1
|
Yates JR. Aberrant glutamatergic systems underlying impulsive behaviors: Insights from clinical and preclinical research. Prog Neuropsychopharmacol Biol Psychiatry 2024; 135:111107. [PMID: 39098647 PMCID: PMC11409449 DOI: 10.1016/j.pnpbp.2024.111107] [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: 05/01/2024] [Revised: 07/07/2024] [Accepted: 07/24/2024] [Indexed: 08/06/2024]
Abstract
Impulsivity is a broad construct that often refers to one of several distinct behaviors and can be measured with self-report questionnaires and behavioral paradigms. Several psychiatric conditions are characterized by one or more forms of impulsive behavior, most notably the impulsive/hyperactive subtype of attention-deficit/hyperactivity disorder (ADHD), mood disorders, and substance use disorders. Monoaminergic neurotransmitters are known to mediate impulsive behaviors and are implicated in various psychiatric conditions. However, growing evidence suggests that glutamate, the major excitatory neurotransmitter of the mammalian brain, regulates important functions that become dysregulated in conditions like ADHD. The purpose of the current review is to discuss clinical and preclinical evidence linking glutamate to separate aspects of impulsivity, specifically motor impulsivity, impulsive choice, and affective impulsivity. Hyperactive glutamatergic activity in the corticostriatal and the cerebro-cerebellar pathways are major determinants of motor impulsivity. Conversely, hypoactive glutamatergic activity in frontal cortical areas and hippocampus and hyperactive glutamatergic activity in anterior cingulate cortex and nucleus accumbens mediate impulsive choice. Affective impulsivity is controlled by similar glutamatergic dysfunction observed for motor impulsivity, except a hyperactive limbic system is also involved. Loss of glutamate homeostasis in prefrontal and nucleus accumbens may contribute to motor impulsivity/affective impulsivity and impulsive choice, respectively. These results are important as they can lead to novel treatments for those with a condition characterized by increased impulsivity that are resistant to conventional treatments.
Collapse
Affiliation(s)
- Justin R Yates
- Department of Psychological Science, Northern Kentucky University, 1 Nunn Drive, Highland Heights, KY 41099, USA.
| |
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
|
Bogdańska-Chomczyk E, Wojtacha P, Tsai ML, Huang ACW, Kozłowska A. Age-related changes in the architecture and biochemical markers levels in motor-related cortical areas of SHR rats-an ADHD animal model. Front Mol Neurosci 2024; 17:1414457. [PMID: 39246601 PMCID: PMC11378348 DOI: 10.3389/fnmol.2024.1414457] [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: 04/08/2024] [Accepted: 06/25/2024] [Indexed: 09/10/2024] Open
Abstract
Introduction Attention-deficit/hyperactivity disorder (ADHD) is a neurodevelopmental disorder whose exact pathophysiology has not been fully understood yet. Numerous studies have suggested disruptions in the cellular architecture and neuronal activity within brain structures of individuals with ADHD, accompanied by imbalances in the immune system, oxidative stress, and metabolism. Methods This study aims to assess two functionally and histologically distinct brain areas involved in motor control and coordination: the motor cortex (MC) and prefrontal cortex (PFC). Namely, the morphometric analysis of the MC throughout the developmental stages of Spontaneously Hypertensive Rats (SHRs) and Wistar Kyoto Rats (WKYs). Additionally, the study aimed to investigate the levels and activities of specific immune, oxidative stress, and metabolic markers in the PFC of juvenile and maturing SHRs in comparison to WKYs. Results The most significant MC volume reductions occurred in juvenile SHRs, accompanied by alterations in neuronal density in these brain areas compared to WKYs. Furthermore, juvenile SHRs exhibit heightened levels and activity of various markers, including interleukin-1α (IL-1α), IL-6, serine/threonine-protein mammalian target of rapamycin, RAC-alpha serine/threonine-protein kinase, glucocorticoid receptor β, malondialdehyde, sulfhydryl groups, superoxide dismutase, peroxidase, glutathione reductase, glutathione S-transferase, glucose, fructosamine, iron, lactic acid, alanine, aspartate transaminase, and lactate dehydrogenase. Discussion Significant changes in the MC morphometry and elevated levels of inflammatory, oxidative, and metabolic markers in PFC might be associated with disrupted brain development and maturation in ADHD.
Collapse
Affiliation(s)
- E Bogdańska-Chomczyk
- Department of Human Physiology and Pathophysiology, School of Medicine, Collegium Medicum, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| | - P Wojtacha
- Department of Psychology and Sociology of Health and Public Health, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| | - M L Tsai
- Department of Biomechatronic Engineering, National Ilan University, Yilan, Taiwan
| | - A C W Huang
- Department of Psychology, Fo Guang University, Yilan, Taiwan
| | - A Kozłowska
- Department of Human Physiology and Pathophysiology, School of Medicine, Collegium Medicum, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| |
Collapse
|
4
|
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
|
5
|
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
|
6
|
Song Y, Hupfeld KE, Davies-Jenkins CW, Zöllner HJ, Murali-Manohar S, Mumuni AN, Crocetti D, Yedavalli V, Oeltzschner G, Alessi N, Batschelett MA, Puts NA, Mostofsky SH, Edden RA. Brain glutathione and GABA+ levels in autistic children. Autism Res 2024; 17:512-528. [PMID: 38279628 PMCID: PMC10963146 DOI: 10.1002/aur.3097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Accepted: 12/28/2023] [Indexed: 01/28/2024]
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental condition characterized by social communication challenges and repetitive behaviors. Altered neurometabolite levels, including glutathione (GSH) and gamma-aminobutyric acid (GABA), have been proposed as potential contributors to the biology underlying ASD. This study investigated whether cerebral GSH or GABA levels differ between a cohort of children aged 8-12 years with ASD (n = 52) and typically developing children (TDC, n = 49). A comprehensive analysis of GSH and GABA levels in multiple brain regions, including the primary motor cortex (SM1), thalamus (Thal), medial prefrontal cortex (mPFC), and supplementary motor area (SMA), was conducted using single-voxel HERMES MR spectroscopy at 3T. The results revealed no significant differences in cerebral GSH or GABA levels between the ASD and TDC groups across all examined regions. These findings suggest that the concentrations of GSH (an important antioxidant and neuromodulator) and GABA (a major inhibitory neurotransmitter) do not exhibit marked alterations in children with ASD compared to TDC. A statistically significant positive correlation was observed between GABA levels in the SM1 and Thal regions with ADHD inattention scores. No significant correlation was found between metabolite levels and hyper/impulsive scores of ADHD, measures of core ASD symptoms (ADOS-2, SRS-P) or adaptive behavior (ABAS-2). While both GSH and GABA have been implicated in various neurological disorders, the current study provides valuable insights into the specific context of ASD and highlights the need for further research to explore other neurochemical alterations that may contribute to the pathophysiology of this complex disorder.
Collapse
Affiliation(s)
- Yulu Song
- The Russel 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
| | - Kathleen E. Hupfeld
- The Russel 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
| | - Christopher W. Davies-Jenkins
- The Russel 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
| | - Helge J. Zöllner
- The Russel 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
| | - Saipavitra Murali-Manohar
- The Russel 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
| | | | - Deana Crocetti
- Center for Neurodevelopmental and Imaging Research, Kennedy Krieger Institute, Baltimore, MD, United States
| | - Vivek Yedavalli
- The Russel H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Georg Oeltzschner
- The Russel 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
| | - Natalie Alessi
- Center for Neurodevelopmental and Imaging Research, Kennedy Krieger Institute, Baltimore, MD, United States
| | - Mitchell A. Batschelett
- Center for Neurodevelopmental and Imaging Research, Kennedy Krieger Institute, Baltimore, MD, United States
| | - Nicolaas A.J. Puts
- Department of Forensic and Neurodevelopmental Sciences, Institute of Psychiatry, Psychology, and Neuroscience, King’s College London, London, United Kingdom
- MRC Center for Neurodevelopmental Disorders, King’s College London, London, United Kingdom
| | - Stewart H. Mostofsky
- Center for Neurodevelopmental and Imaging Research, Kennedy Krieger Institute, Baltimore, MD, United States
- Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Department of Psychiatry and Behavioral Sciences, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Richard A.E. Edden
- The Russel 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
| |
Collapse
|
7
|
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
|
8
|
Patel B, Koysombat K, Mills EG, Tsoutsouki J, Comninos AN, Abbara A, Dhillo WS. The Emerging Therapeutic Potential of Kisspeptin and Neurokinin B. Endocr Rev 2024; 45:30-68. [PMID: 37467734 PMCID: PMC10765167 DOI: 10.1210/endrev/bnad023] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 06/13/2023] [Accepted: 07/11/2023] [Indexed: 07/21/2023]
Abstract
Kisspeptin (KP) and neurokinin B (NKB) are neuropeptides that govern the reproductive endocrine axis through regulating hypothalamic gonadotropin-releasing hormone (GnRH) neuronal activity and pulsatile GnRH secretion. Their critical role in reproductive health was first identified after inactivating variants in genes encoding for KP or NKB signaling were shown to result in congenital hypogonadotropic hypogonadism and a failure of pubertal development. Over the past 2 decades since their discovery, a wealth of evidence from both basic and translational research has laid the foundation for potential therapeutic applications. Beyond KP's function in the hypothalamus, it is also expressed in the placenta, liver, pancreas, adipose tissue, bone, and limbic regions, giving rise to several avenues of research for use in the diagnosis and treatment of pregnancy, metabolic, liver, bone, and behavioral disorders. The role played by NKB in stimulating the hypothalamic thermoregulatory center to mediate menopausal hot flashes has led to the development of medications that antagonize its action as a novel nonsteroidal therapeutic agent for this indication. Furthermore, the ability of NKB antagonism to partially suppress (but not abolish) the reproductive endocrine axis has supported its potential use for the treatment of various reproductive disorders including polycystic ovary syndrome, uterine fibroids, and endometriosis. This review will provide a comprehensive up-to-date overview of the preclinical and clinical data that have paved the way for the development of diagnostic and therapeutic applications of KP and NKB.
Collapse
Affiliation(s)
- Bijal Patel
- Section of Investigative Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College School of Medicine, Imperial College London, London, W12 0NN, UK
| | - Kanyada Koysombat
- Section of Investigative Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College School of Medicine, Imperial College London, London, W12 0NN, UK
- Department of Diabetes and Endocrinology, Imperial College Healthcare NHS Trust, 72 Du Cane Rd, London, W12 0HS, UK
| | - Edouard G Mills
- Section of Investigative Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College School of Medicine, Imperial College London, London, W12 0NN, UK
- Department of Diabetes and Endocrinology, Imperial College Healthcare NHS Trust, 72 Du Cane Rd, London, W12 0HS, UK
| | - Jovanna Tsoutsouki
- Section of Investigative Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College School of Medicine, Imperial College London, London, W12 0NN, UK
| | - Alexander N Comninos
- Section of Investigative Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College School of Medicine, Imperial College London, London, W12 0NN, UK
- Department of Diabetes and Endocrinology, Imperial College Healthcare NHS Trust, 72 Du Cane Rd, London, W12 0HS, UK
| | - Ali Abbara
- Section of Investigative Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College School of Medicine, Imperial College London, London, W12 0NN, UK
- Department of Diabetes and Endocrinology, Imperial College Healthcare NHS Trust, 72 Du Cane Rd, London, W12 0HS, UK
| | - Waljit S Dhillo
- Section of Investigative Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College School of Medicine, Imperial College London, London, W12 0NN, UK
- Department of Diabetes and Endocrinology, Imperial College Healthcare NHS Trust, 72 Du Cane Rd, London, W12 0HS, UK
| |
Collapse
|
9
|
Rudy MJ, Salois G, Cubello J, Newell R, Mayer-Proschel M. Gestational iron deficiency affects the ratio between interneuron subtypes in the postnatal cerebral cortex in mice. Development 2023; 150:dev201068. [PMID: 36805633 PMCID: PMC10110419 DOI: 10.1242/dev.201068] [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: 06/27/2022] [Accepted: 01/30/2023] [Indexed: 02/22/2023]
Abstract
Gestational iron deficiency (gID) is highly prevalent and associated with an increased risk of intellectual and developmental disabilities in affected individuals that are often defined by a disrupted balance of excitation and inhibition (E/I) in the brain. Using a nutritional mouse model of gID, we previously demonstrated a shift in the E/I balance towards increased inhibition in the brains of gID offspring that was refractory to postnatal iron supplementation. We thus tested whether gID affects embryonic progenitor cells that are fated towards inhibitory interneurons. We quantified relevant cell populations during embryonic inhibitory neuron specification and found an increase in the proliferation of Nkx2.1+ interneuron progenitors in the embryonic medial ganglionic eminence at E14 that was associated with increased Shh signaling in gID animals at E12. When we quantified the number of mature inhibitory interneurons that are known to originate from the MGE, we found a persistent disruption of differentiated interneuron subtypes in early adulthood. Our data identify a cellular target that links gID with a disruption of cortical interneurons which play a major role in the establishment of the E/I balance.
Collapse
Affiliation(s)
- Michael J. Rudy
- Department of Biomedical Genetics, University of Rochester, 601 Elmwood Avenue, Rochester, NY 14642, USA
- Department of Neurology, University of Colorado Denver – Anschutz Medical Campus, 13001 East 17th Place, Aurora, CO 80045, USA
| | - Garrick Salois
- Department of Biomedical Genetics, University of Rochester, 601 Elmwood Avenue, Rochester, NY 14642, USA
| | - Janine Cubello
- Department of Biomedical Genetics, University of Rochester, 601 Elmwood Avenue, Rochester, NY 14642, USA
| | - Robert Newell
- Department of Biomedical Genetics, University of Rochester, 601 Elmwood Avenue, Rochester, NY 14642, USA
| | - Margot Mayer-Proschel
- Department of Biomedical Genetics, University of Rochester, 601 Elmwood Avenue, Rochester, NY 14642, USA
| |
Collapse
|
10
|
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
|
11
|
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
|
12
|
Cheng D, Qin ZS, Zheng Y, Xie JY, Liang SS, Zhang JL, Feng YB, Zhang ZJ. Minocycline, a classic antibiotic, exerts psychotropic effects by normalizing microglial neuroinflammation-evoked tryptophan-kynurenine pathway dysregulation in chronically stressed male mice. Brain Behav Immun 2023; 107:305-318. [PMID: 36332817 DOI: 10.1016/j.bbi.2022.10.022] [Citation(s) in RCA: 22] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 10/20/2022] [Accepted: 10/27/2022] [Indexed: 11/06/2022] Open
Abstract
The dysregulation of tryptophan-kynurenine pathway (TKP) is extensively involved in the pathophysiology of Alzheimer's disease, depression, and neurodegenerative disorders. Minocycline, a classic antibiotic, may exert psychotropic effects associated with the modulation of TKP. In this study, we examined the effects of minocycline in improving behaviour and modulating TKP components in chronically stressed male mice. Following repeated treatment with 22.5 mg/kg and 45 mg/kg minocycline for 27 days, the stressed mice particularly with higher dose displayed significant improvement on cognitive impairment, depression- and anxiety-like behaviour. Minocycline suppressed stress-induced overexpression of pro-inflammatory cytokines and restored anti-inflammatory cytokines. Chronic stress dramatically suppressed blood and prefrontal cortical levels of the primary substrate tryptophan (TRP), the neuroprotective metabolite kynurenic acid (KYNA), and KYNA/KYN ratio, but increased the intermediate kynurenine (KYN), 3-hydroxykynurenine (3-HK), KYN/TRP ratio, and the neurotoxic metabolite quinolinic acid (QUIN). Minocycline partially or completely reversed changes in these components. Minocycline also inhibited stress-induced overexpression of QUIN-related enzymes, indoleamine 2, 3-dioxygenase 1(iDO-1), kynureninase (KYNU), kynurenine 3-monooxygenase (KMO), 3-hydroxyanthranilate 3,4-dioxygenase (3-HAO), but rescued the decreased expression of kynurenine aminotransferase (KAT) in brain regions. Behavioral improvements were correlated with multiple TKP metabolites and enzymes. These results suggest that the psychotropic effects of minocycline are mainly associated with the restoration of biodistribution of the primary substrate in the brain and normalization of neuroinflammation-evoked TKP dysregulation.
Collapse
Affiliation(s)
- Dan Cheng
- School of Chinese Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Zong-Shi Qin
- School of Chinese Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Yu Zheng
- School of Chinese Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Jun-Ya Xie
- Department of Statistics and Actuarial Science, Faculty of Science, The University of Hong Kong, Hong Kong, China
| | - Sui-Sha Liang
- School of Public Health, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Jia-Ling Zhang
- School of Chinese Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Yi-Bin Feng
- School of Chinese Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Zhang-Jin Zhang
- School of Chinese Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China; Department of Chinese Medicine, The University of Hong Kong-Shenzhen Hospital (HKU-SZH), Shenzhen, China.
| |
Collapse
|
13
|
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
|
14
|
Ritter C, Buchmann A, Müller ST, Volleberg M, Haynes M, Ghisleni C, Noeske R, Tuura R, Hasler G. Evaluation of Prefrontal γ-Aminobutyric Acid and Glutamate Levels in Individuals With Major Depressive Disorder Using Proton Magnetic Resonance Spectroscopy. JAMA Psychiatry 2022; 79:1209-1216. [PMID: 36260322 PMCID: PMC9582968 DOI: 10.1001/jamapsychiatry.2022.3384] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 08/29/2022] [Indexed: 01/14/2023]
Abstract
Importance Major depressive disorder (MDD) is one of the most prevalent illnesses worldwide. Perturbations of the major inhibitory and excitatory neurotransmitters, γ-aminobutyric acid (GABA) and glutamate (Glu), respectively, as well as Glx (Glu or glutamine [Gln]) have been extensively reported in a multitude of brain areas of individuals with depression, but few studies have examined changes in Gln, the metabolic counterpart of synaptic Glu. Objective To investigate changes in GABA, Glx, Glu, and Gln levels in a voxel in the left dorsolateral prefrontal cortex of participants with no, past, and current MDD using proton magnetic resonance spectroscopy (1H-MRS). Design, Setting, and Participants This community-based study used a cross-sectional design using 3-T 1H-MRS in participants not taking MDD medication recruited from the community. The sample consisted of 251 healthy controls, 98 participants with a history of past MDD, and 47 participants who met the diagnostic criteria for current MDD. Diagnostic groups were comparable regarding age, education, income, and diet. Data were collected from March 2014 to October 2021, and data were analyzed from October 2021 to June 2022. Main Outcomes and Measures GABA, Glx, Glu, and Gln concentrations in the left dorsolateral prefrontal cortex. Results Of 396 included participants, 258 (65.2%) were female, and the mean (SD) age was 25.0 (4.7) years. Compared with healthy controls, those with past MDD and current MDD had lower GABA concentrations (mean [SEM] concentration: healthy controls, 2.70 [0.03] mmol/L; past MDD, 2.49 [0.05] mmol/L; current MDD, 2.54 [0.07] mmol/L; 92 with past MDD vs 236 healthy controls: r = 0.18; P = .002; 44 with current MDD vs 236 healthy controls: r = 0.13; P = .04). Compared with healthy controls, those with past MDD also had lower Glu concentrations (mean [SEM] concentration: healthy controls, 7.52 [0.06] mmol/L; past MDD, 7.23 [0.11] mmol/L; 93 with past MDD vs 234 healthy controls: r = 0.16; P = .01) and higher Gln concentrations (mean [SEM] concentration: healthy controls, 1.63 [0.04] mmol/L; past MDD, 1.84 [0.07] mmol/L; 66 with past MDD 153 healthy controls: r = 0.17; P = .04). Conclusions and Relevance In a large, mostly medication-free community sample, reduced prefrontal GABA concentrations were associated with past MDD, consistent with histopathologic studies reporting reduced glial cell and GABA cell density in the prefrontal cortex in individuals with depression. Patients with MDD also demonstrated increased Gln levels, indicative of increased synaptic Glu release, adding to previous evidence for the Glu hypothesis of MDD.
Collapse
Affiliation(s)
- Christopher Ritter
- Psychiatric University Hospital, University of Bern, Bern, Switzerland
- Unit of Psychiatry Research, University of Fribourg, Villars-sur-Glâne, Switzerland
- Center of MR Research, University Children’s Hospital Zurich, Zurich, Switzerland
| | - Andreas Buchmann
- Psychiatric University Hospital, University of Bern, Bern, Switzerland
- Unit of Psychiatry Research, University of Fribourg, Villars-sur-Glâne, Switzerland
- Center of MR Research, University Children’s Hospital Zurich, Zurich, Switzerland
| | | | - Martin Volleberg
- Division of Clinical Chemistry and Biochemistry, University Children’s Hospital Zurich, Zurich, Switzerland
| | - Melanie Haynes
- Psychiatric University Hospital, University of Bern, Bern, Switzerland
| | - Carmen Ghisleni
- Center of MR Research, University Children’s Hospital Zurich, Zurich, Switzerland
| | | | - Ruth Tuura
- Center of MR Research, University Children’s Hospital Zurich, Zurich, Switzerland
| | - Gregor Hasler
- Unit of Psychiatry Research, University of Fribourg, Villars-sur-Glâne, Switzerland
| |
Collapse
|
15
|
Hooi SL, Dwiyanto J, Rasiti H, Toh KY, Wong RKM, Lee JWJ. A case report of improvement on ADHD symptoms after fecal microbiota transplantation with gut microbiome profiling pre- and post-procedure. Curr Med Res Opin 2022; 38:1977-1982. [PMID: 36164761 DOI: 10.1080/03007995.2022.2129232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
BACKGROUND Recent studies demonstrate the association of the gut microbiome in regulating interactions between the central nervous system and intestinal function. Individuals with attention-deficit hyperactivity disorder (ADHD) have been shown to have unique gut microbial signature, with depletion of beneficial commensal microbes. Fecal microbiota transplant (FMT) restores the imbalanced gut microbiome and may replete missing microbes to increase production of hormones and neurotransmitters regulating human behavior and cognition. RESEARCH DESIGN & METHODS Here, we present an interesting case of a 22-year-old woman treated with FMT primarily to treat recurrent Clostridioides difficile infection, which coincidentally alleviated her ADHD symptoms. We also present the pre- and post-FMT gut microbiota profiles conducted using shotgun metagenomic sequencing on the patient's fecal samples to thereby highlight potential microbial-associated mechanisms associated with the relief of ADHD symptoms. RESULTS & CONCLUSIONS Our case report provides preliminary evidence regarding the use of FMT in a patient with C. difficile and ADHD. We speculate that gut microbiome modulation, in particular the gain or loss of specific microbial species and pathways involving the metabolism of SCFAs, tryptophan and GABA, may merit further exploration as a potential therapeutic strategy for ADHD.
Collapse
Affiliation(s)
| | - Jacky Dwiyanto
- School of Science, Monash University Malaysia, Selangor, Malaysia
| | | | | | - Reuben Kong Min Wong
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Gastroenterology, gutCARE, Singapore
| | - Jonathan Wei Jie Lee
- Data Science, AMILI Pte. Ltd., Singapore
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Division of Gastroenterology and Hepatology, National University Hospital, Singapore, University Medical Cluster, Singapore
| |
Collapse
|
16
|
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
|
17
|
Lee IO, Skuse DH, Constable PA, Marmolejo-Ramos F, Olsen LR, Thompson DA. The electroretinogram b-wave amplitude: a differential physiological measure for Attention Deficit Hyperactivity Disorder and Autism Spectrum Disorder. J Neurodev Disord 2022; 14:30. [PMID: 35524181 PMCID: PMC9077889 DOI: 10.1186/s11689-022-09440-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 04/12/2022] [Indexed: 12/02/2022] Open
Abstract
Background Attention Deficit Hyperactivity Disorder (ADHD) is the most prevalent childhood neurodevelopmental disorder. It shares some genetic risk with Autism Spectrum Disorder (ASD), and the conditions often occur together. Both are potentially associated with abnormal glutamate and GABA neurotransmission, which can be modelled by measuring the synaptic activity in the retina with an electroretinogram (ERG). Reduction of retinal responses in ASD has been reported, but little is known about retinal activity in ADHD. In this study, we compared the light-adapted ERGs of individuals with ADHD, ASD and controls to investigate whether retinal responses differ between these neurodevelopmental conditions. Methods Full field light-adapted ERGs were recorded from 15 ADHD, 57 ASD (without ADHD) and 59 control participants, aged from 5.4 to 27.3 years old. A Troland protocol was used with a random series of nine flash strengths from −0.367 to 1.204 log photopic cd.s.m−2. The time-to-peak and amplitude of the a- and b-waves and the parameters of the Photopic Negative Response (PhNR) were compared amongst the three groups of participants, using generalised estimating equations. Results Statistically significant elevations of the ERG b-wave amplitudes, PhNR responses and faster timings of the b-wave time-to-peak were found in those with ADHD compared with both the control and ASD groups. The greatest elevation in the b-wave amplitudes associated with ADHD were observed at 1.204 log phot cd.s.m−2 flash strength (p < .0001), at which the b-wave amplitude in ASD was significantly lower than that in the controls. Using this measure, ADHD could be distinguished from ASD with an area under the curve of 0.88. Conclusions The ERG b-wave amplitude appears to be a distinctive differential feature for both ADHD and ASD, which produced a reversed pattern of b-wave responses. These findings imply imbalances between glutamate and GABA neurotransmission which primarily regulate the b-wave formation. Abnormalities in the b-wave amplitude could provisionally serve as a biomarker for both neurodevelopmental conditions. Supplementary Information The online version contains supplementary material available at 10.1186/s11689-022-09440-2.
Collapse
Affiliation(s)
- Irene O Lee
- Behavioural and Brain Sciences Unit, Population Policy and Practice Programme, UCL Great Ormond Street Institute of Child Health, University College London, London, UK.
| | - David H Skuse
- Behavioural and Brain Sciences Unit, Population Policy and Practice Programme, UCL Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Paul A Constable
- Caring Futures Institute, College of Nursing and Health Sciences, Flinders University, Adelaide, South Australia, Australia
| | - Fernando Marmolejo-Ramos
- Centre for Change and Complexity in Learning, University of South Australia, Adelaide, Australia
| | - Ludvig R Olsen
- Department of Molecular Medicine (MOMA), Aarhus University, Aarhus, Denmark
| | - Dorothy A Thompson
- The Tony Kriss Visual Electrophysiology Unit, Clinical and Academic Department of Ophthalmology, Sight and Sound Centre, Great Ormond Street Hospital for Children NHS Trust, London, UK.,UCL Great Ormond Street Institute of Child Health, University College London, London, UK
| |
Collapse
|
18
|
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
|
19
|
Miniksar DY, Cansız MA, Kılıç M, Göçmen AY. Relationship between sleep problems and chronotypes of children and adolescents with attention deficit and hyperactivity disorder and serum GABA, glutamate and homocysteine levels. Chronobiol Int 2022; 39:386-397. [PMID: 34961406 DOI: 10.1080/07420528.2021.2018452] [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] [Indexed: 02/07/2023]
Abstract
In this study, we aimed to determine the relationship between chronotype, sleep problems and serum levels of GABA (gamma amminobutyric acid), glutamate and homocysteine in children and adolescents diagnosed with attention deficit and hyperactivity disorder (ADHD) as well as factors affecting this relationship. Sleep problems of 46 children and adolescents aged 7-18 years diagnosed with ADHD and 30 healthy volunteers aged 7-18 years were evaluated with Children's Sleep Habits Questionnaire (CSHQ) while chronotypes were evaluated with Children's Chronotype Questionnaire (CCQ). Serum glutamate, GABA and homocysteine levels were measured using immunosorbent test (ELISA) kits. Sleep problems were significantly more common in the ADHD group compared to the control group (p < .001). Serum GABA, glutamate and homocysteine levels were found to be predictor biomarkers for ADHD, independent of total sleep problem score. When the homocysteine levels were above the cut-off point of 9.445 µmol/L, the sensitivity in early diagnosis of ADHD was 84.8% and the specificity was 70.0%. Although ADHD is a disorder in which sleep problems are common, increased serum GABA, glutamate and homocysteine are important in diagnosing ADHD independent of ADHD-related sleep problems. Homocysteine levels may be an important predictor for the presence of ADHD.
Collapse
Affiliation(s)
| | | | - Mahmut Kılıç
- Department of Public Health, Yozgat Bozok University, Yozgat, Turkey
| | | |
Collapse
|
20
|
Özaslan A, Güney E, Gülbahar Ö, Büyüktaskin D, Arslan B. Increased Serum Level of CCL5 in Children with Attention‑Deficit/Hyperactivity Disorder: First Results about Serum Chemokines. CLINICAL PSYCHOPHARMACOLOGY AND NEUROSCIENCE 2022; 20:109-117. [PMID: 35078953 PMCID: PMC8813316 DOI: 10.9758/cpn.2022.20.1.109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 12/26/2020] [Accepted: 12/30/2020] [Indexed: 12/02/2022]
Abstract
Objective Attention deficit hyperactivity disorder (ADHD) is a neurodevelopmental disorder and its aetiology is not fully understood. This study aimed to determine whether the CCL5 and CCL11 influence the ADHD aetiology by comparing serum CCL5 and CCL11 levels of children with ADHD and typical development. Methods This study included 45 (27 males, mean age = 8.9 ± 1.7 years) treatment-naive patients diagnosed with ADHD and 35 (20 males, mean age = 8.8 ± 1.6 years) healthy controls. Participants ranged in age between 6−12 years and completed the Conners Teacher Rating Scale that assesses ADHD presentation and severity. CCL5 and CCL11 serum levels were also measured using enzyme-linked immunosorbent assay kits. Results Significantly higher serum CCL5 levels were found in children with ADHD compared to healthy controls (p < 0.001). No significant difference was found between the mean serum CC11 level of the patients and controls (p = 0.93). In addition, there was no significant correlation between the serum CCL5 and CCL11 levels and predominant presentations of ADHD and disease severity. Conclusion This study suggests that there are higher levels of serum CCL5 in drug naive children with ADHD, this findings suggest that CCL5 might play a role in the pathophysiology of ADHD. Moreover, these changes in peripheral blood may have therapeutic value. In addition, these results help to understand the role of chemokines in elucidating the etiopathogenesis of ADHD. Our results can be considered as the first step in investigating the role of CCL5 in ADHD, and further research is needed to support these initial findings.
Collapse
Affiliation(s)
- Ahmet Özaslan
- Department of Child and Adolescent Psychiatry, Ankara, Turkey
| | - Esra Güney
- Department of Child and Adolescent Psychiatry, Ankara, Turkey
| | - Özlem Gülbahar
- Department of Medical Biochemistry, Gazi University Medical Faculty, Ankara, Turkey
| | - Dicle Büyüktaskin
- Department of Child and Adolescent Psychiatry, Ankara, Turkey
- Department of Child and Adolescent Psychiatry, Cizre State Hospital, Şırnak, Turkey
| | - Burak Arslan
- Department of Medical Biochemistry, Erciş Şehit Rıdvan Çevik State Hospital, Van, Turkey
| |
Collapse
|
21
|
Mills EG, Yang L, Abbara A, Dhillo WS, Comninos AN. Current Perspectives on Kisspeptins Role in Behaviour. Front Endocrinol (Lausanne) 2022; 13:928143. [PMID: 35757400 PMCID: PMC9225141 DOI: 10.3389/fendo.2022.928143] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 05/11/2022] [Indexed: 11/21/2022] Open
Abstract
The neuropeptide kisspeptin is now well-established as the master regulator of the mammalian reproductive axis. Beyond the hypothalamus, kisspeptin and its cognate receptor are also extensively distributed in extra-hypothalamic brain regions. An expanding pool of animal and human data demonstrates that kisspeptin sits within an extensive neuroanatomical and functional framework through which it can integrate a range of internal and external cues with appropriate neuroendocrine and behavioural responses. In keeping with this, recent studies reveal wide-reaching effects of kisspeptin on key behaviours such as olfactory-mediated partner preference, sexual motivation, copulatory behaviour, bonding, mood, and emotions. In this review, we provide a comprehensive update on the current animal and human literature highlighting the far-reaching behaviour and mood-altering roles of kisspeptin. A comprehensive understanding of this important area in kisspeptin biology is key to the escalating development of kisspeptin-based therapies for common reproductive and related psychological and psychosexual disorders.
Collapse
Affiliation(s)
- Edouard G. Mills
- Section of Endocrinology and Investigative Medicine, Imperial College London, London, United Kingdom
| | - Lisa Yang
- Section of Endocrinology and Investigative Medicine, Imperial College London, London, United Kingdom
| | - Ali Abbara
- Section of Endocrinology and Investigative Medicine, Imperial College London, London, United Kingdom
| | - Waljit S. Dhillo
- Section of Endocrinology and Investigative Medicine, Imperial College London, London, United Kingdom
- Department of Endocrinology, Imperial College Healthcare National Health Service (NHS) Trust, London, United Kingdom
- *Correspondence: Waljit S. Dhillo, ; Alexander N. Comninos,
| | - Alexander N. Comninos
- Section of Endocrinology and Investigative Medicine, Imperial College London, London, United Kingdom
- Department of Endocrinology, Imperial College Healthcare National Health Service (NHS) Trust, London, United Kingdom
- *Correspondence: Waljit S. Dhillo, ; Alexander N. Comninos,
| |
Collapse
|
22
|
De la Torre-Iturbe S, Vázquez-Roque RA, De la Cruz-López F, Flores G, Garcés-Ramírez L. Dendritic and behavioral changes in rats neonatally treated with homocysteine; A proposal as an animal model to study the attention deficit hyperactivity disorder. J Chem Neuroanat 2021; 119:102057. [PMID: 34871732 DOI: 10.1016/j.jchemneu.2021.102057] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 12/01/2021] [Accepted: 12/01/2021] [Indexed: 02/08/2023]
Abstract
Attention deficit hyperactivity disorder (ADHAD) is a neurobehavioral disorder that affects children and adolescents with a high prevalence. Despite its prevalence and an unclear etiology, previous reports suggest that it is closely related to homocysteine metabolism. Male Sprague Dawley rats were administered with homocysteine from postnatal day (PD) 2 to PD 16. Locomotor activity was evaluated at 35 PD (prepuberal age) and 60 PD (adult age) before and after amphetamine administration. In rats evaluated at both ages, homocysteine induced hyperactivity, and the amphetamine administration reduced hyperactivity significantly at 35 PD, but not at 60 PD. In the social interaction test, homocysteine reduced the number of contacts and increased the latency to the first contact only in rats at 35 PD. Homocysteine also had an effect on short term memory at 35D and 60 PD and long-term memory at 60 PD. Morphological changes were found mainly in the shape of dendritic spines in the prefrontal cortex (PFC-3), dorsal hippocampus (CA1), dentate gyrus (DG) and nucleus accumbens (NAcc), in rats administered neonatally with homocysteine at both ages studied. In prepuberal and adult rats, there was an increase in dendritic length in DG and NAcc, respectively. The dendritic spine morphology also was altered at both ages, mainly decreasing the number of mushroom spines in NAcc and CA1 at 30 PD and in all the areas studied at 60 PD rats. Those areas are associated with the processes of attention, learning and memory that were studied, and those alterations are possibly related to changes observed in the behavioral tests. These behavioral and morphological changes in rats at 35 PD administered with homocysteine could be similar to changes found in children diagnosed with ADHD. Moreover, half to two thirds of children diagnosed with ADHD reach adulthood with this disorder. In this study we found similarities with ADHD, finding alterations in both rats at 35 PD and 60 PD. So, this may be proposed as an animal model to study this disorder present in children, adolescents and adults.
Collapse
Affiliation(s)
- Sandra De la Torre-Iturbe
- Laboratorio de Fisiología de la Conducta, Escuela Nacional de Ciencias Biológicas (ENCB), Instituto Politécnico Nacional (IPN), CDMX, Mexico; Laboratorio de Neuropsiquiatría, Instituto de Fisiología, Benemérita Universidad Autónoma de Puebla (BUAP), 14 Sur 6301, Puebla 72570, Mexico
| | - Rubén Antonio Vázquez-Roque
- Laboratorio de Neuropsiquiatría, Instituto de Fisiología, Benemérita Universidad Autónoma de Puebla (BUAP), 14 Sur 6301, Puebla 72570, Mexico
| | - Fidel De la Cruz-López
- Laboratorio de Fisiología de la Conducta, Escuela Nacional de Ciencias Biológicas (ENCB), Instituto Politécnico Nacional (IPN), CDMX, Mexico
| | - Gonzalo Flores
- Laboratorio de Neuropsiquiatría, Instituto de Fisiología, Benemérita Universidad Autónoma de Puebla (BUAP), 14 Sur 6301, Puebla 72570, Mexico
| | - Linda Garcés-Ramírez
- Laboratorio de Fisiología de la Conducta, Escuela Nacional de Ciencias Biológicas (ENCB), Instituto Politécnico Nacional (IPN), CDMX, Mexico.
| |
Collapse
|
23
|
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
|
24
|
Comninos AN, Yang L, O’Callaghan J, Mills EG, Wall MB, Demetriou L, Wing VC, Thurston L, Owen BM, Abbara A, Rabiner EA, Dhillo WS. Kisspeptin modulates gamma-aminobutyric acid levels in the human brain. Psychoneuroendocrinology 2021; 129:105244. [PMID: 33975151 PMCID: PMC8243259 DOI: 10.1016/j.psyneuen.2021.105244] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 03/03/2021] [Accepted: 04/20/2021] [Indexed: 11/29/2022]
Abstract
Gamma-aminobutyric acid (GABA) is a key inhibitory neurotransmitter that has been implicated in the aetiology of common mood and behavioural disorders. By employing proton magnetic resonance spectroscopy in man, we demonstrate that administration of the reproductive neuropeptide, kisspeptin, robustly decreases GABA levels in the limbic system of the human brain; specifically the anterior cingulate cortex (ACC). This finding defines a novel kisspeptin-activated GABA pathway in man, and provides important mechanistic insights into the mood and behaviour-altering effects of kisspeptin seen in rodents and humans. In addition, this work has therapeutic implications as it identifies GABA-signalling as a potential target for the escalating development of kisspeptin-based therapies for common reproductive disorders of body and mind.
Collapse
Affiliation(s)
- Alexander N. Comninos
- Division of Diabetes, Endocrinology & Metabolism, Imperial College London, UK,Department of Endocrinology, Imperial College Healthcare NHS Trust, London, UK
| | - Lisa Yang
- Division of Diabetes, Endocrinology & Metabolism, Imperial College London, UK
| | | | - Edouard G. Mills
- Division of Diabetes, Endocrinology & Metabolism, Imperial College London, UK
| | | | - Lysia Demetriou
- Invicro, London, UK,Nuffield Department of Women’s and Reproductive Health, University of Oxford, UK
| | - Victoria C. Wing
- Division of Diabetes, Endocrinology & Metabolism, Imperial College London, UK
| | - Layla Thurston
- Division of Diabetes, Endocrinology & Metabolism, Imperial College London, UK
| | - Bryn M. Owen
- Division of Diabetes, Endocrinology & Metabolism, Imperial College London, UK
| | - Ali Abbara
- Division of Diabetes, Endocrinology & Metabolism, Imperial College London, UK
| | | | - Waljit S. Dhillo
- Division of Diabetes, Endocrinology & Metabolism, Imperial College London, UK,Department of Endocrinology, Imperial College Healthcare NHS Trust, London, UK,Correspondence to: Division of Diabetes, Endocrinology & Metabolism, Imperial College London, 6th Floor Commonwealth Building, Hammersmith Hospital Campus, London W12 0NN, UK.
| |
Collapse
|
25
|
Porges EC, Jensen G, Foster B, Edden RAE, Puts NAJ. The trajectory of cortical GABA across the lifespan, an individual participant data meta-analysis of edited MRS studies. eLife 2021; 10:e62575. [PMID: 34061022 PMCID: PMC8225386 DOI: 10.7554/elife.62575] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 05/30/2021] [Indexed: 01/18/2023] Open
Abstract
γ-Aminobutyric acid (GABA) is the principal inhibitory neurotransmitter in the human brain and can be measured with magnetic resonance spectroscopy (MRS). Conflicting accounts report decreases and increases in cortical GABA levels across the lifespan. This incompatibility may be an artifact of the size and age range of the samples utilized in these studies. No single study to date has included the entire lifespan. In this study, eight suitable datasets were integrated to generate a model of the trajectory of frontal GABA estimates (as reported through edited MRS; both expressed as ratios and in institutional units) across the lifespan. Data were fit using both a log-normal curve and a nonparametric spline as regression models using a multi-level Bayesian model utilizing the Stan language. Integrated data show that an asymmetric lifespan trajectory of frontal GABA measures involves an early period of increase, followed by a period of stability during early adulthood, with a gradual decrease during adulthood and aging that is described well by both spline and log-normal models. The information gained will provide a general framework to inform expectations of future studies based on the age of the population being studied.
Collapse
Affiliation(s)
- Eric C Porges
- Center for Cognitive Aging and Memory, University of FloridaGainesvilleUnited States
- McKnight Brain Research Foundation, University of FloridaUnited StatesUnited States
- Department of Clinical and Health Psychology, University of FloridaGainesvilleUnited States
| | - Greg Jensen
- Department of Neuroscience, Columbia University Medical CenterNew YorkUnited States
- Zuckerman Mind Brain Behavior Institute, Columbia UniversityNew YorkUnited States
| | - Brent Foster
- Center for Cognitive Aging and Memory, University of FloridaGainesvilleUnited States
- McKnight Brain Research Foundation, University of FloridaUnited StatesUnited States
- Department of Clinical and Health Psychology, University of FloridaGainesvilleUnited States
| | - Richard AE Edden
- Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of MedicineBaltimoreUnited States
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger InstituteBaltimoreUnited States
| | - Nicolaas AJ Puts
- Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of MedicineBaltimoreUnited States
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger InstituteBaltimoreUnited States
- Department of Forensic and Neurodevelopmental Sciences, Sackler Institute for Translational Neurodevelopment, Institute of Psychiatry, Psychology, and Neuroscience, King’s College LondonLondonUnited Kingdom
| |
Collapse
|
26
|
Sukmajaya AC, Lusida MI, Soetjipto, Setiawati Y. Systematic review of gut microbiota and attention-deficit hyperactivity disorder (ADHD). Ann Gen Psychiatry 2021; 20:12. [PMID: 33593384 PMCID: PMC7888126 DOI: 10.1186/s12991-021-00330-w] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 01/11/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Gut-brain axis (GBA) is a system widely studied nowadays, especially in the neuropsychiatry field. It is postulated to correlate with many psychiatric conditions, one of them being attention-deficit hyperactivity disorder (ADHD). ADHD is a disorder that affects many aspects of life, including but not limited to financial, psychosocial, and cultural aspects. Multiple studies have made a comparison of the gut microbiota between ADHD and healthy controls. Our aims were to review the existing studies analyzing the gut microbiota between human samples in ADHD and healthy individuals. METHODS The literature was obtained using Google Scholar, Pubmed, and Science Direct search engine. The keywords used were "ADHD", "gut microbiota", "stool", "gut", and "microbiota". The selected studies were all case-control studies, which identify the gut microbiota between ADHD and healthy individuals. RESULT We found six studies which were eligible for review. The model and methods of each study is different. Forty-nine bacterial taxa were found, yet none of them can explain the precise relationship between ADHD and the gut microbiota. Bifidobacterium was found in higher amount in ADHD patients, but other study stated that the abundance of this genus was lower in ADHD with post-micronutrient treatment. This may suggest that micronutrient can modulate the population of Bifidobacterium and improve the behavior of ADHD patients. Other notable findings include a significantly lower population of Dialister in unmedicated ADHD, which rose after patients were medicated. A smaller amount of Faecalibacterium were also found in ADHD patients. This may explain the pathogenesis of ADHD, as Faecalibacterium is known for its anti-inflammatory products. It is possible the scarcity of this genera could induce overproduction of pro-inflammatory cytokines, which is in accordance with the high level of pro-inflammatory cytokines found in children with ADHD. CONCLUSION There were no studies that examined which bacterial taxa correlated most to ADHD. This might occur due to the different model and methods in each study. Further study is needed to identify the correlation between gut microbiota and ADHD.
Collapse
Affiliation(s)
| | - Maria Inge Lusida
- Institute of Tropical Disease, Universitas Airlangga, Surabaya, Indonesia. .,Department of Microbiology, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia.
| | - Soetjipto
- Department of Psychiatric, Faculty of Medicine Universitas Airlangga, Dr. Soetomo General Hospital, Surabaya, Indonesia
| | - Yunias Setiawati
- Department of Psychiatric, Faculty of Medicine Universitas Airlangga, Dr. Soetomo General Hospital, Surabaya, Indonesia
| |
Collapse
|
27
|
He JL, Wodka E, Tommerdahl M, Edden RAE, Mikkelsen M, Mostofsky SH, Puts NAJ. Disorder-specific alterations of tactile sensitivity in neurodevelopmental disorders. Commun Biol 2021; 4:97. [PMID: 33483581 PMCID: PMC7822903 DOI: 10.1038/s42003-020-01592-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 12/03/2020] [Indexed: 02/06/2023] Open
Abstract
Alterations of tactile processing have long been identified in autism spectrum disorders (ASD) and attention-deficit/hyperactivity disorder (ADHD). However, the extent to which these alterations are disorder-specific, rather than disorder-general, and how they relate to the core symptoms of each disorder, remains unclear. We measured and compared tactile detection, discrimination, and order judgment thresholds between a large sample of children with ASD, ADHD, ASD + ADHD combined and typically developing controls. The pattern of results suggested that while difficulties with tactile detection and order judgement were more common in children with ADHD, difficulties with tactile discrimination were more common in children with ASD. Interestingly, in our subsequent correlation analyses between tactile perception and disorder-specific clinical symptoms, tactile detection and order judgment correlated exclusively with the core symptoms of ADHD, while tactile discrimination correlated exclusively with the symptoms of ASD. When taken together, these results suggest that disorder-specific alterations of lower-level sensory processes exist and are specifically related to higher-level clinical symptoms of each disorder.
Collapse
Affiliation(s)
- Jason L He
- Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
- F. M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD, 21287, USA
- Department of Forensic and Neurodevelopmental Sciences, Sackler Institute for Translational Neurodevelopment, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, SE5 8AB, UK
| | - Ericka Wodka
- Center for Autism and Related Disorders, Kennedy Krieger Institute, Baltimore, MD, USA
- Department of Psychiatry and Behavioral Sciences, The Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Mark Tommerdahl
- Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27514, USA
| | - Richard A E Edden
- Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
- F. M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD, 21287, USA
| | - Mark Mikkelsen
- Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
- F. M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD, 21287, USA
| | - Stewart H Mostofsky
- Center for Autism and Related Disorders, Kennedy Krieger Institute, Baltimore, MD, USA
- Department of Psychiatry and Behavioral Sciences, The Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
- Center for Neurodevelopmental and Imaging Research, Kennedy Krieger Institute, Baltimore, MD, 21287, USA
- Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Nicolaas A J Puts
- Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA.
- F. M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD, 21287, USA.
- Department of Forensic and Neurodevelopmental Sciences, Sackler Institute for Translational Neurodevelopment, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, SE5 8AB, UK.
| |
Collapse
|
28
|
Mamiya PC, Arnett AB, Stein MA. Precision Medicine Care in ADHD: The Case for Neural Excitation and Inhibition. Brain Sci 2021; 11:brainsci11010091. [PMID: 33450814 PMCID: PMC7828220 DOI: 10.3390/brainsci11010091] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Revised: 12/15/2020] [Accepted: 01/11/2021] [Indexed: 12/14/2022] Open
Abstract
Attention-deficit/hyperactivity disorder (ADHD) is a neurodevelopmental disorder that has become increasingly prevalent worldwide. Its core symptoms, including difficulties regulating attention, activity level, and impulses, appear in early childhood and can persist throughout the lifespan. Current pharmacological options targeting catecholamine neurotransmissions have effectively alleviated symptoms in some, but not all affected individuals, leaving clinicians to implement trial-and-error approach to treatment. In this review, we discuss recent experimental evidence from both preclinical and human studies that suggest imbalance of excitation/inhibition (E/I) in the fronto-striatal circuitry during early development may lead to enduring neuroanatomical abnormality of the circuitry, causing persistence of ADHD symptoms in adulthood. We propose a model of precision medicine care that includes E/I balance as a candidate biomarker for ADHD, development of GABA-modulating medications, and use of magnetic resonance spectroscopy and scalp electrophysiology methods to monitor the effects of treatments on shifting E/I balance throughout the lifespan.
Collapse
Affiliation(s)
- Ping C. Mamiya
- Institute for Learning and Brain Sciences, University of Washington, Seattle, WA 98195, USA
- Correspondence:
| | - Anne B. Arnett
- Department of Psychiatry & Behavioral Sciences, University of Washington, Seattle, WA 98195, USA; (A.B.A.); (M.A.S.)
| | - Mark A. Stein
- Department of Psychiatry & Behavioral Sciences, University of Washington, Seattle, WA 98195, USA; (A.B.A.); (M.A.S.)
| |
Collapse
|
29
|
Zöllner HJ, Oeltzschner G, Schnitzler A, Wittsack HJ. In silico GABA+ MEGA-PRESS: Effects of signal-to-noise ratio and linewidth on modeling the 3 ppm GABA+ resonance. NMR IN BIOMEDICINE 2021; 34:e4410. [PMID: 32989890 PMCID: PMC8935357 DOI: 10.1002/nbm.4410] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Revised: 08/11/2020] [Accepted: 08/28/2020] [Indexed: 06/02/2023]
Abstract
To investigate the GABA+ modeling accuracy of MEGA-PRESS GABA+-edited MRS data with various spectral quality scenarios, the influence of varying signal-to-noise ratio (SNR) and linewidth on the model estimates was quantified. MEGA-PRESS data from 46 volunteers were averaged to generate a template MEGA-PRESS spectrum, which was modeled and quantified to generate a GABA+ level ground truth. This spectrum was then manipulated by adding 427 combinations of varying artificial noise levels and line broadening, mimicking variations in GABA+ SNR and B0 homogeneity. GABA+ modeling and quantification was performed with 100 simulated spectra per condition using automated routines in both Gannet 3.0 and Tarquin. The GABA+ estimation error was calculated as the relative deviation to the quantified GABA+ ground truth levels to assess the accuracy of GABA+ modeling. Finally, the accordance between the simulations and different in vivo scenarios was assessed. The GABA+ estimation error was smaller than 5% for all GABA+ SNR values with creatine linewidths lower than 9.7 Hz in Gannet 3.0 or unequal 10.6 Hz in Tarquin. The standard deviation of the GABA+ amplitude over 100 spectra per condition varied between 3.1 and 17% (Gannet 3.0) and between 1 and 11% (Tarquin) over the in vivo relevant GABA+ SNR range between 2.6 and 3.5. GABA+ edited studies might be realized for voxels with low GABA+ SNR at the cost of higher group-level variance. The accuracy of GABA+ modeling had no relation to commonly used quality metrics. The Tarquin algorithm was found to be more robust against linewidth changes than the fitting algorithm in Gannet.
Collapse
Affiliation(s)
- Helge Jörn Zöllner
- institute of Clinical Neuroscience and Medical Psychology, Medical Faculty, Heinrich Heine University Düsseldorf, Germany
- Department of Diagnostic and Interventional Radiology, Medical Faculty, Heinrich Heine University Düsseldorf, Germany
| | - Georg Oeltzschner
- 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
| | - Alfons Schnitzler
- institute of Clinical Neuroscience and Medical Psychology, Medical Faculty, Heinrich Heine University Düsseldorf, Germany
| | - Hans-Jörg Wittsack
- Department of Diagnostic and Interventional Radiology, Medical Faculty, Heinrich Heine University Düsseldorf, Germany
| |
Collapse
|
30
|
Abd-Elhakim YM, Abdel-Motal SM, Malhat SM, Mostafa HI, Moselhy AAA, Beheiry RR, Said EN. Curcumin mitigates neurotoxic and neurobehavioral changes of gentamicin and sodium salicylate in rats by adjusting oxidative stress and apoptosis. Life Sci 2020; 265:118824. [PMID: 33278387 DOI: 10.1016/j.lfs.2020.118824] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2020] [Revised: 11/10/2020] [Accepted: 11/24/2020] [Indexed: 02/08/2023]
Abstract
Currently, antibiotics and salicylates are the most highly consumed medications worldwide. The side effects of these pharmaceuticals on the nervous system have been little investigated. Thus, this study aimed to examine the influence of the gentamicin (GM) and sodium salicylates (SS) on neurobehavioral functions, including locomotors function, memory, and sensorimotor functions together with gamma-aminobutyric acid (GABA) neurotransmitter levels. Also, oxidative stress, lipid peroxidation, and apoptotic indicators of brain tissue were assessed. Additionally, the histopathological architecture of brain tissues was investigated. This study also evaluated the curcumin (CUR) efficacy to counteract the GM or SS induced neurotoxic impacts in rats. For this purpose, seven groups were administered physiological saline (1 ml/rat; orally), olive oil (1 ml/rat; orally), CUR (50 mg/kg bwt; orally), GM (120 mg/kg bwt; intraperitoneally), SS (300 mg /kg bwt; intraperitoneally), CUR + GM, or CUR + SS for consecutive 15 days. The results revealed that GM and SS exposure evoked impaired memory, sensorimotor deficit functions, and depressive-like behavior together with the depletion of GABA. GM and SS exposure elevated malondialdehyde and Caspase-3 levels, but total antioxidant capacity and Bcl-2 levels were reduced. Besides, GM and SS exposure induced distinct pathological perturbations in cerebral cortices and hippocampus tissues. CUR significantly reversed the GM and SS harmful impacts. In conclusion, these findings verified that CUR could be a biologically efficient protective intervention against GM and SS induced neurotoxic impacts and neurobehavioral aberrations.
Collapse
Affiliation(s)
- Yasmina M Abd-Elhakim
- Department of Forensic Medicine and Toxicology, Faculty of Veterinary Medicine, Zagazig University, Zagazig, Egypt.
| | - Sabry M Abdel-Motal
- Department of Pharmacology, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | | | - Hend I Mostafa
- Department of Pharmacology, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | - Attia A A Moselhy
- Department of Anatomy and Embryology, Faculty of Veterinary Medicine, Zagazig University, Egypt
| | - Rasha R Beheiry
- Department of Histology and Cytology, Faculty of Veterinary Medicine, Zagazig University, Zagazig, Egypt
| | - Enas N Said
- Department of Veterinary Public Health, Faculty of Veterinary Medicine, Zagazig University, Zagazig, Egypt
| |
Collapse
|
31
|
Hai T, Swansburg R, Kahl CK, Frank H, Lemay JF, MacMaster FP. Magnetic Resonance Spectroscopy of γ-Aminobutyric Acid and Glutamate Concentrations in Children With Attention-Deficit/Hyperactivity Disorder. JAMA Netw Open 2020; 3:e2020973. [PMID: 33064134 PMCID: PMC7568198 DOI: 10.1001/jamanetworkopen.2020.20973] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
This cohort study assesses concentrations of γ-aminobutyric acid (GABA) and glutamate in children with attention-deficit/hyperactivity disorder (ADHD) compared with typically developing children.
Collapse
Affiliation(s)
- Tasmia Hai
- Werklund School of Education, University of Calgary, Calgary, Alberta, Canada
- Department of Educational Psychology, University of Alberta, Edmonton, Alberta, Canada
| | - Rose Swansburg
- Department of Psychiatry, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Cynthia K. Kahl
- Department of Psychiatry, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Hannah Frank
- Department of Psychology, Mount Royal University, Calgary, Alberta, Canada
| | - Jean-François Lemay
- Department of Pediatrics, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Frank P. MacMaster
- Department of Psychiatry, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
32
|
Kim YS, Choi J, Yoon BE. Neuron-Glia Interactions in Neurodevelopmental Disorders. Cells 2020; 9:cells9102176. [PMID: 32992620 PMCID: PMC7601502 DOI: 10.3390/cells9102176] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Revised: 09/24/2020] [Accepted: 09/24/2020] [Indexed: 12/12/2022] Open
Abstract
Recent studies have revealed synaptic dysfunction to be a hallmark of various psychiatric diseases, and that glial cells participate in synapse formation, development, and plasticity. Glial cells contribute to neuroinflammation and synaptic homeostasis, the latter being essential for maintaining the physiological function of the central nervous system (CNS). In particular, glial cells undergo gliotransmission and regulate neuronal activity in tripartite synapses via ion channels (gap junction hemichannel, volume regulated anion channel, and bestrophin-1), receptors (for neurotransmitters and cytokines), or transporters (GLT-1, GLAST, and GATs) that are expressed on glial cell membranes. In this review, we propose that dysfunction in neuron-glia interactions may contribute to the pathogenesis of neurodevelopmental disorders. Understanding the mechanisms of neuron-glia interaction for synapse formation and maturation will contribute to the development of novel therapeutic targets of neurodevelopmental disorders.
Collapse
Affiliation(s)
- Yoo Sung Kim
- Department of Molecular Biology, Dankook University, Cheonan 31116, Korea; (Y.S.K.); (J.C.)
| | - Juwon Choi
- Department of Molecular Biology, Dankook University, Cheonan 31116, Korea; (Y.S.K.); (J.C.)
| | - Bo-Eun Yoon
- Department of Molecular Biology, Dankook University, Cheonan 31116, Korea; (Y.S.K.); (J.C.)
- Department of Nanobiomedical science, Dankook University, Cheonan 31116, Korea
- Correspondence: ; Tel.: +82-41-529-6085
| |
Collapse
|
33
|
Maier S, Tebartz van Elst L, Philipsen A, Lange T, Feige B, Glauche V, Nickel K, Matthies S, Alm B, Sobanski E, Domschke K, Perlov E, Endres D. Effects of 12-Week Methylphenidate Treatment on Neurometabolism in Adult Patients with ADHD: The First Double-Blind Placebo-Controlled MR Spectroscopy Study. J Clin Med 2020; 9:jcm9082601. [PMID: 32796630 PMCID: PMC7464267 DOI: 10.3390/jcm9082601] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Revised: 06/26/2020] [Accepted: 06/29/2020] [Indexed: 11/27/2022] Open
Abstract
Attention deficit hyperactivity disorder (ADHD) is a frequent neurodevelopmental disorder that often persists into adulthood. Methylphenidate (MPH) is the first-line treatment for ADHD; however, despite its wide usage, little is known about its neurometabolic effects. Until now, no randomized and blinded clinical trials have been conducted addressing the neurometabolic signals of MPH administration in adults with ADHD. In the current study, the authors investigated how MPH intake and group psychotherapy (GPT) influence brain neurometabolism over the course of three months. The authors hypothesized a decrease in the anterior cingulate cortex (ACC) glutamate concentration following MPH administration. This study was part of a double-blind multicenter trial (Comparison of Methylphenidate and Psychotherapy in Adult ADHD Study (COMPAS)) investigating the effects of MPH and GPT in patients with adult ADHD. Using single-voxel magnetic resonance spectroscopy (MRS) of the pregenual ACC and the left cerebellar hemisphere (CHL), we investigated the concentration of glutamate plus glutamine (Glx), N-acetyl-aspartate, creatine, total choline containing compounds, and myo-inositol in patients before and after 12 weeks of treatment. Neither MPH nor GPT significantly influenced the Glx concentration or any of the other metabolite concentrations in the ACC and CHL after 12 weeks. Therefore, contrary to the hypothesis, no change in the prefrontal Glx signal was detected after MPH treatment. Given that MRS does not differentiate between glutamate in the synaptic cleft and in neuronal tissue, MPH-induced down-regulation of glutamatergic neurotransmission in the ACC might only affect the concentration of glutamate in the synaptic cleft, while the general availability of glutamate in the respective neuronal tissue might be unaffected by MPH intake. The observed lack of any MPH-induced normalization in metabolite concentrations is less surprising, considering that the baseline sample did not significantly differ from a healthy control group. Future studies of other regions, such as the basal ganglia, and the use of novel methods, such as whole brain MRS and multimodal imaging approaches, are necessary.
Collapse
Affiliation(s)
- Simon Maier
- Section for Experimental Neuropsychiatry, Department of Psychiatry and Psychotherapy, Medical Center—University of Freiburg, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany; (S.M.); (B.F.); (K.N.); (E.P.); (D.E.)
- Department of Psychiatry and Psychotherapy, Medical Center—University of Freiburg, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany; (S.M.); (K.D.)
| | - Ludger Tebartz van Elst
- Section for Experimental Neuropsychiatry, Department of Psychiatry and Psychotherapy, Medical Center—University of Freiburg, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany; (S.M.); (B.F.); (K.N.); (E.P.); (D.E.)
- Department of Psychiatry and Psychotherapy, Medical Center—University of Freiburg, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany; (S.M.); (K.D.)
- Correspondence:
| | - Alexandra Philipsen
- Department of Psychiatry and Psychotherapy, University Hospital Bonn, 53105 Bonn, Germany;
| | - Thomas Lange
- Department of Radiology, Medical Physics, Medical Center—University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany;
| | - Bernd Feige
- Section for Experimental Neuropsychiatry, Department of Psychiatry and Psychotherapy, Medical Center—University of Freiburg, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany; (S.M.); (B.F.); (K.N.); (E.P.); (D.E.)
- Department of Psychiatry and Psychotherapy, Medical Center—University of Freiburg, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany; (S.M.); (K.D.)
| | - Volkmar Glauche
- Department of Neurology, Medical Center—University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany;
| | - Kathrin Nickel
- Section for Experimental Neuropsychiatry, Department of Psychiatry and Psychotherapy, Medical Center—University of Freiburg, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany; (S.M.); (B.F.); (K.N.); (E.P.); (D.E.)
- Department of Psychiatry and Psychotherapy, Medical Center—University of Freiburg, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany; (S.M.); (K.D.)
| | - Swantje Matthies
- Department of Psychiatry and Psychotherapy, Medical Center—University of Freiburg, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany; (S.M.); (K.D.)
| | - Barbara Alm
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty of Mannheim, University of Heidelberg, 68159 Mannheim, Germany; (B.A.); (E.S.)
| | - Esther Sobanski
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty of Mannheim, University of Heidelberg, 68159 Mannheim, Germany; (B.A.); (E.S.)
- Department of Child and Adolescent Psychiatry, University Medical Center Mainz, 55131 Mainz, Germany
| | - Katharina Domschke
- Department of Psychiatry and Psychotherapy, Medical Center—University of Freiburg, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany; (S.M.); (K.D.)
- Center for Basics in Neuromodulation, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Evgeniy Perlov
- Section for Experimental Neuropsychiatry, Department of Psychiatry and Psychotherapy, Medical Center—University of Freiburg, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany; (S.M.); (B.F.); (K.N.); (E.P.); (D.E.)
- Department of Psychiatry and Psychotherapy, Medical Center—University of Freiburg, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany; (S.M.); (K.D.)
- Clinic for Psychiatry Luzern, St. Urban, 4915 Luzern, Switzerland
| | - Dominique Endres
- Section for Experimental Neuropsychiatry, Department of Psychiatry and Psychotherapy, Medical Center—University of Freiburg, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany; (S.M.); (B.F.); (K.N.); (E.P.); (D.E.)
- Department of Psychiatry and Psychotherapy, Medical Center—University of Freiburg, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany; (S.M.); (K.D.)
| |
Collapse
|
34
|
Puts NA, Ryan M, Oeltzschner G, Horska A, Edden RAE, Mahone EM. Reduced striatal GABA in unmedicated children with ADHD at 7T. Psychiatry Res Neuroimaging 2020; 301:111082. [PMID: 32438277 DOI: 10.1016/j.pscychresns.2020.111082] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 03/30/2020] [Accepted: 03/31/2020] [Indexed: 12/12/2022]
Abstract
Attention-deficit hyperactive disorder (ADHD) is characterized by inattention and increased impulsive and hypermotoric behaviors.Despite the high prevalence and impact of ADHD, little is known about the underlying neurophysiology of ADHD. The main inhibitory and excitatory neurotransmitters γ-aminobutyric acid (GABA) and glutamate are receiving increased attention in ADHD and can be measured using Magnetic Resonance Spectroscopy (MRS). However, MRS studies in ADHD are limited. We measured GABA and glutamate in young unmedicated participants, utilizing high magnetic field strength. Fifty unmedicated children (26 with ADHD, 24 controls) aged 5-9 years completed MRS at 7T and behavioral testing. GABA and glutamate were measured in dorsolateral prefrontal cortex (DLPFC), anterior cingulate cortex (ACC), premotor cortex (PMC), and striatum, and estimated using LCModel. Children with ADHD showed poorer inhibitory control and significantly reduced GABA/Cr in the striatum, but not in ACC, DLPFC, or PMC regions. There were no significant group differences for Glu/Cr levels, or correlations with behavioral manifestations of ADHD. The primary finding of this study is a reduction of striatal GABA levels in unmedicated children with ADHD at 7T. These findings provide guidance for future studies or interventions. Reduced striatal GABA may be a marker for specific GABA-related treatment for ADHD.
Collapse
Affiliation(s)
- Nicolaas A Puts
- Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, 600 N Wolfe St., Baltimore, MD 21287, United States; F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, 707 N Broadway, Baltimore, MD 21205, United States; Department of Forensic and Neurodevelopmental Sciences, Sackler Institute for Translational Neurodevelopment, Institute of Psychiatry, Psychology and Neuroscience, King's College London, 16 De Crespigny Park, London, SE5 8AB, United Kingdom.
| | - Matthew Ryan
- Department of Neuropsychology, Kennedy Krieger Institute, 1750 E. Fairmount Ave., Baltimore, MD 21231 United States
| | - Georg Oeltzschner
- Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, 600 N Wolfe St., Baltimore, MD 21287, United States; F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, 707 N Broadway, Baltimore, MD 21205, United States
| | - Alena Horska
- Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, 600 N Wolfe St., Baltimore, MD 21287, United States
| | - Richard A E Edden
- Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, 600 N Wolfe St., Baltimore, MD 21287, United States; F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, 707 N Broadway, Baltimore, MD 21205, United States
| | - E Mark Mahone
- Department of Neuropsychology, Kennedy Krieger Institute, 1750 E. Fairmount Ave., Baltimore, MD 21231 United States; Department of Psychiatry and Behavioral Sciences, The Johns Hopkins University School of Medicine, 600 N Wolfe St., Baltimore, MD 21287, United States
| |
Collapse
|
35
|
Littlejohn BP, Price DM, Neuendorff DA, Carroll JA, Vann RC, Riggs PK, Riley DG, Long CR, Randel RD, Welsh TH. Influence of prenatal transportation stress-induced differential DNA methylation on the physiological control of behavior and stress response in suckling Brahman bull calves. J Anim Sci 2020; 98:skz368. [PMID: 31807776 PMCID: PMC6986441 DOI: 10.1093/jas/skz368] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2019] [Accepted: 12/05/2019] [Indexed: 02/06/2023] Open
Abstract
The objective of this experiment was to examine potential differential methylation of DNA as a mechanism for altered behavioral and stress responses in prenatally stressed (PNS) compared with nonprenatally stressed (Control) young bull calves. Mature Brahman cows (n = 48) were transported for 2-h periods at 60 ± 5, 80 ± 5, 100 ± 5, 120 ± 5, and 140 ± 5 d of gestation (Transported group) or maintained as nontransported Controls (n = 48). From the offspring born to Transported and Control cows, a subset of 28-d-old intact bulls (n = 7 PNS; n = 7 Control) were evaluated for methylation of DNA of behavior and stress response-associated genes. Methylation of DNA from white blood cells was assessed via reduced representation bisulfite sequencing methods. Because increased methylation of DNA within gene promoter regions has been associated with decreased transcriptional activity of the corresponding gene, differentially methylated (P ≤ 0.05) CG sites (cytosine followed by a guanine nucleotide) located within promoter regions (n = 1,205) were used to predict (using Ingenuity Pathway Analysis software) alterations to canonical pathways in PNS compared with Control bull calves. Among differentially methylated genes (P ≤ 0.05) related to behavior and the stress response were OPRK1, OPRM1, PENK, POMC, NR3C2, TH, DRD1, DRD5, COMT, HTR6, HTR5A, GABRA4, GABRQ, and GAD2. Among altered (P < 0.05) signaling pathways related to behavior and the stress response were Opioid Signaling, Corticotropin-Releasing Hormone Signaling, Dopamine Receptor Signaling, Dopamine-DARPP32 Feedback in cAMP Signaling, Serotonin Receptor Signaling, and GABA Receptor Signaling. Alterations to behavior and stress response-related genes and canonical pathways supported previously observed elevations in temperament score and serum cortisol through weaning in the larger population of PNS calves from which bulls in this study were derived. Differential methylation of DNA and predicted alterations to behavior and stress response-related pathways in PNS compared with Control bull calves suggest epigenetic programming of behavior and the stress response in utero.
Collapse
Affiliation(s)
- Brittni P Littlejohn
- Texas A&M AgriLife Research & Extension Center, Overton, TX
- Department of Animal Science, Texas A&M University, and Texas A&M AgriLife Research, College Station, TX
| | - Deborah M Price
- Texas A&M AgriLife Research & Extension Center, Overton, TX
- Department of Animal Science, Texas A&M University, and Texas A&M AgriLife Research, College Station, TX
| | | | | | - Rhonda C Vann
- Mississippi Agricultural and Forestry Experiment Station, Mississippi State University, Raymond, MS
| | - Penny K Riggs
- Department of Animal Science, Texas A&M University, and Texas A&M AgriLife Research, College Station, TX
| | - David G Riley
- Department of Animal Science, Texas A&M University, and Texas A&M AgriLife Research, College Station, TX
| | - Charles R Long
- Texas A&M AgriLife Research & Extension Center, Overton, TX
- Department of Animal Science, Texas A&M University, and Texas A&M AgriLife Research, College Station, TX
| | | | - Thomas H Welsh
- Department of Animal Science, Texas A&M University, and Texas A&M AgriLife Research, College Station, TX
| |
Collapse
|
36
|
The Potential Influence of the Bacterial Microbiome on the Development and Progression of ADHD. Nutrients 2019; 11:nu11112805. [PMID: 31744191 PMCID: PMC6893446 DOI: 10.3390/nu11112805] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 11/11/2019] [Accepted: 11/13/2019] [Indexed: 02/06/2023] Open
Abstract
The latest research cumulates staggering information about the correlation between the microbiota-gut-brain axis and neurodevelopmental disorders. This review aims to shed light on the potential influence of the microbiome on the development of the most prevalent neurodevelopmental disease, attention-deficit-hyperactive disorder (ADHD). As the etiology and pathophysiology of ADHD are still unclear, finding viable biomarkers and effective treatment still represent a challenge. Therefore, we focused on factors that have been associated with a higher risk of developing ADHD, while simultaneously influencing the microbial composition. We reviewed the effect of a differing microbial makeup on neurotransmitter concentrations important in the pathophysiology of ADHD. Additionally, we deduced factors that correlate with a high prevalence of ADHD, while simultaneously affecting the gut microbiome, such as emergency c-sections, and premature birth as the former leads to a decrease of the gut microbial diversity and the latter causes neuroprotective Lactobacillus levels to be reduced. Also, we assessed nutritional influences, such as breastfeeding, ingestion of short-chain fatty acids (SCFAs) and polyunsaturated fatty acids (PUFAs) on the host′s microbiome and development of ADHD. Finally, we discussed the potential significance of Bifidobacterium as a biomarker for ADHD, the importance of preventing premature birth as prophylaxis and nutrition as a prospective therapeutic measurement against ADHD.
Collapse
|
37
|
Dunn GA, Nigg JT, Sullivan EL. Neuroinflammation as a risk factor for attention deficit hyperactivity disorder. Pharmacol Biochem Behav 2019; 182:22-34. [PMID: 31103523 PMCID: PMC6855401 DOI: 10.1016/j.pbb.2019.05.005] [Citation(s) in RCA: 138] [Impact Index Per Article: 27.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2018] [Revised: 05/08/2019] [Accepted: 05/14/2019] [Indexed: 01/08/2023]
Abstract
Attention Deficit Hyperactivity Disorder (ADHD) is a persistent, and impairing pediatric-onset neurodevelopmental condition. Its high prevalence, and recurrent controversy over its widespread identification and treatment, drive strong interest in its etiology and mechanisms. Emerging evidence for a role for neuroinflammation in ADHD pathophysiology is of great interest. This evidence includes 1) the above-chance comorbidity of ADHD with inflammatory and autoimmune disorders, 2) initial studies indicating an association with ADHD and increased serum cytokines, 3) preliminary evidence from genetic studies demonstrating associations between polymorphisms in genes associated with inflammatory pathways and ADHD, 4) emerging evidence that early life exposure to environmental factors may increase risk for ADHD via an inflammatory mechanism, and 5) mechanistic evidence from animal models of maternal immune activation documenting behavioral and neural outcomes consistent with ADHD. Prenatal exposure to inflammation is associated with changes in offspring brain development including reductions in cortical gray matter volume and the volume of certain cortical areas -parallel to observations associated with ADHD. Alterations in neurotransmitter systems, including the dopaminergic, serotonergic and glutamatergic systems, are observed in ADHD populations. Animal models provide strong evidence that development and function of these neurotransmitters systems are sensitive to exposure to in utero inflammation. In summary, accumulating evidence from human studies and animal models, while still incomplete, support a potential role for neuroinflammation in the pathophysiology of ADHD. Confirmation of this association and the underlying mechanisms have become valuable targets for research. If confirmed, such a picture may be important in opening new intervention routes.
Collapse
Affiliation(s)
| | - Joel T Nigg
- Oregon Health and Science University, United States of America
| | - Elinor L Sullivan
- University of Oregon, United States of America; Oregon Health and Science University, United States of America; Oregon National Primate Research Center, United States of America.
| |
Collapse
|
38
|
Association between prefrontal glutamine levels and neuroticism determined using proton magnetic resonance spectroscopy. Transl Psychiatry 2019; 9:170. [PMID: 31213596 PMCID: PMC6581909 DOI: 10.1038/s41398-019-0500-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Revised: 04/23/2019] [Accepted: 04/29/2019] [Indexed: 12/19/2022] Open
Abstract
There is growing evidence for GABA and glutamate-glutamine dysfunction in the pathogenesis of mood and anxiety disorders. It is important to study this pathology in the early phases of the illness in order to develop new approaches to secondary prevention. New magnetic resonance spectroscopy (MRS) measures allow determining glutamine, the principal metabolite of synaptic glutamate that is directly related to glutamate levels in the synaptic cleft, as well as glutamate and GABA. In contrast to previous investigations, this study used community-based recruitment methods and a combined categorical and dimensional approach to psychopathology. In the study protocol, neuroticism was defined as the primary outcome. Neuroticism shares a large proportion of its genetic variance with mood and anxiety disorders. We examined young adult participants recruited from the general population in a cross-sectional study using 3-T 1H-MRS with one voxel in the left dorsolateral prefrontal cortex (DLPFC). The total sample of N = 110 (61 females) included 18 individuals suffering from MDD and 19 individuals suffering from DSM-IV anxiety disorders. We found that glutamine and glutamine-to-glutamate ratio were correlated with neuroticism in the whole sample (r = 0.263, p = 0.005, and n = 110; respectively, r = 0.252, p = 0.008, and n = 110), even when controlling for depression and anxiety disorder diagnoses (for glutamine: beta = 0.220, p = 0.047, and n = 110). Glutamate and GABA were not significantly correlated with neuroticism (r = 0.087, p = 0.365, and n = 110; r = -0.044, p = 0.645, and n = 110). Lack of self-confidence and emotional instability were the clinical correlates of glutamate-glutamine dysfunction. In conclusion, this study suggests that prefrontal glutamine is increased in early phases of mood and anxiety disorders. Further understanding of glutamate-glutamine dysfunction in stress-related disorders may lead to new therapeutic strategies to prevent and treat these disorders.
Collapse
|
39
|
Sandgren AM, Brummer RJ. ADHD-originating in the gut? The emergence of a new explanatory model. Med Hypotheses 2018; 120:135-145. [DOI: 10.1016/j.mehy.2018.08.022] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Accepted: 08/25/2018] [Indexed: 12/12/2022]
|
40
|
O'Gorman Tuura R, Warnock G, Ametamey S, Treyer V, Noeske R, Buck A, Sommerauer M. Imaging glutamate redistribution after acute N-acetylcysteine administration: A simultaneous PET/MR study. Neuroimage 2018; 184:826-833. [PMID: 30296554 DOI: 10.1016/j.neuroimage.2018.10.017] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 08/31/2018] [Accepted: 10/04/2018] [Indexed: 12/25/2022] Open
Abstract
Glutamate is the most abundant excitatory neurotransmitter in the human brain, but in vivo imaging of acute fluctuations in glutamatergic levels has not been well established. The purpose of this study was to examine acute changes in glutamate after stimulation with N-acetylcysteine (NAC) using a simultaneous positron emission tomography/magnetic resonance spectroscopy (PET/MRS) approach. Ten healthy adult males were examined in two scanning sessions, and 5g NAC was administered 1 h prior to one of the scan sessions. Simultaneous PET/MR data were acquired using an integrated 3T PET/MR scanner. Glutamate (Glu), glutamine (Gln), and glutamate + glutamine (Glx) levels were assessed from MRS data collected from the basal ganglia with PRESS and from the left prefrontal cortex with PRESS and MEGAPRESS, and mGluR5 binding (BPND) was assessed from PET data collected with [18F]PSS232. NAC administration was associated with a significant reduction in Glx and Gln in the basal ganglia spectra, and in Glx in the frontal MEGAPRESS spectra (p < 0.05); no differences in [18F]PSS232 BPND were observed with NAC, although a correlation between pre-/post-treatment Glx and baseline BPnd was found. The MRS-visible Glx signal is sensitive to acute fluctuations in glutamate. The change in Glx was mostly driven by a change in Gln, lending weight to the notion that Gln can provide a proxy marker for neurotransmitter/synaptic glutamate. [18F]PSS232 binding is not sensitive to acute glutamate shifts independently, but was associated with the extent of glutamate liberation upon NAC stimulation.
Collapse
Affiliation(s)
- Ruth O'Gorman Tuura
- Center for MR Research, University Children's Hospital, Steinwiesstrasse 75, CH-8032, Zürich, Switzerland.
| | - Geoff Warnock
- Department of Nuclear Medicine, University Hospital Zürich, Rämistrasse 100, CH-8091, Zürich, Switzerland
| | - Simon Ametamey
- Center for Radiopharmaceutical Sciences, Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology (ETH), Vladimir-Prelog-Weg 4 4, CH-8093, Zürich, Switzerland
| | - Valerie Treyer
- Department of Nuclear Medicine, University Hospital Zürich, Rämistrasse 100, CH-8091, Zürich, Switzerland
| | | | - Alfred Buck
- Department of Nuclear Medicine, University Hospital Zürich, Rämistrasse 100, CH-8091, Zürich, Switzerland
| | - Michael Sommerauer
- Department of Nuclear Medicine, University Hospital Zürich, Rämistrasse 100, CH-8091, Zürich, Switzerland; Department of Neurology, University Hospital Cologne, Kerpener Str. 62, 50937, Cologne, Germany
| |
Collapse
|
41
|
Bauer J, Werner A, Kohl W, Kugel H, Shushakova A, Pedersen A, Ohrmann P. Hyperactivity and impulsivity in adult attention-deficit/hyperactivity disorder is related to glutamatergic dysfunction in the anterior cingulate cortex. World J Biol Psychiatry 2018; 19:538-546. [PMID: 27973969 DOI: 10.1080/15622975.2016.1262060] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
OBJECTIVES Attention-deficit/hyperactivity disorder (ADHD) is closely linked to the dysregulation of dopaminergic and noradrenergic neurotransmission in the fronto-striatal neural network, including the anterior cingulate cortex (ACC) and the dorsolateral prefrontal cortex (DLPFC). Additionally, increasing evidence supports the involvement of the glutamatergic system in the pathophysiology of ADHD. Impulsivity, a core symptom in patients with ADHD, has been repeatedly associated with glutamatergic neurotransmission, and pharmacological treatment of ADHD has been shown to reduce glutamate levels in the prefrontal cortex. METHODS We investigated glutamate levels in the ACC and the DLPFC in 30 adults with ADHD and 30 healthy controls using single-voxel proton magnetic resonance spectroscopy on a 3T scanner. RESULTS The ADHD group showed a significant increase in glutamate in the ACC compared to controls, no significant differences in metabolites were observed in the DLPFC. Overall, glutamate levels in the ACC were positively correlated with ADHD symptomatology, especially hyperactivity and impulsivity symptoms. CONCLUSIONS Increased levels of glutamate in the ACC, which were positively correlated with hyperactivity and impulsivity, support the hypothesis that dysfunctional glutamatergic neurotransmission is at least partially responsible for ADHD symptomatology. Modulation of glutamatergic neurotransmission might therefore be a promising avenue for future pharmacological interventions.
Collapse
Affiliation(s)
- Jochen Bauer
- a Institute for Clinical Radiology, School of Medicine , University of Muenster , Germany
| | - Anne Werner
- b Department of Psychiatry, School of Medicine , University of Muenster , Germany
| | - Waldemar Kohl
- b Department of Psychiatry, School of Medicine , University of Muenster , Germany
| | - Harald Kugel
- a Institute for Clinical Radiology, School of Medicine , University of Muenster , Germany
| | - Anna Shushakova
- c Clinical Psychology and Psychotherapy, Institute of Psychology , University of Kiel , Germany
| | - Anya Pedersen
- c Clinical Psychology and Psychotherapy, Institute of Psychology , University of Kiel , Germany
| | - Patricia Ohrmann
- b Department of Psychiatry, School of Medicine , University of Muenster , Germany
| |
Collapse
|
42
|
Volk C, Jaramillo V, Merki R, O'Gorman Tuura R, Huber R. Diurnal changes in glutamate + glutamine levels of healthy young adults assessed by proton magnetic resonance spectroscopy. Hum Brain Mapp 2018; 39:3984-3992. [PMID: 29885049 DOI: 10.1002/hbm.24225] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Revised: 05/09/2018] [Accepted: 05/10/2018] [Indexed: 11/08/2022] Open
Abstract
The glutamatergic α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor is involved in synaptic plasticity processes, and animal studies have demonstrated altered expression across the sleep wake cycle. Accordingly, glutamate levels are reduced during non-rapid eye movement (NREM) sleep and the rate of this decrease is positively correlated with sleep EEG slow wave activity (SWA). Here, we combined proton magnetic resonance spectroscopy (1 H-MRS) and high-density sleep EEG to assess if 1 H-MRS is sensitive to diurnal changes of glutamate + glutamine (GLX) in healthy young adults and if potential overnight changes of GLX are correlated to SWA. 1 H-MRS was measured in the parietal lobe in the evening and in the subsequent morning. High-density sleep EEG was recorded overnight between the evening and morning scans. Our results revealed a significant overnight reduction in GLX, but no significant changes in other metabolites. The decrease in GLX positively correlated with the decrease of SWA. Our study demonstrates that quantification of diurnal changes in GLX is possible by means of 1 H-MRS and indicates that overnight changes in GLX are related to SWA, a marker that is closely linked to the restorative function of sleep. This relationship might be of particular interest in clinical populations in which sleep is disturbed.
Collapse
Affiliation(s)
- Carina Volk
- Children's Research Center, University Children's Hospital Zurich, Zürich, Switzerland.,Child Development Center, University Children's Hospital Zurich, Zürich, Switzerland
| | - Valeria Jaramillo
- Children's Research Center, University Children's Hospital Zurich, Zürich, Switzerland.,Child Development Center, University Children's Hospital Zurich, Zürich, Switzerland
| | - Renato Merki
- Children's Research Center, University Children's Hospital Zurich, Zürich, Switzerland.,Child Development Center, University Children's Hospital Zurich, Zürich, Switzerland
| | - Ruth O'Gorman Tuura
- Children's Research Center, University Children's Hospital Zurich, Zürich, Switzerland.,MR Center, University Children's Hospital Zurich, Zürich, Switzerland
| | - Reto Huber
- Children's Research Center, University Children's Hospital Zurich, Zürich, Switzerland.,Child Development Center, University Children's Hospital Zurich, Zürich, Switzerland.,Department of Child and Adolescent Psychiatry and Psychotherapy, Psychiatric Hospital University of Zurich, Zürich, Switzerland
| |
Collapse
|
43
|
Binocular rivalry transitions predict inattention symptom severity in adult ADHD. Eur Arch Psychiatry Clin Neurosci 2018; 268:373-382. [PMID: 28409230 DOI: 10.1007/s00406-017-0790-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Accepted: 03/20/2017] [Indexed: 12/13/2022]
Abstract
Attention deficit and hyperactivity disorder (ADHD) is a prevalent childhood disorder that is often maintained throughout the development and persists into adulthood. Established etiology models suggest that deficient inhibition underlies the core ADHD symptoms. While experimental evidence for impaired motor inhibition is overwhelming, little is known about the sensory inhibition processes, their changes throughout the development, and the relationship to ADHD symptoms. Here, we used the well-established binocular rivalry (BR) paradigm to investigate for the very first time the inhibitory processes related to visual perception in adults with ADHD. In BR, perception alternates between two dichoptically presented images throughout the viewing period, with shorter dominant percept durations and longer transition periods indicating poorer suppression/inhibition. Healthy controls (N = 28) and patients with ADHD (N = 32) were presented with two dissimilar images (orthogonal gratings) separately to each eye through a mirror stereoscope and asked to report their perceptual experiences. There were no differences between groups in any of the BR markers. However, an association between transition durations and symptom severity emerged in the ADHD group. Importantly, an exploratory multiple regression analysis revealed that inattention symptoms were the sole predictor for the duration of transition periods. The lack of impairments to sensory inhibition in adult, but not pediatric ADHD may reflect compensatory changes associated with development, while a correlation between inhibition and inattention symptoms may reveal an invariant core of the disorder.
Collapse
|
44
|
Fonseca BM, Cristóvão AC, Alves G. An easy-to-use liquid chromatography method with fluorescence detection for the simultaneous determination of five neuroactive amino acids in different regions of rat brain. J Pharmacol Toxicol Methods 2018; 91:72-79. [DOI: 10.1016/j.vascn.2018.02.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Revised: 01/27/2018] [Accepted: 02/02/2018] [Indexed: 11/17/2022]
|
45
|
Quansah E, Ruiz-Rodado V, Grootveld M, Zetterström TSC. Methylphenidate alters monoaminergic and metabolic pathways in the cerebellum of adolescent rats. Eur Neuropsychopharmacol 2018; 28:513-528. [PMID: 29478746 DOI: 10.1016/j.euroneuro.2018.02.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Revised: 02/01/2018] [Accepted: 02/07/2018] [Indexed: 12/31/2022]
Abstract
Abnormalities in the cerebellar circuitry have been suggested to contribute to some of the symptoms associated with attention deficit hyperactivity disorder (ADHD). The psychostimulant methylphenidate (MPH) is the major drug for treating this condition. Here, the effects of acute (2.0 mg/kg and 5.0 mg/kg) and chronic (2.0 mg/kg, twice daily for 15 days) MPH treatments were investigated in adolescent (35-40 days old) rats on monoaminergic and metabolic markers in the cerebellum. Data acquired indicates that acute MPH treatment (2.0 mg/kg) decreased cerebellar vesicular monoamine transporter (VMAT2) density, while chronic treatment caused an increase. In contrast, protein levels of tyrosine hydroxylase (TH) and the dopamine D1 receptor were not significantly altered by neither acute nor chronic MPH treatment. In addition, while chronic but not acute MPH treatment significantly enhanced dopamine turnover (DOPAC/dopamine) in the cerebellum, levels of dopamine and homovanillic acid (HVA) were not altered. Acute MPH (5.0 mg/kg) significantly modified levels of a range of cerebellar metabolites with similar trends also detected for the lower dose (2.0 mg/kg). In this regard, acute MPH tended to decrease cerebellar metabolites associated with energy consumption and excitatory neurotransmission including glutamate, glutamine, N-acetyl aspartate, and inosine. Conversely, levels of some metabolites associated with inhibitory neurotransmission, including GABA and glycine were reduced by acute (5.0 mg/kg) MPH, together with acetate, aspartate and hypoxanthine. In conclusion, this study demonstrated that MPH alters cerebellar biochemistry, and that this effect depends on both dose and duration of treatment. The therapeutic significance of these results requires further investigation.
Collapse
Affiliation(s)
- Emmanuel Quansah
- Leicester School of Pharmacy, Faculty of Health and Life Sciences, De Montfort University, The Gateway, Leicester LE1 9BH, UK
| | - Victor Ruiz-Rodado
- Neuro-Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Martin Grootveld
- Leicester School of Pharmacy, Faculty of Health and Life Sciences, De Montfort University, The Gateway, Leicester LE1 9BH, UK
| | - Tyra S C Zetterström
- Leicester School of Pharmacy, Faculty of Health and Life Sciences, De Montfort University, The Gateway, Leicester LE1 9BH, UK.
| |
Collapse
|
46
|
Smith KM. Hyperactivity in mice lacking one allele of the glutamic acid decarboxylase 67 gene. ACTA ACUST UNITED AC 2018; 10:267-271. [PMID: 29556956 DOI: 10.1007/s12402-018-0254-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Accepted: 03/08/2018] [Indexed: 12/17/2022]
Abstract
GABAergic interneuron loss, maturational delay or imbalance of glutamatergic to GABAergic signaling has been implicated in several neuropsychiatric disorders including Tourette syndrome and attention-deficit/hyperactivity disorder (ADHD). In schizophrenia, decreases in parvalbumin (PV), somatostatin (Sst) and glutamic acid decarboxylase (GAD) RNA have been observed and seem to indicate a failure in maturation in PV and Sst neurons. In Tourette syndrome, which has a high level of comorbid ADHD, reduced numbers of parvalbumin expressing neurons have been observed in the basal ganglia of affected patients. In addition, polymorphisms in the GAD1 gene that codes for GAD67 protein have been associated with ADHD. We have examined whether mice with a disrupted Gad67 allele, the Gad67 GFP knock-in mice (Gad67-GFP+/-), display abnormal locomotor behavior or altered anxiety behavior on the elevated plus maze. We found that Gad67-GFP+/- mice displayed a mild hyperactivity compared to control littermates.
Collapse
Affiliation(s)
- Karen Müller Smith
- Department of Biology, University of Louisiana at Lafayette, P.O. Box 43602, Lafayette, LA, 70504-3602, USA.
- Child Study Center, Yale University, New Haven, CT, 06520, USA.
| |
Collapse
|
47
|
Rudy M, Mayer-Proschel M. Iron Deficiency Affects Seizure Susceptibility in a Time- and Sex-Specific Manner. ASN Neuro 2017; 9:1759091417746521. [PMID: 29243938 PMCID: PMC5734468 DOI: 10.1177/1759091417746521] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Iron deficiency (ID) affects more than three billion people worldwide making it the most common micronutrient deficiency. ID is most prevalent during gestation and early life, which is of particular concern since its impact on the developing central nervous system is associated with an increased risk of a wide range of different psychiatric disorders later in life. The cause for this association is not known, but many of these same disorders are also associated with an imbalance between excitation and inhibition (E/I) within the brain. Based on this shared impairment, we asked whether ID could contribute to such an imbalance. Disruptions in the E/I balance can be uncovered by the brain’s response to seizure inducing insults. We therefore tested the seizure threshold under different nutritional models of ID. We found that mice which were postnatally exposed to ID (and were acutely ID) had a decreased seizure threshold and increased susceptibility to certain seizure types. In contrast, mice that were exposed to ID only during gestation had an increased seizure threshold and low seizure incidence. We suggest that exposure to ID during gestation might alter the cellular components that contribute to the establishment of a proper E/I balance later in life. In addition, our data highlight the importance of considering the window of vulnerability since gestational ID and postnatal ID have significantly different consequences on seizure probability.
Collapse
Affiliation(s)
- Michael Rudy
- 1 Department of Environmental Medicine, University of Rochester, NY, USA.,2 Department of Biomedical Genetics, University of Rochester, NY, USA
| | - Margot Mayer-Proschel
- 2 Department of Biomedical Genetics, University of Rochester, NY, USA.,3 Department of Neuroscience, University of Rochester, NY, USA
| |
Collapse
|
48
|
Medial Frontal Lobe Neurochemistry in Autism Spectrum Disorder is Marked by Reduced N-Acetylaspartate and Unchanged Gamma-Aminobutyric Acid and Glutamate + Glutamine Levels. J Autism Dev Disord 2017; 48:1467-1482. [PMID: 29177616 DOI: 10.1007/s10803-017-3406-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
49
|
Mikkelsen M, Barker PB, Bhattacharyya PK, Brix MK, Buur PF, Cecil KM, Chan KL, Chen DYT, Craven AR, Cuypers K, Dacko M, Duncan NW, Dydak U, Edmondson DA, Ende G, Ersland L, Gao F, Greenhouse I, Harris AD, He N, Heba S, Hoggard N, Hsu TW, Jansen JFA, Kangarlu A, Lange T, Lebel RM, Li Y, Lin CYE, Liou JK, Lirng JF, Liu F, Ma R, Maes C, Moreno-Ortega M, Murray SO, Noah S, Noeske R, Noseworthy MD, Oeltzschner G, Prisciandaro JJ, Puts NAJ, Roberts TPL, Sack M, Sailasuta N, Saleh MG, Schallmo MP, Simard N, Swinnen SP, Tegenthoff M, Truong P, Wang G, Wilkinson ID, Wittsack HJ, Xu H, Yan F, Zhang C, Zipunnikov V, Zöllner HJ, Edden RAE. Big GABA: Edited MR spectroscopy at 24 research sites. Neuroimage 2017; 159:32-45. [PMID: 28716717 PMCID: PMC5700835 DOI: 10.1016/j.neuroimage.2017.07.021] [Citation(s) in RCA: 121] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2017] [Revised: 06/20/2017] [Accepted: 07/11/2017] [Indexed: 12/14/2022] Open
Abstract
Magnetic resonance spectroscopy (MRS) is the only biomedical imaging method that can noninvasively detect endogenous signals from the neurotransmitter γ-aminobutyric acid (GABA) in the human brain. Its increasing popularity has been aided by improvements in scanner hardware and acquisition methodology, as well as by broader access to pulse sequences that can selectively detect GABA, in particular J-difference spectral editing sequences. Nevertheless, implementations of GABA-edited MRS remain diverse across research sites, making comparisons between studies challenging. This large-scale multi-vendor, multi-site study seeks to better understand the factors that impact measurement outcomes of GABA-edited MRS. An international consortium of 24 research sites was formed. Data from 272 healthy adults were acquired on scanners from the three major MRI vendors and analyzed using the Gannet processing pipeline. MRS data were acquired in the medial parietal lobe with standard GABA+ and macromolecule- (MM-) suppressed GABA editing. The coefficient of variation across the entire cohort was 12% for GABA+ measurements and 28% for MM-suppressed GABA measurements. A multilevel analysis revealed that most of the variance (72%) in the GABA+ data was accounted for by differences between participants within-site, while site-level differences accounted for comparatively more variance (20%) than vendor-level differences (8%). For MM-suppressed GABA data, the variance was distributed equally between site- (50%) and participant-level (50%) differences. The findings show that GABA+ measurements exhibit strong agreement when implemented with a standard protocol. There is, however, increased variability for MM-suppressed GABA measurements that is attributed in part to differences in site-to-site data acquisition. This study's protocol establishes a framework for future methodological standardization of GABA-edited MRS, while the results provide valuable benchmarks for the MRS community.
Collapse
Affiliation(s)
- Mark Mikkelsen
- Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, USA; F. M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD, USA
| | - Peter B Barker
- Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, USA; F. M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD, USA
| | - Pallab K Bhattacharyya
- Imaging Institute, Cleveland Clinic Foundation, Cleveland, OH, USA; Radiology, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, USA
| | - Maiken K Brix
- Department of Radiology, Haukeland University Hospital, Bergen, Norway; Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | - Pieter F Buur
- Spinoza Centre for Neuroimaging, Amsterdam, The Netherlands
| | - Kim M Cecil
- Department of Radiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Kimberly L Chan
- Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, USA; F. M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD, USA; Department of Biomedical Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - David Y-T Chen
- Department of Radiology, Taipei Medical University Shuang Ho Hospital, New Taipei City, Taiwan
| | - Alexander R Craven
- Department of Biological and Medical Psychology, University of Bergen, Bergen, Norway; NORMENT - Norwegian Center for Mental Disorders Research, University of Bergen, Bergen, Norway
| | - Koen Cuypers
- Department of Kinesiology, KU Leuven, Leuven, Belgium; REVAL Rehabilitation Research Center, Hasselt University, Diepenbeek, Belgium
| | - Michael Dacko
- Department of Radiology, Medical Physics, Medical Center - University of Freiburg, Faculty of Medicine, Freiburg, Germany
| | - Niall W Duncan
- Brain and Consciousness Research Centre, Taipei Medical University, Taipei, Taiwan
| | - Ulrike Dydak
- School of Health Sciences, Purdue University, West Lafayette, IN, USA
| | - David A Edmondson
- School of Health Sciences, Purdue University, West Lafayette, IN, USA
| | - Gabriele Ende
- Department of Neuroimaging, Central Institute of Mental Health, Mannheim, Germany
| | - Lars Ersland
- Department of Biological and Medical Psychology, University of Bergen, Bergen, Norway; NORMENT - Norwegian Center for Mental Disorders Research, University of Bergen, Bergen, Norway; Department of Clinical Engineering, Haukeland University Hospital, Bergen, Norway
| | - Fei Gao
- Shandong Medical Imaging Research Institute, Shandong University, Jinan, China
| | - Ian Greenhouse
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA, USA
| | - Ashley D Harris
- Department of Radiology, University of Calgary, Calgary, AB, Canada
| | - Naying He
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Stefanie Heba
- Department of Neurology, BG University Hospital Bergmannsheil, Bochum, Germany
| | - Nigel Hoggard
- Academic Unit of Radiology, University of Sheffield, Sheffield, UK
| | - Tun-Wei Hsu
- Department of Radiology, Taipei Veterans General Hospital, National Yang-Ming University School of Medicine, Taipei, Taiwan
| | - Jacobus F A Jansen
- Department of Radiology, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Alayar Kangarlu
- Department of Psychiatry, Columbia University, New York, NY, USA; New York State Psychiatric Institute, New York, NY, USA
| | - Thomas Lange
- Department of Radiology, Medical Physics, Medical Center - University of Freiburg, Faculty of Medicine, Freiburg, Germany
| | | | - Yan Li
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | | | - Jy-Kang Liou
- Department of Radiology, Taipei Veterans General Hospital, National Yang-Ming University School of Medicine, Taipei, Taiwan
| | - Jiing-Feng Lirng
- Department of Radiology, Taipei Veterans General Hospital, National Yang-Ming University School of Medicine, Taipei, Taiwan
| | - Feng Liu
- New York State Psychiatric Institute, New York, NY, USA
| | - Ruoyun Ma
- School of Health Sciences, Purdue University, West Lafayette, IN, USA
| | - Celine Maes
- Department of Kinesiology, KU Leuven, Leuven, Belgium
| | | | - Scott O Murray
- Department of Psychology, University of Washington, Seattle, WA, USA
| | - Sean Noah
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA, USA
| | | | - Michael D Noseworthy
- Department of Electrical and Computer Engineering, McMaster University, Hamilton, ON, Canada
| | - Georg Oeltzschner
- Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, USA; F. M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD, USA
| | - James J Prisciandaro
- Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC, USA
| | - Nicolaas A J Puts
- Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, USA; F. M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD, USA
| | - Timothy P L Roberts
- Department of Radiology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Markus Sack
- Department of Neuroimaging, Central Institute of Mental Health, Mannheim, Germany
| | - Napapon Sailasuta
- Research Imaging Centre, Centre for Addiction and Mental Health, Toronto, ON, Canada; Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Muhammad G Saleh
- Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, USA; F. M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD, USA
| | | | - Nicholas Simard
- School of Biomedical Engineering, McMaster University, Hamilton, ON, Canada
| | - Stephan P Swinnen
- Department of Kinesiology, KU Leuven, Leuven, Belgium; Leuven Research Institute for Neuroscience & Disease (LIND), KU Leuven, Leuven, Belgium
| | - Martin Tegenthoff
- Department of Neurology, BG University Hospital Bergmannsheil, Bochum, Germany
| | - Peter Truong
- Research Imaging Centre, Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - Guangbin Wang
- Shandong Medical Imaging Research Institute, Shandong University, Jinan, China
| | - Iain D Wilkinson
- Academic Unit of Radiology, University of Sheffield, Sheffield, UK
| | - Hans-Jörg Wittsack
- Department of Diagnostic and Interventional Radiology, Medical Faculty, Heinrich-Heine-University, Duesseldorf, Germany
| | - Hongmin Xu
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fuhua Yan
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chencheng Zhang
- Department of Functional Neurosurgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Vadim Zipunnikov
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Helge J Zöllner
- Department of Diagnostic and Interventional Radiology, Medical Faculty, Heinrich-Heine-University, Duesseldorf, Germany; Institute of Clinical Neuroscience and Medical Psychology, Medical Faculty, Heinrich-Heine-University, Duesseldorf, Germany
| | - Richard A E Edden
- Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, USA; F. M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD, USA.
| |
Collapse
|
50
|
Puts NAJ, Harris AD, Mikkelsen M, Tommerdahl M, Edden RAE, Mostofsky SH. Altered tactile sensitivity in children with attention-deficit hyperactivity disorder. J Neurophysiol 2017; 118:2568-2578. [PMID: 28768738 DOI: 10.1152/jn.00087.2017] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Revised: 08/01/2017] [Accepted: 08/01/2017] [Indexed: 11/22/2022] Open
Abstract
Attention-deficit hyperactivity disorder (ADHD) is characterized by an inability to concentrate, heightened activity, and hypermotoric behavior, but sensory (e.g., tactile) problems are common. The literature on tactile impairments in ADHD is limited, with most work employing clinical observations or questionnaires. We studied tactile processing in children with ADHD and hypothesized that children with ADHD would show reduced performance in tasks closely linked to inhibition. Sixty-seven children with ADHD and 62 typically developing children (TDC) performed a battery of tasks grouped in domains: simple and choice reaction time; static and dynamic detection threshold (probing feedforward inhibition); amplitude discrimination without adaptation and with dual and single-site adaptation (probing lateral inhibition and adaptation); sequential and simultaneous frequency discrimination (previously linked to GABA); and temporal order judgment with and without a synchronous carrier stimulus. Children with ADHD could discriminate different amplitudes without adaptation, suggesting lateral inhibition is intact, but were negatively affected in all adaptation conditions, whereas TDC were only affected during single-site adaptation. Children with ADHD also showed normal frequency discrimination. Children with ADHD showed slower reaction times and higher detection threshold, likely driven by IQ and inattention, because reaction time and detection thresholds correlated with IQ and subtle motor signs. Children with ADHD showed a pattern of altered tactile processing on specific tasks, suggesting that higher cognitive function and cortical mechanisms related to adaptation are affected in ADHD, but no clear conclusion can be drawn toward impaired inhibition.NEW & NOTEWORTHY This manuscript presents the first tactile psychophysical study testing different aspects of tactile processing in attention-deficit hyperactivity disorder (ADHD), using large cohort sizes of 67 children with ADHD and 65 Typically Developing Children. This study demonstrates impaired tactile processing in children with ADHD, on some, but not all tasks (showing this is not just due to attention), related to impaired cortical mechanisms. Furthermore, both IQ and soft motor skill abnormalities (common in ADHD) are correlated with tactile abnormalities.
Collapse
Affiliation(s)
- Nicolaas A J Puts
- Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, Maryland; .,F.M. Kirby Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, Maryland
| | - Ashley D Harris
- Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, Maryland.,F.M. Kirby Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, Maryland.,Department of Radiology, University of Calgary, Calgary, Alberta, Canada.,The Child and Adolescent Imaging Research Program, Alberta Children's Hospital Research Institute, University of Calgary, Alberta Children's Hospital, Calgary, Alberta, Canada.,Hotchkiss Brain Institute and Alberta Children's Hospital Research Institute, Calgary, Alberta, Canada
| | - Mark Mikkelsen
- Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, Maryland.,F.M. Kirby Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, Maryland
| | - Mark Tommerdahl
- Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Richard A E Edden
- Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, Maryland.,F.M. Kirby Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, Maryland
| | - Stewart H Mostofsky
- Center for Neurocognitive and Imaging Research, Kennedy Krieger Institute, Baltimore, Maryland.,Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, Maryland; and.,Department of Behavioral Science and Psychiatry, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|