1
|
Nugiel T, Fogleman ND, Sheridan MA, Cohen JR. Methylphenidate stabilizes dynamic brain network organization during tasks probing attention and reward processing in stimulant-naïve children with ADHD. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.01.27.25321175. [PMID: 39974117 PMCID: PMC11838951 DOI: 10.1101/2025.01.27.25321175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Children with ADHD often exhibit fluctuations in attention and heightened reward sensitivity. Psychostimulants, such as methylphenidate (MPH), improve these behaviors in many, but not all, children with ADHD. Given the extent to which psychostimulants are prescribed for children, coupled with variable efficacy on an individual level, a better understanding of the mechanisms through which MPH changes brain function and behavior is necessary. MPH's primary action is on catecholamines, including dopamine and norepinephrine. Catecholaminergic signaling can influence the tradeoff between flexibility and stability of brain function, which is one candidate mechanism through which MPH may alter brain function and behavior. Time-varying functional connectivity, which models how functional brain networks reconfigure on short timescales, can be used to examine brain flexibility versus stability, and is thus well-suited to test how MPH impacts brain function. Here, we scanned stimulant-naïve children with ADHD (8-12 years) on and off a single dose of MPH. In the MRI machine, participants completed two attention-demanding tasks: 1) a standard go/no-go task and 2) a rewarded go/no-go task. For both tasks, using a within-subjects design, we compared the degree to which brain organization changed throughout the course of the MRI scan, termed whole brain flexibility, on and off MPH. We found that whole brain flexibility decreased on MPH. Further, individuals with greater decreases in whole brain flexibility on MPH exhibited greater improvements in task performance. Together, these results provide novel insights into the neurobiological mechanisms underlying the effectiveness of MPH administration for children with ADHD.
Collapse
|
2
|
Koirala S, Grimsrud G, Mooney MA, Larsen B, Feczko E, Elison JT, Nelson SM, Nigg JT, Tervo-Clemmens B, Fair DA. Neurobiology of attention-deficit hyperactivity disorder: historical challenges and emerging frontiers. Nat Rev Neurosci 2024; 25:759-775. [PMID: 39448818 DOI: 10.1038/s41583-024-00869-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/19/2024] [Indexed: 10/26/2024]
Abstract
Extensive investigations spanning multiple levels of inquiry, from genetic to behavioural studies, have sought to unravel the mechanistic foundations of attention-deficit hyperactivity disorder (ADHD), with the aspiration of developing efficacious treatments for this condition. Despite these efforts, the pathogenesis of ADHD remains elusive. In this Review, we reflect on what has been learned about ADHD while also providing a framework that may serve as a roadmap for future investigations. We emphasize that ADHD is a highly heterogeneous disorder with multiple aetiologies that necessitates a multifactorial dimensional phenotype, rather than a fixed dichotomous conceptualization. We highlight new findings that suggest a more brain-wide, 'global' view of the disorder, rather than the traditional localizationist framework, which asserts that a limited set of brain regions or networks underlie ADHD. Last, we underscore how underpowered studies that have aimed to associate neurobiology with ADHD phenotypes have long precluded the field from making progress. However, a new age of ADHD research with refined phenotypes, advanced methods, creative study designs and adequately powered investigations is beginning to put the field on a good footing. Indeed, the field is at a promising juncture to advance the neurobiological understanding of ADHD and fulfil the promise of clinical utility.
Collapse
Affiliation(s)
- Sanju Koirala
- Institute of Child Development, University of Minnesota, Minneapolis, MN, USA
- Masonic Institute for the Developing Brain, University of Minnesota, Minneapolis, MN, USA
| | - Gracie Grimsrud
- Masonic Institute for the Developing Brain, University of Minnesota, Minneapolis, MN, USA
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA
| | - Michael A Mooney
- Division of Bioinformatics and Computational Biology, Department of Medical Informatics and Clinical Epidemiology, Oregon Health & Science University, Portland, OR, USA
- Departments of Psychiatry, Oregon Health & Science University, Portland, OR, USA
- Center for Mental Health Innovation, Oregon Health & Science University, Portland, OR, USA
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
| | - Bart Larsen
- Masonic Institute for the Developing Brain, University of Minnesota, Minneapolis, MN, USA
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA
| | - Eric Feczko
- Masonic Institute for the Developing Brain, University of Minnesota, Minneapolis, MN, USA
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA
| | - Jed T Elison
- Institute of Child Development, University of Minnesota, Minneapolis, MN, USA
- Masonic Institute for the Developing Brain, University of Minnesota, Minneapolis, MN, USA
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA
| | - Steven M Nelson
- Masonic Institute for the Developing Brain, University of Minnesota, Minneapolis, MN, USA
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA
| | - Joel T Nigg
- Departments of Psychiatry, Oregon Health & Science University, Portland, OR, USA
- Center for Mental Health Innovation, Oregon Health & Science University, Portland, OR, USA
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, USA
| | - Brenden Tervo-Clemmens
- Masonic Institute for the Developing Brain, University of Minnesota, Minneapolis, MN, USA
- Department of Psychiatry and Behavioral Sciences, University of Minnesota, Minneapolis, MN, USA
| | - Damien A Fair
- Institute of Child Development, University of Minnesota, Minneapolis, MN, USA.
- Masonic Institute for the Developing Brain, University of Minnesota, Minneapolis, MN, USA.
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
3
|
Matt SM, Nolan R, Manikandan S, Agarwal Y, Channer B, Oteju O, Daniali M, Canagarajah JA, LuPone T, Mompho K, Runner K, Nickoloff-Bybel E, Li B, Niu M, Schlachetzki JCM, Fox HS, Gaskill PJ. Dopamine-driven Increase in IL-1β in Myeloid Cells is Mediated by Differential Dopamine Receptor Expression and Exacerbated by HIV. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.09.598137. [PMID: 38915663 PMCID: PMC11195146 DOI: 10.1101/2024.06.09.598137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
The catecholamine neurotransmitter dopamine is classically known for regulation of central nervous system (CNS) functions such as reward, movement, and cognition. Increasing evidence also indicates that dopamine regulates critical functions in peripheral organs and is an important immunoregulatory factor. We have previously shown that dopamine increases NF-κB activity, inflammasome activation, and the production of inflammatory cytokines such as IL-1β in human macrophages. As myeloid lineage cells are central to the initiation and resolution of acute inflammatory responses, dopamine-mediated dysregulation of these functions could both impair the innate immune response and exacerbate chronic inflammation. However, the exact pathways by which dopamine drives myeloid inflammation are not well defined, and studies in both rodent and human systems indicate that dopamine can impact the production of inflammatory mediators through both D1-like dopamine receptors (DRD1, DRD5) and D2-like dopamine receptors (DRD2, DRD3, and DRD4). Therefore, we hypothesized that dopamine-mediated production of IL-1β in myeloid cells is regulated by the ratio of different dopamine receptors that are activated. Our data in primary human monocyte-derived macrophages (hMDM) indicate that DRD1 expression is necessary for dopamine-mediated increases in IL-1β, and that changes in the expression of DRD2 and other dopamine receptors can alter the magnitude of the dopamine-mediated increase in IL-1β. Mature hMDM have a high D1-like to D2-like receptor ratio, which is different relative to monocytes and peripheral blood mononuclear cells (PBMCs). We further confirm in human microglia cell lines that a high ratio of D1-like to D2-like receptors promotes dopamine-induced increases in IL-1β gene and protein expression using pharmacological inhibition or overexpression of dopamine receptors. RNA-sequencing of dopamine-treated microglia shows that genes encoding functions in IL-1β signaling pathways, microglia activation, and neurotransmission increased with dopamine treatment. Finally, using HIV as an example of a chronic inflammatory disease that is substantively worsened by comorbid substance use disorders (SUDs) that impact dopaminergic signaling, we show increased effects of dopamine on inflammasome activation and IL-1β in the presence of HIV in both human macrophages and microglia. These data suggest that use of addictive substances and dopamine-modulating therapeutics could dysregulate the innate inflammatory response and exacerbate chronic neuroimmunological conditions like HIV. Thus, a detailed understanding of dopamine-mediated changes in inflammation, in particular pathways regulating IL-1β, will be critical to effectively tailor medication regimens.
Collapse
|
4
|
Kofler MJ, Groves NB, Chan ESM, Marsh CL, Cole AM, Gaye F, Cibrian E, Tatsuki MO, Singh LJ. Working memory and inhibitory control deficits in children with ADHD: an experimental evaluation of competing model predictions. Front Psychiatry 2024; 15:1277583. [PMID: 38779551 PMCID: PMC11110569 DOI: 10.3389/fpsyt.2024.1277583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 03/07/2024] [Indexed: 05/25/2024] Open
Abstract
Introduction Children with ADHD demonstrate difficulties on many different neuropsychological tests. However, it remains unclear whether this pattern reflects a large number of distinct deficits or a small number of deficit(s) that broadly impact test performance. The current study is among the first experiments to systematically manipulate demands on both working memory and inhibition, with implications for competing conceptual models of ADHD pathogenesis. Method A clinically evaluated, carefully phenotyped sample of 110 children with ADHD, anxiety disorders, or co-occurring ADHD+anxiety (Mage=10.35, 44 girls; 69% White Not Hispanic/Latino) completed a counterbalanced, double dissociation experiment, with two tasks each per inhibition (low vs. high) x working memory (low vs. high) condition. Results Bayesian and frequentist models converged in indicating that both manipulations successfully increased demands on their target executive function (BF10>5.33x108, p<.001). Importantly, occupying children's limited capacity working memory system produced slower response times and reduced accuracy on inhibition tasks (BF10>317.42, p<.001, d=0.67-1.53). It also appeared to differentially reduce inhibition (and non-inhibition) accuracy for children with ADHD relative to children with anxiety (BF10=2.03, p=.02, d=0.50). In contrast, there was strong evidence against models that view working memory deficits as secondary outcomes of underlying inhibition deficits in ADHD (BF01=18.52, p=.85). Discussion This pattern indicates that working memory broadly affects children's ability to inhibit prepotent tendencies and maintain fast/accurate performance, and may explain the errors that children with ADHD make on inhibition tests. These findings are broadly consistent with models describing working memory as a causal mechanism that gives rise to secondary impairments. In contrast, these findings provide evidence against models that view disinhibition as a cause of working memory difficulties or view working memory as a non-causal correlate or epiphenomenon in ADHD.
Collapse
Affiliation(s)
- Michael J. Kofler
- Department of Psychology, Florida State University, Tallahassee, FL, United States
| | - Nicole B. Groves
- Department of Psychiatry, Seattle Children’s Hospital, Seattle, WA, United States
| | - Elizabeth S. M. Chan
- Graduate School of Applied and Professional Psychology, Rutgers University, New Brunswick, NJ, United States
| | - Carolyn L. Marsh
- Department of Psychology, Florida State University, Tallahassee, FL, United States
| | - Alissa M. Cole
- Department of Psychology, Florida State University, Tallahassee, FL, United States
| | - Fatou Gaye
- Department of Psychology, Florida State University, Tallahassee, FL, United States
| | - Enrique Cibrian
- Department of Psychology, Florida State University, Tallahassee, FL, United States
| | - Miho O. Tatsuki
- Department of Psychology, Florida State University, Tallahassee, FL, United States
| | - Leah J. Singh
- Department of Psychology, Florida State University, Tallahassee, FL, United States
| |
Collapse
|
5
|
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
|
6
|
Egeland J, Lund O, Kowalik-Gran I, Aarlien AK, Söderlund GBW. Effects of auditory white noise stimulation on sustained attention and response time variability. Front Psychol 2023; 14:1301771. [PMID: 38144987 PMCID: PMC10748431 DOI: 10.3389/fpsyg.2023.1301771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 11/17/2023] [Indexed: 12/26/2023] Open
Abstract
Introduction "The moderate brain arousal model" claims that white noise improves attention by optimizing brain arousal. We analyze Conners' Continuous Performance Test-3 (CCPT-3) performance, expecting to find reduced reaction time variability with noise mediated by decrease under long event-rates and in later parts of the test, indicating that noise reverse fall in phasic and tonic arousal. Methods Sixty-five children with high or lower ADHD-symptoms from a child psychiatric unit, succeeded to complete the CCPT-3 with and without white noise. Results Noise reduced overall variability, improved performance in later parts of the test, and reduced response variability under the longest event rate particularly in the high symptoms group. No overall change in omissions and commissions, but the high symptoms group made fewer omissions during noise compared the low symptom group. Discussion The study indicates an arousal effect of noise but should be replicated with other noise variants and amplitudes to improve effect and compliance.
Collapse
Affiliation(s)
- Jens Egeland
- Divison of Mental Health & Addiction, Vestold Hospital Trust, Tønsberg, Norway
- Department of Psychology, University of Oslo, Oslo, Norway
| | - Olaf Lund
- Divison of Mental Health & Addiction, Vestold Hospital Trust, Tønsberg, Norway
| | - Iwona Kowalik-Gran
- Divison of Mental Health & Addiction, Vestold Hospital Trust, Tønsberg, Norway
| | | | - Göran B. W. Söderlund
- Faculty of Teacher Education Arts and Sports, Western Norway University of Applied Sciences, Sogndal, Norway
- Department of Education and Special Education, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
7
|
van Roessel PJ, Grassi G, Aboujaoude EN, Menchón JM, Van Ameringen M, Rodríguez CI. Treatment-resistant OCD: Pharmacotherapies in adults. Compr Psychiatry 2023; 120:152352. [PMID: 36368186 DOI: 10.1016/j.comppsych.2022.152352] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 09/20/2022] [Accepted: 10/24/2022] [Indexed: 12/13/2022] Open
Abstract
Serotonin reuptake inhibitor (SRI) medications are well established as first-line pharmacotherapeutic treatment for Obsessive-Compulsive Disorder (OCD). However, despite the excellent safety profile and demonstrated efficacy of these medications, a substantial proportion of individuals with OCD fail to attain sufficient benefit from SRIs. In this narrative review, we discuss clinical features of OCD that have been associated with poorer response to SRIs, and we present pharmacotherapeutic interventions that have been explored as augmenting or alternative treatments for treatment-resistant OCD. We additionally highlight non-SRI interventions for OCD that are currently under investigation. Pharmacotherapeutic interventions were identified via expert consensus. To assess the evidence base for individual pharmacotherapies, targeted searches for relevant English-language publications were performed on standard biomedical research databases, including MEDLINE. Information relevant to ongoing registered clinical trials in OCD was obtained by search of ClinicalTrials.gov. Pharmacotherapies are grouped for review in accordance with the general principles of Neuroscience-based Nomenclature (NbN). Clinical features of OCD that may suggest poorer response to SRI treatment include early age of onset, severity of illness, duration of untreated illness, and the presence of symmetry/ordering or hoarding-related symptoms. Based on evolving pathophysiologic models of OCD, diverse agents engaging serotonin, dopamine, norepinephrine, glutamate, and anti-inflammatory pathways have been explored as alternative or adjunctive therapies for treatment-resistant OCD and have at least preliminary evidence of efficacy. Medications with dopamine antagonist activity remain the most robustly evidence-based of augmenting interventions, yet dopamine antagonists benefit only a minority of those who try them and carry elevated risks of adverse effects. Interventions targeting glutamatergic and anti-inflammatory pathways are less well evidenced, but may offer more favorable benefit to risk profiles. Ongoing research should explore whether specific interventions may benefit individuals with particular features of treatment-resistant OCD.
Collapse
Affiliation(s)
- Peter J van Roessel
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA; Sierra Pacific Mental Illness Research, Education, and Clinical Center (MIRECC), VA Palo Alto Health Care System, Palo Alto, CA, USA.
| | | | - Elias N Aboujaoude
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
| | - José M Menchón
- Department of Psychiatry, Bellvitge University Hospital-IDIBELL, University of Barcelona, Cibersam, Barcelona, Spain
| | - Michael Van Ameringen
- Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton, Ontario, Canada
| | - Carolyn I Rodríguez
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA; Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA.
| |
Collapse
|
8
|
van den Bosch R, Lambregts B, Määttä J, Hofmans L, Papadopetraki D, Westbrook A, Verkes RJ, Booij J, Cools R. Striatal dopamine dissociates methylphenidate effects on value-based versus surprise-based reversal learning. Nat Commun 2022; 13:4962. [PMID: 36002446 PMCID: PMC9402573 DOI: 10.1038/s41467-022-32679-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 08/10/2022] [Indexed: 11/13/2022] Open
Abstract
Psychostimulants such as methylphenidate are widely used for their cognitive enhancing effects, but there is large variability in the direction and extent of these effects. We tested the hypothesis that methylphenidate enhances or impairs reward/punishment-based reversal learning depending on baseline striatal dopamine levels and corticostriatal gating of reward/punishment-related representations in stimulus-specific sensory cortex. Young healthy adults (N = 100) were scanned with functional magnetic resonance imaging during a reward/punishment reversal learning task, after intake of methylphenidate or the selective D2/3-receptor antagonist sulpiride. Striatal dopamine synthesis capacity was indexed with [18F]DOPA positron emission tomography. Methylphenidate improved and sulpiride decreased overall accuracy and response speed. Both drugs boosted reward versus punishment learning signals to a greater degree in participants with higher dopamine synthesis capacity. By contrast, striatal and stimulus-specific sensory surprise signals were boosted in participants with lower dopamine synthesis. These results unravel the mechanisms by which methylphenidate gates both attention and reward learning. The mechanisms underpinning the variability in methylphenidate’s effects on cognition remain unclear. Here, the authors show that such effects reflect changes in striatal dopamine-related output gating of task-relevant cortical signals, and that these changes depend on baseline dopamine synthesis capacity.
Collapse
Affiliation(s)
- Ruben van den Bosch
- Radboud University, Donders Institute for Brain, Cognition and Behaviour, Nijmegen, The Netherlands.
| | - Britt Lambregts
- Radboud University Medical Center, Department of Psychiatry, Donders Institute for Brain, Cognition and Behaviour, Nijmegen, The Netherlands
| | - Jessica Määttä
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Lieke Hofmans
- Department of Developmental Psychology, University of Amsterdam, Amsterdam, The Netherlands
| | - Danae Papadopetraki
- Radboud University Medical Center, Department of Psychiatry, Donders Institute for Brain, Cognition and Behaviour, Nijmegen, The Netherlands
| | - Andrew Westbrook
- Cognitive, Linguistic & Psychological Sciences Department, Brown University, Providence, RI, USA
| | - Robbert-Jan Verkes
- Radboud University Medical Center, Department of Psychiatry, Donders Institute for Brain, Cognition and Behaviour, Nijmegen, The Netherlands
| | - Jan Booij
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Centers, location Academic Medical Center, Amsterdam, The Netherlands.,Radboud University Medical Center, Department of Medical Imaging, Nijmegen, The Netherlands
| | - Roshan Cools
- Radboud University Medical Center, Department of Psychiatry, Donders Institute for Brain, Cognition and Behaviour, Nijmegen, The Netherlands
| |
Collapse
|
9
|
Stewart SH, Chinneck A, Thompson K, Afzali MH, Nogueira-Arjona R, Mahu IT, Conrod PJ. Personality to Prescription Drug Misuse in Adolescents: Testing Affect Regulation, Psychological Dysregulation, and Deviance Proneness Pathways. Front Psychiatry 2021; 12:640766. [PMID: 33986700 PMCID: PMC8110923 DOI: 10.3389/fpsyt.2021.640766] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Accepted: 03/17/2021] [Indexed: 11/13/2022] Open
Abstract
Background: Fifteen to 25-year-olds are the age group most likely to misuse prescription drugs. Few studies have tested theory-driven models of adolescent risk for prescription drug misuse. Moreover, rarely are distinct pathways to different forms of prescription drug misuse considered. Methods: We tested mediational paths from personality to mental health symptoms to prescription drug misuse, informed by etiological models of addiction. We specified pathways from particular personality traits to unique forms of prescription drug misuse via specific mental health symptoms. We used semi-longitudinal data collected across two waves of the Co-Venture Trial. Our sample included students from 31 Canadian high schools tested in Grade 9 (n = 3,024) and again in Grade 10 (n = 2,869; 95% retention). Personality (hopelessness, anxiety sensitivity, impulsivity, sensation seeking) was assessed in Grade 9. Mental health symptoms (depression, anxiety, ADHD, conduct disorder) and prescription drug misuse (opioids, sedatives/tranquilizers, stimulants) were assessed at both time points. Results: Consistent with the negative affect regulation model, hopelessness was specifically associated with opioid misuse via depressive symptoms, and anxiety sensitivity was specifically associated with sedative/tranquilizer misuse via anxiety symptoms. Consistent with positive affect regulation, sensation seeking was directly associated with stimulant misuse. Consistent with the psychological dysregulation model, impulsivity was associated with stimulant misuse via ADHD symptoms. And consistent with the deviance proneness model, impulsivity was also associated with unconstrained (i.e., all three forms of) prescription drug misuse via conduct disorder symptoms. Conclusions: Screening for adolescents high in hopelessness, anxiety sensitivity, sensation seeking, or impulsivity and providing them with personality-matched cognitive-behavioral interventions may be helpful in preventing or mitigating prescription drug misuse. Our results point to the specific mental health symptoms that are important to target in each of these personality-matched interventions.
Collapse
Affiliation(s)
- Sherry H. Stewart
- Department of Psychiatry, Dalhousie University, Halifax, NS, Canada
- Department of Psychology & Neuroscience, Dalhousie University, Halifax, NS, Canada
| | - Annie Chinneck
- Department of Psychology & Neuroscience, Dalhousie University, Halifax, NS, Canada
| | - Kara Thompson
- Department of Psychology, St. Francis Xavier University, Antigonish, NS, Canada
| | | | | | - Ioan T. Mahu
- Department of Psychology & Neuroscience, Dalhousie University, Halifax, NS, Canada
| | | |
Collapse
|
10
|
Attention-deficit/hyperactivity disorder and the explore/exploit trade-off. Neuropsychopharmacology 2021; 46:614-621. [PMID: 33040092 PMCID: PMC8027173 DOI: 10.1038/s41386-020-00881-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 09/22/2020] [Accepted: 09/29/2020] [Indexed: 11/09/2022]
Abstract
The ability to maximize rewards and minimize the costs of obtaining them is vital to making advantageous explore/exploit decisions. Exploratory decisions are theorized to be greater among individuals with attention-deficit/hyperactivity disorder (ADHD), potentially due to deficient catecholamine transmission. Here, we examined the effects of ADHD status and methylphenidate, a common ADHD medication, on explore/exploit decisions using a 6-armed bandit task. We hypothesized that ADHD participants would make more exploratory decisions than controls, and that MPH would reduce group differences. On separate study days, adults with (n = 26) and without (n = 23) ADHD completed the bandit task at baseline, and after methylphenidate or placebo in counter-balanced order. Explore/exploit decisions were modeled using reinforcement learning algorithms. ADHD participants made more exploratory decisions (i.e., chose options without the highest expected reward value) and earned fewer points than controls in all three study days, and methylphenidate did not affect these outcomes. Baseline exploratory choices were positively associated with hyperactive ADHD symptoms across all participants. These results support several theoretical models of increased exploratory choices in ADHD and suggest the unexplained variance in ADHD decisions may be due to less value tracking. The inability to suppress actions with little to no reward value may be a key feature of hyperactive ADHD symptoms.
Collapse
|
11
|
Bluschke A, Zink N, Mückschel M, Roessner V, Beste C. A novel approach to intra-individual performance variability in ADHD. Eur Child Adolesc Psychiatry 2021; 30:733-745. [PMID: 32410131 PMCID: PMC8060200 DOI: 10.1007/s00787-020-01555-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Accepted: 05/01/2020] [Indexed: 11/26/2022]
Abstract
Patients with attention deficit/(hyperactivity) disorder (AD(H)D) show increased intra-individual variability (IIV) in behavioral performance. This likely reflects dopaminergic deficiencies. However, the precise performance profile across time and the pattern of fluctuations within it have not yet been considered, partly due to insufficient methods. Yet, such an analysis may yield important theory-based implications for clinical practice. Thus, in a case-control cross-sectional study, we introduce a new method to investigate performance fluctuations in patients with ADD (n = 76) and ADHD (n = 67) compared to healthy controls (n = 45) in a time estimation task. In addition, we also evaluate the effects of methylphenidate (MPH) treatment on this performance pattern in 29 patients with AD(H)D. Trial-by-trial differences in performance between healthy controls and patients with AD(H)D do not persist continuously over longer time periods. Periods during which no differences in performance between healthy controls and patients occur alternate with periods in which such differences are present. AD(H)D subtype and surprisingly also medication status does not affect this pattern. The presented findings likely reflect (phasic) deficiencies of the dopaminergic system in patients with AD(H)D which are not sufficiently ameliorated by first-line pharmacological treatment. The presented findings carry important clinical and scientific implications.
Collapse
Affiliation(s)
- Annet Bluschke
- Cognitive Neurophysiology, Department of Child and Adolescent Psychiatry, Faculty of Medicine, TU Dresden, Fetscherstrasse 74, 01307, Dresden, Germany
| | - Nicolas Zink
- Cognitive Neurophysiology, Department of Child and Adolescent Psychiatry, Faculty of Medicine, TU Dresden, Fetscherstrasse 74, 01307, Dresden, Germany
| | - Moritz Mückschel
- Cognitive Neurophysiology, Department of Child and Adolescent Psychiatry, Faculty of Medicine, TU Dresden, Fetscherstrasse 74, 01307, Dresden, Germany
| | - Veit Roessner
- Cognitive Neurophysiology, Department of Child and Adolescent Psychiatry, Faculty of Medicine, TU Dresden, Fetscherstrasse 74, 01307, Dresden, Germany
| | - Christian Beste
- Cognitive Neurophysiology, Department of Child and Adolescent Psychiatry, Faculty of Medicine, TU Dresden, Fetscherstrasse 74, 01307, Dresden, Germany.
| |
Collapse
|
12
|
Froböse MI, Westbrook A, Bloemendaal M, Aarts E, Cools R. Catecholaminergic modulation of the cost of cognitive control in healthy older adults. PLoS One 2020; 15:e0229294. [PMID: 32084218 PMCID: PMC7034873 DOI: 10.1371/journal.pone.0229294] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Accepted: 02/03/2020] [Indexed: 11/19/2022] Open
Abstract
Catecholamines have long been associated with cognitive control and value-based decision-making. More recently, we have shown that catecholamines also modulate value-based decision-making about whether or not to engage in cognitive control. Yet it is unclear whether catecholamines influence these decisions by altering the subjective value of control. Thus, we tested whether tyrosine, a catecholamine precursor altered the subjective value of performing a demanding working memory task among healthy older adults (60-75 years). Contrary to our prediction, tyrosine administration did not significantly increase the subjective value of conducting an N-back task for reward, as a main effect. Instead, in line with our previous study, exploratory analyses indicated that drug effects varied as a function of participants' trait impulsivity scores. Specifically, tyrosine increased the subjective value of conducting an N-back task in low impulsive participants, while reducing its value in more impulsive participants. One implication of these findings is that the over-the-counter tyrosine supplements may be accompanied by an undermining effect on the motivation to perform demanding cognitive tasks, at least in certain older adults. Taken together, these findings indicate that catecholamines can alter cognitive control by modulating motivation (rather than just the ability) to exert cognitive control.
Collapse
Affiliation(s)
- Monja I. Froböse
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, The Netherlands
- Institute of Experimental Psychology, Heinrich-Heine University, Düsseldorf, Germany
| | - Andrew Westbrook
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, The Netherlands
- Cognitive, Linguistic, and Psychological Sciences, Brown University, Providence, RI, United States of America
| | - Mirjam Bloemendaal
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, The Netherlands
| | - Esther Aarts
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, The Netherlands
| | - Roshan Cools
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, The Netherlands
- Dept Psychiatry, Radboud University Medical Centre, Nijmegen, The Netherlands
| |
Collapse
|
13
|
Furukawa E, da Costa RQM, Bado P, Hoefle S, Vigne P, Monteiro M, Wickens JR, Moll J, Tripp G, Mattos P. Methylphenidate modifies reward cue responses in adults with ADHD: An fMRI study. Neuropharmacology 2020; 162:107833. [DOI: 10.1016/j.neuropharm.2019.107833] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Revised: 10/28/2019] [Accepted: 10/29/2019] [Indexed: 01/01/2023]
|
14
|
Fuller JA, Burrell MH, Yee AG, Liyanagama K, Lipski J, Wickens JR, Hyland BI. Role of homeostatic feedback mechanisms in modulating methylphenidate actions on phasic dopamine signaling in the striatum of awake behaving rats. Prog Neurobiol 2019; 182:101681. [DOI: 10.1016/j.pneurobio.2019.101681] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 07/25/2019] [Accepted: 08/06/2019] [Indexed: 12/13/2022]
|
15
|
Cools R. Chemistry of the Adaptive Mind: Lessons from Dopamine. Neuron 2019; 104:113-131. [DOI: 10.1016/j.neuron.2019.09.035] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 09/19/2019] [Accepted: 09/20/2019] [Indexed: 12/21/2022]
|
16
|
Sethi A, Voon V, Critchley HD, Cercignani M, Harrison NA. A neurocomputational account of reward and novelty processing and effects of psychostimulants in attention deficit hyperactivity disorder. Brain 2019; 141:1545-1557. [PMID: 29547978 PMCID: PMC5917772 DOI: 10.1093/brain/awy048] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Accepted: 01/06/2018] [Indexed: 01/01/2023] Open
Abstract
Computational models of reinforcement learning have helped dissect discrete components of reward-related function and characterize neurocognitive deficits in psychiatric illnesses. Stimulus novelty biases decision-making, even when unrelated to choice outcome, acting as if possessing intrinsic reward value to guide decisions toward uncertain options. Heightened novelty seeking is characteristic of attention deficit hyperactivity disorder, yet how this influences reward-related decision-making is computationally encoded, or is altered by stimulant medication, is currently uncertain. Here we used an established reinforcement-learning task to model effects of novelty on reward-related behaviour during functional MRI in 30 adults with attention deficit hyperactivity disorder and 30 age-, sex- and IQ-matched control subjects. Each participant was tested on two separate occasions, once ON and once OFF stimulant medication. OFF medication, patients with attention deficit hyperactivity disorder showed significantly impaired task performance (P = 0.027), and greater selection of novel options (P = 0.004). Moreover, persistence in selecting novel options predicted impaired task performance (P = 0.025). These behavioural deficits were accompanied by a significantly lower learning rate (P = 0.011) and heightened novelty signalling within the substantia nigra/ventral tegmental area (family-wise error corrected P < 0.05). Compared to effects in controls, stimulant medication improved attention deficit hyperactivity disorder participants' overall task performance (P = 0.011), increased reward-learning rates (P = 0.046) and enhanced their ability to differentiate optimal from non-optimal novel choices (P = 0.032). It also reduced substantia nigra/ventral tegmental area responses to novelty. Preliminary cross-sectional evidence additionally suggested an association between long-term stimulant treatment and a reduction in the rewarding value of novelty. These data suggest that aberrant substantia nigra/ventral tegmental area novelty processing plays an important role in the suboptimal reward-related decision-making characteristic of attention deficit hyperactivity disorder. Compared to effects in controls, abnormalities in novelty processing and reward-related learning were improved by stimulant medication, suggesting that they may be disorder-specific targets for the pharmacological management of attention deficit hyperactivity disorder symptoms.
Collapse
Affiliation(s)
- Arjun Sethi
- Department of Neuroscience, Brighton and Sussex Medical School, University of Sussex, Brighton, UK
| | - Valerie Voon
- Department of Psychiatry, University of Cambridge, Cambridge, UK.,Cambridgeshire and Peterborough NHS Foundation Trust, Cambridge, UK
| | - Hugo D Critchley
- Department of Neuroscience, Brighton and Sussex Medical School, University of Sussex, Brighton, UK.,Sackler Centre for Consciousness Science, University of Sussex, Brighton, UK.,Sussex Partnership NHS Foundation Trust, Brighton, UK
| | - Mara Cercignani
- Department of Neuroscience, Brighton and Sussex Medical School, University of Sussex, Brighton, UK
| | - Neil A Harrison
- Department of Neuroscience, Brighton and Sussex Medical School, University of Sussex, Brighton, UK.,Sackler Centre for Consciousness Science, University of Sussex, Brighton, UK.,Sussex Partnership NHS Foundation Trust, Brighton, UK
| |
Collapse
|
17
|
Zheng H, Jia F, Han H, Wang S, Guo G, Quan D, Li G, Huang H. Combined fluvoxamine and extended-release methylphenidate improved treatment response compared to fluvoxamine alone in patients with treatment-refractory obsessive-compulsive disorder: A randomized double-blind, placebo-controlled study. Eur Neuropsychopharmacol 2019; 29:397-404. [PMID: 30595354 DOI: 10.1016/j.euroneuro.2018.12.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Revised: 11/30/2018] [Accepted: 12/16/2018] [Indexed: 01/04/2023]
Abstract
More effective, tolerable interventions for treatment-refractory obsessive-compulsive disorder (OCD) are needed. Preliminary findings encourage optimism that methylphenidate augmentation may be of benefit in the treatment of OCD. To test modulator methylphenidate (MPH) of extended-release formulations (MPH-ER) a safe and effective add-on therapy for refractory OCD, a pilot randomized, placebo-controlled, double-blind trial was conducted at an outpatient, single-center academic setting. Participants included 44 adults with serotonin reuptake inhibitor (SRI) treatment-refractory OCD and receiving a stable fluvoxamine pharmacotherapy with Yale-Brown Obsessive Compulsive Scale (Y-BOCS) scores higher than 20. Data were analyzed in the intention-to-treat sample. All subjects were randomized into two parallel groups to receive fluvoxamine (250 mg daily) plus MPH-ER (36 mg daily) or fluvoxamine (250 mg daily) plus identical placebo tablets under double-blind conditions and followed for 8 weeks. Forty-four patients (29 [66%] men), with a mean (SD) age of 24.7 (6) years participated; with a mean (SD) duration of episode 5.7 (3) were randomized and forty-one finished the trial. In the intention-to-treat analysis, the improvement in the Y-BOCS total score and Y-BOCS obsession subscale score was more prominent in the fluvoxamine and MPH-ER group compared with those receiving placebo (P < .001). Additionally, cumulative response rates were higher in the MPH-ER vs placebo groups (59% vs 5%; P < .001). MPH-ER was well tolerated; No subjects dropped out due to side effects. In summary, combined treatment with MPH-ER demonstrated an enhanced clinical rate of response compared to placebo. Further trials should examine MPH-ER efficacy in a larger sample.
Collapse
Affiliation(s)
- Huirong Zheng
- Guangdong Mental Health Center, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Affliated School of Medicine of South China University of Technology, No. 123 Huifu Xi Road, Guangzhou 510180, Guangdong, PR China.
| | - Fujun Jia
- Guangdong Mental Health Center, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Affliated School of Medicine of South China University of Technology, No. 123 Huifu Xi Road, Guangzhou 510180, Guangdong, PR China
| | - Hongying Han
- Department of Psychiatry, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, PR China
| | - Shibin Wang
- Guangdong Mental Health Center, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Affliated School of Medicine of South China University of Technology, No. 123 Huifu Xi Road, Guangzhou 510180, Guangdong, PR China
| | - Guangquan Guo
- Guangdong Mental Health Center, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Affliated School of Medicine of South China University of Technology, No. 123 Huifu Xi Road, Guangzhou 510180, Guangdong, PR China
| | - Dongming Quan
- Guangdong Mental Health Center, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Affliated School of Medicine of South China University of Technology, No. 123 Huifu Xi Road, Guangzhou 510180, Guangdong, PR China
| | - Gang Li
- Guangdong Mental Health Center, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Affliated School of Medicine of South China University of Technology, No. 123 Huifu Xi Road, Guangzhou 510180, Guangdong, PR China
| | - Huiyan Huang
- Pharmacy Department of Guangdong General Hospital, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, PR China
| |
Collapse
|
18
|
Beu ND, Burns NR, Baetu I. Polymorphisms in dopaminergic genes predict proactive processes of response inhibition. Eur J Neurosci 2019; 49:1127-1148. [DOI: 10.1111/ejn.14323] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 11/28/2018] [Accepted: 12/12/2018] [Indexed: 01/11/2023]
Affiliation(s)
- Nathan D. Beu
- The School of Psychology University of Adelaide Adelaide South Australia Australia
| | - Nicholas R. Burns
- The School of Psychology University of Adelaide Adelaide South Australia Australia
| | - Irina Baetu
- The School of Psychology University of Adelaide Adelaide South Australia Australia
| |
Collapse
|
19
|
Vogt BA. Cingulate impairments in ADHD: Comorbidities, connections, and treatment. HANDBOOK OF CLINICAL NEUROLOGY 2019; 166:297-314. [PMID: 31731917 DOI: 10.1016/b978-0-444-64196-0.00016-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
The entire cingulate cortex is engaged in the structure/function abnormalities found in attention-deficit/hyperactivity disorder (ADHD). In ADHD, which is the most common developmental disease, impaired impulse control and cognition often trace to anterior midcingulate cortex (aMCC) in Go/No-go tests, decoding and reading, the Stroop Color and Word Test, and the Wisconsin Card Sorting Test (WCST), with volume deficits in anterior cingulate cortex (ACC) and posterior midcingulate cortex (pMCC). Volumes in pMCC correlate positively with the WCST and negatively with total and nonperseverative errors on the WCST. Activation and connectivity on N-back tests show connections for high and low spatial working memory, but patients have increased activation in PCC and decreased connectivity between MCC and PCC for high load. Students struggle in class due to malfunctioning aMCC, pregenual anterior cingulate cortex (pACC), and dorsal posterior cingulate cortex (dPCC), and to core deficits in response/task switching in aMCC. Gene mutations are found in the DA transporter and DA4 and DA5 receptors. Methylphenidate decreases hyperactivity in aMCC. The DA system is controlled by cholinergic receptors in the daMCC and genetics show nAChR mutations in alpha 3, 4, and 7 receptors. At 25 years, a modified Eriksen flanker/No-go task and voxel-based morphometry (VBM) show prenatal smoking, lifetime smoking at 13 years, and novelty seeking. Prenatal exposure to nicotine exhibits weaker responses in aMCC during cognitive tasks for hyperactivity/impulsiveness but not inattention. AZD1446 (ɑ4β2 nAChR agonist) improves the Groton Maze task due to high nAChR in dPCC/RSC engaged in spatial orientation. Environmental factors associated with childhood ADHD relate to pesticides, organochlorine, and air pollutants. Network connection segregation shows increased amygdala local nodal, but decreased ACC and PCC connections, reflecting emphasis on local periamygdala connections at the expense of cortical connections. Thus, ADHD children/adolescents respond impulsively to the significance of stimuli without having cortical inhibition. Finally, controls show negative relationships between aMCC and the default mode network, and ADHD compromises this relationship, showing decreased connectivity between ACC and precuneus/PCC.
Collapse
Affiliation(s)
- Brent A Vogt
- Cingulum Neurosciences Institute, Manlius, NY, United States; Department of Anatomy and Neurobiology, Boston University School of Medicine, Boston, MA, United States.
| |
Collapse
|
20
|
Cools R, Froböse M, Aarts E, Hofmans L. Dopamine and the motivation of cognitive control. HANDBOOK OF CLINICAL NEUROLOGY 2019; 163:123-143. [DOI: 10.1016/b978-0-12-804281-6.00007-0] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
21
|
Boyette-Davis JA, Rice HR, Shoubaki RI, Gonzalez CM, Kunkel MN, Lucero DA, Womble PD, Guarraci FA. A recreational dose of methylphenidate, but not methamphetamine, decreases anxiety-like behavior in female rats. Neurosci Lett 2018; 682:21-26. [DOI: 10.1016/j.neulet.2018.06.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2018] [Revised: 06/01/2018] [Accepted: 06/05/2018] [Indexed: 10/14/2022]
|
22
|
|
23
|
Teixeira MZ. Therapeutic use of the rebound effect of modern drugs: "New homeopathic medicines". Rev Assoc Med Bras (1992) 2017; 63:100-108. [PMID: 28355369 DOI: 10.1590/1806-9282.63.02.100] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Accepted: 07/09/2016] [Indexed: 11/22/2022] Open
Abstract
The homeopathic treatment is based on the principle of therapeutic similitude, employing medicines that cause certain disorders to treat similar manifestations, stimulating a reaction of the organism against its own ailments. The occurrence of this secondary reaction of the organism, opposite in nature to the primary action of the medicines, is evidenced in the study of the rebound (paradoxical) effect of several classes of modern drugs. In this work, in addition to substantiate the principle of similitude before the experimental and clinical pharmacology, we suggest a proposal to employ hundreds of conventional drugs according to homeopathic method, applying the therapeutic similitude between the adverse events of medicines and the clinical manifestations of patients. Describing existing lines of research and a specific method for the therapeutic use of the rebound effect of modern drugs (http://www.newhomeopathicmedicines.com), we hope to minimize prejudices related to the homeopathy and contribute to a broadening of the healing art.
Collapse
Affiliation(s)
- Marcus Zulian Teixeira
- MD, PhD, Postdoctoral Student of the Department of Obstetrics and Gynecology, Faculdade de Medicina da Universidade de São Paulo (FMUSP). Coordinator of the Elective Discipline Fundamentals of Homeopathy (MCM0773), FMUSP, São Paulo, SP, Brazil
| |
Collapse
|
24
|
Gomez A, Petrucci AN, Dance L, Morales-Valenzuela J, Gibbs N, Dahlhausen CC, Villatoro JR, Frohardt RF, Guarraci FA. An acute, non-therapeutic dose of methylphenidate disrupts partner preference in female rats. Pharmacol Biochem Behav 2016; 150-151:100-107. [DOI: 10.1016/j.pbb.2016.09.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Revised: 09/19/2016] [Accepted: 09/21/2016] [Indexed: 11/25/2022]
|
25
|
Modelling ADHD: A review of ADHD theories through their predictions for computational models of decision-making and reinforcement learning. Neurosci Biobehav Rev 2016; 71:633-656. [PMID: 27608958 DOI: 10.1016/j.neubiorev.2016.09.002] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Revised: 08/31/2016] [Accepted: 09/04/2016] [Indexed: 01/13/2023]
Abstract
Attention deficit hyperactivity disorder (ADHD) is characterized by altered decision-making (DM) and reinforcement learning (RL), for which competing theories propose alternative explanations. Computational modelling contributes to understanding DM and RL by integrating behavioural and neurobiological findings, and could elucidate pathogenic mechanisms behind ADHD. This review of neurobiological theories of ADHD describes predictions for the effect of ADHD on DM and RL as described by the drift-diffusion model of DM (DDM) and a basic RL model. Empirical studies employing these models are also reviewed. While theories often agree on how ADHD should be reflected in model parameters, each theory implies a unique combination of predictions. Empirical studies agree with the theories' assumptions of a lowered DDM drift rate in ADHD, while findings are less conclusive for boundary separation. The few studies employing RL models support a lower choice sensitivity in ADHD, but not an altered learning rate. The discussion outlines research areas for further theoretical refinement in the ADHD field.
Collapse
|
26
|
Teixeira MZ, Podgaec S, Baracat EC. Protocol of randomized controlled trial of potentized estrogen in homeopathic treatment of chronic pelvic pain associated with endometriosis. HOMEOPATHY 2016; 105:240-249. [PMID: 27473545 DOI: 10.1016/j.homp.2016.03.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Revised: 03/01/2016] [Accepted: 03/09/2016] [Indexed: 11/30/2022]
Abstract
BACKGROUND Endometriosis is a chronic inflammatory disease that causes difficult-to-treat pelvic pain. Thus being, many patients seek help in complementary and alternative medicine, including homeopathy. The effectiveness of homeopathic treatment for endometriosis is controversial due to the lack of evidences in the literature. The aim of the present randomized controlled trial is to assess the efficacy of potentized estrogen compared to placebo in the treatment of chronic pelvic pain associated with endometriosis. METHODS/DESIGN The present is a randomized, double-blind, placebo-controlled trial of a homeopathic medicine individualized according to program 'New Homeopathic Medicines: use of modern drugs according to the principle of similitude' (http://newhomeopathicmedicines.com). Women with endometriosis, chronic pelvic pain and a set of signs and symptoms similar to the adverse events caused by estrogen were recruited at the Endometriosis Unit of Division of Clinical Gynecology, Clinical Hospital, School of Medicine, University of São Paulo (Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo - HCFMUSP). The participants were selected based on the analysis of their medical records and the application of self-report structured questionnaires. A total of 50 women meeting the eligibility criteria will be randomly allocated to receive potentized estrogen or placebo. The primary clinical outcome measure will be severity of chronic pelvic pain. Statistical analysis will be performed on the intention-to-treat and per-protocol approaches comparing the effect of the homeopathic medicine versus placebo after 24 weeks of intervention. DISCUSSION The present study was approved by the research ethics committee of HCFMUSP and the results are expected in 2016. TRIAL REGISTRATION ClinicalTrials.gov Identifier: https://clinicaltrials.gov/ct2/show/NCT02427386.
Collapse
Affiliation(s)
- Marcus Zulian Teixeira
- Department of Obstetrics and Gynecology, School of Medicine, University of Sao Paulo, Sao Paulo, Brazil.
| | - Sérgio Podgaec
- Department of Obstetrics and Gynecology, School of Medicine, University of Sao Paulo, Sao Paulo, Brazil
| | - Edmund Chada Baracat
- Department of Obstetrics and Gynecology, School of Medicine, University of Sao Paulo, Sao Paulo, Brazil
| |
Collapse
|
27
|
Guarraci FA, Holifield C, Morales-Valenzuela J, Greene K, Brown J, Lopez R, Crandall C, Gibbs N, Vela R, Delgado MY, Frohardt RJ. Exposure to methylphenidate during peri-adolescence affects endocrine functioning and sexual behavior in female Long–Evans rats. Pharmacol Biochem Behav 2016; 142:36-41. [DOI: 10.1016/j.pbb.2015.12.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2015] [Revised: 11/21/2015] [Accepted: 12/13/2015] [Indexed: 10/22/2022]
|
28
|
The role of dopamine in the pathophysiology and treatment of apathy. PROGRESS IN BRAIN RESEARCH 2016; 229:389-426. [DOI: 10.1016/bs.pbr.2016.05.007] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
29
|
Impaired reward processing by anterior cingulate cortex in children with attention deficit hyperactivity disorder. COGNITIVE AFFECTIVE & BEHAVIORAL NEUROSCIENCE 2015; 14:698-714. [PMID: 24874420 DOI: 10.3758/s13415-014-0298-3] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Decades of research have examined the neurocognitive mechanisms of cognitive control, but the motivational factors underlying task selection and performance remain to be elucidated. We recently proposed that anterior cingulate cortex (ACC) utilizes reward prediction error signals carried by the midbrain dopamine system to learn the value of tasks according to the principles of hierarchical reinforcement learning. According to this position, disruption of the ACC-dopamine interface can disrupt the selection and execution of extended, task-related behaviors. To investigate this issue, we recorded the event-related brain potential (ERP) from children with attention deficit hyperactivity disorder (ADHD), which is strongly associated with ACC-dopamine dysfunction, and from typically developing children while they navigated a simple "virtual T-maze" to find rewards. Depending on the condition, the feedback stimuli on each trial indicated that the children earned or failed to earn either money or points. We found that the reward positivity, an ERP component proposed to index the impact of dopamine-related reward signals on ACC, was significantly larger with money feedback than with points feedback for the children with ADHD, but not for the typically developing children. These results suggest that disruption of the ACC-dopamine interface may underlie the impairments in motivational control observed in childhood ADHD.
Collapse
|
30
|
Linssen AMW, Sambeth A, Vuurman EFPM, Riedel WJ. Cognitive effects of methylphenidate and levodopa in healthy volunteers. Eur Neuropsychopharmacol 2014; 24:200-6. [PMID: 24119823 DOI: 10.1016/j.euroneuro.2013.09.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2013] [Revised: 09/16/2013] [Accepted: 09/26/2013] [Indexed: 10/26/2022]
Abstract
Our previous study showed enhanced declarative memory consolidation after acute methylphenidate (MPH) administration. The primary aim of the current study was to investigate the duration of this effect. Secondary, the dopaminergic contribution of MPH effects, the electrophysiological correlates of declarative memory, and the specificity of memory enhancing effects of MPH to declarative memory were assessed. Effects of 40 mg of MPH on memory performance were compared to 100mg of levodopa (LEV) in a placebo-controlled crossover study with 30 healthy volunteers. Memory performance testing included a word learning test, the Sternberg memory scanning task, a paired associates learning task, and a spatial working memory task. During the word learning test, event-related brain potentials (ERPs) were measured. MPH failed to enhance retention of words at a 30 min delay, but it improved 24 h delayed memory recall relative to PLA and LEV. Furthermore, during encoding, the P3b and P600 ERP latencies were prolonged and the P600 amplitude was larger after LEV compared to PLA and MPH. MPH speeded response times on the Sternberg Memory Scanning task and improved performance on the Paired Associates Learning task, relative to LEV, but not PLA. Performance on the Spatial working memory task was not affected by the treatments. These findings suggest that MPH and LEV might have opposite effects on memory.
Collapse
Affiliation(s)
- A M W Linssen
- Department of Neuropsychology and Psychopharmacology, Faculty of Psychology and Neuroscience, Maastricht University, PO Box 616, 6200 MD, Maastricht, The Netherlands.
| | - A Sambeth
- Department of Neuropsychology and Psychopharmacology, Faculty of Psychology and Neuroscience, Maastricht University, PO Box 616, 6200 MD, Maastricht, The Netherlands
| | - E F P M Vuurman
- Department of Neuropsychology and Psychopharmacology, Faculty of Psychology and Neuroscience, Maastricht University, PO Box 616, 6200 MD, Maastricht, The Netherlands
| | - W J Riedel
- Department of Neuropsychology and Psychopharmacology, Faculty of Psychology and Neuroscience, Maastricht University, PO Box 616, 6200 MD, Maastricht, The Netherlands
| |
Collapse
|
31
|
Ramos-Quiroga JA, Corominas M, Castells X, Bosch R, Casas M. OROS methylphenidate for the treatment of adults with attention-deficit/hyperactivity disorder. Expert Rev Neurother 2014; 9:1121-31. [DOI: 10.1586/ern.09.65] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
32
|
Genro JP, Kieling C, Rohde LA, Hutz MH. Attention-deficit/hyperactivity disorder and the dopaminergic hypotheses. Expert Rev Neurother 2014; 10:587-601. [DOI: 10.1586/ern.10.17] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
33
|
Jones Z, Dafny N. Acute and chronic dose-response effect of methylphenidate on ventral tegmental area neurons correlated with animal behavior. J Neural Transm (Vienna) 2013; 121:327-45. [PMID: 24249696 PMCID: PMC4743876 DOI: 10.1007/s00702-013-1101-2] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2013] [Accepted: 10/09/2013] [Indexed: 01/14/2023]
Abstract
Methylphenidate (MPD) is used to treat ADHD and as a cognitive enhancement and recreationally. MPD's effects are not fully understood. One of the sites of psychostimulant action is the ventral tegmental area (VTA). The VTA neuronal activity was recorded from freely behaving rats using a wireless system. 51 animals were divided into groups: saline, 0.6, 2.5, and 10.0 mg/kg MPD. The same repetitive MPD dose can elicit either behavioral sensitization or tolerance; thus the evaluation of the VTA neuronal activity was based on the animals' behavioral response to chronic MPD exposure: animals exhibiting behavioral tolerance or sensitization. Acute MPD elicits dose-related increases in behavioral activity. About half of the animals exhibited behavioral sensitization or tolerance to each of the MPD doses. 361 units were recorded from the VTA and exhibited similar spike shape on experimental day 1 (ED1) and on ED10. 71, 84, and 79 % of VTA units responded to acute 0.6, 2.5, and 10.0 mg/kg MPD, respectively. The neuronal baseline activity at ED10 was significantly modified in 94, 95, and 100 % of VTA units following 0.6, 2.5 and 10.0 mg/kg MPD, respectively. Following chronic MPD exposure, 91, 98, and 100 % exhibit either electrophysiological tolerance or sensitization of 0.6, 2.6, or 10.0 mg/kg MPD, respectively. In conclusion, the chronic administration of the same dose of MPD caused some animals to exhibit behavioral sensitization and other animals to exhibit tolerance. The VTA units recorded from animals exhibiting behavioral sensitization responded significantly differently to MPD from animals that exhibited behavioral tolerance.
Collapse
Affiliation(s)
- Zachary Jones
- Department of Neurobiology and Anatomy, University of Texas Medical School at Houston, Houston, TX 77030,
| | - Nachum Dafny
- Neurobiology and Anatomy University of Texas Medical School at Houston suite 7.208B
| |
Collapse
|
34
|
Teixeira MZ. Rebound effect of modern drugs: serious adverse event unknown by health professionals. Rev Assoc Med Bras (1992) 2013; 59:629-38. [PMID: 24211013 DOI: 10.1016/j.ramb.2013.05.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2012] [Revised: 02/24/2013] [Accepted: 05/21/2013] [Indexed: 12/13/2022] Open
Abstract
OBJECTIVE Supported in the Hippocratic aphorism primum non nocere, the bioethical principle of non-maleficence pray that the medical act cause the least damage or injury to the health of the patient, leaving it to the doctor to assess the risks of a particular therapy through knowledge of possible adverse events of drugs. Among these, the rebound effect represents a common side effect to numerous classes of modern drugs, may cause serious and fatal disorders in patients. This review aims to clarify the health professionals on clinical and epidemiological aspects of rebound phenomenon. METHODS A qualitative, exploratory and bibliographic review was held in the PubMed database using the keywords 'rebound', 'withdrawal', 'paradoxical', 'acetylsalicylic acid', 'anti-inflammatory', 'bronchodilator', 'antidepressant', 'statin', 'proton pump inhibitor' and 'bisphosphonate'. RESULTS The rebound effect occurs after discontinuation of numerous classes of drugs that act contrary to the disease disorders, exacerbating them at levels above those prior to treatment. Regardless of the disease, the drug and duration of treatment, the phenomenon manifests itself in a small proportion of susceptible individuals. However, it may cause serious and fatal adverse events should be considered a public health problem in view of the enormous consumption of drugs by population. CONCLUSION Bringing together a growing and unquestionable body of evidence, the physician needs to have knowledge of the consequences of the rebound effect and how to minimize it, increasing safety in the management of modern drugs. On the other hand, this rebound can be used in a curative way, broadening the spectrum of the modern therapeutics.
Collapse
Affiliation(s)
- Marcus Zulian Teixeira
- Disciplina Fundamentos da Homeopatia, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brasil.
| |
Collapse
|
35
|
Cummins ED, Griffin SB, Burgess KC, Peterson DJ, Watson BD, Buendia MA, Stanwood GD, Brown RW. Methylphenidate place conditioning in adolescent rats: an analysis of sex differences and the dopamine transporter. Behav Brain Res 2013; 257:215-23. [PMID: 24084582 DOI: 10.1016/j.bbr.2013.09.036] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2013] [Revised: 09/02/2013] [Accepted: 09/22/2013] [Indexed: 12/01/2022]
Abstract
In two experiments, we analyzed the effects of methylphenidate (MPH) on conditioned place preference (CPP) in adolescent male and female rats, and the effects of MPH on the dopamine transporter (DAT). In Experiment 1, male and female rats were conditioned for 5 consecutive days from postnatal day (P)44 to P48 with saline, 1, or 5mg/kg MPH. On the post conditioning preference test, the group administered the 1mg/kg dose of MPH resulted in no significant preference compared to controls, whereas the 5mg/kg dose of MPH produced a robust significant preference for the paired context, but there were no sex differences. Analysis of the DAT revealed that animals conditioned with the 5mg/kg dose of MPH demonstrated a significant decrease of the dopamine transporter (DAT) in the nucleus accumbens and striatum compared to controls. In Experiment 2, animals were conditioned using an every second day paradigm from P33-41 to model a previous MPH treatment regimen that had revealed sex differences in behavioral sensitization. MPH produced an increased preference for the paired context on a post-conditioning preference test in Experiment 2, but as in Experiment 1, no sex differences were observed. These data show that a relatively high dose of MPH has rewarding associative effects in both adolescent male and female rats reliably across two different conditioning paradigms and ages in adolescence, but no sex difference. In addition, MPH results in a significant decrease of the DAT in drug reward brain areas which has implications toward plasticity of the brain's reward system.
Collapse
Affiliation(s)
- Elizabeth D Cummins
- Department of Psychology, East Tennessee State University, Johnson City, TN, USA
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Prenatal nicotine exposure mouse model showing hyperactivity, reduced cingulate cortex volume, reduced dopamine turnover, and responsiveness to oral methylphenidate treatment. J Neurosci 2012; 32:9410-8. [PMID: 22764249 DOI: 10.1523/jneurosci.1041-12.2012] [Citation(s) in RCA: 94] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Cigarette smoking, nicotine replacement therapy, and smokeless tobacco use during pregnancy are associated with cognitive disabilities later in life in children exposed prenatally to nicotine. The disabilities include attention deficit hyperactivity disorder (ADHD) and conduct disorder. However, the structural and neurochemical bases of these cognitive deficits remain unclear. Using a mouse model we show that prenatal nicotine exposure produces hyperactivity, selective decreases in cingulate cortical volume, and radial thickness, as well as decreased dopamine turnover in the frontal cortex. The hyperactivity occurs in both male and female offspring and peaks during the "active" or dark phase of the light/dark cycle. These features of the mouse model closely parallel the human ADHD phenotype, whether or not the ADHD is associated with prenatal nicotine exposure. A single oral, but not intraperitoneal, administration of a therapeutic equivalent dose (0.75 mg/kg) of methylphenidate decreases the hyperactivity and increases the dopamine turnover in the frontal cortex of the prenatally nicotine exposed mice, once again paralleling the therapeutic effects of this compound in ADHD subjects. Collectively, our data suggest that the prenatal nicotine exposure mouse model has striking parallels to the ADHD phenotype not only in behavioral, neuroanatomical, and neurochemical features, but also with respect to responsiveness of the behavioral phenotype to methylphenidate treatment. The behavioral, neurochemical, and anatomical biomarkers in the mouse model could be valuable for evaluating new therapies for ADHD and mechanistic investigations into its etiology.
Collapse
|
37
|
Pauls AM, O'Daly OG, Rubia K, Riedel WJ, Williams SCR, Mehta MA. Methylphenidate effects on prefrontal functioning during attentional-capture and response inhibition. Biol Psychiatry 2012; 72:142-9. [PMID: 22552046 DOI: 10.1016/j.biopsych.2012.03.028] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2011] [Revised: 03/01/2012] [Accepted: 03/14/2012] [Indexed: 10/28/2022]
Abstract
BACKGROUND Methylphenidate improves motor response inhibition, typically assessed with the stop-signal task. The exact underlying mechanism for this, however, remains unknown. In addition, recent studies highlight that stop signals can have a confounding attentional-capture effect because of their low frequency in the task. In the current study, we assessed the effects of methylphenidate on neural networks of inhibitory control and attentional-capture within the context of two inhibitory control tasks. METHODS The effects of methylphenidate (40 mg) were assessed using functional magnetic resonance imaging in 16 healthy volunteers in a within-subject, double-blind, placebo-controlled design. RESULTS Methylphenidate significantly reduced activation of different regions within the right inferior frontal gyrus/insula to infrequent stimuli associated with successful inhibition, failed inhibition, and attentional capture. These inferior frontal gyrus regions showed different interregional connections with inhibitory and attention networks. For failed inhibitions, methylphenidate increased activation within performance-monitoring regions, including the superior frontal, anterior cingulate, and parietal-occipital cortices, but only after controlling for attentional capture. CONCLUSIONS Our findings suggest that the improvement of response inhibition seen following methylphenidate administration is due to its influence on underlying attentional mechanisms linked to response control requirements.
Collapse
Affiliation(s)
- Astrid M Pauls
- Department of Neuroimaging, Institute of Psychiatry, King's College London, United Kingdom.
| | | | | | | | | | | |
Collapse
|
38
|
Gilbert JA, Gasteyger C, Raben A, Meier DH, Astrup A, Sjödin A. The effect of tesofensine on appetite sensations. Obesity (Silver Spring) 2012; 20:553-61. [PMID: 21720440 DOI: 10.1038/oby.2011.197] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Tesofensine (TE), an inhibitor of monoamine presynaptic reuptake, has produced twice the weight loss seen with currently marketed drugs. However, its long term effect on appetite in humans has not been studied. A multicentre phase II trial was divided into two parts (24 weeks each). Part 1 had a randomized, double-blind, placebo-controlled design and Part 2, an open-labeled, single-group, uncontrolled design. A drug-free period (12 ± 3 weeks) separated them. In Part 1, participants (n = 158) were assigned to 0.25, 0.5 or 1.0 mg TE, or placebo. Completers of Part 1 were invited to participate in Part 2 (n = 113), during which they all received 0.5 or 1.0 mg TE. Appetite sensations and a composite satiety score (CSS = satiety + fullness + (100 - hunger) + (100 - prospective food consumption) were assessed. In Part 1 TE induced a dose-dependent increase in CSS at week 12 that correlated with weight loss during the 24 weeks (r = 0.36, P < 0.0001). However, CSS diminished over time as weight loss progressed (e.g., for 1.0 mg; 52 ± 17 mm; 64 ± 13 mm; 55 ± 13 mm at baseline, week 12 and week 24, respectively). After drug withdrawal CSS returned to baseline values (50 ± 17 mm, in the whole sample.), despite the participants' reduced-weight state (-7.2 ± 6.7 kg, P < 0.0001). The reintroduction of TE in Part 2 increased CSS again (56 ± 17 mm at week 60), regardless of initial treatment/weight loss. We postulate that enhanced satiety is involved in early weight loss. Whether the attenuated effect on appetite seen after 24 weeks is due to a counteracting effect in the weight reduced state or whether the appetite suppressing effect of TE per se diminishes over time is, however, still unclear.
Collapse
Affiliation(s)
- Jo-Anne Gilbert
- Division of Kinesiology, Department of Preventive and Social Medicine, Faculty of Medicine, Laval University, Quebec City, Quebec, Canada
| | | | | | | | | | | |
Collapse
|
39
|
Nikolaus S, Larisch R, Vosberg H, Beu M, Wirrwar A, Antke C, Kley K, Silva MADS, Huston JP, Müller HW. Pharmacological challenge and synaptic response - assessing dopaminergic function in the rat striatum with small animal single-photon emission computed tomography (SPECT) and positron emission tomography (PET). Rev Neurosci 2011; 22:625-45. [PMID: 22103308 DOI: 10.1515/rns.2011.054] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Disturbances of dopaminergic neurotransmission may be caused by changes in concentrations of synaptic dopamine (DA) and/or availabilities of pre- and post-synaptic transporter and receptor binding sites. We present a series of experiments which focus on the regulatory mechanisms of the dopamin(DA)ergic synapse in the rat striatum. In these studies, DA transporter (DAT) and/or D(2) receptor binding were assessed with either small animal single-photon emission computed tomography (SPECT) or positron emission tomography (PET) after pharmacological challenge with haloperidol, L-DOPA and methylphenidate, and after nigrostriatal 6-hydroxydopamine lesion. Investigations of DAT binding were performed with [(123)I]N-ω-fluoropropyl-2β-carbomethoxy-3β-(4-iodophenyl)nortropane ([(123)I]FP-CIT). D(2) receptor bindingd was assessed with either [(123)I](S)-2-hydroxy-3-iodo-6-methoxy-N-[(1-ethyl-2-pyrrolidinyl)methyl]benzamide ([(123)I]IBZM) or [(18)F]1[3-(4'fluorobenzoyl)propyl]-4-(2-keto-3-methyl-1-benzimidazolinyl)piperidine ([(18)F]FMB). Findings demonstrate that in vivo investigations of transporter and/or receptor binding are feasible with small animal SPECT and PET. Therefore, tracers that are radiolabeled with isotopes of comparatively long half-lives such as (123)I may be employed. Our approach to quantify DAT and/or D(2) receptor binding at baseline and after pharmacological interventions inducing DAT blockade, D(2) receptor blockade, and increases or decreases of endogenous DA concentrations holds promise for the in vivo assessment of synaptic function. This pertains to animal models of diseases associated with pre- or postsynaptic DAergic deficiencies such as Parkinson's disease, Huntington's disease, attention-deficit/hyperactivity disorder, schizophrenia or drug abuse.
Collapse
Affiliation(s)
- Susanne Nikolaus
- Clinic of Nuclear Medicine, University Hospital Düsseldorf, Moorenstr. 5, D-40225 Düsseldorf, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Kidani Y, Ishimatsu M, Akasu T. Methylphenidate enhances inhibitory synaptic transmission by increasing the content of norepinephrine in the locus coeruleus of juvenile rats. Kurume Med J 2011; 57:29-38. [PMID: 21727763 DOI: 10.2739/kurumemedj.57.29] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
The present study examined the effect of methylphenidate (MPH), a psychostimulant, on nor-adrenergic transmission in the locus coeruleus (LC) of juvenile rats. Intracellular recordings showed that MPH (>3 µM) produced a hyperpolarizing response associated with a decrease in the rate of spontaneously firing action potentials. MPH (1 µM) enhanced the amplitude of the inhibitory postsynaptic potential (IPSP) mediated by norepinephrine (NE), but did not change the excitatory postsynaptic potential (EPSP) mediated by excitatory amino acids. Whole-cell patch-clamp recordings showed that MPH (0.3-30 µM) produced an outward current (I(MPH)) and enhanced the inhibitory postsynaptic current (IPSC) in neurons of the juvenile rat LC. MPH (30 µM) enhanced the NE-induced outward current (I(NE)). Bath-application of yohimbine (1 µM) produced an inward current and blocked the MPH-induced enhancement of the IPSC. Yohimbine (1 µM) depressed not only the I(NE) but also the I(MPH) in juvenile rat LC neurons. The current-voltage relationship of the I(MPH) showed inward rectification and reversed polarity at -91.1±4.3 mV (n=5). Ba(2+) (100 µM) blocked the I(MPH), indicating that the I(MPH) is mediated by Ba(2+)-sensitive inward rectifier K(+) current. These results suggest that MPH enhances inhibitory synaptic transmission by increasing the concentration of NE at noradrenergic synapses in juvenile rat LC neurons.
Collapse
Affiliation(s)
- Yuri Kidani
- Department of Physiology, Kurume University School of Medicine, Kurume, Japan
| | | | | |
Collapse
|
41
|
|
42
|
Binding of [123I]iodobenzamide to the rat D2 receptor after challenge with various doses of methylphenidate: an in vivo imaging study with dedicated small animal SPECT. Eur J Nucl Med Mol Imaging 2010; 38:694-701. [PMID: 21110190 DOI: 10.1007/s00259-010-1668-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2010] [Accepted: 11/01/2010] [Indexed: 02/05/2023]
Abstract
PURPOSE The effect of various doses of methylphenidate on the binding of [(123)I]iodobenzamide ([(123)I]IBZM) to the rat D(2) receptor was assessed using small animal SPECT. METHODS D(2) receptor binding was measured at baseline and after pretreatment with various doses of methylphenidate. For baseline and methylphenidate challenge, striatal equilibrium ratios (V(3)″) were computed as an estimation of the binding potential. RESULTS After methylphenidate, striatal V(3)″ was 1.61 ± 0.61 (mean ± SD; 0.3 mg/kg), 0.91 ± 0.44 (3 mg/kg), 1.01 ± 0.44 (10 mg/kg), 0.91 ± 0.34 (30 mg/kg) and 0.99 ± 0.51 (60 mg/kg). Baseline values amounted to 1.73 ± 0.48, 1.32 ± 0.35, 1.50 ± 0.27, 1.82 ± 0.55 and 1.66 ± 0.41, respectively. Differences between baseline and methylphenidate were significant for the doses 3, 10, 30 and 60 mg/kg, whereas no significant difference was obtained for 0.3 mg/kg methylphenidate. Between-group differences of percentage reduction of D(2) receptor binding were only significant for the groups pretreated with 0.3 and 30 mg/kg methylphenidate, respectively. CONCLUSION Methylphenidate between 0.3 and 60 mg/kg decreased D(2) receptor binding with a maximum reduction after 30 mg/kg. As no between-group differences were evident between the groups pretreated with 3, 10, 30 and 60 mg/kg, it may be inferred that doses ≥ 3 mg/kg were sufficient to induce maximum dopamine concentration in the synaptic cleft. Further investigations are needed in order to clarify whether the variation between subjects can be accounted for by different synaptic mechanisms at the presynaptic binding site.
Collapse
|
43
|
Focus on the positive: computational simulations implicate asymmetrical reward prediction error signals in childhood attention-deficit/hyperactivity disorder. Brain Res 2010; 1365:18-34. [PMID: 20875804 DOI: 10.1016/j.brainres.2010.09.065] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2010] [Revised: 08/30/2010] [Accepted: 09/17/2010] [Indexed: 02/02/2023]
Abstract
A number of hypotheses have suggested that the principal neurological dysfunction responsible for the behavioural symptoms associated with Attention-Deficit/Hyperactive Disorder (ADHD) is likely rooted in abnormal phasic signals coded by the firing rate of midbrain dopamine neurons. We present a formal investigation of the impact atypical phasic dopamine signals have on behaviour by applying a TD(λ) reinforcement learning model to simulations of operant conditioning tasks that have been argued to quantify the hyperactive, inattentive and impulsive behaviour associated with ADHD. The results presented here suggest that asymmetrically effective dopamine signals encoded by a punctate increase or decrease in dopamine levels provide the best account for the behaviour of children with ADHD as well as an animal model of ADHD, the spontaneously hypertensive rat (SHR). The biological sources of this asymmetry are considered, as are other computational models of ADHD.
Collapse
|
44
|
Abstract
Disordered dopamine neurotransmission is implicated in mediating impulsiveness across a range of behaviors and disorders including addiction, compulsive gambling, attention-deficit/hyperactivity disorder, and dopamine dysregulation syndrome. Whereas existing theories of dopamine function highlight mechanisms based on aberrant reward learning or behavioral disinhibition, they do not offer an adequate account of the pathological hypersensitivity to temporal delay that forms a crucial behavioral phenotype seen in these disorders. Here we provide evidence that a role for dopamine in controlling the relationship between the timing of future rewards and their subjective value can bridge this explanatory gap. Using an intertemporal choice task, we demonstrate that pharmacologically enhancing dopamine activity increases impulsivity by enhancing the diminutive influence of increasing delay on reward value (temporal discounting) and its corresponding neural representation in the striatum. This leads to a state of excessive discounting of temporally distant, relative to sooner, rewards. Thus our findings reveal a novel mechanism by which dopamine influences human decision-making that can account for behavioral aberrations associated with a hyperfunctioning dopamine system.
Collapse
|
45
|
Engert V, Pruessner JC. Dopaminergic and noradrenergic contributions to functionality in ADHD: the role of methylphenidate. Curr Neuropharmacol 2010; 6:322-8. [PMID: 19587853 PMCID: PMC2701285 DOI: 10.2174/157015908787386069] [Citation(s) in RCA: 91] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2008] [Revised: 07/18/2008] [Accepted: 08/04/2008] [Indexed: 12/23/2022] Open
Abstract
Attention Deficit Hyperactivity Disorder (ADHD) is a childhood psychiatric condition characterized by severe impulsiveness, inattention and overactivity. Methylphenidate (MPH), a psychostimulant affecting both the dopaminergic and the noradrenergic systems, is one of the most frequently prescribed treatments for ADHD. Despite the widespread use of MPH and its proven effectiveness, its precise neurochemical mechanisms of action are under debate. For the most part, MPH’s influence on subcortical dopamine neurotransmission is thought to play a crucial role in its behavioral and cognitive effects. In their hypothesis of biphasic MPH action, Seeman and Madras [42, 43] suggest that therapeutic doses of MPH elevate tonic dopamine while inhibiting phasic transmitter release in subcortical structures, leading to reduced postsynaptic receptor stimulation and psychomotor activation in response to salient stimuli. Volkow and colleagues [56] suggest that by amplifying a weak striatal dopamine signal, MPH increases the perception of a stimulus or task as salient. The enhanced interest for the task is thought to increase attention and improve performance. Recent animal studies have however shown that when administered at doses producing clinically relevant drug plasma levels and enhancing cognitive function, MPH preferentially activates dopamine and noradrenaline efflux within the prefrontal cortex relative to the subcortical structures [5]. Overall, we suggest that the delineated theories of MPH therapeutic action should not be discussed as exclusive. Studies are outlined that allow integrating the different findings and models.
Collapse
Affiliation(s)
- Veronika Engert
- Douglas Mental Health University Institute, Department of Psychiatry, McGill University, Montreal, Quebec, Canada H4H 1R3.
| | | |
Collapse
|
46
|
Turic D, Swanson J, Sonuga-Barke E. DRD4 and DAT1 in ADHD: Functional neurobiology to pharmacogenetics. PHARMACOGENOMICS & PERSONALIZED MEDICINE 2010; 3:61-78. [PMID: 23226043 PMCID: PMC3513209 DOI: 10.2147/pgpm.s6800] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 05/18/2010] [Indexed: 12/26/2022]
Abstract
Attention deficit/hyperactivity disorder (ADHD) is a common and potentially very impairing neuropsychiatric disorder of childhood. Statistical genetic studies of twins have shown ADHD to be highly heritable, with the combination of genes and gene by environment interactions accounting for around 80% of phenotypic variance. The initial molecular genetic studies where candidates were selected because of the efficacy of dopaminergic compounds in the treatment of ADHD were remarkably successful and provided strong evidence for the role of DRD4 and DAT1 variants in the pathogenesis of ADHD. However, the recent application of non-candidate gene strategies (eg, genome-wide association scans) has failed to identify additional genes with substantial genetic main effects, and the effects for DRD4 and DAT1 have not been replicated. This is the usual pattern observed for most other physical and mental disorders evaluated with current state-of-the-art methods. In this paper we discuss future strategies for genetic studies in ADHD, highlighting both the pitfalls and possible solutions relating to candidate gene studies, genome-wide studies, defining the phenotype, and statistical approaches.
Collapse
Affiliation(s)
- Darko Turic
- Institute for Disorders of Impulse and Attention, School of Psychology, University of Southampton, UK
| | | | | |
Collapse
|
47
|
Psychostimulants given in adolescence modulate their effects in adulthood using the open field and the wheel-running assays. Brain Res Bull 2010; 82:208-17. [PMID: 20362034 DOI: 10.1016/j.brainresbull.2010.03.007] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2010] [Revised: 03/23/2010] [Accepted: 03/24/2010] [Indexed: 02/06/2023]
|
48
|
Luman M, Tripp G, Scheres A. Identifying the neurobiology of altered reinforcement sensitivity in ADHD: a review and research agenda. Neurosci Biobehav Rev 2009; 34:744-54. [PMID: 19944715 DOI: 10.1016/j.neubiorev.2009.11.021] [Citation(s) in RCA: 220] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2009] [Revised: 11/20/2009] [Accepted: 11/21/2009] [Indexed: 10/20/2022]
Abstract
ADHD is associated with altered reinforcement sensitivity, despite a number of inconsistent findings. This review focuses on the overlap and differences between seven neurobiologically valid models and lists 15 predictions assessing reinforcement sensitivity in ADHD. When comparing the models it becomes clear that there are great differences in the level of explanation. For example, some models try to explain a single core deficit in terms lower-level reinforcement systems, such as the dopamine transfer to reward back in time. Other models explain multiple deficits, by describing higher-level systems, such as impaired bottom-up prefrontal activation. When reviewing the available experimental evidence in support of the predictions, most experimental studies have been focusing on behavioral changes in the face of reward and response cost over no-reward, and on delay discounting. There is currently a lack in studies that focus on explaining underlying cognitive or neural mechanisms of altered reinforcement sensitivity in ADHD. Additionally, there is a lack in studies that try to understand what subgroup of children with ADHD shows alterations in reinforcement sensitivity. The scarcity in studies testing the neurobiological predictions is explained partly by a lack in knowledge how to test some of these predictions in humans. Nevertheless, we believe that these predictions can serve as a useful guide to the systematic evaluation of altered reinforcement sensitivity in ADHD.
Collapse
Affiliation(s)
- Marjolein Luman
- Department of Clinical Neurpsychology, Vrije Universiteit Amsterdam, Van der Boechorststraat 1, 1081 BT Amsterdam, The Netherlands.
| | | | | |
Collapse
|
49
|
Easton N, Marshall FH, Marsden CA, Fone KCF. Mapping the central effects of methylphenidate in the rat using pharmacological MRI BOLD contrast. Neuropharmacology 2009; 57:653-64. [PMID: 19733553 DOI: 10.1016/j.neuropharm.2009.08.018] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2009] [Revised: 08/10/2009] [Accepted: 08/11/2009] [Indexed: 11/26/2022]
Abstract
Methylphenidate (Ritalin) is a selective dopamine reuptake inhibitor and an effective treatment for attention deficit hyperactivity disorder (ADHD) however the anatomical foci and neuronal circuits involved in these therapeutic benefits are unclear. This study determines the temporal pattern of brain regional activity change produced by systemic administration of a therapeutically relevant dose of methylphenidate in anaesthetised Sprague-Dawley rats using BOLD MRI and a 2.35T Bruker magnet. Following 60 min basal recording separate rats received saline (n = 9) or +/- methylphenidate hydrochloride (2 mg/kg, i.p., n = 9) and BOLD changes were recorded for 90 min using statistical parametric maps. Methylphenidate produced significant positive random BOLD effects in the nucleus accumbens, substantia nigra, entorhinal cortex and medial orbital cortex. Negative random BOLD effects were more widespread and intense, occurring in the motor and somatosensory cortices, caudate putamen, lateral globus pallidus and bed nucleus of the stria terminalis, without accompanying changes in blood pressure or respiratory rate. Methylphenidate-induced negative BOLD in the striatum, and other dopamine terminal areas, may reflect post-synaptic changes produced by blockade of the neuronal dopamine reuptake transporter. While increased positive BOLD in the medial orbital cortex may reflect altered dopamine and/or noradrenaline release indirectly altering striatal activity. The overall pattern of BOLD changes is comparable to that seen in previous studies using guanfacine, amphetamine and atomoxetine, and suggests that although these compounds operate through distinct pharmacological mechanisms the BOLD changes may represent a 'fingerprint pattern' predictive of therapeutic benefit in ADHD.
Collapse
Affiliation(s)
- Neil Easton
- School of Biomedical Sciences, Medical School, Institute of Neuroscience, University of Nottingham, Queen's Medical Centre, Nottingham NG7 2UH, UK
| | | | | | | |
Collapse
|
50
|
Vaidya CJ, Stollstorff M. Cognitive neuroscience of Attention Deficit Hyperactivity Disorder: current status and working hypotheses. ACTA ACUST UNITED AC 2009; 14:261-7. [PMID: 19072750 DOI: 10.1002/ddrr.40] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Cognitive neuroscience studies of Attention Deficit Hyperactivity Disorder (ADHD) suggest multiple loci of pathology with respect to both cognitive domains and neural circuitry. Cognitive deficits extend beyond executive functioning to include spatial, temporal, and lower-level "nonexecutive" functions. Atypical functional anatomy extends beyond frontostriatal circuits to include posterior cortices, limbic regions, and the cerebellum. Pathophysiology includes dopaminergic as well as noradrenergic neurotransmitter systems. We review the major insights gained from functional brain imaging studies in ADHD and discuss working hypotheses regarding their neurochemical underpinnings.
Collapse
Affiliation(s)
- Chandan J Vaidya
- Department of Psychology, Georgetown University, Washington, DC 20057, USA.
| | | |
Collapse
|