1
|
Siebenhühner F, Palva JM, Palva S. Linking the microarchitecture of neurotransmitter systems to large-scale MEG resting state networks. iScience 2024; 27:111111. [PMID: 39524335 PMCID: PMC11544385 DOI: 10.1016/j.isci.2024.111111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 07/06/2024] [Accepted: 10/02/2024] [Indexed: 11/16/2024] Open
Abstract
Neuronal oscillations are ubiquitous in brain activity at all scales and their synchronization dynamics are essential for information processing in neuronal systems. The underlying synaptic mechanisms, while mainly based on GABA- and glutamatergic neurotransmission, are influenced by neuromodulatory systems that have highly variable densities of neurotransmitter receptors and transporters across the cortical mantle. How they constrain the network structures of interacting oscillations has remained a central unaddressed question. We asked here whether the receptor and transporter densities covary with the frequency-specific neuroanatomical patterns of inter-areal phase synchrony (PS) and amplitude correlation (AC) networks in resting-state magnetoencephalography (MEG) data. Network centrality in delta and gamma frequencies covaried positively with GABA-, NMDA-, dopaminergic-, and most serotonergic receptor and transporter densities while covariance was negative in alpha and beta bands. These results show that local receptor microarchitecture shapes macro-scale oscillation networks in spectrally specific patterns.
Collapse
Affiliation(s)
- Felix Siebenhühner
- Neuroscience Center, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
- BioMag Laboratory, HUS Medical Imaging Centre, Helsinki University, Helsinki, Finland
- Department of Neuroscience and Bioengineering (NBE), Aalto University, Espoo, Finland
- Department of Electrical Engineering and Information Technology, Technical University Darmstadt, Darmstadt, Germany
| | - J. Matias Palva
- Neuroscience Center, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
- Department of Neuroscience and Bioengineering (NBE), Aalto University, Espoo, Finland
- Centre for Cognitive Neuroimaging (CCNi), School of Psychology and Neuroscience, University of Glasgow, Glasgow, UK
| | - Satu Palva
- Neuroscience Center, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
- Centre for Cognitive Neuroimaging (CCNi), School of Psychology and Neuroscience, University of Glasgow, Glasgow, UK
- Division of Psychology, VISE, Faculty of Education and Psychology, University of Oulu, Oulu, Finland
| |
Collapse
|
2
|
Souza BR, Codo BC, Romano-Silva MA, Tropepe V. Darpp-32 is regulated by dopamine and is required for the formation of GABAergic neurons in the developing telencephalon. Prog Neuropsychopharmacol Biol Psychiatry 2024; 134:111060. [PMID: 38906412 DOI: 10.1016/j.pnpbp.2024.111060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 04/22/2024] [Accepted: 06/17/2024] [Indexed: 06/23/2024]
Abstract
DARPP-32 (dopamine and cAMP-regulated phosphoprotein Mr. 32 kDa) is a phosphoprotein that is modulated by multiple receptors integrating intracellular pathways and playing roles in various physiological functions. It is regulated by dopaminergic receptors through the cAMP/protein kinase A (PKA) pathway, which modulates the phosphorylation of threonine 34 (Thr34). When phosphorylated at Thr34, DARPP-32 becomes a potent protein phosphatase-1 (PP1) inhibitor. Since dopamine is involved in the development of GABAergic neurons and DARPP-32 is expressed in the developing brain, it is possible that DARPP-32 has a role in GABAergic neuronal development. We cloned the zebrafish darpp-32 gene (ppp1r1b) gene and observed that it is evolutionarily conserved in its inhibitory domain (Thr34 and surrounding residues) and the docking motif (residues 7-11 (KKIQF)). We also characterized darpp-32 protein expression throughout the 5 days post-fertilization (dpf) zebrafish larval brain by immunofluorescence and demonstrated that darpp-32 is mainly expressed in regions that receive dopaminergic projections (pallium, subpallium, preoptic region, and hypothalamus). We demonstrated that dopamine acutely suppressed darpp-32 activity by reducing the levels of p-darpp-32 in the 5dpf zebrafish larval brain. In addition, the knockdown of darpp-32 resulted in a decrease in the number of GABAergic neurons in the subpallium of the 5dpf larval brain, with a concomitant increase in the number of DAergic neurons. Finally, we demonstrated that darpp-32 downregulation during development reduced the motor behavior of 5dpf zebrafish larvae. Thus, our observations suggest that darpp-32 is an evolutionarily conserved regulator of dopamine receptor signaling and is required for the formation of GABAergic neurons in the developing telencephalon.
Collapse
Affiliation(s)
- Bruno Rezende Souza
- Laboratório NeuroDEv, Department of Physiology and Biophysics, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil 31270-901; Laboratório de Neurociências Molecular e Comportamental (LANEC) - Universidade Federal de Minas Gerais, Belo Horizonte, MG 31270-901, Brazil.
| | - Beatriz Campos Codo
- Laboratório NeuroDEv, Department of Physiology and Biophysics, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil 31270-901; Laboratório de Neurociências Molecular e Comportamental (LANEC) - Universidade Federal de Minas Gerais, Belo Horizonte, MG 31270-901, Brazil
| | - Marco Aurélio Romano-Silva
- Laboratório de Neurociências and INCT de Medicina Molecular, Department of Mental Health, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil 30130-100
| | - Vincent Tropepe
- Department of Cell & Systems Biology, University of Toronto, Toronto, ON, Canada M5S 3G5.
| |
Collapse
|
3
|
Moppert S, Mercado E. Contributions of dysfunctional plasticity mechanisms to the development of atypical perceptual processing. Dev Psychobiol 2024; 66:e22504. [PMID: 38837411 DOI: 10.1002/dev.22504] [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: 07/29/2023] [Revised: 03/04/2024] [Accepted: 05/11/2024] [Indexed: 06/07/2024]
Abstract
Experimental studies of sensory plasticity during development in birds and mammals have highlighted the importance of sensory experiences for the construction and refinement of functional neural circuits. We discuss how dysregulation of experience-dependent brain plasticity can lead to abnormal perceptual representations that may contribute to heterogeneous deficits symptomatic of several neurodevelopmental disorders. We focus on alterations of somatosensory processing and the dynamic reorganization of cortical synaptic networks that occurs during early perceptual development. We also discuss the idea that the heterogeneity of strengths and weaknesses observed in children with neurodevelopmental disorders may be a direct consequence of altered plasticity mechanisms during early development. Treating the heterogeneity of perceptual developmental trajectories as a phenomenon worthy of study rather than as an experimental confound that should be overcome may be key to developing interventions that better account for the complex developmental trajectories experienced by modern humans.
Collapse
Affiliation(s)
- Stacy Moppert
- Department of Psychology, University at Buffalo, The State University of New York, Buffalo, New York, USA
| | - Eduardo Mercado
- Department of Psychology, University at Buffalo, The State University of New York, Buffalo, New York, USA
| |
Collapse
|
4
|
Thomson AR, Pasanta D, Arichi T, Puts NA. Neurometabolite differences in Autism as assessed with Magnetic Resonance Spectroscopy: A systematic review and meta-analysis. Neurosci Biobehav Rev 2024; 162:105728. [PMID: 38796123 DOI: 10.1016/j.neubiorev.2024.105728] [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: 01/26/2024] [Revised: 04/23/2024] [Accepted: 05/14/2024] [Indexed: 05/28/2024]
Abstract
1H-Magnetic Resonance Spectroscopy (MRS) is a non-invasive technique that can be used to quantify the concentrations of metabolites in the brain in vivo. MRS findings in the context of autism are inconsistent and conflicting. We performed a systematic review and meta-analysis of MRS studies measuring glutamate and gamma-aminobutyric acid (GABA), as well as brain metabolites involved in energy metabolism (glutamine, creatine), neural and glial integrity (e.g. n-acetyl aspartate (NAA), choline, myo-inositol) and oxidative stress (glutathione) in autism cohorts. Data were extracted and grouped by metabolite, brain region and several other factors before calculation of standardised effect sizes. Overall, we find significantly lower concentrations of GABA and NAA in autism, indicative of disruptions to the balance between excitation/inhibition within brain circuits, as well as neural integrity. Further analysis found these alterations are most pronounced in autistic children and in limbic brain regions relevant to autism phenotypes. Additionally, we show how study outcome varies due to demographic and methodological factors , emphasising the importance of conforming with standardised consensus study designs and transparent reporting.
Collapse
Affiliation(s)
- Alice R Thomson
- Department of Forensic and Neurodevelopmental Sciences, King's College London, UK; MRC Centre for Neurodevelopmental Disorders, King's College London, UK; Centre for the Developing Brain, King's College London, London, UK
| | - Duanghathai Pasanta
- Department of Forensic and Neurodevelopmental Sciences, King's College London, UK
| | - Tomoki Arichi
- MRC Centre for Neurodevelopmental Disorders, King's College London, UK; Centre for the Developing Brain, King's College London, London, UK
| | - Nicolaas A Puts
- Department of Forensic and Neurodevelopmental Sciences, King's College London, UK; MRC Centre for Neurodevelopmental Disorders, King's College London, UK.
| |
Collapse
|
5
|
Li DJ, Tsai SJ, Chen TJ, Liang CS, Chen MH. Risk of major mental disorders in the offspring of parents with migraine. Ann Gen Psychiatry 2024; 23:23. [PMID: 38909222 PMCID: PMC11193281 DOI: 10.1186/s12991-024-00508-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Accepted: 06/14/2024] [Indexed: 06/24/2024] Open
Abstract
BACKGROUND Migraine has been associated with mental disorders, however whether parental migraine is associated with an increased risk of major mental disorders (MMDs) in offspring has not been investigated. We aimed to examine the risk of the development of MMDs in the offspring of parents with migraine compared with those of parents without migraine. METHODS This study used data derived from the Taiwan National Health Insurance Research Database. Offspring of parents with migraine and a control group consisting of offspring of parents without migraine matched for demographic and parental mental disorders were included. Cox regression was used to estimate the risk of MMDs, including schizophrenia, depressive disorder, bipolar disorder, autistic spectrum disorder (ASD), and attention deficit/hyperactivity disorder (ADHD). Sub-analyses stratified by the fathers and mothers were further performed to separately clarify the risks of MMDs among the offspring. RESULTS We included 22,747 offspring of parents with migraine and 227,470 offspring of parents without migraine as the controls. Parental migraine was significantly associated with an increased risk of ADHD (reported as hazard ratios with 95% confidence intervals: 1.37, 1.25-1.50), bipolar disorder (1.35, 1.06-1.71), and depressive disorder (1.33, 1.21-1.47) compared to the offspring of parents without migraine. Importantly, sub-analyses showed that only maternal migraine was significantly associated with these risks. CONCLUSIONS Due to the heavy burden of MMDs, healthcare workers should be aware of the risk of MMDs in the offspring of parents with migraine, particular in mothers.
Collapse
Affiliation(s)
- Dian-Jeng Li
- Department of Addiction Science, Kaohsiung Municipal Kai-Syuan Psychiatric Hospital, Kaohsiung, Taiwan
- Department of Nursing, Meiho University, Pingtung, Taiwan
| | - Shih-Jen Tsai
- Department of Psychiatry, Taipei Veterans General Hospital, Taipei, Taiwan
- Department of Psychiatry, College of Medicine, National Yang Ming Chiao Tung University, No. 201, Sec. 2, Shihpai Road, Beitou District, Taipei, 11217, Taiwan
| | - Tzeng-Ji Chen
- Department of Family Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- Institute of Hospital and Health Care Administration, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Department of Family Medicine, Hsinchu Branch, Taipei Veterans General Hospital, Hsinchu, Taiwan
| | - Chih-Sung Liang
- Department of Psychiatry, Beitou Branch, Tri-Service General Hospital, National Defense Medical School, Taipei, Taiwan.
- Department of Psychiatry, National Defense Medical School, No. 60, Xinmin Road, Beitou District, Taipei, 11243, Taiwan.
| | - Mu-Hong Chen
- Department of Psychiatry, Taipei Veterans General Hospital, Taipei, Taiwan.
- Department of Psychiatry, College of Medicine, National Yang Ming Chiao Tung University, No. 201, Sec. 2, Shihpai Road, Beitou District, Taipei, 11217, Taiwan.
| |
Collapse
|
6
|
Watanangura A, Meller S, Farhat N, Suchodolski JS, Pilla R, Khattab MR, Lopes BC, Bathen-Nöthen A, Fischer A, Busch-Hahn K, Flieshardt C, Gramer M, Richter F, Zamansky A, Volk HA. Behavioral comorbidities treatment by fecal microbiota transplantation in canine epilepsy: a pilot study of a novel therapeutic approach. Front Vet Sci 2024; 11:1385469. [PMID: 38978633 PMCID: PMC11229054 DOI: 10.3389/fvets.2024.1385469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 05/15/2024] [Indexed: 07/10/2024] Open
Abstract
Introduction Anxiety and cognitive dysfunction are frequent, difficult to treat and burdensome comorbidities in human and canine epilepsy. Fecal microbiota transplantation (FMT) has been shown to modulate behavior in rodent models by altering the gastrointestinal microbiota (GIM). This study aims to investigate the beneficial effects of FMT on behavioral comorbidities in a canine translational model of epilepsy. Methods Nine dogs with drug-resistant epilepsy (DRE) and behavioral comorbidities were recruited. The fecal donor had epilepsy with unremarkable behavior, which exhibited a complete response to phenobarbital, resulting in it being seizure-free long term. FMTs were performed three times, two weeks apart, and the dogs had follow-up visits at three and six months after FMTs. Comprehensive behavioral analysis, including formerly validated questionnaires and behavioral tests for attention deficit hyperactivity disorder (ADHD)- and fear- and anxiety-like behavior, as well as cognitive dysfunction, were conducted, followed by objective computational analysis. Blood samples were taken for the analysis of antiseizure drug (ASD) concentrations, hematology, and biochemistry. Urine neurotransmitter concentrations were measured. Fecal samples were subjected to analysis using shallow DNA shotgun sequencing, real-time polymerase chain reaction (qPCR)-based Dysbiosis Index (DI) assessment, and short-chain fatty acid (SCFA) quantification. Results Following FMT, the patients showed improvement in ADHD-like behavior, fear- and anxiety-like behavior, and quality of life. The excitatory neurotransmitters aspartate and glutamate were decreased, while the inhibitory neurotransmitter gamma-aminobutyric acid (GABA) and GABA/glutamate ratio were increased compared to baseline. Only minor taxonomic changes were observed, with a decrease in Firmicutes and a Blautia_A species, while a Ruminococcus species increased. Functional gene analysis, SCFA concentration, blood parameters, and ASD concentrations remained unchanged. Discussion Behavioral comorbidities in canine IE could be alleviated by FMT. This study highlights FMT's potential as a novel approach to improving behavioral comorbidities and enhancing the quality of life in canine patients with epilepsy.
Collapse
Affiliation(s)
- Antja Watanangura
- Department of Small Animal Medicine and Surgery, University of Veterinary Medicine Hannover, Hannover, Germany
- Center for Systems Neuroscience (ZSN), Hannover, Germany
- Veterinary Research and Academic Service, Faculty of Veterinary Medicine, Kasetsart University, Nakhon Pathom, Thailand
| | - Sebastian Meller
- Department of Small Animal Medicine and Surgery, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Nareed Farhat
- Tech4Animals Lab, Information Systems Department, University of Haifa, Haifa, Israel
| | - Jan S. Suchodolski
- Gastrointestinal Laboratory, Department of Small Animal Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, TX, United States
| | - Rachel Pilla
- Gastrointestinal Laboratory, Department of Small Animal Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, TX, United States
| | - Mohammad R. Khattab
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO, United States
| | - Bruna C. Lopes
- Gastrointestinal Laboratory, Department of Small Animal Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, TX, United States
| | | | - Andrea Fischer
- Centre for Clinical Veterinary Medicine, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Kathrin Busch-Hahn
- Centre for Clinical Veterinary Medicine, Ludwig-Maximilians-Universität München, Munich, Germany
| | | | - Martina Gramer
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Franziska Richter
- Center for Systems Neuroscience (ZSN), Hannover, Germany
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Anna Zamansky
- Tech4Animals Lab, Information Systems Department, University of Haifa, Haifa, Israel
| | - Holger A. Volk
- Department of Small Animal Medicine and Surgery, University of Veterinary Medicine Hannover, Hannover, Germany
- Center for Systems Neuroscience (ZSN), Hannover, Germany
| |
Collapse
|
7
|
Ferranti AS, Luessen DJ, Niswender CM. Novel pharmacological targets for GABAergic dysfunction in ADHD. Neuropharmacology 2024; 249:109897. [PMID: 38462041 DOI: 10.1016/j.neuropharm.2024.109897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Revised: 02/26/2024] [Accepted: 03/04/2024] [Indexed: 03/12/2024]
Abstract
Attention deficit/hyperactivity disorder (ADHD) is a neurodevelopment disorder that affects approximately 5% of the population. The disorder is characterized by impulsivity, hyperactivity, and deficits in attention and cognition, although symptoms vary across patients due to the heterogenous and polygenic nature of the disorder. Stimulant medications are the standard of care treatment for ADHD patients, and their effectiveness has led to the dopaminergic hypothesis of ADHD in which deficits in dopaminergic signaling, especially in cortical brain regions, mechanistically underly ADHD pathophysiology. Despite their effectiveness in many individuals, almost one-third of patients do not respond to stimulant treatments and the long-term negative side effects of these medications remain unclear. Emerging clinical evidence is beginning to highlight an important role of dysregulated excitatory/inhibitory (E/I) balance in ADHD. These deficits in E/I balance are related to functional abnormalities in glutamate and Gamma-Aminobutyric Acid (GABA) signaling in the brain, with increasing emphasis placed on GABAergic interneurons driving specific aspects of ADHD pathophysiology. Recent genome-wide association studies (GWAS) have also highlighted how genes associated with GABA function are mutated in human populations with ADHD, resulting in the generation of several new genetic mouse models of ADHD. This review will discuss how GABAergic dysfunction underlies ADHD pathophysiology, and how specific receptors/proteins related to GABAergic interneuron dysfunction may be pharmacologically targeted to treat ADHD in subpopulations with specific comorbidities and symptom domains. This article is part of the Special Issue on "PFC circuit function in psychiatric disease and relevant models".
Collapse
Affiliation(s)
- Anthony S Ferranti
- Department of Pharmacology, Vanderbilt University, Nashville, TN, 37232, USA; Warren Center for Neuroscience Drug Discovery, Nashville, TN, 37232, USA
| | - Deborah J Luessen
- Department of Pharmacology, Vanderbilt University, Nashville, TN, 37232, USA; Warren Center for Neuroscience Drug Discovery, Nashville, TN, 37232, USA
| | - Colleen M Niswender
- Department of Pharmacology, Vanderbilt University, Nashville, TN, 37232, USA; Warren Center for Neuroscience Drug Discovery, Nashville, TN, 37232, USA; Vanderbilt Kennedy Center, Vanderbilt University Medical Center, Nashville, TN, 37232, USA; Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, 37232, USA; Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN, 37232, USA.
| |
Collapse
|
8
|
Chen Z, Xu T, Liu X, Becker B, Li W, Xia L, Zhao W, Zhang R, Huo Z, Hu B, Tang Y, Xiao Z, Feng Z, Chen J, Feng T. Cortical gradient perturbation in attention deficit hyperactivity disorder correlates with neurotransmitter-, cell type-specific and chromosome- transcriptomic signatures. Psychiatry Clin Neurosci 2024; 78:309-321. [PMID: 38334172 DOI: 10.1111/pcn.13649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 01/16/2024] [Accepted: 01/18/2024] [Indexed: 02/10/2024]
Abstract
AIMS This study aimed to illuminate the neuropathological landscape of attention deficit hyperactivity disorder (ADHD) by a multiscale macro-micro-molecular perspective from in vivo neuroimaging data. METHODS The "ADHD-200 initiative" repository provided multi-site high-quality resting-state functional connectivity (rsfc-) neuroimaging for ADHD children and matched typically developing (TD) cohort. Diffusion mapping embedding model to derive the functional connectome gradient detecting biologically plausible neural pattern was built, and the multivariate partial least square method to uncover the enrichment of neurotransmitomic, cellular and chromosomal gradient-transcriptional signatures of AHBA enrichment and meta-analytic decoding. RESULTS Compared to TD, ADHD children presented connectopic cortical gradient perturbations in almost all the cognition-involved brain macroscale networks (all pBH <0.001), but not in the brain global topology. As an intermediate phenotypic variant, such gradient perturbation was spatially enriched into distributions of GABAA/BZ and 5-HT2A receptors (all pBH <0.01) and co-varied with genetic transcriptional expressions (e.g. DYDC2, ATOH7, all pBH <0.01), associated with phenotypic variants in episodic memory and emotional regulations. Enrichment models demonstrated such gradient-transcriptional variants indicated the risk of both cell-specific and chromosome- dysfunctions, especially in enriched expression of oligodendrocyte precursors and endothelial cells (all pperm <0.05) as well enrichment into chromosome 18, 19 and X (pperm <0.05). CONCLUSIONS Our findings bridged brain macroscale neuropathological patterns to microscale/cellular biological architectures for ADHD children, demonstrating the neurobiologically pathological mechanism of ADHD into the genetic and molecular variants in GABA and 5-HT systems as well brain-derived enrichment of specific cellular/chromosomal expressions.
Collapse
Affiliation(s)
- Zhiyi Chen
- Experimental Research Center of Medical and Psychological Science, School of Psychology, Third Military Medical University, Chongqing, China
- Key Laboratory of Cognition and Personality, Ministry of Education, Faculty of Psychology, Southwest University, Chongqing, China
| | - Ting Xu
- Department of Psychology, The University of Hong Kong, Hong Kong, China
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Laboratory for Neuroinformation, University of Electronic Science and Technology of China, Chengdu, China
| | - Xuerong Liu
- Experimental Research Center of Medical and Psychological Science, School of Psychology, Third Military Medical University, Chongqing, China
| | - Benjamin Becker
- Department of Psychology, The University of Hong Kong, Hong Kong, China
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Laboratory for Neuroinformation, University of Electronic Science and Technology of China, Chengdu, China
| | - Wei Li
- Experimental Research Center of Medical and Psychological Science, School of Psychology, Third Military Medical University, Chongqing, China
| | - Lei Xia
- Experimental Research Center of Medical and Psychological Science, School of Psychology, Third Military Medical University, Chongqing, China
| | - Wenqi Zhao
- Experimental Research Center of Medical and Psychological Science, School of Psychology, Third Military Medical University, Chongqing, China
| | - Rong Zhang
- Key Laboratory of Cognition and Personality, Ministry of Education, Faculty of Psychology, Southwest University, Chongqing, China
| | - Zhenzhen Huo
- Key Laboratory of Cognition and Personality, Ministry of Education, Faculty of Psychology, Southwest University, Chongqing, China
| | - Bowen Hu
- Key Laboratory of Cognition and Personality, Ministry of Education, Faculty of Psychology, Southwest University, Chongqing, China
| | - Yancheng Tang
- School of Business and Management, Shanghai International Studies University, Shanghai, China
| | - Zhibing Xiao
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing, China
| | - Zhengzhi Feng
- Experimental Research Center of Medical and Psychological Science, School of Psychology, Third Military Medical University, Chongqing, China
- Key Laboratory of Cognition and Personality, Ministry of Education, Faculty of Psychology, Southwest University, Chongqing, China
| | - Ji Chen
- Department of Psychology and Behavioral Sciences, Zhejiang University, Hangzhou, China
- Department of Psychiatry, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, China
| | - Tingyong Feng
- Key Laboratory of Cognition and Personality, Ministry of Education, Faculty of Psychology, Southwest University, Chongqing, China
| |
Collapse
|
9
|
Aschner M, Martins AC, Oliveira-Paula GH, Skalny AV, Zaitseva IP, Bowman AB, Kirichuk AA, Santamaria A, Tizabi Y, Tinkov AA. Manganese in autism spectrum disorder and attention deficit hyperactivity disorder: The state of the art. Curr Res Toxicol 2024; 6:100170. [PMID: 38737010 PMCID: PMC11088232 DOI: 10.1016/j.crtox.2024.100170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 03/27/2024] [Accepted: 04/23/2024] [Indexed: 05/14/2024] Open
Abstract
The objective of the present narrative review was to synthesize existing clinical and epidemiological findings linking manganese (Mn) exposure biomarkers to autism spectrum disorder (ASD) and attention deficit hyperactivity disorder (ADHD), and to discuss key pathophysiological mechanisms of neurodevelopmental disorders that may be affected by this metal. Existing epidemiological data demonstrated both direct and inverse association between Mn body burden and ASD, or lack of any relationship. In contrast, the majority of studies revealed significantly higher Mn levels in subjects with ADHD, as well as direct relationship between Mn body burden with hyperactivity and inattention scores in children, although several studies reported contradictory results. Existing laboratory studies demonstrated that impaired attention and hyperactivity in animals following Mn exposure was associated with dopaminergic dysfunction and neuroinflammation. Despite lack of direct evidence on Mn-induced neurobiological alterations in patients with ASD and ADHD, a plethora of studies demonstrated that neurotoxic effects of Mn overexposure may interfere with key mechanisms of pathogenesis inherent to these neurodevelopmental disorders. Specifically, Mn overload was shown to impair not only dopaminergic neurotransmission, but also affect metabolism of glutamine/glutamate, GABA, serotonin, noradrenaline, thus affecting neuronal signaling. In turn, neurotoxic effects of Mn may be associated with its ability to induce oxidative stress, apoptosis, and neuroinflammation, and/or impair neurogenesis. Nonetheless, additional detailed studies are required to evaluate the association between environmental Mn exposure and/or Mn body burden and neurodevelopmental disorders at a wide range of concentrations to estimate the potential dose-dependent effects, as well as environmental and genetic factors affecting this association.
Collapse
Affiliation(s)
- Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, New York, NY 10461, USA
| | - Airton C. Martins
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, New York, NY 10461, USA
| | | | - Anatoly V. Skalny
- Department of Medical Elementology, and Department of Human Ecology and Bioelementology, Peoples' Friendship University of Russia (RUDN University), Moscow 117198, Russia
- Center of Bioelementology and Human Ecology, IM Sechenov First Moscow State Medical University (Sechenov University), Moscow 119435, Russia
| | - Irina P. Zaitseva
- Laboratory of Ecobiomonitoring and Quality Control, Yaroslavl State University, Yaroslavl 150003, Russia
| | - Aaron B. Bowman
- School of Health Sciences, Purdue University, West Lafayette, IN 47907-2051, USA
| | - Anatoly A. Kirichuk
- Department of Medical Elementology, and Department of Human Ecology and Bioelementology, Peoples' Friendship University of Russia (RUDN University), Moscow 117198, Russia
| | - Abel Santamaria
- Facultad de Ciencias, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
- Laboratorio de Nanotecnología y Nanomedicina, Departamento de Cuidado de la Salud, Universidad Autónoma Metropolitana-Xochimilco, Mexico City 04960, Mexico
| | - Yousef Tizabi
- Department of Pharmacology, Howard University College of Medicine, Washington, DC 20059, USA
| | - Alexey A. Tinkov
- Department of Medical Elementology, and Department of Human Ecology and Bioelementology, Peoples' Friendship University of Russia (RUDN University), Moscow 117198, Russia
- Center of Bioelementology and Human Ecology, IM Sechenov First Moscow State Medical University (Sechenov University), Moscow 119435, Russia
- Laboratory of Ecobiomonitoring and Quality Control, Yaroslavl State University, Yaroslavl 150003, Russia
| |
Collapse
|
10
|
Muscò A, Martini D, Digregorio M, Broccoli V, Andreazzoli M. Shedding a Light on Dark Genes: A Comparative Expression Study of PRR12 Orthologues during Zebrafish Development. Genes (Basel) 2024; 15:492. [PMID: 38674426 PMCID: PMC11050278 DOI: 10.3390/genes15040492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Revised: 04/06/2024] [Accepted: 04/09/2024] [Indexed: 04/28/2024] Open
Abstract
Haploinsufficiency of the PRR12 gene is implicated in a human neuro-ocular syndrome. Although identified as a nuclear protein highly expressed in the embryonic mouse brain, PRR12 molecular function remains elusive. This study explores the spatio-temporal expression of zebrafish PRR12 co-orthologs, prr12a and prr12b, as a first step to elucidate their function. In silico analysis reveals high evolutionary conservation in the DNA-interacting domains for both orthologs, with significant syntenic conservation observed for the prr12b locus. In situ hybridization and RT-qPCR analyses on zebrafish embryos and larvae reveal distinct expression patterns: prr12a is expressed early in zygotic development, mainly in the central nervous system, while prr12b expression initiates during gastrulation, localizing later to dopaminergic telencephalic and diencephalic cell clusters. Both transcripts are enriched in the ganglion cell and inner neural layers of the 72 hpf retina, with prr12b widely distributed in the ciliary marginal zone. In the adult brain, prr12a and prr12b are found in the cerebellum, amygdala and ventral telencephalon, which represent the main areas affected in autistic patients. Overall, this study suggests PRR12's potential involvement in eye and brain development, laying the groundwork for further investigations into PRR12-related neurobehavioral disorders.
Collapse
Affiliation(s)
- Alessia Muscò
- Cell and Developmental Biology Unit, University of Pisa, 56126 Pisa, Italy (D.M.)
| | - Davide Martini
- Cell and Developmental Biology Unit, University of Pisa, 56126 Pisa, Italy (D.M.)
| | - Matteo Digregorio
- Cell and Developmental Biology Unit, University of Pisa, 56126 Pisa, Italy (D.M.)
| | - Vania Broccoli
- Stem Cell and Neurogenesis Unit, Division of Neuroscience, San Raffaele Scientific Institute, 20132 Milan, Italy
- CNR Institute of Neuroscience, 20132 Milan, Italy
| | | |
Collapse
|
11
|
Braga JD, Thongngam M, Kumrungsee T. Gamma-aminobutyric acid as a potential postbiotic mediator in the gut-brain axis. NPJ Sci Food 2024; 8:16. [PMID: 38565567 PMCID: PMC10987602 DOI: 10.1038/s41538-024-00253-2] [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: 07/26/2023] [Accepted: 02/01/2024] [Indexed: 04/04/2024] Open
Abstract
Gamma-aminobutyric acid (GABA) plays a crucial role in the central nervous system as an inhibitory neurotransmitter. Imbalances of this neurotransmitter are associated with neurological diseases, such as Alzheimer's and Parkinson's disease, and psychological disorders, including anxiety, depression, and stress. Since GABA has long been believed to not cross the blood-brain barrier, the effects of circulating GABA on the brain are neglected. However, emerging evidence has demonstrated that changes in both circulating and brain levels of GABA are associated with changes in gut microbiota composition and that changes in GABA levels and microbiota composition play a role in modulating mental health. This recent research has raised the possibility that GABA may be a potent mediator of the gut-brain axis. This review article will cover up-to-date information about GABA-producing microorganisms isolated from human gut and food sources, explanation why those microorganisms produce GABA, food factors inducing gut-GABA production, evidence suggesting GABA as a mediator linking between gut microbiota and mental health, including anxiety, depression, stress, epilepsy, autism spectrum disorder, and attention deficit hyperactivity disorder, and novel information regarding homocarnosine-a predominant brain peptide that is a putative downstream mediator of GABA in regulating brain functions. This review will help us to understand how the gut microbiota and GABA-homocarnosine metabolism play a significant role in brain functions. Nonetheless, it could support further research on the use of GABA production-inducing microorganisms and food factors as agents to treat neurological and psychological disorders.
Collapse
Affiliation(s)
- Jason D Braga
- Laboratory of Molecular Nutrition, Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima, 739-8527, Japan
- Institute of Food Science and Technology, College of Agriculture, Food, Environment and Natural Resources, Cavite State University, Indang, Cavite, 4122, Philippines
| | - Masubon Thongngam
- Department of Food Science and Technology, Faculty of Agro-Industry, Kasetsart University, Bangkok, 10900, Thailand
| | - Thanutchaporn Kumrungsee
- Laboratory of Molecular Nutrition, Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima, 739-8527, Japan.
- Smart Agriculture, Graduate School of Innovation and Practice for Smart Society, Hiroshima University, Hiroshima, 739-8527, Japan.
| |
Collapse
|
12
|
Bogdańska-Chomczyk E, Równiak M, Huang ACW, Kozłowska A. Parvalbumin interneuron deficiency in the prefrontal and motor cortices of spontaneously hypertensive rats: an attention-deficit hyperactivity disorder animal model insight. Front Psychiatry 2024; 15:1359237. [PMID: 38600979 PMCID: PMC11005678 DOI: 10.3389/fpsyt.2024.1359237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 02/15/2024] [Indexed: 04/12/2024] Open
Abstract
Background Attention deficit hyperactivity disorder (ADHD) is characterized by impairments in developmental-behavioral inhibition, resulting in impulsivity and hyperactivity. Recent research has underscored cortical inhibition deficiencies in ADHD via the gamma-aminobutyric acid (GABA)ergic system, which is crucial for maintaining excitatory-inhibitory balance in the brain. This study explored postnatal changes in parvalbumin (PV) immunoreactivity, indicating GABAergic interneuron types, in the prefrontal (PFC) and motor (MC) cortices of spontaneously hypertensive rats (SHRs), an ADHD animal model. Methods Examining PV- positive (PV+) cells associated with dopamine D2 receptors (D2) and the impact of dopamine on GABA synthesis, we also investigated changes in the immunoreactivity of D2 and tyrosine hydroxylase (TH). Brain sections from 4- to 10-week-old SHRs and Wistar Kyoto rats (WKYs) were immunohistochemically analyzed, comparing PV+, D2+ cells, and TH+ fiber densities across age-matched SHRs and WKYs in specific PFC/MC regions. Results The results revealed significantly reduced PV+ cell density in SHRs: prelimbic (~20% less), anterior cingulate (~15% less), primary (~15% less), and secondary motor (~17% less) cortices. PV+ deficits coincided with the upregulation of D2 in prepubertal SHRs and the downregulation of TH predominantly in pubertal/postpubertal SHRs. Conclusion Reduced PV+ cells in various PFC regions could contribute to inattention/behavioral alterations in ADHD, while MC deficits could manifest as motor hyperactivity. D2 upregulation and TH deficits may impact GABA synthesis, exacerbating behavioral deficits in ADHD. These findings not only shed new light on ADHD pathophysiology but also pave the way for future research endeavors.
Collapse
Affiliation(s)
- Ewelina Bogdańska-Chomczyk
- Department of Human Physiology and Pathophysiology, School of Medicine, University of Warmia and Mazury, Olsztyn, Poland
| | - Maciej Równiak
- Department of Animal Anatomy and Physiology, Faculty of Biology and Biotechnology, University of Warmia and Mazury, Olsztyn, Poland
| | | | - Anna Kozłowska
- Department of Human Physiology and Pathophysiology, School of Medicine, University of Warmia and Mazury, Olsztyn, Poland
| |
Collapse
|
13
|
You DD, Huang YM, Wang XY, Li W, Li F. Long-term low-dose lamotrigine for paroxysmal kinesigenic dyskinesia: a two-year investigation of cognitive function in children. Front Psychiatry 2024; 15:1368289. [PMID: 38528979 PMCID: PMC10961978 DOI: 10.3389/fpsyt.2024.1368289] [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: 01/10/2024] [Accepted: 02/27/2024] [Indexed: 03/27/2024] Open
Abstract
Objective While low-dose lamotrigine has shown effectiveness in managing paroxysmal kinesigenic dyskinesia (PKD) in pediatric populations, the cognitive consequences of extended use are yet to be fully elucidated. This study seeks to assess the evolution of cognitive functions and the amelioration of attention deficit and hyperactivity disorder (ADHD) symptoms following a two-year lamotrigine treatment in children. Methods This investigation employed an open-label, uncontrolled trial design. Between January 2008 and December 2021, thirty-one participants, ranging in age from 6.5 to 14.1 years, were enrolled upon receiving a new diagnosis of PKD, as defined by the clinical diagnostic criteria set by Bruno in 2004. Comprehensive evaluation of PRRT2 variants and 16p11.2 microdeletion was achieved using whole-exome sequencing (WES) and bioinformatics analysis of copy number variant (CNV) for all subjects. Immediately after diagnosis, participants commenced treatment with low-dose lamotrigine. Cognitive function was assessed using the Wechsler Intelligence Scale for Children-Chinese Revised (WISC-CR) at baseline and after 2 years, with ADHD diagnoses and symptom severity simultaneously assessed by experts in accordance with the DSM-IV diagnostic criteria for ADHD and the ADHD Rating Scale-IV (ADHD-RS-IV). Results Initially, twelve out of 31 patients (38.7%) presented with comorbid ADHD. The latency to treatment initiation was notably longer in PKD patients with ADHD (30.75 ± 12.88 months) than in those without ADHD (11.66 ± 9.08 months), t = 4.856, p<0.001. Notably, patients with a latency exceeding 2 years exhibited a heightened risk for comorbid ADHD (OR = 4.671, P=0.015) in comparison to those with shorter latency. Out of the cohort, twenty-five patients saw the clinical trial to its completion. These individuals demonstrated a marked elevation in WISC-CR scores at the 2-year mark relative to the outset across FSIQ (baseline mean: 108.72 ± 10.45 vs 24 months: 110.56 ± 10.03, p=0.001), VIQ (baseline mean: 109.44 ± 11.15 vs 24 months: 110.80 ± 10.44, p=0.028), and PIQ domains (baseline mean: 106.52 ± 9.74 vs 24 months: 108.24 ± 9.38, p=0.012). Concurrently, a substantial mitigation was observed in ADHD inattention at 2 years compared to baseline (p<0.001), with an average total subscale scores decrement from 9.04 ± 4.99 to 6.24 ± 4.05. Conclusion Prolonged duration of untreated PKD in children may elevate the risk of ADHD comorbidity. Notably, following a 2-year lamotrigine regimen, enhancements were observed in both cognitive test outcomes and ADHD symptomatology.
Collapse
Affiliation(s)
- Dong-dong You
- Department of Neonatology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yu-mei Huang
- Department of Neonatology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xiao-yu Wang
- Department of Pediatric Neurology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Wei Li
- Department of Pediatric Neurology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Feng Li
- Department of Pediatric Neurology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
14
|
You W, Li Q, Chen L, He N, Li Y, Long F, Wang Y, Chen Y, McNamara RK, Sweeney JA, DelBello MP, Gong Q, Li F. Common and distinct cortical thickness alterations in youth with autism spectrum disorder and attention-deficit/hyperactivity disorder. BMC Med 2024; 22:92. [PMID: 38433204 PMCID: PMC10910790 DOI: 10.1186/s12916-024-03313-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 02/22/2024] [Indexed: 03/05/2024] Open
Abstract
BACKGROUND Autism spectrum disorder (ASD) and attention-deficit/hyperactivity disorder (ADHD) are neurodevelopmental disorders with overlapping behavioral features and genetic etiology. While brain cortical thickness (CTh) alterations have been reported in ASD and ADHD separately, the degree to which ASD and ADHD are associated with common and distinct patterns of CTh changes is unclear. METHODS We searched PubMed, Web of Science, Embase, and Science Direct from inception to 8 December 2023 and included studies of cortical thickness comparing youth (age less than 18) with ASD or ADHD with typically developing controls (TDC). We conducted a comparative meta-analysis of vertex-based studies to identify common and distinct CTh alterations in ASD and ADHD. RESULTS Twelve ASD datasets involving 458 individuals with ASD and 10 ADHD datasets involving 383 individuals with ADHD were included in the analysis. Compared to TDC, ASD showed increased CTh in bilateral superior frontal gyrus, left middle temporal gyrus, and right superior parietal lobule (SPL) and decreased CTh in right temporoparietal junction (TPJ). ADHD showed decreased CTh in bilateral precentral gyri, right postcentral gyrus, and right TPJ relative to TDC. Conjunction analysis showed both disorders shared reduced TPJ CTh located in default mode network (DMN). Comparative analyses indicated ASD had greater CTh in right SPL and TPJ located in dorsal attention network and thinner CTh in right TPJ located in ventral attention network than ADHD. CONCLUSIONS These results suggest shared thinner TPJ located in DMN is an overlapping neurobiological feature of ASD and ADHD. This alteration together with SPL alterations might be related to altered biological motion processing in ASD, while abnormalities in sensorimotor systems may contribute to behavioral control problems in ADHD. The disorder-specific thinner TPJ located in disparate attention networks provides novel insight into distinct symptoms of attentional deficits associated with the two neurodevelopmental disorders. TRIAL REGISTRATION PROSPERO CRD42022370620. Registered on November 9, 2022.
Collapse
Affiliation(s)
- Wanfang You
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Lmaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China
- Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, 610041, Sichuan, People's Republic of China
- Department of Radiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, Zhejiang, People's Republic of China
| | - Qian Li
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Lmaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China
- Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, 610041, Sichuan, People's Republic of China
| | - Lizhou Chen
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Lmaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China
- Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, 610041, Sichuan, People's Republic of China
| | - Ning He
- Department of Psychiatry, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China
| | - Yuanyuan Li
- Department of Psychiatry, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China
| | - Fenghua Long
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Lmaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China
- Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, 610041, Sichuan, People's Republic of China
| | - Yaxuan Wang
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Lmaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China
- Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, 610041, Sichuan, People's Republic of China
| | - Yufei Chen
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Lmaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China
- Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, 610041, Sichuan, People's Republic of China
| | - Robert K McNamara
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, Cincinnati, OH, 45219, USA
| | - John A Sweeney
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Lmaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, Cincinnati, OH, 45219, USA
| | - Melissa P DelBello
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, Cincinnati, OH, 45219, USA
| | - Qiyong Gong
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Lmaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China
- Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, 610041, Sichuan, People's Republic of China
| | - Fei Li
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Lmaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China.
- Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, 610041, Sichuan, People's Republic of China.
| |
Collapse
|
15
|
Nashaat NH, Elrouby I, Zeidan HM, Kilany A, Abdelraouf ER, Hashish AF, Abdelhady HS, ElKeblawy MM, Shadi MS. Childhood Apraxia of Speech: Exploring Gluten Sensitivity and Changes in Glutamate and Gamma-Aminobutyric Acid Plasma Levels. Pediatr Neurol 2024; 151:104-110. [PMID: 38154236 DOI: 10.1016/j.pediatrneurol.2023.11.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 11/16/2023] [Accepted: 11/25/2023] [Indexed: 12/30/2023]
Abstract
BACKGROUND Individuals with childhood apraxia of speech (CAS) were reported to have genetic variations related to gluten sensitivity and some neuroanatomic changes, which could be associated with alterations in neurotransmitters levels such as glutamate and gamma-aminobutyric acid (GABA). The aim was to measure the levels of antigliadin immunoglobulin A (IgA) antibody, glutamate, and GABA in the plasma of children with CAS compared with children with delayed language development (DLD) and neurotypical (NT) children. METHODS The participants (N = 120) were in three groups: Group I for CAS (N = 30), Group II for DLD (N = 60), and Group III for NT (N = 30). The abilities of children in Groups I and II were evaluated. The plasma levels of antigliadin IgA, glutamate, and GABA were determined by enzyme-linked immunosorbent assay. RESULTS The intelligence quotient and expressive language age in Group I were low compared with Group II (P = 0.001; 0.004). The levels of antigliadin IgA and glutamate in Group I were higher compared with the other two groups, whereas the level of GABA was lower (P < 0.0001). An imbalance between glutamate and GABA was found in Group I. In Group II, no measures differed from NTs except lower GABA levels (P = 0.0007). CONCLUSIONS The elevated levels of antigliadin IgA antibody and glutamate demonstrated high sensitivity and specificity, differentiating children with CAS from children with DLD and NT children. The low levels of GABA contributed to the imbalance between the excitatory and inhibitory neurotransmitters' levels detected in children with CAS.
Collapse
Affiliation(s)
- Neveen Hassan Nashaat
- Children with Special Needs Research Department, Medical Research and Clinical Studies Institute, National Research Centre, Cairo, Egypt; Learning Disability and Neurorehabilitation Research Field, Medical Research Centre of Excellence, National Research Centre, Cairo, Egypt.
| | - Iman Elrouby
- Phoniatrics Department, Hearing and Speech Institute, Giza, Egypt
| | - Hala M Zeidan
- Children with Special Needs Research Department, Medical Research and Clinical Studies Institute, National Research Centre, Cairo, Egypt
| | - Ayman Kilany
- Children with Special Needs Research Department, Medical Research and Clinical Studies Institute, National Research Centre, Cairo, Egypt
| | - Ehab Ragaa Abdelraouf
- Children with Special Needs Research Department, Medical Research and Clinical Studies Institute, National Research Centre, Cairo, Egypt; Learning Disability and Neurorehabilitation Research Field, Medical Research Centre of Excellence, National Research Centre, Cairo, Egypt
| | - Adel F Hashish
- Children with Special Needs Research Department, Medical Research and Clinical Studies Institute, National Research Centre, Cairo, Egypt
| | - Hebatallah Sherif Abdelhady
- Children with Special Needs Research Department, Medical Research and Clinical Studies Institute, National Research Centre, Cairo, Egypt
| | - Mohamed M ElKeblawy
- Children with Special Needs Research Department, Medical Research and Clinical Studies Institute, National Research Centre, Cairo, Egypt
| | - Mariam S Shadi
- Unit of Phoniatrics, Otorhinolaryngology Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| |
Collapse
|
16
|
Xiao Y, Lanz B, Lim SI, Tkáč I, Xin L. Improved reproducibility of γ-aminobutyric acid measurement from short-echo-time proton MR spectroscopy by linewidth-matched basis sets in LCModel. NMR IN BIOMEDICINE 2024; 37:e5056. [PMID: 37839823 DOI: 10.1002/nbm.5056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 09/05/2023] [Accepted: 09/13/2023] [Indexed: 10/17/2023]
Abstract
γ-Aminobutyric acid (GABA), as the primary inhibitory neurotransmitter, is extremely important for maintaining healthy brain function, and deviations from GABA homeostasis are related to various brain diseases. Short-echo-time (short-TE) proton MR spectroscopy (1 H-MRS) has been employed to measure GABA concentration from various human brain regions at high magnetic fields. The aim of this study was to investigate the effect of spectral linewidth on GABA quantification and explore the application of an optimized basis-set preparation approach using a spectral-linewidth-matched (LM) basis set in LCModel to improve the reproducibility of GABA quantification from short-TE 1 H-MRS. In contrast to the fixed-linewidth basis-set approach, the LM basis-set preparation approach, where all metabolite basis spectra were simulated with a linewidth 4 Hz narrower than that of water, showed a smaller standard deviation of estimated GABA concentration from synthetic spectra with varying linewidths and lineshapes. The test-retest reproducibility was assessed by the mean within-subject coefficient of variation, which improved from 19.2% to 12.0% in the thalamus, from 27.9% to 14.9% in the motor cortex, and from 9.7% to 2.8% in the medial prefrontal cortex using LM basis sets at 7 T. We conclude that spectral linewidth has a large effect on GABA quantification from short-TE 1 H-MRS data and that using LM basis sets in LCModel can improve the reproducibility of GABA quantification.
Collapse
Affiliation(s)
- Ying Xiao
- Center for Biomedical Imaging (CIBM), Lausanne, Switzerland
- Animal Imaging and Technology, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Bernard Lanz
- Center for Biomedical Imaging (CIBM), Lausanne, Switzerland
- Animal Imaging and Technology, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Song-I Lim
- Center for Biomedical Imaging (CIBM), Lausanne, Switzerland
- Animal Imaging and Technology, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Ivan Tkáč
- Center for Magnetic Resonance Research, University of Minnesota, Minneapolis, Minnesota, USA
| | - Lijing Xin
- Center for Biomedical Imaging (CIBM), Lausanne, Switzerland
- Animal Imaging and Technology, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| |
Collapse
|
17
|
Liu H, Wang G, Zhang J, Lu B, Li D, Chen J. Inhalation of diesel exhaust particulate matter induces locomotor hyperactivity and its relationship with brain and gut metabolism. ENVIRONMENT INTERNATIONAL 2024; 183:108359. [PMID: 38056096 DOI: 10.1016/j.envint.2023.108359] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Revised: 11/21/2023] [Accepted: 11/28/2023] [Indexed: 12/08/2023]
Abstract
Diesel exhaust particulate matter (DEPM) are important components of urban air pollution worldwide. Recent studies proved that airborne DEPM can enter the human brain, which was associated with brain and mental diseases. In this study, we investigated the effects of DEPM exposure on behavior, and explored potential mechanisms from the perspective of metabolism in specific brain regions and short chain fatty acids (SCFAs) in the gut using mice. The results showed that inhalation of DEPM induced locomotor hyperactivity and a tendency for memory decline in mice. Exposure to DEPM disrupted motor behavior generation related cerebellar Purkinje cells, induced widespread reduction of neurotransmitters in the frontal cortex, and downregulated expression of genes encoding Brain-derived neurotrophic factor (BDNF) and involved in the Brain-blood-barrier (BBB) in the hippocampus. Moreover, there was a DEPM dose-dependent increase in fecal SCFA levels. Correlation analysis showed that DEPM-induced locomotor hyperactivity was mainly associated with decreased neurotransmission in the frontal cortex and increased gut SCFAs, and those associations were discussed. This study provides new insights into the mechanisms underpinning behavioral changes caused by air pollution, and extends our knowledge on the toxicity and health effects of airborne pollutants.
Collapse
Affiliation(s)
- Hou Liu
- Shanghai Key Laboratory of Atmospheric Particle Pollution and Prevention (LAP(3)), Department of Environmental Science and Engineering, Fudan University, Shanghai, 200433, China
| | - Guicheng Wang
- Institute of Developmental Biology and Molecular Medicine, Fudan University, Shanghai, 200433, China
| | - Jin Zhang
- Shanghai Key Laboratory of Atmospheric Particle Pollution and Prevention (LAP(3)), Department of Environmental Science and Engineering, Fudan University, Shanghai, 200433, China
| | - Bingjie Lu
- Shanghai Key Laboratory of Atmospheric Particle Pollution and Prevention (LAP(3)), Department of Environmental Science and Engineering, Fudan University, Shanghai, 200433, China
| | - Dan Li
- Shanghai Key Laboratory of Atmospheric Particle Pollution and Prevention (LAP(3)), Department of Environmental Science and Engineering, Fudan University, Shanghai, 200433, China.
| | - Jianmin Chen
- Shanghai Key Laboratory of Atmospheric Particle Pollution and Prevention (LAP(3)), Department of Environmental Science and Engineering, Fudan University, Shanghai, 200433, China
| |
Collapse
|
18
|
Imrie G, Gray MB, Raghuraman V, Farhy-Tselnicker I. Gene Expression at the Tripartite Synapse: Bridging the Gap Between Neurons and Astrocytes. ADVANCES IN NEUROBIOLOGY 2024; 39:95-136. [PMID: 39190073 DOI: 10.1007/978-3-031-64839-7_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/28/2024]
Abstract
Astrocytes, a major class of glial cells, are an important element at the synapse where they engage in bidirectional crosstalk with neurons to regulate numerous aspects of neurotransmission, circuit function, and behavior. Mutations in synapse-related genes expressed in both neurons and astrocytes are central factors in a vast number of neurological disorders, making the proteins that they encode prominent targets for therapeutic intervention. Yet, while the roles of many of these synaptic proteins in neurons are well established, the functions of the same proteins in astrocytes are largely unknown. This gap in knowledge must be addressed to refine therapeutic approaches. In this chapter, we integrate multiomic meta-analysis and a comprehensive overview of current literature to show that astrocytes express an astounding number of genes that overlap with the neuronal and synaptic transcriptomes. Further, we highlight recent reports that characterize the expression patterns and potential novel roles of these genes in astrocytes in both physiological and pathological conditions, underscoring the importance of considering both cell types when investigating the function and regulation of synaptic proteins.
Collapse
Affiliation(s)
- Gillian Imrie
- Department of Biology, Texas A&M University, College Station, TX, USA
| | - Madison B Gray
- Department of Biology, Texas A&M University, College Station, TX, USA
| | - Vishnuvasan Raghuraman
- Department of Biology, Texas A&M University, College Station, TX, USA
- Department of Computer Science and Engineering, Texas A&M University, College Station, TX, USA
| | - Isabella Farhy-Tselnicker
- Department of Biology, Texas A&M University, College Station, TX, USA.
- Texas A&M Institute for Neuroscience (TAMIN), Texas A&M University, College Station, TX, USA.
- Center for Biological Clocks Research, Texas A&M University, College Station, TX, USA.
| |
Collapse
|
19
|
Rabeh N, Hajjar B, Maraka JO, Sammanasunathan AF, Khan M, Alkhaaldi SMI, Mansour S, Almheiri RT, Hamdan H, Abd-Elrahman KS. Targeting mGluR group III for the treatment of neurodegenerative diseases. Biomed Pharmacother 2023; 168:115733. [PMID: 37862967 DOI: 10.1016/j.biopha.2023.115733] [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: 08/21/2023] [Revised: 10/11/2023] [Accepted: 10/13/2023] [Indexed: 10/22/2023] Open
Abstract
Glutamate, an excitatory neurotransmitter, is essential for neuronal function, and it acts on ionotropic or metabotropic glutamate receptors (mGluRs). A disturbance in glutamatergic signaling is a hallmark of many neurodegenerative diseases. Developing disease-modifying treatments for neurodegenerative diseases targeting glutamate receptors is a promising avenue. The understudied group III mGluR 4, 6-8 are commonly found in the presynaptic membrane, and their activation inhibits glutamate release. Thus, targeted mGluRs therapies could aid in treating neurodegenerative diseases. This review describes group III mGluRs and their pharmacological ligands in the context of amyotrophic lateral sclerosis, Parkinson's, Alzheimer's, and Huntington's diseases. Attempts to evaluate the efficacy of these drugs in clinical trials are also discussed. Despite a growing list of group III mGluR-specific pharmacological ligands, research on the use of these drugs in neurodegenerative diseases is limited, except for Parkinson's disease. Future efforts should focus on delineating the contribution of group III mGluR to neurodegeneration and developing novel ligands with superior efficacy and a favorable side effect profile for the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Nadia Rabeh
- Department of Physiology and Immunology, College of Medicine and Health Sciences, Khalifa University, Abu Dhabi 127788, United Arab Emirates; Department of Anesthesiology, Pharmacology and Therapeutics, and Djavad Mowafaghian Center for Brain Health, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada
| | - Baraa Hajjar
- Department of Physiology and Immunology, College of Medicine and Health Sciences, Khalifa University, Abu Dhabi 127788, United Arab Emirates
| | - Jude O Maraka
- Department of Physiology and Immunology, College of Medicine and Health Sciences, Khalifa University, Abu Dhabi 127788, United Arab Emirates
| | - Ashwin F Sammanasunathan
- Department of Physiology and Immunology, College of Medicine and Health Sciences, Khalifa University, Abu Dhabi 127788, United Arab Emirates
| | - Mohammed Khan
- Department of Physiology and Immunology, College of Medicine and Health Sciences, Khalifa University, Abu Dhabi 127788, United Arab Emirates
| | - Saif M I Alkhaaldi
- Department of Physiology and Immunology, College of Medicine and Health Sciences, Khalifa University, Abu Dhabi 127788, United Arab Emirates
| | - Samy Mansour
- Department of Physiology and Immunology, College of Medicine and Health Sciences, Khalifa University, Abu Dhabi 127788, United Arab Emirates
| | - Rashed T Almheiri
- Department of Physiology and Immunology, College of Medicine and Health Sciences, Khalifa University, Abu Dhabi 127788, United Arab Emirates
| | - Hamdan Hamdan
- Department of Physiology and Immunology, College of Medicine and Health Sciences, Khalifa University, Abu Dhabi 127788, United Arab Emirates; Healthcare Engineering Innovation Center (HEIC), Khalifa University of Science and Technology, Abu Dhabi 127788, United Arab Emirates
| | - Khaled S Abd-Elrahman
- Department of Anesthesiology, Pharmacology and Therapeutics, and Djavad Mowafaghian Center for Brain Health, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada; Department of Pharmacology and Therapeutics, College of Medicine and Health Science, Khalifa University, Abu Dhabi 127788, United Arab Emirates; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt.
| |
Collapse
|
20
|
Ou Y, Rots E, Belzer C, Smidt H, de Weerth C. Gut microbiota and child behavior in early puberty: does child sex play a role? Gut Microbes 2023; 15:2278222. [PMID: 37943628 PMCID: PMC10731618 DOI: 10.1080/19490976.2023.2278222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 10/27/2023] [Indexed: 11/12/2023] Open
Abstract
A growing number of studies have indicated relations between the gut microbiota and mental health. However, to date, there is a scarcity of microbiota studies in community samples in early puberty. The current preregistered study (https://osf.io/wu2vt) investigated gut microbiota composition in relation to sex in low-risk children and explored behavioral associations with gut microbiota composition and metabolites in the same samples, together with the potential role of sex. Fecal microbiota composition was analyzed in 12-year-old children (N = 137) by 16S rRNA gene sequencing and quantitative PCR. Modest sex differences were observed in beta diversity. Generalized linear models showed consistent behavioral relations to both relative and absolute abundances of individual taxa, including positive associations between Parasutterella and mother-reported internalizing behavior, and negative associations between Odoribacter and mother-reported externalizing behavior. Additionally, Prevotella 9 was positively related to mother-reported externalizing behavior, confirming earlier findings on the same cohort at 5 years of age. Sex-related differences were found in behavioral relations to Ruminiclostridium 5, Alistipes, Streptococcus, Ruminiclostridium 9, Ruminococcaceae UCG-5, and Dialister, for relative abundances, as well as to Family XIII AD3011 group and an unidentified bacterium within the Tenericutes, for absolute abundances. Limited behavioral relations were observed regarding alpha diversity and fecal metabolites. Our findings describe links between the gut microbiota and child behavior, together with differences between child sexes in these relations, in low-risk early pubertal children. Importantly, this study confirmed earlier findings in this cohort of positive relations between Prevotella 9 and externalizing behavior at age 10 years. Results also show the merit of including absolute abundances in microbiota studies.
Collapse
Affiliation(s)
- Yangwenshan Ou
- Laboratory of Microbiology, Wageningen University & Research, Wageningen, The Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Department of Cognitive Neuroscience, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Eline Rots
- Laboratory of Microbiology, Wageningen University & Research, Wageningen, The Netherlands
| | - Clara Belzer
- Laboratory of Microbiology, Wageningen University & Research, Wageningen, The Netherlands
| | - Hauke Smidt
- Laboratory of Microbiology, Wageningen University & Research, Wageningen, The Netherlands
| | - Carolina de Weerth
- Donders Institute for Brain, Cognition and Behaviour, Department of Cognitive Neuroscience, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
21
|
Zacharopoulos G, Sella F, Emir U, Cohen Kadosh R. Dissecting the chain of information processing and its interplay with neurochemicals and fluid intelligence across development. eLife 2023; 12:e84086. [PMID: 37772958 PMCID: PMC10541179 DOI: 10.7554/elife.84086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 08/23/2023] [Indexed: 09/30/2023] Open
Abstract
Previous research has highlighted the role of glutamate and gamma-aminobutyric acid (GABA) in perceptual, cognitive, and motor tasks. However, the exact involvement of these neurochemical mechanisms in the chain of information processing, and across human development, is unclear. In a cross-sectional longitudinal design, we used a computational approach to dissociate cognitive, decision, and visuomotor processing in 293 individuals spanning early childhood to adulthood. We found that glutamate and GABA within the intraparietal sulcus (IPS) explained unique variance in visuomotor processing, with higher glutamate predicting poorer visuomotor processing in younger participants but better visuomotor processing in mature participants, while GABA showed the opposite pattern. These findings, which were neurochemically, neuroanatomically and functionally specific, were replicated ~21 mo later and were generalized in two further different behavioral tasks. Using resting functional MRI, we revealed that the relationship between IPS neurochemicals and visuomotor processing is mediated by functional connectivity in the visuomotor network. We then extended our findings to high-level cognitive behavior by predicting fluid intelligence performance. We present evidence that fluid intelligence performance is explained by IPS GABA and glutamate and is mediated by visuomotor processing. However, this evidence was obtained using an uncorrected alpha and needs to be replicated in future studies. These results provide an integrative biological and psychological mechanistic explanation that links cognitive processes and neurotransmitters across human development and establishes their potential involvement in intelligent behavior.
Collapse
Affiliation(s)
- George Zacharopoulos
- Wellcome Centre for Integrative Neuroimaging, Department of Experimental Psychology, University of OxfordOxfordUnited Kingdom
- School of Psychology, Swansea UniversitySwanseaUnited Kingdom
| | - Francesco Sella
- Wellcome Centre for Integrative Neuroimaging, Department of Experimental Psychology, University of OxfordOxfordUnited Kingdom
- Centre for Mathematical Cognition, Loughborough UniversityLoughboroughUnited Kingdom
| | - Uzay Emir
- Wellcome Centre for Integrative Neuroimaging, Department of Experimental Psychology, University of OxfordOxfordUnited Kingdom
- School of Health Sciences, College of Health and Human Sciences, Purdue UniversityWest LafayetteUnited States
| | - Roi Cohen Kadosh
- Wellcome Centre for Integrative Neuroimaging, Department of Experimental Psychology, University of OxfordOxfordUnited Kingdom
- School of Psychology, University of SurreyGuildfordUnited Kingdom
| |
Collapse
|
22
|
Kuś J, Saramowicz K, Czerniawska M, Wiese W, Siwecka N, Rozpędek-Kamińska W, Kucharska-Lusina A, Strzelecki D, Majsterek I. Molecular Mechanisms Underlying NMDARs Dysfunction and Their Role in ADHD Pathogenesis. Int J Mol Sci 2023; 24:12983. [PMID: 37629164 PMCID: PMC10454781 DOI: 10.3390/ijms241612983] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 08/17/2023] [Accepted: 08/18/2023] [Indexed: 08/27/2023] Open
Abstract
Attention deficit hyperactivity disorder (ADHD) is one of the most common neurodevelopmental disorders, although the aetiology of ADHD is not yet understood. One proposed theory for developing ADHD is N-methyl-D-aspartate receptors (NMDARs) dysfunction. NMDARs are involved in regulating synaptic plasticity and memory function in the brain. Abnormal expression or polymorphism of some genes associated with ADHD results in NMDAR dysfunction. Correspondingly, NMDAR malfunction in animal models results in ADHD-like symptoms, such as impulsivity and hyperactivity. Currently, there are no drugs for ADHD that specifically target NMDARs. However, NMDAR-stabilizing drugs have shown promise in improving ADHD symptoms with fewer side effects than the currently most widely used psychostimulant in ADHD treatment, methylphenidate. In this review, we outline the molecular and genetic basis of NMDAR malfunction and how it affects the course of ADHD. We also present new therapeutic options related to treating ADHD by targeting NMDAR.
Collapse
Affiliation(s)
- Justyna Kuś
- Department of Clinical Chemistry and Biochemistry, Medical University of Lodz, Mazowiecka 5, 92-215 Lodz, Poland; (J.K.); (K.S.); (M.C.); (W.W.); (N.S.); (W.R.-K.); (A.K.-L.)
| | - Kamil Saramowicz
- Department of Clinical Chemistry and Biochemistry, Medical University of Lodz, Mazowiecka 5, 92-215 Lodz, Poland; (J.K.); (K.S.); (M.C.); (W.W.); (N.S.); (W.R.-K.); (A.K.-L.)
| | - Maria Czerniawska
- Department of Clinical Chemistry and Biochemistry, Medical University of Lodz, Mazowiecka 5, 92-215 Lodz, Poland; (J.K.); (K.S.); (M.C.); (W.W.); (N.S.); (W.R.-K.); (A.K.-L.)
| | - Wojciech Wiese
- Department of Clinical Chemistry and Biochemistry, Medical University of Lodz, Mazowiecka 5, 92-215 Lodz, Poland; (J.K.); (K.S.); (M.C.); (W.W.); (N.S.); (W.R.-K.); (A.K.-L.)
| | - Natalia Siwecka
- Department of Clinical Chemistry and Biochemistry, Medical University of Lodz, Mazowiecka 5, 92-215 Lodz, Poland; (J.K.); (K.S.); (M.C.); (W.W.); (N.S.); (W.R.-K.); (A.K.-L.)
| | - Wioletta Rozpędek-Kamińska
- Department of Clinical Chemistry and Biochemistry, Medical University of Lodz, Mazowiecka 5, 92-215 Lodz, Poland; (J.K.); (K.S.); (M.C.); (W.W.); (N.S.); (W.R.-K.); (A.K.-L.)
| | - Aleksandra Kucharska-Lusina
- Department of Clinical Chemistry and Biochemistry, Medical University of Lodz, Mazowiecka 5, 92-215 Lodz, Poland; (J.K.); (K.S.); (M.C.); (W.W.); (N.S.); (W.R.-K.); (A.K.-L.)
| | - Dominik Strzelecki
- Department of Affective and Psychotic Disorders, Medical University of Lodz, Czechoslowacka 8/10, 92-216 Lodz, Poland;
| | - Ireneusz Majsterek
- Department of Clinical Chemistry and Biochemistry, Medical University of Lodz, Mazowiecka 5, 92-215 Lodz, Poland; (J.K.); (K.S.); (M.C.); (W.W.); (N.S.); (W.R.-K.); (A.K.-L.)
| |
Collapse
|
23
|
Liu Y, Yang C, Meng Y, Dang Y, Yang L. Ketogenic diet ameliorates attention deficit hyperactivity disorder in rats via regulating gut microbiota. PLoS One 2023; 18:e0289133. [PMID: 37585373 PMCID: PMC10431618 DOI: 10.1371/journal.pone.0289133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 07/11/2023] [Indexed: 08/18/2023] Open
Abstract
Attention deficit hyperactivity disorder (ADHD) is a common mental behavioral disorder in children. Alterations in gut microbiota composition are associated with neurological disorders. We aimed to investigate whether a ketogenic diet (KD) can be an alternative therapy for ADHD by altering the gut microbiota. Male spontaneously hypertensive rats (SHR) and Wistar Kyoto (WKY) rats were randomly allocated to the normal diet (ND), methylphenidate (MPH), and KD groups. SHR in groups KD and MPH exhibited a significant increase in behavioral characteristics of ADHD, such as distance moved and immobility time. KD and MPH treatment led to a significant elevation in concentrations of 5-HT, AC, cAMP, and NE of brain tissue and the expression of DRD1, DAT, PKA, DARPP32, and cAMP at the protein level in WKY rats and SHR. KD and MPH significantly increased the richness and diversity of gut microbiota in SHR. The abundance of Ruminococcus_gauvreauii_group, Bacteroides, Bifidobacterium, and Blautia significantly increased, whereas that of Lactobacillus, Romboutsia, Facklamia, and Turicibacter significantly declined in the KD group compared with the ND group. The gut microbiota in the KD group of SHR mainly participated in amino acid metabolism- and sugar metabolism-related pathways. KD might alleviate behavioral disorders in ADHD by regulating gut microbiota. This study provides novel insights for the use of KD in treating ADHD.
Collapse
Affiliation(s)
- Yu Liu
- Department of Pediatrics, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Changhong Yang
- Department of Pediatrics, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Yingxue Meng
- Department of Pediatrics, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Yonghui Dang
- College of Medicine and Forensics, Xi’an Jiaotong University Health Science Center, Xi’an, Shaanxi, China
| | - Lin Yang
- Department of Pediatrics, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| |
Collapse
|
24
|
Gutiérrez-Casares JR, Quintero J, Segú-Vergés C, Rodríguez Monterde P, Pozo-Rubio T, Coma M, Montoto C. In silico clinical trial evaluating lisdexamfetamine's and methylphenidate's mechanism of action computational models in an attention-deficit/hyperactivity disorder virtual patients' population. Front Psychiatry 2023; 14:939650. [PMID: 37333910 PMCID: PMC10273406 DOI: 10.3389/fpsyt.2023.939650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 04/21/2023] [Indexed: 06/20/2023] Open
Abstract
Introduction Attention-deficit/hyperactivity disorder (ADHD) is an impairing psychiatric condition with the stimulants, lisdexamfetamine (LDX), and methylphenidate (MPH), as the first lines pharmacological treatment. Methods Herein, we applied a novel in silico method to evaluate virtual LDX (vLDX) and vMPH as treatments for ADHD applying quantitative systems pharmacology (QSP) models. The objectives were to evaluate the model's output, considering the model characteristics and the information used to build them, to compare both virtual drugs' efficacy mechanisms, and to assess how demographic (age, body mass index, and sex) and clinical characteristics may affect vLDX's and vMPH's relative efficacies. Results and Discussion We molecularly characterized the drugs and pathologies based on a bibliographic search, and generated virtual populations of adults and children-adolescents totaling 2,600 individuals. For each virtual patient and virtual drug, we created physiologically based pharmacokinetic and QSP models applying the systems biology-based Therapeutic Performance Mapping System technology. The resulting models' predicted protein activity indicated that both virtual drugs modulated ADHD through similar mechanisms, albeit with some differences. vMPH induced several general synaptic, neurotransmitter, and nerve impulse-related processes, whereas vLDX seemed to modulate neural processes more specific to ADHD, such as GABAergic inhibitory synapses and regulation of the reward system. While both drugs' models were linked to an effect over neuroinflammation and altered neural viability, vLDX had a significant impact on neurotransmitter imbalance and vMPH on circadian system deregulation. Among demographic characteristics, age and body mass index affected the efficacy of both virtual treatments, although the effect was more marked for vLDX. Regarding comorbidities, only depression negatively impacted both virtual drugs' efficacy mechanisms and, while that of vLDX were more affected by the co-treatment of tic disorders, the efficacy mechanisms of vMPH were disturbed by wide-spectrum psychiatric drugs. Our in silico results suggested that both drugs could have similar efficacy mechanisms as ADHD treatment in adult and pediatric populations and allowed raising hypotheses for their differential impact in specific patient groups, although these results require prospective validation for clinical translatability.
Collapse
Affiliation(s)
- José Ramón Gutiérrez-Casares
- Unidad Ambulatoria de Psiquiatría y Salud Mental de la Infancia, Niñez y Adolescencia, Hospital Perpetuo Socorro, Badajoz, Spain
| | - Javier Quintero
- Servicio de Psiquiatría, Hospital Universitario Infanta Leonor, Universidad Complutense, Madrid, Spain
| | - Cristina Segú-Vergés
- Anaxomics Biotech, Barcelona, Spain
- Structural Bioinformatics Group, Research Programme on Biomedical Informatics, Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | | | | | | | - Carmen Montoto
- Medical Department, Takeda Farmacéutica España, Madrid, Spain
| |
Collapse
|
25
|
Pang EW, Hammill C, Taylor MJ, Near J, Schachar R, Crosbie J, Arnold PD, Anagnostou E, Lerch JP. Cerebellar gamma-aminobutyric acid: Investigation of group effects in neurodevelopmental disorders. Autism Res 2023; 16:535-542. [PMID: 36626308 DOI: 10.1002/aur.2888] [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: 03/23/2022] [Accepted: 12/24/2022] [Indexed: 01/11/2023]
Abstract
Neurodevelopmental disorders (NDDs) including autism spectrum disorder (ASD), attention-deficit hyperactivity disorder (ADHD) and obsessive-compulsive disorder (OCD) are thought to arise in part from the disruption in the excitatory/inhibitory balance of gamma-aminobutyric acid (GABA) and glutamate in the brain. Recent evidence has shown the involvement of the cerebellum in cognition and affect regulation, and cerebellar atypical function or damage is reported frequently in NDDs. Magnetic resonance spectroscopy studies have reported decreases in GABA in cortical brain areas in the NDDs, however, GABA levels in the cerebellum have not been examined. To determine possible group effects, we used a MEGA-PRESS acquisition to investigate GABA+ levels in a cerebellar voxel in 343 individuals (aged 2.5-22 years) with ASD, ADHD, OCD and controls. Using a mixed effects model, we found no significant differences between groups in GABA+ concentration. Our findings suggest that cerebellar GABA+ levels do not differentiate NDD groups.
Collapse
Affiliation(s)
- Elizabeth W Pang
- Division of Neurology/Neuroscience and Mental Health, Hospital for Sick Children, Toronto, Canada
| | - Chris Hammill
- Mouse Imaging Centre, The Hospital for Sick Children, Toronto, Canada
| | - Margot J Taylor
- Diagnostic Imaging/Neuroscience and Mental Health, Hospital for Sick Children, Toronto and Departments of Medical Imaging and Psychology, University of Toronto, Toronto, Canada
| | - Jamie Near
- Physical Sciences, Sunnybrook Research Institute, Toronto, Canada
| | - Russell Schachar
- Department of Psychiatry/Neuroscience and Mental Health, Hospital for Sick Children, Toronto, Canada
| | - Jennifer Crosbie
- Department of Psychiatry/Neuroscience and Mental Health, Hospital for Sick Children, Toronto, Canada
| | - Paul D Arnold
- The Mathison Centre for Mental Health Research and Education, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Canada
| | - Evdokia Anagnostou
- Autism Research Centre, Holland Bloorview Kids Rehabilitation Hospital, Toronto, Canada
| | - Jason P Lerch
- Mouse Imaging Centre, The Hospital for Sick Children, Toronto, Canada.,Wellcome Centre for Integrative Neuroimaging, FMRIB, Nuffield Department of Clinical Neuroscience, The University of Oxford, Oxford, UK
| |
Collapse
|
26
|
Event-related potentials during mental rotation of body-related stimuli in spinal cord injury population. Neuropsychologia 2023; 179:108447. [PMID: 36521630 DOI: 10.1016/j.neuropsychologia.2022.108447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 11/13/2022] [Accepted: 12/10/2022] [Indexed: 12/14/2022]
Abstract
Mental rotations of body-related stimuli are known to engage the motor system and activate body schema. Sensorimotor deficits following spinal cord injury (SCI) alter the representation of the body with a negative impact on the performance during motor-related tasks, such as mental rotation of body parts. Here we investigated the relationship between event-related potentials in SCI participants and the difficulty in mentally rotating a body-part. Participants with SCI and healthy control subjects performed a laterality judgment task, in which left or right images of hands, feet or animals (as a control stimulus) were presented in two different orientation angles (75° and 150°), and participants reported the laterality of the stimulus. We found that reaction times of participants with SCI were slower for the rotation of body-related stimuli compared to non-body-related stimuli and healthy controls. At the brain level, we found that relative to healthy controls SCI participants show: 1) reduced amplitudes of the posterior P100 and anterior N100 and larger amplitudes of the anterior P200 for overall stimuli; 2) an absence of the modulation of the rotation related negativity by stimulus type and rotation angles. Our results show that body representation changes after SCI affecting both components of early stimulus processing and late components that process high-order cognitive aspects of body-representation and task complexity.
Collapse
|
27
|
da Silva BS, Grevet EH, Silva LCF, Ramos JKN, Rovaris DL, Bau CHD. An overview on neurobiology and therapeutics of attention-deficit/hyperactivity disorder. DISCOVER MENTAL HEALTH 2023; 3:2. [PMID: 37861876 PMCID: PMC10501041 DOI: 10.1007/s44192-022-00030-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Accepted: 12/29/2022] [Indexed: 10/21/2023]
Abstract
Attention-Deficit/Hyperactivity Disorder (ADHD) is a prevalent psychiatric condition characterized by developmentally inappropriate symptoms of inattention and/or hyperactivity/impulsivity, which leads to impairments in the social, academic, and professional contexts. ADHD diagnosis relies solely on clinical assessment based on symptom evaluation and is sometimes challenging due to the substantial heterogeneity of the disorder in terms of clinical and pathophysiological aspects. Despite the difficulties imposed by the high complexity of ADHD etiology, the growing body of research and technological advances provide good perspectives for understanding the neurobiology of the disorder. Such knowledge is essential to refining diagnosis and identifying new therapeutic options to optimize treatment outcomes and associated impairments, leading to improvements in all domains of patient care. This review is intended to be an updated outline that addresses the etiological and neurobiological aspects of ADHD and its treatment, considering the impact of the "omics" era on disentangling the multifactorial architecture of ADHD.
Collapse
Affiliation(s)
- Bruna Santos da Silva
- ADHD and Developmental Psychiatry Programs, Hospital de Clínicas de Porto Alegre, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, Brazil
- Department of Genetics and Graduate Program in Genetics and Molecular Biology, Instituto de Biociências, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, Brazil
- Department of Physiology and Biophysics, Instituto de Ciencias Biomedicas Universidade de Sao Paulo, São Paulo, Brazil
- Laboratory of Physiological Genomics of Mental Health (PhysioGen Lab), Institute of Biomedical Sciences, University of Sao Paulo, São Paulo, Brazil
| | - Eugenio Horacio Grevet
- ADHD and Developmental Psychiatry Programs, Hospital de Clínicas de Porto Alegre, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, Brazil
- Department of Psychiatry and Graduate Program in Psychiatry and Behavioral Sciences, Faculdade de Medicina, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, Brazil
| | - Luiza Carolina Fagundes Silva
- ADHD and Developmental Psychiatry Programs, Hospital de Clínicas de Porto Alegre, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, Brazil
- Department of Psychiatry and Graduate Program in Psychiatry and Behavioral Sciences, Faculdade de Medicina, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, Brazil
| | - João Kleber Neves Ramos
- Department of Physiology and Biophysics, Instituto de Ciencias Biomedicas Universidade de Sao Paulo, São Paulo, Brazil
- Laboratory of Physiological Genomics of Mental Health (PhysioGen Lab), Institute of Biomedical Sciences, University of Sao Paulo, São Paulo, Brazil
| | - Diego Luiz Rovaris
- Department of Physiology and Biophysics, Instituto de Ciencias Biomedicas Universidade de Sao Paulo, São Paulo, Brazil
- Laboratory of Physiological Genomics of Mental Health (PhysioGen Lab), Institute of Biomedical Sciences, University of Sao Paulo, São Paulo, Brazil
| | - Claiton Henrique Dotto Bau
- ADHD and Developmental Psychiatry Programs, Hospital de Clínicas de Porto Alegre, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, Brazil.
- Department of Genetics and Graduate Program in Genetics and Molecular Biology, Instituto de Biociências, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, Brazil.
- Department of Psychiatry and Graduate Program in Psychiatry and Behavioral Sciences, Faculdade de Medicina, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, Brazil.
| |
Collapse
|
28
|
Chen X, Gong T, Chen T, Xu C, Li Y, Song Q, Lin L, Oeltzschner G, Edden RAE, Xia Z, Wang G. Altered glutamate-glutamine and amide proton transfer-weighted values in the hippocampus of patients with amnestic mild cognitive impairment: A novel combined imaging diagnostic marker. Front Neurosci 2023; 17:1089300. [PMID: 36908797 PMCID: PMC9995585 DOI: 10.3389/fnins.2023.1089300] [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: 11/04/2022] [Accepted: 01/30/2023] [Indexed: 02/25/2023] Open
Abstract
Background and purpose Early diagnosis of amnestic mild cognitive impairment (aMCI) and timely management to delay the onset of Alzheimer's disease (AD) would benefit patients. Pathological metabolic changes of excitatory/inhibitory neurotransmitters and abnormal protein deposition in the hippocampus of aMCI may provide a new clue to imaging diagnosis. However, the diagnostic performance using these hippocampal metabolite measurements is still unclear. We aimed to quantify right hippocampal glutamate-glutamine (Glx) and gamma-aminobutyric acid (GABA) levels as well as protein-based amide proton transfer-weighted (APTw) signals of patients with aMCI and investigate the diagnostic performance of these metabolites. Methods In this cross-sectional study, 20 patients with aMCI and 20 age- and gender-matched healthy controls (HCs) underwent MEGA Point Resolved Spectroscopy (MEGA-PRESS) and APTw MR imaging at 3 T. GABA+, Glx, and APTw signals were measured in the right hippocampus. The GABA+ levels, Glx levels, Glx/GABA+ ratios, and APTw values were compared between the HCs and aMCI groups using the Mann-Whitney U test. Binary logistic regression and receiver operating characteristic (ROC) curve analyses were used to evaluate MEGA-PRESS and APTw parameters' diagnostic performance. Results Compared with HCs, patients with aMCI had significantly lower Glx levels in the right hippocampus (7.02 ± 1.41 i.u. vs. 5.81 ± 1.33 i.u., P = 0.018). No significant changes in the GABA+ levels were observed in patients with aMCI (HCs vs. aMCI: 2.54 ± 0.28 i.u. vs. 2.47 ± 0.36 i.u., P = 0.620). In addition, Glx/GABA+ ratios between the two groups (HCs vs. aMCI: 2.79 ± 0.60 vs. 2.37 ± 0.55, P = 0.035) were significantly different. Compared with HCs, patients with aMCI showed higher APTw values in the right hippocampus (0.99 ± 0.26% vs. 1.26% ± 0.28, P = 0.006). The ROC curve analysis showed that Glx, GABA+, Glx/GABA+, and APTw values had an area under the curve (AUC) of 0.72, 0.55, 0.70, and 0.75, respectively, for diagnosing aMCI. In the ROC curve analysis, the AUC of the combination of the parameters increased to 0.88, which is much higher than that observed in the univariate analysis (P < 0.05). Conclusion The combination of right hippocampal Glx levels and APTw values improved the diagnostic performance for aMCI, indicating it as a promising combined imaging diagnostic marker. Our study provided a potential imaging diagnostic strategy of aMCI, which may promote early detection of aMCI and facilitate timely intervention to delay the pathological progress toward AD.
Collapse
Affiliation(s)
- Xin Chen
- Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China.,Department of Neurology, Liaocheng People's Hospital, Liaocheng, China.,Department of Radiology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Tao Gong
- Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China.,Department of Radiology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Tong Chen
- Department of Radiology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Changyuan Xu
- Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Yuchao Li
- Department of Ultrasound, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Qingxu Song
- Department of Radiation Oncology, Qilu Hospital of Shandong University, Jinan, China
| | | | - Georg Oeltzschner
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, United States.,F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD, United States
| | - Richard A E Edden
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, United States.,F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD, United States
| | - Zhangyong Xia
- Department of Neurology, Liaocheng People's Hospital, Liaocheng, China.,Department of Neurology, Liaocheng Clinical School of Shandong First Medical University, Liaocheng, China
| | - Guangbin Wang
- Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China.,Department of Radiology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| |
Collapse
|
29
|
Bakhshi S, Tehrani-Doost M, Batouli SAH. Evaluation of fronto-cerebellar neurometabolites in youth with ADHD compared to the healthy group and their associations with cognitive and behavioral characteristics: A proton magnetic spectroscopy study. Int J Psychophysiol 2022; 182:190-199. [DOI: 10.1016/j.ijpsycho.2022.10.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 10/10/2022] [Accepted: 10/12/2022] [Indexed: 11/06/2022]
|
30
|
Emerging findings of glutamate-glutamine imbalance in the medial prefrontal cortex in attention deficit/hyperactivity disorder: systematic review and meta-analysis of spectroscopy studies. Eur Arch Psychiatry Clin Neurosci 2022; 272:1395-1411. [PMID: 35322293 DOI: 10.1007/s00406-022-01397-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Accepted: 03/01/2022] [Indexed: 12/11/2022]
Abstract
One of the main challenges in investigating the neurobiology of ADHD is our limited capacity to study its neurochemistry in vivo. Magnetic resonance spectroscopy (MRS) estimates metabolite concentrations within the brain, but approaches and findings have been heterogeneous. To assess differences in brain metabolites between patients with ADHD and healthy controls, we searched 12 databases screening for MRS studies. Studies were divided into 'children and adolescents' and 'adults' and meta-analyses were performed for each brain region with more than five studies. The quality of studies was assessed by the Newcastle-Ottawa Scale. Thirty-three studies met our eligibility criteria, including 874 patients with ADHD. Primary analyses revealed that the right medial frontal area of children with ADHD presented higher concentrations of a composite of glutamate and glutamine (p = 0.02, SMD = 0.53). Glutamate might be implicated in pruning and neurodegenerative processes as an excitotoxin, while glutamine excess might signal a glutamate depletion that could hinder neurotrophic activity. Both neuro metabolites could be implicated in the differential cortical thinning observed in patients with ADHD across all ages. Notably, more homogeneous designs and reporting guidelines are the key factors to determine how suitable MRS is for research and, perhaps, for clinical psychiatry. Results of this meta-analysis provided an overall map of the brain regions evaluated so far, addressed the role of glutamatergic metabolites in the pathophysiology of ADHD, and pointed to new perspectives for consistent use of the tool in the field.
Collapse
|
31
|
Attention-Deficit/Hyperactivity Disorder and the Gut Microbiota–Gut–Brain Axis: Closing Research Gaps through Female Inclusion in Study Design. WOMEN 2022. [DOI: 10.3390/women2030023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The gastrointestinal tract harbors a densely populated community of microbes that exhibits sexual dimorphism. Dysbiosis of this community has been associated with chronic human disease states ranging from metabolic diseases to neuropsychiatric disorders (NPDs). The gut microbiota–gut–brain axis (GMGBA) is a bi-directional pathway that facilitates the interaction of the gut microflora with host physiological functions. Recently, research surrounding the potential roles of the GMGBA in the development of NPDs (e.g., depression, anxiety, and autism spectrum disorders (ASDs)) has increased. However, the role of the GMGBA in attention-deficit/hyperactivity disorder (ADHD), an NPD that affects an estimated 8.4% of children (5.1% of female and 11.5% of male children) and 4% of adults (with a male–female odds ratio of 1.6) in the United States, remains understudied. Herein, we synthesize the current literature regarding the GMGBA, ADHD, and the potentially relevant intersections between the GMGBA and ADHD. Recommendations are presented for pathways of future research into the role(s) of the GMGBA in ADHD etiology and symptomatology. Particular focus is given to the potential for the variable of host sex to act as an outcome modifier of the relationship between the GMGBA and ADHD.
Collapse
|
32
|
Quintero J, Gutiérrez-Casares JR, Álamo C. Molecular Characterisation of the Mechanism of Action of Stimulant Drugs Lisdexamfetamine and Methylphenidate on ADHD Neurobiology: A Review. Neurol Ther 2022; 11:1489-1517. [PMID: 35951288 DOI: 10.1007/s40120-022-00392-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 07/28/2022] [Indexed: 10/15/2022] Open
Abstract
Attention-deficit/hyperactivity disorder (ADHD) is a common childhood-onset neurodevelopmental disorder characterised by persistent inattention, hyperactivity and impulsivity. Moreover, ADHD is commonly associated with other comorbid diseases (depression, anxiety, bipolar disorder, etc.). The ADHD symptomatology interferes with subject function and development. The treatment of ADHD requires a multidisciplinary approach based on a combination of non-pharmacological and pharmacological treatments with the aim of ameliorating the symptomatology; among first-line pharmacological treatments are stimulants [such as methylphenidate (MPH) and lisdexamfetamine dimesylate (LDX)]. In this review we explored recent ADHD- and stimulants-related literature, with the aim of compiling available descriptions of molecular pathways altered in ADHD, and molecular mechanisms of current first-line stimulants MPH and LDX. While conducting the narrative review, we applied structured search strategies covering PubMed/MEDLINE database and performed handsearching of reference lists on the results of those searches. The aetiology and pathophysiology of ADHD are incompletely understood; both genetic and environmental factors have been associated with the disorder and its grade of burden, and also the relationship between the molecular mechanisms of pharmacological treatments and their clinical implications. The lack of comprehensive understanding of the underlying molecular pathology makes both the diagnosis and treatment difficult. Few published studies evaluating molecular data on the mechanism of action (MoA) of MPH and LDX on ADHD are available and most of them are based on animal models. Further studies are necessary to improve the knowledge of ADHD pathophysiology and how the MoAs of MPH and LDX differentially modulate ADHD pathophysiology and control ADHD symptomatology.
Collapse
Affiliation(s)
- Javier Quintero
- Servicio de Psiquiatría y Salud Mental, Hospital Universitario Infanta Leonor, Universidad Complutense, Madrid, Spain
| | - José R Gutiérrez-Casares
- Unidad Ambulatoria de Psiquiatría y Salud Mental de la Infancia, Niñez y Adolescencia, Hospital Perpetuo Socorro, Badajoz, Spain.
| | - Cecilio Álamo
- Facultad de Medicina y Ciencias de la Salud, Universidad de Alcalá, Alcalá de Henares, Madrid, Spain
| |
Collapse
|
33
|
Zetterström TSC, Quansah E, Grootveld M. Effects of Methylphenidate on the Dopamine Transporter and Beyond. Curr Top Behav Neurosci 2022; 57:127-157. [PMID: 35507284 DOI: 10.1007/7854_2022_333] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The dopamine transporter (DAT) is the main target of methylphenidate (MPH), which remains the number one drug prescribed worldwide for the treatment of Attention-Deficit Hyperactivity Disorder (ADHD). In addition, abnormalities of the DAT have been widely associated with ADHD. Based on clinical and preclinical studies, the direction of DAT abnormalities in ADHD are, however, still unclear. Moreover, chronic treatment of MPH has been shown to increase brain DAT expression in both animals and ADHD patients, suggesting that findings of overexpressed levels of DAT in ADHD patients are possibly attributable to the effects of long-term MPH treatment rather than the pathology of the condition itself. In this chapter, we will discuss some of the effects exerted by MPH, which are related to its actions on catecholamine protein targets and brain metabolites, together with genes and proteins mediating neuronal plasticity. For this purpose, we present data from biochemical, proton nuclear magnetic resonance spectroscopy (1H-NMR) and gene/protein expression studies. Overall, results of the studies discussed in this chapter show that MPH has a complex biological/pharmacological action well beyond the DAT.
Collapse
Affiliation(s)
- Tyra S C Zetterström
- Pharmacology and Neuroscience Research Group, Leicester School of Pharmacy, Faculty of Health and Life Sciences, De Montfort University, Leicester, UK.
| | - Emmanuel Quansah
- Pharmacology and Neuroscience Research Group, Leicester School of Pharmacy, Faculty of Health and Life Sciences, De Montfort University, Leicester, UK
| | - Martin Grootveld
- Pharmacology and Neuroscience Research Group, Leicester School of Pharmacy, Faculty of Health and Life Sciences, De Montfort University, Leicester, UK
| |
Collapse
|
34
|
Investigating the Role of GABA in Neural Development and Disease Using Mice Lacking GAD67 or VGAT Genes. Int J Mol Sci 2022; 23:ijms23147965. [PMID: 35887307 PMCID: PMC9318753 DOI: 10.3390/ijms23147965] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 07/12/2022] [Accepted: 07/16/2022] [Indexed: 11/18/2022] Open
Abstract
Normal development and function of the central nervous system involves a balance between excitatory and inhibitory neurotransmission. Activity of both excitatory and inhibitory neurons is modulated by inhibitory signalling of the GABAergic and glycinergic systems. Mechanisms that regulate formation, maturation, refinement, and maintenance of inhibitory synapses are established in early life. Deviations from ideal excitatory and inhibitory balance, such as down-regulated inhibition, are linked with many neurological diseases, including epilepsy, schizophrenia, anxiety, and autism spectrum disorders. In the mammalian forebrain, GABA is the primary inhibitory neurotransmitter, binding to GABA receptors, opening chloride channels and hyperpolarizing the cell. We review the involvement of down-regulated inhibitory signalling in neurological disorders, possible mechanisms for disease progression, and targets for therapeutic intervention. We conclude that transgenic models of disrupted inhibitory signalling—in GAD67+/− and VGAT−/− mice—are useful for investigating the effects of down-regulated inhibitory signalling in a range of neurological diseases.
Collapse
|
35
|
Kahl CK, Swansburg R, Hai T, Wrightson JG, Bell T, Lemay JF, Kirton A, MacMaster FP. Differences in neurometabolites and transcranial magnetic stimulation motor maps in children with attention-deficit/hyperactivity disorder. J Psychiatry Neurosci 2022; 47:E239-E249. [PMID: 35793906 PMCID: PMC9262400 DOI: 10.1503/jpn.210186] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 03/01/2022] [Accepted: 03/04/2022] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND Although much is known about cognitive dysfunction in attention-deficit/hyperactivity disorder (ADHD), few studies have examined the pathophysiology of disordered motor circuitry. We explored differences in neurometabolite levels and transcranial magnetic stimulation (TMS)-derived corticomotor representations among children with ADHD and typically developing children. METHODS We used magnetic resonance spectroscopy (MRS) protocols to measure excitatory (glutamate + glutamine [Glx]) and inhibitory (γ-aminobutyric acid [GABA]) neurometabolite levels in the dominant primary motor cortex (M1) and the supplementary motor area (SMA) in children with ADHD and typically developing children. We used robotic neuronavigated TMS to measure corticospinal excitability and create corticomotor maps. RESULTS We collected data from 26 medication-free children with ADHD (aged 7-16 years) and 25 typically developing children (11-16 years). Children with ADHD had lower M1 Glx (p = 0.044, d = 0.6); their mean resting motor threshold was lower (p = 0.029, d = 0.8); their map area was smaller (p = 0.044, d = 0.7); and their hotspot density was higher (p = 0.008, d = 0.9). M1 GABA levels were associated with motor map area (p = 0.036).Limitations: Some TMS data were lost because the threshold of some children exceeded 100% of the machine output. The relatively large MRS voxel required to obtain sufficient signal-to-noise ratio and reliably measure GABA levels encompassed tissue beyond the M1, making this measure less anatomically specific. CONCLUSION The neurochemistry and neurophysiology of key nodes in the motor network may be altered in children with ADHD, and the differences appear to be related to each other. These findings suggest potentially novel neuropharmacological and neuromodulatory targets for ADHD.
Collapse
Affiliation(s)
- Cynthia K Kahl
- From the Department of Psychiatry, Cumming School of Medicine, University of Calgary, Calgary, Alta. (Kahl, Swansburg, MacMaster); the Department of Pediatrics, Cumming School of Medicine, University of Calgary, Calgary, Alta. (Kahl, Swansburg, Lemay, Kirton, MacMaster); the Hotchkiss Brain Institute, University of Calgary, Calgary, Alta. (Kahl, Wrightson, Bell, Kirton, MacMaster); the Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alta. (Kahl, Bell, Kirton, MacMaster); the Department of Educational Psychology, University of Alberta, Edmonton, Alta. (Hai); the Department of Radiology, Cumming School of Medicine, University of Calgary, Calgary, Alta. (Bell); the Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, Alta. (Kirton); and the Strategic Clinical Network for Addictions and Mental Health, Calgary, Alta. (MacMaster)
| | - Rose Swansburg
- From the Department of Psychiatry, Cumming School of Medicine, University of Calgary, Calgary, Alta. (Kahl, Swansburg, MacMaster); the Department of Pediatrics, Cumming School of Medicine, University of Calgary, Calgary, Alta. (Kahl, Swansburg, Lemay, Kirton, MacMaster); the Hotchkiss Brain Institute, University of Calgary, Calgary, Alta. (Kahl, Wrightson, Bell, Kirton, MacMaster); the Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alta. (Kahl, Bell, Kirton, MacMaster); the Department of Educational Psychology, University of Alberta, Edmonton, Alta. (Hai); the Department of Radiology, Cumming School of Medicine, University of Calgary, Calgary, Alta. (Bell); the Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, Alta. (Kirton); and the Strategic Clinical Network for Addictions and Mental Health, Calgary, Alta. (MacMaster)
| | - Tasmia Hai
- From the Department of Psychiatry, Cumming School of Medicine, University of Calgary, Calgary, Alta. (Kahl, Swansburg, MacMaster); the Department of Pediatrics, Cumming School of Medicine, University of Calgary, Calgary, Alta. (Kahl, Swansburg, Lemay, Kirton, MacMaster); the Hotchkiss Brain Institute, University of Calgary, Calgary, Alta. (Kahl, Wrightson, Bell, Kirton, MacMaster); the Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alta. (Kahl, Bell, Kirton, MacMaster); the Department of Educational Psychology, University of Alberta, Edmonton, Alta. (Hai); the Department of Radiology, Cumming School of Medicine, University of Calgary, Calgary, Alta. (Bell); the Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, Alta. (Kirton); and the Strategic Clinical Network for Addictions and Mental Health, Calgary, Alta. (MacMaster)
| | - James G Wrightson
- From the Department of Psychiatry, Cumming School of Medicine, University of Calgary, Calgary, Alta. (Kahl, Swansburg, MacMaster); the Department of Pediatrics, Cumming School of Medicine, University of Calgary, Calgary, Alta. (Kahl, Swansburg, Lemay, Kirton, MacMaster); the Hotchkiss Brain Institute, University of Calgary, Calgary, Alta. (Kahl, Wrightson, Bell, Kirton, MacMaster); the Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alta. (Kahl, Bell, Kirton, MacMaster); the Department of Educational Psychology, University of Alberta, Edmonton, Alta. (Hai); the Department of Radiology, Cumming School of Medicine, University of Calgary, Calgary, Alta. (Bell); the Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, Alta. (Kirton); and the Strategic Clinical Network for Addictions and Mental Health, Calgary, Alta. (MacMaster)
| | - Tiffany Bell
- From the Department of Psychiatry, Cumming School of Medicine, University of Calgary, Calgary, Alta. (Kahl, Swansburg, MacMaster); the Department of Pediatrics, Cumming School of Medicine, University of Calgary, Calgary, Alta. (Kahl, Swansburg, Lemay, Kirton, MacMaster); the Hotchkiss Brain Institute, University of Calgary, Calgary, Alta. (Kahl, Wrightson, Bell, Kirton, MacMaster); the Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alta. (Kahl, Bell, Kirton, MacMaster); the Department of Educational Psychology, University of Alberta, Edmonton, Alta. (Hai); the Department of Radiology, Cumming School of Medicine, University of Calgary, Calgary, Alta. (Bell); the Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, Alta. (Kirton); and the Strategic Clinical Network for Addictions and Mental Health, Calgary, Alta. (MacMaster)
| | - Jean-François Lemay
- From the Department of Psychiatry, Cumming School of Medicine, University of Calgary, Calgary, Alta. (Kahl, Swansburg, MacMaster); the Department of Pediatrics, Cumming School of Medicine, University of Calgary, Calgary, Alta. (Kahl, Swansburg, Lemay, Kirton, MacMaster); the Hotchkiss Brain Institute, University of Calgary, Calgary, Alta. (Kahl, Wrightson, Bell, Kirton, MacMaster); the Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alta. (Kahl, Bell, Kirton, MacMaster); the Department of Educational Psychology, University of Alberta, Edmonton, Alta. (Hai); the Department of Radiology, Cumming School of Medicine, University of Calgary, Calgary, Alta. (Bell); the Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, Alta. (Kirton); and the Strategic Clinical Network for Addictions and Mental Health, Calgary, Alta. (MacMaster)
| | - Adam Kirton
- From the Department of Psychiatry, Cumming School of Medicine, University of Calgary, Calgary, Alta. (Kahl, Swansburg, MacMaster); the Department of Pediatrics, Cumming School of Medicine, University of Calgary, Calgary, Alta. (Kahl, Swansburg, Lemay, Kirton, MacMaster); the Hotchkiss Brain Institute, University of Calgary, Calgary, Alta. (Kahl, Wrightson, Bell, Kirton, MacMaster); the Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alta. (Kahl, Bell, Kirton, MacMaster); the Department of Educational Psychology, University of Alberta, Edmonton, Alta. (Hai); the Department of Radiology, Cumming School of Medicine, University of Calgary, Calgary, Alta. (Bell); the Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, Alta. (Kirton); and the Strategic Clinical Network for Addictions and Mental Health, Calgary, Alta. (MacMaster)
| | - Frank P MacMaster
- From the Department of Psychiatry, Cumming School of Medicine, University of Calgary, Calgary, Alta. (Kahl, Swansburg, MacMaster); the Department of Pediatrics, Cumming School of Medicine, University of Calgary, Calgary, Alta. (Kahl, Swansburg, Lemay, Kirton, MacMaster); the Hotchkiss Brain Institute, University of Calgary, Calgary, Alta. (Kahl, Wrightson, Bell, Kirton, MacMaster); the Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alta. (Kahl, Bell, Kirton, MacMaster); the Department of Educational Psychology, University of Alberta, Edmonton, Alta. (Hai); the Department of Radiology, Cumming School of Medicine, University of Calgary, Calgary, Alta. (Bell); the Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, Alta. (Kirton); and the Strategic Clinical Network for Addictions and Mental Health, Calgary, Alta. (MacMaster)
| |
Collapse
|
36
|
Xiao W, Manyi G, Khaleghi A. Deficits in auditory and visual steady-state responses in adolescents with bipolar disorder. J Psychiatr Res 2022; 151:368-376. [PMID: 35551068 DOI: 10.1016/j.jpsychires.2022.04.041] [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: 09/26/2021] [Revised: 04/06/2022] [Accepted: 04/28/2022] [Indexed: 10/18/2022]
Abstract
BACKGROUND Many aspects of steady-state responses of the brain remain unclear in bipolar disorder (BD) due to the small number of auditory steady-state response (ASSR) studies and the lack of steady-state visual evoked potential (SSVEP) studies on this complex disorder. Therefore, we assessed the patterns of SSVEP and ASSR in adolescents with BD during an active task to detect possible deficits in these important brain responses compared to normal subjects. METHODS 27 adolescents with BD and 30 healthy adolescents were assessed in this study. The blinking background of the monitor presented at 15 Hz and the tone signal stimulation at 40 Hz evoked SSVEPs and ASSRs, respectively. The phase and amplitude of the steady-state responses were calculated in the auditory and visual conditions. RESULTS Patients exhibited a substantially worse performance in the motor control inhibition task during both auditory and visual modalities. Patients showed increased SSVEP amplitude and phase in the frontal region compared to control adolescents. Also, patients exhibited decreased ASSR amplitude in the prefrontal and increased ASSR amplitude in the right-frontal and centro-parietal areas compared to healthy adolescents. CONCLUSIONS impairments in the production and preservation of SSVEP and ASSR are evident in BD, implicating abnormalities in visual and auditory pathways. Neurophysiological deficits and worse performance in BD adolescents may imply that visual and auditory pathways cannot well transfer the pertinent information from arriving sensory data to the visual and auditory cortices, and the frontal cortex cannot well integrate incoming signals into a unified and coherent perceptual action.
Collapse
Affiliation(s)
- Wang Xiao
- School of Humanities and Management, Southwest Medical University, Luzhou City, Sichuan Province, 646000, China
| | - Gu Manyi
- School of Humanities and Management, Southwest Medical University, Luzhou City, Sichuan Province, 646000, China.
| | - Ali Khaleghi
- Psychiatry and Psychology Research Center, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
37
|
Shortland N, Portnoy J, McGarry P, Perliger A, Gordon T, Anastasio N. A Reinforcement Sensitivity Theory of Violent Extremist Propaganda: The Motivational Pathways Underlying Movement Toward and Away From Violent Extremist Action. Front Psychol 2022; 13:858392. [PMID: 35664206 PMCID: PMC9160867 DOI: 10.3389/fpsyg.2022.858392] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 04/19/2022] [Indexed: 11/13/2022] Open
Abstract
Anecdotal evidence supports than engaging with violent extremist content online facilitates the radicalization process. However, there is a consistent lack of empirically grounded research to provide insight into the psychological process through which this influence occurs (if at all). As such, most theories often fail to accommodate both the multifinality (the concept that many people are exposed to violent extremist material, yet never engage in violent extremism), and equifinality (the concept that people can view a range violent extremist content, yet all end up engaging in violent extremism) that naturally is observed in those who engage with violent extremist content online and those who engage in violent extremist behavior. This paper presents Reinforcement Sensitivity Theory (RST) as a theoretical framework to inform understanding of the process that governs the interaction between violent extremist material online and engaging with violent extremism. RST is a motivational theory which has been applied to a range of benevolent and deviant behaviors. Specifically, we argue that RST is suitable to explain the effect of violent extremist content online because (1) it outlines multiple differentiated motivational pathways that can account for multifinality and equifinality observed in those who engage in violent extremist behavior and (2) the extant neurological and psychophysiological research using RST provides a empirically supported framework for developing both research methods and verifiable hypotheses to advance our understanding of how, if at all, violent extremist content online contributes to the process of radicalization.
Collapse
Affiliation(s)
- Neil Shortland
- School of Criminology and Justice Studies, University of Massachusetts Lowell, Lowell, MA, United States
| | | | | | | | | | | |
Collapse
|
38
|
Transcranial Direct Current Stimulation as an Approach to Mitigate Neurodevelopmental Disorders Affecting Excitation/Inhibition Balance: Focus on Autism Spectrum Disorder, Schizophrenia, and Attention Deficit/Hyperactivity Disorder. J Clin Med 2022; 11:jcm11102839. [PMID: 35628965 PMCID: PMC9143428 DOI: 10.3390/jcm11102839] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 05/12/2022] [Accepted: 05/14/2022] [Indexed: 02/04/2023] Open
Abstract
Transcranial direct current stimulation (tDCS) has been proposed as a promising therapy for rehabilitation of neurodevelopmental disorders. In this review, we discuss studies on the impact of tDCS as a therapy for autism, schizophrenia, and attention deficit/hyperactivity disorder, as well as the tDCS' mechanism of action, and propose future paths of research to optimize tDCS treatment protocols. The mechanism underlying tDCS effects is the modulation of excitatory and/or inhibitory activity, making it a valuable tool for restoring the excitation/inhibition (E/I) balance which is disrupted in many neurodevelopmental disorders. Clinical studies have shown that tDCS therapy is well-tolerated by patients and seems to ameliorate behavior and cognitive functions. Alterations in early development of neuronal circuits lead to disruptions in brain activity in neurodevelopmental disorders. An increasing amount of research into the effects of tDCS on neuronal activity has provided a foundation for its use as a therapy for behavior and cognitive characteristics of neurodevelopmental disorders. Clinical studies show that tDCS appears to ameliorate behavioral and cognitive outcomes of patients with autism, schizophrenia, and attention deficit/hyperactivity disorder. More research is needed to understand the mechanisms of action of tDCS and to optimize treatment protocols.
Collapse
|
39
|
Hsu T, Chen M, Chu C, Tsai S, Bai Y, Su T, Chen T, Liang C. Attention deficit hyperactivity disorder and risk of migraine: A nationwide longitudinal study. Headache 2022; 62:634-641. [DOI: 10.1111/head.14306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 03/06/2022] [Accepted: 03/10/2022] [Indexed: 11/30/2022]
Affiliation(s)
- Tien‐Wei Hsu
- Department of Psychiatry Kaohsiung Veterans General Hospital Kaohsiung City Taiwan
| | - Mu‐Hong Chen
- Department of Psychiatry Taipei Veterans General Hospital Taipei Taiwan
- Department of Psychiatry College of Medicine National Yang Ming Chiao Tung University Taipei Taiwan
| | - Che‐Sheng Chu
- Department of Psychiatry Kaohsiung Veterans General Hospital Kaohsiung City Taiwan
| | - Shih‐Jen Tsai
- Department of Psychiatry Taipei Veterans General Hospital Taipei Taiwan
- Department of Psychiatry College of Medicine National Yang Ming Chiao Tung University Taipei Taiwan
| | - Ya‐Mei Bai
- Department of Psychiatry Taipei Veterans General Hospital Taipei Taiwan
- Department of Psychiatry College of Medicine National Yang Ming Chiao Tung University Taipei Taiwan
| | - Tung‐Ping Su
- Department of Psychiatry Taipei Veterans General Hospital Taipei Taiwan
- Department of Psychiatry College of Medicine National Yang Ming Chiao Tung University Taipei Taiwan
| | - Tzeng‐Ji Chen
- Department of Family Medicine Taipei Veterans General Hospital Taipei Taiwan
- Institute of Hospital and Health Care Administration National Yang Ming Chiao Tung University Taipei Taiwan
| | - Chih‐Sung Liang
- Department of Psychiatry Beitou Branch Tri‐Service General Hospital Taipei Taiwan
- Graduate Institute of Medical Sciences National Defense Medical Center Taipei Taiwan
| |
Collapse
|
40
|
Mamiya PC, Richards TL, Edden RAE, Lee AKC, Stein MA, Kuhl PK. Reduced Glx and GABA Inductions in the Anterior Cingulate Cortex and Caudate Nucleus Are Related to Impaired Control of Attention in Attention-Deficit/Hyperactivity Disorder. Int J Mol Sci 2022; 23:ijms23094677. [PMID: 35563067 PMCID: PMC9100027 DOI: 10.3390/ijms23094677] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 04/17/2022] [Accepted: 04/20/2022] [Indexed: 11/16/2022] Open
Abstract
Attention-deficit/hyperactivity disorder (ADHD) is a neurodevelopmental disorder that impairs the control of attention and behavioral inhibition in affected individuals. Recent genome-wide association findings have revealed an association between glutamate and GABA gene sets and ADHD symptoms. Consistently, people with ADHD show altered glutamate and GABA content in the brain circuitry that is important for attention control function. Yet, it remains unknown how glutamate and GABA content in the attention control circuitry change when people are controlling their attention, and whether these changes can predict impaired attention control in people with ADHD. To study these questions, we recruited 18 adults with ADHD (31-51 years) and 16 adults without ADHD (28-54 years). We studied glutamate + glutamine (Glx) and GABA content in the fronto-striatal circuitry while participants performed attention control tasks. We found that Glx and GABA concentrations at rest did not differ between participants with ADHD or without ADHD. However, while participants were performing the attention control tasks, participants with ADHD showed smaller Glx and GABA increases than participants without ADHD. Notably, smaller GABA increases in participants with ADHD significantly predicted their poor task performance. Together, these findings provide the first demonstration showing that attention control deficits in people with ADHD may be related to insufficient responses of the GABAergic system in the fronto-striatal circuitry.
Collapse
Affiliation(s)
- Ping C. Mamiya
- Institute for Learning & Brain Sciences, University of Washington, Seattle, WA 98195, USA;
- Correspondence:
| | - Todd L. Richards
- Department of Radiology, University of Washington, Seattle, WA 98195, USA;
| | - Richard A. E. Edden
- Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, MD 21205, USA;
| | - Adrian K. C. Lee
- Department of Speech and Hearing Sciences, University of Washington, Seattle, WA 98195, USA;
| | - Mark A. Stein
- Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, WA 98195, USA;
| | - Patricia K. Kuhl
- Institute for Learning & Brain Sciences, University of Washington, Seattle, WA 98195, USA;
| |
Collapse
|
41
|
Frenquelli R, Ratcliff M, Villar de Onis J, Fernandes M, Barros FC, Hirst JE, Papageorghiou AT, Kennedy SH, Villar J. Complex Perinatal Syndromes Affecting Early Human Growth and Development: Issues to Consider to Understand Their Aetiology and Postnatal Effects. Front Neurosci 2022; 16:856886. [PMID: 35509448 PMCID: PMC9058100 DOI: 10.3389/fnins.2022.856886] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 03/11/2022] [Indexed: 12/17/2022] Open
Abstract
Complex perinatal syndromes (CPS) affecting pregnancy and childhood, such as preterm birth, and intra- and extra-uterine growth restriction, have multiple, diverse contexts of complexity and interaction that determine the short- and long-term growth, health and development of all human beings. Early in life, genetically-guided somatic and cerebral development occurs alongside a psychism "in statu nascendi," with the neural structures subjected to the effects of the intra- and extra-uterine environments in preparation for optimal postnatal functioning. Different trajectories of fetal cranial and abdominal growth have been identified before 25 weeks' gestation, tracking differential growth and neurodevelopment at 2 years of age. Similarly, critical time-windows exist in the first 5-8 months of postnatal life because of interactions between the newborn and their environment, mother/care-givers and feeding practices. Understanding these complex relational processes requires abandoning classical, linear and mechanistic interpretations that are placed in rigid, artificial biological silos. Instead, we need to conduct longitudinal, interdisciplinary research and integrate the resulting new knowledge into clinical practice. An ecological-systemic approach is required to understand early human growth and development, based on a dynamic multidimensional process from the molecular or genomic level to the socio-economic-environmental context. For this, we need theoretical and methodological tools that permit a global understanding of CPS, delineating temporal trajectories and their conditioning factors, updated by the incorporation of new scientific discoveries. The potential to optimize human growth and development across chronological age and geographical locations - by implementing interventions or "treatments" during periods of greatest instability or vulnerability - should be recognized. Hence, it is imperative to take a holistic view of reproductive and perinatal issues, acknowledging at all levels the complexity and interactions of CPS and their sensitive periods, laying the foundations for further improvements in growth and development of populations, to maximize global human potential. We discuss here conceptual issues that should be considered for the development and implementation of such a strategy aimed at addressing the perinatal health problems of the new millenium.
Collapse
Affiliation(s)
- Roberto Frenquelli
- Master Program in Child Psychoanalysis and Neuropsychological, Developmental Psychology Unit, Faculty of Psychology, National University of Rosario, Rosario, Argentina
| | - Marc Ratcliff
- Faculty of Psychology and Educational Science, Centre Jean Piaget, University of Geneva, Geneva, Switzerland
| | - Jimena Villar de Onis
- Oxford Maternal & Perinatal Health Institute, Green Templeton College, University of Oxford, Oxford, United Kingdom
- Geneva Foundation for Medical Education and Research, Geneva, Switzerland
| | - Michelle Fernandes
- Oxford Maternal & Perinatal Health Institute, Green Templeton College, University of Oxford, Oxford, United Kingdom
- MRC Lifecourse Epidemiology Centre, Human Development and Health Academic Unit, Department of Paediatrics, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
- Nuffield Department of Women’s & Reproductive Health, University of Oxford, Oxford, United Kingdom
| | - Fernando C. Barros
- Post Graduate Course on Health in the Vital Cycle, Universidade Católica de Pelotas, Pelotas, Brazil
| | - Jane E. Hirst
- Nuffield Department of Women’s & Reproductive Health, University of Oxford, Oxford, United Kingdom
| | - Aris T. Papageorghiou
- Oxford Maternal & Perinatal Health Institute, Green Templeton College, University of Oxford, Oxford, United Kingdom
- Nuffield Department of Women’s & Reproductive Health, University of Oxford, Oxford, United Kingdom
| | - Stephen H. Kennedy
- Oxford Maternal & Perinatal Health Institute, Green Templeton College, University of Oxford, Oxford, United Kingdom
- Nuffield Department of Women’s & Reproductive Health, University of Oxford, Oxford, United Kingdom
| | - Jose Villar
- Oxford Maternal & Perinatal Health Institute, Green Templeton College, University of Oxford, Oxford, United Kingdom
- Nuffield Department of Women’s & Reproductive Health, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
42
|
Ostlund B, Donoghue T, Anaya B, Gunther KE, Karalunas SL, Voytek B, Pérez-Edgar KE. Spectral parameterization for studying neurodevelopment: How and why. Dev Cogn Neurosci 2022; 54:101073. [PMID: 35074579 PMCID: PMC8792072 DOI: 10.1016/j.dcn.2022.101073] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 12/07/2021] [Accepted: 01/15/2022] [Indexed: 12/12/2022] Open
Abstract
A growing body of literature suggests that the explicit parameterization of neural power spectra is important for the appropriate physiological interpretation of periodic and aperiodic electroencephalogram (EEG) activity. In this paper, we discuss why parameterization is an imperative step for developmental cognitive neuroscientists interested in cognition and behavior across the lifespan, as well as how parameterization can be readily accomplished with an automated spectral parameterization ("specparam") algorithm (Donoghue et al., 2020a). We provide annotated code for power spectral parameterization, via specparam, in Jupyter Notebook and R Studio. We then apply this algorithm to EEG data in childhood (N = 60; Mage = 9.97, SD = 0.95) to illustrate its utility for developmental cognitive neuroscientists. Ultimately, the explicit parameterization of EEG power spectra may help us refine our understanding of how dynamic neural communication contributes to normative and aberrant cognition across the lifespan. Data and annotated analysis code for this manuscript are available on GitHub as a supplement to the open-access specparam toolbox.
Collapse
Affiliation(s)
- Brendan Ostlund
- Department of Psychology, The Pennsylvania State University, USA.
| | - Thomas Donoghue
- Department of Cognitive Science, University of California, San Diego, USA
| | - Berenice Anaya
- Department of Psychology, The Pennsylvania State University, USA
| | - Kelley E Gunther
- Department of Psychology, The Pennsylvania State University, USA
| | | | - Bradley Voytek
- Department of Cognitive Science, University of California, San Diego, USA; Halıcıoğlu Data Science Institute, University of California, San Diego, USA; Neurosciences Graduate Program, University of California, San Diego, USA
| | | |
Collapse
|
43
|
No effects of transcranial direct current stimulation on visual evoked potential and peak gamma frequency. Cogn Process 2022; 23:235-254. [PMID: 35099659 DOI: 10.1007/s10339-022-01076-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Accepted: 01/12/2022] [Indexed: 11/03/2022]
Abstract
Evidence suggests that the visual evoked potential (VEP) and gamma oscillations elicited by visual stimuli reflect the balance of excitatory and inhibitory (E-I) cortical processes. As tDCS has been shown to modulate E-I balance, the current study investigated whether amplitudes of VEP components (N1 and P2) and peak gamma frequency are modulated by transcranial direct current stimulation (tDCS). Healthy adults underwent two electroencephalography (EEG) recordings while viewing stimuli designed to elicit a robust visual response. Between the two recordings, participants were randomly assigned to three tDCS conditions (anodal-, cathodal-, and sham-tDCS) or received no-tDCS. tDCS electrodes were placed over the occipital cortex (Oz) and the left cheek with an intensity of 2 mA for 10 min. Data of 39 participants were analysed for VEP amplitudes and peak gamma frequency using mixed-model ANOVAs. The results showed no main effects of tDCS in any metric. Possible explanations for the absence of tDCS effects are discussed.
Collapse
|
44
|
An Overview of Bioprocesses Employing Specifically Selected Microbial Catalysts for γ-Aminobutyric Acid Production. Microorganisms 2021; 9:microorganisms9122457. [PMID: 34946060 PMCID: PMC8704203 DOI: 10.3390/microorganisms9122457] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 11/22/2021] [Accepted: 11/25/2021] [Indexed: 11/17/2022] Open
Abstract
Gamma-aminobutyric acid (GABA) is an important chemical compound in the human brain. GABA acts as an inhibitory neurotransmitter by inducing hyperpolarization of cellular membranes. Usually, this pharmaceutically important compound is synthesized using a chemical process, but in this short overview we have only analysed microbial processes, which have been studied for the biosynthesis of this commercially important compound. The content of this article includes the following summarised information: the search for biological processes showed a number of lactic acid bacteria and certain species of fungi, which could be effectively used for the production of GABA. Strains found to possess GABA-producing pathways include Lactobacillus brevis CRL 1942, L. plantarum FNCC 260, Streptococcus salivarius subsp. thermophilus Y2, Bifidobacterium strains, Monascus spp., and Rhizopus spp. Each of these strains required specific growth conditions. However, several factors were common among these strains, such as the use of two main supplements in their fermentation medium—monosodium glutamate and pyridoxal phosphate—and maintaining an acidic pH. Optimization studies of GABA production were comprised of altering the media constituents, modifying growth conditions, types of cultivation system, and genetic manipulation. Some strains increased the production of GABA under anaerobic conditions. Genetic manipulation focused on silencing some genes or overexpression of gadB and gadC. The conclusion, based on the review of information available in published research, is that the targeted manipulation of selected microorganisms, as well as the culture conditions for an optimised bioprocess, should be adopted for an increased production of GABA to meet its increasing demand for food and pharmaceutical applications.
Collapse
|
45
|
Ahmadi ZZ, DiBacco ML, Pearl PL. Speech Motor Function and Auditory Perception in Succinic Semialdehyde Dehydrogenase Deficiency: Toward Pre-Supplementary Motor Area (SMA) and SMA-Proper Dysfunctions. J Child Neurol 2021; 36:1210-1217. [PMID: 33757330 DOI: 10.1177/08830738211001210] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
This study reviews the fundamental roles of pre-supplementary motor area (SMA) and SMA-proper responsible for speech-motor functions and auditory perception in succinic semialdehyde dehydrogenase (SSADH) deficiency. We comprehensively searched the databases of PubMed, Google Scholar, and the electronic journals Springer, PreQuest, and Science Direct associated with keywords SSADHD, SMA, auditory perception, speech, and motor with AND operator. Transcranial magnetic stimulation emerged for assessing excitability/inhibitory M1 functions, but its role in pre-SMA and SMA proper dysfunction remains unknown. There was a lack of data on resting-state and task-based functional magnetic resonance imaging (MRI), with a focus on passive and active tasks for both speech and music, in terms of analysis of SMA-related cortex and its connections. Children with SSADH deficiency likely experience a dysfunction in connectivity between SMA portions with cortical and subcortical areas contributing to disabilities in speech-motor functions and auditory perception. Early diagnosis of auditory-motor disabilities in children with SSADH deficiency by neuroimaging techniques invites opportunities for utilizing sensory-motor integration as future interventional strategies.
Collapse
Affiliation(s)
- Zohreh Ziatabar Ahmadi
- Department of Speech Therapy, School of Rehabilitation, Babol University of Medical Sciences, Babol, I.R. Iran
| | - Melissa L DiBacco
- Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Phillip L Pearl
- Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
46
|
Scheuer T, dem Brinke EA, Grosser S, Wolf SA, Mattei D, Sharkovska Y, Barthel PC, Endesfelder S, Friedrich V, Bührer C, Vida I, Schmitz T. Reduction of cortical parvalbumin-expressing GABAergic interneurons in a rodent hyperoxia model of preterm birth brain injury with deficits in social behavior and cognition. Development 2021; 148:272278. [PMID: 34557899 DOI: 10.1242/dev.198390] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 09/17/2021] [Indexed: 12/18/2022]
Abstract
The inhibitory GABAergic system in the brain is involved in the etiology of various psychiatric problems, including autism spectrum disorders (ASD), attention deficit hyperactivity disorder (ADHD) and others. These disorders are influenced not only by genetic but also by environmental factors, such as preterm birth, although the underlying mechanisms are not known. In a translational hyperoxia model, exposing mice pups at P5 to 80% oxygen for 48 h to mimic a steep rise of oxygen exposure caused by preterm birth from in utero into room air, we documented a persistent reduction of cortical mature parvalbumin-expressing interneurons until adulthood. Developmental delay of cortical myelin was observed, together with decreased expression of oligodendroglial glial cell-derived neurotrophic factor (GDNF), a factor involved in interneuronal development. Electrophysiological and morphological properties of remaining interneurons were unaffected. Behavioral deficits were observed for social interaction, learning and attention. These results demonstrate that neonatal oxidative stress can lead to decreased interneuron density and to psychiatric symptoms. The obtained cortical myelin deficit and decreased oligodendroglial GDNF expression indicate that an impaired oligodendroglial-interneuronal interplay contributes to interneuronal damage.
Collapse
Affiliation(s)
- Till Scheuer
- Department of Neonatology, Charité - Universitätsmedizin Berlin, Berlin 13353, Germany
| | - Elena Auf dem Brinke
- Department of Neonatology, Charité - Universitätsmedizin Berlin, Berlin 13353, Germany
| | - Sabine Grosser
- Institute for Integrative Neuroanatomy, NeuroCure Cluster of Excellence, Charité - Universitätsmedizin Berlin, Berlin 10117, Germany
| | - Susanne A Wolf
- Cellular Neurocience, Max-Delbrueck-Center for Molecular Medicine in the Helmholtz Association, Berlin 13125, Germany.,Department of Experimental Ophthalmology, Charité - Universitätsmedizin Berlin, Berlin 13353, Germany
| | - Daniele Mattei
- Cellular Neurocience, Max-Delbrueck-Center for Molecular Medicine in the Helmholtz Association, Berlin 13125, Germany.,Institute of Pharmacology and Toxicology, University of Zurich-Vetsuisse, Zurich CH-8057, Switzerland
| | - Yuliya Sharkovska
- Department of Neonatology, Charité - Universitätsmedizin Berlin, Berlin 13353, Germany.,Institute for Cell and Neurobiology, Center for Anatomy, Charité - Universitätsmedizin Berlin, Berlin 10117, Germany.,Berlin Institute of Health (BIH), Berlin 10178, Germany
| | - Paula C Barthel
- Department of Neonatology, Charité - Universitätsmedizin Berlin, Berlin 13353, Germany.,Institute for Cell and Neurobiology, Center for Anatomy, Charité - Universitätsmedizin Berlin, Berlin 10117, Germany
| | - Stefanie Endesfelder
- Department of Neonatology, Charité - Universitätsmedizin Berlin, Berlin 13353, Germany
| | - Vivien Friedrich
- Department of Neonatology, Charité - Universitätsmedizin Berlin, Berlin 13353, Germany.,Berlin Institute of Health (BIH), Berlin 10178, Germany
| | - Christoph Bührer
- Department of Neonatology, Charité - Universitätsmedizin Berlin, Berlin 13353, Germany
| | - Imre Vida
- Institute for Integrative Neuroanatomy, NeuroCure Cluster of Excellence, Charité - Universitätsmedizin Berlin, Berlin 10117, Germany
| | - Thomas Schmitz
- Department of Neonatology, Charité - Universitätsmedizin Berlin, Berlin 13353, Germany
| |
Collapse
|
47
|
Increased locomotor activity via regulation of GABAergic signalling in foxp2 mutant zebrafish-implications for neurodevelopmental disorders. Transl Psychiatry 2021; 11:529. [PMID: 34650032 PMCID: PMC8517032 DOI: 10.1038/s41398-021-01651-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 09/08/2021] [Accepted: 09/29/2021] [Indexed: 12/14/2022] Open
Abstract
Recent advances in the genetics of neurodevelopmental disorders (NDDs) have identified the transcription factor FOXP2 as one of numerous risk genes, e.g. in autism spectrum disorders (ASD) and attention-deficit/hyperactivity disorder (ADHD). FOXP2 function is suggested to be involved in GABAergic signalling and numerous studies demonstrate that GABAergic function is altered in NDDs, thus disrupting the excitation/inhibition balance. Interestingly, GABAergic signalling components, including glutamate-decarboxylase 1 (Gad1) and GABA receptors, are putative transcriptional targets of FOXP2. However, the specific role of FOXP2 in the pathomechanism of NDDs remains elusive. Here we test the hypothesis that Foxp2 affects behavioural dimensions via GABAergic signalling using zebrafish as model organism. We demonstrate that foxp2 is expressed by a subset of GABAergic neurons located in brain regions involved in motor functions, including the subpallium, posterior tuberculum, thalamus and medulla oblongata. Using CRISPR/Cas9 gene-editing we generated a novel foxp2 zebrafish loss-of-function mutant that exhibits increased locomotor activity. Further, genetic and/or pharmacological disruption of Gad1 or GABA-A receptors causes increased locomotor activity, resembling the phenotype of foxp2 mutants. Application of muscimol, a GABA-A receptor agonist, rescues the hyperactive phenotype induced by the foxp2 loss-of-function. By reverse translation of the therapeutic effect on hyperactive behaviour exerted by methylphenidate, we note that application of methylphenidate evokes different responses in wildtype compared to foxp2 or gad1b loss-of-function animals. Together, our findings support the hypothesis that foxp2 regulates locomotor activity via GABAergic signalling. This provides one targetable mechanism, which may contribute to behavioural phenotypes commonly observed in NDDs.
Collapse
|
48
|
Chan KL, Hock A, Edden RAE, MacMillan EL, Henning A. Improved prospective frequency correction for macromolecule-suppressed GABA editing with metabolite cycling at 3T. Magn Reson Med 2021; 86:2945-2956. [PMID: 34431549 DOI: 10.1002/mrm.28950] [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: 03/03/2021] [Revised: 07/14/2021] [Accepted: 07/15/2021] [Indexed: 11/08/2022]
Abstract
PURPOSE To combine metabolite cycling with J-difference editing (MC MEGA) to allow for prospective frequency correction at each transient without additional acquisitions and compare it to water-suppressed MEGA-PRESS (WS MEGA) editing with intermittent prospective frequency correction. METHODS Macromolecule-suppressed gamma aminobutyric acid (GABA)-edited experiments were performed in a phantom and in the occipital lobe (OCC) (n = 12) and medial prefrontal cortex (mPFC) (n = 8) of the human brain. Water frequency consistency and average offset over acquisition time were compared. GABA multiplet patterns, signal intensities, and choline subtraction artifacts were evaluated. In vivo GABA concentrations were compared and related to frequency offset in the OCC. RESULTS MC MEGA was more stable with 21% and 32% smaller water frequency SDs in the OCC and mPFC, respectively. MC MEGA also had 39% and 40% smaller average frequency offsets in the OCC and mPFC, respectively. Phantom GABA multiplet patterns and signal intensities were similar. In vivo GABA concentrations were smaller in MC MEGA than in WS MEGA, with median (interquartile range) of 2.52 (0.27) and 2.29 (0.19) institutional units (i.u.), respectively in the OCC scans without prior DTI, and 0.99 (0.3) and 1.72 (0.5), respectively in the mPFC. OCC WS MEGA GABA concentrations, but not MC MEGA GABA concentrations were moderately correlated with frequency offset. mPFC WS MEGA spectra contained significantly more subtraction artifacts than MC MEGA spectra. CONCLUSION MC MEGA is feasible and allows for prospective frequency correction at every transient. MC MEGA GABA concentrations were not biased by frequency offsets and contained less subtraction artifacts compared to WS MEGA.
Collapse
Affiliation(s)
- Kimberly L Chan
- Advanced Imaging Research Center, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Andreas Hock
- MR Clinical Science, Philips Health Systems, Horgen, Switzerland
| | - Richard A E Edden
- Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,F. M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, Maryland, USA
| | - Erin L MacMillan
- UBC MRI Research Centre, University of British Columbia, Vancouver, British Columbia, Canada.,SFU ImageTech Lab, Simon Fraser University, Surrey, British Columbia, Canada.,MR Clinical Science, Philips Healthcare, Markham, Ontario, Canada
| | - Anke Henning
- Advanced Imaging Research Center, University of Texas Southwestern Medical Center, Dallas, Texas, USA.,Max Planck Institute for Biological Cybernetics, Tübingen, Germany
| |
Collapse
|
49
|
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
|
50
|
Nikolaus S, Mamlins E, Giesel FL, Schmitt D, Müller HW. Monoaminergic hypo- or hyperfunction in adolescent and adult attention-deficit hyperactivity disorder? Rev Neurosci 2021; 33:347-364. [PMID: 34378877 DOI: 10.1515/revneuro-2021-0083] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 06/25/2021] [Indexed: 12/13/2022]
Abstract
Disturbances of dopamine (DA), serotonin (5-HT) and/or norepinephrine (NE) functions are implied in attention-deficit hyperactivity disorder (ADHD). However, the precise cortical and subcortical mechanisms are still not fully understood. In the present survey, we conducted a PUBMED search, which provided 37 in vivo investigations with PET and SPECT on 419 ADHD patients and 490 controls. The retrospective analysis revealed increased striatal DA transporter (DAT) in adolescent as well as adult medication-naïve and not acutely medicated patients. In acutely medicated adults, DAT was not different from controls. Midbrain DAT was normal in adults, but decreased in adolescents. Striatal D2 receptor (R) binding was normal in both adolescents (not acutely medicated) and adults (acutely medicated and not acutely medicated). In medication-naïve adults, DA synthesis was decreased in putamen and amygdala, but normal in the whole striatum and midbrain. In not acutely medicated adults, DA synthesis was reduced in putamen, whole striatum, prefrontal cortex, frontal cortex, amygdala and midbrain, whereas, in adolescents, no regional differences were observed. In adult (not acutely medicated) subjects, cingulate D1R was reduced. 5-HT transporter (SERT) binding was decreased in striatum and thalamus, but normal in midbrain, neocortex and limbic regions, whereas, in medication-naïve adults, SERT was diminished in striatum and midbrain, but normal in thalamus and neocortex. The findings suggest transient stages of synaptic DA shortage as well as DA surplus in individual brain regions, which elicit presynaptic as well as postsynaptic compensatory mechanisms, striving to attain functional homeostasis. Thereby, it remains a matter of debate, whether ADHD may be characterized by a general hypo- or hyperactivity of DA and/or 5-HT function.
Collapse
Affiliation(s)
- Susanne Nikolaus
- Department of Nuclear Medicine, University Hospital Düsseldorf, Heinrich-Heine University, Moorenstr. 5, 40225 Düsseldorf, Germany
| | - Eduards Mamlins
- Department of Nuclear Medicine, University Hospital Düsseldorf, Heinrich-Heine University, Moorenstr. 5, 40225 Düsseldorf, Germany
| | - Frederik L Giesel
- Department of Nuclear Medicine, University Hospital Düsseldorf, Heinrich-Heine University, Moorenstr. 5, 40225 Düsseldorf, Germany
| | - Dominik Schmitt
- Department of Nuclear Medicine, University Hospital Düsseldorf, Heinrich-Heine University, Moorenstr. 5, 40225 Düsseldorf, Germany
| | - Hans-Wilhelm Müller
- Department of Nuclear Medicine, University Hospital Düsseldorf, Heinrich-Heine University, Moorenstr. 5, 40225 Düsseldorf, Germany
| |
Collapse
|