critical role in synaptic function, learning, and memory. Abnormalities in PNNs have been closely linked to a series of neurological diseases, such as addiction. However, the exact role of PNNs in MA-induced related behaviors remains elusive. Here, we established a MA-induced conditioned place preference (CPP) paradigm in female mice and found that the number and average optical density of PNNs increased significantly in the medial prefrontal cortex (mPFC) of mice during the acquisition, extinction, and reinstatement stages of CPP. Notably, the removal of PNNs in the mPFC via chondroitinase ABC (ChABC) before extinction training not only facilitated the extinction of MA-induced CPP and attenuated the relapse of extinguished MA preference but also significantly reduced the activation of c-Fos in the mPFC. Similarly, the ablation of PNNs in the mPFC before reinstatement markedly lessened the reinstatement of MA-induced CPP, which was accompanied by the decreased expression of c-Fos in the mPFC. Collectively, our results provide more evidence for the implication of degradation of PNNs in facilitating extinction and preventing relapse of MA-induced CPP, which indicate that targeting PNNs may be an effective therapeutic option for MA-induced CPP memories.


Subject(s)
Extinction, Psychological , Methamphetamine , Mice, Inbred C57BL , Prefrontal Cortex , Animals , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Methamphetamine/pharmacology , Female , Extinction, Psychological/drug effects , Extinction, Psychological/physiology , Mice , Extracellular Matrix/metabolism , Extracellular Matrix/drug effects , Central Nervous System Stimulants/pharmacology , Conditioning, Classical/drug effects , Conditioning, Classical/physiology , Drug-Seeking Behavior/drug effects , Drug-Seeking Behavior/physiology , Nerve Net/drug effects , Nerve Net/metabolism , Chondroitin ABC Lyase/pharmacology
15.
Physiol Behav ; 284: 114646, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-39053627

ABSTRACT

Clavulanic acid (CLAV) is a component of Augmentin® that preserves antibiotic efficacy by inhibiting ß-lactamase activity. It also enhances cellular glutamate uptake and is a potential CNS therapeutic. Because increased glutamate transmission in brain reward circuits facilitates methamphetamine (METH) locomotor activation and sensitization, we tested the hypothesis that CLAV inhibits acute and sensitized locomotor responses to METH in mice and investigated effects of CLAV on METH-induced changes in glutaminase, the major glutamate-producing enzyme in the brain. Acute METH (3 mg/kg) produced hyperlocomotion that was reduced by CLAV (20 mg/kg but not 10 mg/kg). Mice injected with METH (3 mg/kg) every other day for 9 d and then challenged with METH 27 d later displayed locomotor sensitization. CLAV (10 mg/kg), when injected 15 min before each METH injection during the 9-d exposure interval, blocked locomotor sensitization induced by METH challenge. In METH-sensitized mice, mRNA levels of both isoforms of glutaminase (GLS and GLS2) were altered in the nucleus accumbens compared to mice exposed to a single injection of METH (i.e., GLS decreased and GLS2 increased). CLAV normalized the METH-induced GLS deficit but not the increase in GLS2. In summary, CLAV reduced acute and sensitized locomotor responses to METH and normalized the METH-induced reduction of GLS gene expression in the NAC. Given that glutaminases belong to the ß-lactamase superfamily and CLAV is a ß-lactamase inhibitor, our data point toward studying glutaminase as a therapeutic target of CLAV.


Subject(s)
Central Nervous System Stimulants , Clavulanic Acid , Glutaminase , Methamphetamine , Nucleus Accumbens , RNA, Messenger , Animals , Methamphetamine/pharmacology , Nucleus Accumbens/drug effects , Nucleus Accumbens/metabolism , Glutaminase/metabolism , Male , Clavulanic Acid/pharmacology , RNA, Messenger/metabolism , RNA, Messenger/drug effects , Central Nervous System Stimulants/pharmacology , Mice, Inbred C57BL , Mice , Locomotion/drug effects , Motor Activity/drug effects , Dose-Response Relationship, Drug
16.
Article in English | MEDLINE | ID: mdl-38880464

ABSTRACT

Methylphenidate (MPH) is a central nervous system stimulant drug and a first order prescription in the treatment of Attention Deficit Hyperactivity Disorder (ADHD). Although MPH biochemistry in neurodevelopment is not completely understood, studies showed it alters energy metabolism in rat brains. ADHD prevalence during neurodevelopment is related to males and the investigation has been mainly done in these subjects, therefore, little is known about MPH action in females and, consequently, about sexual dimorphism. In the present study we evaluated markers of mitochondrial dynamics (DRP1 and MFN2, fission and fusion, respectively), biogenesis (mtTFA) and bioenergetics (respiratory chain complexes) in prefrontal cortex of male and female juvenile rats submitted to exposure to MPH to better understand MPH effect during postnatal neurodevelopment. ATP and oxidative stress levels were also evaluated. Wistar rats received intraperitoneal injection of MPH (2.0 mg/kg) or control (saline), once a day, from 15th to 45th day of age. Results showed that MPH increased DRP1 and decreased MFN2, as well as increased mtTFA in prefrontal cortex of male rats. In female, MPH decreased NRF1 and increased Parkin, which are mitochondrial regulatory proteins. Respiratory chain complexes (complex I, SDH, complexes III and IV), ATP production and oxidative stress parameters were altered and shown to be sex-dependent. Taken together, results suggest that chronic MPH exposure at an early age in healthy animals changes mitochondrial dynamics, biogenesis and bioenergetics differently depending on the sex of the subjects.


Subject(s)
Central Nervous System Stimulants , Dynamins , Energy Metabolism , Methylphenidate , Mitochondrial Dynamics , Oxidative Stress , Prefrontal Cortex , Rats, Wistar , Animals , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Methylphenidate/pharmacology , Male , Mitochondrial Dynamics/drug effects , Female , Central Nervous System Stimulants/pharmacology , Energy Metabolism/drug effects , Oxidative Stress/drug effects , Dynamins/metabolism , Rats , Sex Characteristics , Adenosine Triphosphate/metabolism , GTP Phosphohydrolases/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondrial Proteins , Ubiquitin-Protein Ligases
17.
Brain Behav Immun ; 120: 167-180, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38834156

ABSTRACT

It is widely believed that the activation of the central dopamine (DA) system is crucial to the rewarding effects of methamphetamine (METH) and to the behavioral outcomes of METH use disorder. It was reported that METH exposure induced gasdermin D (GSDMD)-dependent pyroptosis in rats. The membrane pore formation caused by METH-induced pyroptosis may also contribute to the overflow of DA into the extracellular space and subsequently increase the DA levels in the brain. The present study firstly investigated whether the membrane pore information induced by GSDMD-dependent pyroptosis was associated with the increased DA levels in the ventral tegmental area (VAT) and nucleus accumbens (NAc) of rats self-administering METH and SY-SH5Y cells treated by METH. Subsequently, the effect of pore formation blockade or genetic inhibition of GSDMD on the reinforcing and motivational effect of METH was determined in rats, using the animal model of METH self-administration (SA). METH exposure significantly increased the activity of NLRP1/Cas-1/GSDMD pathway and the presence of pyroptosis, accompanied by the significantly increased DA levels in VTA and NAc. Moreover, intraperitoneal injections of disulfiram (DSF) or microinjection of rAAV-shGSDMD into VTA/NAc significantly reduced the reinforcing and motivational effect of METH, accompanied by the decreased level of DA in VTA and NAc. The results provided novel evidence that METH-induced pyroptosis could increase DA release in VTA and NAc via the NLRP1/Cas-1/GSDMD pathway. Additionally, membrane pores or GSDMD blockade could significantly reduce the reinforcing and motivational effect of METH. In conclusion, blocking GSDMD and membrane pore formation could be a promising potential target for the development of agents to treat METH use disorder.


Subject(s)
Dopamine , Methamphetamine , Nucleus Accumbens , Phosphate-Binding Proteins , Pyroptosis , Self Administration , Ventral Tegmental Area , Animals , Methamphetamine/pharmacology , Methamphetamine/administration & dosage , Pyroptosis/drug effects , Male , Phosphate-Binding Proteins/metabolism , Rats , Nucleus Accumbens/metabolism , Nucleus Accumbens/drug effects , Dopamine/metabolism , Ventral Tegmental Area/metabolism , Ventral Tegmental Area/drug effects , Intracellular Signaling Peptides and Proteins/metabolism , Rats, Sprague-Dawley , Humans , Central Nervous System Stimulants/pharmacology , Central Nervous System Stimulants/administration & dosage , Gasdermins
18.
Int J Neuropsychopharmacol ; 27(7)2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38875132

ABSTRACT

BACKGROUND: A compelling hypothesis about attention-deficit/hyperactivity disorder (ADHD) etiopathogenesis is that the ADHD phenotype reflects a delay in cortical maturation. Slow-wave activity (SWA) of non-rapid eye movement (NREM) sleep electroencephalogram (EEG) is an electrophysiological index of sleep intensity reflecting cortical maturation. Available data on ADHD and SWA are conflicting, and developmental differences, or the effect of pharmacological treatment, are relatively unknown. METHODS: We examined, in samples (Mage = 16.4, SD = 1.2), of ever-medicated adolescents at risk for ADHD (n = 18; 72% boys), medication-naïve adolescents at risk for ADHD (n = 15, 67% boys), and adolescents not at risk for ADHD (n = 31, 61% boys) matched for chronological age and controlling for non-ADHD pharmacotherapy, whether ADHD pharmacotherapy modulates the association between NREM SWA and ADHD risk in home sleep. RESULTS: Findings indicated medication-naïve adolescents at risk for ADHD exhibited greater first sleep cycle and entire night NREM SWA than both ever-medicated adolescents at risk for ADHD and adolescents not at risk for ADHD and no difference between ever-medicated, at-risk adolescents, and not at-risk adolescents. CONCLUSIONS: Results support atypical cortical maturation in medication-naïve adolescents at risk for ADHD that appears to be normalized by ADHD pharmacotherapy in ever-medicated adolescents at risk for ADHD. Greater NREM SWA may reflect a compensatory mechanism in middle-later adolescents at risk for ADHD that normalizes an earlier occurring developmental delay.


Subject(s)
Attention Deficit Disorder with Hyperactivity , Electroencephalography , Humans , Attention Deficit Disorder with Hyperactivity/physiopathology , Attention Deficit Disorder with Hyperactivity/drug therapy , Adolescent , Male , Female , Sleep, Slow-Wave/physiology , Sleep, Slow-Wave/drug effects , Central Nervous System Stimulants/pharmacology , Sleep Stages/drug effects , Sleep Stages/physiology
19.
Neurosci Biobehav Rev ; 163: 105774, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38914177

ABSTRACT

To provide an overview of treatments in the pipeline for adults with attention-deficit/hyperactivity disorder (ADHD), we searched https://clinicaltrials.gov/and and https://www.clinicaltrialsregister.eu/ from 01/01/2010-10/18/2023 for ongoing or completed phase 2 or 3 randomised controlled trials (RCTs), assessing pharmacological or non-pharmacological interventions for adults with ADHD with no current regulatory approval. We found 90 eligible RCTs. Of these, 24 (27 %) reported results with statistical analysis for primary efficacy endpoints. While several pharmacological and non-pharmacological interventions had evidence of superiority compared to the control condition from a single RCT, centanafadine (norepinephrine, dopamine, and serotonin re-uptake inhibitor) was the only treatment with evidence of efficacy on ADHD core symptoms (small effect size=0.28-0.40) replicated in at least one additional RCT, alongside reasonable tolerability. Overall, the body of ongoing RCTs in adults with ADHD is insufficient, without any intervention on the horizon to match the efficacy of stimulant treatment or atomoxetine and with better tolerability profile. Additional effective and well tolerated treatments for adults with ADHD require development and testing.


Subject(s)
Attention Deficit Disorder with Hyperactivity , Attention Deficit Disorder with Hyperactivity/drug therapy , Attention Deficit Disorder with Hyperactivity/therapy , Humans , Adult , Randomized Controlled Trials as Topic , Central Nervous System Stimulants/therapeutic use , Central Nervous System Stimulants/pharmacology
20.
Pharmacol Biochem Behav ; 241: 173793, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38823543

ABSTRACT

OBJECTIVE: Caffeine and modafinil are used to reverse effects of sleep deprivation. Nicotinic alpha-7 receptor and AMPA receptor positive allosteric modulators (PAM) are also potential substances in this context. Our objective is to evaluate the effects of caffeine, modafinil, AVL-3288 (nicotinic alpha-7 PAM) and CX516 (AMPA receptor PAM) on cognition and mood in a model of sleep deprivation. METHOD: Modified multiple platform model is used to sleep-deprive mice for 24 days, for 8 h/day. Vehicle, Modafinil (40 mg/kg), Caffeine (5 mg/kg), CX516 (10 mg/kg), and AVL3288 (1 mg/kg) were administered intraperitoneally daily. A cognitive test battery was applied every six days for four times. The battery that included elevated plus maze, novel object recognition, and sucrose preference tests was administered on consecutive days. RESULTS: Sleep deprivation decreased novel object recognition skill, but no significant difference was found in anxiety and depressive mood. Caffeine administration decreased anxiety-like behavior in short term, but this effect disappeared in chronic administration. Caffeine administration increased memory performance in chronic period. AVL group showed better memory performance in short term, but this effect disappeared in the rest of experiment. Although, in the modafinil group, no significant change in mood and memory was observed, anhedonia was observed in the chronic period in vehicle, caffeine and modafinil groups, but not in AVL-3288 and CX-516 groups. CONCLUSION: Caffeine has anxiolytic effect in acute administration. The improvement of memory in chronic period may be associated with the neuroprotective effects of caffeine. AVL-3288 had a short-term positive effect on memory, but tolerance to these effects developed over time. Furthermore, no anhedonia was observed in AVL-3288 and CX516 groups in contrast to vehicle, caffeine and modafinil groups. This indicates that AVL-3288 and CX516 may show protective effect against depression.


Subject(s)
Affect , Caffeine , Cognition , Modafinil , Sleep Deprivation , Animals , Sleep Deprivation/psychology , Sleep Deprivation/drug therapy , Sleep Deprivation/complications , Modafinil/pharmacology , Modafinil/administration & dosage , Mice , Male , Cognition/drug effects , Caffeine/pharmacology , Caffeine/administration & dosage , Affect/drug effects , Disease Models, Animal , Central Nervous System Stimulants/pharmacology , Central Nervous System Stimulants/administration & dosage , Benzhydryl Compounds/pharmacology , Benzhydryl Compounds/administration & dosage , Time Factors , Anxiety/drug therapy
SELECTION OF CITATIONS
SEARCH DETAIL