1
|
Hrabak M, Ahmed R, Soriano MG, Powell A, Thanos PK, Steiner H. Vilazodone, a Novel SSRI Antidepressant with 5-HT1A Partial Agonist Properties: Diminished Potentiation of Chronic Oral Methylphenidate-Induced Dynorphin Expression in the Striatum in Adolescent Male Rats. Mol Neurobiol 2024:10.1007/s12035-024-04569-8. [PMID: 39466575 DOI: 10.1007/s12035-024-04569-8] [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: 05/15/2024] [Accepted: 10/18/2024] [Indexed: 10/30/2024]
Abstract
Selective serotonin reuptake inhibitor (SSRI) antidepressants such as fluoxetine are used in combination with the medical psychostimulant methylphenidate (Ritalin) in a variety of treatments in children and adults. Unintended co-exposure to these psychotropic medications also occurs in patients on SSRIs who abuse methylphenidate as a "cognitive enhancer" or recreational drug. Preclinical research shows that SSRIs such as fluoxetine when given in conjunction with methylphenidate potentiate addiction-related gene regulation by methylphenidate in the striatum, consistent with the known facilitatory role for serotonin in psychostimulant-induced neuronal and behavioral changes. Moreover, fluoxetine combined with methylphenidate also facilitated subsequent acquisition of cocaine self-administration in adolescent rats, suggesting an increased addiction liability for methylphenidate. In the present study, we investigated the impact of a novel SSRI, vilazodone, on methylphenidate-induced gene regulation in adolescent male rats. In contrast to prototypical SSRIs such as fluoxetine, vilazodone also acts as a partial 5-HT1A serotonin receptor agonist and is thus proposed to temper serotonin input to the striatum. We compared the effects of chronic treatment (4 weeks) with vilazodone (10 mg/kg, twice daily) with those of fluoxetine (5 mg/kg, twice daily) on striatal dynorphin expression induced by oral methylphenidate treatment (30/60 mg/kg/day in drinking water, 8 h access daily). Our results demonstrate that, in contrast to fluoxetine, vilazodone had minimal or no potentiating effects on methylphenidate-induced dynorphin expression. This diminished impact on gene regulation was seen throughout the striatum, including the nucleus accumbens, where increased dynorphin expression has previously been associated with various aspects of addiction. Our findings suggest that vilazodone may serve as a better adjunct SSRI with reduced addiction-facilitating properties.
Collapse
Affiliation(s)
- Michael Hrabak
- Stanson Toshok Center for Brain Function and Repair, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | - Rania Ahmed
- Behavioral Neuropharmacology and Neuroimaging Laboratory, Clinical Research Institute on Addictions, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
| | - Michelle G Soriano
- Stanson Toshok Center for Brain Function and Repair, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | - Aidan Powell
- Behavioral Neuropharmacology and Neuroimaging Laboratory, Clinical Research Institute on Addictions, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
| | - Panayotis K Thanos
- Behavioral Neuropharmacology and Neuroimaging Laboratory, Clinical Research Institute on Addictions, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
| | - Heinz Steiner
- Stanson Toshok Center for Brain Function and Repair, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA.
- Discipline of Cellular and Molecular Pharmacology, The Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA.
| |
Collapse
|
2
|
Fesenko Z, Ptukha M, da Silva MM, de Carvalho RSM, Tsytsarev V, Gainetdinov RR, Faber J, Volnova AB. Electrophysiological and Behavioral Markers of Hyperdopaminergia in DAT-KO Rats. Biomedicines 2024; 12:2114. [PMID: 39335627 PMCID: PMC11428849 DOI: 10.3390/biomedicines12092114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Revised: 09/10/2024] [Accepted: 09/14/2024] [Indexed: 09/30/2024] Open
Abstract
Background/Objectives: Dopamine dysfunction (DA) is a hallmark of many neurological disorders. In this case, the mechanism of changes in dopamine transmission on behavior remains unclear. This study is a look into the intricate link between disrupted DA signaling, neuronal activity patterns, and behavioral abnormalities in a hyperdopaminergic animal model. Methods: To study the relationship between altered DA levels, neuronal activity, and behavioral deficits, local field potentials (LFPs) were recorded during four different behaviors in dopamine transporter knockout rats (DAT-KO). At the same time, local field potentials were recorded in the striatum and prefrontal cortex. Correlates of LFP and accompanying behavioral patterns in genetically modified (DAT-KO) and control animals were studied. Results: DAT-KO rats exhibited desynchronization between LFPs of the striatum and prefrontal cortex, particularly during exploratory behavior. A suppressive effect of high dopamine levels on the striatum was also observed. Wild-type rats showed greater variability in LFP patterns across certain behaviors, while DAT-KO rats showed more uniform patterns. Conclusions: The decisive role of the synchrony of STR and PFC neurons in the organization of motor acts has been revealed. The greater variability of control animals in certain forms of behavior probably suggests greater adaptability. More uniform patterns in DAT-KO rats, indicating a loss of striatal flexibility when adapting to specific motor tasks. It is likely that hyperdopaminergy in the DAT-KO rat reduces the efficiency of information processing due to less synchronized activity during active behavior.
Collapse
Affiliation(s)
- Zoia Fesenko
- Institute of Translational Biomedicine, Saint Petersburg State University, Saint Petersburg 199034, Russia
| | - Maria Ptukha
- Centre for Youth Mental Health, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Victoria 3010, Australia;
| | - Marcelo M. da Silva
- Department of Biophysics, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo 04039-032, Brazil
| | - Raquel S. Marques de Carvalho
- Department of Biophysics, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo 04039-032, Brazil
| | - Vassiliy Tsytsarev
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Raul R. Gainetdinov
- Institute of Translational Biomedicine, Saint Petersburg State University, Saint Petersburg 199034, Russia
- Saint Petersburg University Hospital, Saint Petersburg 190121, Russia
| | - Jean Faber
- Department of Neurology and Neurosurgery, Division of Neuroscience, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo 04023-900, Brazil
| | - Anna B. Volnova
- Institute of Translational Biomedicine, Saint Petersburg State University, Saint Petersburg 199034, Russia
- Biological Faculty, Saint Petersburg State University, Saint Petersburg 199034, Russia
| |
Collapse
|
3
|
Liu T, Yu Y, Mi L, Zhao Z, Liu M, Wang J, Wang X, Sha Z, Nie M, Jiang W, Wu C, Yuan J, Lv C, Zhao B, Lin K, Li Z, Luo Z, Liu X, Qian Y, Jiang R. Efficacy and safety of compound porcine cerebroside and ganglioside injection (CPCGI) versus piracetam on cognition and functional outcomes for adults with traumatic brain injury: A study protocol for randomized controlled trial. Heliyon 2024; 10:e37296. [PMID: 39319135 PMCID: PMC11419906 DOI: 10.1016/j.heliyon.2024.e37296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Revised: 08/28/2024] [Accepted: 08/30/2024] [Indexed: 09/26/2024] Open
Abstract
Background Traumatic brain injury (TBI) is a common neurosurgical disease in emergency rooms with poor prognosis, imposing severe burdens on patients and their families. Evidence indicates that piracetam and compound porcine cerebroside and ganglioside injection (CPCGI) can improve cognitive levels in TBI patients to enhance functional prognosis, but there is still a research gap regarding the efficacy of CPCGI. This study aims to determine the effectiveness and safety of CPCGI in improving cognitive and functional outcomes in TBI patients. Methods This study is a multicenter, randomized, parallel-group, double-blind trial aiming to recruit 900 adult patients with mild to moderate TBI. After providing informed consent, 600 patients will be randomly assigned to the CPCGI group (20 ml/d, for 14 days), and 300 patients will be randomized to the piracetam group as a control (20 ml/d, for 14 days), followed up for 3 months after treatment. The primary outcome is the change in the Montreal Cognitive Assessment (MoCA) score from baseline after 3 months. The main secondary outcome measures include Mini-Mental State Examination (MMSE) scores, Glasgow Outcome Scale-Extended (GOS-E), and the Barthel Index at 1 and 3 months. Discussion This multi-center clinical trial aims to provide high-quality evidence on the efficacy and safety of CPCGI in improving cognitive and functional outcomes in mild to moderate TBI patients. Trial registration ChiCTR2000040466, date of registration: November 28, 2020.
Collapse
Affiliation(s)
- Tao Liu
- Department of Neurosurgery, Tianjin Neurological Institute, State Key Laboratory of Experimental Hematology, Key Laboratory of Post-Neuroinjury Neurorepair and Regeneration in Central Nervous System Tianjin & Ministry of Education, Tianjin Medical University General Hospital, Tianjin, China
- The George Institute for Global Health, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
| | - Yunhu Yu
- Department of Neurosurgery, Tianjin Neurological Institute, State Key Laboratory of Experimental Hematology, Key Laboratory of Post-Neuroinjury Neurorepair and Regeneration in Central Nervous System Tianjin & Ministry of Education, Tianjin Medical University General Hospital, Tianjin, China
- Department of Clinical Research Center for Neurological Disease, the People's Hospital of HongHuaGang District of ZunYi, Guizhou, China
| | - Liang Mi
- Department of Neurosurgery, Tianjin Neurological Institute, State Key Laboratory of Experimental Hematology, Key Laboratory of Post-Neuroinjury Neurorepair and Regeneration in Central Nervous System Tianjin & Ministry of Education, Tianjin Medical University General Hospital, Tianjin, China
| | - Zhihao Zhao
- Department of Neurosurgery, Tianjin Neurological Institute, State Key Laboratory of Experimental Hematology, Key Laboratory of Post-Neuroinjury Neurorepair and Regeneration in Central Nervous System Tianjin & Ministry of Education, Tianjin Medical University General Hospital, Tianjin, China
| | - Mingqi Liu
- Department of Rehabilitation Medicine, Zhejiang Provincial People's Hospital, Hangzhou, China
| | - Jiao Wang
- Department of Epidemiology, College of Preventive Medicine, Third Military Medical University, Chongqing, China
| | - Xin Wang
- Department of Neurology, The First Affiliated Hospital of China Medical University, Liaoning, China
| | - Zhuang Sha
- Department of Neurosurgery, Tianjin Neurological Institute, State Key Laboratory of Experimental Hematology, Key Laboratory of Post-Neuroinjury Neurorepair and Regeneration in Central Nervous System Tianjin & Ministry of Education, Tianjin Medical University General Hospital, Tianjin, China
| | - Meng Nie
- Department of Neurosurgery, Tianjin Neurological Institute, State Key Laboratory of Experimental Hematology, Key Laboratory of Post-Neuroinjury Neurorepair and Regeneration in Central Nervous System Tianjin & Ministry of Education, Tianjin Medical University General Hospital, Tianjin, China
| | - Weiwei Jiang
- Department of Neurosurgery, Tianjin Neurological Institute, State Key Laboratory of Experimental Hematology, Key Laboratory of Post-Neuroinjury Neurorepair and Regeneration in Central Nervous System Tianjin & Ministry of Education, Tianjin Medical University General Hospital, Tianjin, China
| | - Chenrui Wu
- Department of Neurosurgery, Tianjin Neurological Institute, State Key Laboratory of Experimental Hematology, Key Laboratory of Post-Neuroinjury Neurorepair and Regeneration in Central Nervous System Tianjin & Ministry of Education, Tianjin Medical University General Hospital, Tianjin, China
| | - Jiangyuan Yuan
- Department of Neurosurgery, Tianjin Neurological Institute, State Key Laboratory of Experimental Hematology, Key Laboratory of Post-Neuroinjury Neurorepair and Regeneration in Central Nervous System Tianjin & Ministry of Education, Tianjin Medical University General Hospital, Tianjin, China
| | - Chuanxiang Lv
- Department of Neurosurgery, The First Hospital of Jilin University, Changchun, China
| | - Biao Zhao
- Department of Neurosurgery, the Second Affiliated Hospital of Bengbu Medical University, Bengbu, China
| | - Kun Lin
- Department of Neurosurgery, Fujian Provincial Hospital, Fujian, China
| | - Zhanying Li
- Department of Neurosurgery, Kailuan General Hospital, Hebei, China
| | - Zhenyu Luo
- Department of Neurosurgery, Shandong Provincial Third Hospital, Jinan, China
| | - Xuanhui Liu
- Department of Neurosurgery, Tianjin Neurological Institute, State Key Laboratory of Experimental Hematology, Key Laboratory of Post-Neuroinjury Neurorepair and Regeneration in Central Nervous System Tianjin & Ministry of Education, Tianjin Medical University General Hospital, Tianjin, China
| | - Yu Qian
- Department of Neurosurgery, Tianjin Neurological Institute, State Key Laboratory of Experimental Hematology, Key Laboratory of Post-Neuroinjury Neurorepair and Regeneration in Central Nervous System Tianjin & Ministry of Education, Tianjin Medical University General Hospital, Tianjin, China
| | - Rongcai Jiang
- Department of Neurosurgery, Tianjin Neurological Institute, State Key Laboratory of Experimental Hematology, Key Laboratory of Post-Neuroinjury Neurorepair and Regeneration in Central Nervous System Tianjin & Ministry of Education, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
4
|
Kirschner H, Molla HM, Nassar MR, de Wit H, Ullsperger M. Methamphetamine-induced adaptation of learning rate dynamics depend on baseline performance. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.04.602054. [PMID: 39026741 PMCID: PMC11257491 DOI: 10.1101/2024.07.04.602054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
The ability to calibrate learning according to new information is a fundamental component of an organism's ability to adapt to changing conditions. Yet, the exact neural mechanisms guiding dynamic learning rate adjustments remain unclear. Catecholamines appear to play a critical role in adjusting the degree to which we use new information over time, but individuals vary widely in the manner in which they adjust to changes. Here, we studied the effects of a low dose of methamphetamine (MA), and individual differences in these effects, on probabilistic reversal learning dynamics in a within-subject, double-blind, randomized design. Participants first completed a reversal learning task during a drug-free baseline session to provide a measure of baseline performance. Then they completed the task during two sessions, one with MA (20 mg oral) and one with placebo (PL). First, we showed that, relative to PL, MA modulates the ability to dynamically adjust learning from prediction errors. Second, this effect was more pronounced in participants who performed poorly at baseline. These results present novel evidence for the involvement of catecholaminergic transmission on learning flexibility and highlights that baseline performance modulates the effect of the drug.
Collapse
Affiliation(s)
- Hans Kirschner
- Institute of Psychology, Otto-von-Guericke University, D-39106 Magdeburg, Germany
| | - Hanna M Molla
- Department of Psychiatry and Behavioral Neuroscience, University of Chicago, Chicago, Illinois, USA
| | - Matthew R Nassar
- Robert J. and Nancy D. Carney Institute for Brain Science, Brown University, Providence RI 02912-1821, USA
- Department of Neuroscience, Brown University, Providence RI 02912-1821, USA
| | - Harriet de Wit
- Department of Psychiatry and Behavioral Neuroscience, University of Chicago, Chicago, Illinois, USA
| | - Markus Ullsperger
- Institute of Psychology, Otto-von-Guericke University, D-39106 Magdeburg, Germany
- Center for Behavioral Brain Sciences, D-39106 Magdeburg, Germany
- German Center for Mental Health (DZPG), Center for Intervention and Research on Adaptive and Maladaptive Brain Circuits Underlying Mental Health (C-I-R-C), Halle-Jena-Magdeburg, Germany
| |
Collapse
|
5
|
Mayer FP, Stewart A, Varman DR, Moritz AE, Foster JD, Owens AW, Areal LB, Gowrishankar R, Velez M, Wickham K, Phelps H, Katamish R, Rabil M, Jayanthi LD, Vaughan RA, Daws LC, Blakely RD, Ramamoorthy S. Kappa Opioid Receptor Antagonism Restores Phosphorylation, Trafficking and Behavior induced by a Disease Associated Dopamine Transporter Variant. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.05.03.539310. [PMID: 37205452 PMCID: PMC10187322 DOI: 10.1101/2023.05.03.539310] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Aberrant dopamine (DA) signaling is implicated in schizophrenia, bipolar disorder (BPD), autism spectrum disorder (ASD), substance use disorder, and attention-deficit/hyperactivity disorder (ADHD). Treatment of these disorders remains inadequate, as exemplified by the therapeutic use of d-amphetamine and methylphenidate for the treatment of ADHD, agents with high abuse liability. In search for an improved and non-addictive therapeutic approach for the treatment of DA-linked disorders, we utilized a preclinical mouse model expressing the human DA transporter (DAT) coding variant DAT Val559, previously identified in individuals with ADHD, ASD, or BPD. DAT Val559, like several other disease-associated variants of DAT, exhibits anomalous DA efflux (ADE) that can be blocked by d-amphetamine and methylphenidate. Kappa opioid receptors (KORs) are expressed by DA neurons and modulate DA release and clearance, suggesting that targeting KORs might also provide an alternative approach to normalizing DA-signaling disrupted by perturbed DAT function. Here we demonstrate that KOR stimulation leads to enhanced surface trafficking and phosphorylation of Thr53 in wildtype DAT, effects achieved constitutively by the Val559 mutant. Moreover, these effects can be rescued by KOR antagonism of DAT Val559 in ex vivo preparations. Importantly, KOR antagonism also corrected in vivo DA release as well as sex-dependent behavioral abnormalities observed in DAT Val559 mice. Given their low abuse liability, our studies with a construct valid model of human DA associated disorders reinforce considerations of KOR antagonism as a pharmacological strategy to treat DA associated brain disorders.
Collapse
Affiliation(s)
- Felix P. Mayer
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, Jupiter, FL, USA
- Stiles-Nicholson Brain Institute, Florida Atlantic University, Jupiter, FL, USA
| | - Adele Stewart
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, Jupiter, FL, USA
- Stiles-Nicholson Brain Institute, Florida Atlantic University, Jupiter, FL, USA
| | - Durairaj Ragu Varman
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, USA
| | - Amy E. Moritz
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, USA
| | - James D. Foster
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, USA
| | - Anthony W. Owens
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio, TX, USA
| | - Lorena B. Areal
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, Jupiter, FL, USA
| | - Raajaram Gowrishankar
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, Jupiter, FL, USA
| | - Michelle Velez
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, Jupiter, FL, USA
| | - Kyria Wickham
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, Jupiter, FL, USA
| | - Hannah Phelps
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, Jupiter, FL, USA
| | - Rania Katamish
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, Jupiter, FL, USA
| | - Maximilian Rabil
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, Jupiter, FL, USA
| | - Lankupalle D. Jayanthi
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, USA
| | - Roxanne A. Vaughan
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, USA
| | - Lynette C. Daws
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio, TX, USA
- Department of Pharmacology, University of Texas Health Science Center, San Antonio, TX, USA
| | - Randy D. Blakely
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, Jupiter, FL, USA
- Stiles-Nicholson Brain Institute, Florida Atlantic University, Jupiter, FL, USA
| | - Sammanda Ramamoorthy
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, USA
| |
Collapse
|
6
|
Carucci S, Zuddas A, Lampis A, Man KKC, Balia C, Buitelaar J, Danckaerts M, Dittmann RW, Donno F, Falissard B, Gagliano A, Garas P, Häge A, Hollis C, Inglis SK, Konrad K, Kovshoff H, Liddle E, McCarthy S, Neubert A, Nagy P, Rosenthal E, Sonuga-Barke EJS, Wong ICK, Banaschewski T, Coghill D. The Impact of Methylphenidate on Pubertal Maturation and Bone Age in ADHD Children and Adolescents: Results from the ADHD Drugs Use Chronic Effects (ADDUCE) Project. J Atten Disord 2024; 28:722-739. [PMID: 38366816 DOI: 10.1177/10870547241226726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/18/2024]
Abstract
OBJECTIVE The short-term safety of methylphenidate (MPH) has been widely demonstrated; however the long-term safety is less clear. The aim of this study was to investigate the safety of MPH in relation to pubertal maturation and to explore the monitoring of bone age. METHOD Participants from ADDUCE, a two-year observational longitudinal study with three parallel cohorts (MPH group, no-MPH group, and a non-ADHD control group), were compared with respect to Tanner staging. An Italian subsample of medicated-ADHD was further assessed by the monitoring of bone age. RESULTS The medicated and unmedicated ADHD groups did not differ in Tanner stages indicating no higher risk of sexual maturational delay in the MPH-treated patients. The medicated subsample monitored for bone age showed a slight acceleration of the bone maturation after 24 months, however their predicted adult height remained stable. CONCLUSION Our results do not suggest safety concerns on long-term treatment with MPH in relation to pubertal maturation and growth.
Collapse
Affiliation(s)
- Sara Carucci
- University of Cagliari, Italy
- ASL Cagliari, Italy
| | | | | | - Kenneth K C Man
- Donders Institute for Brain, Cognition and Behavior, Nijmegen, The Netherlands
- Hong Kong Science Park, China
- University College London Hospitals NHS Foundation Trust, UK
- The University of Hong Kong, China
| | - Carla Balia
- University of Cagliari, Italy
- ASL Cagliari, Italy
| | - Jan Buitelaar
- Donders Institute for Brain, Cognition and Behavior, Nijmegen, The Netherlands
- Karakter Child and Adolescent Psychiatry University Centre, Nijmegen, The Netherlands
| | | | | | | | | | | | | | | | | | | | - Kerstin Konrad
- University Hospital RWTH Aachen, Germany
- RWTH Aachen and Research Centre Jülich, Germany
| | | | | | | | | | - Peter Nagy
- Bethesda Children's Hospital, Budapest, Hungary
| | | | | | - Ian C K Wong
- University College London, UK
- Hong Kong Science Park, China
- University College London Hospitals NHS Foundation Trust, UK
- The University of Hong Kong, China
- Aston University, Birmingham, UK
| | | | - David Coghill
- University of Dundee, UK
- University of Melbourne, VIC, Australia
| |
Collapse
|
7
|
Peattie ARD, Manktelow AE, Sahakian BJ, Menon DK, Stamatakis EA. Methylphenidate Ameliorates Behavioural and Neurobiological Deficits in Executive Function for Patients with Chronic Traumatic Brain Injury. J Clin Med 2024; 13:771. [PMID: 38337465 PMCID: PMC10856064 DOI: 10.3390/jcm13030771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 01/18/2024] [Accepted: 01/23/2024] [Indexed: 02/12/2024] Open
Abstract
(1) Background: Traumatic brain injury (TBI) often results in cognitive impairments, including in visuospatial planning and executive function. Methylphenidate (MPh) demonstrates potential improvements in several cognitive domains in patients with TBI. The Tower of London (TOL) is a visuospatial planning task used to assess executive function. (2) Methods: Volunteers with a history of TBI (n = 16) participated in a randomised, double-blinded, placebo-controlled, fMRI study to investigate the neurobiological correlates of visuospatial planning and executive function, on and off MPh. (3) Results: Healthy controls (HCs) (n = 18) and patients on placebo (TBI-placebo) differed significantly in reaction time (p < 0.0005) and accuracy (p < 0.0001) when considering all task loads, but especially for high cognitive loads for reaction time (p < 0.001) and accuracy (p < 0.005). Across all task loads, TBI-MPh were more accurate than TBI-placebo (p < 0.05) but remained less accurate than HCs (p < 0.005). TBI-placebo substantially improved in accuracy with MPh administration (TBI-MPh) to a level statistically comparable to HCs at low (p = 0.443) and high (p = 0.175) cognitive loads. Further, individual patients that performed slower on placebo at low cognitive loads were faster with MPh (p < 0.05), while individual patients that performed less accurately on placebo were more accurate with MPh at both high and low cognitive loads (p < 0.005). TBI-placebo showed reduced activity in the bilateral inferior frontal gyri (IFG) and insulae versus HCs. MPh normalised these regional differences. MPh enhanced within-network connectivity (between parietal, striatal, insula, and cerebellar regions) and enhanced beyond-network connectivity (between parietal, thalamic, and cerebellar regions). Finally, individual changes in cerebellar-thalamic (p < 0.005) and cerebellar-parietal (p < 0.05) connectivity with MPh related to individual changes in accuracy with MPh. (4) Conclusions: This work highlights behavioural and neurofunctional differences between HCs and patients with chronic TBI, and that adverse differences may benefit from MPh treatment.
Collapse
Affiliation(s)
- Alexander R. D. Peattie
- Division of Anaesthesia, University of Cambridge, Addenbrooke’s Hospital, Box 93, Hills Road, Cambridge CB2 0QQ, UK; (A.E.M.); (D.K.M.)
- Department of Clinical Neurosciences, University of Cambridge, Addenbrooke’s Hospital, Box 165, Hills Road, Cambridge CB2 0QQ, UK
| | - Anne E. Manktelow
- Division of Anaesthesia, University of Cambridge, Addenbrooke’s Hospital, Box 93, Hills Road, Cambridge CB2 0QQ, UK; (A.E.M.); (D.K.M.)
- Department of Clinical Neurosciences, University of Cambridge, Addenbrooke’s Hospital, Box 165, Hills Road, Cambridge CB2 0QQ, UK
| | - Barbara J. Sahakian
- Department of Psychiatry, University of Cambridge, Herchel Smith Building for Brain and Mind Sciences, Forvie Site, Robinson Way, Cambridge CB2 0SZ, UK;
| | - David K. Menon
- Division of Anaesthesia, University of Cambridge, Addenbrooke’s Hospital, Box 93, Hills Road, Cambridge CB2 0QQ, UK; (A.E.M.); (D.K.M.)
- Wolfson Brain Imaging Centre, University of Cambridge, Cambridge Biomedical Campus, Box 65, Cambridge CB2 0QQ, UK
| | - Emmanuel A. Stamatakis
- Division of Anaesthesia, University of Cambridge, Addenbrooke’s Hospital, Box 93, Hills Road, Cambridge CB2 0QQ, UK; (A.E.M.); (D.K.M.)
- Department of Clinical Neurosciences, University of Cambridge, Addenbrooke’s Hospital, Box 165, Hills Road, Cambridge CB2 0QQ, UK
| |
Collapse
|
8
|
Tomasi D, Manza P, Yan W, Shokri-Kojori E, Demiral ŞB, Yonga MV, McPherson K, Biesecker C, Dennis E, Johnson A, Zhang R, Wang GJ, Volkow ND. Examining the role of dopamine in methylphenidate's effects on resting brain function. Proc Natl Acad Sci U S A 2023; 120:e2314596120. [PMID: 38109535 PMCID: PMC10756194 DOI: 10.1073/pnas.2314596120] [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: 08/23/2023] [Accepted: 11/14/2023] [Indexed: 12/20/2023] Open
Abstract
The amplitude of low-frequency fluctuations (ALFF) and global functional connectivity density (gFCD) are fMRI (Functional MRI) metrics widely used to assess resting brain function. However, their differential sensitivity to stimulant-induced dopamine (DA) increases, including the rate of DA rise and the relationship between them, have not been investigated. Here we used, simultaneous PET-fMRI to examine the association between dynamic changes in striatal DA and brain activity as assessed by ALFF and gFCD, following placebo, intravenous (IV), or oral methylphenidate (MP) administration, using a within-subject double-blind placebo-controlled design. In putamen, MP significantly reduced D2/3 receptor availability and strongly reduced ALFF and increased gFCD in the brain for IV-MP (Cohen's d > 1.6) but less so for oral-MP (Cohen's d < 0.6). Enhanced gFCD was associated with both the level and the rate of striatal DA increases, whereas decreased ALFF was only associated with the level of DA increases. These findings suggest distinct representations of neurovascular activation with ALFF and gFCD by stimulant-induced DA increases with differential sensitivity to the rate and the level of DA increases. We also observed an inverse association between gFCD and ALFF that was markedly enhanced during IV-MP, which could reflect an increased contribution from MP's vasoactive properties.
Collapse
Affiliation(s)
- Dardo Tomasi
- Laboratory of Neuroimaging (LNI), National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, MD20892
| | - Peter Manza
- Laboratory of Neuroimaging (LNI), National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, MD20892
| | - Weizheng Yan
- Laboratory of Neuroimaging (LNI), National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, MD20892
| | - Ehsan Shokri-Kojori
- Laboratory of Neuroimaging (LNI), National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, MD20892
| | - Şükrü Barış Demiral
- Laboratory of Neuroimaging (LNI), National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, MD20892
| | - Michele-Vera Yonga
- Laboratory of Neuroimaging (LNI), National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, MD20892
| | - Katherine McPherson
- Laboratory of Neuroimaging (LNI), National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, MD20892
| | - Catherine Biesecker
- Laboratory of Neuroimaging (LNI), National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, MD20892
| | - Evan Dennis
- Laboratory of Neuroimaging (LNI), National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, MD20892
| | - Allison Johnson
- Laboratory of Neuroimaging (LNI), National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, MD20892
| | - Rui Zhang
- Laboratory of Neuroimaging (LNI), National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, MD20892
| | - Gene-Jack Wang
- Laboratory of Neuroimaging (LNI), National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, MD20892
| | - Nora D. Volkow
- Laboratory of Neuroimaging (LNI), National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, MD20892
| |
Collapse
|
9
|
Senior D, McCarthy M, Ahmed R, Klein S, Lee WX, Hadjiargyrou M, Komatsu D, Steiner H, Thanos PK. Chronic oral methylphenidate plus fluoxetine treatment in adolescent rats increases cocaine self-administration. ADDICTION NEUROSCIENCE 2023; 8:100127. [PMID: 38274857 PMCID: PMC10809890 DOI: 10.1016/j.addicn.2023.100127] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/27/2024]
Abstract
Background Depression and attention deficit hyperactivity disorder are known to be comorbid. Treatment of these commonly coexisting diseases typically involves the combined prescription of methylphenidate (MP), a psychostimulant, and fluoxetine (FLX), a selective serotonin reuptake inhibitor (SSRI). MP and cocaine have similar mechanisms of action and this study examined the effects of chronic treatment of MP combined with FLX on cocaine consumption in rats. Methods Four groups of rats received access to drinking solutions of water (control), MP (30/60 mg/kg/day), FLX (20 mg/kg/day), or the combination of MP (30/60 mg/kg/day) plus FLX (20 mg/kg/day), during 8 h per day for one month. Following these drug treatments, rats were allowed to self-administer cocaine for 14 days. Results Our results showed that, during the first week of cocaine self-administration, the MP-treated rats had significantly greater numbers of active lever presses (plus 127%) and increased consumption of cocaine compared to the control rats. In contrast, during week two of cocaine self-administration, the rats treated with the MP + FLX combination showed significantly more lever presses (plus 198%) and significantly greater cocaine consumption (plus 84%) compared to the water controls. Conclusion Chronic oral treatment during adolescence with the combination of MP plus FLX resulted in increased cocaine use after 2 weeks of cocaine self-administration in rats. These novel findings suggest that the combined exposure to these two drugs chronically, during adolescence, may produce increased vulnerability towards cocaine abuse during young adulthood.
Collapse
Affiliation(s)
- Daniela Senior
- Behavioral Neuropharmacology and Neuroimaging Laboratory (BNNL), Clinical Research Institute on Addictions, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14051, USA
| | - Madison McCarthy
- Behavioral Neuropharmacology and Neuroimaging Laboratory (BNNL), Clinical Research Institute on Addictions, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14051, USA
| | - Rania Ahmed
- Behavioral Neuropharmacology and Neuroimaging Laboratory (BNNL), Clinical Research Institute on Addictions, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14051, USA
- Department of Psychology, University at Buffalo, Buffalo, NY, 14203, USA
| | - Shannon Klein
- Behavioral Neuropharmacology and Neuroimaging Laboratory (BNNL), Clinical Research Institute on Addictions, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14051, USA
| | - Wen Xuan Lee
- Behavioral Neuropharmacology and Neuroimaging Laboratory (BNNL), Clinical Research Institute on Addictions, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14051, USA
| | - Michael Hadjiargyrou
- Department of Biological and Chemical Sciences, New York Institute of Technology, Old Westbury, NY, USA
| | - David Komatsu
- Department of Orthopedics, Stony Brook University, Stony Brook, NY, USA
| | - Heinz Steiner
- Stanson Toshok Center for Brain Function and Repair, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064, USA
- Discipline of Cellular and Molecular Pharmacology, The Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064, USA
| | - Panayotis K. Thanos
- Behavioral Neuropharmacology and Neuroimaging Laboratory (BNNL), Clinical Research Institute on Addictions, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14051, USA
- Department of Psychology, University at Buffalo, Buffalo, NY, 14203, USA
| |
Collapse
|
10
|
Zammit N, Muscat R. Alpha/beta-gamma decoupling in methylphenidate medicated ADHD patients. Front Neurosci 2023; 17:1267901. [PMID: 37841679 PMCID: PMC10570420 DOI: 10.3389/fnins.2023.1267901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 09/18/2023] [Indexed: 10/17/2023] Open
Abstract
There is much interest to understand how different neural rhythms function, interact and are regulated. Here, we focus on WM delay gamma to investigate its coupling with alpha/beta rhythms and its neuromodulation by methylphenidate. We address this through the use of human EEG conducted in healthy and ADHD subjects which revealed ADHD-specific electrophysiological deficits and MPH-induced normalization of gamma amplitude and its coupling with alpha/beta rhythms. Decreased alpha/beta-gamma coupling is known to facilitate memory representations via disinhibition of gamma ensembles coding the maintained stimuli. Here, we present EEG evidence which suggests that these dynamics are sensitive to catecholaminergic neuromodulation. MPH decreased alpha/beta-gamma coupling and this was related to the increase in delay-relevant gamma activity evoked by the same drug. These results add further to the neuromodulatory findings that reflect an electrophysiological dimension to the well-known link between WM delay and catecholaminergic transmission.
Collapse
Affiliation(s)
- Nowell Zammit
- Centre for Molecular Medicine and Biobanking, University of Malta, Msida, Malta
| | - Richard Muscat
- Centre for Molecular Medicine and Biobanking, University of Malta, Msida, Malta
- Department of Physiology and Biochemistry, University of Malta, Msida, Malta
| |
Collapse
|
11
|
Grunze H. The role of the D3 dopamine receptor and its partial agonist cariprazine in patients with schizophrenia and substance use disorder. Expert Opin Pharmacother 2023; 24:1985-1992. [PMID: 37817489 DOI: 10.1080/14656566.2023.2266359] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 09/29/2023] [Indexed: 10/12/2023]
Abstract
INTRODUCTION Comorbidity of substance use disorder (SUD) with schizophrenia, referred to as dual disorder (DD), significantly increases morbidity and mortality compared to schizophrenia alone. A dopaminergic dysregulation seems to be a common pathophysiological basis of the comorbidity. AREAS COVERED This article reports the current evidence on the role of dopamine dysregulations in DD, the pharmacological profile of cariprazine, a partial agonist of D3 and D2 dopamine receptors, and first clinical observations that may support its usefulness in the therapy of DD. PubMed/MEDLINE was searched for the keywords 'cariprazine,' 'schizophrenia,' 'dual disorder,' 'dopamine,' and 'dopamine receptor.' Preclinical and clinical studies, and reviews published in English were retrieved. EXPERT OPINION Although the management of DD remains challenging, and the evidence for pharmacologic treatments is still unsatisfactory, cariprazine may be a candidate medication in DD due to its unique mechanism of action. Preliminary clinical experiences suggest that cariprazine has both antipsychotic and anticraving properties and should be considered early in patients with DD.
Collapse
Affiliation(s)
- Heinz Grunze
- Psychiatrie Schwäbisch Hall, Schwäbisch Hall, Germany
- Department of Psychiatry, Paracelsus Medical University Nuremberg, Nuremberg, Germany
| |
Collapse
|
12
|
Panfil K, Small R, Kirkpatrick K. Effects of methylphenidate on impulsive choice and delay aversion in Lewis rats. Behav Pharmacol 2023; 34:169-Btii. [PMID: 36752349 PMCID: PMC10006322 DOI: 10.1097/fbp.0000000000000719] [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] [Indexed: 02/09/2023]
Abstract
Attention-deficit/hyperactivity disorder (ADHD), a common behavioral disorder in children and young adults, is characterized by symptoms of impulsivity, inattention, and hyperactivity. The purpose of this study was to evaluate the Lewis rat strain as a model of ADHD by testing their impulsive choices. Lewis rats were compared to their source strain, the Wistar rat, on an impulsive choice task. Rats completed the tasks on and off methylphenidate, a commonly prescribed medication for ADHD. Off methylphenidate, Lewis rats made more impulsive choices than Wistar rats. Analyses of acquisition of choice behavior suggested that both strains were able to discriminate reward sizes, but Lewis rats still chose the smaller-sooner option more than the larger-later (LL) option when the delays to reward were the same. This may be due to an aversion to the LL lever, which was associated with the longest delays to reward. Higher doses of methylphenidate increased LL choices in Lewis rats but decreased LL choices in Wistar rats. Altogether, these results suggest Lewis rats may be a viable model for ADHD in individuals whose symptoms are characterized by impulsive choices.
Collapse
Affiliation(s)
- Kelsey Panfil
- Department of Psychological Sciences, Kansas State University, Manhattan, Kansas, USA
| | | | | |
Collapse
|
13
|
Soufsaf S, Robaey P, Nekka F. An exploratory analysis of the performance of methylphenidate regimens based on a PKPD model of dopamine and norepinephrine transporter occupancy. J Pharmacokinet Pharmacodyn 2023:10.1007/s10928-023-09854-y. [PMID: 36930337 DOI: 10.1007/s10928-023-09854-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 02/23/2023] [Indexed: 03/18/2023]
Abstract
Methylphenidate (MPH) is a psychostimulant which inhibits the uptake of dopamine and norepinephrine transporters, DAT and NET, and is mostly used to treat Attention Deficit/Hyperactivity Disorder. The current dose optimization is done through titration, a cumbersome approach for patients. To assess the therapeutic performance of MPH regimens, we introduce an in silico framework composed of (i) a population pharmacokinetic model of MPH, (ii) a pharmacodynamic (PD) model of DAT and NET occupancy, (iii) a therapeutic box delimited by time and DAT occupancy, and (iv) a performance score computation. DAT occupancy data was digitized (n = 152) and described with Emax models. NET occupancy was described with a KPD model. We used this integrative framework to simulate the performance of extended-release (18-99 mg) and tid MPH regimens (25-40 mg). Early blood samples of MPH seem to lead to higher DAT occupancy, consistent with an acute tolerance observed in clinical rating scales. An Emax model with a time-dependent tolerance was fitted to available data to assess the observed clockwise hysteresis. Peak performance is observed at 63 mg. While our analysis does not deny the existence of an acute tolerance, data precision in terms of formulation and sampling times does not allow a definite confirmation of this phenomenon. This work justifies the need for a more systematic collection of DAT and NET occupancy data to further investigate the presence of acute tolerance and assess the impact of low MPH doses on its efficacy.
Collapse
|
14
|
Yuan A, Kharas N, King N, Yang P, Dafny N. Methylphenidate cross-sensitization with amphetamine is dose dependent but not age dependent. Behav Brain Res 2023; 438:114178. [PMID: 36341913 DOI: 10.1016/j.bbr.2022.114178] [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: 08/17/2022] [Revised: 10/21/2022] [Accepted: 10/24/2022] [Indexed: 12/29/2022]
Abstract
Psychostimulants such as methylphenidate (MPD) and amphetamine (AMP) are often prescribed to young children and adolescents to treat behavioral disorders, or used to improve their intellectual performance in our competitive society. This is concerning as the temporal effects of how MPD exposure at a young age influences the response to MPD and AMP administration later in adulthood remains unclear. The objective of this study was to test whether MPD has the characteristics of substances that elicit behavioral symptoms of dependence and whether those effects are influenced by the initial age of MPD exposure. Three control and nine experimental groups of male rats were used. They were exposed to repetitive (chronic) 0.6, 2.5, or 10.0 mg/kg MPD in adolescence only, adulthood only, or adolescence and adulthood respectively. Then all groups were subsequently re-challenged with a single AMP dose in adulthood to test whether cross-sensitization between MPD and AMP was expressed, potentially as a result of prior MPD consumption. Exposure to 2.5 mg/kg and 10.0 mg/kg MPD in adolescence and adulthood or in adulthood alone led to cross-sensitization with AMP while exposure to 0.6 mg/kg MPD in adolescence and adulthood or in adulthood alone did not lead to cross-sensitization with AMP. Thus, these results indicate that MPD cross-sensitization with AMP is dose dependent.
Collapse
Affiliation(s)
- Anthony Yuan
- Department of Neurobiology and Anatomy, University of Texas Health at the McGovern Medical School, 6431 Fannin Street, Houston TX 77030, United States
| | - Natasha Kharas
- Department of Neurobiology and Anatomy, University of Texas Health at the McGovern Medical School, 6431 Fannin Street, Houston TX 77030, United States
| | - Nicholas King
- Department of Neurobiology and Anatomy, University of Texas Health at the McGovern Medical School, 6431 Fannin Street, Houston TX 77030, United States
| | - Pamela Yang
- Department of Neurobiology and Anatomy, University of Texas Health at the McGovern Medical School, 6431 Fannin Street, Houston TX 77030, United States
| | - Nachum Dafny
- Department of Neurobiology and Anatomy, University of Texas Health at the McGovern Medical School, 6431 Fannin Street, Houston TX 77030, United States.
| |
Collapse
|
15
|
Time-varying SUVr reflects the dynamics of dopamine increases during methylphenidate challenges in humans. Commun Biol 2023; 6:166. [PMID: 36765261 PMCID: PMC9918528 DOI: 10.1038/s42003-023-04545-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 02/01/2023] [Indexed: 02/12/2023] Open
Abstract
Dopamine facilitates cognition and is implicated in reward processing. Methylphenidate, a dopamine transporter blocker widely used to treat attention-deficit/hyperactivity disorder, can have rewarding and addictive effects if injected. Since methylphenidate's brain uptake is much faster after intravenous than oral intake, we hypothesize that the speed of dopamine increases in the striatum in addition to its amplitude underly drug reward. To test this we use simulations and PET data of [11C]raclopride's binding displacement with oral and intravenous methylphenidate challenges in 20 healthy controls. Simulations suggest that the time-varying difference in standardized uptake value ratios for [11C]raclopride between placebo and methylphenidate conditions is a proxy for the time-varying dopamine increases induced by methylphenidate. Here we show that the dopamine increase induced by intravenous methylphenidate (0.25 mg/kg) in the striatum is significantly faster than that by oral methylphenidate (60 mg), and its time-to-peak is strongly associated with the intensity of the self-report of "high". We show for the first time that the "high" is associated with the fast dopamine increases induced by methylphenidate.
Collapse
|
16
|
Pandita P, Bhalla R, Saini A, Mani I. Emerging tools for studying receptor endocytosis and signaling. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2023; 194:19-48. [PMID: 36631193 DOI: 10.1016/bs.pmbts.2022.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Ligands, agonists, or antagonists use receptor-mediated endocytosis (RME) to reach their intracellular targets. After the internalization of ligand-receptor complexes, it traffics through different subcellular organelles such as early endosome, recycling endosome, lysosome, etc. Further, after the ligand binding to the receptor, different second messengers are generated, such as cGMP, cAMP, IP3, etc. Several methods have been used, such as radioligand binding assay, western blotting, co-immunoprecipitation (co-IP), qRT-PCR, immunofluorescence and confocal microscopy, microRNA/siRNA, and bioassays to understand the various events, such as internalization, subcellular trafficking, signaling, metabolic degradation, etc. This chapter briefly discusses the key principles and methods used to study internalization, subcellular trafficking, signaling, and metabolic degradation of numerous receptors.
Collapse
Affiliation(s)
- Pratiksha Pandita
- Faculty of Medicine, Department of Infectious Disease, Imperial College London, London, United Kingdom
| | - Rhea Bhalla
- ICMR-National Institute of Virology, Pune, Maharashtra, India
| | - Ashok Saini
- Department of Microbiology, Institute of Home Economics, University of Delhi, New Delhi, India
| | - Indra Mani
- Department of Microbiology, Gargi College, University of Delhi, New Delhi, India.
| |
Collapse
|
17
|
Yamamoto M, Inada T. Positron emission tomography studies in adult patients with attention-deficit/hyperactivity disorder. Jpn J Radiol 2022; 41:382-392. [PMID: 36480104 DOI: 10.1007/s11604-022-01368-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 11/28/2022] [Indexed: 12/13/2022]
Abstract
Attention-deficit/hyperactivity disorder (ADHD) is a neurodevelopmental disorder characterized by inattention, motor hyperactivity, impulsivity, and psychosocial as well as cognitive dysfunction. Although characteristic clinical manifestations have been described, no definitive biomarkers to diagnose ADHD have been established. In this review article, we summarize positron emission tomography (PET) studies conducted in adult patients with ADHD. We found that, although, disturbances of dopamine, serotonin, and norepinephrine functions have been implicated in ADHD, no characteristic findings have been identified from PET studies in patients with ADHD. Several previous PET studies on the central dopaminergic transmission-related ligands in patients with ADHD have shown altered binding of dopamine markers in the basal ganglia. However, no consistent results were observed in the binding characteristics for dopamine transporters and receptors. Findings from PET studies with ligands related to serotonin and norepinephrine pathways showed either unclear clinical significance or low replicability. Therefore, whether alterations of monoamine function may be involved in the pathophysiological mechanism remains to be clarified. The limitations of previous PET studies include their small sample sizes, focus on several kinds of existing ligands, and a questionable validity of the diagnosis (lack of biological diagnostic criteria). To determine the characteristic findings for diagnosing ADHD, further research is needed, and particularly, studies that evaluate new active ligands with specific binding to monoamine pathways should be undertaken.
Collapse
Affiliation(s)
- Maeri Yamamoto
- Department of Psychiatry, Nagoya University Hospital, 65 Tsurumai-Cho, Showa-Ku, Nagoya-Shi, Aichi, 466-8560, Japan
| | - Toshiya Inada
- Department of Psychiatry, Graduate School of Medicine, Nagoya University, 65 Tsurumai-Cho, Showa-Ku, Nagoya-Shi, Aichi, 466-8550, Japan.
| |
Collapse
|
18
|
Zakiniaeiz Y, Hoye J, Ryan Petrulli J, LeVasseur B, Stanley G, Gao H, Najafzadeh S, Ropchan J, Nabulsi N, Huang Y, Chen MK, Matuskey D, Barron DS, Kelmendi B, Fulbright RK, Hampson M, Cosgrove KP, Morris ED. Systemic inflammation enhances stimulant-induced striatal dopamine elevation in tobacco smokers. Brain Behav Immun 2022; 106:262-269. [PMID: 36058419 PMCID: PMC10097458 DOI: 10.1016/j.bbi.2022.08.016] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 08/25/2022] [Accepted: 08/29/2022] [Indexed: 02/04/2023] Open
Abstract
Immune-brain interactions influence the pathophysiology of addiction. Lipopolysaccharide (LPS)-induced systemic inflammation produces effects on reward-related brain regions and the dopamine system. We previously showed that LPS amplifies dopamine elevation induced by methylphenidate (MP), compared to placebo (PBO), in eight healthy controls. However, the effects of LPS on the dopamine system of tobacco smokers have not been explored. The goal of Study 1 was to replicate previous findings in an independent cohort of tobacco smokers. The goal of Study 2 was to combine tobacco smokers with the aforementioned eight healthy controls to examine the effect of LPS on dopamine elevation in a heterogenous sample for power and effect size determination. Eight smokers were each scanned with [11C]raclopride positron emission tomography three times-at baseline, after administration of LPS (0.8 ng/kg, intravenously) and MP (40 mg, orally), and after administration of PBO and MP, in a double-blind, randomized order. Dopamine elevation was quantified as change in [11C]raclopride binding potential (ΔBPND) from baseline. A repeated-measures ANOVA was conducted to compare LPS and PBO conditions. Smokers and healthy controls were well-matched for demographics, drug dosing, and scanning parameters. In Study 1, MP-induced striatal dopamine elevation was significantly higher following LPS than PBO (p = 0.025, 18 ± 2.9 % vs 13 ± 2.7 %) for smokers. In Study 2, MP-induced striatal dopamine elevation was also significantly higher under LPS than under PBO (p < 0.001, 18 ± 1.6 % vs 11 ± 1.5 %) in the combined sample. Smoking status did not interact with the effect of condition. This is the first study to translate the phenomenon of amplified dopamine elevation after experimental activation of the immune system to an addicted sample which may have implications for drug reinforcement, seeking, and treatment.
Collapse
Affiliation(s)
- Yasmin Zakiniaeiz
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA; Yale Positron Emission Tomography (PET) Center, Yale School of Medicine, New Haven, CT, USA.
| | - Jocelyn Hoye
- Yale Positron Emission Tomography (PET) Center, Yale School of Medicine, New Haven, CT, USA; Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, USA
| | - Joseph Ryan Petrulli
- Yale Positron Emission Tomography (PET) Center, Yale School of Medicine, New Haven, CT, USA; Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, USA
| | | | - Gelsina Stanley
- Yale Positron Emission Tomography (PET) Center, Yale School of Medicine, New Haven, CT, USA; Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, USA
| | - Hong Gao
- Yale Positron Emission Tomography (PET) Center, Yale School of Medicine, New Haven, CT, USA; Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, USA
| | - Soheila Najafzadeh
- Yale Positron Emission Tomography (PET) Center, Yale School of Medicine, New Haven, CT, USA; Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, USA
| | - Jim Ropchan
- Yale Positron Emission Tomography (PET) Center, Yale School of Medicine, New Haven, CT, USA; Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, USA
| | - Nabeel Nabulsi
- Yale Positron Emission Tomography (PET) Center, Yale School of Medicine, New Haven, CT, USA; Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, USA
| | - Yiyun Huang
- Yale Positron Emission Tomography (PET) Center, Yale School of Medicine, New Haven, CT, USA; Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, USA
| | - Ming-Kai Chen
- Yale Positron Emission Tomography (PET) Center, Yale School of Medicine, New Haven, CT, USA; Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, USA
| | - David Matuskey
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA; Yale Positron Emission Tomography (PET) Center, Yale School of Medicine, New Haven, CT, USA; Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, USA
| | - Daniel S Barron
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA; Department of Psychiatry, Brigham & Women's Hospital, Boston, MA, USA; Department of Anesthesiology, Perioperative, and Pain Medicine, Brigham & Women's Hospital, Boston, MA, USA
| | - Benjamin Kelmendi
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
| | - Robert K Fulbright
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, USA
| | - Michelle Hampson
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA; Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, USA; Department of Biomedical Engineering, Yale School of Medicine, New Haven, CT, USA
| | - Kelly P Cosgrove
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA; Yale Positron Emission Tomography (PET) Center, Yale School of Medicine, New Haven, CT, USA
| | - Evan D Morris
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA; Yale Positron Emission Tomography (PET) Center, Yale School of Medicine, New Haven, CT, USA; Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, USA; Department of Psychiatry, Brigham & Women's Hospital, Boston, MA, USA
| |
Collapse
|
19
|
Avram M, Müller F, Rogg H, Korda A, Andreou C, Holze F, Vizeli P, Ley L, Liechti ME, Borgwardt S. Characterizing thalamocortical (dys)connectivity following d-amphetamine, LSD, and MDMA administration. BIOLOGICAL PSYCHIATRY. COGNITIVE NEUROSCIENCE AND NEUROIMAGING 2022; 7:885-894. [PMID: 35500840 DOI: 10.1016/j.bpsc.2022.04.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 03/28/2022] [Accepted: 04/13/2022] [Indexed: 12/19/2022]
Abstract
BACKGROUND Patients with psychotic disorders present alterations in thalamocortical intrinsic functional connectivity (iFC) as measured by resting-state functional magnetic resonance imaging (rs-fMRI). Specifically, thalamic iFC is increased with sensorimotor cortices (hyperconnectivity) and decreased with prefrontal-limbic cortices (hypoconnectivity). Intriguingly, psychedelics such as lysergic acid diethylamide (LSD) elicit similar thalamocortical-hyperconnectivity with sensorimotor areas in healthy volunteers. It is unclear whether LSD also induces thalamocortical-hypoconnectivity with prefrontal-limbic cortices as current findings are equivocal. Notably, thalamocortical-hyperconnectivity was associated with psychotic symptoms in patients and substance-induced altered states of consciousness in healthy volunteers. Thalamocortical dysconnectivity is likely evoked by altered neurotransmission, e.g., via dopaminergic excess in psychotic disorders and serotonergic agonism in psychedelic-induced states. It is unclear whether thalamocortical dysconnectivity is also elicited by amphetamine-type substances, broadly releasing monoamines (i.e., dopamine, norepinephrine) but producing fewer perceptual effects than psychedelics. METHODS We administrated LSD, d-amphetamine, and 3,4-methylenedioxymethamphetamine (MDMA) in 28 healthy volunteers and investigated their effects on thalamic iFC with two brain networks (auditory-sensorimotor (ASM) and salience (SAL) - corresponding to sensorimotor and prefrontal-limbic cortices, respectively), using a double-blind, placebo-controlled, cross-over design. RESULTS All active substances elicited ASM-thalamic-hyperconnectivity compared to placebo, despite predominantly distinct pharmacological actions and subjective effects. LSD-induced effects correlated with subjective changes in perception, indicating a link between hyperconnectivity and psychedelic-type perceptual alterations. Unlike d-amphetamine and MDMA, which induced hypoconnectivity with SAL, LSD elicited hyperconnectivity. D-amphetamine and MDMA evoked similar thalamocortical dysconnectivity patterns. CONCLUSIONS Psychedelics, empathogens, and psychostimulants evoke thalamocortical-hyperconnectivity with sensorimotor areas, akin to findings in patients with psychotic disorders.
Collapse
Affiliation(s)
- Mihai Avram
- Translational Psychiatry, Department of Psychiatry and Psychotherapy, University of Lübeck, Lübeck, 23538, Germany.
| | - Felix Müller
- Department of Psychiatry (UPK), University of Basel, Basel, 4012, Switzerland
| | - Helena Rogg
- Translational Psychiatry, Department of Psychiatry and Psychotherapy, University of Lübeck, Lübeck, 23538, Germany
| | - Alexandra Korda
- Translational Psychiatry, Department of Psychiatry and Psychotherapy, University of Lübeck, Lübeck, 23538, Germany
| | - Christina Andreou
- Translational Psychiatry, Department of Psychiatry and Psychotherapy, University of Lübeck, Lübeck, 23538, Germany
| | - Friederike Holze
- Division of Clinical Pharmacology and Toxicology, Department of Clinical Research, University Hospital Basel, University of Basel, Basel, 4031, Switzerland
| | - Patrick Vizeli
- Division of Clinical Pharmacology and Toxicology, Department of Clinical Research, University Hospital Basel, University of Basel, Basel, 4031, Switzerland
| | - Laura Ley
- Division of Clinical Pharmacology and Toxicology, Department of Clinical Research, University Hospital Basel, University of Basel, Basel, 4031, Switzerland
| | - Matthias E Liechti
- Division of Clinical Pharmacology and Toxicology, Department of Clinical Research, University Hospital Basel, University of Basel, Basel, 4031, Switzerland
| | - Stefan Borgwardt
- Translational Psychiatry, Department of Psychiatry and Psychotherapy, University of Lübeck, Lübeck, 23538, Germany
| |
Collapse
|
20
|
Reith MEA, Kortagere S, Wiers CE, Sun H, Kurian MA, Galli A, Volkow ND, Lin Z. The dopamine transporter gene SLC6A3: multidisease risks. Mol Psychiatry 2022; 27:1031-1046. [PMID: 34650206 PMCID: PMC9008071 DOI: 10.1038/s41380-021-01341-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 09/28/2021] [Accepted: 10/01/2021] [Indexed: 02/02/2023]
Abstract
The human dopamine transporter gene SLC6A3 has been consistently implicated in several neuropsychiatric diseases but the disease mechanism remains elusive. In this risk synthesis, we have concluded that SLC6A3 represents an increasingly recognized risk with a growing number of familial mutants associated with neuropsychiatric and neurological disorders. At least five loci were related to common and severe diseases including alcohol use disorder (high activity variant), attention-deficit/hyperactivity disorder (low activity variant), autism (familial proteins with mutated networking) and movement disorders (both regulatory variants and familial mutations). Association signals depended on genetic markers used as well as ethnicity examined. Strong haplotype selection and gene-wide epistases support multimarker assessment of functional variations and phenotype associations. Inclusion of its promoter region's functional markers such as DNPi (rs67175440) and 5'VNTR (rs70957367) may help delineate condensate-based risk action, testing a locus-pathway-phenotype hypothesis for one gene-multidisease etiology.
Collapse
Affiliation(s)
- Maarten E A Reith
- Department of Psychiatry, New York University School of Medicine, New York City, NY, 10016, USA
| | - Sandhya Kortagere
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, 19129, USA
| | - Corinde E Wiers
- Laboratory of Neuroimaging, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, 20817, USA
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Hui Sun
- Laboratory of Neuroimaging, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, 20817, USA
| | - Manju A Kurian
- Molecular Neurosciences, Developmental Neurosciences, Zayed Centre for Research into Rare Diseases in Children, UCL Great Ormond Street Institute of Child Health, and Department of Neurology, Great Ormond Street Hospital, London, WC1N 1EH, UK
| | - Aurelio Galli
- Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Nora D Volkow
- Laboratory of Neuroimaging, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, 20817, USA
- National Institute on Drug Abuse, Bethesda, MD, 20817, USA
| | - Zhicheng Lin
- Laboratory of Psychiatric Neurogenomics, McLean Hospital, and Department of Psychiatry, Harvard Medical School, Belmont, MA, 02478, USA.
| |
Collapse
|
21
|
Sex differences in methylphenidate-induced dopamine increases in ventral striatum. Mol Psychiatry 2022; 27:939-946. [PMID: 34707237 PMCID: PMC9043036 DOI: 10.1038/s41380-021-01294-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 08/16/2021] [Accepted: 09/08/2021] [Indexed: 12/02/2022]
Abstract
Sex differences in the prevalence of dopamine-related neuropsychiatric diseases and in the sensitivity to dopamine-boosting drugs such as stimulants is well recognized. Here we assessed whether there are sex differences in the brain dopamine system in humans that could contribute to these effects. We analyzed data from two independent [11C]raclopride PET brain imaging studies that measured methylphenidate-induced dopamine increases in the striatum using different routes of administration (Cohort A = oral 60 mg; Cohort B = intravenous 0.5 mg/kg; total n = 95; 65 male, 30 female), in blinded placebo-controlled designs. Females when compared to males reported stronger feeling of "drug effects" and showed significantly greater dopamine release in the ventral striatum (where nucleus accumbens is located) to both oral and intravenous methylphenidate. In contrast, there were no significant differences in methylphenidate-induced increases in dorsal striatum for either oral or intravenous administration nor were there differences in levels of methylphenidate in plasma. The greater dopamine increases with methylphenidate in ventral but not dorsal striatum in females compared to males suggests an enhanced sensitivity specific to the dopamine reward system that might underlie sex differences in the vulnerability to substance use disorders and to attention-deficit/hyperactivity disorder (ADHD).
Collapse
|
22
|
Abstract
Attention-Deficit Hyperactivity Disorder (ADHD) is a prevalent neurodevelopmental condition associated with impaired function and increased risk of poor outcomes in children, young people and adults with the condition. Currently approved pharmacological treatments for ADHD include a range of stimulant (methylphenidate, amphetamine) and nonstimulant (atomoxetine, guanfacine, clonidine) medications. All have been shown to be effective in treating the symptoms of ADHD and improving other functional outcomes including quality of life, academic performance, rates of accidents and injuries, and do not appear to be associated with significant adverse outcomes or side effects. In this chapter, we review medications for ADHD by summarising the mechanisms of action of each of the two main classes of compounds (stimulants and nonstimulants), the formulations of the most commonly prescribed medications within each class, their efficacy in treating ADHD symptoms and other outcomes, and other factors that influence treatment decisions including side effects and tolerability, comorbidities and medical history. We conclude with a summary of the treatment decisions made by clinicians and suggest some next steps for research. Further research is needed to understand the mechanisms of action of these medications and how exactly they improve symptoms, and to examine their effects on commonly occurring comorbidities.
Collapse
Affiliation(s)
- Madeleine J Groom
- Academic Unit of Mental Health and Clinical Neurosciences, School of Medicine, Institute of Mental Health, University of Nottingham, Nottingham, UK.
| | - Samuele Cortese
- Faculty of Environmental and Life Sciences, Center for Innovation in Mental Health, School of Psychology, University of Southampton, Southampton, UK
- Clinical and Experimental Sciences (CNS and Psychiatry), Faculty of Medicine, University of Southampton, Southampton, UK
- Solent NHS Trust, Southampton, UK
- Hassenfeld Children's Hospital at NYU Langone, New York University Child Study Center, New York, NY, USA
- Centre for ADHD and Neurodevelopmental Disorders Across the Lifespan, Institute of Mental Health, University of Nottingham, Nottingham, UK
| |
Collapse
|
23
|
Moon C, Marion M, Thanos PK, Steiner H. Fluoxetine Potentiates Oral Methylphenidate-Induced Gene Regulation in the Rat Striatum. Mol Neurobiol 2021; 58:4856-4870. [PMID: 34213723 DOI: 10.1007/s12035-021-02466-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 06/21/2021] [Indexed: 11/24/2022]
Abstract
Methylphenidate (MP) is combined with selective serotonin reuptake inhibitors (SSRIs) such as fluoxetine (FLX) to treat various disorders. MP, a dopamine reuptake inhibitor, helps manage attention-deficit hyperactivity disorder (ADHD) and is abused as a cognitive enhancer; it has a reduced addiction liability. We showed that combining FLX (serotonin) with MP potentiates MP-induced gene regulation in the striatum. These studies used intraperitoneal drug administration, which is relevant for MP abuse. Clinically, MP and FLX are taken orally (slower bioavailability). Here, we investigated whether chronic oral administration of MP and FLX also altered striatal gene regulation. MP (30/60 mg/kg/day), FLX (20 mg/kg/day), and MP + FLX were administered in rats' drinking water for 8 h/day over 4 weeks. We assessed the expression of dynorphin and substance P (both markers for striatal direct pathway neurons) and enkephalin (indirect pathway) by in situ hybridization histochemistry. Chronic oral MP alone produced a tendency for increased dynorphin and substance P expression and no changes in enkephalin expression. Oral FLX alone did not increase gene expression. In contrast, when given together, FLX greatly enhanced MP-induced expression of dynorphin and substance P and to a lesser degree enkephalin. Thus, FLX potentiated oral MP-induced gene regulation predominantly in direct pathway neurons, mimicking cocaine effects. The three functional domains of the striatum were differentially affected. MP + SSRI concomitant therapies are indicated in ADHD/depression comorbidity and co-exposure occurs with MP misuse as a cognitive enhancer by patients on SSRIs. Our findings indicate that MP + SSRI combinations, even given orally, may enhance addiction-related gene regulation.
Collapse
Affiliation(s)
- Connor Moon
- Stanson Toshok Center for Brain Function and Repair, Rosalind Franklin University of Medicine and Science, North Chicago, IL, 60064, USA
| | - Matt Marion
- Behavioral Neuropharmacology and Neuroimaging Laboratory, Clinical Research Institute On Addictions, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University At Buffalo, Buffalo, NY, 14203, USA
| | - Panayotis K Thanos
- Behavioral Neuropharmacology and Neuroimaging Laboratory, Clinical Research Institute On Addictions, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University At Buffalo, Buffalo, NY, 14203, USA
| | - Heinz Steiner
- Stanson Toshok Center for Brain Function and Repair, Rosalind Franklin University of Medicine and Science, North Chicago, IL, 60064, USA. .,Discipline of Cellular and Molecular Pharmacology, The Chicago Medical School, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Road, North Chicago, IL, 60064, USA.
| |
Collapse
|
24
|
Chang JC, Lin HY, Lv J, Tseng WYI, Gau SSF. Regional brain volume predicts response to methylphenidate treatment in individuals with ADHD. BMC Psychiatry 2021; 21:26. [PMID: 33430830 PMCID: PMC7798216 DOI: 10.1186/s12888-021-03040-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Accepted: 12/24/2020] [Indexed: 12/01/2022] Open
Abstract
BACKGROUND Despite the effectiveness of methylphenidate for treating ADHD, up to 30% of individuals with ADHD show poor responses to methylphenidate. Neuroimaging biomarkers to predict medication responses remain elusive. This study characterized neuroanatomical features that differentiated between clinically good and poor methylphenidate responders with ADHD. METHODS Using a naturalistic observation design selected from a larger cohort, we included 79 drug-naive individuals (aged 6-42 years) with ADHD without major psychiatric comorbidity, who had acceptable baseline structural MRI data quality. Based on a retrospective chart review, we defined responders by individuals' responses to at least one-month treatment with methylphenidate. A nonparametric mass-univariate voxel-based morphometric analysis was used to compare regional gray matter volume differences between good and poor responders. A multivariate pattern recognition based on the support vector machine was further implemented to identify neuroanatomical indicators to predict an individual's response. RESULTS 63 and 16 individuals were classified in the good and poor responder group, respectively. Using the small-volume correction procedure based on the hypothesis-driven striatal and default-mode network masks, poor responders had smaller regional volumes of the left putamen as well as larger precuneus volumes compared to good responders at baseline. The machine learning approach identified that volumetric information among these two regions alongside the left frontoparietal regions, occipital lobes, and posterior/inferior cerebellum could predict clinical responses to methylphenidate in individuals with ADHD. CONCLUSION Our results suggest regional striatal and precuneus gray matter volumes play a critical role in mediating treatment responses in individuals with ADHD.
Collapse
Affiliation(s)
- Jung-Chi Chang
- grid.412094.a0000 0004 0572 7815Department of Psychiatry, National Taiwan University Hospital, Taipei, Taiwan ,grid.412094.a0000 0004 0572 7815Department of Psychiatry, National Taiwan University Hospital, Hsin-Chu Branch, Hsin-Chu, Taiwan ,grid.19188.390000 0004 0546 0241Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Hsiang-Yuan Lin
- grid.155956.b0000 0000 8793 5925Azrieli Adult Neurodevelopmental Centre and Adult Neurodevelopment and Geriatric Psychiatry Division, Centre for Addiction and Mental Health, Toronto, Ontario Canada ,grid.17063.330000 0001 2157 2938Department of Psychiatry, University of Toronto, Toronto, Ontario Canada
| | - Junglei Lv
- grid.1013.30000 0004 1936 834XSydney Imaging and School of Biomedical Engineering, University of Sydney, Camperdown, NSW Australia
| | - Wen-Yih Issac Tseng
- grid.19188.390000 0004 0546 0241Graduate Institute of Brain and Mind Sciences, National Taiwan University College of Medicine, Taipei, Taiwan ,grid.19188.390000 0004 0546 0241Institute of Medical Device and Imaging, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Susan Shur-Fen Gau
- Department of Psychiatry, National Taiwan University Hospital, Taipei, Taiwan. .,Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan. .,Graduate Institute of Brain and Mind Sciences, National Taiwan University College of Medicine, Taipei, Taiwan. .,Department of Psychiatry, College of Medicine, National Taiwan University, Taipei, Taiwan.
| |
Collapse
|
25
|
Effects of methylphenidate on reinforcement learning depend on working memory capacity. Psychopharmacology (Berl) 2021; 238:3569-3584. [PMID: 34676440 PMCID: PMC8629893 DOI: 10.1007/s00213-021-05974-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 08/25/2021] [Indexed: 11/25/2022]
Abstract
RATIONALE Brain catecholamines have long been implicated in reinforcement learning, exemplified by catecholamine drug and genetic effects on probabilistic reversal learning. However, the mechanisms underlying such effects are unclear. OBJECTIVES AND METHODS Here we investigated effects of an acute catecholamine challenge with methylphenidate (20 mg, oral) on a novel probabilistic reversal learning paradigm in a within-subject, double-blind randomised design. The paradigm was designed to disentangle effects on punishment avoidance from effects on reward perseveration. Given the known large individual variability in methylphenidate's effects, we stratified our effects by working memory capacity and trait impulsivity, putatively modulating the effects of methylphenidate, in a large sample (n = 102) of healthy volunteers. RESULTS Contrary to our prediction, methylphenidate did not alter performance in the reversal phase of the task. Our key finding is that methylphenidate altered learning of choice-outcome contingencies in a manner that depended on individual variability in working memory span. Specifically, methylphenidate improved performance by adaptively reducing the effective learning rate in participants with higher working memory capacity. CONCLUSIONS This finding emphasises the important role of working memory in reinforcement learning, as reported in influential recent computational modelling and behavioural work, and highlights the dependence of this interplay on catecholaminergic function.
Collapse
|
26
|
Womack KB, Dubiel R, Callender L, Dunklin C, Dahdah M, Harris TS, Devous MD, Juengst SB, Bell K, Diaz-Arrastia R, Ding K. 123I-Iofluopane Single-Photon Emission Computed Tomography as an Imaging Biomarker of Pre-Synaptic Dopaminergic System after Moderate-to-Severe Traumatic Brain Injury. J Neurotrauma 2020; 37:2113-2119. [PMID: 32216525 DOI: 10.1089/neu.2019.6892] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Dopaminergic (DA) system function is frequently disrupted after traumatic brain injury (TBI). However, published interventions that target the DA system with the hope of enhancing functional outcomes are inconclusive, partially because of the lack of DA signaling biomarkers that can be used to select patients likely to benefit from DA-directed therapies or to monitor treatment efficacy. The aim of this study was to evaluate the feasibility of using 123I-iofluopane single-photon emission computerized tomography (SPECT) to assess pre-synaptic DA system dysfunction after severe TBI. Eighteen patients with severe TBI were enrolled in this study. 123I-iofluopane SPECT imaging was performed at baseline and again 2.5 h after a single dose of methylphenidate (MP) administered enterally. DA transporter (DAT) specific binding ratio (SBR) before and after MP was measured. Functional outcomes included the Disability Rating Scale, JFK Coma Recovery Scale-Revised, Functional Independence Measure, and Functional Assessment Measure. Thirteen of 18 patients completed the study. Average time from injury to SPECT scan was 48 days (standard deviation [SD], 24 days; median, 31). Baseline ioflupane striatal SBR was 1.51 ± 0.46 (median, 1.67). A 43.1% (SD, 16; median, 46.5) displacement of ioflupane from pre-synaptic DAT was observed after MP administration. Baseline SBR positively correlated with functional status at baseline and 4 weeks after completion of the study. Serum MP levels correlated with relative change in SBR (rs = 0.60; p = 0.04). Our findings suggest that 123I-iofluopane SPECT is a promising tool to determine the severity of pre-synaptic DA terminal disruption and for monitoring pharmacokinetics and pharmacodynamics of therapeutic interventions targeting the DA system.
Collapse
Affiliation(s)
- Kyle B Womack
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, Texas, USA.,Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Rosemary Dubiel
- Baylor Scott & White Institute for Rehabilitation, Dallas, Texas, USA.,Baylor Scott & White Health, Dallas, Texas, USA
| | - Librada Callender
- Baylor Scott & White Institute for Rehabilitation, Dallas, Texas, USA.,Baylor Scott & White Health, Dallas, Texas, USA
| | - Cynthia Dunklin
- Baylor Scott & White Institute for Rehabilitation, Dallas, Texas, USA.,Baylor Scott & White Health, Dallas, Texas, USA
| | | | - Thomas S Harris
- Avid Radiopharmaceuticals Inc, Philadelphia, Pennsylvania, USA
| | | | - Shannon B Juengst
- Department of Physical Medicine and Rehabilitation, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Kathleen Bell
- Department of Physical Medicine and Rehabilitation, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Ramon Diaz-Arrastia
- Department of Neurology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Kan Ding
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
27
|
The rate of dasotraline brain entry is slow following intravenous administration. Psychopharmacology (Berl) 2020; 237:3435-3446. [PMID: 32813030 PMCID: PMC7651685 DOI: 10.1007/s00213-020-05623-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 07/27/2020] [Indexed: 12/03/2022]
Abstract
RATIONALE Drugs that rapidly increase dopamine levels have an increased risk of abuse. Dasotraline (DAS) is a dopamine and norepinephrine reuptake inhibitor characterized by slow oral absorption with low potential for abuse. However, it remains unclear whether intravenous (i.v.) administration would facilitate the rapid elevation of dopamine levels associated with stimulant drugs. OBJECTIVE To assess the kinetics of DAS across the blood-brain barrier and time to onset of dopamine transporters (DAT) inhibition. METHODS We compared the onset of DAT occupancy and the associated elevation of synaptic dopamine levels in rhesus monkey following i.v. administration of DAS or methylphenidate (MPH) using positron emission tomography (PET). Brain entry times were estimated by reductions in [18F]-FE-PE2I binding to DAT in rhesus monkeys. Elevations of synaptic dopamine were estimated by reductions in [11C]-Raclopride binding to D2 receptors. RESULTS Intravenous administration of DAS (0.1 and 0.2 mg/kg) resulted in striatal DAT occupancies of 54% and 68%, respectively; i.v. administered MPH (0.1 and 0.5 mg/kg) achieved occupancies of 69% and 88% respectively. Brain entry times of DAS (22 and 15 min, respectively) were longer than for MPH (3 and 2 min). Elevations in synaptic dopamine were similar for both DAS and MPH however the time for half-maximal displacement by MPH (t = 23 min) was 4-fold more rapid than for DAS (t = 88 min). CONCLUSIONS These results demonstrate that the pharmacodynamics effects of DAS on DAT occupancy and synaptic dopamine levels are more gradual in onset than those of MPH even with i.v. administration that is favored by recreational drug abusers.
Collapse
|
28
|
Lövdén M, Karalija N, Andersson M, Wåhlin A, Axelsson J, Köhncke Y, Jonasson LS, Rieckman A, Papenberg G, Garrett DD, Guitart-Masip M, Salami A, Riklund K, Bäckman L, Nyberg L, Lindenberger U. Latent-Profile Analysis Reveals Behavioral and Brain Correlates of Dopamine-Cognition Associations. Cereb Cortex 2019; 28:3894-3907. [PMID: 29028935 DOI: 10.1093/cercor/bhx253] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Accepted: 09/07/2017] [Indexed: 01/14/2023] Open
Abstract
Evidence suggests that associations between the neurotransmitter dopamine and cognition are nonmonotonic and open to modulation by various other factors. The functional implications of a given level of dopamine may therefore differ from person to person. By applying latent-profile analysis to a large (n = 181) sample of adults aged 64-68 years, we probabilistically identified 3 subgroups that explain the multivariate associations between dopamine D2/3R availability (probed with 11C-raclopride-PET, in cortical, striatal, and hippocampal regions) and cognitive performance (episodic memory, working memory, and perceptual speed). Generally, greater receptor availability was associated with better cognitive performance. However, we discovered a subgroup of individuals for which high availability, particularly in striatum, was associated with poor performance, especially for working memory. Relative to the rest of the sample, this subgroup also had lower education, higher body-mass index, and lower resting-state connectivity between caudate nucleus and dorsolateral prefrontal cortex. We conclude that a smaller subset of individuals induces a multivariate non-linear association between dopamine D2/3R availability and cognitive performance in this group of older adults, and discuss potential reasons for these differences that await further empirical scrutiny.
Collapse
Affiliation(s)
- Martin Lövdén
- Aging Research Center, Karolinska Institutet and Stockholm University, Stockholm, Sweden
| | - Nina Karalija
- Department of Radiation Sciences, Umeå University, Umeå, Sweden.,Umeå Center for Functional Brain Imaging (UFBI), Umeå University, Umeå, Sweden
| | - Micael Andersson
- Umeå Center for Functional Brain Imaging (UFBI), Umeå University, Umeå, Sweden.,Department of Integrative Medical Biology, Umeå University, Umeå, Sweden
| | - Anders Wåhlin
- Department of Radiation Sciences, Umeå University, Umeå, Sweden
| | - Jan Axelsson
- Department of Radiation Sciences, Umeå University, Umeå, Sweden.,Umeå Center for Functional Brain Imaging (UFBI), Umeå University, Umeå, Sweden
| | - Ylva Köhncke
- Aging Research Center, Karolinska Institutet and Stockholm University, Stockholm, Sweden
| | - Lars S Jonasson
- Department of Radiation Sciences, Umeå University, Umeå, Sweden.,Umeå Center for Functional Brain Imaging (UFBI), Umeå University, Umeå, Sweden.,Center for Aging and Demographic Research, CEDAR, Umeå University, Umeå, Sweden
| | - Anna Rieckman
- Department of Radiation Sciences, Umeå University, Umeå, Sweden.,Umeå Center for Functional Brain Imaging (UFBI), Umeå University, Umeå, Sweden
| | - Goran Papenberg
- Aging Research Center, Karolinska Institutet and Stockholm University, Stockholm, Sweden
| | - Douglas D Garrett
- Max Planck UCL Centre for Computational Psychiatry and Ageing Research, Berlin, Germany.,Center for Lifespan Psychology, Max Planck Institute for Human Development, Berlin, Germany
| | - Marc Guitart-Masip
- Aging Research Center, Karolinska Institutet and Stockholm University, Stockholm, Sweden
| | - Alireza Salami
- Aging Research Center, Karolinska Institutet and Stockholm University, Stockholm, Sweden
| | - Katrine Riklund
- Department of Radiation Sciences, Umeå University, Umeå, Sweden.,Umeå Center for Functional Brain Imaging (UFBI), Umeå University, Umeå, Sweden
| | - Lars Bäckman
- Aging Research Center, Karolinska Institutet and Stockholm University, Stockholm, Sweden
| | - Lars Nyberg
- Department of Radiation Sciences, Umeå University, Umeå, Sweden.,Umeå Center for Functional Brain Imaging (UFBI), Umeå University, Umeå, Sweden.,Department of Integrative Medical Biology, Umeå University, Umeå, Sweden
| | - Ulman Lindenberger
- Max Planck UCL Centre for Computational Psychiatry and Ageing Research, Berlin, Germany.,Center for Lifespan Psychology, Max Planck Institute for Human Development, Berlin, Germany.,European University Institute, San Domenico di Fiesole (FI), Italy
| |
Collapse
|
29
|
Cook JL, Swart JC, Froböse MI, Diaconescu AO, Geurts DEM, den Ouden HEM, Cools R. Catecholaminergic modulation of meta-learning. eLife 2019; 8:e51439. [PMID: 31850844 PMCID: PMC6974360 DOI: 10.7554/elife.51439] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Accepted: 12/18/2019] [Indexed: 01/03/2023] Open
Abstract
The remarkable expedience of human learning is thought to be underpinned by meta-learning, whereby slow accumulative learning processes are rapidly adjusted to the current learning environment. To date, the neurobiological implementation of meta-learning remains unclear. A burgeoning literature argues for an important role for the catecholamines dopamine and noradrenaline in meta-learning. Here, we tested the hypothesis that enhancing catecholamine function modulates the ability to optimise a meta-learning parameter (learning rate) as a function of environmental volatility. 102 participants completed a task which required learning in stable phases, where the probability of reinforcement was constant, and volatile phases, where probabilities changed every 10-30 trials. The catecholamine transporter blocker methylphenidate enhanced participants' ability to adapt learning rate: Under methylphenidate, compared with placebo, participants exhibited higher learning rates in volatile relative to stable phases. Furthermore, this effect was significant only with respect to direct learning based on the participants' own experience, there was no significant effect on inferred-value learning where stimulus values had to be inferred. These data demonstrate a causal link between catecholaminergic modulation and the adjustment of the meta-learning parameter learning rate.
Collapse
Affiliation(s)
- Jennifer L Cook
- School of PsychologyUniversity of BirminghamBirminghamUnited Kingdom
| | - Jennifer C Swart
- Donders Institute for Brain, Cognition and Behaviour, Centre for Cognitive NeuroimagingRadboud UniversityNijmegenNetherlands
| | - Monja I Froböse
- Donders Institute for Brain, Cognition and Behaviour, Centre for Cognitive NeuroimagingRadboud UniversityNijmegenNetherlands
| | - Andreea O Diaconescu
- Translational Neuromodeling Unit, Institute for Biomedical EngineeringUniversity of Zurich and ETH ZurichZurichSwitzerland
- Department of PsychiatryUniversity of BaselBaselSwitzerland
- Krembil Centre for Neuroinformatics,CAMHUniversity of TorontoTorontoCanada
| | - Dirk EM Geurts
- Donders Institute for Brain, Cognition and Behaviour, Centre for Cognitive NeuroimagingRadboud UniversityNijmegenNetherlands
- Department of PsychiatryRadboud University Medical CentreNijmegenNetherlands
| | - Hanneke EM den Ouden
- Donders Institute for Brain, Cognition and Behaviour, Centre for Cognitive NeuroimagingRadboud UniversityNijmegenNetherlands
| | - Roshan Cools
- Donders Institute for Brain, Cognition and Behaviour, Centre for Cognitive NeuroimagingRadboud UniversityNijmegenNetherlands
- Department of PsychiatryRadboud University Medical CentreNijmegenNetherlands
| |
Collapse
|
30
|
Wiers CE, Lohoff FW, Lee J, Muench C, Freeman C, Zehra A, Marenco S, Lipska BK, Auluck PK, Feng N, Sun H, Goldman D, Swanson JM, Wang GJ, Volkow ND. Methylation of the dopamine transporter gene in blood is associated with striatal dopamine transporter availability in ADHD: A preliminary study. Eur J Neurosci 2019; 48:1884-1895. [PMID: 30033547 DOI: 10.1111/ejn.14067] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Revised: 06/12/2018] [Accepted: 07/04/2018] [Indexed: 12/23/2022]
Abstract
Dopamine transporters (DAT) are implicated in the pathogenesis and treatment of attention-deficit hyperactivity disorder (ADHD) and are upregulated by chronic treatment with methylphenidate, commonly prescribed for ADHD. Methylation of the DAT1 gene in brain and blood has been associated with DAT expression in rodents' brains. Here we tested the association between methylation of the DAT1 promoter derived from blood and DAT availability in the striatum of unmedicated ADHD adult participants and in that of healthy age-matched controls (HC) using Positron Emission Tomography (PET) and [11 C]cocaine. Results showed no between-group differences in DAT1 promoter methylation or striatal DAT availability. However, the degree of methylation in the promoter region of DAT1 correlated negatively with DAT availability in caudate in ADHD participants only. DAT availability in VS correlated with inattention scores in ADHD participants. We verified in a postmortem cohort with ADHD diagnosis and without, that DAT1 promoter methylation in peripheral blood correlated positively with DAT1 promoter methylation extracted from substantia nigra (SN) in both groups. In the cohort without ADHD diagnosis, DAT1 gene expression in SN further correlated positively with DAT protein expression in caudate; however, the sample size of the cohort with ADHD was insufficient to investigate DAT1 and DAT expression levels. Overall, these findings suggest that peripheral DAT1 promoter methylation may be predictive of striatal DAT availability in adults with ADHD. Due to the small sample size, more work is needed to validate whether DAT1 methylation in blood predicts DAT1 methylation in SN in ADHD and controls.
Collapse
Affiliation(s)
- Corinde E Wiers
- National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland
| | - Falk W Lohoff
- National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland
| | - Jisoo Lee
- National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland
| | - Christine Muench
- National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland
| | - Clara Freeman
- National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland
| | - Amna Zehra
- National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland
| | - Stefano Marenco
- National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland
| | - Barbara K Lipska
- National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland
| | - Pavan K Auluck
- National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland
| | - Ningping Feng
- National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland
| | - Hui Sun
- National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland
| | - David Goldman
- National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland
| | - James M Swanson
- Child Development Center, University of California, Irvine, California
| | - Gene-Jack Wang
- National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland
| | - Nora D Volkow
- National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland.,National Institute on Drug Abuse, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
31
|
Moreno-Salinas AL, Avila-Zozaya M, Ugalde-Silva P, Hernández-Guzmán DA, Missirlis F, Boucard AA. Latrophilins: A Neuro-Centric View of an Evolutionary Conserved Adhesion G Protein-Coupled Receptor Subfamily. Front Neurosci 2019; 13:700. [PMID: 31354411 PMCID: PMC6629964 DOI: 10.3389/fnins.2019.00700] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Accepted: 06/20/2019] [Indexed: 12/21/2022] Open
Abstract
The adhesion G protein-coupled receptors latrophilins have been in the limelight for more than 20 years since their discovery as calcium-independent receptors for α-latrotoxin, a spider venom toxin with potent activity directed at neurotransmitter release from a variety of synapse types. Latrophilins are highly expressed in the nervous system. Although a substantial amount of studies has been conducted to describe the role of latrophilins in the toxin-mediated action, the recent identification of endogenous ligands for these receptors helped confirm their function as mediators of adhesion events. Here we hypothesize a role for latrophilins in inter-neuronal contacts and the formation of neuronal networks and we review the most recent information on their role in neurons. We explore molecular, cellular and behavioral aspects related to latrophilin adhesion function in mice, zebrafish, Drosophila melanogaster and Caenorhabditis elegans, in physiological and pathophysiological conditions, including autism spectrum, bipolar, attention deficit and hyperactivity and substance use disorders.
Collapse
Affiliation(s)
- Ana L. Moreno-Salinas
- Department of Cell Biology, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Mexico City, Mexico
| | - Monserrat Avila-Zozaya
- Department of Cell Biology, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Mexico City, Mexico
| | - Paul Ugalde-Silva
- Department of Cell Biology, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Mexico City, Mexico
| | - David A. Hernández-Guzmán
- Department of Physiology, Biophysics and Neurosciences, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Mexico City, Mexico
| | - Fanis Missirlis
- Department of Physiology, Biophysics and Neurosciences, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Mexico City, Mexico
| | - Antony A. Boucard
- Department of Cell Biology, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Mexico City, Mexico
| |
Collapse
|
32
|
Addicott MA, Schechter JC, Sapyta JJ, Selig JP, Kollins SH, Weiss MD. Methylphenidate increases willingness to perform effort in adults with ADHD. Pharmacol Biochem Behav 2019; 183:14-21. [PMID: 31226260 DOI: 10.1016/j.pbb.2019.06.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 05/31/2019] [Accepted: 06/16/2019] [Indexed: 10/26/2022]
Abstract
BACKGROUND A reduced willingness to perform effort based on the magnitude and probability of potential rewards has been associated with diminished dopamine function and may be relevant to attention-deficit/hyperactivity disorder (ADHD). Here, we investigated the influence of ADHD status and methylphenidate on effort-based decisions. We hypothesized that ADHD participants would make fewer high-effort selections than non-ADHD subjects, and that methylphenidate would increase the number of high-effort selections. Furthermore, we hypothesized there would be associations among ADHD severity and methylphenidate-related changes in effort-based and attentional performance across all participants. METHODS AND PARTICIPANTS ADHD (n = 23) and non-ADHD (n = 23) adults completed the Effort Expenditure for Rewards Task in which participants select between low-effort and high-effort options to receive monetary rewards at varying levels of reward magnitude and probability. A test of attentional performance was also completed. RESULTS Overall, participants made more high-effort selections as potential reward magnitude and probability increased. ADHD participants did not make fewer high-effort selections than non-ADHD participants, but ADHD participants showed greater methylphenidate-related increases in high-effort selections. ADHD participants had worse attentional performance than non-ADHD participants. ADHD severity was associated with methylphenidate-related changes in high-effort selections, but not changes in attentional performance. CONCLUSIONS These results indicate that methylphenidate increases the willingness to perform effort in individuals with ADHD, possibly due to disorder-related motivational deficits. This provides support for theories of insufficient effort allocation among individuals with ADHD. TRIAL REGISTRATION Clinicaltrials.gov Identifier, NCT02630017.
Collapse
Affiliation(s)
| | | | | | - James P Selig
- University of Arkansas for Medical Sciences, United States of America
| | - Scott H Kollins
- Duke University School of Medicine, United States of America
| | - Margaret D Weiss
- University of Arkansas for Medical Sciences, United States of America
| |
Collapse
|
33
|
Rățală CE, Fallon SJ, van der Schaaf ME, Ter Huurne N, Cools R, Sanfey AG. Catecholaminergic modulation of trust decisions. Psychopharmacology (Berl) 2019; 236:1807-1816. [PMID: 30706097 PMCID: PMC6602982 DOI: 10.1007/s00213-019-5165-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 01/03/2019] [Indexed: 01/19/2023]
Abstract
RATIONALE Trust is a key component of social interactions. In order to assess the trustworthiness of others, people rely on both information learned from previous encounters, as well as on implicit biases associated with specific facial features. OBJECTIVE Here, we investigated the role of catecholamine (dopamine and noradrenaline) transmission on trust decisions as a function of both experienced behavior and facial features. METHODS To increase catecholamine levels, methylphenidate (MPH, i.e., Ritalin®, 20 mg) was administered to participants (N = 24) prior to their playing a well-studied economic task, namely the Trust Game (Berg et al. 1995). We measured the amount of money invested with a variety of game partners. Across game partners, we manipulated two aspects of trust: the facial trust level (high facial trust, low facial trust, and non-social) and the likelihood of reciprocation (high, low). RESULTS Results demonstrated no main effect of MPH on investments, but rather a selective lowering of investments under MPH as compared with placebo with the game partners who were low on facial trustworthiness and were low reciprocators. CONCLUSION These results provide evidence that MPH administration impacts social trust decision-making, but does so in a context-specific manner.
Collapse
Affiliation(s)
- Cătălina E Rățală
- Rotterdam School of Management, Erasmus University Rotterdam, Rotterdam, 3062, PA, The Netherlands.
- Donders Institute for Brain, Cognition and Behaviour, Centre for Cognitive Neuroimaging, Radboud University, 6500, HB, Nijmegen, The Netherlands.
| | - Sean J Fallon
- Donders Institute for Brain, Cognition and Behaviour, Centre for Cognitive Neuroimaging, Radboud University, 6500, HB, Nijmegen, The Netherlands
| | - Marieke E van der Schaaf
- Donders Institute for Brain, Cognition and Behaviour, Centre for Cognitive Neuroimaging, Radboud University, 6500, HB, Nijmegen, The Netherlands
- Department of Psychiatry, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Niels Ter Huurne
- Donders Institute for Brain, Cognition and Behaviour, Centre for Cognitive Neuroimaging, Radboud University, 6500, HB, Nijmegen, The Netherlands
- Karakter Child and Adolescent Psychiatry University Centre, Nijmegen, The Netherlands
| | - Roshan Cools
- Donders Institute for Brain, Cognition and Behaviour, Centre for Cognitive Neuroimaging, Radboud University, 6500, HB, Nijmegen, The Netherlands
- Department of Psychiatry, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Alan G Sanfey
- Donders Institute for Brain, Cognition and Behaviour, Centre for Cognitive Neuroimaging, Radboud University, 6500, HB, Nijmegen, The Netherlands
- Behavioral Science Institute, Radboud University, Nijmegen, The Netherlands
| |
Collapse
|
34
|
PharmGKB summary: methylphenidate pathway, pharmacokinetics/pharmacodynamics. Pharmacogenet Genomics 2019; 29:136-154. [PMID: 30950912 DOI: 10.1097/fpc.0000000000000376] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
35
|
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: 1.2] [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
|
36
|
Wang GJ, Wiers CE, Shumay E, Tomasi D, Yuan K, Wong CT, Logan J, Fowler JS, Volkow ND. Expectation effects on brain dopamine responses to methylphenidate in cocaine use disorder. Transl Psychiatry 2019; 9:93. [PMID: 30770780 PMCID: PMC6377670 DOI: 10.1038/s41398-019-0421-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Accepted: 01/04/2019] [Indexed: 11/29/2022] Open
Abstract
The response to drugs of abuse is affected by expectation, which is modulated in part by dopamine (DA), which encodes for a reward prediction error. Here we assessed the effect of expectation on methylphenidate (MP)-induced striatal DA changes in 23 participants with an active cocaine use disorder (CUD) and 23 healthy controls (HC) using [11C]raclopride and PET both after placebo (PL) and after MP (0.5 mg/kg, i.v.). Brain dopamine D2 and D3 receptor availability (D2R: non-displaceable binding potential (BPND)) was measured under four conditions in randomized order: (1) expecting PL/receiving PL, (2) expecting PL/receiving MP, (3) expecting MP/receiving PL, and (4) expecting MP/receiving MP. Expecting MP increased pulse rate compared to expecting PL. Receiving MP decreased D2R in striatum compared to PL, indicating MP-induced striatal DA release, and this effect was significantly blunted in CUD versus HC consistent with prior findings of decreased striatal dopamine responses both in active and detoxified CUD. There was a group × challenge × expectation effect in caudate and midbrain, with expectation of MP increasing MP-induced DA release in HC but not in CUD, and expectation of PL showing a trend to increase MP-induced DA release in CUD but not in HC. These results are consistent with the role of DA in reward prediction error in the human brain: decreasing DA signaling when rewards are less than expected (blunted DA increases to MP in CUD) and increasing them when greater than expected (for PL in CUD reflecting conditioned responses to injection). Our findings also document disruption of the expectation of drug effects in dopamine signaling in participants with CUD compared to non-addicted individuals.
Collapse
Affiliation(s)
- Gene-Jack Wang
- Laboratory of Neuroimaging, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, 20892-1013, USA.
| | - Corinde E. Wiers
- 0000 0004 0481 4802grid.420085.bLaboratory of Neuroimaging, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD 20892-1013 USA
| | - Elena Shumay
- 0000 0004 0481 4802grid.420085.bLaboratory of Neuroimaging, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD 20892-1013 USA
| | - Dardo Tomasi
- 0000 0004 0481 4802grid.420085.bLaboratory of Neuroimaging, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD 20892-1013 USA
| | - Kai Yuan
- 0000 0004 0481 4802grid.420085.bLaboratory of Neuroimaging, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD 20892-1013 USA ,0000 0001 0707 115Xgrid.440736.2School of Life Science and Technology, Xidian University, 710071 Xi’an, Shaanxi China
| | - Christopher T. Wong
- 0000 0004 0481 4802grid.420085.bLaboratory of Neuroimaging, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD 20892-1013 USA
| | - Jean Logan
- 0000 0004 1936 8753grid.137628.9Department of Radiology, New York University, New York, NY 11793 USA
| | - Joanna S. Fowler
- 0000 0004 0481 4802grid.420085.bLaboratory of Neuroimaging, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD 20892-1013 USA ,0000 0001 2188 4229grid.202665.5Brookhaven National Laboratory, Upton, NY 11973 USA
| | - Nora D. Volkow
- 0000 0004 0481 4802grid.420085.bLaboratory of Neuroimaging, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD 20892-1013 USA ,0000 0001 2297 5165grid.94365.3dNational Institute on Drug Abuse, National Institutes of Health, Bethesda, MD 20892 USA
| |
Collapse
|
37
|
Luo Y, Weibman D, Halperin JM, Li X. A Review of Heterogeneity in Attention Deficit/Hyperactivity Disorder (ADHD). Front Hum Neurosci 2019; 13:42. [PMID: 30804772 PMCID: PMC6378275 DOI: 10.3389/fnhum.2019.00042] [Citation(s) in RCA: 152] [Impact Index Per Article: 30.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Accepted: 01/25/2019] [Indexed: 12/24/2022] Open
Abstract
Attention-deficit/hyperactivity disorder (ADHD) is a neurodevelopmental disorder that affects approximately 8%–12% of children worldwide. Throughout an individual’s lifetime, ADHD can significantly increase risk for other psychiatric disorders, educational and occupational failure, accidents, criminality, social disability and addictions. No single risk factor is necessary or sufficient to cause ADHD. The multifactorial causation of ADHD is reflected in the heterogeneity of this disorder, as indicated by its diversity of psychiatric comorbidities, varied clinical profiles, patterns of neurocognitive impairment and developmental trajectories, and the wide range of structural and functional brain anomalies. Although evidence-based treatments can reduce ADHD symptoms in a substantial portion of affected individuals, there is yet no curative treatment for ADHD. A number of theoretical models of the emergence and developmental trajectories of ADHD have been proposed, aimed at providing systematic guides for clinical research and practice. We conducted a comprehensive review of the current status of research in understanding the heterogeneity of ADHD in terms of etiology, clinical profiles and trajectories, and neurobiological mechanisms. We suggest that further research focus on investigating the impact of the etiological risk factors and their interactions with developmental neural mechanisms and clinical profiles in ADHD. Such research would have heuristic value for identifying biologically homogeneous subgroups and could facilitate the development of novel and more tailored interventions that target underlying neural anomalies characteristic of more homogeneous subgroups.
Collapse
Affiliation(s)
- Yuyang Luo
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, United States
| | - Dana Weibman
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, United States
| | - Jeffrey M Halperin
- Department of Psychology, Queens College of the City University of New York, Flushing, NY, United States
| | - Xiaobo Li
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, United States.,Department of Electric and Computer Engineering, New Jersey Institute of Technology, Newark, NJ, United States
| |
Collapse
|
38
|
Dopaminergic Mechanisms Underlying Normal Variation in Trait Anxiety. J Neurosci 2019; 39:2735-2744. [PMID: 30737306 DOI: 10.1523/jneurosci.2382-18.2019] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 01/03/2019] [Accepted: 01/29/2019] [Indexed: 02/07/2023] Open
Abstract
Trait anxiety has been associated with altered activity within corticolimbic pathways connecting the amygdala and rostral anterior cingulate cortex (rACC), which receive rich dopaminergic input. Though the popular culture uses the term "chemical imbalance" to describe the pathophysiology of psychiatric conditions such as anxiety disorders, we know little about how individual differences in human dopamine neurochemistry are related to variation in anxiety and activity within corticolimbic circuits. We addressed this issue by examining interindividual variability in dopamine release at rest using [11C]raclopride positron emission tomography (PET), functional connectivity between amygdala and rACC using resting-state functional magnetic resonance imaging (fMRI), and trait anxiety measures in healthy adult male and female humans. To measure endogenous dopamine release, we collected two [11C]raclopride PET scans per participant. We contrasted baseline [11C]raclopride D2/3 receptor binding and D2/3 receptor binding following oral methylphenidate administration. Methylphenidate blocks the dopamine transporter, which increases extracellular dopamine and leads to reduced [11C]raclopride D2/3 receptor binding via competitive displacement. We found that individuals with higher dopamine release in the amygdala and rACC self-reported lower trait anxiety. Lower trait anxiety was also associated with reduced rACC-amygdala functional connectivity at baseline. Further, functional connectivity showed a modest negative relationship with dopamine release such that reduced rACC-amygdala functional connectivity was accompanied by higher levels of dopamine release in these regions. Together, these findings contribute to hypodopaminergic models of anxiety and support the utility of combining fMRI and PET measures of neurochemical function to advance our understanding of basic affective processes in humans.SIGNIFICANCE STATEMENT It is common wisdom that individuals vary in their baseline levels of anxiety. We all have a friend or colleague we know to be more "tightly wound" than others, or, perhaps, we are the ones marveling at others' ability to "just go with the flow." Although such observations about individual differences within nonclinical populations are commonplace, the neural mechanisms underlying normal variation in trait anxiety have not been established. Using multimodal brain imaging in humans, this study takes initial steps in linking intrinsic measures of neuromodulator release and functional connectivity within regions implicated in anxiety disorders. Our findings suggest that in healthy adults, higher levels of trait anxiety may arise, at least in part, from reduced dopamine neurotransmission.
Collapse
|
39
|
Farias TL, Marinho V, Carvalho V, Rocha K, da Silva PRA, Silva F, Teles AS, Gupta D, Ribeiro P, Velasques B, Cagy M, Bastos VH, Silva-Junior F, Teixeira S. Methylphenidate modifies activity in the prefrontal and parietal cortex accelerating the time judgment. Neurol Sci 2019; 40:829-837. [PMID: 30693423 DOI: 10.1007/s10072-018-3699-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Accepted: 12/31/2018] [Indexed: 12/30/2022]
Abstract
Methylphenidate produces its effects via actions on cortical areas involved with attention and working memory, which have a direct role in time estimation judgment tasks. In particular, the prefrontal and parietal cortex has been the target of several studies to understand the effect of methylphenidate on executive functions and time interval perception. However, it has not yet been studied whether acute administration of methylphenidate influences performance in time estimation task and the changes in alpha band absolute power in the prefrontal and parietal cortex. The current study investigates the influence of the acute use of methylphenidate in both performance and judgment in the time estimation interpretation through the alpha band absolute power activity in the prefrontal and parietal cortex. This is a double-blind, crossover study with a sample of 32 subjects under control (placebo) and experimental (methylphenidate) conditions with absolute alpha band power analysis during a time estimation task. We observed that methylphenidate does not influence task performance (p > 0.05), but it increases the time interval underestimation by over 7 s (p < 0.001) with a concomitant decrease in absolute alpha band power in the ventrolateral prefrontal cortex and dorsolateral prefrontal cortex and parietal cortex (p < 0.001). Acute use of methylphenidate increases the time interval underestimation, consistent with reduced accuracy of the internal clock mechanisms. Furthermore, acute use of methylphenidate influences the absolute alpha band power over the dorsolateral prefrontal cortex, ventrolateral prefrontal cortex, and parietal cortex.
Collapse
Affiliation(s)
- Tiago Lopes Farias
- Neuro-innovation Technology and Brain Mapping Laboratory, Federal University of Piauí, Av. São Sebastião, 2819, Bairro São Benedito, Parnaíba, Piauí, CEP: 64202-020, Brazil.
| | - Victor Marinho
- Neuro-innovation Technology and Brain Mapping Laboratory, Federal University of Piauí, Av. São Sebastião, 2819, Bairro São Benedito, Parnaíba, Piauí, CEP: 64202-020, Brazil. .,The Northeast Biotechnology Network, Federal University of Piauí, Teresina, Brazil.
| | - Valécia Carvalho
- Neuro-innovation Technology and Brain Mapping Laboratory, Federal University of Piauí, Av. São Sebastião, 2819, Bairro São Benedito, Parnaíba, Piauí, CEP: 64202-020, Brazil.,The Northeast Biotechnology Network, Federal University of Piauí, Teresina, Brazil
| | - Kaline Rocha
- Neuro-innovation Technology and Brain Mapping Laboratory, Federal University of Piauí, Av. São Sebastião, 2819, Bairro São Benedito, Parnaíba, Piauí, CEP: 64202-020, Brazil.,The Northeast Biotechnology Network, Federal University of Piauí, Teresina, Brazil
| | - Paulo Ramiler Alves da Silva
- Neuro-innovation Technology and Brain Mapping Laboratory, Federal University of Piauí, Av. São Sebastião, 2819, Bairro São Benedito, Parnaíba, Piauí, CEP: 64202-020, Brazil.,Masters Programs in Biotechnology, Federal University of Piauí, Parnaíba, Brazil
| | - Francisca Silva
- Neuro-innovation Technology and Brain Mapping Laboratory, Federal University of Piauí, Av. São Sebastião, 2819, Bairro São Benedito, Parnaíba, Piauí, CEP: 64202-020, Brazil
| | - Ariel Soares Teles
- Neuro-innovation Technology and Brain Mapping Laboratory, Federal University of Piauí, Av. São Sebastião, 2819, Bairro São Benedito, Parnaíba, Piauí, CEP: 64202-020, Brazil
| | - Daya Gupta
- Department of Biology, Camden County College, Blackwood, NJ, USA
| | - Pedro Ribeiro
- Brain Mapping and Sensory Motor Integration Laboratory, Institute of Psychiatry of Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Bruna Velasques
- Brain Mapping and Sensory Motor Integration Laboratory, Institute of Psychiatry of Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Mauricio Cagy
- Brain Mapping and Sensory Motor Integration Laboratory, Institute of Psychiatry of Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Victor Hugo Bastos
- Brain Mapping and Functionality Laboratory, Federal University of Piauí, Parnaíba, Brazil
| | - Fernando Silva-Junior
- Neuro-innovation Technology and Brain Mapping Laboratory, Federal University of Piauí, Av. São Sebastião, 2819, Bairro São Benedito, Parnaíba, Piauí, CEP: 64202-020, Brazil
| | - Silmar Teixeira
- Neuro-innovation Technology and Brain Mapping Laboratory, Federal University of Piauí, Av. São Sebastião, 2819, Bairro São Benedito, Parnaíba, Piauí, CEP: 64202-020, Brazil.,The Northeast Biotechnology Network, Federal University of Piauí, Teresina, Brazil.,Masters Programs in Biotechnology, Federal University of Piauí, Parnaíba, Brazil
| |
Collapse
|
40
|
Castrellon JJ, Seaman KL, Crawford JL, Young JS, Smith CT, Dang LC, Hsu M, Cowan RL, Zald DH, Samanez-Larkin GR. Individual Differences in Dopamine Are Associated with Reward Discounting in Clinical Groups But Not in Healthy Adults. J Neurosci 2019; 39:321-332. [PMID: 30446530 PMCID: PMC6325254 DOI: 10.1523/jneurosci.1984-18.2018] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2018] [Revised: 09/20/2018] [Accepted: 11/04/2018] [Indexed: 01/01/2023] Open
Abstract
Some people are more willing to make immediate, risky, or costly reward-focused choices than others, which has been hypothesized to be associated with individual differences in dopamine (DA) function. In two studies using PET imaging, one empirical (Study 1: N = 144 males and females across 3 samples) and one meta-analytic (Study 2: N = 307 across 12 samples), we sought to characterize associations between individual differences in DA and time, probability, and physical effort discounting in human adults. Study 1 demonstrated that individual differences in DA D2-like receptors were not associated with time or probability discounting of monetary rewards in healthy humans, and associations with physical effort discounting were inconsistent across adults of different ages. Meta-analytic results for temporal discounting corroborated our empirical finding for minimal effect of DA measures on discounting in healthy individuals but suggested that associations between individual differences in DA and reward discounting depend on clinical features. Addictions were characterized by negative correlations between DA and discounting, but other clinical conditions, such as Parkinson's disease, obesity, and attention-deficit/hyperactivity disorder, were characterized by positive correlations between DA and discounting. Together, the results suggest that trait differences in discounting in healthy adults do not appear to be strongly associated with individual differences in D2-like receptors. The difference in meta-analytic correlation effects between healthy controls and individuals with psychopathology suggests that individual difference findings related to DA and reward discounting in clinical samples may not be reliably generalized to healthy controls, and vice versa.SIGNIFICANCE STATEMENT Decisions to forgo large rewards for smaller ones due to increasing time delays, uncertainty, or physical effort have been linked to differences in dopamine (DA) function, which is disrupted in some forms of psychopathology. It remains unclear whether alterations in DA function associated with psychopathology also extend to explaining associations between DA function and decision making in healthy individuals. We show that individual differences in DA D2 receptor availability are not consistently related to monetary discounting of time, probability, or physical effort in healthy individuals across a broad age range. By contrast, we suggest that psychopathology accounts for observed inconsistencies in the relationship between measures of DA function and reward discounting behavior.
Collapse
Affiliation(s)
- Jaime J Castrellon
- Department of Psychology and Neuroscience,
- Center for Cognitive Neuroscience
| | - Kendra L Seaman
- Center for Cognitive Neuroscience
- Center for the Study of Aging and Human Development, Duke University, Durham, North Carolina 27708
- Department of Psychology, Yale University, New Haven, Connecticut 06511
| | | | - Jacob S Young
- Department of Psychology, Vanderbilt University, Nashville, Tennessee 37240
| | | | - Linh C Dang
- Department of Psychology, Vanderbilt University, Nashville, Tennessee 37240
| | - Ming Hsu
- Haas School of Business, University of California Berkeley, Berkeley, California 94720
| | - Ronald L Cowan
- Department of Psychology, Vanderbilt University, Nashville, Tennessee 37240
- Department of Psychiatry and Behavioral Sciences, Vanderbilt University School of Medicine, and Nashville, Tennessee 37212
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, Tennessee 37232
| | - David H Zald
- Department of Psychology, Vanderbilt University, Nashville, Tennessee 37240
- Department of Psychiatry and Behavioral Sciences, Vanderbilt University School of Medicine, and Nashville, Tennessee 37212
| | - Gregory R Samanez-Larkin
- Department of Psychology and Neuroscience
- Center for Cognitive Neuroscience
- Center for the Study of Aging and Human Development, Duke University, Durham, North Carolina 27708
- Department of Psychology, Yale University, New Haven, Connecticut 06511
| |
Collapse
|
41
|
Changes in Endogenous Dopamine Induced by Methylphenidate Predict Functional Connectivity in Nonhuman Primates. J Neurosci 2018; 39:1436-1444. [PMID: 30530859 DOI: 10.1523/jneurosci.2513-18.2018] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 11/09/2018] [Accepted: 12/04/2018] [Indexed: 11/21/2022] Open
Abstract
Dopamine (DA) levels in the striatum are increased by many therapeutic drugs, such as methylphenidate (MPH), which also alters behavioral and cognitive functions thought to be controlled by the PFC dose-dependently. We linked DA changes and functional connectivity (FC) using simultaneous [18F]fallypride PET and resting-state fMRI in awake male rhesus monkeys after oral administration of various doses of MPH. We found a negative correlation between [18F]fallypride nondisplaceable binding potential (BPND) and MPH dose in the head of the caudate (hCd), demonstrating increased extracellular DA resulting from MPH administration. The decreased BPND was negatively correlated with FC between the hCd and the PFC. Subsequent voxelwise analyses revealed negative correlations with FC between the hCd and the dorsolateral PFC, hippocampus, and precuneus. These results, showing that MPH-induced changes in DA levels in the hCd predict resting-state FC, shed light on a mechanism by which changes in striatal DA could influence function in the PFC.SIGNIFICANCE STATEMENT Dopamine transmission is thought to play an essential role in shaping large scale-neural networks that underlie cognitive functions. It is the target of therapeutic drugs, such as methylphenidate (Ritalin), which blocks the dopamine transporter, thereby increasing extracellular dopamine levels. Methylphenidate is used extensively to treat attention deficit hyperactivity disorder, even though its effects on cognitive functions and their underlying neural mechanisms are not well understood. To date, little is known about the link between changes in dopamine levels and changes in functional brain organization. Using simultaneous PET/MR imaging, we show that methylphenidate-induced changes in endogenous dopamine levels in the head of the caudate predict changes in resting-state functional connectivity between this structure and the prefrontal cortex, precuneus, and hippocampus.
Collapse
|
42
|
Withdrawal-Emergent Dyskinesia After Acute Discontinuation of Risperidone in a Child With Autism Spectrum Disorder. J Clin Psychopharmacol 2018; 38:640-642. [PMID: 30285999 DOI: 10.1097/jcp.0000000000000958] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
43
|
Akay AP, Kaya GÇ, Kose S, Yazıcıoğlu ÇE, Erkuran HÖ, Güney SA, Oğuz K, Keskin D, Baykara B, Emiroğlu Nİ, Eren MŞ, Kızıldağ S, Ertay T, Özsoylu D, Miral S, Durak H, Gönül AS, Rohde LA. Genetic imaging study with [Tc- 99m] TRODAT-1 SPECT in adolescents with ADHD using OROS-methylphenidate. Prog Neuropsychopharmacol Biol Psychiatry 2018; 86:294-300. [PMID: 29684537 DOI: 10.1016/j.pnpbp.2018.04.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 04/11/2018] [Accepted: 04/18/2018] [Indexed: 01/13/2023]
Abstract
AIM To examine theeffects on the brain of 2-month treatment withamethylphenidate extended-release formulation (OROS-MPH) using [Tc-99m] TRODAT-1SPECT in a sample of treatment-naïve adolescents with Attention Deficit/Hyperactivity Disorder (ADHD). In addition, to assess whether risk alleles (homozygosity for 10-repeat allele at the DAT1 gene were associated with alterations in striatal DAT availability. METHODS Twenty adolescents with ADHD underwent brain single-photon emission computed tomography (SPECT) scans with [Tc-99m] TRODAT-1 at baseline and two months after starting OROS-MPH treatment with dosages up to 1 mg/kg/day. Severity of illness was estimated using the Clinical Global Impression Scale (CGI-S) and DuPaul ADHD Rating Scale-Clinician version (ARS) before treatment,1 month and 2 months after initiating OROS-MPH treatment. RESULTS Decreased DAT availability was found in both the right caudate (pretreatment DAT binding: 224.76 ± 33.77, post-treatment DAT binding: 208.86 ± 28.75, p = 0.02) and right putamen (pre-treatment DAT binding: 314.41 ± 55.24, post-treatment DAT binding: 285.66 ± 39.20, p = 0.05) in adolescents with ADHD receiving OROS-MPH treatment. Adolescents with ADHD who showed a robust response to OROS-MPH (n = 7) had significantly greater reduction of DAT density in the right putamen than adolescents who showed less robust response to OROS-MPH (n = 13) (p = 0.02). However, between-group differences by treatment responses were not related with DAT density in the right caudate. Risk alleles (homozygosity for the 10-repeat allele of DAT1 gene) in the DAT1 gene were not associated with alterations in striatal DAT availability. CONCLUSION Two months of OROS-MPH treatment decreased DAT availability in both the right caudate and putamen. Adolescents with ADHD who showed a robust response to OROS-MPH had greater reduction of DAT density in the right putamen. However,our findings did not support an association between homozygosity for a 10-repeat allele in the DAT1 gene and DAT density, assessedusing[Tc-99m] TRODAT-1SPECT.
Collapse
Affiliation(s)
- Aynur Pekcanlar Akay
- Deparment of Child and Adolescent Psychiatry, Dokuz Eylul University, Izmir, Turkey.
| | - Gamze Çapa Kaya
- Deparment of Nuclear Medicine, Dokuz Eylul University, Izmir, Turkey
| | - Samet Kose
- Department of Psychology, H. Kalyoncu University, Gaziantep, Turkey; Medical School of Houston, Center for Neurobehavioral Research on Addictions, University of Texas, Houston, TX, USA
| | | | - Handan Özek Erkuran
- Dr.Behçet Uz Pediatrics, Pediatric Surgery Research andTraining Hospital, Deparment of Child and Adolescent Psychiatry, Izmir, Turkey.
| | - Sevay Alşen Güney
- Deparment of Child and Adolescent Psychiatry, Dokuz Eylul University, Izmir, Turkey
| | - Kaya Oğuz
- Department of Computer Engineering, Izmir University of Economics, Izmir, Turkey
| | - Duygu Keskin
- Deparment of Psychiatry, EgianUniversity, Izmir, Turkey
| | - Burak Baykara
- Deparment of Child and Adolescent Psychiatry, Dokuz Eylul University, Izmir, Turkey
| | | | - Mine Şencan Eren
- Deparment of Nuclear Medicine, Dokuz Eylul University, Izmir, Turkey
| | - Sefa Kızıldağ
- Deparment of Molecular Biology and Genetics, Dokuz Eylul University, Izmir,Turkey
| | - Türkan Ertay
- Deparment of Nuclear Medicine, Dokuz Eylul University, Izmir, Turkey
| | - Dua Özsoylu
- Deparment of Molecular Biology and Genetics, Dokuz Eylul University, Izmir,Turkey
| | - Süha Miral
- Deparment of Child and Adolescent Psychiatry, Dokuz Eylul University, Izmir, Turkey
| | - Hatice Durak
- Deparment of Nuclear Medicine, Dokuz Eylul University, Izmir, Turkey
| | | | - Luis Augusto Rohde
- Department of Psychiatry, Federal University of Rio Grande do Sul, Brazil; ADHD Program, Hospital de Clínicas de Porto Alegre, Brazil
| |
Collapse
|
44
|
Dopamine Synthesis Capacity is Associated with D2/3 Receptor Binding but Not Dopamine Release. Neuropsychopharmacology 2018; 43:1201-1211. [PMID: 28816243 PMCID: PMC5916345 DOI: 10.1038/npp.2017.180] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Revised: 08/07/2017] [Accepted: 08/10/2017] [Indexed: 01/08/2023]
Abstract
Positron Emission Tomography (PET) imaging allows the estimation of multiple aspects of dopamine function including dopamine synthesis capacity, dopamine release, and D2/3 receptor binding. Though dopaminergic dysregulation characterizes a number of neuropsychiatric disorders including schizophrenia and addiction, there has been relatively little investigation into the nature of relationships across dopamine markers within healthy individuals. Here we used PET imaging in 40 healthy adults to compare, within individuals, the estimates of dopamine synthesis capacity (Ki) using 6-[18F]fluoro-l-m-tyrosine ([18F]FMT; a substrate for aromatic amino acid decarboxylase), baseline D2/3 receptor-binding potential using [11C]raclopride (a weak competitive D2/3 receptor antagonist), and dopamine release using [11C]raclopride paired with oral methylphenidate administration. Methylphenidate increases synaptic dopamine by blocking the dopamine transporter. We estimated dopamine release by contrasting baseline D2/3 receptor binding and D2/3 receptor binding following methylphenidate. Analysis of relationships among the three measurements within striatal regions of interest revealed a positive correlation between [18F]FMT Ki and the baseline (placebo) [11C]raclopride measure, such that participants with greater synthesis capacity showed higher D2/3 receptor-binding potential. In contrast, there was no relationship between [18F]FMT and methylphenidate-induced [11C]raclopride displacement. These findings shed light on the nature of regulation between pre- and postsynaptic dopamine function in healthy adults, which may serve as a template from which to identify and describe alteration with disease.
Collapse
|
45
|
Faraone SV. The pharmacology of amphetamine and methylphenidate: Relevance to the neurobiology of attention-deficit/hyperactivity disorder and other psychiatric comorbidities. Neurosci Biobehav Rev 2018; 87:255-270. [PMID: 29428394 DOI: 10.1016/j.neubiorev.2018.02.001] [Citation(s) in RCA: 311] [Impact Index Per Article: 51.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Revised: 01/25/2018] [Accepted: 02/05/2018] [Indexed: 12/20/2022]
Abstract
Psychostimulants, including amphetamines and methylphenidate, are first-line pharmacotherapies for individuals with attention-deficit/hyperactivity disorder (ADHD). This review aims to educate physicians regarding differences in pharmacology and mechanisms of action between amphetamine and methylphenidate, thus enhancing physician understanding of psychostimulants and their use in managing individuals with ADHD who may have comorbid psychiatric conditions. A systematic literature review of PubMed was conducted in April 2017, focusing on cellular- and brain system-level effects of amphetamine and methylphenidate. The primary pharmacologic effect of both amphetamine and methylphenidate is to increase central dopamine and norepinephrine activity, which impacts executive and attentional function. Amphetamine actions include dopamine and norepinephrine transporter inhibition, vesicular monoamine transporter 2 (VMAT-2) inhibition, and monoamine oxidase activity inhibition. Methylphenidate actions include dopamine and norepinephrine transporter inhibition, agonist activity at the serotonin type 1A receptor, and redistribution of the VMAT-2. There is also evidence for interactions with glutamate and opioid systems. Clinical implications of these actions in individuals with ADHD with comorbid depression, anxiety, substance use disorder, and sleep disturbances are discussed.
Collapse
Affiliation(s)
- Stephen V Faraone
- Departments of Psychiatry and of Neuroscience and Physiology, State University of New York (SUNY) Upstate Medical University, Syracuse, NY, United States; K.G. Jebsen Centre for Research on Neuropsychiatric Disorders, University of Bergen, Bergen, Norway.
| |
Collapse
|
46
|
Rubio Morell B, Hernández Expósito S. Differential long-term medication impact on executive function and delay aversion in ADHD. APPLIED NEUROPSYCHOLOGY-CHILD 2017; 8:140-157. [DOI: 10.1080/21622965.2017.1407653] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Affiliation(s)
- Belen Rubio Morell
- Child Psychiatry Unit, Hospital Universitario de Canarias, La Laguna, Spain
| | | |
Collapse
|
47
|
Bartl J, Palazzesi F, Parrinello M, Hommers L, Riederer P, Walitza S, Grünblatt E. The impact of methylphenidate and its enantiomers on dopamine synthesis and metabolism in vitro. Prog Neuropsychopharmacol Biol Psychiatry 2017; 79:281-288. [PMID: 28690202 DOI: 10.1016/j.pnpbp.2017.07.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Revised: 06/02/2017] [Accepted: 07/03/2017] [Indexed: 12/19/2022]
Abstract
Methylphenidate (MPH), a psychostimulant, is an effective first-line treatment for the symptoms associated with Attention-Deficit/Hyperactivity Disorder (ADHD). Although most MPH formulations are composed of the racemic 1:1 mixture of the two enantiomers (d- and l-threo), converging lines of evidence indicate that d-threo MPH seems to be superior to the l-isomer. We aimed to investigate whether MPH racemic mixture or pure enantiomers influence the enzyme activity of tyrosine hydroxylase (TH), monoamine oxidase B (MAO-B), catechol-O-methyltransferase (COMT), and aldehyde dehydrogenase (ALDH) in vitro in homogenates of rat PC12 cells incubated with racemic, d- and l-threo MPH (1nM up to 100μM), or a vehicle for control. We could observe dose dependent enhancement of TH activity with d-threo MPH, probably due to its higher affinity to the enzyme, which we could confirm for d-threo versus l-threo MPH via docking and molecular dynamic simulations analysis. MAO-B enzyme activity was found to be enhanced when incubated with both d- and l-isomers but not with the racemic mixture. This conflicting result was hypothesized to be due to possible aggregation of the two enantiomers or other molecular conformations. Such a possible interaction was observed indirectly, when TH was incubated with constant d-threo MPH while increasing l-isomer (increasing total MPH concentrations). Hence, TH activity was slightly decreased with increased l-isomer. In conclusion, the current in vitro investigation points to the stereoselectivity of the investigated enzymes and pharmacological effects of MPH enantiomers.
Collapse
Affiliation(s)
- Jasmin Bartl
- Department of Child and Adolescent Psychiatry and Psychotherapy, University Hospital of Psychiatry Zurich, University of Zurich, Switzerland
| | - Ferruccio Palazzesi
- Department of Chemistry and Applied Biosciences, Eidgenössische Technische Hochschule (ETH) Zurich, 8093 Zurich, Switzerland; Facoltà di Informatica, Istituto di Scienze Computazionali, Università della Svizzera Italiana, 6900 Lugano, Switzerland
| | - Michele Parrinello
- Department of Chemistry and Applied Biosciences, Eidgenössische Technische Hochschule (ETH) Zurich, 8093 Zurich, Switzerland; Facoltà di Informatica, Istituto di Scienze Computazionali, Università della Svizzera Italiana, 6900 Lugano, Switzerland
| | - Leif Hommers
- Center of Mental Health, Department of Psychiatry, Psychosomatics and Psychotherapy, University Hospital of Wuerzburg, Wuerzburg, Germany
| | - Peter Riederer
- Center of Mental Health, Department of Psychiatry, Psychosomatics and Psychotherapy, University Hospital of Wuerzburg, Wuerzburg, Germany; Unit of Psychiatry, University of Southern Denmark, Odense, Denmark
| | - Susanne Walitza
- Department of Child and Adolescent Psychiatry and Psychotherapy, University Hospital of Psychiatry Zurich, University of Zurich, Switzerland; Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland; Neuroscience Center Zurich, University of Zurich and ETH Zurich, Switzerland
| | - Edna Grünblatt
- Department of Child and Adolescent Psychiatry and Psychotherapy, University Hospital of Psychiatry Zurich, University of Zurich, Switzerland; Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland; Neuroscience Center Zurich, University of Zurich and ETH Zurich, Switzerland.
| |
Collapse
|
48
|
Verbeeck W, Bekkering GE, Van den Noortgate W, Kramers C. Bupropion for attention deficit hyperactivity disorder (ADHD) in adults. Cochrane Database Syst Rev 2017; 10:CD009504. [PMID: 28965364 PMCID: PMC6485546 DOI: 10.1002/14651858.cd009504.pub2] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
BACKGROUND Attention deficit hyperactivity disorder (ADHD) is a prevalent neurobiological condition, characterised by behavioral and cognitive symptoms such as inattention, impulsivity and/or excessive activity. The syndrome is commonly accompanied by psychiatric comorbidities and is associated with educational and occupational underachievement.Although psychostimulant medications are the mainstay of treatment for ADHD, not all adults respond optimally to, or can tolerate, these medicines. Thus, alternative non-stimulant treatment approaches for ADHD have been explored. One of these alternatives is bupropion, an aminoketone antidepressant and non-competitive antagonism of nicotinic acetylcholine receptors. Bupropion is registered for the treatment of depression and smoking cessation, but is also used off-label to treat ADHD. OBJECTIVES To assess the effects and safety of bupropion for the treatment of adults with ADHD. SEARCH METHODS We searched the Cochrane Central Register of Controlled Trials (CENTRAL), MEDLINE, Embase, and seven other databases in February 2017. We also searched three trials registers and three online theses portals. In addition, we checked references of included studies and contacted study authors to identify potentially relevant studies that were missed by our search. SELECTION CRITERIA We included all randomised controlled trials (RCTs) that evaluated the effects (including adverse effects) of bupropion compared to placebo in adults with ADHD. DATA COLLECTION AND ANALYSIS Two review authors (WV, GB) independently screened records and extracted data using a data extraction sheet that we tested in a pilot study. We extracted all relevant data on study characteristics and results. We assessed risks of bias using the Cochrane 'Risk of bias' tool, and assessed the overall quality of evidence using the GRADE approach. We used a fixed-effect model to pool the results across studies. MAIN RESULTS We included six studies with a total of 438 participants. Five studies were conducted in the USA, and one in Iran. All studies evaluated a long-acting version of bupropion, with the dosage ranging from 150 mg up to 450 mg daily. Study intervention length varied from six to 10 weeks. Four studies explicitly excluded participants with psychiatric comorbidity and one study included only participants with opioid dependency. Four studies were funded by industry, but the impact of this on study results is unknown. Two studies were publicly funded and in one of these studies, the lead author was a consultant for several pharmaceutical companies and also received investigator-driven funding from two companies, however none of these companies manufacture bupropion. We judged none of the studies to be free of bias because for most risk of bias domains the study reports failed to provide sufficient details. Using the GRADE approach, we rated the overall quality of evidence as low. We downgraded the quality of the evidence because of serious risk of bias and serious imprecision due to small sample sizes.We found low-quality evidence that bupropion decreased the severity of ADHD symptoms (standardised mean difference -0.50, 95% confidence interval (CI) -0.86 to -0.15, 3 studies, 129 participants), and increased the proportion of participants achieving clinical improvement (risk ratio (RR) 1.50, 95% CI 1.13 to 1.99, 4 studies, 315 participants), and reporting an improvement on the Clinical Global Impression - Improvement scale (RR 1.78, 95% CI 1.27 to 2.50, 5 studies, 337 participants). There was low-quality evidence that the proportion of participants who withdrew due to any adverse effect was similar in the bupropion and placebo groups (RR 1.20, 95% CI 0.35 to 4.10, 3 studies, 253 participants). The results were very similar when using a random-effects model and when we analysed only studies that excluded participants with a psychiatric comorbidity. AUTHORS' CONCLUSIONS The findings of this review, which compared bupropion to placebo for adult ADHD, indicate a possible benefit of bupropion. We found low-quality evidence that bupropion decreased the severity of ADHD symptoms and moderately increased the proportion of participants achieving a significant clinical improvement in ADHD symptoms. Furthermore, we found low-quality evidence that the tolerability of bupropion is similar to that of placebo. In the pharmacological treatment of adults with ADHD, extended- or sustained-release bupropion may be an alternative to stimulants. The low-quality evidence indicates uncertainty with respect to the pooled effect estimates. Further research is very likely to change these estimates. More research is needed to reach more definite conclusions as well as clarifying the optimal target population for this medicine. Treatment response remains to be reported in a DSM5-diagnosed population. There is also a lack of knowledge on long-term outcomes.
Collapse
Affiliation(s)
- Wim Verbeeck
- Centrum ADHD/ASS, GGZ Vincent van Gogh Instituut Venray, Noordsingel 39, Venray, Netherlands, 5801 GJ
| | | | | | | |
Collapse
|
49
|
Abstract
Drugs for psychosis and mood that bind dopamine D2 receptors can be classified not only by whether they also block serotonin 2A receptors, but by whether they also bind D3 or D1 receptors.
Collapse
|
50
|
Swart JC, Froböse MI, Cook JL, Geurts DEM, Frank MJ, Cools R, den Ouden HEM. Catecholaminergic challenge uncovers distinct Pavlovian and instrumental mechanisms of motivated (in)action. eLife 2017; 6:e22169. [PMID: 28504638 PMCID: PMC5432212 DOI: 10.7554/elife.22169] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Accepted: 04/24/2017] [Indexed: 01/06/2023] Open
Abstract
Catecholamines modulate the impact of motivational cues on action. Such motivational biases have been proposed to reflect cue-based, 'Pavlovian' effects. Here, we assess whether motivational biases may also arise from asymmetrical instrumental learning of active and passive responses following reward and punishment outcomes. We present a novel paradigm, allowing us to disentangle the impact of reward and punishment on instrumental learning from Pavlovian response biasing. Computational analyses showed that motivational biases reflect both Pavlovian and instrumental effects: reward and punishment cues promoted generalized (in)action in a Pavlovian manner, whereas outcomes enhanced instrumental (un)learning of chosen actions. These cue- and outcome-based biases were altered independently by the catecholamine enhancer melthylphenidate. Methylphenidate's effect varied across individuals with a putative proxy of baseline dopamine synthesis capacity, working memory span. Our study uncovers two distinct mechanisms by which motivation impacts behaviour, and helps refine current models of catecholaminergic modulation of motivated action.
Collapse
Affiliation(s)
- Jennifer C Swart
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, The Netherlands
| | - Monja I Froböse
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, The Netherlands
| | - Jennifer L Cook
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, The Netherlands
- School of Psychology, University of Birmingham, Birmingham, United Kingdom
| | - Dirk EM Geurts
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, The Netherlands
- Department of Psychiatry, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Michael J Frank
- Department of Cognitive, Linguistic and Psychological Sciences, Brown University, Providence, United States
- Brown Institute for Brain Sciences, Brown University, Providence, United States
| | - Roshan Cools
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, The Netherlands
- Department of Psychiatry, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Hanneke EM den Ouden
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, The Netherlands
| |
Collapse
|