1
|
Parlatini V, Bellato A, Murphy D, Cortese S. From neurons to brain networks, pharmacodynamics of stimulant medication for ADHD. Neurosci Biobehav Rev 2024; 164:105841. [PMID: 39098738 DOI: 10.1016/j.neubiorev.2024.105841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 07/25/2024] [Accepted: 08/01/2024] [Indexed: 08/06/2024]
Abstract
Stimulants represent the first line pharmacological treatment for attention-deficit/hyperactivity disorder (ADHD) and are among the most prescribed psychopharmacological treatments. Their mechanism of action at synaptic level has been extensively studied. However, it is less clear how their mechanism of action determines clinically observed benefits. To help bridge this gap, we provide a comprehensive review of stimulant effects, with an emphasis on nuclear medicine and magnetic resonance imaging (MRI) findings. There is evidence that stimulant-induced modulation of dopamine and norepinephrine neurotransmission optimizes engagement of task-related brain networks, increases perceived saliency, and reduces interference from the default mode network. An acute administration of stimulants may reduce brain alterations observed in untreated individuals in fronto-striato-parieto-cerebellar networks during tasks or at rest. Potential effects of prolonged treatment remain controversial. Overall, neuroimaging has fostered understanding on stimulant mechanism of action. However, studies are often limited by small samples, short or no follow-up, and methodological heterogeneity. Future studies should address age-related and longer-term effects, potential differences among stimulants, and predictors of treatment response.
Collapse
Affiliation(s)
- Valeria Parlatini
- School of Psychology, University of Southampton, Southampton, United Kingdom; Centre for Innovation in Mental Health, University of Southampton, Southampton, United Kingdom; Institute for Life Sciences, University of Southampton, Southampton, United Kingdom; Institute of Translational Neurodevelopment, Department of Forensic and Neurodevelopmental Sciences, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE5 8AF, United Kingdom; Department of Forensic and Neurodevelopmental Sciences, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE5 8AF, United Kingdom; Solent NHS Trust, Southampton, United Kingdom.
| | - Alessio Bellato
- School of Psychology, University of Southampton, Southampton, United Kingdom; Centre for Innovation in Mental Health, University of Southampton, Southampton, United Kingdom; Institute for Life Sciences, University of Southampton, Southampton, United Kingdom; Solent NHS Trust, Southampton, United Kingdom; School of Psychology, University of Nottingham, Semenyih, Malaysia
| | - Declan Murphy
- Institute of Translational Neurodevelopment, Department of Forensic and Neurodevelopmental Sciences, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE5 8AF, United Kingdom; Department of Forensic and Neurodevelopmental Sciences, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE5 8AF, United Kingdom
| | - Samuele Cortese
- School of Psychology, University of Southampton, Southampton, United Kingdom; Centre for Innovation in Mental Health, University of Southampton, Southampton, United Kingdom; Institute for Life Sciences, University of Southampton, Southampton, United Kingdom; Solent NHS Trust, Southampton, United Kingdom; Mind and Neurodevelopment (MiND) Research Group, University of Nottingham, Semenyih, Malaysia; Clinical and Experimental Sciences (CNS and Psychiatry), Faculty of Medicine, University of Southampton, Southampton, United Kingdom; Hassenfeld Children's Hospital at NYU Langone, New York University Child Study Center, New York, NY, USA
| |
Collapse
|
2
|
Akyuz E, Arulsamy A, Aslan FS, Sarisözen B, Guney B, Hekimoglu A, Yilmaz BN, Retinasamy T, Shaikh MF. An Expanded Narrative Review of Neurotransmitters on Alzheimer's Disease: The Role of Therapeutic Interventions on Neurotransmission. Mol Neurobiol 2024:10.1007/s12035-024-04333-y. [PMID: 39012443 DOI: 10.1007/s12035-024-04333-y] [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: 07/06/2023] [Accepted: 06/24/2024] [Indexed: 07/17/2024]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease. The accumulation of amyloid-β (Aβ) plaques and tau neurofibrillary tangles are the key players responsible for the pathogenesis of the disease. The accumulation of Aβ plaques and tau affect the balance in chemical neurotransmitters in the brain. Thus, the current review examined the role of neurotransmitters in the pathogenesis of Alzheimer's disease and discusses the alterations in the neurochemical activity and cross talk with their receptors and transporters. In the presence of Aβ plaques and neurofibrillary tangles, changes may occur in the expression of neuronal receptors which in turn triggers excessive release of glutamate into the synaptic cleft contributing to cell death and neuronal damage. The GABAergic system may also be affected by AD pathology in a similar way. In addition, decreased receptors in the cholinergic system and dysfunction in the dopamine neurotransmission of AD pathology may also contribute to the damage to cognitive function. Moreover, the presence of deficiencies in noradrenergic neurons within the locus coeruleus in AD suggests that noradrenergic stimulation could be useful in addressing its pathophysiology. The regulation of melatonin, known for its effectiveness in enhancing cognitive function and preventing Aβ accumulation, along with the involvement of the serotonergic system and histaminergic system in cognition and memory, becomes remarkable for promoting neurotransmission in AD. Additionally, nitric oxide and adenosine-based therapeutic approaches play a protective role in AD by preventing neuroinflammation. Overall, neurotransmitter-based therapeutic strategies emerge as pivotal for addressing neurotransmitter homeostasis and neurotransmission in the context of AD. This review discussed the potential for neurotransmitter-based drugs to be effective in slowing and correcting the neurodegenerative processes in AD by targeting the neurochemical imbalance in the brain. Therefore, neurotransmitter-based drugs could serve as a future therapeutic strategy to tackle AD.
Collapse
Affiliation(s)
- Enes Akyuz
- Department of Biophysics, International School of Medicine, University of Health Sciences, Istanbul, Turkey
- Department of Pediatrics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Alina Arulsamy
- Neuropharmacology Research Laboratory, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, 47500, Bandar Sunway, Selangor, Malaysia.
| | | | - Bugra Sarisözen
- School of Medicine, Tekirdağ Namık Kemal University, Tekirdağ, Turkey
| | - Beyzanur Guney
- International School of Medicine, University of Health Sciences, Istanbul, Turkey
| | | | - Beyza Nur Yilmaz
- International School of Medicine, University of Health Sciences, Istanbul, Turkey
| | - Thaarvena Retinasamy
- Neuropharmacology Research Laboratory, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, 47500, Bandar Sunway, Selangor, Malaysia
| | - Mohd Farooq Shaikh
- Neuropharmacology Research Laboratory, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, 47500, Bandar Sunway, Selangor, Malaysia.
- School of Dentistry and Medical Sciences, Charles Sturt University, Orange, New South Wales, 2800, Australia.
| |
Collapse
|
3
|
Higuchi T, Chen X, Werner RA. Navigating new horizons: Prospects of NET-targeted radiopharmaceuticals in precision medicine. Theranostics 2024; 14:3178-3192. [PMID: 38855189 PMCID: PMC11155404 DOI: 10.7150/thno.96743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 04/23/2024] [Indexed: 06/11/2024] Open
Abstract
In the evolving landscape of precision medicine, NET-targeted radiopharmaceuticals are emerging as pivotal tools for the diagnosis and treatment of a range of conditions, from heart failure and neurodegenerative disorders to neuroendocrine cancers. This review evaluates the advancements offered by 18F-labeled PET tracers and 211At alpha-particle therapy, juxtaposed with current 123I-MIBG SPECT and 131I-MIBG therapies. The enhanced spatial resolution and capability for quantitative analysis render 18F-labeled PET tracers potential candidates for improved detection and management of diseases. Alpha-particle therapy with 211At may offer increased specificity and tumoricidal efficacy, pointing towards a shift in therapeutic protocols. While preliminary data is promising, these innovative approaches require thorough validation against current modalities. Ongoing clinical trials are pivotal to confirm the expected clinical benefits and to address safety concerns. This review underscores the need for rigorous research to verify the clinical utility of NET-targeted radiopharmaceuticals, which may redefine precision medicine paradigms and significantly impact patient care.
Collapse
Affiliation(s)
- Takahiro Higuchi
- Department of Nuclear Medicine and Comprehensive Heart Failure Center, University Hospital of Würzburg, Würzburg, Germany
- Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Xinyu Chen
- Nuclear Medicine, Faculty of Medicine, University of Augsburg, Augsburg, Germany
| | - Rudolf A Werner
- DZHK (German Centre for Cardiovascular Research), Partner Site Frankfurt Rhine-Main, Frankfurt, Germany
- Goethe University Frankfurt, Department of Nuclear Medicine, Clinic for Radiology and Nuclear Medicine, Frankfurt, Germany
- German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz and German Cancer Research Center (DKFZ), Heidelberg, Germany
- The Russell H Morgan Department of Radiology and Radiological Sciences, Division of Nuclear Medicine and Molecular Imaging, Johns Hopkins School of Medicine, Baltimore, MD, United States
| |
Collapse
|
4
|
Zhao X, Song L, Yang A, Zhang Z, Zhang J, Yang YT, Zhao XM. Prioritizing genes associated with brain disorders by leveraging enhancer-promoter interactions in diverse neural cells and tissues. Genome Med 2023; 15:56. [PMID: 37488639 PMCID: PMC10364416 DOI: 10.1186/s13073-023-01210-6] [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: 02/07/2023] [Accepted: 07/10/2023] [Indexed: 07/26/2023] Open
Abstract
BACKGROUND Prioritizing genes that underlie complex brain disorders poses a considerable challenge. Despite previous studies have found that they shared symptoms and heterogeneity, it remained difficult to systematically identify the risk genes associated with them. METHODS By using the CAGE (Cap Analysis of Gene Expression) read alignment files for 439 human cell and tissue types (including primary cells, tissues and cell lines) from FANTOM5 project, we predicted enhancer-promoter interactions (EPIs) of 439 cell and tissue types in human, and examined their reliability. Then we evaluated the genetic heritability of 17 diverse brain disorders and behavioral-cognitive phenotypes in each neural cell type, brain region, and developmental stage. Furthermore, we prioritized genes associated with brain disorders and phenotypes by leveraging the EPIs in each neural cell and tissue type, and analyzed their pleiotropy and functionality for different categories of disorders and phenotypes. Finally, we characterized the spatiotemporal expression dynamics of these associated genes in cells and tissues. RESULTS We found that identified EPIs showed activity specificity and network aggregation in cell and tissue types, and enriched TF binding in neural cells played key roles in synaptic plasticity and nerve cell development, i.e., EGR1 and SOX family. We also discovered that most neurological disorders exhibit heritability enrichment in neural stem cells and astrocytes, while psychiatric disorders and behavioral-cognitive phenotypes exhibit enrichment in neurons. Furthermore, our identified genes recapitulated well-known risk genes, which exhibited widespread pleiotropy between psychiatric disorders and behavioral-cognitive phenotypes (i.e., FOXP2), and indicated expression specificity in neural cell types, brain regions, and developmental stages associated with disorders and phenotypes. Importantly, we showed the potential associations of brain disorders with brain regions and developmental stages that have not been well studied. CONCLUSIONS Overall, our study characterized the gene-enhancer regulatory networks and genetic mechanisms in the human neural cells and tissues, and illustrated the value of reanalysis of publicly available genomic datasets.
Collapse
Affiliation(s)
- Xingzhong Zhao
- Institute of Science and Technology for Brain-Inspired Intelligence, and Department of Neurology of Zhongshan Hospital, Fudan University, 220 Handan Road, Shanghai, 200433, China
- MOE Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, 200433, China
| | - Liting Song
- Institute of Science and Technology for Brain-Inspired Intelligence, and Department of Neurology of Zhongshan Hospital, Fudan University, 220 Handan Road, Shanghai, 200433, China
- MOE Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, 200433, China
| | - Anyi Yang
- Institute of Science and Technology for Brain-Inspired Intelligence, and Department of Neurology of Zhongshan Hospital, Fudan University, 220 Handan Road, Shanghai, 200433, China
- MOE Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, 200433, China
| | - Zichao Zhang
- Institute of Science and Technology for Brain-Inspired Intelligence, and Department of Neurology of Zhongshan Hospital, Fudan University, 220 Handan Road, Shanghai, 200433, China
- MOE Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, 200433, China
| | - Jinglong Zhang
- Institute of Science and Technology for Brain-Inspired Intelligence, and Department of Neurology of Zhongshan Hospital, Fudan University, 220 Handan Road, Shanghai, 200433, China
- MOE Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, 200433, China
| | - Yucheng T Yang
- Institute of Science and Technology for Brain-Inspired Intelligence, and Department of Neurology of Zhongshan Hospital, Fudan University, 220 Handan Road, Shanghai, 200433, China.
- MOE Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, 200433, China.
| | - Xing-Ming Zhao
- Institute of Science and Technology for Brain-Inspired Intelligence, and Department of Neurology of Zhongshan Hospital, Fudan University, 220 Handan Road, Shanghai, 200433, China.
- MOE Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, 200433, China.
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, Fudan University, Shanghai, 200032, China.
- Internatioal Human Phenome Institutes (Shanghai), Shanghai, 200433, China.
| |
Collapse
|
5
|
Galgani A, Bartolini E, D’Amora M, Faraguna U, Giorgi FS. The Central Noradrenergic System in Neurodevelopmental Disorders: Merging Experimental and Clinical Evidence. Int J Mol Sci 2023; 24:5805. [PMID: 36982879 PMCID: PMC10055776 DOI: 10.3390/ijms24065805] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 03/15/2023] [Accepted: 03/16/2023] [Indexed: 03/30/2023] Open
Abstract
The aim of this article is to highlight the potential role of the locus-coeruleus-noradrenergic (LC-NA) system in neurodevelopmental disorders (NdDs). The LC is the main brain noradrenergic nucleus, key in the regulation of arousal, attention, and stress response, and its early maturation and sensitivity to perinatal damage make it an interesting target for translational research. Clinical data shows the involvement of the LC-NA system in several NdDs, suggesting a pathogenetic role in the development of such disorders. In this context, a new neuroimaging tool, LC Magnetic Resonance Imaging (MRI), has been developed to visualize the LC in vivo and assess its integrity, which could be a valuable tool for exploring morphological alterations in NdD in vivo in humans. New animal models may be used to test the contribution of the LC-NA system to the pathogenic pathways of NdD and to evaluate the efficacy of NA-targeting drugs. In this narrative review, we provide an overview of how the LC-NA system may represent a common pathophysiological and pathogenic mechanism in NdD and a reliable target for symptomatic and disease-modifying drugs. Further research is needed to fully understand the interplay between the LC-NA system and NdD.
Collapse
Affiliation(s)
- Alessandro Galgani
- Department of Translational Research and of New Surgical and Medical Technologies, University of Pisa, 56126 Pisa, Italy; (A.G.)
| | - Emanuele Bartolini
- Department of Developmental Neuroscience, IRCCS Fondazione Stella Maris, 56128 Pisa, Italy
- Tuscany PhD Programme in Neurosciences, 50121 Florence, Italy
| | - Marta D’Amora
- Department of Biology, University of Pisa, 56125 Pisa, Italy
- Istituto Italiano di Tecnologia, 16163 Genova, Italy
| | - Ugo Faraguna
- Department of Translational Research and of New Surgical and Medical Technologies, University of Pisa, 56126 Pisa, Italy; (A.G.)
- Department of Developmental Neuroscience, IRCCS Fondazione Stella Maris, 56128 Pisa, Italy
| | - Filippo Sean Giorgi
- Department of Translational Research and of New Surgical and Medical Technologies, University of Pisa, 56126 Pisa, Italy; (A.G.)
| |
Collapse
|
6
|
Zhang Y, Chen Y, Xin Y, Peng B, Liu S. Norepinephrine system at the interface of attention and reward. Prog Neuropsychopharmacol Biol Psychiatry 2023; 125:110751. [PMID: 36933778 DOI: 10.1016/j.pnpbp.2023.110751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 03/08/2023] [Accepted: 03/14/2023] [Indexed: 03/20/2023]
Abstract
Reward learning is key to survival for individuals. Attention plays an important role in the rapid recognition of reward cues and establishment of reward memories. Reward history reciprocally guides attention to reward stimuli. However, the neurological processes of the interplay between reward and attention remain largely elusive, due to the diversity of the neural substrates that participate in these two processes. In this review, we delineate the complex and differentiated locus coeruleus norepinephrine (LC-NE) system in relation to different behavioral and cognitive substrates of reward and attention. The LC receives reward related sensory, perceptual, and visceral inputs, releases NE, glutamate, dopamine and various neuropeptides, forms reward memories, drives attentional bias and selects behavioral strategies for reward. Preclinical and clinical studies have found that abnormalities in the LC-NE system are involved in a variety of psychiatric conditions marked by disturbed functions in reward and attention. Therefore, we propose that the LC-NE system is an important hub in the interplay between reward and attention as well as a critical therapeutic target for psychiatric disorders characterized by compromised functions in reward and attention.
Collapse
Affiliation(s)
- Yuxiao Zhang
- Shanghai Key Laboratory of Brain Functional Genomics (Ministry of Education), Affiliated Mental Health Center (ECNU), School of Psychology and Cognitive Science, East China Normal University, Shanghai 200062, China; Shanghai Changning Mental Health Center, Shanghai 200335, China; NYU-ECNU Institute of Brain and Cognitive Science at NYU Shanghai, Shanghai 200062, China
| | - Yan Chen
- Shanghai Key Laboratory of Brain Functional Genomics (Ministry of Education), Affiliated Mental Health Center (ECNU), School of Psychology and Cognitive Science, East China Normal University, Shanghai 200062, China; Shanghai Changning Mental Health Center, Shanghai 200335, China; NYU-ECNU Institute of Brain and Cognitive Science at NYU Shanghai, Shanghai 200062, China
| | - Yushi Xin
- Shanghai Key Laboratory of Brain Functional Genomics (Ministry of Education), Affiliated Mental Health Center (ECNU), School of Psychology and Cognitive Science, East China Normal University, Shanghai 200062, China
| | - Beibei Peng
- Shanghai Key Laboratory of Brain Functional Genomics (Ministry of Education), Affiliated Mental Health Center (ECNU), School of Psychology and Cognitive Science, East China Normal University, Shanghai 200062, China; Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, MA, 02478, USA
| | - Shuai Liu
- Shanghai Key Laboratory of Brain Functional Genomics (Ministry of Education), Affiliated Mental Health Center (ECNU), School of Psychology and Cognitive Science, East China Normal University, Shanghai 200062, China; Shanghai Changning Mental Health Center, Shanghai 200335, China; NYU-ECNU Institute of Brain and Cognitive Science at NYU Shanghai, Shanghai 200062, China.
| |
Collapse
|
7
|
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
|
8
|
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
|
9
|
McBride M, Appling C, Ferguson B, Gonzalez A, Schaeffer A, Zand A, Wang D, Sam A, Hart E, Tosh A, Fontcha I, Parmacek S, Beversdorf D. Effects of stimulant medication on divergent and convergent thinking tasks related to creativity in adults with attention-deficit hyperactivity disorder. Psychopharmacology (Berl) 2021; 238:3533-3541. [PMID: 34477886 DOI: 10.1007/s00213-021-05970-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 08/19/2021] [Indexed: 11/29/2022]
Abstract
RATIONALE Common pharmacological treatments for attention-deficit hyperactivity disorder (ADHD) are central nervous system stimulants acting as norepinephrine-dopamine reuptake inhibitors. The noradrenergic and dopaminergic systems have been shown to impact performance on tasks assessing creativity. Some previous studies suggest higher performance on creativity tasks in ADHD. Stimulant medication has been shown to differentially impact creativity in those without ADHD. However, the full range of effects of stimulant medication on creativity in those with ADHD is not known. OBJECTIVES This study examined the effects of stimulants on convergent and divergent tasks associated with creativity in adults with ADHD. METHOD Seventeen adults diagnosed with ADHD who were prescribed stimulant medication attended two counterbalanced sessions: one after taking their prescribed stimulant dose and one after the dose was withheld. Participants completed convergent problem-solving (anagrams, Compound Remote Associates) and divergent generative (letter/semantic fluency, Torrance Test for Creative Thinking (TTCT)-Verbal) tasks. RESULTS There was a significant increase in words generated on the semantic fluency task for the stimulant session. Additionally, significant increases were found in the stimulant session for originality, flexibility, and fluency scores on the TTCT. Stimulant medication did not have an effect on any of the problem-solving tasks. CONCLUSIONS Stimulant medication enhanced verbal fluency in adults with ADHD but had no effect on convergent abilities. Furthermore, stimulants enhanced fluency, flexibility, and originality scores on the TTCT. Therefore, stimulants appear to have positive effects on divergent task performance in adults with ADHD, but not convergent tasks. This finding warrants further studies into the specific roles of norepinephrine and dopamine in this effect.
Collapse
Affiliation(s)
- Molly McBride
- School of Medicine, University of Missouri, Columbia, MO, USA
| | - Carrina Appling
- University of Missouri Interdisciplinary Neuroscience Program, Columbia, MO, USA
| | - Bradley Ferguson
- University of Missouri Interdisciplinary Neuroscience Program, Columbia, MO, USA.,Thompson Center for Autism & Neurodevelopmental Disorders, University of Missouri, Columbia, MO, USA.,Department of Health Psychology, University of Missouri, Columbia, MO, USA
| | | | | | - Amanda Zand
- School of Medicine, University of Missouri, Columbia, MO, USA
| | - David Wang
- School of Medicine, Emory University, Atlanta, GA, USA
| | - Alinna Sam
- School of Medicine, University of Missouri, Columbia, MO, USA
| | - Eric Hart
- Department of Health Psychology, University of Missouri, Columbia, MO, USA
| | - Aneesh Tosh
- Department of Child Health, University of Missouri, Columbia, MO, USA
| | - Ivan Fontcha
- Department of Biological Sciences, University of Missouri, Columbia, MO, USA
| | - Sophia Parmacek
- Department of Psychological Sciences, University of Missouri, Columbia, MO, USA
| | - David Beversdorf
- School of Medicine, University of Missouri, Columbia, MO, USA. .,University of Missouri Interdisciplinary Neuroscience Program, Columbia, MO, USA. .,Thompson Center for Autism & Neurodevelopmental Disorders, University of Missouri, Columbia, MO, USA. .,Department of Health Psychology, University of Missouri, Columbia, MO, USA. .,Departments of Radiology, Neurology, and Psychological Sciences, University of Missouri, Columbia, USA.
| |
Collapse
|
10
|
Neuroimaging in Attention-Deficit/Hyperactivity Disorder: Recent Advances. AJR. AMERICAN JOURNAL OF ROENTGENOLOGY 2021; 218:321-332. [PMID: 34406053 DOI: 10.2214/ajr.21.26316] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Attention-deficit/hyperactivity disorder (ADHD) is a common neurodevelopmental condition, leading to impaired attention and impulsive behaviors diagnosed in, but not limited to, children. ADHD can cause symptoms throughout life. This article summarizes structural (conventional, volumetric, and diffusion tensor imaging MRI) and functional [task-based functional MRI (fMRI), resting state fMRI, PET, and MR spectroscopy] brain findings in patients with ADHD. Consensus is lacking regarding altered anatomic or functional imaging findings of the brain in children with ADHD, likely because of the disorder's heterogeneity. Most anatomic studies report abnormalities in the frontal lobes, basal ganglia, and corpus callosum; decreased surface area in the left ventral frontal and right prefrontal cortex; thinner medial temporal lobes; and smaller caudate nuclei. Using fMRI, researchers have focused on the prefrontal and temporal regions, reflecting perception-action mapping alterations. Artificial intelligence models evaluating brain anatomy have highlighted changes in cortical thickness and shape of the inferior frontal cortex, bilateral sensorimotor cortex, left temporal lobe, and insula. Early intervention and/or normal brain maturation can alter imaging patterns and convert functional imaging studies to a normal pattern. While the imaging findings provide insight into the disease's neuropathophysiology, no definitive structural or functional pattern defines the disorder from a neuroradiologic perspective.
Collapse
|
11
|
Mei X, Wang L, Yang B, Li X. Sex differences in noradrenergic modulation of attention and impulsivity in rats. Psychopharmacology (Berl) 2021; 238:2167-2177. [PMID: 33834255 DOI: 10.1007/s00213-021-05841-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 03/29/2021] [Indexed: 01/01/2023]
Abstract
RATIONALE Noradrenaline (NE) is closely related to attentive performance and impulsive control. However, the potential sex differences regarding attention and impulsivity under the noradrenergic modulation have been largely neglected. Therefore, our study aimed to investigate whether male and female rats exhibit differential responses to NE-related drugs during the five-choice serial reaction time task (5CSRT). METHODS Male and female rats were trained in 5CSRT and administered with different NE drugs after obtaining stable baseline performance: atipamezole, a highly selective α2 receptor antagonist; prazosin, an α1 receptor antagonist; and atomoxetine, a selective NE reuptake inhibitor. Later, prazosin was selected to co-administration with atomoxetine. RESULTS Male and female rats exhibited equal learning speed, and no significant baseline differences were found as measured by the 5CSRT. Atomoxetine decreased premature responses in both sexes, but the extent of this reduction was different, with the reduction greater in males. Besides, atomoxetine (1.8 mg/kg) increased the error of omissions in females. The high dose of prazosin (0.5 mg/kg) decreased the accuracy only in male rats, but this was ameliorated by the co-administration with atomoxetine. CONCLUSIONS Atomoxetine showed significant improvement in impulsivity, but atomoxetine had less beneficial effects on impulsive control in females than in males, and it even impaired attentional performance in female rats. The α1 receptors were mainly responsible for NE drug-related sex differences in attention rather than impulsivity. The results obtained in this study indicate that the sex differences exist in both attention and impulsivity by the modulation of noradrenaline and raise the concern to improve sex-specific treatments.
Collapse
Affiliation(s)
- Xiaolin Mei
- College of Psychology, Capital Normal University, Beijing, 100048, China
- Department of Psychology, Sun Yat-Sen University, Guangzhou, 510275, China
| | - Lutong Wang
- College of Psychology, Capital Normal University, Beijing, 100048, China
| | - Bo Yang
- Department of Psychology, Sun Yat-Sen University, Guangzhou, 510275, China
| | - Xinwang Li
- College of Psychology, Capital Normal University, Beijing, 100048, China.
| |
Collapse
|
12
|
Association of norepinephrine transporter methylation with in vivo NET expression and hyperactivity-impulsivity symptoms in ADHD measured with PET. Mol Psychiatry 2021; 26:1009-1018. [PMID: 31383926 PMCID: PMC7910214 DOI: 10.1038/s41380-019-0461-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Revised: 04/04/2019] [Accepted: 04/11/2019] [Indexed: 01/29/2023]
Abstract
Attention deficit hyperactivity disorder (ADHD) is a common neurodevelopmental disorder with a robust genetic influence. The norepinephrine transporter (NET) is of particular interest as it is one of the main targets in treatment of the disorder. As ADHD is a complex and polygenetic condition, the possible regulation by epigenetic processes has received increased attention. We sought to determine possible differences in NET promoter DNA methylation between patients with ADHD and healthy controls. DNA methylation levels in the promoter region of the NET were determined in 23 adult patients with ADHD and 23 healthy controls. A subgroup of 18 patients with ADHD and 18 healthy controls underwent positron emission tomography (PET) with the radioligand (S,S)-[18F]FMeNER-D2 to quantify the NET in several brain areas in vivo. Analyses revealed significant differences in NET methylation levels at several cytosine-phosphate-guanine (CpG) sites between groups. A defined segment of the NET promoter ("region 1") was hypermethylated in patients in comparison with controls. In ADHD patients, a negative correlation between methylation of a CpG site in this region and NET distribution in the thalamus, locus coeruleus, and the raphe nuclei was detected. Furthermore, methylation of several sites in region 1 was negatively associated with the severity of hyperactivity-impulsivity symptoms. Our results point to an epigenetic dysregulation in ADHD, possibly due to a compensatory mechanisms or additional factors involved in transcriptional processing.
Collapse
|
13
|
Changes of central noradrenaline transporter availability in immunotherapy-naïve multiple sclerosis patients. Sci Rep 2020; 10:14651. [PMID: 32887904 PMCID: PMC7474089 DOI: 10.1038/s41598-020-70732-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 07/17/2020] [Indexed: 02/07/2023] Open
Abstract
The neurotransmitter noradrenaline (NA) mediates arousal, attention and mood, and exerts anti-inflammatory and neuroprotective effects. Alterations of monoamine signalling were reported in multiple sclerosis (MS) and psychiatric illness and may account for the high prevalence of comorbid depression and fatigue in MS patients. We assessed central noradrenaline transporter (NAT) availability using positron emission tomography (PET) and the NAT selective radiotracer S,S-[11C]O-methylreboxetine in immunotherapy-naïve patients with relapsing–remitting MS (RRMS; n = 11) compared to healthy controls (HC; n = 12), and its association to lesion load, time since manifestation, the expanded disability status scale (EDSS), the fatigue scale Würzburger Erschöpfungsinventar bei MS (WEIMuS) and Beck Depression Inventory (BDI). We found NAT availability to be increased in the thalamus, amygdala, putamen and pons/midbrain of MS patients. No relation to clinical or psychometric variables was found. These first data indicate higher NAT availability in subcortical brain regions of immunotherapy-naïve RRMS patients. If these changes of noradrenergic neurotransmission predispose to psychiatric symptoms or associate with disease activity needs to be investigated in longitudinal studies or a larger sample which allows subgroup analyses.
Collapse
|
14
|
Kubota M, Fujino J, Tei S, Takahata K, Matsuoka K, Tagai K, Sano Y, Yamamoto Y, Shimada H, Takado Y, Seki C, Itahashi T, Aoki YY, Ohta H, Hashimoto RI, Zhang MR, Suhara T, Nakamura M, Takahashi H, Kato N, Higuchi M. Binding of Dopamine D1 Receptor and Noradrenaline Transporter in Individuals with Autism Spectrum Disorder: A PET Study. Cereb Cortex 2020; 30:6458-6468. [DOI: 10.1093/cercor/bhaa211] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 06/26/2020] [Accepted: 07/14/2020] [Indexed: 11/13/2022] Open
Abstract
Abstract
Although previous studies have suggested the involvement of dopamine (DA) and noradrenaline (NA) neurotransmissions in the autism spectrum disorder (ASD) pathophysiology, few studies have examined these neurotransmissions in individuals with ASD in vivo. Here, we investigated DA D1 receptor (D1R) and noradrenaline transporter (NAT) binding in adults with ASD (n = 18) and neurotypical controls (n = 20) by utilizing two different PET radioligands, [11C]SCH23390 and (S,S)-[18F]FMeNER-D2, respectively. We found no significant group differences in DA D1R (striatum, anterior cingulate cortex, and temporal cortex) or NAT (thalamus and pons) binding. However, in the ASD group, there were significant negative correlations between DA D1R binding (striatum, anterior cingulate cortex and temporal cortex) and the “attention to detail” subscale score of the Autism Spectrum Quotient. Further, there was a significant positive correlation between DA D1R binding (temporal cortex) and emotion perception ability assessed by the neurocognitive battery. Associations of NAT binding with empathic abilities and executive function were found in controls, but were absent in the ASD group. Although a lack of significant group differences in binding might be partly due to the heterogeneity of ASD, our results indicate that central DA and NA function might play certain roles in the clinical characteristics of ASD.
Collapse
Affiliation(s)
- Manabu Kubota
- Department of Functional Brain Imaging, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Chiba 263-8555, Japan
- Medical Institute of Developmental Disabilities Research, Showa University, Tokyo 157-8577, Japan
- Department of Psychiatry, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan
| | - Junya Fujino
- Medical Institute of Developmental Disabilities Research, Showa University, Tokyo 157-8577, Japan
- Department of Psychiatry and Behavioral Sciences, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo 113-8510, Japan
| | - Shisei Tei
- Medical Institute of Developmental Disabilities Research, Showa University, Tokyo 157-8577, Japan
- Department of Psychiatry, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan
- Institute of Applied Brain Sciences, Waseda University, Saitama 359-1192, Japan
- School of Human and Social Sciences, Tokyo International University, Saitama 350-1198, Japan
| | - Keisuke Takahata
- Department of Functional Brain Imaging, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Chiba 263-8555, Japan
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Kiwamu Matsuoka
- Department of Functional Brain Imaging, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Chiba 263-8555, Japan
| | - Kenji Tagai
- Department of Functional Brain Imaging, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Chiba 263-8555, Japan
| | - Yasunori Sano
- Department of Functional Brain Imaging, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Chiba 263-8555, Japan
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Yasuharu Yamamoto
- Department of Functional Brain Imaging, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Chiba 263-8555, Japan
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Hitoshi Shimada
- Department of Functional Brain Imaging, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Chiba 263-8555, Japan
| | - Yuhei Takado
- Department of Functional Brain Imaging, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Chiba 263-8555, Japan
| | - Chie Seki
- Department of Functional Brain Imaging, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Chiba 263-8555, Japan
| | - Takashi Itahashi
- Medical Institute of Developmental Disabilities Research, Showa University, Tokyo 157-8577, Japan
| | - Yuta Y Aoki
- Medical Institute of Developmental Disabilities Research, Showa University, Tokyo 157-8577, Japan
| | - Haruhisa Ohta
- Medical Institute of Developmental Disabilities Research, Showa University, Tokyo 157-8577, Japan
- Department of Psychiatry, School of Medicine, Showa University, Tokyo 157-8577, Japan
| | - Ryu-ichiro Hashimoto
- Medical Institute of Developmental Disabilities Research, Showa University, Tokyo 157-8577, Japan
- Department of Language Sciences, Graduate School of Humanities, Tokyo Metropolitan University, Tokyo 192-0397, Japan
| | - Ming-Rong Zhang
- Department of Radiopharmaceuticals Development, National Institute of Radiological Sciences, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Chiba 263-8555, Japan
| | - Tetsuya Suhara
- Department of Functional Brain Imaging, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Chiba 263-8555, Japan
| | - Motoaki Nakamura
- Medical Institute of Developmental Disabilities Research, Showa University, Tokyo 157-8577, Japan
- Kanagawa Psychiatric Center, Yokohama, Kanagawa 233-0006, Japan
| | - Hidehiko Takahashi
- Medical Institute of Developmental Disabilities Research, Showa University, Tokyo 157-8577, Japan
- Department of Psychiatry, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan
- Department of Psychiatry and Behavioral Sciences, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo 113-8510, Japan
| | - Nobumasa Kato
- Medical Institute of Developmental Disabilities Research, Showa University, Tokyo 157-8577, Japan
| | - Makoto Higuchi
- Department of Functional Brain Imaging, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Chiba 263-8555, Japan
| |
Collapse
|
15
|
Recent advances in radiotracers targeting norepinephrine transporter: structural development and radiolabeling improvements. J Neural Transm (Vienna) 2020; 127:851-873. [PMID: 32274584 PMCID: PMC7223405 DOI: 10.1007/s00702-020-02180-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Accepted: 03/21/2020] [Indexed: 12/13/2022]
Abstract
The norepinephrine transporter (NET) is a major target for the evaluation of the cardiac sympathetic nerve system in patients with heart failure and Parkinson's disease. It is also used in the therapeutic applications against certain types of neuroendocrine tumors, as exemplified by the clinically used 123/131I-MIBG as theranostic single-photon emission computed tomography (SPECT) agent. With the development of more advanced positron emission tomography (PET) technology, more radiotracers targeting NET have been reported, with superior temporal and spatial resolutions, along with the possibility of functional and kinetic analysis. More recently, fluorine-18-labelled NET tracers have drawn increasing attentions from researchers, due to their longer radiological half-life relative to carbon-11 (110 min vs. 20 min), reduced dependence on on-site cyclotrons, and flexibility in the design of novel tracer structures. In the heart, certain NET tracers provide integral diagnostic information on sympathetic innervation and the nerve status. In the central nervous system, such radiotracers can reveal NET distribution and density in pathological conditions. Most radiotracers targeting cardiac NET-function for the cardiac application consistent of derivatives of either norepinephrine or MIBG with its benzylguanidine core structure, e.g. 11C-HED and 18F-LMI1195. In contrast, all NET tracers used in central nervous system applications are derived from clinically used antidepressants. Lastly, possible applications of NET as selective tracers over organic cation transporters (OCTs) in the kidneys and other organs controlled by sympathetic nervous system will also be discussed.
Collapse
|
16
|
Atomoxetine Does Not Improve Complex Attention in Idiopathic Parkinson's Disease Patients with Cognitive Deficits: A Meta-Analysis. PARKINSONS DISEASE 2020; 2020:4853590. [PMID: 32211146 PMCID: PMC7049416 DOI: 10.1155/2020/4853590] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Accepted: 12/23/2019] [Indexed: 12/11/2022]
Abstract
Objectives To evaluate the effects of atomoxetine on complex attention and other neurocognitive domains in idiopathic Parkinson's disease (PD). Methods Interventional trials reporting changes in complex attention and other neurocognitive functions (Diagnostic and Statistical Manual of Mental Disorders-5) following administration of atomoxetine for at least 8 weeks in adults with idiopathic PD were included. Effect sizes (Cohen's d), the standardized mean difference in the scores of each cognitive domain, were compared using a random-effects model (MetaXL version 5.3). Results Three studies were included in the final analysis. For a change in complex attention in PD with mild cognitive impairment (MCI), the estimated effect size was small and nonsignificant (0.16 (95% CI: −0.09, 0.42), n = 42). For changes in executive function, perceptual-motor function, language, social cognition, and learning and memory, the estimated effect sizes were small and medium, but nonsignificant. A deteriorative trend in executive function was observed after atomoxetine treatment in PD with MCI. For a change in global cognitive function in PD without MCI, the estimated effect size was large and significant. Conclusion In idiopathic PD with MCI, atomoxetine does not improve complex attention. Also, a deteriorative trend in the executive function was noted.
Collapse
|
17
|
Adorjan K, Pogarell O. [What needs to be considered in diagnostics and therapy]. MMW Fortschr Med 2020; 162:42-51. [PMID: 32221872 DOI: 10.1007/s15006-020-0005-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Affiliation(s)
- Kristina Adorjan
- Institut für Psychiatrische Phänomik und Genomik (IPPG), Klinik für Psychiatrie und Psychotherapie, Klinikum der Universität München (LMU), Nussbaumstr. 7, D-80336, München, Deutschland.
| | - Oliver Pogarell
- Klinik für Psychiatrie und Psychotherapie, Klinikum der Universität München (LMU), Deutschland
| |
Collapse
|
18
|
Wanke N, Müller JC, Wiedemann K, Schwabe L. (Lack of) Effects of noradrenergic stimulation on human working memory performance. Psychopharmacology (Berl) 2020; 237:3033-3046. [PMID: 32583013 PMCID: PMC7524848 DOI: 10.1007/s00213-020-05590-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 06/11/2020] [Indexed: 11/29/2022]
Abstract
RATIONALE Working memory depends on prefrontal cortex functioning, which is particularly sensitive to levels of noradrenaline. Studies in non-human primates have shown that modest levels of noradrenaline improve working memory, and that higher levels of noradrenaline impair working memory performance. However, research in humans provided inconsistent findings concerning noradrenergic effects on working memory. OBJECTIVE The present study aimed at assessing dose-dependent effects of yohimbine, an alpha-2 adrenoceptor antagonist, on working memory performance in healthy humans. We further aimed to explore a potential interactive effect between noradrenergic arousal and lack of control over aversive events on working memory performance. METHODS We used a double-blind, fully crossed, placebo-controlled, between-subject design. Participants (N = 121) performed an adaptive n-back task before and after oral administration of either a placebo, 20 mg, or 40 mg yohimbine and a manipulation of controllability, during which participants could either learn to avoid electric shocks (controllability groups), had no instrumental control over shock administration (uncontrollability groups), or did not receive any shocks (no-shock control group). RESULTS While no significant results of noradrenergic stimulation through yohimbine were obtained using conventional frequentist analyses, additional Bayesian analyses provided strong evidence for the absence of an association between pharmacological treatment and working memory performance. We further observed no effect of controllability and no interaction between noradrenergic stimulation and the manipulation of controllability. CONCLUSIONS Our results suggest that noradrenergic stimulation through yohimbine does not affect (non-spatial) working memory in healthy human participants.
Collapse
Affiliation(s)
- Nadine Wanke
- Department of Cognitive Psychology, University of Hamburg, 20146 Hamburg, Germany
| | | | - Klaus Wiedemann
- Department of Psychiatry, University Clinic Hamburg-Eppendorf, Hamburg, Germany
| | - Lars Schwabe
- Department of Cognitive Psychology, University of Hamburg, 20146, Hamburg, Germany.
| |
Collapse
|
19
|
Ulke C, Rullmann M, Huang J, Luthardt J, Becker GA, Patt M, Meyer PM, Tiepolt S, Hesse S, Sabri O, Strauß M. Adult attention-deficit/hyperactivity disorder is associated with reduced norepinephrine transporter availability in right attention networks: a (S,S)-O-[ 11C]methylreboxetine positron emission tomography study. Transl Psychiatry 2019; 9:301. [PMID: 31732713 PMCID: PMC6858438 DOI: 10.1038/s41398-019-0619-y] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2019] [Revised: 09/12/2019] [Accepted: 10/03/2019] [Indexed: 12/22/2022] Open
Abstract
The norepinephrine transporter (NET) has been suggested to play a critical role in attention-deficit/hyperactivity disorder (ADHD). In this prospective controlled study we tested the a-priori-hypothesis that central NET availability is altered in adult ADHD patients compared to healthy controls. Study participants underwent single positron emission tomography-magnetic resonance imaging (PET-MRI). MRI sequences included high resolution T1-MPRAGE data for regions of interest (ROI) delineation and voxel-based morphometry (VBM) and T2-weighted fluid-attenuated inversion-recovery for detection and exclusion of pathological abnormalities. NET availability was assessed by NET-selective (S,S)-O-[11C]methylreboxetine; regional distribution volume ratios (DVR) were calculated based on individual PET-MRI data co-registration and a multi-linear reference tissue model with two constraints (MRTM2; reference region: occipital cortex). VBM analysis revealed no difference in local distribution of gray matter between the 20 ADHD patients (9 females, age 31.8 ± 7.9 years, 488 ± 8 MBq injected activity) and the 20 age-matched and sex-matched control participants (9 females, age 32.3 ± 7.9 years, 472 ± 72 MBq). In mixed-model repeated-measures analysis with NET availability as dependent and ROI as repeated measure we found a significant main effect group in fronto-parietal-thalamic-cerebellar regions (regions on the right: F1,25 = 12.30, p = .002; regions on the left: F1,41 = 6.80, p = .013) indicating a reduced NET availability in ADHD patients. None of the other investigated brain regions yielded significant differences in NET availability between groups after applying a Benjamini-Hochberg correction at a significance level of 0.05. Overall our findings demonstrate the pathophysiological involvement of NET availability in adult ADHD.
Collapse
Affiliation(s)
- Christine Ulke
- Department of Psychiatry and Psychotherapy, University of Leipzig Medical Center, 04103, Leipzig, Germany.
| | - Michael Rullmann
- Department of Nuclear Medicine, University of Leipzig Medical Center, 04103, Leipzig, Germany
| | - Jue Huang
- Department of Psychiatry and Psychotherapy, University of Leipzig Medical Center, 04103, Leipzig, Germany
| | - Julia Luthardt
- Department of Nuclear Medicine, University of Leipzig Medical Center, 04103, Leipzig, Germany
| | - Georg-Alexander Becker
- Department of Nuclear Medicine, University of Leipzig Medical Center, 04103, Leipzig, Germany
| | - Marianne Patt
- Department of Nuclear Medicine, University of Leipzig Medical Center, 04103, Leipzig, Germany
| | - Philipp M Meyer
- Department of Nuclear Medicine, University of Leipzig Medical Center, 04103, Leipzig, Germany
| | - Solveig Tiepolt
- Department of Nuclear Medicine, University of Leipzig Medical Center, 04103, Leipzig, Germany
| | - Swen Hesse
- Department of Nuclear Medicine, University of Leipzig Medical Center, 04103, Leipzig, Germany
| | - Osama Sabri
- Department of Nuclear Medicine, University of Leipzig Medical Center, 04103, Leipzig, Germany
| | - Maria Strauß
- Department of Psychiatry and Psychotherapy, University of Leipzig Medical Center, 04103, Leipzig, Germany
| |
Collapse
|
20
|
Dyer-Reaves K, Goodman AM, Nelson AR, McMahon LL. Alpha1-Adrenergic Receptor Mediated Long-Term Depression at CA3-CA1 Synapses Can Be Induced via Accumulation of Endogenous Norepinephrine and Is Preserved Following Noradrenergic Denervation. Front Synaptic Neurosci 2019; 11:27. [PMID: 31649525 PMCID: PMC6794465 DOI: 10.3389/fnsyn.2019.00027] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2019] [Accepted: 09/20/2019] [Indexed: 12/27/2022] Open
Abstract
Locus coeruleus (LC) provides the sole source of noradrenergic (NA) innervation to hippocampus, and it undergoes significant degeneration early in Alzheimer's disease (AD). Norepinephrine (NE) modulates synaptic transmission and plasticity at hippocampal synapses which likely contributes to hippocampus-dependent learning and memory. We previously reported that pharmacological activation of α1 adrenergic receptors (α1ARs) induces long-term depression (LTD) at CA3-CA1 synapses. Here, we investigated whether accumulation of endogenous NE via pharmacological blockade of norepinephrine transporters (NETs) and the NE degradative enzyme, monoamine oxidase (MAO), can induce α1AR LTD, as these inhibitors are used clinically. Further, we sought to determine how degeneration of hippocampal NA innervation, as occurs in AD, impacts α1AR function and α1AR LTD. Bath application of NET and MAO inhibitors in slices from control rats reliably induced α1AR LTD when β adrenergic receptors were inhibited. To induce degeneration of LC-NA innervation, rats were treated with the specific NA neurotoxin DSP-4 and recordings performed 1-3 weeks later when NA axon degeneration had stabilized. Even with 85% loss of hippocampal NA innervation, α1AR LTD was successfully induced using either the α1AR agonist phenylephrine or the combined NET and MAO inhibitors, and importantly, the LTD magnitude was not different from saline-treated control. These data suggest that despite significant decreases in NA input to hippocampus, the mechanisms necessary for the induction of α1AR LTD remain functional. Furthermore, we posit that α1AR activation could be a viable therapeutic target for pharmacological intervention in AD and other diseases involving malfunctions of NA neurotransmission.
Collapse
Affiliation(s)
- Katie Dyer-Reaves
- Department of Cell, Developmental, and Integrative Biology (CDIB), School of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Anthoni M. Goodman
- Department of Cell, Developmental, and Integrative Biology (CDIB), School of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
- Department of Psychology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Amy R. Nelson
- Department of Cell, Developmental, and Integrative Biology (CDIB), School of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Lori L. McMahon
- Department of Cell, Developmental, and Integrative Biology (CDIB), School of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
21
|
Adorjan K, Karch S, Martin G, Plörer D, Winter C, Hantschk I, Koller G, Erbas B, Pogarell O. [Adult attention deficit hyperactivity disorder (ADHD) with comorbid addictive disorder. Therapeutic challenges using the example of a survey of clinical practice]. MMW Fortschr Med 2019; 161:7-12. [PMID: 31313269 DOI: 10.1007/s15006-019-0740-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Accepted: 05/06/2019] [Indexed: 06/10/2023]
Abstract
BACKGROUND ADHD in childhood and adolescence is characterized by the symptoms hyperactivity, impulsivity, and inattentiveness; these symptoms may persist into adulthood or may manifest as restlessness, emotional instability, and disorganized behavior. In adults ADHD often occurs with increased substance use and is associated with an early onset of substance use, development of severe addiction, and decreased treatment effectiveness. METHOD This overview will present and critically discuss current study results and evidence-based and consensus-oriented recommendations that ensure the most adequate care for patients with ADHD and addictive disorder. RESULTS AND CONCLUSIONS For drug therapy, the current S3 guideline recommends methylphenidate, amphetamine salts, and atomoxetine, among others. Treatment of adult patients with ADHD and addiction with stimulants tends to be viewed critically; if required, long-acting medications should be used. Integrated treatment of ADHD and addiction, consisting of a combination of pharmacotherapy and psychotherapy, is recommended.
Collapse
Affiliation(s)
- Kristina Adorjan
- Klinik für Psychiatrie und Psychotherapie, Klinikum der Universität München (LMU), Nußbaumstraße 7, 80336, München, Deutschland.
- Institut für Psychiatrische Phänomik und Genomik (IPPG), Klinikum der Universität München (LMU), München, Deutschland.
| | - Susanne Karch
- Klinik für Psychiatrie und Psychotherapie, Klinikum der Universität München (LMU), Nußbaumstraße 7, 80336, München, Deutschland
| | - Gabi Martin
- Klinik für Psychiatrie und Psychotherapie, Klinikum der Universität München (LMU), Nußbaumstraße 7, 80336, München, Deutschland
| | - Diana Plörer
- Klinik für Psychiatrie und Psychotherapie, Klinikum der Universität München (LMU), Nußbaumstraße 7, 80336, München, Deutschland
| | - Catja Winter
- Klinik für Psychiatrie und Psychotherapie, Klinikum der Universität München (LMU), Nußbaumstraße 7, 80336, München, Deutschland
| | - Irmgard Hantschk
- Klinik für Psychiatrie und Psychotherapie, Klinikum der Universität München (LMU), Nußbaumstraße 7, 80336, München, Deutschland
| | - Gabi Koller
- Klinik für Psychiatrie und Psychotherapie, Klinikum der Universität München (LMU), Nußbaumstraße 7, 80336, München, Deutschland
| | - Beate Erbas
- Bayerische Akademie für Sucht- und Gesundheitsfragen (BAS) München, München, Deutschland
| | - Oliver Pogarell
- Klinik für Psychiatrie und Psychotherapie, Klinikum der Universität München (LMU), Nußbaumstraße 7, 80336, München, Deutschland
- Bayerische Akademie für Sucht- und Gesundheitsfragen (BAS) München, München, Deutschland
| |
Collapse
|
22
|
G. Béroule D. Autism-modifying therapy based on the promotion of a brain enzyme: An introductory case-report. AIMS MOLECULAR SCIENCE 2019. [DOI: 10.3934/molsci.2019.3.52] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
23
|
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
|
24
|
Wang P, Fu T, Zhang X, Yang F, Zheng G, Xue W, Chen Y, Yao X, Zhu F. Differentiating physicochemical properties between NDRIs and sNRIs clinically important for the treatment of ADHD. Biochim Biophys Acta Gen Subj 2017; 1861:2766-2777. [PMID: 28757337 DOI: 10.1016/j.bbagen.2017.07.022] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Revised: 07/23/2017] [Accepted: 07/26/2017] [Indexed: 12/25/2022]
Abstract
BACKGROUND Drugs available for treating attention-deficit hyperactivity disorder (ADHD) are mainly selective norepinephrine (sNRIs) and dual norepinephrine-dopamine (NDRIs) reuptake inhibitors. The major problem of sNRIs lines in their delayed onset of action and partial- or non-responses, which makes NDRIs distinguished in drug efficacy. Understanding of the differential binding modes of these 2 types of drugs to their corresponding targets can give great insights into the discovery of privileged drug-like scaffolds with improved efficacy. So far, no such study has been carried out. METHODS A combinatorial computational strategy, integrating homology modeling, molecular docking, molecular dynamics (MD) and binding free energy calculation, was employed to analyze the binding modes of 8 clinically important ADHD drugs in their targets. RESULTS Binding modes of 2 types of ADHD drugs (sNRIs and NDRIs) in their targets was identified for the first time by MD simulation, and 15 hot spot residues were discovered as crucial for NDRIs' binding in hNET and hDAT. Comparing to sNRIs, a clear reduction in the hydrophobic property of NDRIs' one functional group was observed, and the depth of drugs' aromatic ring stretched into the pocket of both targets was further identified as key contributors to drugs' selectivity. CONCLUSIONS The hydrophobic property of NDRI ADHD drugs' one functional group contributes to their selectivity when bind hNET and hDAT. GENERAL SIGNIFICANCE These results provide insights into NDRI ADHD drugs' binding mechanisms, which could be utilized as structural blueprints for assessing and discovering more efficacious drugs for ADHD therapy.
Collapse
Affiliation(s)
- Panpan Wang
- Innovative Drug Research and Bioinformatics Group, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; College of Chemistry and Pharmaceutical Engineering, Huanghuai University, Zhumadian 463000, China; School of Pharmaceutical Sciences and Collaborative Innovation Center for Brain Science, Chongqing University, Chongqing 401331, China
| | - Tingting Fu
- Innovative Drug Research and Bioinformatics Group, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; School of Pharmaceutical Sciences and Collaborative Innovation Center for Brain Science, Chongqing University, Chongqing 401331, China
| | - Xiaoyu Zhang
- Innovative Drug Research and Bioinformatics Group, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; School of Pharmaceutical Sciences and Collaborative Innovation Center for Brain Science, Chongqing University, Chongqing 401331, China
| | - Fengyuan Yang
- Innovative Drug Research and Bioinformatics Group, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; School of Pharmaceutical Sciences and Collaborative Innovation Center for Brain Science, Chongqing University, Chongqing 401331, China
| | - Guoxun Zheng
- Innovative Drug Research and Bioinformatics Group, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; School of Pharmaceutical Sciences and Collaborative Innovation Center for Brain Science, Chongqing University, Chongqing 401331, China
| | - Weiwei Xue
- School of Pharmaceutical Sciences and Collaborative Innovation Center for Brain Science, Chongqing University, Chongqing 401331, China.
| | - Yuzong Chen
- Bioinformatics and Drug Design Group, Department of Pharmacy, National University of Singapore, Singapore 117543, Singapore
| | - Xiaojun Yao
- State Key Laboratory of Applied Organic Chemistry, Department of Chemistry, Lanzhou University, Lanzhou 730000, China
| | - Feng Zhu
- Innovative Drug Research and Bioinformatics Group, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; School of Pharmaceutical Sciences and Collaborative Innovation Center for Brain Science, Chongqing University, Chongqing 401331, China.
| |
Collapse
|
25
|
Kautzky A, James GM, Philippe C, Baldinger-Melich P, Kraus C, Kranz GS, Vanicek T, Gryglewski G, Wadsak W, Mitterhauser M, Rujescu D, Kasper S, Lanzenberger R. The influence of the rs6295 gene polymorphism on serotonin-1A receptor distribution investigated with PET in patients with major depression applying machine learning. Transl Psychiatry 2017; 7:e1150. [PMID: 28608854 PMCID: PMC5537636 DOI: 10.1038/tp.2017.108] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2016] [Revised: 04/08/2017] [Accepted: 04/20/2017] [Indexed: 12/25/2022] Open
Abstract
Major depressive disorder (MDD) is the most common neuropsychiatric disease and despite extensive research, its genetic substrate is still not sufficiently understood. The common polymorphism rs6295 of the serotonin-1A receptor gene (HTR1A) is affecting the transcriptional regulation of the 5-HT1A receptor and has been closely linked to MDD. Here, we used positron emission tomography (PET) exploiting advances in data mining and statistics by using machine learning in 62 healthy subjects and 19 patients with MDD, which were scanned with PET using the radioligand [carbonyl-11C]WAY-100635. All the subjects were genotyped for rs6295 and genotype was grouped in GG vs C allele carriers. Mixed model was applied in a ROI-based (region of interest) approach. ROI binding potential (BPND) was divided by dorsal raphe BPND as a specific measure to highlight rs6295 effects (BPDiv). Mixed model produced an interaction effect of ROI and genotype in the patients' group but no effects in healthy controls. Differences of BPDiv was demonstrated in seven ROIs; parahippocampus, hippocampus, fusiform gyrus, gyrus rectus, supplementary motor area, inferior frontal occipital gyrus and lingual gyrus. For classification of genotype, 'RandomForest' and Support Vector Machines were used, however, no model with sufficient predictive capability could be computed. Our results are in line with preclinical data, mouse model knockout studies as well as previous clinical analyses, demonstrating the two-pronged effect of the G allele on 5-HT1A BPND for, we believe, the first time. Future endeavors should address epigenetic effects and allosteric heteroreceptor complexes. Replication in larger samples of MDD patients is necessary to substantiate our findings.
Collapse
Affiliation(s)
- A Kautzky
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria
| | - G M James
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria
| | - C Philippe
- Department of Biomedical Imaging and Image-Guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Vienna, Austria
| | - P Baldinger-Melich
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria
| | - C Kraus
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria
| | - G S Kranz
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria
| | - T Vanicek
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria
| | - G Gryglewski
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria
| | - W Wadsak
- Department of Biomedical Imaging and Image-Guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Vienna, Austria,Center for Biomarker Research in Medicine (CBmed), Graz, Austria
| | - M Mitterhauser
- Department of Biomedical Imaging and Image-Guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Vienna, Austria,Ludwig Boltzmann Institute for Applied Diagnostics, Vienna, Austria
| | - D Rujescu
- University Clinic for Psychiatry, Psychotherapy and Psychosomatic, Martin-Luther-University Halle-Wittenberg, Halle, Germany
| | - S Kasper
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria
| | - R Lanzenberger
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria,Department of Psychiatry and Psychotherapy, Medical University of Vienna, Währinger Gürtel 18-20, Vienna A-1090, Austria. E-mail:
| |
Collapse
|
26
|
Noradrenergic Modulation of Cognition in Health and Disease. Neural Plast 2017; 2017:6031478. [PMID: 28596922 PMCID: PMC5450174 DOI: 10.1155/2017/6031478] [Citation(s) in RCA: 136] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Accepted: 04/18/2017] [Indexed: 12/15/2022] Open
Abstract
Norepinephrine released by the locus coeruleus modulates cellular processes and synaptic transmission in the central nervous system through its actions at a number of pre- and postsynaptic receptors. This transmitter system facilitates sensory signal detection and promotes waking and arousal, processes which are necessary for navigating a complex and dynamic sensory environment. In addition to its effects on sensory processing and waking behavior, norepinephrine is now recognized as a contributor to various aspects of cognition, including attention, behavioral flexibility, working memory, and long-term mnemonic processes. Two areas of dense noradrenergic innervation, the prefrontal cortex and the hippocampus, are particularly important with regard to these functions. Due to its role in mediating normal cognitive function, it is reasonable to expect that noradrenergic transmission becomes dysfunctional in a number of neuropsychiatric and neurodegenerative diseases characterized by cognitive deficits. In this review, we summarize the unique role that norepinephrine plays in prefrontal cortical and hippocampal function and how its interaction with its various receptors contribute to cognitive behaviors. We further assess the changes that occur in the noradrenergic system in Alzheimer's disease, Parkinson's disease, attention-deficit/hyperactivity disorder, and schizophrenia and how these changes contribute to cognitive decline in these pathologies.
Collapse
|
27
|
James GM, Baldinger-Melich P, Philippe C, Kranz GS, Vanicek T, Hahn A, Gryglewski G, Hienert M, Spies M, Traub-Weidinger T, Mitterhauser M, Wadsak W, Hacker M, Kasper S, Lanzenberger R. Effects of Selective Serotonin Reuptake Inhibitors on Interregional Relation of Serotonin Transporter Availability in Major Depression. Front Hum Neurosci 2017; 11:48. [PMID: 28220069 PMCID: PMC5292566 DOI: 10.3389/fnhum.2017.00048] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Accepted: 01/23/2017] [Indexed: 11/13/2022] Open
Abstract
Selective serotonin reuptake inhibitors (SSRIs) modulate serotonergic neurotransmission by blocking reuptake of serotonin from the extracellular space. Up to now, it remains unclear how SSRIs achieve their antidepressant effect. However, task-based and resting state functional magnetic resonance imaging studies, have demonstrated connectivity changes between brain regions. Here, we use positron emission tomography (PET) to quantify SSRI's main target, the serotonin transporter (SERT), and assess treatment-induced molecular changes in the interregional relation of SERT binding potential (BPND). Nineteen out-patients with major depressive disorder (MDD) and 19 healthy controls (HC) were included in this study. Patients underwent three PET measurements with the radioligand [11C]DASB: (1) at baseline, (2) after a first SSRI dose; and (3) following at least 3 weeks of daily intake. Controls were measured once with PET. Correlation analyses were restricted to brain regions repeatedly implicated in MDD pathophysiology. After 3 weeks of daily SSRI administration a significant increase in SERT BPND correlations of anterior cingulate cortex and insula with the amygdala, midbrain, hippocampus, pallidum and putamen (p < 0.05; false discovery rate, FDR corrected) was revealed. No significant differences were found when comparing MDD patients and HC at baseline. These findings are in line with the clinical observation that treatment response to SSRIs is often achieved only after a latency of several weeks. The elevated associations in interregional SERT associations may be more closely connected to clinical outcomes than regional SERT occupancy measures and could reflect a change in the regional interaction of serotonergic neurotransmission during antidepressant treatment.
Collapse
Affiliation(s)
- Gregory M James
- Department of Psychiatry and Psychotherapy, Medical University of Vienna Vienna, Austria
| | - Pia Baldinger-Melich
- Department of Psychiatry and Psychotherapy, Medical University of Vienna Vienna, Austria
| | - Cecile Philippe
- Department of Biomedical Imaging and Image-Guided Therapy, Division of Nuclear Medicine, Medical University of Vienna Vienna, Austria
| | - Georg S Kranz
- Department of Psychiatry and Psychotherapy, Medical University of Vienna Vienna, Austria
| | - Thomas Vanicek
- Department of Psychiatry and Psychotherapy, Medical University of Vienna Vienna, Austria
| | - Andreas Hahn
- Department of Psychiatry and Psychotherapy, Medical University of Vienna Vienna, Austria
| | - Gregor Gryglewski
- Department of Psychiatry and Psychotherapy, Medical University of Vienna Vienna, Austria
| | - Marius Hienert
- Department of Psychiatry and Psychotherapy, Medical University of Vienna Vienna, Austria
| | - Marie Spies
- Department of Psychiatry and Psychotherapy, Medical University of Vienna Vienna, Austria
| | - Tatjana Traub-Weidinger
- Department of Biomedical Imaging and Image-Guided Therapy, Division of Nuclear Medicine, Medical University of Vienna Vienna, Austria
| | - Markus Mitterhauser
- Department of Biomedical Imaging and Image-Guided Therapy, Division of Nuclear Medicine, Medical University of Vienna Vienna, Austria
| | - Wolfgang Wadsak
- Department of Biomedical Imaging and Image-Guided Therapy, Division of Nuclear Medicine, Medical University of Vienna Vienna, Austria
| | - Marcus Hacker
- Department of Biomedical Imaging and Image-Guided Therapy, Division of Nuclear Medicine, Medical University of Vienna Vienna, Austria
| | - Siegfried Kasper
- Department of Psychiatry and Psychotherapy, Medical University of Vienna Vienna, Austria
| | - Rupert Lanzenberger
- Department of Psychiatry and Psychotherapy, Medical University of Vienna Vienna, Austria
| |
Collapse
|
28
|
Central noradrenaline transporter availability in highly obese, non-depressed individuals. Eur J Nucl Med Mol Imaging 2017; 44:1056-1064. [PMID: 28066877 DOI: 10.1007/s00259-016-3590-3] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Accepted: 11/28/2016] [Indexed: 10/20/2022]
Abstract
PURPOSE The brain noradrenaline (NA) system plays an important role in the central nervous control of energy balance and is thus implicated in the pathogenesis of obesity. The specific processes modulated by this neurotransmitter which lead to obesity and overeating are still a matter of debate. METHODS We tested the hypothesis that in vivo NA transporter (NAT) availability is changed in obesity by using positron emission tomography (PET) and S,S-[11C]O-methylreboxetine (MRB) in twenty subjects comprising ten highly obese (body mass index BMI > 35 kg/m2), metabolically healthy, non-depressed individuals and ten non-obese (BMI < 30 kg/m2) healthy controls. RESULTS Overall, we found no significant differences in binding potential (BPND) values between obese and non-obese individuals in the investigated brain regions, including the NAT-rich thalamus (0.40 ± 0.14 vs. 0.41 ± 0.18; p = 0.84) though additional discriminant analysis correctly identified individual group affiliation based on regional BPND in all but one (control) case. Furthermore, inter-regional correlation analyses indicated different BPND patterns between both groups but this did not survive testing for multiple comparions. CONCLUSIONS Our data do not find an overall involvement of NAT changes in human obesity. However, preliminary secondary findings of distinct regional and associative patterns warrant further investigation.
Collapse
|
29
|
Moriguchi S, Kimura Y, Ichise M, Arakawa R, Takano H, Seki C, Ikoma Y, Takahata K, Nagashima T, Yamada M, Mimura M, Suhara T. PET Quantification of the Norepinephrine Transporter in Human Brain with (S,S)-18F-FMeNER-D2. J Nucl Med 2016; 58:1140-1145. [DOI: 10.2967/jnumed.116.178913] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Accepted: 12/05/2016] [Indexed: 11/16/2022] Open
|
30
|
Vanicek T, Kutzelnigg A, Philippe C, Sigurdardottir HL, James GM, Hahn A, Kranz GS, Höflich A, Kautzky A, Traub-Weidinger T, Hacker M, Wadsak W, Mitterhauser M, Kasper S, Lanzenberger R. Altered interregional molecular associations of the serotonin transporter in attention deficit/hyperactivity disorder assessed with PET. Hum Brain Mapp 2016; 38:792-802. [PMID: 27770470 DOI: 10.1002/hbm.23418] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Revised: 08/17/2016] [Accepted: 09/20/2016] [Indexed: 02/05/2023] Open
Abstract
Altered serotonergic neurotransmission has been found to cause impulsive and aggressive behavior, as well as increased motor activity, all exemplifying key symptoms of ADHD. The main objectives of this positron emission tomography (PET) study were to investigate the serotonin transporter binding potential (SERT BPND ) in patients with ADHD and to assess associations of SERT BPND between the brain regions. 25 medication-free patients with ADHD (age ± SD; 32.39 ± 10.15; 10 females) without any psychiatric comorbidity and 25 age and sex matched healthy control subjects (33.74 ± 10.20) were measured once with PET and the highly selective and specific radioligand [11 C]DASB. SERT BPND maps in nine a priori defined ROIs exhibiting high SERT binding were compared between groups by means of a linear mixed model. Finally, adopted from structural and functional connectivity analyses, we performed correlational analyses using regional SERT binding potentials to examine molecular interregional associations between all selected ROIs. We observed significant differences in the interregional correlations between the precuneus and the hippocampus in patients with ADHD compared to healthy controls, using SERT BPND of the investigated ROIs (P < 0.05; Bonferroni corrected). When correlating SERT BPND and age in the ADHD and the healthy control group, we confirmed an age-related decline in brain SERT binding in the thalamus and insula (R2 = 0.284, R2 = 0.167, Ps < 0.05; Bonferroni corrected). The results show significantly different interregional molecular associations of the SERT expression for the precuneus with hippocampus in patients with ADHD, indicating presumably altered functional coupling. Altered interregional coupling between brain regions might be a sensitive approach to demonstrate functional and molecular alterations in psychiatric conditions. Hum Brain Mapp 38:792-802, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Thomas Vanicek
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Austria
| | - Alexandra Kutzelnigg
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Austria
| | - Cecile Philippe
- Division of Nuclear Medicine, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Austria
| | | | - Gregory M James
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Austria
| | - Andreas Hahn
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Austria
| | - Georg S Kranz
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Austria
| | - Anna Höflich
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Austria
| | - Alexander Kautzky
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Austria
| | - Tatjana Traub-Weidinger
- Division of Nuclear Medicine, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Austria
| | - Marcus Hacker
- Division of Nuclear Medicine, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Austria
| | - Wolfgang Wadsak
- Division of Nuclear Medicine, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Austria
| | - Markus Mitterhauser
- Division of Nuclear Medicine, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Austria
| | - Siegfried Kasper
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Austria
| | - Rupert Lanzenberger
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Austria
| |
Collapse
|
31
|
Methylphenidate does not enhance visual working memory but benefits motivation in macaque monkeys. Neuropharmacology 2016; 109:223-235. [DOI: 10.1016/j.neuropharm.2016.06.019] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Revised: 06/14/2016] [Accepted: 06/17/2016] [Indexed: 02/04/2023]
|
32
|
[(18)F]FMeNER-D2: A systematic in vitro analysis of radio-metabolism. Nucl Med Biol 2016; 43:490-5. [PMID: 27236284 DOI: 10.1016/j.nucmedbio.2016.05.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Revised: 05/06/2016] [Accepted: 05/10/2016] [Indexed: 11/23/2022]
Abstract
INTRODUCTION The norepinephrine transporter (NET) presents an important target for therapy and diagnosis of ADHD and other neurodegenerative and psychiatric diseases. Thus, PET is the diagnostic method of choice, using radiolabeled NET-ligands derived from reboxetine. So far, [(18)F]FMeNER-D2 showed best pharmacokinetic and -dynamic properties. However, the disadvantage of reboxetine derived PET tracers is their high metabolic cleavage-resulting in impeding signals in the PET scans, which hamper a proper quantification of the NET in cortical areas. METHODS Metabolic stability testing was performed in vitro using a plethora of human and murine enzymes. RESULTS No metabolism was observed using monoamine oxidase A and B or catechol-O-methyl transferase. Incubation of [(18)F]FMeNER-D2 with CYP450-enzymes, predominantly located in the liver, led to a significant and fast metabolism of the tracer. Moreover, the arising three radiometabolites were found to be more polar than [(18)F]FMeNER-D2. Surprisingly, definitely no formation of free [(18)F]fluoride was observed. CONCLUSION According to our in vitro data, the interfering uptake in cortical regions might be attributed to these emerging radiometabolites but does not reflect bonding in bone due to defluorination. Further research on these radiometabolites is necessary to elucidate the in vivo situation. This might include an analysis of human blood samples after injection of [(18)F]FMeNER-D2, to enable a better correction of the PET-input function.
Collapse
|
33
|
Adhikarla V, Zeng F, Votaw JR, Goodman MM, Nye JA. Compartmental modeling of [(11)C]MENET binding to the norepinephrine transporter in the healthy human brain. Nucl Med Biol 2016; 43:318-23. [PMID: 27150035 DOI: 10.1016/j.nucmedbio.2016.02.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Revised: 02/24/2016] [Accepted: 02/24/2016] [Indexed: 11/24/2022]
Abstract
INTRODUCTION Dysregulation of the noradrenergic system has been implicated in a number of neurological conditions such as Parkinson's and Alzheimer's. [(11)C]MENET is a novel PET radiotracer with high affinity and selectivity for the norepinephrine transporter. The applicability of different kinetic models on [(11)C]MENET PET image quantification in healthy population is evaluated. METHODS Six healthy volunteers (mean age: 54years) were recruited for the study, five of whom underwent arterial sampling for measurement of the input function. Ninety minute dynamic PET scans were obtained on a high resolution research tomograph with 15mCi of [(11)C]MENET injected at the scan start time. Regions of interest were delineated on the PET scan aided by the corresponding MRI image for anatomical guidance. Distribution volumes and their ratios (DVRs) with respect to the occipital reference tissue were calculated using the full arterial model (FAM), the simplified reference tissue model (SRTM) and the multilinear reference tissue model (MRTM2). RESULTS Among the FAMs, the single-tissue model was found to be statistically superior to the two-tissue model. [(11)C]MENET focal uptake was observed in the NET-rich regions of the brainstem and subcortical regions including the thalamus, locus cereleus and the raphe nuclei. Highest DVRs were observed in the locus cereleus (mean±standard deviation: 1.39±0.25) and red nucleus (1.35±0.25). DVRs of the thalamus were in good agreement between FAM (1.26±0.13), SRTM (1.23±0.15) and MRTM2 (1.21±0.14). Comparing the FAM to the SRTM and MRTM2, DVRs were underestimated in the thalamus by 3 and 4% on average, respectively. CONCLUSION The single-tissue compartmental model was sufficient in describing the [(11)C]MENET kinetics in the healthy human brain. SRTM and MRTM2 present themselves as attractive options for estimating NET DVR using an occipital reference region.
Collapse
Affiliation(s)
- Vikram Adhikarla
- Department of Radiology and Imaging Sciences, Emory University, Atlanta, GA, 30322, USA.
| | - Fanxing Zeng
- Department of Radiology and Imaging Sciences, Emory University, Atlanta, GA, 30322, USA
| | - John R Votaw
- Department of Radiology and Imaging Sciences, Emory University, Atlanta, GA, 30322, USA
| | - Mark M Goodman
- Department of Radiology and Imaging Sciences, Emory University, Atlanta, GA, 30322, USA
| | - Jonathon A Nye
- Department of Radiology and Imaging Sciences, Emory University, Atlanta, GA, 30322, USA.
| |
Collapse
|
34
|
MUW researcher of the month Januar 2016. Wien Klin Wochenschr 2016; 128:82-4. [DOI: 10.1007/s00508-016-0950-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
35
|
Sigurdardottir HL, Kranz GS, Rami-Mark C, James GM, Vanicek T, Gryglewski G, Kautzky A, Hienert M, Traub-Weidinger T, Mitterhauser M, Wadsak W, Hacker M, Rujescu D, Kasper S, Lanzenberger R. Effects of norepinephrine transporter gene variants on NET binding in ADHD and healthy controls investigated by PET. Hum Brain Mapp 2015; 37:884-95. [PMID: 26678348 PMCID: PMC4949568 DOI: 10.1002/hbm.23071] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Revised: 11/18/2015] [Accepted: 11/18/2015] [Indexed: 01/08/2023] Open
Abstract
Attention deficit hyperactivity disorder (ADHD) is a heterogeneous disorder with a strong genetic component. The norepinephrine transporter (NET) is a key target for ADHD treatment and the NET gene has been of high interest as a possible modulator of ADHD pathophysiology. Therefore, we conducted an imaging genetics study to examine possible effects of single nucleotide polymorphisms (SNPs) within the NET gene on NET nondisplaceable binding potential (BPND ) in patients with ADHD and healthy controls (HCs). Twenty adult patients with ADHD and 20 HCs underwent (S,S)-[18F]FMeNER-D2 positron emission tomography (PET) and were genotyped on a MassARRAY MALDI-TOF platform using the Sequenom iPLEX assay. Linear mixed models analyses revealed a genotype-dependent difference in NET BPND between groups in the thalamus and cerebellum. In the thalamus, a functional promoter SNP (-3081 A/T) and a 5'-untranslated region (5'UTR) SNP (-182 T/C), showed higher binding in ADHD patients compared to HCs depending on the major allele. Furthermore, we detected an effect of genotype in HCs, with major allele carriers having lower binding. In contrast, for two 3'UTR SNPs (*269 T/C, *417 A/T), ADHD subjects had lower binding in the cerebellum compared to HCs depending on the major allele. Additionally, symptoms of hyperactivity and impulsivity correlated with NET BPND in the cerebellum depending on genotype. Symptoms correlated positively with cerebellar NET BPND for the major allele, while symptoms correlated negatively to NET BPND in minor allele carriers. Our findings support the role of genetic influence of the NE system on NET binding to be pertubated in ADHD.
Collapse
Affiliation(s)
- Helen L Sigurdardottir
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria
| | - Georg S Kranz
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria
| | - Christina Rami-Mark
- Department of Biomedical Imaging and Image-Guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Vienna, Austria
| | - Gregory M James
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria
| | - Thomas Vanicek
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria
| | - Gregor Gryglewski
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria
| | - Alexander Kautzky
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria
| | - Marius Hienert
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria
| | - Tatjana Traub-Weidinger
- Department of Biomedical Imaging and Image-Guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Vienna, Austria
| | - Markus Mitterhauser
- Department of Biomedical Imaging and Image-Guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Vienna, Austria
| | - Wolfgang Wadsak
- Department of Biomedical Imaging and Image-Guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Vienna, Austria
| | - Marcus Hacker
- Department of Biomedical Imaging and Image-Guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Vienna, Austria
| | - Dan Rujescu
- Department of Psychiatry, University of Halle, Halle, Germany
| | - Siegfried Kasper
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria
| | - Rupert Lanzenberger
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
36
|
1-(3-Amino-1-phenylpropyl)-3-(2-fluorophenyl)-1,3-dihydro-2H-benzimidazol-2-one. MOLBANK 2015. [DOI: 10.3390/m867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
37
|
Bello NT. Clinical utility of guanfacine extended release in the treatment of ADHD in children and adolescents. Patient Prefer Adherence 2015; 9:877-85. [PMID: 26170637 PMCID: PMC4494608 DOI: 10.2147/ppa.s73167] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Attention deficit hyperactivity disorder (ADHD) is the most common psychiatric illness in children and adolescents. Several stimulant medications, such as methylphenidate and amphetamine derivatives, are available to treat ADHD in pediatric patients. Nonstimulant medications are more preferred by some parents, other caregivers, and patients because they lack the abuse potential of stimulant medications. In the US, one available nonstimulant option is guanfacine extended release (XR). As a selective α2A adrenergic receptor, guanfacine acts on the central noradrenergic pathways and cortical noradrenergic targets to improve working memory and attention. The XR formulation of guanfacine, compared with the immediate-release formulation, is more effective for the long-term management of ADHD and is associated with fewer adverse effects. Available data also indicate that guanfacine XR is superior to atomoxetine and is as effective as the nonselective α2 adrenergic receptor agonist, clonidine XR. The most common adverse effects associated with guanfacine XR are somnolence, fatigue, bradycardia, and hypotension. Somnolence is the most often cited reason for discontinuation. Guanfacine XR is also labeled for use as an adjuvant to stimulant treatment for ADHD. A similar profile of adverse effects as reported with monotherapy is reported when guanfacine XR is "added on" to stimulant therapy with somnolence as the most commonly reported adverse event. This review discusses the clinical efficacy and patient preference of guanfacine XR based on available published data on the safety, relative effectiveness, and tolerance of this medication to treat ADHD.
Collapse
Affiliation(s)
- Nicholas T Bello
- Department of Animal Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
| |
Collapse
|