1
|
Li YT, Huang YL, Chen JJJ, Hyland BI, Wickens JR. Phasic dopamine signals are reduced in the spontaneously hypertensive rat and increased by methylphenidate. Eur J Neurosci 2024; 59:1567-1584. [PMID: 38314648 DOI: 10.1111/ejn.16269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Revised: 12/27/2023] [Accepted: 01/17/2024] [Indexed: 02/06/2024]
Abstract
The spontaneously hypertensive rat (SHR) is a selectively bred animal strain that is frequently used to model attention-deficit hyperactivity disorder (ADHD) because of certain genetically determined behavioural characteristics. To test the hypothesis that the characteristically altered response to positive reinforcement in SHRs may be due to altered phasic dopamine response to reward, we measured phasic dopamine signals in the SHRs and Sprague Dawley (SD) rats using in vivo fast-scan cyclic voltammetry. The effects of the dopamine reuptake inhibitor, methylphenidate, on these signals were also studied. Phasic dopamine signals during the pairing of a sensory cue with electrical stimulation of midbrain dopamine neurons were significantly smaller in the SHRs than in the SD rats. Over repeated pairings, the dopamine response to the sensory cue increased, whereas the response to the electrical stimulation of dopamine neurons decreased, similarly in both strains. However, the final amplitude of the response to the sensory cue after pairing was significantly smaller in SHRs than in the SD rats. Methylphenidate increased responses to sensory cues to a significantly greater extent in the SHRs than in the SD rats, due largely to differences in the low dose effect. At a higher dose, methylphenidate increased responses to sensory cues and electrical stimulation similarly in SHRs and SD rats. The smaller dopamine responses may explain the reduced salience of reward-predicting cues previously reported in the SHR, whereas the action of methylphenidate on the cue response suggests a potential mechanism for the therapeutic effects of low-dose methylphenidate in ADHD.
Collapse
Affiliation(s)
- Yu-Ting Li
- Neurobiology Research Unit, Okinawa Institute of Science and Technology, Okinawa, Japan
- Taiwan Instrument Research Institute, National Applied Research Laboratories, Hsinchu, Taiwan
| | - Yi-Ling Huang
- Neurobiology Research Unit, Okinawa Institute of Science and Technology, Okinawa, Japan
| | - Jia-Jin Jason Chen
- Department of Biomedical Engineering, National Cheng Kung University, Tainan, Taiwan
| | - Brian Ian Hyland
- Department of Physiology, Brain Health Research Centre, University of Otago, Dunedin, New Zealand
| | - Jeffery R Wickens
- Neurobiology Research Unit, Okinawa Institute of Science and Technology, Okinawa, Japan
| |
Collapse
|
2
|
Miyazaki K, Uchiyama M. An adolescent boy with kleptomania and attention-deficit hyperactivity disorder treated with methylphenidate and guanfacine: A case report. Neuropsychopharmacol Rep 2023; 43:650-653. [PMID: 37921065 PMCID: PMC10739163 DOI: 10.1002/npr2.12394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 10/02/2023] [Accepted: 10/18/2023] [Indexed: 11/04/2023] Open
Abstract
We present the case of a patient, a boy of 16 years of age at initial presentation, with kleptomania, an impulse disorder characterized by an impulse to steal unneeded items, and attention-deficit hyperactivity disorder (ADHD). The patient's parents reported that he would frequently impulsively steal items and money that he did not need. Cognitive and physical assessments revealed no abnormalities, and the patient had no history of substance abuse. The patient was diagnosed with kleptomania and ADHD. The patient was started on Osmotic Release Oral System Methylphenidate (OROS-MPH), a medication commonly used to treat ADHD, and experienced improvement in ADHD symptoms and stealing behavior. At 19 years of age, it was discovered that the patient's behavioral symptoms were uncontrolled during times of the day when the blood concentration of MPH was likely to have waned. After starting an additional dose of guanfacine at night, his symptoms during these times of day improved. While existing research is not definitive, there may be a connection between ADHD and kleptomania. Further, there are some reports that treatment of ADHD with MPH also reduced stealing behavior, aligning with our present findings. We discuss the potential mechanisms behind these improvements and further present the first evidence of the efficacy of guanfacine in the treatment of kleptomania.
Collapse
Affiliation(s)
- Kensuke Miyazaki
- Department of NeuropsychiatryHirosaki‐Aiseikai HospitalHirosaki‐shi, AomoriJapan
| | - Michiko Uchiyama
- Department of Hospital PharmacyHirosaki‐Aiseikai HospitalHirosaki‐shi, AomoriJapan
| |
Collapse
|
3
|
Soufsaf S, Robaey P, Nekka F. An exploratory analysis of the performance of methylphenidate regimens based on a PKPD model of dopamine and norepinephrine transporter occupancy. J Pharmacokinet Pharmacodyn 2023:10.1007/s10928-023-09854-y. [PMID: 36930337 DOI: 10.1007/s10928-023-09854-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 02/23/2023] [Indexed: 03/18/2023]
Abstract
Methylphenidate (MPH) is a psychostimulant which inhibits the uptake of dopamine and norepinephrine transporters, DAT and NET, and is mostly used to treat Attention Deficit/Hyperactivity Disorder. The current dose optimization is done through titration, a cumbersome approach for patients. To assess the therapeutic performance of MPH regimens, we introduce an in silico framework composed of (i) a population pharmacokinetic model of MPH, (ii) a pharmacodynamic (PD) model of DAT and NET occupancy, (iii) a therapeutic box delimited by time and DAT occupancy, and (iv) a performance score computation. DAT occupancy data was digitized (n = 152) and described with Emax models. NET occupancy was described with a KPD model. We used this integrative framework to simulate the performance of extended-release (18-99 mg) and tid MPH regimens (25-40 mg). Early blood samples of MPH seem to lead to higher DAT occupancy, consistent with an acute tolerance observed in clinical rating scales. An Emax model with a time-dependent tolerance was fitted to available data to assess the observed clockwise hysteresis. Peak performance is observed at 63 mg. While our analysis does not deny the existence of an acute tolerance, data precision in terms of formulation and sampling times does not allow a definite confirmation of this phenomenon. This work justifies the need for a more systematic collection of DAT and NET occupancy data to further investigate the presence of acute tolerance and assess the impact of low MPH doses on its efficacy.
Collapse
|
4
|
Kaiser A, Broeder C, Cohen JR, Douw L, Reneman L, Schrantee A. Effects of a single-dose methylphenidate challenge on resting-state functional connectivity in stimulant-treatment naive children and adults with ADHD. Hum Brain Mapp 2022; 43:4664-4675. [PMID: 35781371 PMCID: PMC9491277 DOI: 10.1002/hbm.25981] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 05/17/2022] [Accepted: 05/27/2022] [Indexed: 11/09/2022] Open
Abstract
Prior studies suggest that methylphenidate, the primary pharmacological treatment for attention-deficit/hyperactivity disorder (ADHD), alters functional brain connectivity. As the neurotransmitter systems targeted by methylphenidate undergo significant alterations throughout development, the effects of methylphenidate on functional connectivity may also be modulated by age. Therefore, we assessed the effects of a single methylphenidate challenge on brain network connectivity in stimulant-treatment naïve children and adults with ADHD. We obtained resting-state functional MRI from 50 boys (10-12 years of age) and 49 men (23-40 years of age) with ADHD (DSM IV, all subtypes), before and after an oral challenge with 0.5 mg/kg methylphenidate; and from 11 boys and 12 men as typically developing controls. Connectivity strength (CS), eigenvector centrality (EC), and betweenness centrality (BC) were calculated for the striatum, thalamus, dorsal anterior cingulate cortex (dACC), and prefrontal cortex (PFC). In line with our hypotheses, we found that methylphenidate decreased measures of connectivity and centrality in the striatum and thalamus in children with ADHD, but increased the same metrics in adults with ADHD. Surprisingly, we found no major effects of methylphenidate in the dACC and PFC in either children or adults. Interestingly, pre-methylphenidate, participants with ADHD showed aberrant connectivity and centrality compared to controls predominantly in frontal regions. Our findings demonstrate that methylphenidate's effects on connectivity of subcortical regions are age-dependent in stimulant-treatment naïve participants with ADHD, likely due to ongoing maturation of dopamine and noradrenaline systems. These findings highlight the importance for future studies to take a developmental perspective when studying the effects of methylphenidate treatment.
Collapse
Affiliation(s)
- Antonia Kaiser
- Department of Radiology and Nuclear Medicine, Amsterdam Neuroscience, Amsterdam UMCUniversity of AmsterdamAmsterdamThe Netherlands
| | - Caroline Broeder
- Department of Radiology and Nuclear Medicine, Amsterdam Neuroscience, Amsterdam UMCUniversity of AmsterdamAmsterdamThe Netherlands
| | - Jessica R. Cohen
- Department of Psychology and NeuroscienceUniversity of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
| | - Linda Douw
- Department of Anatomy and Neurosciences, Amsterdam Neuroscience, Amsterdam UMCVrije Universiteit AmsterdamAmsterdamThe Netherlands
| | - Liesbeth Reneman
- Department of Radiology and Nuclear Medicine, Amsterdam Neuroscience, Amsterdam UMCUniversity of AmsterdamAmsterdamThe Netherlands
| | - Anouk Schrantee
- Department of Radiology and Nuclear Medicine, Amsterdam Neuroscience, Amsterdam UMCUniversity of AmsterdamAmsterdamThe Netherlands
| |
Collapse
|
5
|
Shram MJ, Setnik B, Webster L, Guenther S, Mickle TC, Braeckman R, Kanski J, Martin A, Kelsh D, Vince BD, Barrett AC. Oral, intranasal, and intravenous abuse potential of serdexmethylphenidate, a novel prodrug of d-methylphenidate. Curr Med Res Opin 2022; 38:1237-1250. [PMID: 35570699 DOI: 10.1080/03007995.2022.2076474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
OBJECTIVES Serdexmethylphenidate (SDX) chloride (Cl) is a novel prodrug of d-methylphenidate (d-MPH). These studies evaluated the abuse potential of SDX Cl when administered orally, intranasally (IN), and intravenously (IV). METHODS Three randomized, double-blind, placebo- and active-controlled crossover studies were conducted in recreational drug users to evaluate the abuse-related effects of oral SDX (120 and 240 mg) vs. extended-release (ER) d-MPH (80 mg) and phentermine (60 mg); IN SDX (80 mg) vs. d-MPH (40 mg), and IV SDX (30 mg) vs. d-MPH (15 mg). Abuse-related subjective measures, pharmacokinetics, and safety were assessed. RESULTS The primary endpoint of maximum (Emax) Drug Liking (DL) (0-100-point scale) was significantly higher following d-MPH vs. placebo, validating the studies. In the oral study, DL Emax was significantly higher following 80 mg ER d-MPH (Emax = 81.5) than 120 mg SDX (Emax = 62.8, p < .001) and 240 mg SDX (Emax = 63.8, p = .006); and following 60 mg phentermine (Emax = 80.2) than 120 mg SDX (p = .0195), but not 240 mg SDX (p = .0665). DL Emax scores were significantly higher following IN d-MPH vs SDX (Emax = 93.2 vs. 71.0, p < .0001) and following IV d-MPH vs. SDX (Emax = 84.3 vs. 56.6, p = .001). Intravenous SDX was non-inferior to placebo (p = .001) for DL Emax. Secondary endpoints (e.g. Take Drug Again) were generally consistent with the primary endpoint. Maximal and overall d-MPH exposure was lower for SDX than d-MPH for all routes. Adverse events typical of stimulants were more frequent with d-MPH than SDX. CONCLUSIONS These findings indicate that the novel d-MPH prodrug, SDX, has lower abuse potential than d-MPH and support its classification as a C-IV controlled substance.
Collapse
Affiliation(s)
- Megan J Shram
- Altreos Research Partners, Inc, Toronto, Canada
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Canada
| | - Beatrice Setnik
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Canada
- Altasciences, Laval, Canada
| | | | | | | | | | | | | | | | | | | |
Collapse
|
6
|
Contreras D, Piña R, Carvallo C, Godoy F, Ugarte G, Zeise M, Rozas C, Morales B. Methylphenidate Restores Behavioral and Neuroplasticity Impairments in the Prenatal Nicotine Exposure Mouse Model of ADHD: Evidence for Involvement of AMPA Receptor Subunit Composition and Synaptic Spine Morphology in the Hippocampus. Int J Mol Sci 2022; 23:ijms23137099. [PMID: 35806103 PMCID: PMC9266648 DOI: 10.3390/ijms23137099] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 06/22/2022] [Accepted: 06/22/2022] [Indexed: 02/04/2023] Open
Abstract
In ADHD treatment, methylphenidate (MPH) is the most frequently used medication. The present work provides evidence that MPH restored behavioral impairments and neuroplasticity due to changes in AMPAR subunit composition and distribution, as well as maturation of dendritic spines, in a prenatal nicotine exposure (PNE) ADHD mouse model. PNE animals and controls were given a single oral dose of MPH (1 mg/kg), and their behavior was tested for attention, hyperactivity, and working memory. Long-term potentiation (LTP) was induced and analyzed at the CA3/CA1 synapse in hippocampal slices taken from the same animals tested behaviorally, measuring fEPSPs and whole-cell patch-clamp EPSCs. By applying crosslinking and Western blots, we estimated the LTP effects on AMPAR subunit composition and distribution. The density and types of dendritic spines were quantified by using the Golgi staining method. MPH completely restored the behavioral impairments of PNE mice. Reduced LTP and AMPA-receptor-mediated EPSCs were also restored. EPSC amplitudes were tightly correlated with numbers of GluA1/GluA1 AMPA receptors at the cell surface. Finally, we found a lower density of dendritic spines in hippocampal pyramidal neurons in PNE mice, with a higher fraction of thin-type immature spines and a lower fraction of mushroom mature spines; the latter effect was also reversed by MPH.
Collapse
Affiliation(s)
- Darwin Contreras
- Laboratory of Neuroscience, Faculty of Chemistry and Biology, University of Santiago de Chile, Alameda 3363, Santiago 9170022, Chile; (D.C.); (F.G.); (G.U.)
| | - Ricardo Piña
- Departamento de Biología, Facultad de Ciencias Básicas, Universidad Metropolitana de Ciencias de la Educación, Santiago 7760197, Chile;
- Departamento de Ciencias Pedagógicas, Facultad de Educación, Universidad Bernardo O’Higgins, Santiago 8370993, Chile
| | - Claudia Carvallo
- Centro de investigación e innovación en Gerontología Aplicada (CIGAP), Facultad de Salud, Universidad Santo Tomás, Santiago 8370003, Chile;
| | - Felipe Godoy
- Laboratory of Neuroscience, Faculty of Chemistry and Biology, University of Santiago de Chile, Alameda 3363, Santiago 9170022, Chile; (D.C.); (F.G.); (G.U.)
| | - Gonzalo Ugarte
- Laboratory of Neuroscience, Faculty of Chemistry and Biology, University of Santiago de Chile, Alameda 3363, Santiago 9170022, Chile; (D.C.); (F.G.); (G.U.)
| | - Marc Zeise
- School of Psychology, Faculty of Humanities, University of Santiago de Chile, Santiago 9170022, Chile;
| | - Carlos Rozas
- Laboratory of Neuroscience, Faculty of Chemistry and Biology, University of Santiago de Chile, Alameda 3363, Santiago 9170022, Chile; (D.C.); (F.G.); (G.U.)
- Correspondence: (C.R.); (B.M.)
| | - Bernardo Morales
- Laboratory of Neuroscience, Faculty of Chemistry and Biology, University of Santiago de Chile, Alameda 3363, Santiago 9170022, Chile; (D.C.); (F.G.); (G.U.)
- Correspondence: (C.R.); (B.M.)
| |
Collapse
|
7
|
Braeckman R, Guenther S, Mickle TC, Barrett AC, Smith A, Oh C. Dose Proportionality and Steady-State Pharmacokinetics of Serdexmethylphenidate/Dexmethylphenidate, a Novel Prodrug Combination to Treat Attention-Deficit/Hyperactivity Disorder. J Child Adolesc Psychopharmacol 2022; 32:288-295. [PMID: 35666231 PMCID: PMC9245728 DOI: 10.1089/cap.2022.0015] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Objective: The study was designed to determine (1) the pharmacokinetic (PK) profile of dexmethylphenidate (d-MPH) after oral administration of three dosage strengths of a new treatment containing d-MPH and a novel prodrug, serdexmethylphenidate (SDX); (2) the dose proportionality of the different SDX/d-MPH dosages; and (3) the steady-state PK profile of d-MPH and SDX after multiple dosing of SDX/d-MPH. Methods: Twenty-three healthy volunteers (aged 18-55 years) under fasted conditions received in a crossover design SDX/d-MPH 26.1/5.2 mg (Treatment A), 39.2/7.8 mg (Treatment B), and 52.3/10.4 mg (Treatment C) for a total d-MPH hydrochloride equivalent dose of 20, 30, and 40 mg, respectively. After a 96-hour washout period, all participants received four consecutive daily doses of SDX/d-MPH 52.3/10.4 mg. Blood samples were collected for measurement of plasma d-MPH and SDX and for PK analysis. Results: Administration of all three doses of SDX/d-MPH resulted in a rapid rise and slow decline in the plasma concentration of d-MPH. For Treatments A, B, and C, mean (± standard deviation) maximum concentrations (Cmax) were 7.1 ± 2.1, 9.8 ± 2.8, and 13.8 ± 3.8 ng/mL, and overall exposures (AUC0-last) were 97.2 ± 28.8, 142.5 ± 41.2, and 199.8 ± 57.2 h*ng/mL, respectively. Dose-normalized Cmax, AUC0-last, and AUC0-inf for d-MPH were similar when comparing the high and low doses versus the middle dose. Power model regression analysis revealed that Cmax and AUC0-inf proportionally increased with an increase in SDX/d-MPH dose. In the multiple-dose study, d-MPH reached steady state before the third dose, and SDX after the first dose. Conclusion: The PK profile of SDX/d-MPH is characterized by a rapid rise and a gradual decline in d-MPH concentration, with proportional Cmax and AUC0-inf across doses. The PK attributes of SDX/d-MPH may optimize symptom control from early morning to early evening, while the demonstrated dose proportionality may facilitate initial dose titration and ongoing dose adjustment.
Collapse
Affiliation(s)
- Rene Braeckman
- KemPharm, Inc., Celebration, Florida, USA.,Address correspondence to: Rene Braeckman, PhD, KemPharm, Inc., 1180 Celebration Blvd, Suite 103, Celebration, FL 34747, USA
| | | | | | | | - Adam Smith
- KemPharm, Inc., Celebration, Florida, USA
| | - Charles Oh
- Corium, Inc., Grand Rapids, Michigan, USA
| |
Collapse
|
8
|
Adam LC, Repantis D, Konrad BN, Dresler M, Kühn S. Memory enhancement with stimulants: Differential neural effects of methylphenidate, modafinil, and caffeine. A pilot study. Brain Cogn 2021; 154:105802. [PMID: 34592684 DOI: 10.1016/j.bandc.2021.105802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 09/19/2021] [Accepted: 09/19/2021] [Indexed: 11/18/2022]
Abstract
Human memory is susceptible to manipulation in many respects. While consolidation is well known to be prone to disruption, there is also growing evidence for the enhancement of memory function. Beside cognitive strategies and mnemonic training, the use of stimulants may improve memory processing in healthy adults. In this single-dose, double-blind, within-subject, randomized, placebo-controlled pilot study, 20 mg methylphenidate (N = 13) or 200 mg modafinil (N = 12) or 200 mg caffeine (N = 14) were administrated to in total 39 healthy participants while performing a declarative memory task. Each participant received only one substance and functional magnetic resonance imaging (fMRI) was used to assess drug-dependent memory effects of the substance for encoding and recognition compared to task-related activation under placebo. While methylphenidate showed some behavioral effect regarding memory recall performance, on the neural level, methylphenidate-dependent deactivations were found in fronto-parietal and temporal regions during recognition of previously learned words. No BOLD alterations were seen during encoding. Caffeine led to deactivations in the precentral gyrus during encoding whereas modafinil did not show any BOLD signal alterations at all. These results should be interpreted with caution since this a pilot study with several limitations, most importantly the small number of participants per group. However, our main finding of task-related deactivations may point to a drug-dependent increase of efficiency in physiological response to memory processing.
Collapse
Affiliation(s)
- Lucas C Adam
- Lise Meitner Group for Environmental Neuroscience, Max Planck Institute for Human Development, Berlin, Germany
| | - Dimitris Repantis
- Lise Meitner Group for Environmental Neuroscience, Max Planck Institute for Human Development, Berlin, Germany; Charité - Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Psychiatry and Psychotherapy, Campus Benjamin Franklin, Berlin, Germany.
| | - Boris N Konrad
- Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Centre, Nijmegen, the Netherlands
| | - Martin Dresler
- Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Centre, Nijmegen, the Netherlands
| | - Simone Kühn
- Lise Meitner Group for Environmental Neuroscience, Max Planck Institute for Human Development, Berlin, Germany; University Medical Center Hamburg-Eppendorf (UKE), Department of Psychiatry and Psychotherapy, Hamburg, Germany
| |
Collapse
|
9
|
Moon C, Marion M, Thanos PK, Steiner H. Fluoxetine Potentiates Oral Methylphenidate-Induced Gene Regulation in the Rat Striatum. Mol Neurobiol 2021; 58:4856-4870. [PMID: 34213723 DOI: 10.1007/s12035-021-02466-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 06/21/2021] [Indexed: 11/24/2022]
Abstract
Methylphenidate (MP) is combined with selective serotonin reuptake inhibitors (SSRIs) such as fluoxetine (FLX) to treat various disorders. MP, a dopamine reuptake inhibitor, helps manage attention-deficit hyperactivity disorder (ADHD) and is abused as a cognitive enhancer; it has a reduced addiction liability. We showed that combining FLX (serotonin) with MP potentiates MP-induced gene regulation in the striatum. These studies used intraperitoneal drug administration, which is relevant for MP abuse. Clinically, MP and FLX are taken orally (slower bioavailability). Here, we investigated whether chronic oral administration of MP and FLX also altered striatal gene regulation. MP (30/60 mg/kg/day), FLX (20 mg/kg/day), and MP + FLX were administered in rats' drinking water for 8 h/day over 4 weeks. We assessed the expression of dynorphin and substance P (both markers for striatal direct pathway neurons) and enkephalin (indirect pathway) by in situ hybridization histochemistry. Chronic oral MP alone produced a tendency for increased dynorphin and substance P expression and no changes in enkephalin expression. Oral FLX alone did not increase gene expression. In contrast, when given together, FLX greatly enhanced MP-induced expression of dynorphin and substance P and to a lesser degree enkephalin. Thus, FLX potentiated oral MP-induced gene regulation predominantly in direct pathway neurons, mimicking cocaine effects. The three functional domains of the striatum were differentially affected. MP + SSRI concomitant therapies are indicated in ADHD/depression comorbidity and co-exposure occurs with MP misuse as a cognitive enhancer by patients on SSRIs. Our findings indicate that MP + SSRI combinations, even given orally, may enhance addiction-related gene regulation.
Collapse
Affiliation(s)
- Connor Moon
- Stanson Toshok Center for Brain Function and Repair, Rosalind Franklin University of Medicine and Science, North Chicago, IL, 60064, USA
| | - Matt Marion
- Behavioral Neuropharmacology and Neuroimaging Laboratory, Clinical Research Institute On Addictions, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University At Buffalo, Buffalo, NY, 14203, USA
| | - Panayotis K Thanos
- Behavioral Neuropharmacology and Neuroimaging Laboratory, Clinical Research Institute On Addictions, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University At Buffalo, Buffalo, NY, 14203, USA
| | - Heinz Steiner
- Stanson Toshok Center for Brain Function and Repair, Rosalind Franklin University of Medicine and Science, North Chicago, IL, 60064, USA. .,Discipline of Cellular and Molecular Pharmacology, The Chicago Medical School, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Road, North Chicago, IL, 60064, USA.
| |
Collapse
|
10
|
Oliveira-Campos D, Reis HS, Libarino-Santos M, Cata-Preta EG, Dos Santos TB, Dos Anjos-Santos A, Oliveira TS, de Brito ACL, Patti CL, Marinho EAV, de Oliveira Lima AJ. The influence of early exposure to methylphenidate on addiction-related behaviors in mice. Pharmacol Biochem Behav 2021; 206:173208. [PMID: 34022293 DOI: 10.1016/j.pbb.2021.173208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Revised: 04/22/2021] [Accepted: 05/13/2021] [Indexed: 10/21/2022]
Abstract
Methylphenidate (MET) has a putative cognitive enhancer effect that has led adolescents and young adults to increase and indiscriminate its use aiming to ameliorate their productivity. However, the impacts of MET on addiction-related behaviors, emotional levels, and cognition are still not fully understood. To investigate the influence of chronic treatment with MET during adolescence on addiction-like behaviors, memory, and anxiety in adult mice. Thirty-day-old female mice received i.p. 10 mg/kg MET or Veh injections for 10 consecutive days. Forty days after the treatment (mice were 70-days-old), animals were submitted to the behavioral evaluation under the effects of MET, which included: MET-induced conditioned place preference (CPP), behavioral sensitization, and plus-maze discriminative avoidance task. Pre-exposure to MET during adolescence promoted an early expression of CPP and also facilitated the development of MET-induced behavioral sensitization during adulthood. These addictive-like behaviors were accompanied by anxiogenic effects of MET but not by any memory-enhancing effect. We demonstrated that exposure to MET during adolescence can increase the vulnerability to addiction-like behaviors and anxiety during adulthood. Our results reinforce the necessity of a more efficient system to control MET indiscriminate use, thus avoiding its potential tardive addictive effects.
Collapse
Affiliation(s)
| | - Henrique Sousa Reis
- Department of Health Sciences, Universidade Estadual de Santa Cruz (UESC), Ilhéus, BA, Brazil
| | - Matheus Libarino-Santos
- Department of Health Sciences, Universidade Estadual de Santa Cruz (UESC), Ilhéus, BA, Brazil
| | | | | | - Alexia Dos Anjos-Santos
- Department of Health Sciences, Universidade Estadual de Santa Cruz (UESC), Ilhéus, BA, Brazil
| | - Thaynara Silva Oliveira
- Department of Health Sciences, Universidade Estadual de Santa Cruz (UESC), Ilhéus, BA, Brazil
| | | | - Camilla L Patti
- Department of Pharmacology, Universidade Federal de São Paulo (UNIFESP), São Paulo, SP, Brazil
| | | | | |
Collapse
|
11
|
Repantis D, Bovy L, Ohla K, Kühn S, Dresler M. Cognitive enhancement effects of stimulants: a randomized controlled trial testing methylphenidate, modafinil, and caffeine. Psychopharmacology (Berl) 2021; 238:441-451. [PMID: 33201262 PMCID: PMC7826302 DOI: 10.1007/s00213-020-05691-w] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 10/23/2020] [Indexed: 12/14/2022]
Abstract
RATIONAL At all times humans have made attempts to improve their cognitive abilities by different means, among others, with the use of stimulants. Widely available stimulants such as caffeine, but also prescription substances such as methylphenidate and modafinil, are being used by healthy individuals to enhance cognitive performance. OBJECTIVES There is a lack of knowledge on the effects of prescription stimulants when taken by healthy individuals (as compared with patients) and especially on the effects of different substances across different cognitive domains. METHODS We conducted a pilot study with three arms in which male participants received placebo and one of three stimulants (caffeine, methylphenidate, modafinil) and assessed cognitive performance with a test battery that captures various cognitive domains. RESULTS Our study showed some moderate effects of the three stimulants tested. Methylphenidate had positive effects on self-reported fatigue as well as on declarative memory 24 hours after learning; caffeine had a positive effect on sustained attention; there was no significant effect of modafinil in any of the instruments of our test battery. All stimulants were well tolerated, and no trade-off negative effects on other cognitive domains were found. CONCLUSIONS The few observed significant positive effects of the tested stimulants were domain-specific and of rather low magnitude. The results can inform the use of stimulants for cognitive enhancement purposes as well as direct further research to investigate the effects of stimulants on specific cognitive domains that seem most promising, possibly by using tasks that are more demanding.
Collapse
Affiliation(s)
- Dimitris Repantis
- Department of Psychiatry and Psychotherapy, Campus Benjamin Franklin, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Hindenburgdamm 30, 12203, Berlin, Germany.
- Lise Meitner Group for Environmental Neuroscience, Max Planck Institute for Human Development, Berlin, Germany.
| | - Leonore Bovy
- Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Kathrin Ohla
- Institute of Neuroscience and Medicine (INM-3), Jülich Research Centre, Jülich, Germany
| | - Simone Kühn
- Lise Meitner Group for Environmental Neuroscience, Max Planck Institute for Human Development, Berlin, Germany
- Department of Psychiatry and Psychotherapy, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Martin Dresler
- Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
12
|
Edlow BL, Barra ME, Zhou DW, Foulkes AS, Snider SB, Threlkeld ZD, Chakravarty S, Kirsch JE, Chan ST, Meisler SL, Bleck TP, Fins JJ, Giacino JT, Hochberg LR, Solt K, Brown EN, Bodien YG. Personalized Connectome Mapping to Guide Targeted Therapy and Promote Recovery of Consciousness in the Intensive Care Unit. Neurocrit Care 2020; 33:364-375. [PMID: 32794142 PMCID: PMC8336723 DOI: 10.1007/s12028-020-01062-7] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 04/18/2020] [Indexed: 01/05/2023]
Abstract
There are currently no therapies proven to promote early recovery of consciousness in patients with severe brain injuries in the intensive care unit (ICU). For patients whose families face time-sensitive, life-or-death decisions, treatments that promote recovery of consciousness are needed to reduce the likelihood of premature withdrawal of life-sustaining therapy, facilitate autonomous self-expression, and increase access to rehabilitative care. Here, we present the Connectome-based Clinical Trial Platform (CCTP), a new paradigm for developing and testing targeted therapies that promote early recovery of consciousness in the ICU. We report the protocol for STIMPACT (Stimulant Therapy Targeted to Individualized Connectivity Maps to Promote ReACTivation of Consciousness), a CCTP-based trial in which intravenous methylphenidate will be used for targeted stimulation of dopaminergic circuits within the subcortical ascending arousal network (ClinicalTrials.gov NCT03814356). The scientific premise of the CCTP and the STIMPACT trial is that personalized brain network mapping in the ICU can identify patients whose connectomes are amenable to neuromodulation. Phase 1 of the STIMPACT trial is an open-label, safety and dose-finding study in 22 patients with disorders of consciousness caused by acute severe traumatic brain injury. Patients in Phase 1 will receive escalating daily doses (0.5-2.0 mg/kg) of intravenous methylphenidate over a 4-day period and will undergo resting-state functional magnetic resonance imaging and electroencephalography to evaluate the drug's pharmacodynamic properties. The primary outcome measure for Phase 1 relates to safety: the number of drug-related adverse events at each dose. Secondary outcome measures pertain to pharmacokinetics and pharmacodynamics: (1) time to maximal serum concentration; (2) serum half-life; (3) effect of the highest tolerated dose on resting-state functional MRI biomarkers of connectivity; and (4) effect of each dose on EEG biomarkers of cerebral cortical function. Predetermined safety and pharmacodynamic criteria must be fulfilled in Phase 1 to proceed to Phase 2A. Pharmacokinetic data from Phase 1 will also inform the study design of Phase 2A, where we will test the hypothesis that personalized connectome maps predict therapeutic responses to intravenous methylphenidate. Likewise, findings from Phase 2A will inform the design of Phase 2B, where we plan to enroll patients based on their personalized connectome maps. By selecting patients for clinical trials based on a principled, mechanistic assessment of their neuroanatomic potential for a therapeutic response, the CCTP paradigm and the STIMPACT trial have the potential to transform the therapeutic landscape in the ICU and improve outcomes for patients with severe brain injuries.
Collapse
Affiliation(s)
- Brian L Edlow
- Department of Neurology, Center for Neurotechnology and Neurorecovery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, USA.
| | - Megan E Barra
- Department of Neurology, Center for Neurotechnology and Neurorecovery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Pharmacy, Massachusetts General Hospital, Boston, MA, USA
| | - David W Zhou
- Department of Neurology, Center for Neurotechnology and Neurorecovery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Andrea S Foulkes
- Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Samuel B Snider
- Department of Neurology, Center for Neurotechnology and Neurorecovery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Zachary D Threlkeld
- Department of Neurology, Center for Neurotechnology and Neurorecovery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Neurology and Neurological Sciences, Stanford School of Medicine, Stanford, CA, USA
| | - Sourish Chakravarty
- Department of Neurology, Center for Neurotechnology and Neurorecovery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA, USA
- The Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - John E Kirsch
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, USA
| | - Suk-Tak Chan
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, USA
| | - Steven L Meisler
- Department of Neurology, Center for Neurotechnology and Neurorecovery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Thomas P Bleck
- Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Joseph J Fins
- Division of Medical Ethics and Consortium for the Advanced Study of Brain Injury (CASBI), Weill Cornell Medical College, New York, NY, USA
- The Rockefeller University, New York, NY, USA
- Solomon Center for Health Law and Policy, Yale Law School, New Haven, CT, USA
| | - Joseph T Giacino
- Department of Physical Medicine and Rehabilitation, Spaulding Rehabilitation Hospital, Boston, MA, USA
| | - Leigh R Hochberg
- Department of Neurology, Center for Neurotechnology and Neurorecovery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- School of Engineering and Carney Institute for Brain Science, Brown University, Providence, RI, USA
- Veterans Affairs RR&D Center for Neurorestoration and Neurotechnology, VA Medical Center, Providence, RI, USA
| | - Ken Solt
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Emery N Brown
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA, USA
- The Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Yelena G Bodien
- Department of Neurology, Center for Neurotechnology and Neurorecovery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Physical Medicine and Rehabilitation, Spaulding Rehabilitation Hospital, Boston, MA, USA
| |
Collapse
|
13
|
Intrinsic connections between thalamic sub-regions and the lateral prefrontal cortex are differentially impacted by acute methylphenidate. Psychopharmacology (Berl) 2020; 237:1873-1883. [PMID: 32307560 PMCID: PMC7437544 DOI: 10.1007/s00213-020-05505-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Accepted: 03/10/2020] [Indexed: 10/24/2022]
Abstract
BACKGROUND The thalamus is a major target of dopaminergic projections and is densely connected with the prefrontal cortex. A better understanding of how dopamine changes thalamo-cortical communication may shed light on how dopamine supports cognitive function. Methylphenidate has been shown to facilitate cognitive processing and reduce connectivity between the thalamus and lateral prefrontal cortex. AIMS The thalamus is a heterogeneous structure, and the present study sought to clarify how the intrinsic connections of thalamic sub-regions are differentially impacted by acute dopamine transporter blockade. METHODS Sixty healthy volunteers were orally administered either 20 mg of methylphenidate (N = 29) or placebo (N = 31) in a double-blind, randomized, between-subject design. Multi-echo fMRI was used to assess intrinsic functional connectivity of sub-regions of the thalamus during a resting state scan. An N-back working-memory paradigm provided a measure of cognitive performance. RESULTS Acute methylphenidate significantly reduced connectivity of the lateral prefrontal cortex with the motor and somatosensory sub-regions of the thalamus and reduced connectivity with the parietal and visual sub-regions at a trend level. Connectivity with the premotor, prefrontal, and temporal sub-regions was not impacted. The intrinsic connectivity between the thalamus and the lateral prefrontal cortex was not associated with working-memory performance. CONCLUSIONS Methylphenidate decreases functional connections between the lateral prefrontal cortex and thalamus broadly, while sparing intrinsic connectivity with thalamic sub-regions involved with working-memory and language related processes. Collectively, our results suggest that the dopamine transporter regulates functional connections between the prefrontal cortex and non-cognitive areas of the thalamus.
Collapse
|
14
|
Westbrook A, van den Bosch R, Määttä JI, Hofmans L, Papadopetraki D, Cools R, Frank MJ. Dopamine promotes cognitive effort by biasing the benefits versus costs of cognitive work. Science 2020; 367:1362-1366. [PMID: 32193325 DOI: 10.1126/science.aaz5891] [Citation(s) in RCA: 170] [Impact Index Per Article: 42.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2019] [Accepted: 01/20/2020] [Indexed: 12/22/2022]
Abstract
Stimulants such as methylphenidate are increasingly used for cognitive enhancement but precise mechanisms are unknown. We found that methylphenidate boosts willingness to expend cognitive effort by altering the benefit-to-cost ratio of cognitive work. Willingness to expend effort was greater for participants with higher striatal dopamine synthesis capacity, whereas methylphenidate and sulpiride, a selective D2 receptor antagonist, increased cognitive motivation more for participants with lower synthesis capacity. A sequential sampling model informed by momentary gaze revealed that decisions to expend effort are related to amplification of benefit-versus-cost information attended early in the decision process, whereas the effect of benefits is strengthened with higher synthesis capacity and by methylphenidate. These findings demonstrate that methylphenidate boosts the perceived benefits versus costs of cognitive effort by modulating striatal dopamine signaling.
Collapse
Affiliation(s)
- A Westbrook
- Department of Cognitive, Linguistic, and Psychological Sciences, Brown University, Providence, RI, USA. .,Radboud University, Donders Institute for Brain, Cognition and Behaviour, Centre for Cognitive Neuroimaging, Nijmegen, Netherlands.,Radboud University Medical Centre, Department of Psychiatry, Nijmegen, Netherlands
| | - R van den Bosch
- Radboud University, Donders Institute for Brain, Cognition and Behaviour, Centre for Cognitive Neuroimaging, Nijmegen, Netherlands.,Radboud University Medical Centre, Department of Psychiatry, Nijmegen, Netherlands
| | - J I Määttä
- Radboud University, Donders Institute for Brain, Cognition and Behaviour, Centre for Cognitive Neuroimaging, Nijmegen, Netherlands.,Radboud University Medical Centre, Department of Psychiatry, Nijmegen, Netherlands
| | - L Hofmans
- Radboud University, Donders Institute for Brain, Cognition and Behaviour, Centre for Cognitive Neuroimaging, Nijmegen, Netherlands.,Radboud University Medical Centre, Department of Psychiatry, Nijmegen, Netherlands
| | - D Papadopetraki
- Radboud University, Donders Institute for Brain, Cognition and Behaviour, Centre for Cognitive Neuroimaging, Nijmegen, Netherlands.,Radboud University Medical Centre, Department of Psychiatry, Nijmegen, Netherlands
| | - R Cools
- Radboud University, Donders Institute for Brain, Cognition and Behaviour, Centre for Cognitive Neuroimaging, Nijmegen, Netherlands.,Radboud University Medical Centre, Department of Psychiatry, Nijmegen, Netherlands
| | - M J Frank
- Department of Cognitive, Linguistic, and Psychological Sciences, Brown University, Providence, RI, USA.,Carney Institute for Brain Science, Brown University, Providence, RI, USA
| |
Collapse
|
15
|
Neurotoxicity of nanoparticles entering the brain via sensory nerve-to-brain pathways: injuries and mechanisms. Arch Toxicol 2020; 94:1479-1495. [DOI: 10.1007/s00204-020-02701-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Accepted: 03/05/2020] [Indexed: 12/15/2022]
|
16
|
Rățală CE, Fallon SJ, van der Schaaf ME, Ter Huurne N, Cools R, Sanfey AG. Catecholaminergic modulation of trust decisions. Psychopharmacology (Berl) 2019; 236:1807-1816. [PMID: 30706097 PMCID: PMC6602982 DOI: 10.1007/s00213-019-5165-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 01/03/2019] [Indexed: 01/19/2023]
Abstract
RATIONALE Trust is a key component of social interactions. In order to assess the trustworthiness of others, people rely on both information learned from previous encounters, as well as on implicit biases associated with specific facial features. OBJECTIVE Here, we investigated the role of catecholamine (dopamine and noradrenaline) transmission on trust decisions as a function of both experienced behavior and facial features. METHODS To increase catecholamine levels, methylphenidate (MPH, i.e., Ritalin®, 20 mg) was administered to participants (N = 24) prior to their playing a well-studied economic task, namely the Trust Game (Berg et al. 1995). We measured the amount of money invested with a variety of game partners. Across game partners, we manipulated two aspects of trust: the facial trust level (high facial trust, low facial trust, and non-social) and the likelihood of reciprocation (high, low). RESULTS Results demonstrated no main effect of MPH on investments, but rather a selective lowering of investments under MPH as compared with placebo with the game partners who were low on facial trustworthiness and were low reciprocators. CONCLUSION These results provide evidence that MPH administration impacts social trust decision-making, but does so in a context-specific manner.
Collapse
Affiliation(s)
- Cătălina E Rățală
- Rotterdam School of Management, Erasmus University Rotterdam, Rotterdam, 3062, PA, The Netherlands.
- Donders Institute for Brain, Cognition and Behaviour, Centre for Cognitive Neuroimaging, Radboud University, 6500, HB, Nijmegen, The Netherlands.
| | - Sean J Fallon
- Donders Institute for Brain, Cognition and Behaviour, Centre for Cognitive Neuroimaging, Radboud University, 6500, HB, Nijmegen, The Netherlands
| | - Marieke E van der Schaaf
- Donders Institute for Brain, Cognition and Behaviour, Centre for Cognitive Neuroimaging, Radboud University, 6500, HB, Nijmegen, The Netherlands
- Department of Psychiatry, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Niels Ter Huurne
- Donders Institute for Brain, Cognition and Behaviour, Centre for Cognitive Neuroimaging, Radboud University, 6500, HB, Nijmegen, The Netherlands
- Karakter Child and Adolescent Psychiatry University Centre, Nijmegen, The Netherlands
| | - Roshan Cools
- Donders Institute for Brain, Cognition and Behaviour, Centre for Cognitive Neuroimaging, Radboud University, 6500, HB, Nijmegen, The Netherlands
- Department of Psychiatry, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Alan G Sanfey
- Donders Institute for Brain, Cognition and Behaviour, Centre for Cognitive Neuroimaging, Radboud University, 6500, HB, Nijmegen, The Netherlands
- Behavioral Science Institute, Radboud University, Nijmegen, The Netherlands
| |
Collapse
|
17
|
Schrantee A, Bouziane C, Bron EE, Klein S, Bottelier MA, Kooij JJS, Rombouts SARB, Reneman L. Long-term effects of stimulant exposure on cerebral blood flow response to methylphenidate and behavior in attention-deficit hyperactivity disorder. Brain Imaging Behav 2019; 12:402-410. [PMID: 28321605 PMCID: PMC5880865 DOI: 10.1007/s11682-017-9707-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Stimulant prescription rates for attention deficit hyperactivity disorder (ADHD) are increasing, even though potential long-term effects on the developing brain have not been well-studied. A previous randomized clinical trial showed short-term age-dependent effects of stimulants on the DA system. We here assessed the long-term modifying effects of age-of-first-stimulant treatment on the human brain and behavior. 81 male adult ADHD patients were stratified into three groups: 1) early stimulant treatment (EST; <16 years of age) 2) late stimulant treatment (LST: ≥23 years of age) and 3) stimulant treatment naive (STN; no history of stimulant treatment). We used pharmacological magnetic resonance imaging (phMRI) to assess the cerebral blood flow (CBF) response to an oral methylphenidate challenge (MPH, 0.5 mg/kg), as an indirect measure of dopamine function in fronto-striatal areas. In addition, mood and anxiety scores, and recreational drug use were assessed. Baseline ACC CBF was lower in the EST than the STN group (p = 0.03), although CBF response to MPH was similar between the three groups (p = 0.23). ADHD symptom severity was higher in the STN group compared to the other groups (p < 0.01). In addition, the EST group reported more depressive symptoms (p = 0.04), but not anxiety (p = 0.26), and less recreational drug use (p = 0.04). In line with extensive pre-clinical data, our data suggest that early, but not late, stimulant treatment long-lastingly affects the human brain and behavior, possibly indicating fundamental changes in the dopamine system.
Collapse
Affiliation(s)
- Anouk Schrantee
- Department of Radiology, Academic Medical Center, University of Amsterdam, P.O. Box 22660, 1105, AZ, Amsterdam, the Netherlands.
| | - C Bouziane
- Department of Radiology, Academic Medical Center, University of Amsterdam, P.O. Box 22660, 1105, AZ, Amsterdam, the Netherlands
| | - E E Bron
- Biomedical Imaging Group Rotterdam, Departments of Medical Informatics and Radiology, Erasmus MC, P.O. Box 2040, 3000, CA, Rotterdam, The Netherlands
| | - S Klein
- Biomedical Imaging Group Rotterdam, Departments of Medical Informatics and Radiology, Erasmus MC, P.O. Box 2040, 3000, CA, Rotterdam, The Netherlands
| | - M A Bottelier
- Department of Radiology, Academic Medical Center, University of Amsterdam, P.O. Box 22660, 1105, AZ, Amsterdam, the Netherlands.,Department of Child- and Adolescent Psychiatry, Triversum, Kees Boekestraat 5, 1817, EZ, Alkmaar, The Netherlands
| | - J J S Kooij
- Expertise Center Adult ADHD, PsyQ, Psycho-Medical Programs, Carel Reinierszkade 197, 2593, HR, The Hague, The Netherlands
| | - S A R B Rombouts
- Institute of Psychology, Leiden University, P.O. Box 9555, 2300, RB, Leiden, The Netherlands.,Department of Radiology, LUMC, P.O. Box 9600, 2300 RC, Leiden, The Netherlands
| | - L Reneman
- Department of Radiology, Academic Medical Center, University of Amsterdam, P.O. Box 22660, 1105, AZ, Amsterdam, the Netherlands
| |
Collapse
|
18
|
Abstract
Understanding how the brain translates a structured sequence of sounds, such as music, into a pleasant and rewarding experience is a fascinating question which may be crucial to better understand the processing of abstract rewards in humans. Previous neuroimaging findings point to a challenging role of the dopaminergic system in music-evoked pleasure. However, there is a lack of direct evidence showing that dopamine function is causally related to the pleasure we experience from music. We addressed this problem through a double blind within-subject pharmacological design in which we directly manipulated dopaminergic synaptic availability while healthy participants (n = 27) were engaged in music listening. We orally administrated to each participant a dopamine precursor (levodopa), a dopamine antagonist (risperidone), and a placebo (lactose) in three different sessions. We demonstrate that levodopa and risperidone led to opposite effects in measures of musical pleasure and motivation: while the dopamine precursor levodopa, compared with placebo, increased the hedonic experience and music-related motivational responses, risperidone led to a reduction of both. This study shows a causal role of dopamine in musical pleasure and indicates that dopaminergic transmission might play different or additive roles than the ones postulated in affective processing so far, particularly in abstract cognitive activities.
Collapse
|
19
|
Carvallo C, Contreras D, Ugarte G, Delgado R, Pancetti F, Rozas C, Piña R, Constandil L, Zeise ML, Morales B. Single and Repeated Administration of Methylphenidate Modulates Synaptic Plasticity in Opposite Directions via Insertion of AMPA Receptors in Rat Hippocampal Neurons. Front Pharmacol 2018; 9:1485. [PMID: 30618772 PMCID: PMC6305740 DOI: 10.3389/fphar.2018.01485] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 12/04/2018] [Indexed: 01/20/2023] Open
Abstract
Methylphenidate (MPH) is widely used in the treatment of Attention Deficit Hyperactivity Disorder. Several lines of evidence support that MPH can modulate learning and memory processes in different ways including improvement and impairment of test performances. A relevant factor in the efficacy of treatment is whether administration is performed once or several times. In this study we demonstrate opposite effects of MPH on performance of preadolescent rats in the Morris Water Maze test. Animals treated with a single dose (1 mg/kg) performed significantly better compared to controls, while in animals treated with repetitive administration at the same concentration performance was reduced. We found that hippocampal LTP in slices from rats treated with a single dose was increased, while LTP from rats treated with repetitive injections of MPH was lower than in controls. Using Western blot of CA1 areas from potentiated slices of rats treated with a single dose we found a significant increase of phosphorylation at Ser845 of GluA1 subunits, associated to an increased insertion of GluA1-containing AMPARs in the plasma membrane. These receptors were functional, because AMPA-dependent EPSCs recorded on CA1 were enhanced, associated to a significant increase in short-term plasticity. In contrast, CA1 samples from rats injected with MPH during six consecutive days, showed a significant decrease in the phosphorylation at Ser845 of GluA1 subunits associated to a lower insertion of GluA1-containing AMPARs. Accordingly, a reduction of the AMPA-mediated EPSCs and short-term plasticity was also observed. Taken together, our results demonstrate that single and repeated doses with MPH can induce opposite effects at behavioral, cellular, and molecular levels. The mechanisms demonstrated here in preadolescent rats are relevant to understand the effects of this psychostimulant in the treatment of ADHD.
Collapse
Affiliation(s)
- Claudia Carvallo
- Laboratory of Neuroscience, Faculty of Chemistry and Biology, Universidad de Santiago de Chile, Santiago, Chile
| | - Darwin Contreras
- Laboratory of Neuroscience, Faculty of Chemistry and Biology, Universidad de Santiago de Chile, Santiago, Chile
| | - Gonzalo Ugarte
- Laboratory of Neuroscience, Faculty of Chemistry and Biology, Universidad de Santiago de Chile, Santiago, Chile
| | - Ricardo Delgado
- Department of Biology, Faculty of Sciences, Universidad de Chile, Santiago, Chile
| | - Floria Pancetti
- Laboratory of Environmental Neurotoxicology, Faculty of Medicine, Universidad Católica del Norte, Coquimbo, Chile
| | - Carlos Rozas
- Laboratory of Neuroscience, Faculty of Chemistry and Biology, Universidad de Santiago de Chile, Santiago, Chile
| | - Ricardo Piña
- Laboratory of Neuroscience, Faculty of Chemistry and Biology, Universidad de Santiago de Chile, Santiago, Chile
| | - Luis Constandil
- Laboratory of Neuroscience, Faculty of Chemistry and Biology, Universidad de Santiago de Chile, Santiago, Chile
| | - Marc L Zeise
- School of Psychology, Faculty of Humanities, Universidad de Santiago de Chile, Santiago, Chile
| | - Bernardo Morales
- Laboratory of Neuroscience, Faculty of Chemistry and Biology, Universidad de Santiago de Chile, Santiago, Chile
| |
Collapse
|
20
|
Lee NR, Zheng G, Crooks PA, Bardo MT, Dwoskin LP. New Scaffold for Lead Compounds to Treat Methamphetamine Use Disorders. AAPS JOURNAL 2018; 20:29. [PMID: 29427069 DOI: 10.1208/s12248-018-0192-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/28/2017] [Accepted: 01/13/2018] [Indexed: 01/08/2023]
Abstract
Despite increased methamphetamine use worldwide, pharmacotherapies are not available to treat methamphetamine use disorder. The vesicular monoamine transporter-2 (VMAT2) is an important pharmacological target for discovery of treatments for methamphetamine use disorder. VMAT2 inhibition by the natural product, lobeline, reduced methamphetamine-evoked dopamine release, methamphetamine-induced hyperlocomotion, and methamphetamine self-administration in rats. Compared to lobeline, lobelane exhibited improved affinity and selectivity for VMAT2 over nicotinic acetylcholine receptors. Lobelane inhibited neurochemical and behavioral effects of methamphetamine, but tolerance developed to its behavioral efficacy in reducing methamphetamine self-administration, preventing further development. The lobelane analog, R-N-(1,2-dihydroxypropyl)-2,6-cis-di-(4-methoxyphenethyl)piperidine hydrochloride (GZ-793A), potently and selectively inhibited VMAT2 function and reduced neurochemical and behavioral effects of methamphetamine. However, GZ-793A exhibited potential to induce ventricular arrhythmias interacting with human-ether-a-go-go (hERG) channels. Herein, a new lead, R-3-(4-methoxyphenyl)-N-(1-phenylpropan-2-yl)propan-1-amine (GZ-11610), from the novel scaffold (N-alkyl(1-methyl-2-phenylethyl)amine) was evaluated as a VMAT2 inhibitor and potential therapeutic for methamphetamine use disorder. GZ-11610 was 290-fold selective for VMAT2 over dopamine transporters, suggesting that it may lack abuse liability. GZ-11610 was 640- to 3500-fold selective for VMAT2 over serotonin transporters and nicotinic acetylcholine receptors. GZ-11610 exhibited > 1000-fold selectivity for VMAT2 over hERG, representing a robust improvement relative to our previous VMAT2 inhibitors. GZ-11610 (3-30 mg/kg, s.c. or 56-300 mg/kg, oral) reduced methamphetamine-induced hyperactivity in methamphetamine-sensitized rats. Thus, GZ-11610 is a potent and selective inhibitor of VMAT2, may have low abuse liability and low cardiotoxicity, and after oral administration is effective and specific in inhibiting the locomotor stimulant effects of methamphetamine, suggesting further investigation as a potential therapeutic for methamphetamine use disorder.
Collapse
Affiliation(s)
- Na-Ra Lee
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, 465 TODD Building, 789 South Limestone, Lexington, Kentucky, 40536-0596, USA
| | - Guangrong Zheng
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Peter A Crooks
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Michael T Bardo
- Department of Psychology, College of Arts and Sciences, University of Kentucky, Lexington, Kentucky, USA
| | - Linda P Dwoskin
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, 465 TODD Building, 789 South Limestone, Lexington, Kentucky, 40536-0596, USA.
| |
Collapse
|
21
|
Strand MC, Mørland J, Slørdal L, Riedel B, Innerdal C, Aamo T, Mathisrud G, Vindenes V. Conversion factors for assessment of driving impairment after exposure to multiple benzodiazepines/z-hypnotics or opioids. Forensic Sci Int 2017; 281:29-36. [PMID: 29101905 DOI: 10.1016/j.forsciint.2017.10.022] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2017] [Revised: 09/06/2017] [Accepted: 10/11/2017] [Indexed: 11/18/2022]
Abstract
AIMS Norway has introduced legal concentration limits in blood for 28 non-alcohol drugs in driving under the influence cases. As of 2016 this legislation also regulates the assessment of combined effects of multiple benzodiazepines and opioids. We herein describe the employed methodology for the equivalence tables for concentrations of benzodiazepines/z-hypnotics and opioids implemented in the Norwegian Road Traffic Act. METHODS Legislative limits corresponding to impairment at blood alcohol concentrations (BAC) of 0.02%, 0.05% and 0.12% were established for 15 different benzodiazepines and opioids. This was based on a concept of a linear relationship between blood drug concentration and impairment in drug naïve users. Concentration ratios between these drugs were used to establish conversion factors and calculate net impairment using diazepam and morphine equivalents. RESULTS Conversion factors were established for 14 benzodiazepines/z-hypnotics (alprazolam, bromazepam, clobazam, clonazepam, etizolam, flunitrazepam, lorazepam, nitrazepam, nordiazepam, oxazepam, phenazepam, temazepam, zolpidem and zopiclone) and two opioids (methadone and oxycodone). CONCLUSIONS Conversion factors to calculate diazepam and morphine equivalents for benzodiazepines/z-hypnotics and selected opioids, respectively, have been operative in the Norwegian Road Traffic Act as of February 2016. Calculated equivalents can be applied by the courts to meter out sanctions.
Collapse
Affiliation(s)
| | - Jørg Mørland
- Norwegian Institute of Public Health, Division of Health Data and Digitalization, Oslo, Norway.
| | - Lars Slørdal
- Department of Laboratory Medicine, Children's and Women's Health, Norwegian University of Science and Technology, Trondheim, Norway; Department of Clinical Pharmacology, St. Olav University Hospital, Trondheim, Norway.
| | - Bettina Riedel
- University of Bergen, Faculty of Medicine and Dentistry, Department of Clinical Science, Bergen, Norway; Haukeland University Hospital, Laboratory of Clinical Biochemistry, Bergen, Norway.
| | | | - Trond Aamo
- Department of Clinical Pharmacology, St. Olav University Hospital, Trondheim, Norway.
| | - Grete Mathisrud
- Norwegian Ministry of Transport and Communications, Department of Public Roads and Traffic Safety, Oslo, Norway.
| | - Vigdis Vindenes
- Oslo University Hospital, Department of Forensic Medicine, Oslo, Norway; Center of Drug and Addiction Research, Faculty of Medicine, University of Oslo, Norway.
| |
Collapse
|
22
|
Verbeeck W, Bekkering GE, Van den Noortgate W, Kramers C. Bupropion for attention deficit hyperactivity disorder (ADHD) in adults. Cochrane Database Syst Rev 2017; 10:CD009504. [PMID: 28965364 PMCID: PMC6485546 DOI: 10.1002/14651858.cd009504.pub2] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
BACKGROUND Attention deficit hyperactivity disorder (ADHD) is a prevalent neurobiological condition, characterised by behavioral and cognitive symptoms such as inattention, impulsivity and/or excessive activity. The syndrome is commonly accompanied by psychiatric comorbidities and is associated with educational and occupational underachievement.Although psychostimulant medications are the mainstay of treatment for ADHD, not all adults respond optimally to, or can tolerate, these medicines. Thus, alternative non-stimulant treatment approaches for ADHD have been explored. One of these alternatives is bupropion, an aminoketone antidepressant and non-competitive antagonism of nicotinic acetylcholine receptors. Bupropion is registered for the treatment of depression and smoking cessation, but is also used off-label to treat ADHD. OBJECTIVES To assess the effects and safety of bupropion for the treatment of adults with ADHD. SEARCH METHODS We searched the Cochrane Central Register of Controlled Trials (CENTRAL), MEDLINE, Embase, and seven other databases in February 2017. We also searched three trials registers and three online theses portals. In addition, we checked references of included studies and contacted study authors to identify potentially relevant studies that were missed by our search. SELECTION CRITERIA We included all randomised controlled trials (RCTs) that evaluated the effects (including adverse effects) of bupropion compared to placebo in adults with ADHD. DATA COLLECTION AND ANALYSIS Two review authors (WV, GB) independently screened records and extracted data using a data extraction sheet that we tested in a pilot study. We extracted all relevant data on study characteristics and results. We assessed risks of bias using the Cochrane 'Risk of bias' tool, and assessed the overall quality of evidence using the GRADE approach. We used a fixed-effect model to pool the results across studies. MAIN RESULTS We included six studies with a total of 438 participants. Five studies were conducted in the USA, and one in Iran. All studies evaluated a long-acting version of bupropion, with the dosage ranging from 150 mg up to 450 mg daily. Study intervention length varied from six to 10 weeks. Four studies explicitly excluded participants with psychiatric comorbidity and one study included only participants with opioid dependency. Four studies were funded by industry, but the impact of this on study results is unknown. Two studies were publicly funded and in one of these studies, the lead author was a consultant for several pharmaceutical companies and also received investigator-driven funding from two companies, however none of these companies manufacture bupropion. We judged none of the studies to be free of bias because for most risk of bias domains the study reports failed to provide sufficient details. Using the GRADE approach, we rated the overall quality of evidence as low. We downgraded the quality of the evidence because of serious risk of bias and serious imprecision due to small sample sizes.We found low-quality evidence that bupropion decreased the severity of ADHD symptoms (standardised mean difference -0.50, 95% confidence interval (CI) -0.86 to -0.15, 3 studies, 129 participants), and increased the proportion of participants achieving clinical improvement (risk ratio (RR) 1.50, 95% CI 1.13 to 1.99, 4 studies, 315 participants), and reporting an improvement on the Clinical Global Impression - Improvement scale (RR 1.78, 95% CI 1.27 to 2.50, 5 studies, 337 participants). There was low-quality evidence that the proportion of participants who withdrew due to any adverse effect was similar in the bupropion and placebo groups (RR 1.20, 95% CI 0.35 to 4.10, 3 studies, 253 participants). The results were very similar when using a random-effects model and when we analysed only studies that excluded participants with a psychiatric comorbidity. AUTHORS' CONCLUSIONS The findings of this review, which compared bupropion to placebo for adult ADHD, indicate a possible benefit of bupropion. We found low-quality evidence that bupropion decreased the severity of ADHD symptoms and moderately increased the proportion of participants achieving a significant clinical improvement in ADHD symptoms. Furthermore, we found low-quality evidence that the tolerability of bupropion is similar to that of placebo. In the pharmacological treatment of adults with ADHD, extended- or sustained-release bupropion may be an alternative to stimulants. The low-quality evidence indicates uncertainty with respect to the pooled effect estimates. Further research is very likely to change these estimates. More research is needed to reach more definite conclusions as well as clarifying the optimal target population for this medicine. Treatment response remains to be reported in a DSM5-diagnosed population. There is also a lack of knowledge on long-term outcomes.
Collapse
Affiliation(s)
- Wim Verbeeck
- Centrum ADHD/ASS, GGZ Vincent van Gogh Instituut Venray, Noordsingel 39, Venray, Netherlands, 5801 GJ
| | | | | | | |
Collapse
|
23
|
Ethanol Interactions With Dexmethylphenidate and dl-Methylphenidate Spheroidal Oral Drug Absorption Systems in Healthy Volunteers. J Clin Psychopharmacol 2017; 37:419-428. [PMID: 28590363 PMCID: PMC5484776 DOI: 10.1097/jcp.0000000000000721] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
BACKGROUND/PURPOSE Ethanol coadministered with immediate-release dl-methylphenidate (dl-MPH) or dexmethylphenidate (d-MPH) significantly increases the geomean maximum plasma concentration (Cmax) of d-MPH 22% and 15%, respectively, and elevates overall drug exposure and psychostimulant effects. We asked the question: Are these ethanol-MPH interactions based more fundamentally on (1) inhibition of postabsorption d-MPH metabolism or (2) acceleration of MPH formulation gastric dissolution by ethanol in the stomach? This was investigated using the pulsatile, distinctly biphasic, spheroidal oral drug absorption systems of dl-MPH and d-MPH. METHODS In a randomized, 4-way crossover study, 14 healthy subjects received pulsatile dl-MPH (40 mg) or d-MPH (20 mg), with or without ethanol (0.6 g/kg), dosed 4 hours later. These 4 hours allowed the delayed-release second MPH pulse to reach a more distal region of the gut to preclude gastric biopharmaceutical influences. Plasma was analyzed using a highly sensitive chiral method. Subjective/physiological effects were recorded. FINDINGS/RESULTS Ethanol increased the second pulse of d-MPH Cmax for dl-MPH by 35% (P < 0.01) and the partial area under the plasma concentration curve from 4 to 8 hours by 25% (P < 0.05). The respective values for enantiopure d-MPH were 27% (P = 0.001) and 20% (P < 0.01). The carboxylesterase 1-mediated transesterification metabolite ethylphenidate served as a biomarker for coexposure. Ethanol significantly potentiated stimulant responses to either formulation. IMPLICATIONS/CONCLUSIONS These findings support drug dispositional interactions between ethanol and MPH as dominant over potential biopharmaceutical considerations. Understanding the pharmacology underlying the frequent coabuse of MPH-ethanol provides rational guidance in the selection of first-line pharmacotherapy for comorbid attention-deficit/hyperactivity disorder-alcohol use disorder.
Collapse
|
24
|
Naltrexone modulates dopamine release following chronic, but not acute amphetamine administration: a translational study. Transl Psychiatry 2017; 7:e1104. [PMID: 28440810 PMCID: PMC5416714 DOI: 10.1038/tp.2017.79] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Accepted: 03/08/2017] [Indexed: 01/16/2023] Open
Abstract
The opioid antagonist naltrexone has been shown to attenuate the subjective effects of amphetamine. However, the mechanisms behind this modulatory effect are currently unknown. We hypothesized that naltrexone would diminish the striatal dopamine release induced by amphetamine, which is considered an important mechanism behind many of its stimulant properties. We used positron emission tomography and the dopamine D2-receptor radioligand [11C]raclopride in healthy subjects to study the dopaminergic effects of an amphetamine injection after pretreatment with naltrexone or placebo. In a rat model, we used microdialysis to study the modulatory effects of naltrexone on dopamine levels after acute and chronic amphetamine exposure. In healthy humans, naltrexone attenuated the subjective effects of amphetamine, confirming our previous results. Amphetamine produced a significant reduction in striatal radioligand binding, indicating increased levels of endogenous dopamine. However, there was no statistically significant effect of naltrexone on dopamine release. The same pattern was observed in rats, where an acute injection of amphetamine caused a significant rise in striatal dopamine levels, with no effect of naltrexone pretreatment. However, in a chronic model, naltrexone significantly attenuated the dopamine release caused by reinstatement of amphetamine. Collectively, these data suggest that the opioid system becomes engaged during the more chronic phase of drug use, evidenced by the modulatory effect of naltrexone on dopamine release following chronic amphetamine administration. The importance of opioid-dopamine interactions in the reinforcing and addictive effects of amphetamine is highlighted by the present findings and may help to facilitate medication development in the field of stimulant dependence.
Collapse
|
25
|
Wagner IC, van Buuren M, Bovy L, Morris RG, Fernández G. Methylphenidate during early consolidation affects long-term associative memory retrieval depending on baseline catecholamines. Psychopharmacology (Berl) 2017; 234:657-669. [PMID: 28013352 PMCID: PMC5263224 DOI: 10.1007/s00213-016-4502-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Accepted: 12/05/2016] [Indexed: 12/03/2022]
Abstract
RATIONALE Synaptic memory consolidation is thought to rely on catecholaminergic signaling. Eventually, it is followed by systems consolidation, which embeds memories in a neocortical network. Although this sequence was demonstrated in rodents, it is unclear how catecholamines affect memory consolidation in humans. OBJECTIVES Here, we tested the effects of catecholaminergic modulation on synaptic and subsequent systems consolidation. We expected enhanced memory performance and increased neocortical engagement during delayed retrieval. Additionally, we tested if this effect was modulated by individual differences in a cognitive proxy measure of baseline catecholamine synthesis capacity. METHODS Fifty-three healthy males underwent a between-subjects, double-blind, placebo-controlled procedure across 2 days. On day 1, subjects studied and retrieved object-location associations and received 20 mg of methylphenidate or placebo. Drug intake was timed so that methylphenidate was expected to affect early consolidation but not encoding or retrieval. Memory was tested again while subjects were scanned three days later. RESULTS Methylphenidate did not facilitate memory performance, and there was no significant group difference in activation during delayed retrieval. However, memory representations differed between groups depending on baseline catecholamines. The placebo group showed increased activation in occipito-temporal regions but decreased connectivity with the hippocampus, associated with lower baseline catecholamine synthesis capacity. The methylphenidate group showed stronger activation in the postcentral gyrus, associated with higher baseline catecholamine synthesis capacity. CONCLUSIONS Altogether, methylphenidate during early consolidation did not foster long-term memory performance, but it affected retrieval-related neural processes depending on individual levels of baseline catecholamines.
Collapse
Affiliation(s)
- Isabella C Wagner
- Donders Institute for Brain, Cognition and Behaviour, Radboud University Nijmegen Medical Center, Kapittelweg 29, Nijmegen, 6525 EN, The Netherlands.
| | - Mariët van Buuren
- Donders Institute for Brain, Cognition and Behaviour, Radboud University Nijmegen Medical Center, Kapittelweg 29, Nijmegen, 6525 EN, The Netherlands
| | - Leonore Bovy
- Donders Institute for Brain, Cognition and Behaviour, Radboud University Nijmegen Medical Center, Kapittelweg 29, Nijmegen, 6525 EN, The Netherlands
| | - Richard G Morris
- Centre for Cognitive and Neural Systems, University of Edinburgh, Edinburgh, EH8 9JZ, UK
| | - Guillén Fernández
- Donders Institute for Brain, Cognition and Behaviour, Radboud University Nijmegen Medical Center, Kapittelweg 29, Nijmegen, 6525 EN, The Netherlands
| |
Collapse
|
26
|
Schrantee A, Mutsaerts HJMM, Bouziane C, Tamminga HGH, Bottelier MA, Reneman L. The age-dependent effects of a single-dose methylphenidate challenge on cerebral perfusion in patients with attention-deficit/hyperactivity disorder. Neuroimage Clin 2016; 13:123-129. [PMID: 27942455 PMCID: PMC5137172 DOI: 10.1016/j.nicl.2016.11.021] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Revised: 11/18/2016] [Accepted: 11/19/2016] [Indexed: 10/26/2022]
Abstract
Methylphenidate (MPH) is a stimulant drug and an effective treatment for attention-deficit/hyperactivity disorder (ADHD) in both children and adults. Pre-clinical studies suggest that the response to stimulants is dependent on age, which may reflect the ontogeny of the dopamine (DA) system, which continues to develop throughout childhood and adolescence. Therefore, the aim of this study was to investigate the modulating effect of age on the cerebral blood flow (CBF) response to MPH in stimulant treatment-naive children and adults with ADHD. Ninety-eight stimulant treatment-naive male pediatric (10-12 years) and adult (23-40 years) patients with ADHD were included in this study. The CBF response to an acute challenge with MPH (0.5 mg/kg) was measured using arterial spin labeling (ASL) pharmacological magnetic resonance imaging, as a proxy for DA function. Region-of-interest (ROI) analyses were carried out for the striatum, thalamus and medial prefrontal cortex and in addition voxel-wise analyses were conducted. An acute challenge with MPH decreased CBF in both children and adults in cortical areas, although to a greater extent in adults. In contrast, ROI analyses showed that MPH decreased thalamic CBF only in children, but not adults. Our findings highlight the importance of taking the developmental perspective into account when studying the effects of stimulants in ADHD patients.
Collapse
Affiliation(s)
- A Schrantee
- Department of Radiology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Brain and Cognition, University of Amsterdam, Amsterdam, The Netherlands
| | - HJMM Mutsaerts
- Department of Radiology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
- Sunnybrook Research Institute, University of Toronto, Toronto, ON, Canada
| | - C Bouziane
- Department of Radiology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - HGH Tamminga
- Department of Radiology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
- Department of Psychology, University of Amsterdam, Amsterdam, The Netherlands
| | - MA Bottelier
- Department of Child- and Adolescent Psychiatry, Triversum, Alkmaar, The Netherlands
| | - L Reneman
- Department of Radiology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Brain and Cognition, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
27
|
Hung CL, Huang CJ, Tsai YJ, Chang YK, Hung TM. Neuroelectric and Behavioral Effects of Acute Exercise on Task Switching in Children with Attention-Deficit/Hyperactivity Disorder. Front Psychol 2016; 7:1589. [PMID: 27790182 PMCID: PMC5062018 DOI: 10.3389/fpsyg.2016.01589] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2016] [Accepted: 09/29/2016] [Indexed: 11/17/2022] Open
Abstract
The main purpose of this two-part study was to examine the effects of acute, moderate intensity exercise on task switching in children with attention-deficit/hyperactivity disorder (ADHD). In Study 1, we compared the task switching performance of children with and without ADHD. Twenty children with ADHD and 20 matched controls performed the task switching paradigm, in which the behavioral indices and P3 component of event-related potentials elicited by task-switching were assessed simultaneously. The amplitude and latency of P3 reflected the amount of attention resource allocated to task-relevant stimulus in the environment and the efficiency of stimulus detection and evaluation, respectively. The task switching included two conditions; the pure condition required participants to perform the task on the same rule (e.g., AAAA or BBBB) whereas the mixed condition required participants to perform the task on two alternating rules (e.g., AABBAA…). The results indicated that children with ADHD had significantly longer RTs, less accuracy, and larger global switch cost for accuracy than controls. Additionally, ADHD participants showed smaller amplitudes and longer P3 latencies in global switch effects. In Study 2, we further examined the effects of an acute aerobic exercise session on task switching in children with ADHD. Thirty-four children with ADHD performed a task switching paradigm after 30 min of moderate-intensity aerobic exercise on a treadmill and after control sessions (watching videos while seated). The results revealed that following exercise, children with ADHD exhibited smaller global switch costs in RT compared with after control sessions. The P3 amplitude only increased following exercise in the mixed condition relative to the pure condition, whereas no effects were found in the control session. These findings suggest that single bouts of moderate intensity aerobic exercise may have positive effects on the working memory of children with ADHD.
Collapse
Affiliation(s)
- Chiao-Ling Hung
- Department of Athletic, National Taiwan University Taipei, Taiwan
| | - Chung-Ju Huang
- Graduate Institute of Sport Pedagogy, University of Taipei Taipei, Taiwan
| | - Yu-Jung Tsai
- Department of Physical Education, National Taiwan Normal University Taipei, Taiwan
| | - Yu-Kai Chang
- Graduate Institute of Athletics and Coaching Science, National Taiwan Sport University Taoyuan, Taiwan
| | - Tsung-Min Hung
- Department of Physical Education, National Taiwan Normal University Taipei, Taiwan
| |
Collapse
|
28
|
Methylphenidate does not enhance visual working memory but benefits motivation in macaque monkeys. Neuropharmacology 2016; 109:223-235. [DOI: 10.1016/j.neuropharm.2016.06.019] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Revised: 06/14/2016] [Accepted: 06/17/2016] [Indexed: 02/04/2023]
|
29
|
Schrantee A, Tamminga HGH, Bouziane C, Bottelier MA, Bron EE, Mutsaerts HJMM, Zwinderman AH, Groote IR, Rombouts SARB, Lindauer RJL, Klein S, Niessen WJ, Opmeer BC, Boer F, Lucassen PJ, Andersen SL, Geurts HM, Reneman L. Age-Dependent Effects of Methylphenidate on the Human Dopaminergic System in Young vs Adult Patients With Attention-Deficit/Hyperactivity Disorder: A Randomized Clinical Trial. JAMA Psychiatry 2016; 73:955-62. [PMID: 27487479 PMCID: PMC5267166 DOI: 10.1001/jamapsychiatry.2016.1572] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
IMPORTANCE Although numerous children receive methylphenidate hydrochloride for the treatment of attention-deficit/hyperactivity disorder (ADHD), little is known about age-dependent and possibly lasting effects of methylphenidate on the human dopaminergic system. OBJECTIVES To determine whether the effects of methylphenidate on the dopaminergic system are modified by age and to test the hypothesis that methylphenidate treatment of young but not adult patients with ADHD induces lasting effects on the cerebral blood flow response to dopamine challenge, a noninvasive probe for dopamine function. DESIGN, SETTING, AND PARTICIPANTS A randomized, double-blind, placebo-controlled trial (Effects of Psychotropic Drugs on Developing Brain-Methylphenidate) among ADHD referral centers in the greater Amsterdam area in the Netherlands between June 1, 2011, and June 15, 2015. Additional inclusion criteria were male sex, age 10 to 12 years or 23 to 40 years, and stimulant treatment-naive status. INTERVENTIONS Treatment with either methylphenidate or a matched placebo for 16 weeks. MAIN OUTCOMES AND MEASURES Change in the cerebral blood flow response to an acute challenge with methylphenidate, noninvasively assessed using pharmacological magnetic resonance imaging, between baseline and 1 week after treatment. Data were analyzed using intent-to-treat analyses. RESULTS Among 131 individuals screened for eligibility, 99 patients met DSM-IV criteria for ADHD, and 50 participants were randomized to receive methylphenidate and 49 to placebo. Sixteen weeks of methylphenidate treatment increased the cerebral blood flow response to methylphenidate within the thalamus (mean difference, 6.5; 95% CI, 0.4-12.6; P = .04) of children aged 10 to 12 years old but not in adults or in the placebo group. In the striatum, the methylphenidate condition differed significantly from placebo in children but not in adults (mean difference, 7.7; 95% CI, 0.7-14.8; P = .03). CONCLUSIONS AND RELEVANCE We confirm preclinical data and demonstrate age-dependent effects of methylphenidate treatment on human extracellular dopamine striatal-thalamic circuitry. Given its societal relevance, these data warrant replication in larger groups with longer follow-up. TRIAL REGISTRATION identifier: NL34509.000.10 and trialregister.nl identifier: NTR3103.
Collapse
Affiliation(s)
- Anouk Schrantee
- Department of Radiology, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands2Brain Imaging Center, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands3Amsterdam Brain and Cognition, University of Amsterdam, Amsterdam, the Netherlands
| | - Hyke G. H. Tamminga
- Department of Radiology, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands4d’Arc (Dutch Autism and Attention-Deficit/Hyperactivity Disorder Research Center), Department of Brain and Cognition, University of Amsterdam, Amsterdam, the Netherlands
| | - Cheima Bouziane
- Department of Radiology, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands2Brain Imaging Center, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Marco A. Bottelier
- Department of Radiology, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands5Department of Child and Adolescent Psychiatry, Triversum, Alkmaar, the Netherlands
| | - Esther E. Bron
- Biomedical Imaging Group Rotterdam, Department of Medical Informatics, Erasmus Medical Center, Rotterdam, the Netherlands7Biomedical Imaging Group Rotterdam, Department of Radiology, Erasmus MC, Rotterdam, the Netherlands
| | - Henk-Jan M. M. Mutsaerts
- Department of Radiology, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands2Brain Imaging Center, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Aeilko H. Zwinderman
- Department of Clinical Epidemiology and Biostatistics, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Inge R. Groote
- Institute of Psychology, Department of Social Sciences, University of Oslo, Oslo, Norway
| | - Serge A. R. B. Rombouts
- Institute of Psychology, Leiden University, Leiden, the Netherlands11Department of Radiology, Leids Universitair Medisch Centrum, Leiden, the Netherlands
| | - Ramon J. L. Lindauer
- Department of Child and Adolescent Psychiatry, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands13De Bascule Academic Center for Child and Adolescent Psychiatry, Amsterdam, the Netherlands
| | - Stefan Klein
- Biomedical Imaging Group Rotterdam, Department of Medical Informatics, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Wiro J. Niessen
- Biomedical Imaging Group Rotterdam, Department of Medical Informatics, Erasmus Medical Center, Rotterdam, the Netherlands14Department of Imaging Physics, Faculty of Applied Sciences, Delft University of Technology, Delft, the Netherlands
| | - Brent C. Opmeer
- Clinical Research Unit, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Frits Boer
- Department of Child and Adolescent Psychiatry, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands13De Bascule Academic Center for Child and Adolescent Psychiatry, Amsterdam, the Netherlands
| | - Paul J. Lucassen
- Swammerdam Institute for Life Sciences, Center for Neuroscience, University of Amsterdam, Amsterdam, the Netherlands
| | - Susan L. Andersen
- Department of Psychiatry, Harvard Medical School and McLean Hospital, Belmont, Massachusetts
| | - Hilde M. Geurts
- d’Arc (Dutch Autism and Attention-Deficit/Hyperactivity Disorder Research Center), Department of Brain and Cognition, University of Amsterdam, Amsterdam, the Netherlands
| | - Liesbeth Reneman
- Department of Radiology, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands2Brain Imaging Center, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands3Amsterdam Brain and Cognition, University of Amsterdam, Amsterdam, the Netherlands
| |
Collapse
|
30
|
Abstract
Children/adolescents with attention-deficit/hyperactivity disorder (ADHD) may have a poor or inadequate response to psychostimulants or be unable to tolerate their side-effects; furthermore, stimulants may be inappropriate because of co-existing conditions. Only one non-stimulant ADHD pharmacotherapy, the noradrenaline transporter inhibitor atomoxetine, is currently approved for use in Europe. We review recent advances in understanding of the pathophysiology of ADHD with a focus on the roles of catecholamine receptors in context of the α2A-adrenergic receptor agonist guanfacine extended release (GXR), a new non-stimulant treatment option in Europe. Neuroimaging studies of children/adolescents with ADHD show impaired brain maturation, and structural and functional anomalies in brain regions and networks. Neurobiological studies in ADHD and medication response patterns support involvement of monoaminergic neurotransmitters (primarily dopamine and noradrenaline). Guanfacine is a selective α2A-adrenergic receptor agonist that has been shown to improve prefrontal cortical cognitive function, including working memory. The hypothesized mode of action of guanfacine centres on direct stimulation of post-synaptic α2A-adrenergic receptors to enhance noradrenaline neurotransmission. Preclinical data suggest that guanfacine also influences dendritic spine growth and maturation. Clinical trials have demonstrated the efficacy of GXR in ADHD, and it is approved as monotherapy or adjunctive therapy to stimulants in Canada and the USA (for children and adolescents). GXR was approved recently in Europe for the treatment of ADHD in children and adolescents for whom stimulants are not suitable, not tolerated or have been shown to be ineffective. GXR may provide particular benefit for children/adolescents who have specific co-morbidities such as chronic tic disorders or oppositional defiant disorder (or oppositional symptoms) that have failed to respond to first-line treatment options.
Collapse
|
31
|
Batistela S, Bueno OFA, Vaz LJ, Galduróz JCF. Methylphenidate as a cognitive enhancer in healthy young people. Dement Neuropsychol 2016; 10:134-142. [PMID: 29213444 PMCID: PMC5642404 DOI: 10.1590/s1980-5764-2016dn1002009] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The so-called cognitive enhancers have been widely and increasingly used by healthy individuals who seek improvements in cognitive performance despite having no pathologies. One drug used for this purpose is methylphenidate, a first-line drug for the treatment of attention deficit hyperactivity disorder (ADHD). Objective The aim of the present study was to test the effect of acute administration of varying doses of methylphenidate (10 mg, 20 mg, 40 mg and placebo) on a wide range of cognitive functions in healthy young people. Methods A total of 36 young university students and graduates participated in the study. The participants underwent tests of attention and of episodic, and working memory. Results No differences in performance were observed on any of the tests. There was a dose-dependent (40 mg > placebo) effect on self-reported wellbeing. Conclusions According to the recent literature, psychostimulant medications, such as methylphenidate, improve performance when cognitive processes are below an optimal level, which was not the case for the subjects of the present study. We suggest the impression that methylphenidate enhances cognitive performance in healthy young people, justifying its use, may be due to improvements in subjective wellbeing promoted by the drug.
Collapse
Affiliation(s)
- Silmara Batistela
- MS, Departamento de Psicobiologia - Universidade Federal de São Paulo, São Paulo SP, Brazil
| | | | - Leonardo José Vaz
- PhD,Departamento de Psicobiologia - Universidade Federal de São Paulo, São Paulo SP, Brazil
| | | |
Collapse
|
32
|
Koblan KS, Hopkins SC, Sarma K, Gallina N, Jin F, Levy-Cooperman N, Schoedel KA, Loebel A. Assessment of human abuse potential of dasotraline compared to methylphenidate and placebo in recreational stimulant users. Drug Alcohol Depend 2016; 159:26-34. [PMID: 26794682 DOI: 10.1016/j.drugalcdep.2015.10.029] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Revised: 10/06/2015] [Accepted: 10/07/2015] [Indexed: 10/22/2022]
Abstract
AIMS The aim of this study was to evaluate the abuse potential of dasotraline, a novel dopamine and norepinephrine reuptake inhibitor with slow absorption (tmax, 10-12h) and elimination (t1/2=47-77 h) that is in development for the treatment of attention deficit hyperactivity disorder (ADHD). METHODS Recreational stimulant users (N=48) who had specific experience with cocaine, and who were able to distinguish methylphenidate (60 mg) versus placebo in a qualification session, were randomized, in a 6-period, double-blind, crossover design, to receive single doses of dasotraline 8 mg, 16 mg, and 36 mg, methylphenidate (MPH) 40 mg and 80 mg, and placebo. The primary endpoint was the Drug Liking Visual Analog Scale (VAS) score at the time of peak effect (Emax). RESULTS There were no significant differences between the 3 doses of dasotraline and placebo on the drug liking VAS at Emax, and on most secondary endpoints. Both doses of MPH had significantly higher VAS-drug liking scores at Emax relative to both placebo (P<0.001 for all comparisons) and dasotraline 8 mg (P<0.001), 16 mg (P<0.001) and 36 mg (P<0.01). The increase in heart rate for MPH and dasotraline 36 mg showed a time-course that closely matched subject-rated measures such as Any Effects VAS. CONCLUSIONS In this study, dasotraline was found to have low potential for abuse, which may be, in part, related to its established pharmacokinetics (PK) profile, which is characterized by slow absorption and gradual elimination.
Collapse
Affiliation(s)
- K S Koblan
- Sunovion Pharmaceuticals Inc., Marlborough, MA, USA.
| | - S C Hopkins
- Sunovion Pharmaceuticals Inc., Marlborough, MA, USA
| | - K Sarma
- Sunovion Pharmaceuticals Inc., Marlborough, MA, USA
| | - N Gallina
- Sunovion Pharmaceuticals Inc., Marlborough, MA, USA
| | - F Jin
- Sunovion Pharmaceuticals Inc., Marlborough, MA, USA
| | | | | | - A Loebel
- Sunovion Pharmaceuticals Inc., Marlborough, MA, USA
| |
Collapse
|
33
|
Comparative Ethanol-Induced Potentiation of Stimulatory Responses to Dexmethylphenidate Versus Methylphenidate. J Clin Psychopharmacol 2015; 35:464-7. [PMID: 26075488 PMCID: PMC4485535 DOI: 10.1097/jcp.0000000000000348] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
The potentiation of positive subjective responses to immediate-release dexmethylphenidate (d-MPH) or dl-methylphenidate (dl-MPH) by ethanol was investigated over the time course of maximal drug exposure after a single dose. In a 4-way, randomized, crossover study design, 12 men and 12 women normal volunteers received d-MPH (0.15 mg/kg) or dl-MPH (0.3 mg/kg) with or without ethanol (0.6 g/kg). Serial visual analog scales were used as surrogates for drug abuse liability ("high," "good," "like," "stimulated," and "any drug effect"). Combining pure d-MPH with ethanol significantly (P < 0.005) increased the area under the effect curves (AUC(0-5.25h)) of all 5 subscales. The dl-MPH-ethanol combination significantly (P < 0.05) increased these AUCs with the exception of like (P = 0.08). Effects of the pure d-MPH-ethanol combination exhibited delayed potentiation relative to dl-MPH-ethanol. A pharmacokinetic interaction between the l-isomer of dl-MPH and ethanol has previously been shown to increase early exposure to d-MPH. Administration of the pure isomer d-MPH precludes this absorption phase pharmacokinetic interaction with ethanol. This notwithstanding, the pure d-MPH-ethanol combination resulted in comparable, if not greater, cumulative stimulant potentiation than the dl-MPH-ethanol combination. These findings provide evidence of a pharmacodynamic component to d-MPH-ethanol synergistic interactions and carry implications for the rational drug individualization in the treatment of attention-deficit/hyperactivity disorder.
Collapse
|
34
|
Rozas C, Carvallo C, Contreras D, Carreño M, Ugarte G, Delgado R, Zeise ML, Morales B. Methylphenidate amplifies long-term potentiation in rat hippocampus CA1 area involving the insertion of AMPA receptors by activation of β-adrenergic and D1/D5 receptors. Neuropharmacology 2015; 99:15-27. [PMID: 26165920 DOI: 10.1016/j.neuropharm.2015.07.003] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Revised: 07/02/2015] [Accepted: 07/04/2015] [Indexed: 12/01/2022]
Abstract
Methylphenidate (MPH, Ritalin©) is widely used in the treatment of Attention Deficit Hyperactivity Disorder and recently as a drug of abuse. Although the effect of MPH has been studied in brain regions such as striatum and prefrontal cortex (PFC), the hippocampus has received relatively little attention. It is known that MPH increases the TBS-dependent Long Term Potentiation (LTP) in the CA1 area. However, the cellular and molecular mechanisms involved in this process are still unknown. Using field potential recordings and western blot analysis in rat hippocampal slices of young rats, we found that acute application of MPH enhances LTP in CA3-CA1 synapses in a dose-dependent manner with an EC50 of 73.44±6.32 nM. Using specific antagonists and paired-pulse facilitation protocols, we observed that the MPH-dependent increase of LTP involves not only β-adrenergic receptors activation but also post-synaptic D1/D5 dopamine receptors. The inhibition of PKA with PKI, suppressed the facilitation of LTP induced by MPH consistent with an involvement of the adenyl cyclase-cAMP-PKA dependent cascade downstream of the activation of D1/D5 receptors. In addition, samples of CA1 areas taken from slices potentiated with MPH presented an increase in the phosphorylation of the Ser845 residue of the GluA1 subunit of AMPA receptors compared to control slices. This effect was reverted by SCH23390, antagonist of D1/D5 receptors, and PKI. Moreover, we found an increase of surface-associated functional AMPA receptors. We propose that MPH increases TBS-dependent LTP in CA3-CA1 synapses through a polysynaptic mechanism involving activation of β-adrenergic and D1/D5 dopaminergic receptors and promoting the trafficking and insertion of functional AMPA receptors to the plasma membrane.
Collapse
Affiliation(s)
- C Rozas
- Laboratorio de Neurociencia, Departamento de Biología, Universidad de Santiago de Chile, Chile
| | - C Carvallo
- Laboratorio de Neurociencia, Departamento de Biología, Universidad de Santiago de Chile, Chile
| | - D Contreras
- Laboratorio de Neurociencia, Departamento de Biología, Universidad de Santiago de Chile, Chile
| | - M Carreño
- Laboratorio de Neurociencia, Departamento de Biología, Universidad de Santiago de Chile, Chile
| | - G Ugarte
- Laboratorio de Neurociencia, Departamento de Biología, Universidad de Santiago de Chile, Chile
| | - R Delgado
- Laboratorio de Neurociencia, Departamento de Biología, Universidad de Santiago de Chile, Chile
| | - M L Zeise
- Escuela de Psicología, Universidad de Santiago de Chile, Chile
| | - B Morales
- Laboratorio de Neurociencia, Departamento de Biología, Universidad de Santiago de Chile, Chile.
| |
Collapse
|
35
|
Ponchio R, Teodorov E, Kirsten T, Coelho C, Oshiro A, Florio J, Bernardi M. Repeated methylphenidate administration during lactation reduces maternal behavior, induces maternal tolerance, and increases anxiety-like behavior in pups in adulthood. Neurotoxicol Teratol 2015; 50:64-72. [DOI: 10.1016/j.ntt.2015.05.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2015] [Revised: 05/02/2015] [Accepted: 05/18/2015] [Indexed: 12/13/2022]
|
36
|
Di Miceli M, Gronier B. Psychostimulants and atomoxetine alter the electrophysiological activity of prefrontal cortex neurons, interaction with catecholamine and glutamate NMDA receptors. Psychopharmacology (Berl) 2015; 232:2191-205. [PMID: 25572531 DOI: 10.1007/s00213-014-3849-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2014] [Accepted: 12/12/2014] [Indexed: 12/29/2022]
Abstract
RATIONALE Attention-deficit hyperactivity disorder (ADHD) is the most frequently diagnosed neuropsychiatric disorder in childhood. Currently available ADHD drugs include the psychostimulants methylphenidate (MPH) and D-amphetamine (D-AMP), acting on norepinephrine and dopamine transporters/release, and atomoxetine (ATX), a selective norepinephrine uptake inhibitor. Recent evidence suggests an involvement of glutamate neurotransmission in the pathology and treatment of ADHD, via mechanisms to be clarified. OBJECTIVE We have investigated how ADHD drugs could modulate, through interaction with catecholamine receptors, basal and glutamate-induced excitability of pyramidal neurons in the prefrontal cortex (PFC), a region which plays a major role in control of attention and impulsivity. METHODS We have used the technique of extracellular single-unit recording in anaesthetised rats coupled with microiontophoresis. RESULTS Both MPH (1-3 mg/kg) and D-AMP (1-9 mg/kg) increased the firing activity of PFC neurons in a dopamine D1 receptor-dependent manner. ATX administration (1-6 mg/kg) also increased the firing of neurons, but this effect is not significantly reversed by D1 (SCH 23390) or alpha1 (prazosin) receptor antagonists but potentiated by alpha2 antagonist (yohimbine). All drugs induced a clear potentiation of the excitatory response of PFC neurons to the microiontophoretic application of the glutamate agonist N-methyl-D-aspartate (NMDA), but not to the glutamate agonist α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA). The potentiating effect of D-AMP on NMDA-induced activation of PFC neurons was partially reversed or prevented by dopamine D1 receptor blockade. CONCLUSION Our data shows that increase in excitability of PFC neurons in basal conditions and via NMDA receptor activation may be involved in the therapeutic response to ADHD drugs.
Collapse
Affiliation(s)
- Mathieu Di Miceli
- Leicester School of Pharmacy, De Montfort University, The Gateway, Leicester, LE1 9BH, UK
| | | |
Collapse
|
37
|
Burgos H, Cofré C, Hernández A, Sáez-Briones P, Agurto R, Castillo A, Morales B, Zeise ML. Methylphenidate has long-lasting metaplastic effects in the prefrontal cortex of adolescent rats. Behav Brain Res 2015; 291:112-117. [PMID: 25997580 DOI: 10.1016/j.bbr.2015.05.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2014] [Revised: 05/05/2015] [Accepted: 05/08/2015] [Indexed: 02/06/2023]
Abstract
Methylphenidate (MPH) is widely used as a "nootropic" agent and in the treatment of disorders of attention, and has been shown to modulate synaptic plasticity in vitro. Here we present in vivo evidence that this MPH-induced metaplasticity can last long after the end of treatment. MPH (0, 0.2, 1 and 5mg/kg) was administered daily to male rats from postnatal day 42 for 15 days. The animals were tested daily in a radial maze. Long-term potentiation (LTP), a marker of neural plasticity, was induced in vivo in the prefrontal cortex after 2-3h, 15-18 days or 5 months without treatment. The behavioral performance of the 1mg/kg group improved, while that of animals that had received 5mg/kg deteriorated. In the 1 and 5mg/kg groups LTP induced 2-3h after the last MPH treatment was twice as large as in the controls. Further, 15-18 days after the last MPH administration, in groups receiving 1 and 5mg/kg, LTP was about fourfold higher than in controls. However, 5 months later, LTP in the 1mg/kg group was similar to controls and in the 5mg/kg group LTP could not be induced at all. No significant changes of LTP were seen in the low-dose group of animals (0.2mg/kg). Thus, firstly, doses of MPH that improve learning coincide approximately with those that augment LTP. Secondly, MPH-induced increases in LTP can last for several weeks, but these may disappear over longer periods or deteriorate at high doses.
Collapse
Affiliation(s)
- H Burgos
- Laboratorio de Biopsicología, Escuela de Psicología, FAHU, USACH, Chile; Escuela de Psicología, FACSO, Universidad Central de Chile, FAHU, USACH, Chile
| | - C Cofré
- Laboratorio de Biopsicología, Escuela de Psicología, FAHU, USACH, Chile
| | - A Hernández
- Laboratorio de Neurobiología, Facultad de Química y Biología, FAHU, USACH, Chile
| | - P Sáez-Briones
- Laboratorio de Neurofarmacología y Comportamiento, Facultad de Ciencias Médicas, FAHU, USACH, Chile
| | - R Agurto
- Escuela de Psicología, FCJS, Universidad Autónoma de Chile, FAHU, USACH, Chile
| | - A Castillo
- Escuela de Psicología, FCJS, Universidad Autónoma de Chile, FAHU, USACH, Chile
| | - B Morales
- Laboratorio de Neurociencias, Facultad de Química y Biología, Universidad de Santiago de Chile, FAHU, USACH, Chile.
| | - M L Zeise
- Laboratorio de Biopsicología, Escuela de Psicología, FAHU, USACH, Chile.
| |
Collapse
|
38
|
Skoglund C, Chen Q, Franck J, Lichtenstein P, Larsson H. Attention-deficit/hyperactivity disorder and risk for substance use disorders in relatives. Biol Psychiatry 2015; 77:880-6. [PMID: 25457227 DOI: 10.1016/j.biopsych.2014.10.006] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2014] [Revised: 10/14/2014] [Accepted: 10/15/2014] [Indexed: 12/31/2022]
Abstract
BACKGROUND Previous research indicates that attention-deficit/hyperactivity disorder (ADHD) is highly associated with substance use disorders (SUD). However, these studies have failed to clarify the nature of the overlap. The main aim of this study was to explore whether the overlap between ADHD and SUD could be explained by shared genetic and environmental factors or by harmful effects of ADHD medication. METHODS We employed a matched cohort design across different levels of family relatedness recorded from 1973-2009. By linking longitudinal Swedish national registers, 62,015 ADHD probands and first-degree and second-degree relatives were identified and matched 1:10 with control subjects without ADHD and their corresponding relatives. Any record of SUD was defined by discharge diagnoses of the International Classification of Diseases or a purchase of any drug used in the treatment of SUD. RESULTS First-degree relatives of ADHD probands were at elevated risk for SUD (odds ratios 2.2 and 1.8) compared with relatives of control subjects. The corresponding relative risk in second-degree relatives was substantially lower (odd ratios 1.4 and 1.4). The familial aggregation patterns remained similar for first-degree and second-degree relatives after excluding individuals with coexisting disorders such as schizophrenia, bipolar disorder, depression, and conduct disorder. CONCLUSIONS Our findings suggest that the co-occurrence of ADHD and SUD is due to genetic factors shared between the two disorders, rather than to a general propensity for psychiatric disorders or harmful effects of ADHD medication.
Collapse
Affiliation(s)
- Charlotte Skoglund
- Departments of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden..
| | - Qi Chen
- Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Johan Franck
- Departments of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Paul Lichtenstein
- Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Henrik Larsson
- Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
39
|
Baskin BM, Dwoskin LP, Kantak KM. Methylphenidate treatment beyond adolescence maintains increased cocaine self-administration in the spontaneously hypertensive rat model of attention deficit/hyperactivity disorder. Pharmacol Biochem Behav 2015; 131:51-6. [PMID: 25643872 DOI: 10.1016/j.pbb.2015.01.019] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Revised: 01/21/2015] [Accepted: 01/26/2015] [Indexed: 10/24/2022]
Abstract
Past research with the spontaneously hypertensive rat (SHR) model of attention deficit/hyperactivity disorder showed that adolescent methylphenidate treatment enhanced cocaine abuse risk in SHR during adulthood. The acquisition of cocaine self-administration was faster, and cocaine dose-response functions were shifted upward under fixed-ratio and progressive ratio schedules compared to adult SHR that received adolescent vehicle treatment or to control strains that received adolescent methylphenidate treatment. The current study determined if extending treatment beyond adolescence would ameliorate long-term consequences of adolescent methylphenidate treatment on cocaine abuse risk in adult SHR. Treatments (vehicle or 1.5mg/kg/day oral methylphenidate) began on postnatal day 28. Groups of male SHR were treated with vehicle during adolescence and adulthood, with methylphenidate during adolescence and vehicle during adulthood, or with methylphenidate during adolescence and adulthood. The group receiving adolescent-only methylphenidate was switched to vehicle on P56. Cocaine self-administration began on postnatal day 77, and groups receiving methylphenidate during adolescence and adulthood were treated either 1-h before or 1-h after daily sessions. At baseline under a fixed-ratio 1 schedule, cocaine self-administration (2h sessions; 0.3mg/kg unit dose) did not differ among the four treatment groups. Under a progressive ratio schedule (4.5h maximum session length; 0.01-1.0mg/kg unit doses), breakpoints for self-administered cocaine in SHR receiving the adult methylphenidate treatment 1-h pre-session were not different from the vehicle control group. However, compared to the vehicle control group, breakpoints for self-administered cocaine at the 0.3 and 1.0mg/kg unit doses were greater in adult SHR that received adolescent-only methylphenidate or received methylphenidate that was continued into adulthood and administered 1-h post-session. These findings suggest that extending methylphenidate treatment beyond adolescence does not ameliorate explicitly the long-term consequences of adolescent methylphenidate treatment. Pre-session methylphenidate may mask temporarily the detection of an increase in cocaine self-administration following chronic methylphenidate treatment.
Collapse
Affiliation(s)
- Britahny M Baskin
- Department of Psychological and Brain Sciences, Boston University, Boston, MA 02215, USA..
| | - Linda P Dwoskin
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY 40536 USA.
| | - Kathleen M Kantak
- Department of Psychological and Brain Sciences, Boston University, Boston, MA 02215, USA..
| |
Collapse
|
40
|
Jenson D, Yang K, Acevedo-Rodriguez A, Levine A, Broussard JI, Tang J, Dani JA. Dopamine and norepinephrine receptors participate in methylphenidate enhancement of in vivo hippocampal synaptic plasticity. Neuropharmacology 2014; 90:23-32. [PMID: 25445492 DOI: 10.1016/j.neuropharm.2014.10.029] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2014] [Revised: 10/29/2014] [Accepted: 10/30/2014] [Indexed: 11/30/2022]
Abstract
Attention-deficit hyperactive disorder (ADHD) is the most commonly studied and diagnosed psychiatric disorder in children. Methylphenidate (MPH, e.g., Ritalin) has been used to treat ADHD for over 50 years. It is the most commonly prescribed treatment for ADHD, and in the past decade it was the drug most commonly prescribed to teenagers. In addition, MPH has become one of the most widely abused drugs on college campuses. In this study, we examined the effects of MPH on hippocampal synaptic plasticity, which serves as a measurable quantification of memory mechanisms. Field potentials were recorded with permanently implanted electrodes in freely-moving mice to quantify MPH modulation of perforant path synaptic transmission onto granule cells of the dentate gyrus. Our hypothesis was that MPH affects hippocampal synaptic plasticity underlying learning because MPH boosts catecholamine signaling by blocking the dopamine and norepinephrine transporters (DAT and NET respectively). In vitro hippocampal slice experiments indicated MPH enhances perforant path plasticity, and this MPH enhancement arose from action via D1-type dopamine receptors and β-type adrenergic receptors. Similarly, MPH boosted in vivo initiation of long-term potentiation (LTP). While there was an effect via both dopamine and adrenergic receptors in vivo, LTP induction was more dependent on the MPH-induced action via D1-type dopamine receptors. Under biologically reasonable experimental conditions, MPH enhances hippocampal synaptic plasticity via catecholamine receptors.
Collapse
Affiliation(s)
- Daniel Jenson
- Department of Neuroscience, Center on Addiction, Learning, Memory, Baylor College of Medicine, Houston, TX 77030, USA
| | - Kechun Yang
- Department of Neuroscience, Mahoney Institute for Neurosciences, Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Alexandra Acevedo-Rodriguez
- Department of Neuroscience, Center on Addiction, Learning, Memory, Baylor College of Medicine, Houston, TX 77030, USA
| | - Amber Levine
- Department of Neuroscience, Center on Addiction, Learning, Memory, Baylor College of Medicine, Houston, TX 77030, USA
| | - John I Broussard
- Department of Neuroscience, Center on Addiction, Learning, Memory, Baylor College of Medicine, Houston, TX 77030, USA
| | - Jianrong Tang
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
| | - John A Dani
- Department of Neuroscience, Mahoney Institute for Neurosciences, Perelman School of Medicine, Philadelphia, PA 19104, USA.
| |
Collapse
|
41
|
Lepelletier FX, Tauber C, Nicolas C, Solinas M, Castelnau P, Belzung C, Emond P, Cortese S, Faraone SV, Chalon S, Galineau L. Prenatal exposure to methylphenidate affects the dopamine system and the reactivity to natural reward in adulthood in rats. Int J Neuropsychopharmacol 2014; 18:pyu044. [PMID: 25522388 PMCID: PMC4360227 DOI: 10.1093/ijnp/pyu044] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2014] [Revised: 06/24/2014] [Accepted: 07/08/2014] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Methylphenidate (MPH) is a commonly-used medication for the treatment of children with Attention-Deficit/Hyperactivity Disorders (ADHD). However, its prescription to adults with ADHD and narcolepsy raises the question of how the brain is impacted by MPH exposure during pregnancy. The goal of this study was to elucidate the long-term neurobiological consequences of prenatal exposure to MPH using a rat model. METHODS We focused on the effects of such treatment on the adult dopamine (DA) system and on the reactivity of animals to natural rewards. RESULTS This study shows that adult male rats prenatally exposed to MPH display elevated expression of presynaptic DA markers in the DA cell bodies and the striatum. Our results also suggest that MPH-treated animals could exhibit increased tonic DA activity in the mesolimbic pathway, altered signal-to-noise ratio after a pharmacological stimulation, and decreased reactivity to the locomotor effects of cocaine. Finally, we demonstrated that MPH rats display a decreased preference and motivation for sucrose. CONCLUSIONS This is the first preclinical study reporting long-lasting neurobiological alterations of DA networks as well as alterations in motivational behaviors for natural rewards after a prenatal exposure to MPH. These results raise concerns about the possible neurobiological consequences of MPH treatment during pregnancy.
Collapse
Affiliation(s)
- François-Xavier Lepelletier
- Université François-Rabelais de Tours, Inserm, Imagerie et Cerveau UMR 930, Tours, France (Drs Lepelletier, Tauber, Castelnau, Belzung, Emond, Chalon, and Galineau); Experimental and Clinical Neurosciences Laboratory, INSERM U1084, Poitiers, France (Drs Nicolas and Solinas); University of Poitiers, Poitiers, France (Drs Nicolas and Solinas); Child Neurology Unit, University Hospital, University of Tours, Tours, France (Dr Castelnau); Department of Neurosciences, University François Rabelais of Tours, Tours, France (Drs Belzung and Galineau); Institute for Pediatric Neuroscience, NYU Child Study Center, Langone Medical Center, NY (Dr Cortese); Child Psychiatry Centre, University Hospital, University of Tours, Tours, France (Dr Cortese); Departments of Psychiatry and of Neuroscience and Physiology, Upstate Medical University, State University of New York, Syracuse, NY (Dr Faraone)
| | - Clovis Tauber
- Université François-Rabelais de Tours, Inserm, Imagerie et Cerveau UMR 930, Tours, France (Drs Lepelletier, Tauber, Castelnau, Belzung, Emond, Chalon, and Galineau); Experimental and Clinical Neurosciences Laboratory, INSERM U1084, Poitiers, France (Drs Nicolas and Solinas); University of Poitiers, Poitiers, France (Drs Nicolas and Solinas); Child Neurology Unit, University Hospital, University of Tours, Tours, France (Dr Castelnau); Department of Neurosciences, University François Rabelais of Tours, Tours, France (Drs Belzung and Galineau); Institute for Pediatric Neuroscience, NYU Child Study Center, Langone Medical Center, NY (Dr Cortese); Child Psychiatry Centre, University Hospital, University of Tours, Tours, France (Dr Cortese); Departments of Psychiatry and of Neuroscience and Physiology, Upstate Medical University, State University of New York, Syracuse, NY (Dr Faraone)
| | - Céline Nicolas
- Université François-Rabelais de Tours, Inserm, Imagerie et Cerveau UMR 930, Tours, France (Drs Lepelletier, Tauber, Castelnau, Belzung, Emond, Chalon, and Galineau); Experimental and Clinical Neurosciences Laboratory, INSERM U1084, Poitiers, France (Drs Nicolas and Solinas); University of Poitiers, Poitiers, France (Drs Nicolas and Solinas); Child Neurology Unit, University Hospital, University of Tours, Tours, France (Dr Castelnau); Department of Neurosciences, University François Rabelais of Tours, Tours, France (Drs Belzung and Galineau); Institute for Pediatric Neuroscience, NYU Child Study Center, Langone Medical Center, NY (Dr Cortese); Child Psychiatry Centre, University Hospital, University of Tours, Tours, France (Dr Cortese); Departments of Psychiatry and of Neuroscience and Physiology, Upstate Medical University, State University of New York, Syracuse, NY (Dr Faraone)
| | - Marcello Solinas
- Université François-Rabelais de Tours, Inserm, Imagerie et Cerveau UMR 930, Tours, France (Drs Lepelletier, Tauber, Castelnau, Belzung, Emond, Chalon, and Galineau); Experimental and Clinical Neurosciences Laboratory, INSERM U1084, Poitiers, France (Drs Nicolas and Solinas); University of Poitiers, Poitiers, France (Drs Nicolas and Solinas); Child Neurology Unit, University Hospital, University of Tours, Tours, France (Dr Castelnau); Department of Neurosciences, University François Rabelais of Tours, Tours, France (Drs Belzung and Galineau); Institute for Pediatric Neuroscience, NYU Child Study Center, Langone Medical Center, NY (Dr Cortese); Child Psychiatry Centre, University Hospital, University of Tours, Tours, France (Dr Cortese); Departments of Psychiatry and of Neuroscience and Physiology, Upstate Medical University, State University of New York, Syracuse, NY (Dr Faraone)
| | - Pierre Castelnau
- Université François-Rabelais de Tours, Inserm, Imagerie et Cerveau UMR 930, Tours, France (Drs Lepelletier, Tauber, Castelnau, Belzung, Emond, Chalon, and Galineau); Experimental and Clinical Neurosciences Laboratory, INSERM U1084, Poitiers, France (Drs Nicolas and Solinas); University of Poitiers, Poitiers, France (Drs Nicolas and Solinas); Child Neurology Unit, University Hospital, University of Tours, Tours, France (Dr Castelnau); Department of Neurosciences, University François Rabelais of Tours, Tours, France (Drs Belzung and Galineau); Institute for Pediatric Neuroscience, NYU Child Study Center, Langone Medical Center, NY (Dr Cortese); Child Psychiatry Centre, University Hospital, University of Tours, Tours, France (Dr Cortese); Departments of Psychiatry and of Neuroscience and Physiology, Upstate Medical University, State University of New York, Syracuse, NY (Dr Faraone)
| | - Catherine Belzung
- Université François-Rabelais de Tours, Inserm, Imagerie et Cerveau UMR 930, Tours, France (Drs Lepelletier, Tauber, Castelnau, Belzung, Emond, Chalon, and Galineau); Experimental and Clinical Neurosciences Laboratory, INSERM U1084, Poitiers, France (Drs Nicolas and Solinas); University of Poitiers, Poitiers, France (Drs Nicolas and Solinas); Child Neurology Unit, University Hospital, University of Tours, Tours, France (Dr Castelnau); Department of Neurosciences, University François Rabelais of Tours, Tours, France (Drs Belzung and Galineau); Institute for Pediatric Neuroscience, NYU Child Study Center, Langone Medical Center, NY (Dr Cortese); Child Psychiatry Centre, University Hospital, University of Tours, Tours, France (Dr Cortese); Departments of Psychiatry and of Neuroscience and Physiology, Upstate Medical University, State University of New York, Syracuse, NY (Dr Faraone)
| | - Patrick Emond
- Université François-Rabelais de Tours, Inserm, Imagerie et Cerveau UMR 930, Tours, France (Drs Lepelletier, Tauber, Castelnau, Belzung, Emond, Chalon, and Galineau); Experimental and Clinical Neurosciences Laboratory, INSERM U1084, Poitiers, France (Drs Nicolas and Solinas); University of Poitiers, Poitiers, France (Drs Nicolas and Solinas); Child Neurology Unit, University Hospital, University of Tours, Tours, France (Dr Castelnau); Department of Neurosciences, University François Rabelais of Tours, Tours, France (Drs Belzung and Galineau); Institute for Pediatric Neuroscience, NYU Child Study Center, Langone Medical Center, NY (Dr Cortese); Child Psychiatry Centre, University Hospital, University of Tours, Tours, France (Dr Cortese); Departments of Psychiatry and of Neuroscience and Physiology, Upstate Medical University, State University of New York, Syracuse, NY (Dr Faraone)
| | - Samuele Cortese
- Université François-Rabelais de Tours, Inserm, Imagerie et Cerveau UMR 930, Tours, France (Drs Lepelletier, Tauber, Castelnau, Belzung, Emond, Chalon, and Galineau); Experimental and Clinical Neurosciences Laboratory, INSERM U1084, Poitiers, France (Drs Nicolas and Solinas); University of Poitiers, Poitiers, France (Drs Nicolas and Solinas); Child Neurology Unit, University Hospital, University of Tours, Tours, France (Dr Castelnau); Department of Neurosciences, University François Rabelais of Tours, Tours, France (Drs Belzung and Galineau); Institute for Pediatric Neuroscience, NYU Child Study Center, Langone Medical Center, NY (Dr Cortese); Child Psychiatry Centre, University Hospital, University of Tours, Tours, France (Dr Cortese); Departments of Psychiatry and of Neuroscience and Physiology, Upstate Medical University, State University of New York, Syracuse, NY (Dr Faraone)
| | - Stephen V Faraone
- Université François-Rabelais de Tours, Inserm, Imagerie et Cerveau UMR 930, Tours, France (Drs Lepelletier, Tauber, Castelnau, Belzung, Emond, Chalon, and Galineau); Experimental and Clinical Neurosciences Laboratory, INSERM U1084, Poitiers, France (Drs Nicolas and Solinas); University of Poitiers, Poitiers, France (Drs Nicolas and Solinas); Child Neurology Unit, University Hospital, University of Tours, Tours, France (Dr Castelnau); Department of Neurosciences, University François Rabelais of Tours, Tours, France (Drs Belzung and Galineau); Institute for Pediatric Neuroscience, NYU Child Study Center, Langone Medical Center, NY (Dr Cortese); Child Psychiatry Centre, University Hospital, University of Tours, Tours, France (Dr Cortese); Departments of Psychiatry and of Neuroscience and Physiology, Upstate Medical University, State University of New York, Syracuse, NY (Dr Faraone)
| | - Sylvie Chalon
- Université François-Rabelais de Tours, Inserm, Imagerie et Cerveau UMR 930, Tours, France (Drs Lepelletier, Tauber, Castelnau, Belzung, Emond, Chalon, and Galineau); Experimental and Clinical Neurosciences Laboratory, INSERM U1084, Poitiers, France (Drs Nicolas and Solinas); University of Poitiers, Poitiers, France (Drs Nicolas and Solinas); Child Neurology Unit, University Hospital, University of Tours, Tours, France (Dr Castelnau); Department of Neurosciences, University François Rabelais of Tours, Tours, France (Drs Belzung and Galineau); Institute for Pediatric Neuroscience, NYU Child Study Center, Langone Medical Center, NY (Dr Cortese); Child Psychiatry Centre, University Hospital, University of Tours, Tours, France (Dr Cortese); Departments of Psychiatry and of Neuroscience and Physiology, Upstate Medical University, State University of New York, Syracuse, NY (Dr Faraone)
| | - Laurent Galineau
- Université François-Rabelais de Tours, Inserm, Imagerie et Cerveau UMR 930, Tours, France (Drs Lepelletier, Tauber, Castelnau, Belzung, Emond, Chalon, and Galineau); Experimental and Clinical Neurosciences Laboratory, INSERM U1084, Poitiers, France (Drs Nicolas and Solinas); University of Poitiers, Poitiers, France (Drs Nicolas and Solinas); Child Neurology Unit, University Hospital, University of Tours, Tours, France (Dr Castelnau); Department of Neurosciences, University François Rabelais of Tours, Tours, France (Drs Belzung and Galineau); Institute for Pediatric Neuroscience, NYU Child Study Center, Langone Medical Center, NY (Dr Cortese); Child Psychiatry Centre, University Hospital, University of Tours, Tours, France (Dr Cortese); Departments of Psychiatry and of Neuroscience and Physiology, Upstate Medical University, State University of New York, Syracuse, NY (Dr Faraone).
| |
Collapse
|
42
|
Patrick KS, Corbin TR, Murphy CE. Ethylphenidate as a selective dopaminergic agonist and methylphenidate-ethanol transesterification biomarker. J Pharm Sci 2014; 103:3834-3842. [PMID: 25303048 DOI: 10.1002/jps.24202] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2014] [Revised: 09/18/2014] [Accepted: 09/18/2014] [Indexed: 01/19/2023]
Abstract
We review the pharmaceutical science of ethylphenidate (EPH) in the contexts of drug discovery, drug interactions, biomarker for dl-methylphenidate (MPH)-ethanol exposure, potentiation of dl-MPH abuse liability, contemporary "designer drug," pertinence to the newer transdermal and chiral switch MPH formulations, as well as problematic internal standard. d-EPH selectively targets the dopamine transporter, whereas d-MPH exhibits equipotent actions at dopamine and norepinephrine transporters. This selectivity carries implications for the advancement of tailored attention-deficit/hyperactivity disorder (ADHD) pharmacotherapy in the era of genome-based diagnostics. Abuse of dl-MPH often involves ethanol coabuse. Carboxylesterase 1 enantioselectively transesterifies l-MPH with ethanol to yield l-EPH accompanied by significantly increased early exposure to d-MPH and rapid potentiation of euphoria. The pharmacokinetic component of this drug interaction can largely be avoided using dexmethylphenidate (dexMPH). This notwithstanding, maximal potentiated euphoria occurs following dexMPH-ethanol. C57BL/6 mice model dl-MPH-ethanol interactions: an otherwise depressive dose of ethanol synergistically increases dl-MPH stimulation; a substimulatory dose of dl-MPH potentiates a low, stimulatory dose of ethanol; ethanol elevates blood, brain, and urinary d-MPH concentrations while forming l-EPH. Integration of EPH preclinical neuropharmacology with clinical studies of MPH-ethanol interactions provides a translational approach toward advancement of ADHD personalized medicine and management of comorbid alcohol use disorder.
Collapse
Affiliation(s)
- Kennerly S Patrick
- Department of Drug Discovery and Biomedical Sciences, Medical University of South Carolina, Charleston, South Carolina 29425-1400.
| | - Timothy R Corbin
- Department of Drug Discovery and Biomedical Sciences, Medical University of South Carolina, Charleston, South Carolina 29425-1400
| | - Cristina E Murphy
- Department of Drug Discovery and Biomedical Sciences, Medical University of South Carolina, Charleston, South Carolina 29425-1400
| |
Collapse
|
43
|
Yang X, Morris SM, Gearhart JM, Ruark CD, Paule MG, Slikker W, Mattison DR, Vitiello B, Twaddle NC, Doerge DR, Young JF, Fisher JW. Development of a physiologically based model to describe the pharmacokinetics of methylphenidate in juvenile and adult humans and nonhuman primates. PLoS One 2014; 9:e106101. [PMID: 25184666 PMCID: PMC4153582 DOI: 10.1371/journal.pone.0106101] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2014] [Accepted: 07/28/2014] [Indexed: 11/18/2022] Open
Abstract
The widespread usage of methylphenidate (MPH) in the pediatric population has received considerable attention due to its potential effect on child development. For the first time a physiologically based pharmacokinetic (PBPK) model has been developed in juvenile and adult humans and nonhuman primates to quantitatively evaluate species- and age-dependent enantiomer specific pharmacokinetics of MPH and its primary metabolite ritalinic acid. The PBPK model was first calibrated in adult humans using in vitro enzyme kinetic data of MPH enantiomers, together with plasma and urine pharmacokinetic data with MPH in adult humans. Metabolism of MPH in the small intestine was assumed to account for the low oral bioavailability of MPH. Due to lack of information, model development for children and juvenile and adult nonhuman primates primarily relied on intra- and interspecies extrapolation using allometric scaling. The juvenile monkeys appear to metabolize MPH more rapidly than adult monkeys and humans, both adults and children. Model prediction performance is comparable between juvenile monkeys and children, with average root mean squared error values of 4.1 and 2.1, providing scientific basis for interspecies extrapolation of toxicity findings. Model estimated human equivalent doses in children that achieve similar internal dose metrics to those associated with pubertal delays in juvenile monkeys were found to be close to the therapeutic doses of MPH used in pediatric patients. This computational analysis suggests that continued pharmacovigilance assessment is prudent for the safe use of MPH.
Collapse
Affiliation(s)
- Xiaoxia Yang
- National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, Arkansas, United States of America
- * E-mail:
| | - Suzanne M. Morris
- National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, Arkansas, United States of America
| | - Jeffery M. Gearhart
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Wright-Patterson Air Force Base, Ohio, United States of America
| | - Christopher D. Ruark
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Wright-Patterson Air Force Base, Ohio, United States of America
| | - Merle G. Paule
- National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, Arkansas, United States of America
| | - William Slikker
- National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, Arkansas, United States of America
| | - Donald R. Mattison
- Risk Sciences International, Ottawa, Ontario, Canada
- University of Ottawa, Ottawa, Ontario, Canada
| | - Benedetto Vitiello
- National Institute of Mental Health, Bethesda, Maryland, United States of America
| | - Nathan C. Twaddle
- National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, Arkansas, United States of America
| | - Daniel R. Doerge
- National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, Arkansas, United States of America
| | - John F. Young
- National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, Arkansas, United States of America
| | - Jeffrey W. Fisher
- National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, Arkansas, United States of America
| |
Collapse
|
44
|
Kishikawa Y, Kawahara Y, Yamada M, Kaneko F, Kawahara H, Nishi A. The spontaneously hypertensive rat/Izm (SHR/Izm) shows attention deficit/hyperactivity disorder-like behaviors but without impulsive behavior: therapeutic implications of low-dose methylphenidate. Behav Brain Res 2014; 274:235-42. [PMID: 25151620 DOI: 10.1016/j.bbr.2014.08.026] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2014] [Revised: 08/06/2014] [Accepted: 08/11/2014] [Indexed: 01/20/2023]
Abstract
The spontaneously hypertensive rat (SHR) has been used as a genetic animal model of attention deficit/hyperactivity disorder (ADHD). SHR/Izm is derived from stroke-resistant SHR as SHR/NIH and SHR/NCrl but from 22nd to 23rd generation descendants of the SHR/NIH ancestor and therefore may show different behavioral phenotypes compared to other SHR sub-strains. In this study, ADHD-like behaviors in SHR/Izm were evaluated compared to Wistar rats. SHR/Izm showed high locomotor activity in the habituation phase in a novel environment, although locomotor activity in the initial exploratory phase was low. In a behavioral test for attention, spontaneous alternation behavior in the Y-maze test was impaired in SHR/Izm. However, impulsive behavior in the elevated-plus maze test, which is designed to detect anxiety-related behavior but also reflects impulsivity for novelty seeking, was comparable to Wistar rats. Hyperactivity and inattention, detected as ADHD-like behaviors in SHR/Izm, were ameliorated with methylphenidate at a low dose (0.05mg/kg, i.p.). Therefore, SHR/Izm represents a unique animal model of ADHD without anxiety-related impulsive behavior.
Collapse
Affiliation(s)
- Yuki Kishikawa
- Department of Pharmacology, Kurume University School of Medicine, Asahi-machi 67, Kurume, Fukuoka 830-0011, Japan.
| | - Yukie Kawahara
- Department of Pharmacology, Kurume University School of Medicine, Asahi-machi 67, Kurume, Fukuoka 830-0011, Japan.
| | - Makiko Yamada
- Department of Pharmacology, Kurume University School of Medicine, Asahi-machi 67, Kurume, Fukuoka 830-0011, Japan; Department of Psychiatry, Tokyo Women's Medical University, Kawada-Cho 8-1, Shinjuku-ku, Tokyo 168-8666, Japan.
| | - Fumi Kaneko
- Department of Pharmacology, Kurume University School of Medicine, Asahi-machi 67, Kurume, Fukuoka 830-0011, Japan.
| | - Hiroshi Kawahara
- Department of Dental Anesthesiology, School of Dentistry, Tsurumi University, Tsurumi 2-1-3, Tsurumi-ku, Yokohama, Kanagawa 230-8501, Japan.
| | - Akinori Nishi
- Department of Pharmacology, Kurume University School of Medicine, Asahi-machi 67, Kurume, Fukuoka 830-0011, Japan.
| |
Collapse
|
45
|
Mohamed AD. Neuroethical issues in pharmacological cognitive enhancement. WILEY INTERDISCIPLINARY REVIEWS. COGNITIVE SCIENCE 2014; 5:533-549. [PMID: 26308743 DOI: 10.1002/wcs.1306] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2014] [Revised: 05/31/2014] [Accepted: 06/22/2014] [Indexed: 12/11/2022]
Abstract
UNLABELLED Neuroethics is an emerging field that in general deals with the ethics of neuroscience and the neuroscience of ethics. In particular, it is concerned with the ethical issues in the translation of neuroscience to clinical practice and in the public domain. Numerous ethical issues arise when healthy individuals use pharmacological substances known as pharmacological cognitive enhancers (PCEs) for non-medical purposes in order to boost higher-order cognitive processes such as memory, attention, and executive functions. However, information regarding their actual use, benefits, and harms to healthy individuals is currently lacking. Neuroethical issues that arise from their use include the unknown side effects that are associated with these drugs, concerns about the modification of authenticity and personhood, and as a result of inequality of access to these drugs, the lack of distributive justice and competitive fairness that they may cause in society. Healthy individuals might be coerced by social institutions that force them to take these drugs to function better. These drugs might enable or hinder healthy individuals to gain better moral and self-understanding and autonomy. However, how these drugs might achieve this still remains speculative and unknown. Hence, before concrete policy decisions are made, the cognitive effects of these drugs should be determined. The initiation of accurate surveys to determine the actual usage of these drugs by healthy individuals from different sections of the society is proposed. In addition, robust empirical research need to be conducted to delineate not only whether or not these drugs modify complex higher-order cognitive processes but also how they might alter important human virtues such as empathy, moral reasoning, creativity, and motivation in healthy individuals. WIREs Cogn Sci 2014, 5:533-549. doi: 10.1002/wcs.1306 For further resources related to this article, please visit the WIREs website. CONFLICT OF INTEREST The author has declared no conflicts of interest for this article.
Collapse
Affiliation(s)
- Ahmed Dahir Mohamed
- The School of Psychology, Cognitive and Sensory Systems Group, Faculty of Science, University of Nottingham, Royal Selangor, Malaysia.,Department of Psychiatry, Cambridge School of Clinical Medicine, University of Cambridge, Cambridge, UK.,Clare Hall College, Cambridge, UK
| |
Collapse
|
46
|
Heal DJ, Gosden J, Smith SL. Dopamine reuptake transporter (DAT) "inverse agonism"--a novel hypothesis to explain the enigmatic pharmacology of cocaine. Neuropharmacology 2014; 87:19-40. [PMID: 24953830 DOI: 10.1016/j.neuropharm.2014.06.012] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2014] [Revised: 05/07/2014] [Accepted: 06/09/2014] [Indexed: 12/20/2022]
Abstract
The long held view is cocaine's pharmacological effects are mediated by monoamine reuptake inhibition. However, drugs with rapid brain penetration like sibutramine, bupropion, mazindol and tesofensine, which are equal to or more potent than cocaine as dopamine reuptake inhibitors, produce no discernable subjective effects such as drug "highs" or euphoria in drug-experienced human volunteers. Moreover they are dysphoric and aversive when given at high doses. In vivo experiments in animals demonstrate that cocaine's monoaminergic pharmacology is profoundly different from that of other prescribed monoamine reuptake inhibitors, with the exception of methylphenidate. These findings led us to conclude that the highly unusual stimulant profile of cocaine and related compounds, eg methylphenidate, is not mediated by monoamine reuptake inhibition alone. We describe the experimental findings which suggest cocaine serves as a negative allosteric modulator to alter the function of the dopamine reuptake transporter (DAT) and reverse its direction of transport. This results in a firing-dependent, retro-transport of dopamine into the synaptic cleft. The proposed mechanism of cocaine is, therefore, different from other small molecule negative allostereric modulators of the monoamine reuptake transporters, eg SoRI-6238, which merely reduce the rate of inward transport. Because the physiological role of DAT is to remove dopamine from the synapse and the action of cocaine is the opposite of this, we have postulated that cocaine's effect is analogous to an inverse agonist. If this hypothesis is validated then cocaine is the prototypical compound that exemplifies a new class of monoaminergic drugs; DAT "inverse agonists". This article is part of the Special Issue entitled 'CNS Stimulants'.
Collapse
Affiliation(s)
- David J Heal
- RenaSci Limited, BioCity, Pennyfoot Street, Nottingham NG1 1GF, UK.
| | - Jane Gosden
- RenaSci Limited, BioCity, Pennyfoot Street, Nottingham NG1 1GF, UK
| | - Sharon L Smith
- RenaSci Limited, BioCity, Pennyfoot Street, Nottingham NG1 1GF, UK.
| |
Collapse
|
47
|
Mueller S, Costa A, Keeser D, Pogarell O, Berman A, Coates U, Reiser MF, Riedel M, Möller HJ, Ettinger U, Meindl T. The effects of methylphenidate on whole brain intrinsic functional connectivity. Hum Brain Mapp 2014; 35:5379-88. [PMID: 24862742 DOI: 10.1002/hbm.22557] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2013] [Revised: 04/25/2014] [Accepted: 05/07/2014] [Indexed: 12/29/2022] Open
Abstract
Methylphenidate (MPH) is an indirect dopaminergic and noradrenergic agonist that is used to treat attention deficit hyperactivity disorder and that has shown therapeutic potential in neuropsychiatric diseases such as depression, dementia, and Parkinson's disease. While effects of MPH on task-induced brain activation have been investigated, little is known about how MPH influences the resting brain. To investigate the effects of 40 mg of oral MPH on intrinsic functional connectivity, we used resting state fMRI in 54 healthy male subjects in a double-blind, randomized, placebo-controlled study. Functional connectivity analysis employing ICA revealed seven resting state networks (RSN) of interest. Connectivity strength between the dorsal attention network and the thalamus was increased after MPH intake. Other RSN located in association cortex areas, such as the left and right frontoparietal networks and the executive control network, showed MPH-induced connectivity increase to sensory-motor and visual cortex regions and connectivity decrease to cortical and subcortical components of cortico-striato-thalamo-cortical circuits (CST). RSN located in sensory-motor cortex areas showed the opposite pattern with MPH-induced connectivity increase to CST components and connectivity decrease to sensory-motor and visual cortex regions. Our results provide evidence that MPH does not only alter intrinsic connectivity between brain areas involved in sustained attention, but that it also induces significant changes in the cortico-cortical and cortico-subcortical connectivity of many other cognitive and sensory-motor RSN.
Collapse
Affiliation(s)
- Sophia Mueller
- Institute of Clinical Radiology, Ludwig-Maximilians University Munich, 81377, Munich, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Human abuse liability evaluation of CNS stimulant drugs. Neuropharmacology 2014; 87:81-90. [PMID: 24793872 DOI: 10.1016/j.neuropharm.2014.04.014] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2014] [Revised: 04/01/2014] [Accepted: 04/16/2014] [Indexed: 01/16/2023]
Abstract
Psychoactive drugs that increase alertness, attention and concentration and energy, while also elevating mood, heart rate and blood pressure are referred to as stimulants. Despite some overlapping similarities, stimulants cannot be easily categorized by their chemical structure, mechanism of action, receptor binding profile, effects on monoamine uptake, behavioral pharmacology (e.g., effects on locomotion, temperature, and blood pressure), therapeutic indication or efficacy. Because of their abuse liability, a pre-market assessment of abuse potential is required for drugs that show stimulant properties; this review article focuses on the clinical aspects of this evaluation. This includes clinical trial adverse events, evidence of diversion or tampering, overdoses and the results of a human abuse potential study. While there are different types of human experimental studies that can be employed to evaluate stimulant abuse potential (e.g., drug discrimination, self-administration), only the human abuse potential study and clinical trial adverse event data are required for drug approval. The principal advances that have improved human abuse potential studies include using study enrichment strategies (pharmacologic qualification), larger sample sizes, better selection of endpoints and measurement strategies and more carefully considered interpretation of data. Because of the methodological advances, comparisons of newer studies with historical data is problematic and may contribute to a biased regulatory framework for the evaluation of newer stimulant-like drugs, such as A2 antagonists. This article is part of the Special Issue entitled 'CNS Stimulants'.
Collapse
|
49
|
Sripada C, Kessler D, Jonides J. Methylphenidate blocks effort-induced depletion of regulatory control in healthy volunteers. Psychol Sci 2014; 25:1227-34. [PMID: 24756766 DOI: 10.1177/0956797614526415] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2013] [Accepted: 01/23/2014] [Indexed: 11/15/2022] Open
Abstract
A recent wave of studies--more than 100 conducted over the last decade--has shown that exerting effort at controlling impulses or behavioral tendencies leaves a person depleted and less able to engage in subsequent rounds of regulation. Regulatory depletion is thought to play an important role in everyday problems (e.g., excessive spending, overeating) as well as psychiatric conditions, but its neurophysiological basis is poorly understood. Using a placebo-controlled, double-blind design, we demonstrated that the psychostimulant methylphenidate (commonly known as Ritalin), a catecholamine reuptake blocker that increases dopamine and norepinephrine at the synaptic cleft, fully blocks effort-induced depletion of regulatory control. Spectral analysis of trial-by-trial reaction times revealed specificity of methylphenidate effects on regulatory depletion in the slow-4 frequency band. This band is associated with the operation of resting-state brain networks that produce mind wandering, which raises potential connections between our results and recent brain-network-based models of control over attention.
Collapse
Affiliation(s)
| | | | - John Jonides
- Department of Psychology, University of Michigan
| |
Collapse
|
50
|
Graf WD, Miller G, Nagel SK. Addressing the problem of ADHD medication as neuroenhancements. Expert Rev Neurother 2014; 14:569-81. [PMID: 24738763 DOI: 10.1586/14737175.2014.908707] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The prevalence of attention deficit hyperactivity disorder (ADHD) diagnoses is rising. ADHD is closely linked to its treatment with medications such as methylphenidate and amphetamines, which have popular appeal as neuroenhancement drugs by persons without a neurological disorder. The three main reasons for the increase in ADHD medication demand, production, and consumption are a) the inclusion of milder ADHD diagnoses; b) the vast marketing of ADHD medications by the pharmaceutical industry; and c) the illegal diversion of controlled ADHD medication to consumers seeking stimulants as neuroenhancements. Rapidly rising rates of any neurological disorder - especially a behaviorly-defined disorder closely linked to potent medications currently prescribed to more than 5% of the population - deserves ongoing scrutiny. Major social and ethical problems arise from vague-symptom medicalization, neurological disorder trivialization, medication overuse, and controlled substances diversion to healthy persons for nonmedical purposes. We argue against the 'spectrumization' of ADHD in an effort to curtail further diagnosis creep.
Collapse
Affiliation(s)
- William D Graf
- Departments of Pediatrics and Neurology, Yale University, PO Box 208064, 333 Cedar Street, New Haven, CT 06510, USA
| | | | | |
Collapse
|