1
|
Fujioka Y, Shiura H, Ishii M, Ono R, Endo T, Kiyonari H, Hirate Y, Ito H, Kanai-Azuma M, Kohda T, Kaneko-Ishino T, Ishino F. Targeting of retrovirus-derived Rtl8a/ 8b causes late-onset obesity, reduced social response and increased apathy-like behaviour. Open Biol 2025; 15:240279. [PMID: 39875098 PMCID: PMC11774587 DOI: 10.1098/rsob.240279] [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: 09/26/2024] [Revised: 11/18/2024] [Accepted: 11/28/2024] [Indexed: 01/30/2025] Open
Abstract
Retrotransposon Gag-like (RTL) 8A, 8B and 8C are eutherian-specific genes derived from a certain retrovirus. They cluster as a triplet of genes on the X chromosome, but their function remains unknown. Here, we demonstrate that Rtl8a and Rtl8b play important roles in the brain: their double knockout (DKO) mice not only exhibit reduced social responses and increased apathy-like behaviour, but also become obese from young adulthood, similar to patients with late Prader-Willi syndrome (PWS), a neurodevelopmental genomic imprinting disorder. Mouse RTL8A/8B proteins are expressed in the prefrontal cortex and hypothalamus and localize to both the nucleus and cytoplasm of neurons, presumably due to the N-terminal nuclear localization signal-like sequence at the N-terminus. An RNAseq study in the cerebral cortex revealed reduced expression of several GABA type A receptor subunit genes in DKO, in particular Gabrb2, which encodes its β2 subunit. We confirmed the reduction of GABRB2 protein in the DKO cerebral cortex by western blotting. As GABRB2 has been implicated in the aetiology of several neurodevelopmental and neuropsychiatric disorders, it is likely that the reduction of GABRB2 is one of the major causes of the neuropsychiatric defects in the DKO mice.
Collapse
Affiliation(s)
- Yoshifumi Fujioka
- Department of Epigenetics, Medical Research Institute (MRI), Tokyo Medical and Dental University (TMDU), Tokyo113-8510, Japan
- Center for Experimental Animals, TMDU, Tokyo113-8510, Japan
| | - Hirosuke Shiura
- Department of Epigenetics, Medical Research Institute (MRI), Tokyo Medical and Dental University (TMDU), Tokyo113-8510, Japan
- Faculty of Life and Environmental Sciences, University of Yamanashi, Kohfu,Yamanashi 400-8510, Japan
| | - Masayuki Ishii
- Department of Epigenetics, Medical Research Institute (MRI), Tokyo Medical and Dental University (TMDU), Tokyo113-8510, Japan
| | - Ryuichi Ono
- Department of Epigenetics, Medical Research Institute (MRI), Tokyo Medical and Dental University (TMDU), Tokyo113-8510, Japan
- Division of Cellular and Molecular Toxicology, Center for Biological Safety and Research, National Institute of Health Sciences (NIHS), Kawasaki, Kanagawa210-9501, Japan
| | - Tsutomu Endo
- Center for Experimental Animals, TMDU, Tokyo113-8510, Japan
| | - Hiroshi Kiyonari
- Laboratory for Animal Resources and Genetic Engineering, RIKEN Center for Biosystems Dynamics Research, Kobe, Hyogo650-0047, Japan
| | | | - Hikaru Ito
- Center for Experimental Animals, TMDU, Tokyo113-8510, Japan
- Research Facility Center for Science and Technology, Kagawa University, Takamatsu,Kagawa 761-0793, Japan
| | | | - Takashi Kohda
- Faculty of Life and Environmental Sciences, University of Yamanashi, Kohfu,Yamanashi 400-8510, Japan
| | - Tomoko Kaneko-Ishino
- Faculty of Nursing, Tokai University School of Medicine, Isehara, Kanagawa259-1193, Japan
| | - Fumitoshi Ishino
- Department of Epigenetics, Medical Research Institute (MRI), Tokyo Medical and Dental University (TMDU), Tokyo113-8510, Japan
- Center for Experimental Animals, TMDU, Tokyo113-8510, Japan
| |
Collapse
|
2
|
Li X, Guo S, Sun Y, Ding J, Chen C, Wu Y, Li P, Sun T, Wang X. GABRG2 mutations in genetic epilepsy with febrile seizures plus: structure, roles, and molecular genetics. J Transl Med 2024; 22:767. [PMID: 39143639 PMCID: PMC11323400 DOI: 10.1186/s12967-024-05387-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 06/10/2024] [Indexed: 08/16/2024] Open
Abstract
Genetic epilepsy with febrile seizures plus (GEFS+) is a genetic epilepsy syndrome characterized by a marked hereditary tendency inherited as an autosomal dominant trait. Patients with GEFS+ may develop typical febrile seizures (FS), while generalized tonic-clonic seizures (GTCSs) with fever commonly occur between 3 months and 6 years of age, which is generally followed by febrile seizure plus (FS+), with or without absence seizures, focal seizures, or GTCSs. GEFS+ exhibits significant genetic heterogeneity, with polymerase chain reaction, exon sequencing, and single nucleotide polymorphism analyses all showing that the occurrence of GEFS+ is mainly related to mutations in the gamma-aminobutyric acid type A receptor gamma 2 subunit (GABRG2) gene. The most common mutations in GABRG2 are separated in large autosomal dominant families, but their pathogenesis remains unclear. The predominant types of GABRG2 mutations include missense (c.983A → T, c.245G → A, p.Met199Val), nonsense (R136*, Q390*, W429*), frameshift (c.1329delC, p.Val462fs*33, p.Pro59fs*12), point (P83S), and splice site (IVS6+2T → G) mutations. All of these mutations types can reduce the function of ion channels on the cell membrane; however, the degree and mechanism underlying these dysfunctions are different and could be linked to the main mechanism of epilepsy. The γ2 subunit plays a special role in receptor trafficking and is closely related to its structural specificity. This review focused on investigating the relationship between GEFS+ and GABRG2 mutation types in recent years, discussing novel aspects deemed to be great significance for clinically accurate diagnosis, anti-epileptic treatment strategies, and new drug development.
Collapse
Affiliation(s)
- Xinxiao Li
- Department of Neurosurgery, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, People's Republic of China.
| | - Shengnan Guo
- Department of Rehabilitative Medicine, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, People's Republic of China
| | - Yangyang Sun
- Ningxia Key Laboratory of Cerebrocranial Disease, The Incubation Base of National Key Laboratory, Ningxia Medical University, Yinchuan, Ningxia, 750001, People's Republic of China
| | - Jiangwei Ding
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, People's Republic of China
| | - Chao Chen
- Department of Neurosurgery, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, People's Republic of China
| | - Yuehui Wu
- Department of Neurosurgery, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, People's Republic of China
| | - Peidong Li
- Department of Neurosurgery, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, People's Republic of China
| | - Tao Sun
- Ningxia Key Laboratory of Cerebrocranial Disease, The Incubation Base of National Key Laboratory, Ningxia Medical University, Yinchuan, Ningxia, 750001, People's Republic of China.
| | - Xinjun Wang
- Department of Neurosurgery, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, People's Republic of China.
- Department of Neurosurgery, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, People's Republic of China.
| |
Collapse
|
3
|
Steele JW, Krishnan V, Finnell RH. Mechanisms of neurodevelopmental toxicity of topiramate. Crit Rev Toxicol 2024; 54:465-475. [PMID: 38995641 PMCID: PMC11296906 DOI: 10.1080/10408444.2024.2368552] [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: 02/27/2024] [Revised: 05/03/2024] [Accepted: 06/11/2024] [Indexed: 07/13/2024]
Abstract
Prescriptions for antiseizure medications (ASMs) have been rapidly growing over the last several decades due, in part, to an expanding list of clinical indications for which they are now prescribed. This trend has raised concern for potential adverse neurodevelopmental outcomes in ASM-exposed pregnancies. Recent large scale population studies have suggested that the use of topiramate (TOPAMAX, Janssen-Cilag), when prescribed for seizure control, migraines, and/or weight management, is associated with an increased risk for autism spectrum disorder (ASD), intellectual disability, and attention-deficit/hyperactivity disorder (ADHD) in exposed offspring. Here, we critically review epidemiologic evidence demonstrating the neurobehavioral teratogenicity of topiramate and speculate on the neuromolecular mechanisms by which prenatal exposure may perturb neurocognitive development. Specifically, we explore the potential role of topiramate's pharmacological interactions with ligand- and voltage-gated ion channels, especially GABAergic signaling, its effects on DNA methylation and histone acetylation, whether topiramate induces oxidative stress, and its association with fetal growth restriction as possible mechanisms contributing to neurodevelopmental toxicity. Resolving this biology will be necessary to reduce the risk of adverse pregnancy outcomes caused by topiramate or other ASMs.
Collapse
Affiliation(s)
- John W. Steele
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, TX, USA
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Vaishnav Krishnan
- Departments of Neurology, Neuroscience and Psychiatry, and Behavioral Sciences, Baylor College of Medicine, Houston, TX, USA
| | - Richard H. Finnell
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, TX, USA
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
- Departments of Molecular and Cellular Biology and Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
4
|
Smith SS, Benanni S, Jones Q, Kenney L, Evrard MR. Manipulation of α4βδ GABA A receptors alters synaptic pruning in layer 3 prelimbic prefrontal cortex and impairs temporal order recognition: Implications for schizophrenia and autism. Brain Res 2024; 1835:148929. [PMID: 38599510 DOI: 10.1016/j.brainres.2024.148929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 04/04/2024] [Accepted: 04/06/2024] [Indexed: 04/12/2024]
Abstract
Temporal order memory is impaired in autism spectrum disorder (ASD) and schizophrenia (SCZ). These disorders, more prevalent in males, result in abnormal dendritic spine pruning during adolescence in layer 3 (L3) medial prefrontal cortex (mPFC), yielding either too many (ASD) or too few (SCZ) spines. Here we tested whether altering spine density in neural circuits including the mPFC could be associated with impaired temporal order memory in male mice. We have shown that α4βδ GABAA receptors (GABARs) emerge at puberty on spines of L5 prelimbic mPFC (PL) where they trigger pruning. We show here that α4βδ receptors also increase at puberty in L3 PL (P < 0.0001) and used these receptors as a target to manipulate spine density here. Pubertal injection (14 d) of the GABA agonist gaboxadol, at a dose (3 mg/kg) selective for α4βδ, reduced L3 spine density by half (P < 0.0001), while α4 knock-out increased spine density ∼ 40 % (P < 0.0001), mimicking spine densities in SCZ and ASD, respectively. In both cases, performance on the mPFC-dependent temporal order recognition task was impaired, resulting in decreases in the discrimination ratio which assesses preference for the novel object: -0.39 ± 0.15, gaboxadol versus 0.52 ± 0.09, vehicle; P = 0.0002; -0.048 ± 0.10, α4 KO versus 0.49 ± 0.04, wild-type; P < 0.0001. In contrast, the number of approaches was unaltered, reflecting unchanged locomotion. These data suggest that altering α4βδ GABAR expression/activity alters spine density in L3 mPFC and impairs temporal order memory to mimic changes in ASD and SCZ. These findings may provide insight into these disorders.
Collapse
Affiliation(s)
- Sheryl S Smith
- Department of Physiology and Pharmacology, SUNY Downstate Medical Center, 450 Clarkson Ave., Brooklyn, NY 11203, USA.
| | - Safae Benanni
- Department of Physiology and Pharmacology, SUNY Downstate Medical Center, 450 Clarkson Ave., Brooklyn, NY 11203, USA.
| | - Quiana Jones
- Department of Physiology and Pharmacology, SUNY Downstate Medical Center, 450 Clarkson Ave., Brooklyn, NY 11203, USA.
| | - Lindsay Kenney
- Department of Physiology and Pharmacology, SUNY Downstate Medical Center, 450 Clarkson Ave., Brooklyn, NY 11203, USA.
| | - Matthew R Evrard
- Department of Physiology and Pharmacology, SUNY Downstate Medical Center, 450 Clarkson Ave., Brooklyn, NY 11203, USA.
| |
Collapse
|
5
|
Tekin M, Shen H, Smith SS. Sex differences in motor learning flexibility are accompanied by sex differences in mushroom spine pruning of the mouse primary motor cortex during adolescence. Front Neurosci 2024; 18:1420309. [PMID: 39040633 PMCID: PMC11262054 DOI: 10.3389/fnins.2024.1420309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 06/24/2024] [Indexed: 07/24/2024] Open
Abstract
Background Although males excel at motor tasks requiring strength, females exhibit greater motor learning flexibility. Cognitive flexibility is associated with low baseline mushroom spine densities achieved by pruning which can be triggered by α4βδ GABAA receptors (GABARs); defective synaptic pruning impairs this process. Methods We investigated sex differences in adolescent pruning of mushroom spine pruning of layer 5 pyramidal cells of primary motor cortex (L5M1), a site essential for motor learning, using microscopic evaluation of Golgi stained sections. We assessed α4GABAR expression using immunohistochemical and electrophysiological techniques (whole cell patch clamp responses to 100 nM gaboxadol, selective for α4βδ GABARs). We then compared performance of groups with different post-pubertal mushroom spine densities on motor learning (constant speed) and learning flexibility (accelerating speed following constant speed) rotarod tasks. Results Mushroom spines in proximal L5M1 of female mice decreased >60% from PND35 (puberty onset) to PND56 (Pubertal: 2.23 ± 0.21 spines/10 μm; post-pubertal: 0.81 ± 0.14 spines/10 μm, P < 0.001); male mushroom spine density was unchanged. This was due to greater α4βδ GABAR expression in the female (P < 0.0001) because α4 -/- mice did not exhibit mushroom spine pruning. Although motor learning was similar for all groups, only female wild-type mice (low mushroom spine density) learned the accelerating rotarod task after the constant speed task (P = 0.006), a measure of motor learning flexibility. Conclusions These results suggest that optimal motor learning flexibility of female mice is associated with low baseline levels of post-pubertal mushroom spine density in L5M1 compared to male and female α4 -/- mice.
Collapse
Affiliation(s)
- Michael Tekin
- Department of Physiology and Pharmacology, SUNY Downstate Medical Center, Brooklyn, NY, United States
- Graduate Program in Neural and Behavioral Science, SUNY Downstate Medical Center, Brooklyn, NY, United States
| | - Hui Shen
- School of Biomedical Engineering, Tianjin Medical University, Tianjin, China
| | - Sheryl S. Smith
- Department of Physiology and Pharmacology, SUNY Downstate Medical Center, Brooklyn, NY, United States
| |
Collapse
|
6
|
Abedini SS, Akhavantabasi S, Liang Y, Heng JIT, Alizadehsani R, Dehzangi I, Bauer DC, Alinejad-Rokny H. A critical review of the impact of candidate copy number variants on autism spectrum disorder. MUTATION RESEARCH. REVIEWS IN MUTATION RESEARCH 2024; 794:108509. [PMID: 38977176 DOI: 10.1016/j.mrrev.2024.108509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Revised: 04/14/2024] [Accepted: 07/02/2024] [Indexed: 07/10/2024]
Abstract
Autism spectrum disorder (ASD) is a complex neurodevelopmental disorder (NDD) influenced by genetic, epigenetic, and environmental factors. Recent advancements in genomic analysis have shed light on numerous genes associated with ASD, highlighting the significant role of both common and rare genetic mutations, as well as copy number variations (CNVs), single nucleotide polymorphisms (SNPs) and unique de novo variants. These genetic variations disrupt neurodevelopmental pathways, contributing to the disorder's complexity. Notably, CNVs are present in 10 %-20 % of individuals with autism, with 3 %-7 % detectable through cytogenetic methods. While the role of submicroscopic CNVs in ASD has been recently studied, their association with genomic loci and genes has not been thoroughly explored. In this review, we focus on 47 CNV regions linked to ASD, encompassing 1632 genes, including protein-coding genes and long non-coding RNAs (lncRNAs), of which 659 show significant brain expression. Using a list of ASD-associated genes from SFARI, we detect 17 regions harboring at least one known ASD-related protein-coding gene. Of the remaining 30 regions, we identify 24 regions containing at least one protein-coding gene with brain-enriched expression and a nervous system phenotype in mouse mutants, and one lncRNA with both brain-enriched expression and upregulation in iPSC to neuron differentiation. This review not only expands our understanding of the genetic diversity associated with ASD but also underscores the potential of lncRNAs in contributing to its etiology. Additionally, the discovered CNVs will be a valuable resource for future diagnostic, therapeutic, and research endeavors aimed at prioritizing genetic variations in ASD.
Collapse
Affiliation(s)
- Seyedeh Sedigheh Abedini
- UNSW BioMedical Machine Learning Lab (BML), The Graduate School of Biomedical Engineering, UNSW Sydney, Sydney, NSW 2052, Australia; School of Biotechnology & Biomolecular Sciences, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Shiva Akhavantabasi
- Department of Molecular Biology and Genetics, Yeni Yuzyil University, Istanbul, Turkey; Ghiaseddin Jamshid Kashani University, Andisheh University Town, Danesh Blvd, 3441356611, Abyek, Qazvin, Iran
| | - Yuheng Liang
- UNSW BioMedical Machine Learning Lab (BML), The Graduate School of Biomedical Engineering, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Julian Ik-Tsen Heng
- Curtin Health Innovation Research Institute, Curtin University, Bentley 6845, Australia
| | - Roohallah Alizadehsani
- Institute for Intelligent Systems Research and Innovation (IISRI), Deakin University, Victoria, Australia
| | - Iman Dehzangi
- Center for Computational and Integrative Biology, Rutgers University, Camden, NJ 08102, USA; Department of Computer Science, Rutgers University, Camden, NJ 08102, USA
| | - Denis C Bauer
- Transformational Bioinformatics, Commonwealth Scientific and Industrial Research Organisation (CSIRO), Sydney, Australia; Applied BioSciences, Faculty of Science and Engineering, Macquarie University, Macquarie Park, Australia
| | - Hamid Alinejad-Rokny
- UNSW BioMedical Machine Learning Lab (BML), The Graduate School of Biomedical Engineering, UNSW Sydney, Sydney, NSW 2052, Australia; Tyree Institute of Health Engineering (IHealthE), UNSW Sydney, Sydney, NSW 2052, Australia.
| |
Collapse
|
7
|
Issahaku AR, Wilhelm A, Schutte-Smith M, Erasmus E, Visser H. Elucidating the binding mechanisms of GABA and Muscimol as an avenue to discover novel GABA-mimetic small molecules. J Biomol Struct Dyn 2024:1-16. [PMID: 38520326 DOI: 10.1080/07391102.2024.2331088] [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: 01/04/2024] [Accepted: 03/10/2024] [Indexed: 03/25/2024]
Abstract
Gamma-aminobutyric acid (GABA) signaling is the principal inhibitory pathway in the central nervous system. It is critical in neuronal cell proliferation and fate determination. Any aberration in GABA inhibition results in psychiatric and neurological diseases. Thus, modulating GABAergic neurotransmission has become the basis of drug therapy for psychiatric and several neurological diseases. Though GABA and muscimol are classical inhibitors of GABA receptors, the search for novel inhibitors continues unabated. In this study, the binding mechanism of GABA and muscimol was elucidated and applied in the search for small molecule GABAergic inhibitors using comprehensive computational techniques. It was revealed that a high-affinity binding of GABA and muscimol was mediated by a water molecule involving α1Thr129 and then stabilized by strong interactions including salt bridges with β2Glu155 and α1Arg66 amidst hydrogen bonds, π-π stacking, and π -cation interactions with other residues. The binding of GABA and muscimol was also characterized by stability and deeper penetration into the hydrophobic core of the protein which resulted in conformational changes of the binding pocket and domain, by inducing correlated motions of the residues. Thermodynamics analysis showed GABA and muscimol exhibited total binding free energies of -19.85 ± 8.83 Kcal/mol and -26.55 ± 3.42 Kcal/mol, respectively. A pharmacophore model search, based on the energy contributions of implicating binding residues, resulted in the identification of ZINC68604167, ZINC19735138, ZINC04202466, ZINC00901626, and ZINC01532854 as potential GABA-mimetic compounds from metabolites and natural products libraries. This study has elucidated the binding mechanisms of GABA and muscimol and successfully applied in the identification of GABA-mimetic compounds.
Collapse
Affiliation(s)
| | - Anke Wilhelm
- Department of Chemistry, University of the Free State, Bloemfontein, South Africa
| | | | - Elizabeth Erasmus
- Department of Chemistry, University of the Free State, Bloemfontein, South Africa
| | - Hendrik Visser
- Department of Chemistry, University of the Free State, Bloemfontein, South Africa
| |
Collapse
|
8
|
Platonov M, Maximyuk O, Rayevsky A, Iegorova O, Hurmach V, Holota Y, Bulgakov E, Cherninskyi A, Karpov P, Ryabukhin S, Krishtal O, Volochnyuk D. Integrated workflow for the identification of new GABA A R positive allosteric modulators based on the in silico screening with further in vitro validation. Case study using Enamine's stock chemical space. Mol Inform 2024; 43:e202300156. [PMID: 37964718 DOI: 10.1002/minf.202300156] [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: 06/11/2023] [Revised: 11/10/2023] [Accepted: 11/14/2023] [Indexed: 11/16/2023]
Abstract
Numerous studies reported an association between GABAA R subunit genes and epilepsy, eating disorders, autism spectrum disorders, neurodevelopmental disorders, and bipolar disorders. This study was aimed to find some potential positive allosteric modulators and was performed by combining the in silico approach with further in vitro evaluation of its real activity. We started from the GABAA R-diazepam complexes and assembled a lipid embedded protein ensemble to refine it via molecular dynamics (MD) simulation. Then we focused on the interaction of α1β2γ2 with some Z-drugs (non-benzodiazepine compounds) using an Induced Fit Docking (IFD) into the relaxed binding site to generate a pharmacophore model. The pharmacophore model was validated with a reference set and applied to decrease the pre-filtered Enamine database before the main docking procedure. Finally, we succeeded in identifying a set of compounds, which met all features of the docking model. The aqueous solubility and stability of these compounds in mouse plasma were assessed. Then they were tested for the biological activity using the rat Purkinje neurons and CHO cells with heterologously expressed human α1β2γ2 GABAA receptors. Whole-cell patch clamp recordings were used to reveal the GABA induced currents. Our study represents a convenient and tunable model for the discovery of novel positive allosteric modulators of GABAA receptors. A High-throughput virtual screening of the largest available database of chemical compounds resulted in the selection of 23 compounds. Further electrophysiological tests allowed us to determine a set of 3 the most outstanding active compounds. Considering the structural features of leader compounds, the study can develop into the MedChem project soon.
Collapse
Affiliation(s)
- Maksym Platonov
- Institute of molecular biology and genetics, Natl. Academy of Sciences of Ukraine, Zabolotnogo Str., 150, Kyiv, 03143, Ukraine
- Enamine Ltd., 78 Chervonotkatska Str., 02660, Kyiv, Ukraine
| | - Oleksandr Maximyuk
- Bogomoletz Institute of Physiology, Natl. Academy of Sciences of Ukraine, 4 Bogomoletz Str., 01024, Kyiv, Ukraine
| | - Alexey Rayevsky
- Institute of molecular biology and genetics, Natl. Academy of Sciences of Ukraine, Zabolotnogo Str., 150, Kyiv, 03143, Ukraine
- Enamine Ltd., 78 Chervonotkatska Str., 02660, Kyiv, Ukraine
- Institute of Food Biotechnology and Genomics, Natl. Academy of Sciences of Ukraine, Osypovskoho Str., 2 A, Kyiv, 04123, Ukraine
| | - Olena Iegorova
- Bogomoletz Institute of Physiology, Natl. Academy of Sciences of Ukraine, 4 Bogomoletz Str., 01024, Kyiv, Ukraine
| | - Vasyl Hurmach
- Institute of molecular biology and genetics, Natl. Academy of Sciences of Ukraine, Zabolotnogo Str., 150, Kyiv, 03143, Ukraine
- Enamine Ltd., 78 Chervonotkatska Str., 02660, Kyiv, Ukraine
| | - Yuliia Holota
- Enamine Ltd., 78 Chervonotkatska Str., 02660, Kyiv, Ukraine
| | - Elijah Bulgakov
- Enamine Ltd., 78 Chervonotkatska Str., 02660, Kyiv, Ukraine
- Institute of Food Biotechnology and Genomics, Natl. Academy of Sciences of Ukraine, Osypovskoho Str., 2 A, Kyiv, 04123, Ukraine
| | - Andrii Cherninskyi
- Bogomoletz Institute of Physiology, Natl. Academy of Sciences of Ukraine, 4 Bogomoletz Str., 01024, Kyiv, Ukraine
| | - Pavel Karpov
- Institute of Food Biotechnology and Genomics, Natl. Academy of Sciences of Ukraine, Osypovskoho Str., 2 A, Kyiv, 04123, Ukraine
| | - Sergey Ryabukhin
- Enamine Ltd., 78 Chervonotkatska Str., 02660, Kyiv, Ukraine
- Institute of High Technologies, Taras Shevchenko National University of Kyiv., Glushkova Ave, 03022, Kyiv, Ukraine
- Institute of organic chemistry NAS of Ukraine, 5 Murmanska Str., 02660, Kyiv, Ukraine
| | - Oleg Krishtal
- Bogomoletz Institute of Physiology, Natl. Academy of Sciences of Ukraine, 4 Bogomoletz Str., 01024, Kyiv, Ukraine
| | - Dmitriy Volochnyuk
- Enamine Ltd., 78 Chervonotkatska Str., 02660, Kyiv, Ukraine
- Institute of High Technologies, Taras Shevchenko National University of Kyiv., Glushkova Ave, 03022, Kyiv, Ukraine
- Institute of organic chemistry NAS of Ukraine, 5 Murmanska Str., 02660, Kyiv, Ukraine
| |
Collapse
|
9
|
Cano ACSS, Santos D, Beltrão-Braga PCB. The Interplay of Astrocytes and Neurons in Autism Spectrum Disorder. ADVANCES IN NEUROBIOLOGY 2024; 39:269-284. [PMID: 39190079 DOI: 10.1007/978-3-031-64839-7_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/28/2024]
Abstract
Autism spectrum disorder (ASD) comprises a complex neurodevelopmental condition characterized by an impairment in social interaction, involving communication deficits and specific patterns of behaviors, like repetitive behaviors. ASD is clinically diagnosed and usually takes time, typically occurring not before four years of age. Genetic mutations affecting synaptic transmission, such as neuroligin and neurexin, are associated with ASD and contribute to behavioral and cognitive deficits. Recent research highlights the role of astrocytes, the brain's most abundant glial cells, in ASD pathology. Aberrant Ca2+ signaling in astrocytes is linked to behavioral deficits and neuroinflammation. Notably, the cytokine IL-6 overexpression by astrocytes impacts synaptogenesis. Altered neurotransmitter levels, disruptions in the blood-brain barrier, and cytokine dysregulation further contribute to ASD complexity. Understanding these astrocyte-related mechanisms holds promise for identifying ASD subtypes and developing targeted therapies.
Collapse
Affiliation(s)
- Amanda C S S Cano
- Laboratory of Disease Modeling, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Debora Santos
- Laboratory of Disease Modeling, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Patricia C B Beltrão-Braga
- Laboratory of Disease Modeling, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil.
- Institut Pasteur de São Paulo, São Paulo, Brazil.
| |
Collapse
|
10
|
Parrella NF, Hill AT, Dipnall LM, Loke YJ, Enticott PG, Ford TC. Inhibitory dysfunction and social processing difficulties in autism: A comprehensive narrative review. J Psychiatr Res 2024; 169:113-125. [PMID: 38016393 DOI: 10.1016/j.jpsychires.2023.11.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 09/04/2023] [Accepted: 11/15/2023] [Indexed: 11/30/2023]
Abstract
The primary inhibitory neurotransmitter γ-aminobutyric acid (GABA) has a prominent role in regulating neural development and function, with disruption to GABAergic signalling linked to behavioural phenotypes associated with neurodevelopmental disorders, particularly autism. Such neurochemical disruption, likely resulting from diverse genetic and molecular mechanisms, particularly during early development, can subsequently affect the cellular balance of excitation and inhibition in neuronal circuits, which may account for the social processing difficulties observed in autism and related conditions. This comprehensive narrative review integrates diverse streams of research from several disciplines, including molecular neurobiology, genetics, epigenetics, and systems neuroscience. In so doing it aims to elucidate the relevance of inhibitory dysfunction to autism, with specific focus on social processing difficulties that represent a core feature of this disorder. Many of the social processing difficulties experienced in autism have been linked to higher levels of the excitatory neurotransmitter glutamate and/or lower levels of inhibitory GABA. While current therapeutic options for social difficulties in autism are largely limited to behavioural interventions, this review highlights the psychopharmacological studies that explore the utility of GABA modulation in alleviating such difficulties.
Collapse
Affiliation(s)
| | - Aron T Hill
- Cognitive Neuroscience Unit, School of Psychology, Deakin University, Geelong, Australia; Department of Psychiatry, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Lillian M Dipnall
- Cognitive Neuroscience Unit, School of Psychology, Deakin University, Geelong, Australia; Early Life Epigenetics Group, Deakin University, Geelong, Australia
| | - Yuk Jing Loke
- Epigenetics Group, Murdoch Children's Research Institute, Melbourne, Victoria, Australia; Department of Paediatrics, The University of Melbourne, Melbourne, Victoria, Australia
| | - Peter G Enticott
- Cognitive Neuroscience Unit, School of Psychology, Deakin University, Geelong, Australia
| | - Talitha C Ford
- Cognitive Neuroscience Unit, School of Psychology, Deakin University, Geelong, Australia; Centre for Human Psychopharmacology, Faculty of Health, Arts and Design, Swinburne University of Technology, Melbourne, Victoria, Australia
| |
Collapse
|
11
|
Gillespie B, Panthi S, Sundram S, Hill RA. The impact of maternal immune activation on GABAergic interneuron development: A systematic review of rodent studies and their translational implications. Neurosci Biobehav Rev 2024; 156:105488. [PMID: 38042358 DOI: 10.1016/j.neubiorev.2023.105488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 11/09/2023] [Accepted: 11/27/2023] [Indexed: 12/04/2023]
Abstract
Mothers exposed to infections during pregnancy disproportionally birth children who develop autism and schizophrenia, disorders associated with altered GABAergic function. The maternal immune activation (MIA) model recapitulates this risk factor, with many studies also reporting disruptions to GABAergic interneuron expression, protein, cellular density and function. However, it is unclear if there are species, sex, age, region, or GABAergic subtype specific vulnerabilities to MIA. Furthermore, to fully comprehend the impact of MIA on the GABAergic system a synthesised account of molecular, cellular, electrophysiological and behavioural findings was required. To this end we conducted a systematic review of GABAergic interneuron changes in the MIA model, focusing on the prefrontal cortex and hippocampus. We reviewed 102 articles that revealed robust changes in a number of GABAergic markers that present as gestationally-specific, region-specific and sometimes sex-specific. Disruptions to GABAergic markers coincided with distinct behavioural phenotypes, including memory, sensorimotor gating, anxiety, and sociability. Findings suggest the MIA model is a valid tool for testing novel therapeutics designed to recover GABAergic function and associated behaviour.
Collapse
Affiliation(s)
- Brendan Gillespie
- Department of Psychiatry, School of Clinical Sciences, Monash University, Clayton, VIC 3168, Australia
| | - Sandesh Panthi
- Department of Psychiatry, School of Clinical Sciences, Monash University, Clayton, VIC 3168, Australia
| | - Suresh Sundram
- Department of Psychiatry, School of Clinical Sciences, Monash University, Clayton, VIC 3168, Australia
| | - Rachel A Hill
- Department of Psychiatry, School of Clinical Sciences, Monash University, Clayton, VIC 3168, Australia.
| |
Collapse
|
12
|
Miyahara K, Tatehana M, Kikkawa T, Osumi N. Investigating the impact of paternal aging on murine sperm miRNA profiles and their potential link to autism spectrum disorder. Sci Rep 2023; 13:20608. [PMID: 38062235 PMCID: PMC10703820 DOI: 10.1038/s41598-023-47878-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 11/17/2023] [Indexed: 12/18/2023] Open
Abstract
Paternal aging has consistently been linked to an increased risk of neurodevelopmental disorders, including autism spectrum disorder (ASD), in offspring. Recent evidence has highlighted the involvement of epigenetic factors. In this study, we aimed to investigate age-related alterations in microRNA (miRNA) profiles of mouse sperm and analyze target genes regulated by differentially expressed miRNAs (DEmiRNAs). Microarray analyses were conducted on sperm samples from mice at different ages: 3 months (3 M), over 12 M, and beyond 20 M. We identified 26 miRNAs with differential expression between the 3 and 20 M mice, 34 miRNAs between the 12 and 20 M mice, and 2 miRNAs between the 3 and 12 M mice. The target genes regulated by these miRNAs were significantly associated with apoptosis/ferroptosis pathways and the nervous system. We revealed alterations in sperm miRNA profiles due to aging and suggest that the target genes regulated by these DEmiRNAs are associated with apoptosis and the nervous system, implying a potential link between paternal aging and an increased risk of neurodevelopmental disorders such as ASD. The observed age-related changes in sperm miRNA profiles have the potential to impact sperm quality and subsequently affect offspring development.
Collapse
Affiliation(s)
- Kazusa Miyahara
- Department of Developmental Neuroscience, Center for Advanced Research and Translational Medicine (ART), Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Misako Tatehana
- Department of Developmental Neuroscience, Center for Advanced Research and Translational Medicine (ART), Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Takako Kikkawa
- Department of Developmental Neuroscience, Center for Advanced Research and Translational Medicine (ART), Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Noriko Osumi
- Department of Developmental Neuroscience, Center for Advanced Research and Translational Medicine (ART), Tohoku University Graduate School of Medicine, Sendai, Japan.
| |
Collapse
|
13
|
Meng X, Yao D, Imaizumi K, Chen X, Kelley KW, Reis N, Thete MV, Arjun McKinney A, Kulkarni S, Panagiotakos G, Bassik MC, Pașca SP. Assembloid CRISPR screens reveal impact of disease genes in human neurodevelopment. Nature 2023; 622:359-366. [PMID: 37758944 PMCID: PMC10567561 DOI: 10.1038/s41586-023-06564-w] [Citation(s) in RCA: 58] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 08/22/2023] [Indexed: 09/29/2023]
Abstract
The assembly of cortical circuits involves the generation and migration of interneurons from the ventral to the dorsal forebrain1-3, which has been challenging to study at inaccessible stages of late gestation and early postnatal human development4. Autism spectrum disorder and other neurodevelopmental disorders (NDDs) have been associated with abnormal cortical interneuron development5, but which of these NDD genes affect interneuron generation and migration, and how they mediate these effects remains unknown. We previously developed a platform to study interneuron development and migration in subpallial organoids and forebrain assembloids6. Here we integrate assembloids with CRISPR screening to investigate the involvement of 425 NDD genes in human interneuron development. The first screen aimed at interneuron generation revealed 13 candidate genes, including CSDE1 and SMAD4. We subsequently conducted an interneuron migration screen in more than 1,000 forebrain assembloids that identified 33 candidate genes, including cytoskeleton-related genes and the endoplasmic reticulum-related gene LNPK. We discovered that, during interneuron migration, the endoplasmic reticulum is displaced along the leading neuronal branch before nuclear translocation. LNPK deletion interfered with this endoplasmic reticulum displacement and resulted in abnormal migration. These results highlight the power of this CRISPR-assembloid platform to systematically map NDD genes onto human development and reveal disease mechanisms.
Collapse
Affiliation(s)
- Xiangling Meng
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
- Stanford Brain Organogenesis Program, Wu Tsai Neurosciences Institute and Bio-X, Stanford, CA, USA
| | - David Yao
- Department of Genetics, Stanford University, Stanford, CA, USA
| | - Kent Imaizumi
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
- Stanford Brain Organogenesis Program, Wu Tsai Neurosciences Institute and Bio-X, Stanford, CA, USA
| | - Xiaoyu Chen
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
- Stanford Brain Organogenesis Program, Wu Tsai Neurosciences Institute and Bio-X, Stanford, CA, USA
| | - Kevin W Kelley
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
- Stanford Brain Organogenesis Program, Wu Tsai Neurosciences Institute and Bio-X, Stanford, CA, USA
| | - Noah Reis
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
- Stanford Brain Organogenesis Program, Wu Tsai Neurosciences Institute and Bio-X, Stanford, CA, USA
| | - Mayuri Vijay Thete
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
- Stanford Brain Organogenesis Program, Wu Tsai Neurosciences Institute and Bio-X, Stanford, CA, USA
| | - Arpana Arjun McKinney
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA, USA
- Departments of Psychiatry and Neuroscience, Black Family Stem Cell Institute, Seaver Autism Center for Research and Treatment, Alper Center for Neural Development and Regeneration, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Shravanti Kulkarni
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
- Stanford Brain Organogenesis Program, Wu Tsai Neurosciences Institute and Bio-X, Stanford, CA, USA
| | - Georgia Panagiotakos
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA, USA
- Department of Biochemistry and Biophysics, University of California, San Francisco, CA, USA
- Departments of Psychiatry and Neuroscience, Black Family Stem Cell Institute, Seaver Autism Center for Research and Treatment, Alper Center for Neural Development and Regeneration, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Sergiu P Pașca
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA.
- Stanford Brain Organogenesis Program, Wu Tsai Neurosciences Institute and Bio-X, Stanford, CA, USA.
| |
Collapse
|
14
|
Wang X, Zhao Z, Guo J, Mei D, Duan Y, Zhang Y, Gou L. GABA B1 receptor knockdown in prefrontal cortex induces behavioral aberrations associated with autism spectrum disorder in mice. Brain Res Bull 2023; 202:110755. [PMID: 37678443 DOI: 10.1016/j.brainresbull.2023.110755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 08/31/2023] [Accepted: 09/05/2023] [Indexed: 09/09/2023]
Abstract
Autism spectrum disorder (ASD) is a set of heterogeneous neurodevelopmental disorders, characterized by social interaction deficit, stereotyped or repetitive behaviors. Apart from these core symptoms, a great number of individuals with ASD exhibit higher levels of anxiety and memory deficits. Previous studies demonstrate pronounced decrease of γ-aminobutyric acid B1 receptor (GABAB1R) protein level of frontal lobe in both ASD patients and animal models. The aim of the present study was to determine the role of GABAB1R in ASD-related behavioral aberrations. Herein, the protein and mRNA levels of GABAB1R in the prefrontal cortex (PFC) of sodium valproic acid (VPA)-induced mouse ASD model were determined by Western blot and qRT-PCR analysis, respectively. Moreover, the behavioral abnormalities in naive mice with GABAB1R knockdown mediated by recombinant adeno-associated virus (rAAV) were assessed in a comprehensive test battery consisted of social interaction, marble burying, self-grooming, open-field, Y-maze and novel object recognition tests. Furthermore, the action potential changes induced by GABAB1R deficiency were examined in neurons within the PFC of mouse. The results show that the mRNA and protein levels of GABAB1R in the PFC of prenatal VPA-induced mouse ASD model were decreased. Concomitantly, naive mice with GABAB1R knockdown exhibited ASD-like behaviors, such as impaired social interaction and communication, elevated stereotypes, anxiety and memory deficits. Patch-clamp recordings also revealed that GABAB1R knockdown provoked enhanced neuronal excitability by increasing action potential discharge frequencies. Overall, these findings support a notion that GABAB1R deficiency might contribute to ASD-like phenotypes, with the pathogenesis most likely resulting from enhanced neuronal excitability. SUBHEADINGS: GABAB1 Knockdown Induces Behavioral Aberrations with ASD.
Collapse
Affiliation(s)
- Xiaona Wang
- Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Henan Key Laboratory of Children's Genetics and Metabolic Diseases, Henan Children's Neurodevelopment Engineering Research Center, Zhengzhou, China.
| | - Zhengqin Zhao
- Department of Nuclear Medicine, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Jisheng Guo
- School of Basic Medical Sciences, Yantai Campus of Binzhou Medical University, Yantai City, Shandong, China
| | - Daoqi Mei
- Department of Neurology, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| | - Yongtao Duan
- Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Henan Key Laboratory of Children's Genetics and Metabolic Diseases, Henan Children's Neurodevelopment Engineering Research Center, Zhengzhou, China
| | - Yaodong Zhang
- Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Henan Key Laboratory of Children's Genetics and Metabolic Diseases, Henan Children's Neurodevelopment Engineering Research Center, Zhengzhou, China.
| | - Lingshan Gou
- Peninsula Cancer Research Center, Binzhou Medical University, Yantai, Shandong, China.
| |
Collapse
|
15
|
Pedraza N, Monserrat MV, Ferrezuelo F, Torres-Rosell J, Colomina N, Miguez-Cabello F, Párraga JP, Soto D, López-Merino E, García-Vilela C, Esteban JA, Egea J, Garí E. Cyclin D1-Cdk4 regulates neuronal activity through phosphorylation of GABAA receptors. Cell Mol Life Sci 2023; 80:280. [PMID: 37684532 PMCID: PMC10491536 DOI: 10.1007/s00018-023-04920-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 08/10/2023] [Accepted: 08/12/2023] [Indexed: 09/10/2023]
Abstract
Nuclear Cyclin D1 (Ccnd1) is a main regulator of cell cycle progression and cell proliferation. Interestingly, Ccnd1 moves to the cytoplasm at the onset of differentiation in neuronal precursors. However, cytoplasmic functions and targets of Ccnd1 in post-mitotic neurons are unknown. Here we identify the α4 subunit of gamma-aminobutyric acid (GABA) type A receptors (GABAARs) as an interactor and target of Ccnd1-Cdk4. Ccnd1 binds to an intracellular loop in α4 and, together with Cdk4, phosphorylates the α4 subunit at threonine 423 and serine 431. These modifications upregulate α4 surface levels, increasing the response of α4-containing GABAARs, measured in whole-cell patch-clamp recordings. In agreement with this role of Ccnd1-Cdk4 in neuronal signalling, inhibition of Cdk4 or expression of the non-phosphorylatable α4 decreases synaptic and extra-synaptic currents in the hippocampus of newborn rats. Moreover, according to α4 functions in synaptic pruning, CCND1 knockout mice display an altered pattern of dendritic spines that is rescued by the phosphomimetic α4. Overall, our findings molecularly link Ccnd1-Cdk4 to GABAARs activity in the central nervous system and highlight a novel role for this G1 cyclin in neuronal signalling.
Collapse
Affiliation(s)
- Neus Pedraza
- Cell Cycle, Department of Basic Medical Sciences, Institut de Recerca Biomèdica de Lleida (IRBLLEIDA), University of Lleida, Lleida, Spain.
| | - Ma Ventura Monserrat
- Cell Cycle, Department of Basic Medical Sciences, Institut de Recerca Biomèdica de Lleida (IRBLLEIDA), University of Lleida, Lleida, Spain
| | - Francisco Ferrezuelo
- Cell Cycle, Department of Basic Medical Sciences, Institut de Recerca Biomèdica de Lleida (IRBLLEIDA), University of Lleida, Lleida, Spain
| | - Jordi Torres-Rosell
- Cell Cycle, Department of Basic Medical Sciences, Institut de Recerca Biomèdica de Lleida (IRBLLEIDA), University of Lleida, Lleida, Spain
| | - Neus Colomina
- Cell Cycle, Department of Basic Medical Sciences, Institut de Recerca Biomèdica de Lleida (IRBLLEIDA), University of Lleida, Lleida, Spain
| | - Federico Miguez-Cabello
- Laboratori de Neurofisiologia, Departament de Biomedicina, Facultat de Medicina i Ciències de la Salut, Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic, Universitat de Barcelona, Barcelona, Spain
| | - Javier Picañol Párraga
- Laboratori de Neurofisiologia, Departament de Biomedicina, Facultat de Medicina i Ciències de la Salut, Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic, Universitat de Barcelona, Barcelona, Spain
| | - David Soto
- Laboratori de Neurofisiologia, Departament de Biomedicina, Facultat de Medicina i Ciències de la Salut, Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic, Universitat de Barcelona, Barcelona, Spain
| | - Esperanza López-Merino
- Department of Molecular Neurobiology, Centro de Biología Molecular 'Severo Ochoa', Consejo Superior de Investigaciones Científicas (CSIC)/Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - Celia García-Vilela
- Department of Molecular Neurobiology, Centro de Biología Molecular 'Severo Ochoa', Consejo Superior de Investigaciones Científicas (CSIC)/Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - José A Esteban
- Department of Molecular Neurobiology, Centro de Biología Molecular 'Severo Ochoa', Consejo Superior de Investigaciones Científicas (CSIC)/Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - Joaquim Egea
- Molecular and Developmental Neurobiology, Dept. Ciències Mèdiques Bàsiques, Facultat de Medicina, Universitat de Lleida/IRBLLEIDA, Rovira Roure 80, 25198, Lleida, Spain
| | - Eloi Garí
- Cell Cycle, Department of Basic Medical Sciences, Institut de Recerca Biomèdica de Lleida (IRBLLEIDA), University of Lleida, Lleida, Spain.
| |
Collapse
|
16
|
Wang J, Gao Y, Xiao L, Lin Y, Huang L, Chen J, Liang G, Li W, Yi W, Lao J, Zhang B, Gao TM, Zhong M, Yang X. Increased NMDARs in neurons and glutamine synthetase in astrocytes underlying autistic-like behaviors of Gabrb1-/- mice. iScience 2023; 26:107476. [PMID: 37599823 PMCID: PMC10433130 DOI: 10.1016/j.isci.2023.107476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 06/16/2023] [Accepted: 07/21/2023] [Indexed: 08/22/2023] Open
Abstract
Mutations of the GABA-A receptor subunit β1 (GABRB1) gene are found in autism patients. However, it remains unclear how mutations in Gabrb1 may lead to autism. We generated Gabrb1-/- mouse model, which showed autistic-like behaviors. We carried out RNA-seq on the hippocampus and found glutamatergic pathway may be involved. We further carried out single-cell RNA sequencing on the whole brain followed by qRT-PCR, immunofluorescence, electrophysiology, and metabolite detection on specific cell types. We identified the up-regulated Glul/Slc38a3 in astrocytes, Grin1/Grin2b in neurons, glutamate, and the ratio of Glu/GABA in the hippocampus. Consistent with these results, increased NMDAR-currents and reduced GABAAR-currents in the CA1 neurons were detected in Gabrb1-/- mice. NMDAR antagonist memantine or Glul inhibitor methionine sulfoximine could rescue the abnormal behaviors in Gabrb1-/- mice. Our data reveal that upregulation of the glutamatergic synapse pathway, including NMDARs at neuronal synapses and glutamine exported by astrocytes, may lead to autistic-like behaviors.
Collapse
Affiliation(s)
- Jing Wang
- Center for Genetics and Developmental Systems Biology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
- Department of Obstetrics & Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
- State Key Laboratory of Organ Failure Research, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Yue Gao
- Center for Genetics and Developmental Systems Biology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
- Department of Obstetrics & Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
- State Key Laboratory of Organ Failure Research, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
- Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
- Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
- Guangdong Key Laboratory of Psychiatric Disorders, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Liuyan Xiao
- Center for Genetics and Developmental Systems Biology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
- Department of Obstetrics & Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
- State Key Laboratory of Organ Failure Research, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
- Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
- Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
- Guangdong Key Laboratory of Psychiatric Disorders, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Yanmei Lin
- Center for Genetics and Developmental Systems Biology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
- Department of Obstetrics & Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
- State Key Laboratory of Organ Failure Research, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
- Department of Psychiatry, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Lang Huang
- Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
- Guangdong Key Laboratory of Psychiatric Disorders, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Jinfa Chen
- Center for Genetics and Developmental Systems Biology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
- Department of Obstetrics & Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
- State Key Laboratory of Organ Failure Research, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
- Department of Psychiatry, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
- Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
- Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Guanmei Liang
- Center for Genetics and Developmental Systems Biology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
- Department of Obstetrics & Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
- State Key Laboratory of Organ Failure Research, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
- Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Weiming Li
- Department of Psychiatry, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Wenjuan Yi
- Center for Genetics and Developmental Systems Biology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
- Department of Obstetrics & Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
- State Key Laboratory of Organ Failure Research, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Jianpei Lao
- Center for Genetics and Developmental Systems Biology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
- Department of Obstetrics & Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
- State Key Laboratory of Organ Failure Research, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
- Department of Psychiatry, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
- Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
- Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Bin Zhang
- Department of Psychiatry, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Tian-Ming Gao
- Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
- Guangdong Key Laboratory of Psychiatric Disorders, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Mei Zhong
- Department of Obstetrics & Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Xinping Yang
- Center for Genetics and Developmental Systems Biology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
- Department of Obstetrics & Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
- State Key Laboratory of Organ Failure Research, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
- Department of Psychiatry, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
- Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
- Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
17
|
Vaz R, Edwards S, Dueñas-Rey A, Hofmeister W, Lindstrand A. Loss of ctnnd2b affects neuronal differentiation and behavior in zebrafish. Front Neurosci 2023; 17:1205653. [PMID: 37465584 PMCID: PMC10351287 DOI: 10.3389/fnins.2023.1205653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 06/15/2023] [Indexed: 07/20/2023] Open
Abstract
Delta-catenin (CTNND2) is an adhesive junction associated protein belonging to the family of p120 catenins. The human gene is located on the short arm of chromosome 5, the region deleted in Cri-du-chat syndrome (OMIM #123450). Heterozygous loss of CTNND2 has been linked to a wide spectrum of neurodevelopmental disorders such as autism, schizophrenia, and intellectual disability. Here we studied how heterozygous loss of ctnnd2b affects zebrafish embryonic development, and larvae and adult behavior. First, we observed a disorganization of neuronal subtypes in the developing forebrain, namely the presence of ectopic isl1-expressing cells and a local reduction of GABA-positive neurons in the optic recess region. Next, using time-lapse analysis, we found that the disorganized distribution of is1l-expressing forebrain neurons resulted from an increased specification of Isl1:GFP neurons. Finally, we studied the swimming patterns of both larval and adult heterozygous zebrafish and observed an increased activity compared to wildtype animals. Overall, this data suggests a role for ctnnd2b in the differentiation cascade of neuronal subtypes in specific regions of the vertebrate brain, with repercussions in the animal's behavior.
Collapse
Affiliation(s)
- Raquel Vaz
- Department of Molecular Medicine and Surgery and Centre of Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Steven Edwards
- Department of Applied Physics and Science for Life Laboratory, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Alfredo Dueñas-Rey
- Department of Molecular Medicine and Surgery and Centre of Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Wolfgang Hofmeister
- Department of Molecular Medicine and Surgery and Centre of Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Anna Lindstrand
- Department of Molecular Medicine and Surgery and Centre of Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
- Department of Clinical Genetics, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
18
|
Günay Ç, Aykol D, Özsoy Ö, Sönmezler E, Hanci YS, Kara B, Akkoyunlu Sünnetçi D, Cine N, Deniz A, Özer T, Ölçülü CB, Yilmaz Ö, Kanmaz S, Yilmaz S, Tekgül H, Yildiz N, Acar Arslan E, Cansu A, Olgaç Dündar N, Kusgoz F, Didinmez E, Gençpinar P, Aksu Uzunhan T, Ertürk B, Gezdirici A, Ayaz A, Ölmez A, Ayanoğlu M, Tosun A, Topçu Y, Kiliç B, Aydin K, Çağlar E, Ersoy Kosvali Ö, Okuyaz Ç, Besen Ş, Tekin Orgun L, Erol İ, Yüksel D, Sezer A, Atasoy E, Toprak Ü, Güngör S, Ozgor B, Karadağ M, Dilber C, Şahinoğlu B, Uyur Yalçin E, Eldes Hacifazlioglu N, Yaramiş A, Edem P, Gezici Tekin H, Yilmaz Ü, Ünalp A, Turay S, Biçer D, Gül Mert G, Dokurel Çetin İ, Kirik S, Öztürk G, Karal Y, Sanri A, Aksoy A, Polat M, Özgün N, Soydemir D, Sarikaya Uzan G, Ülker Üstebay D, Gök A, Yeşilmen MC, Yiş U, Karakülah G, Bursali A, Oktay Y, Hiz Kurul S. Shared Biological Pathways and Processes in Patients with Intellectual Disability: A Multicenter Study. Neuropediatrics 2023. [PMID: 36787800 DOI: 10.1055/a-2034-8528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
Abstract
BACKGROUND Although the underlying genetic causes of intellectual disability (ID) continue to be rapidly identified, the biological pathways and processes that could be targets for a potential molecular therapy are not yet known. This study aimed to identify ID-related shared pathways and processes utilizing enrichment analyses. METHOD In this multicenter study, causative genes of patients with ID were used as input for Disease Ontology (DO), Gene Ontology (GO), and Kyoto Encyclopedia of Genes and Genomes enrichment analysis. RESULTS Genetic test results of 720 patients from 27 centers were obtained. Patients with chromosomal deletion/duplication, non-ID genes, novel genes, and results with changes in more than one gene were excluded. A total of 558 patients with 341 different causative genes were included in the study. Pathway-based enrichment analysis of the ID-related genes via ClusterProfiler revealed 18 shared pathways, with lysine degradation and nicotine addiction being the most common. The most common of the 25 overrepresented DO terms was ID. The most frequently overrepresented GO biological process, cellular component, and molecular function terms were regulation of membrane potential, ion channel complex, and voltage-gated ion channel activity/voltage-gated channel activity, respectively. CONCLUSION Lysine degradation, nicotine addiction, and thyroid hormone signaling pathways are well-suited to be research areas for the discovery of new targeted therapies in ID patients.
Collapse
Affiliation(s)
- Çağatay Günay
- Department of Pediatric Neurology, Dokuz Eylul University Faculty of Medicine, Izmir, Turkey
| | - Duygu Aykol
- Department of Pediatric Neurology, Dokuz Eylul University Faculty of Medicine, Izmir, Turkey
| | - Özlem Özsoy
- Department of Pediatric Neurology, Dokuz Eylul University Faculty of Medicine, Izmir, Turkey
| | - Ece Sönmezler
- Izmir Biomedicine and Genome Center, Dokuz Eylul University Health Campus, Izmir, Turkey
| | - Yaren Sena Hanci
- Department of Pediatric Neurology, Dokuz Eylul University Faculty of Medicine, Izmir, Turkey
| | - Bülent Kara
- Department of Pediatric Neurology, Kocaeli University School of Medicine, Kocaeli, Turkey
| | | | - Naci Cine
- Department of Medical Genetics, Kocaeli University School of Medicine, Kocaeli, Turkey
| | - Adnan Deniz
- Department of Pediatric Neurology, Kocaeli University School of Medicine, Kocaeli, Turkey
| | - Tolgahan Özer
- Department of Medical Genetics, Kocaeli University School of Medicine, Kocaeli, Turkey
| | - Cemile Büşra Ölçülü
- Department of Child Neurology, Ege University Faculty of Medicine, Izmir, Turkey
| | - Özlem Yilmaz
- Department of Child Neurology, Ege University Faculty of Medicine, Izmir, Turkey
| | - Seda Kanmaz
- Department of Child Neurology, Ege University Faculty of Medicine, Izmir, Turkey
| | - Sanem Yilmaz
- Department of Child Neurology, Ege University Faculty of Medicine, Izmir, Turkey
| | - Hasan Tekgül
- Department of Child Neurology, Ege University Faculty of Medicine, Izmir, Turkey
| | - Nihal Yildiz
- Department of Pediatric Neurology, Karadeniz Technical University, Faculty of Medicine, Farabi Hospital, Trabzon, Turkey
| | - Elif Acar Arslan
- Department of Pediatric Neurology, Karadeniz Technical University, Faculty of Medicine, Farabi Hospital, Trabzon, Turkey
| | - Ali Cansu
- Department of Pediatric Neurology, Karadeniz Technical University, Faculty of Medicine, Farabi Hospital, Trabzon, Turkey
| | - Nihal Olgaç Dündar
- Department of Pediatric Neurology, İzmir Katip Çelebi University, Izmir, Turkey
| | - Fatma Kusgoz
- Department of Pediatric Neurology, Tepecik Research and Training Hospital, Izmir, Turkey
| | - Elif Didinmez
- Department of Pediatric Neurology, Tepecik Research and Training Hospital, Izmir, Turkey
| | - Pınar Gençpinar
- Department of Pediatric Neurology, İzmir Katip Çelebi University, Izmir, Turkey
| | - Tuğçe Aksu Uzunhan
- Department of Pediatric Neurology, Prof Dr Cemil Tascioglu City Hospital, Istanbul, Turkey
| | - Biray Ertürk
- Department of Pediatric Neurology, Prof Dr Cemil Tascioglu City Hospital, Istanbul, Turkey
| | - Alper Gezdirici
- Department of Medical Genetics, Başakşehir Çam and Sakura City Hospital, Istanbul, Turkey
| | - Akif Ayaz
- Department of Medical Genetics, Istanbul Medipol University School of Medicine, Istanbul, Turkey
| | - Akgün Ölmez
- Denizli Pediatric Neurology Clinic, Denizli, Turkey
| | - Müge Ayanoğlu
- Department of Child Neurology, Adnan Menderes University School of Medicine, Aydın, Turkey
| | - Ayşe Tosun
- Department of Child Neurology, Adnan Menderes University School of Medicine, Aydın, Turkey
| | - Yasemin Topçu
- Department of Pediatric Neurology, Istanbul Medipol University Faculty of Medicine, Istanbul, Turkey
| | - Betül Kiliç
- Department of Pediatric Neurology, Istanbul Medipol University Faculty of Medicine, Istanbul, Turkey
| | - Kürşad Aydin
- Department of Pediatric Neurology, Istanbul Medipol University Faculty of Medicine, Istanbul, Turkey
| | - Ezgi Çağlar
- Departments of Pediatric Neurology, Mersin University Faculty of Medicine, Mersin, Turkey
| | - Özlem Ersoy Kosvali
- Departments of Pediatric Neurology, Mersin University Faculty of Medicine, Mersin, Turkey
| | - Çetin Okuyaz
- Departments of Pediatric Neurology, Mersin University Faculty of Medicine, Mersin, Turkey
| | - Şeyda Besen
- Division of Pediatric Neurology, Başkent University Adana Medical and Research Center Faculty of Medicine, Adana, Turkey
| | - Leman Tekin Orgun
- Division of Pediatric Neurology, Başkent University Adana Medical and Research Center Faculty of Medicine, Adana, Turkey
| | - İlknur Erol
- Division of Pediatric Neurology, Başkent University Adana Medical and Research Center Faculty of Medicine, Adana, Turkey
| | - Deniz Yüksel
- Department of Pediatric Neurology, University of Health Sciences Faculty of Medicine, Dr Sami Ulus Maternity Child Health and Diseases Training and Research Hospital, Ankara, Turkey
| | - Abdullah Sezer
- Department of Genetics, University of Health Sciences Faculty of Medicine, Dr Sami Ulus Maternity Child Health and Diseases Training and Research Hospital, Ankara, Turkey
| | - Ergin Atasoy
- Department of Pediatric Neurology, University of Health Sciences Faculty of Medicine, Dr Sami Ulus Maternity Child Health and Diseases Training and Research Hospital, Ankara, Turkey
| | - Ülkühan Toprak
- Department of Pediatric Neurology, University of Health Sciences Faculty of Medicine, Dr Sami Ulus Maternity Child Health and Diseases Training and Research Hospital, Ankara, Turkey
| | - Serdal Güngör
- Department of Paediatric Neurology, Inonu University Faculty of Medicine, Turgut Ozal Research Center, Malatya, Turkey
| | - Bilge Ozgor
- Department of Paediatric Neurology, Inonu University Faculty of Medicine, Turgut Ozal Research Center, Malatya, Turkey
| | - Meral Karadağ
- Department of Paediatric Neurology, Inonu University Faculty of Medicine, Turgut Ozal Research Center, Malatya, Turkey
| | - Cengiz Dilber
- Department of Pediatric Neurology, Kahramanmaras Sutcu Imam University Faculty of Medicine, Kahramanmaraş, Turkey
| | - Bahtiyar Şahinoğlu
- Deparment of Genetics, Dr Ersin Arslan Traning and Research Hospital, Gaziantep, Turkey
| | - Emek Uyur Yalçin
- Departments of Pediatrics and Pediatric Neurology, University of Health Sciences, Zeynep Kamil Maternity and Children's Diseases Hospital, Istanbul, Turkey
| | - Nilüfer Eldes Hacifazlioglu
- Departments of Pediatrics and Pediatric Neurology, University of Health Sciences, Zeynep Kamil Maternity and Children's Diseases Hospital, Istanbul, Turkey
| | - Ahmet Yaramiş
- Diyarbakır Pediatric Neurology Clinic, Diyarbakır, Turkey
| | - Pınar Edem
- Department of Pediatric Neurology, Bakırcay University, Cigli District Training Hospital, Izmir, Turkey
| | - Hande Gezici Tekin
- Department of Pediatric Neurology, Bakırcay University, Cigli District Training Hospital, Izmir, Turkey
| | - Ünsal Yilmaz
- Department of Pediatric Neurology, Dr. Behcet Uz Children's Hospital, Izmir, Turkey
| | - Aycan Ünalp
- Department of Pediatric Neurology, Dr. Behcet Uz Children's Hospital, Izmir, Turkey
| | - Sevim Turay
- Department of Pediatric Neurology, Duzce University Faculty of Medicine, Düzce, Turkey
| | - Didem Biçer
- Department of Pediatric Neurology, Çukurova University Faculty of Medicine, Adana, Turkey
| | - Gülen Gül Mert
- Department of Pediatric Neurology, Çukurova University Faculty of Medicine, Adana, Turkey
| | - İpek Dokurel Çetin
- Department of Pediatric Neurology, Balıkesir Atatürk Training and Research Hospital, Balıkesir, Turkey
| | - Serkan Kirik
- Fırat University School of Medicine, Pediatric Neurology, Elazığ, Turkey
| | - Gülten Öztürk
- Department of Pediatric Neurology, Marmara University School of Medicine, Istanbul, Turkey
| | - Yasemin Karal
- Department of Pediatric Neurology, Trakya University, Faculty of Medicine, Edirne, Turkey
| | - Aslıhan Sanri
- Department of Pediatric Genetics, University of Health Sciences, Samsun Training and Research Hospital, Samsun, Turkey
| | - Ayşe Aksoy
- Department of Pediatric Neurology, Ondokuz Mayıs University, Samsun, Turkey
| | - Muzaffer Polat
- Department of Pediatric Neurology, Celal Bayar University School of Medicine, Manisa, Turkey
| | - Nezir Özgün
- Department of Pediatric Neurology, Mardin Artuklu University, Faculty of Health Sciences, Mardin, Turkey
| | - Didem Soydemir
- Department of Pediatric Neurology, Dokuz Eylul University Faculty of Medicine, Izmir, Turkey
| | - Gamze Sarikaya Uzan
- Department of Pediatric Neurology, Dokuz Eylul University Faculty of Medicine, Izmir, Turkey
| | - Döndü Ülker Üstebay
- Department of Pediatric Neurology, Dokuz Eylul University Faculty of Medicine, Izmir, Turkey
| | - Ayşen Gök
- Department of Pediatric Neurology, Dokuz Eylul University Faculty of Medicine, Izmir, Turkey
| | - Mehmet Can Yeşilmen
- Department of Pediatric Neurology, Dokuz Eylul University Faculty of Medicine, Izmir, Turkey
| | - Uluç Yiş
- Department of Pediatric Neurology, Dokuz Eylul University Faculty of Medicine, Izmir, Turkey
| | - Gökhan Karakülah
- Izmir Biomedicine and Genome Center, Dokuz Eylul University Health Campus, Izmir, Turkey
| | - Ahmet Bursali
- Izmir Biomedicine and Genome Center, Dokuz Eylul University Health Campus, Izmir, Turkey
| | - Yavuz Oktay
- Izmir Biomedicine and Genome Center, Dokuz Eylul University Health Campus, Izmir, Turkey
| | - Semra Hiz Kurul
- Department of Pediatric Neurology, Dokuz Eylul University Faculty of Medicine, Izmir, Turkey
- Izmir Biomedicine and Genome Center, Dokuz Eylul University Health Campus, Izmir, Turkey
| |
Collapse
|
19
|
Adak P, Banerjee N, Sinha S, Bandyopadhyay AK. Gamma-Aminobutyric Acid Type A Receptor Variants are Associated with Autism Spectrum Disorders. J Mol Neurosci 2023; 73:237-249. [PMID: 36943547 DOI: 10.1007/s12031-023-02113-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 03/01/2023] [Indexed: 03/23/2023]
Abstract
Despite several efforts to identify the causes of autism spectrum disorders (ASD), its etiology remains still unclear. Among other aspects, genes that encode neurotransmitter receptors are strong candidates for autism. Here, we wanted to study some genetic variants of gamma-aminobutyric acid (GABA) receptor subunit genes GABRB3, GABRG3, and GABRA5, located on chromosome 15q11-q13 that might contribute to the etiology of ASD in the affected children of West Bengal. rs7180158, rs2081648 (GABRB3); rs12910555 (GABRG3); rs35399885, rs35832850 (GABRA5) were analyzed in 316 children with ASD and 227 healthy controls. Phenotypic associations were evaluated by Childhood Autism Rating Scale (CARS). Gene expression levels were measured by quantitative real-time PCR. ASD probands showed a higher frequency of "A" allele for rs7180158, "G" allele for rs12901555, and "T" allele for rs35399885. The GA + AA genotypes (rs7180158) and CT + TT genotypes (rs35399885) were found to confer significant risk towards ASD. rs2081648 was found to have transmission bias in the family. Additionally, these variants were found to be associated with one or more of ASD-associated phenotypic traits. Multifactor dimensionality reduction (MDR) analyses showed mostly independent contributory effects of some of the variants. Again, the gene expression levels of GABRB3, GABRG3, and GABRA5 were downregulated in the cases than the controls. ForGABRA5 rs35399885, the CC genotypes corresponded to higher expression levels compared to the other groups. This study reveals that genetic variants of GABAA receptor subunit genes are significantly associated with ASD. No data for the mentioned variants are found in the population of West Bengal, India.
Collapse
Affiliation(s)
- Pallabi Adak
- Manovikas Biomedical Research and Diagnostic Centre, Manovikas Kendra Rehabilitation and Research Institute for the Handicapped, 700107, Kolkata, West Bengal, India
| | - Nilanjana Banerjee
- Manovikas Biomedical Research and Diagnostic Centre, Manovikas Kendra Rehabilitation and Research Institute for the Handicapped, 700107, Kolkata, West Bengal, India.
| | - Swagata Sinha
- Out Patient Department, Manovikas Kendra Rehabilitation and Research Institute for the Handicapped, 700107, Kolkata, West Bengal, India
| | - Apurba Kumar Bandyopadhyay
- Out Patient Department, Manovikas Kendra Rehabilitation and Research Institute for the Handicapped, 700107, Kolkata, West Bengal, India
| |
Collapse
|
20
|
Chaudhary R, Steinson E. Genes and their Involvement in the Pathogenesis of Autism Spectrum Disorder: Insights from Earlier Genetic Studies. NEUROBIOLOGY OF AUTISM SPECTRUM DISORDERS 2023:375-415. [DOI: 10.1007/978-3-031-42383-3_18] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
21
|
Yadav A, Verhaegen S, Filis P, Domanska D, Lyle R, Sundaram AYM, Leithaug M, Østby GC, Aleksandersen M, Berntsen HF, Zimmer KE, Fowler PA, Paulsen RE, Ropstad E. Exposure to a human relevant mixture of persistent organic pollutants or to perfluorooctane sulfonic acid alone dysregulates the developing cerebellum of chicken embryo. ENVIRONMENT INTERNATIONAL 2022; 166:107379. [PMID: 35792514 DOI: 10.1016/j.envint.2022.107379] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 05/07/2022] [Accepted: 06/24/2022] [Indexed: 06/15/2023]
Abstract
Prenatal exposure to persistent organic pollutants (POPs) is associated with neurodevelopmental disorders. In the present study, we explored whether a human-relevant POP mixture affects the development of chicken embryo cerebellum. We used a defined mixture of 29 POPs, with chemical composition and concentrations based on blood levels in the Scandinavian population. We also evaluated exposure to a prominent compound in the mixture, perfluorooctane sulfonic acid (PFOS), alone. Embryos (n = 7-9 per exposure group) were exposed by injection directly into the allantois at embryonic day 13 (E13). Cerebella were isolated at E17 and subjected to morphological, RNA-seq and shot-gun proteomics analyses. There was a reduction in thickness of the molecular layer of cerebellar cortex in both exposure scenarios. Exposure to the POP mixture significantly affected expression of 65 of 13,800 transcripts, and 43 of 2,568 proteins, when compared to solvent control. PFOS alone affected expression of 80 of 13,859 transcripts, and 69 of 2,555 proteins. Twenty-five genes and 15 proteins were common for both exposure groups. These findings point to alterations in molecular events linked to retinoid X receptor (RXR) signalling, neuronal cell proliferation and migration, cellular stress responses including unfolded protein response, lipid metabolism, and myelination. Exposure to the POP mixture increased methionine oxidation, whereas PFOS decreased oxidation. Several of the altered genes and proteins are involved in a wide variety of neurological disorders. We conclude that POP exposure can interfere with fundamental aspects of neurodevelopment, altering molecular pathways that are associated with adverse neurocognitive and behavioural outcomes.
Collapse
Affiliation(s)
- Ajay Yadav
- Department of Production Animal Clinical Sciences, Norwegian University of Life Sciences, P.O. Box 5003, NO-1432 Ås, Norway; Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, P.O. Box 1068, Blindern, NO-0316 Oslo, Norway.
| | - Steven Verhaegen
- Department of Production Animal Clinical Sciences, Norwegian University of Life Sciences, P.O. Box 5003, NO-1432 Ås, Norway.
| | - Panagiotis Filis
- Institute of Medical Sciences, School of Medicine, Medical Sciences & Nutrition, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, UK.
| | - Diana Domanska
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Department of Pathology, Oslo University Hospital-Rikshospitalet, Oslo, Norway.
| | - Robert Lyle
- Department of Medical Genetics, Oslo University Hospital and University of Oslo, Oslo, Norway; Centre for Fertility and Health, Norwegian Institute of Public Health, Oslo, Norway.
| | - Arvind Y M Sundaram
- Department of Medical Genetics, Oslo University Hospital and University of Oslo, Oslo, Norway.
| | - Magnus Leithaug
- Department of Medical Genetics, Oslo University Hospital and University of Oslo, Oslo, Norway.
| | - Gunn Charlotte Østby
- Department of Production Animal Clinical Sciences, Norwegian University of Life Sciences, P.O. Box 5003, NO-1432 Ås, Norway.
| | - Mona Aleksandersen
- Department of Preclinical Sciences and Pathology, Norwegian University of Life Sciences, P.O. Box 5003, NO-1432 Ås, Norway.
| | - Hanne Friis Berntsen
- Department of Production Animal Clinical Sciences, Norwegian University of Life Sciences, P.O. Box 5003, NO-1432 Ås, Norway; National Institute of Occupational Health, P.O. Box 5330 Majorstuen, NO-0304, Oslo, Norway.
| | - Karin Elisabeth Zimmer
- Department of Preclinical Sciences and Pathology, Norwegian University of Life Sciences, P.O. Box 5003, NO-1432 Ås, Norway.
| | - Paul A Fowler
- Institute of Medical Sciences, School of Medicine, Medical Sciences & Nutrition, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, UK.
| | - Ragnhild Elisabeth Paulsen
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, P.O. Box 1068, Blindern, NO-0316 Oslo, Norway.
| | - Erik Ropstad
- Department of Production Animal Clinical Sciences, Norwegian University of Life Sciences, P.O. Box 5003, NO-1432 Ås, Norway.
| |
Collapse
|
22
|
Mehta R, Kuhad A, Bhandari R. Nitric oxide pathway as a plausible therapeutic target in autism spectrum disorders. Expert Opin Ther Targets 2022; 26:659-679. [DOI: 10.1080/14728222.2022.2100252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Affiliation(s)
- Rishab Mehta
- Pharmacology Research Laboratory, University Institute of Pharmaceutical Sciences, UGC-Centre of Advanced Study, Panjab University, Chandigarh – 160 014 India
| | - Anurag Kuhad
- Pharmacology Research Laboratory, University Institute of Pharmaceutical Sciences, UGC-Centre of Advanced Study, Panjab University, Chandigarh – 160 014 India
| | - Ranjana Bhandari
- Pharmacology Research Laboratory, University Institute of Pharmaceutical Sciences, UGC-Centre of Advanced Study, Panjab University, Chandigarh – 160 014 India
| |
Collapse
|
23
|
Ali ZA, Yasseen AA, McAllister KA, Al-Dujailli A, Al-Karaqully AJ, Jumaah AS. SNP-PCR genotyping links alterations in the GABAA receptor (GABRG3: rs208129) and RELN (rs73670) genes to autism spectrum disorder among peadiatric Iraqi Arabs. Mol Biol Rep 2022; 49:6019-6028. [PMID: 35403940 PMCID: PMC9270290 DOI: 10.1007/s11033-022-07388-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 03/16/2022] [Indexed: 11/22/2022]
Abstract
INTRODUCTION Autism spectrum disorder (ASD) is an increasing concern among the Iraqi Arab population. The genetic alterations that cause ASD are likely to converge at the synapse. This study investigated polymorphisms in the GABAA receptor subunit (GABRG3) and the RELN gene as putative biomarkers of ASD in a pediatric population in Iraq. METHODS The case control study included 60 patients with a clinical diagnosis of ASD (mild, moderate, or severe) according to DSM-IV criteria and matched healthy controls (n = 60). Blood samples were collected for DNA genotyping of SNPs rs736707 and rs208129 for RELN and GABRG3 using allele specific PCR. Assessment of genotype and allele distributions in patient groups used odd ratios (OR) with 95% confidence intervals and the Chi-square test. All statistical analysis was performed used SPSS software. RESULT The patient cohort was highly consanguineous, with increased ratio (p > 0.05) of males to females (3:1) in both ASD (mean age, 6.66 ± 3.05) and controls (mean age, 5.76 ± 2.3). Both GABRG3 rs208129 genotypes TT (OR 4.33, p = 0.0015) and TA (OR 0.259, P = 0.008), and the T and A alleles were significantly associated with ASD. The RELN rs736707 TC genotype (OR 2.626, P = 0.034) was the only significant association with ASD. CONCLUSION GABRG3 SNP rs208129 is a leading biomarker to predict genetic vulnerability to ASD in Iraqi Arabs. Expanded SNP panels and increased sample sizes are required for future GABRG3 studies, and to reach a consensus on RELN utility. Future ASD screening programs in Iraq should include genetic metrics in addition to clinical phenotype assessments.
Collapse
Affiliation(s)
- Zainab A. Ali
- Department of Pathology and Forensic Medicine, Faculty of Medicine, University of Kerbala, Kerbala Governorate, Iraq
| | - Akeel A. Yasseen
- Department of Pathology and Forensic Medicine, Faculty of Medicine, University of Kufa, P.O. Box 21, Kufa, Iraq
| | | | - Arafat Al-Dujailli
- Department of Pathology and Forensic Medicine, Faculty of Medicine, University of Kufa, P.O. Box 21, Kufa, Iraq
| | | | - Alaa S. Jumaah
- Department of Pathology and Forensic Medicine, Faculty of Medicine, University of Kufa, P.O. Box 21, Kufa, Iraq
| |
Collapse
|
24
|
Park S, Zikopoulos B, Yazdanbakhsh A. Visual illusion susceptibility in autism: A neural model. Eur J Neurosci 2022; 56:4246-4265. [PMID: 35701859 PMCID: PMC9541695 DOI: 10.1111/ejn.15739] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 04/04/2022] [Accepted: 06/06/2022] [Indexed: 11/26/2022]
Abstract
While atypical sensory perception is reported among individuals with autism spectrum disorder (ASD), the underlying neural mechanisms of autism that give rise to disruptions in sensory perception remain unclear. We developed a neural model with key physiological, functional and neuroanatomical parameters to investigate mechanisms underlying the range of representations of visual illusions related to orientation perception in typically developed subjects compared to individuals with ASD. Our results showed that two theorized autistic traits, excitation/inhibition imbalance and weakening of top‐down modulation, could be potential candidates for reduced susceptibility to some visual illusions. Parametric correlation between cortical suppression, balance of excitation/inhibition, feedback from higher visual areas on one hand and susceptibility to a class of visual illusions related to orientation perception on the other hand provide the opportunity to investigate the contribution and complex interactions of distinct sensory processing mechanisms in ASD. The novel approach used in this study can be used to link behavioural, functional and neuropathological studies; estimate and predict perceptual and cognitive heterogeneity in ASD; and form a basis for the development of novel diagnostics and therapeutics.
Collapse
Affiliation(s)
- Sangwook Park
- Computational Neuroscience and Vision Laboratory, Boston University, Boston, Massachusetts, USA
| | - Basilis Zikopoulos
- Human Systems Neuroscience Laboratory, Department of Health Sciences, Boston University, Boston, Massachusetts, USA.,Department of Anatomy and Neurobiology, Boston University School of Medicine, Boston, Massachusetts, USA.,Center for Systems Neuroscience, Boston University, Boston, Massachusetts, USA.,Graduate Program for Neuroscience, Boston University, Boston, Massachusetts, USA
| | - Arash Yazdanbakhsh
- Computational Neuroscience and Vision Laboratory, Boston University, Boston, Massachusetts, USA.,Center for Systems Neuroscience, Boston University, Boston, Massachusetts, USA.,Graduate Program for Neuroscience, Boston University, Boston, Massachusetts, USA.,Department of Psychological and Brain Sciences, Boston University, Boston, Massachusetts, USA
| |
Collapse
|
25
|
Gill PS, Dweep H, Rose S, Wickramasinghe PJ, Vyas KK, McCullough S, Porter-Gill PA, Frye RE. Integrated microRNA–mRNA Expression Profiling Identifies Novel Targets and Networks Associated with Autism. J Pers Med 2022; 12:jpm12060920. [PMID: 35743705 PMCID: PMC9225282 DOI: 10.3390/jpm12060920] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 05/23/2022] [Accepted: 05/27/2022] [Indexed: 01/27/2023] Open
Abstract
Autism spectrum disorder (ASD) is a complex neurodevelopmental disorder, with mutations in hundreds of genes contributing to its risk. Herein, we studied lymphoblastoid cell lines (LCLs) from children diagnosed with autistic disorder (n = 10) and controls (n = 7) using RNA and miRNA sequencing profiles. The sequencing analysis identified 1700 genes and 102 miRNAs differentially expressed between the ASD and control LCLs (p ≤ 0.05). The top upregulated genes were GABRA4, AUTS2, and IL27, and the top upregulated miRNAs were hsa-miR-6813-3p, hsa-miR-221-5p, and hsa-miR-21-5p. The RT-qPCR analysis confirmed the sequencing results for randomly selected candidates: AUTS2, FMR1, PTEN, hsa-miR-15a-5p, hsa-miR-92a-3p, and hsa-miR-125b-5p. The functional enrichment analysis showed pathways involved in ASD control proliferation of neuronal cells, cell death of immune cells, epilepsy or neurodevelopmental disorders, WNT and PTEN signaling, apoptosis, and cancer. The integration of mRNA and miRNA sequencing profiles by miRWalk2.0 identified correlated changes in miRNAs and their targets’ expression. The integration analysis found significantly dysregulated miRNA–gene pairs in ASD. Overall, these findings suggest that mRNA and miRNA expression profiles in ASD are greatly altered in LCLs and reveal numerous miRNA–gene interactions that regulate critical pathways involved in the proliferation of neuronal cells, cell death of immune cells, and neuronal development.
Collapse
Affiliation(s)
- Pritmohinder S. Gill
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72202, USA;
- Arkansas Children′s Research Institute, Little Rock, AR 72202, USA; (K.K.V.); (S.M.); (P.A.P.-G.)
- Correspondence: ; Tel.: +1-501-364-2743
| | - Harsh Dweep
- The Wistar Institute, 3601 Spruce St., Philadelphia, PA 19104, USA; (H.D.); (P.J.W.)
| | - Shannon Rose
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72202, USA;
- Arkansas Children′s Research Institute, Little Rock, AR 72202, USA; (K.K.V.); (S.M.); (P.A.P.-G.)
| | | | - Kanan K. Vyas
- Arkansas Children′s Research Institute, Little Rock, AR 72202, USA; (K.K.V.); (S.M.); (P.A.P.-G.)
| | - Sandra McCullough
- Arkansas Children′s Research Institute, Little Rock, AR 72202, USA; (K.K.V.); (S.M.); (P.A.P.-G.)
| | - Patricia A. Porter-Gill
- Arkansas Children′s Research Institute, Little Rock, AR 72202, USA; (K.K.V.); (S.M.); (P.A.P.-G.)
| | - Richard E. Frye
- Barrow Neurological Institute at Phoenix Children′s Hospital, Phoenix, AZ 85016, USA;
- Department of Child Health, University of Arizona College of Medicine, Phoenix, AZ 85004, USA
| |
Collapse
|
26
|
Santrač A, Bijelić D, Stevanović V, Banićević M, Aranđelović J, Batinić B, Sharmin D, Cook JM, Savić MM. Postweaning positive modulation of α5GABAA receptors improves autism-like features in prenatal valproate rat model in a sex-specific manner. Autism Res 2022; 15:806-820. [PMID: 35266641 DOI: 10.1002/aur.2699] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Revised: 02/11/2022] [Accepted: 02/20/2022] [Indexed: 12/27/2022]
Abstract
Autism spectrum disorder (ASD), as a common neurodevelopmental disorder that encompasses impairments in social communication and interaction, as well as repetitive and restrictive behavior, still awaits an effective treatment strategy. The involvement of GABAergic neurotransmission, and especially a deficit of GABAA receptors that contain the α5 subunits, were implicated in pathogenesis of ASD. Therefore, we tested MP-III-022, a positive allosteric modulator (PAM) selective for α5GABAA receptors, in Wistar rats prenatally exposed to valproic acid, as an animal model useful for studying ASD. Postweaning rats of both sexes were treated for 7 days with vehicle or MP-III-022 at two doses pharmacokinetically determined as selective, and thereafter tested in a behavioral battery (social interaction test, elevated plus maze, spontaneous locomotor activity, and standard and reverse Morris water maze). Additional rats were used for establishing a primary neuronal culture and performing calcium imaging, and determination of hippocampal mRNA levels of GABRA5, NKCC1, and KCC2. MP-III-022 prevented impairments in many parameters connected with social, repetitive and restrictive behavioral domains. The lower and higher dose was more effective in males and females, respectively. Intriguingly, MP-III-022 elicited certain changes in control animals similar to those manifested in valproate animals themselves. Behavioral results were mirrored in GABA switch and spontaneous neuronal activity, assessed with calcium imaging, and also in expression changes of three genes analyzed. Our data support a role of α5GABAA receptors in pathophysiology of ASD, and suggest a potential application of selective PAMs in its treatment, that needs to be researched in a sex-specific manner. LAY SUMMARY: In rats prenatally exposed to valproate as a model of autism, a modulator of α5GABAA receptors ameliorated social, repetitive and restrictive impairments, and, intriguingly, elicited certain autism-like changes in control rats. Behavioral results were mirrored in GABA switch and spontaneous neuronal activity, and partly in gene expression changes. This shows a role of α5GABAA receptors in pathophysiology of ASD, and a potential application of their selective modulators in its treatment.
Collapse
Affiliation(s)
- Anja Santrač
- Department of Pharmacology, University of Belgrade - Faculty of Pharmacy, Belgrade, Serbia
| | - Dunja Bijelić
- Centre for Laser Microscopy, Institute of Physiology and Biochemistry "Jean Giaja", University of Belgrade - Faculty of Biology, Belgrade, Serbia
| | - Vladimir Stevanović
- Department of Pharmacology, University of Belgrade - Faculty of Pharmacy, Belgrade, Serbia
| | - Marija Banićević
- Department of Pharmacology, University of Belgrade - Faculty of Pharmacy, Belgrade, Serbia.,Institute of Pharmacy and Molecular Biotechnology, Bioinformatics and Functional Genomics, Ruprecht-Karls University Heidelberg, Heidelberg, Germany
| | - Jovana Aranđelović
- Department of Pharmacology, University of Belgrade - Faculty of Pharmacy, Belgrade, Serbia
| | - Bojan Batinić
- Department of Physiology, University of Belgrade - Faculty of Pharmacy, Belgrade, Serbia
| | - Dishary Sharmin
- Department of Chemistry and Biochemistry, University of Wisconsin-Milwaukee and the Milwaukee Institute of Drug Discovery, Milwaukee, Wisconsin, USA
| | - James M Cook
- Department of Chemistry and Biochemistry, University of Wisconsin-Milwaukee and the Milwaukee Institute of Drug Discovery, Milwaukee, Wisconsin, USA
| | - Miroslav M Savić
- Department of Pharmacology, University of Belgrade - Faculty of Pharmacy, Belgrade, Serbia
| |
Collapse
|
27
|
Zhao H, Mao X, Zhu C, Zou X, Peng F, Yang W, Li B, Li G, Ge T, Cui R. GABAergic System Dysfunction in Autism Spectrum Disorders. Front Cell Dev Biol 2022; 9:781327. [PMID: 35198562 PMCID: PMC8858939 DOI: 10.3389/fcell.2021.781327] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 12/16/2021] [Indexed: 12/19/2022] Open
Abstract
Autism spectrum disorder (ASD) refers to a series of neurodevelopmental diseases characterized by two hallmark symptoms, social communication deficits and repetitive behaviors. Gamma-aminobutyric acid (GABA) is one of the most important inhibitory neurotransmitters in the central nervous system (CNS). GABAergic inhibitory neurotransmission is critical for the regulation of brain rhythm and spontaneous neuronal activities during neurodevelopment. Genetic evidence has identified some variations of genes associated with the GABA system, indicating an abnormal excitatory/inhibitory (E/I) neurotransmission ratio implicated in the pathogenesis of ASD. However, the specific molecular mechanism by which GABA and GABAergic synaptic transmission affect ASD remains unclear. Transgenic technology enables translating genetic variations into rodent models to further investigate the structural and functional synaptic dysregulation related to ASD. In this review, we summarized evidence from human neuroimaging, postmortem, and genetic and pharmacological studies, and put emphasis on the GABAergic synaptic dysregulation and consequent E/I imbalance. We attempt to illuminate the pathophysiological role of structural and functional synaptic dysregulation in ASD and provide insights for future investigation.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Ranji Cui
- *Correspondence: Tongtong Ge, ; Ranji Cui,
| |
Collapse
|
28
|
Barki M, Xue H. GABRB2, a key player in neuropsychiatric disorders and beyond. Gene 2022; 809:146021. [PMID: 34673206 DOI: 10.1016/j.gene.2021.146021] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2020] [Revised: 08/05/2021] [Accepted: 09/14/2021] [Indexed: 01/11/2023]
Abstract
The GABA receptors represent the main inhibitory system in the central nervous system that ensure synaptogenesis, neurogenesis, and the regulation of neuronal plasticity and learning. GABAA receptors are pentameric in structure and belong to the Cys-loop superfamily. The GABRB2 gene, located on chromosome 5q34, encodes the β2 subunit that combines with the α and γ subunits to form the major subtype of GABAA receptors, which account for 43% of all GABAA receptors in the mammalian brain. Each subunit probably consists of an extracellular N-terminal domain, four membrane-spanning segments, a large intracellular loop between TM3 and TM4, and an extracellular C-terminal domain. Alternative splicing of the RNA transcript of the GABRB2 gene gives rise at least to four long and short isoforms with dissimilar electrophysiological properties. Furthermore, GABRB2 is imprinted and subjected to epigenetic regulation and positive selection. It has been associated with schizophrenia first in Han Chinese, and subsequently validated in other populations. Gabrb2 knockout mice also exhibited schizophrenia-like behavior and neuroinflammation that were ameliorated by the antipsychotic drug risperidone. GABRB2 was also associated with other neuropsychiatric disorders including bipolar disorder, epilepsy, autism spectrum disorder, Alzheimer's disease, frontotemporal dementia, substance dependence, depression, internet gaming disorder, and premenstrual dysphoric disorder. Recently, it has been postulated that GABRB2 might be a potential marker for different cancer types. As GABRB2 has a pivotal role in the central nervous system and is increasingly recognized to contribute to human diseases, further understanding of its structure and function may expedite the generation of new therapeutic approaches.
Collapse
Affiliation(s)
- Manel Barki
- Center for Cancer Genomics, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Hong Xue
- Center for Cancer Genomics, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China; Division of Life Science and Applied Genomics Center, Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China.
| |
Collapse
|
29
|
Aykan S, Puglia MH, Kalaycıoğlu C, Pelphrey KA, Tuncalı T, Nalçacı E. Right Anterior Theta Hypersynchrony as a Quantitative Measure Associated with Autistic Traits and K-Cl Cotransporter KCC2 Polymorphism. J Autism Dev Disord 2022; 52:61-72. [PMID: 33635423 DOI: 10.1007/s10803-021-04924-x] [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] [Accepted: 02/10/2021] [Indexed: 10/22/2022]
Abstract
Our aim was to use theta coherence as a quantitative trait to investigate the relation of the polymorphisms in NKCC1 (rs3087889) and KCC2 (rs9074) channel protein genes to autistic traits (AQ) in neurotypicals. Coherence values for candidate connection regions were calculated from eyes-closed resting EEGs in two independent groups. Hypersynchrony within the right anterior region was related to AQ in both groups (p < 0.05), and variability in this hypersynchrony was related to the rs9074 polymorphism in the total group (p < 0.05). In conclusion, theta hypersynchrony within the right anterior region during eyes-closed rest can be considered a quantitative measure for autistic traits. Replicating our findings in two independent populations with different backgrounds strengthens the validity of the current study.
Collapse
Affiliation(s)
- Simge Aykan
- Department of Physiology, Ankara University School of Medicine, Ankara, Turkey.
| | - Meghan H Puglia
- Department of Neurology, University of Virginia, Charlottesville, VA, USA
- Department of Psychology, University of Virginia, Charlottesville, VA, USA
| | - Canan Kalaycıoğlu
- Department of Physiology, Ankara University School of Medicine, Ankara, Turkey
| | - Kevin A Pelphrey
- Department of Neurology, University of Virginia, Charlottesville, VA, USA
| | - Timur Tuncalı
- Department of Medical Genetics, Ankara University School of Medicine, Ankara, Turkey
| | - Erhan Nalçacı
- Department of Physiology, Ankara University School of Medicine, Ankara, Turkey
| |
Collapse
|
30
|
Lee J, Ha S, Ahn J, Lee ST, Choi JR, Cheon KA. The Role of Ion Channel-Related Genes in Autism Spectrum Disorder: A Study Using Next-Generation Sequencing. Front Genet 2021; 12:595934. [PMID: 34712263 PMCID: PMC8546317 DOI: 10.3389/fgene.2021.595934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 09/21/2021] [Indexed: 11/25/2022] Open
Abstract
The clinical heterogeneity of autism spectrum disorder (ASD) is closely associated with the diversity of genes related to ASD pathogenesis. With their low effect size, it has been hard to define the role of common variants of genes in ASD phenotype. In this study, we reviewed genetic results and clinical scores widely used for ASD diagnosis to investigate the role of genes in ASD phenotype considering their functions in molecular pathways. Genetic data from next-generation sequencing (NGS) were collected from 94 participants with ASD. We analyzed enrichment of cellular processes and gene ontology using the Database for Annotation, Visualization, and Integrated Discovery (DAVID). We compared clinical characteristics according to genetic functional characteristics. We found 266 genes containing nonsense, frame shift, missense, and splice site mutations. Results from DAVID revealed significant enrichment for “ion channel” with an enrichment score of 8.84. Moreover, ASD participants carrying mutations in ion channel-related genes showed higher total IQ (p = 0.013) and lower repetitive, restricted behavior (RRB)-related scores (p = 0.003) and mannerism subscale of social responsiveness scale scores, compared to other participants. Individuals with variants in ion channel genes showed lower RRB scores, suggesting that ion channel genes might be relatively less associated with RRB pathogenesis. These results contribute to understanding of the role of common variants in ASD and could be important in the development of precision medicine of ASD.
Collapse
Affiliation(s)
- Junghan Lee
- Division of Child and Adolescent Psychiatry, Department of Psychiatry, Severance Hospital, Institute of Behavioral Science in Medicine, Yonsei University College of Medicine, Seoul, South Korea
| | - Sungji Ha
- Department of Psychiatry, Institute of Behavioral Science in Medicine, Yonsei University College of Medicine, Seoul, South Korea
| | - Jaeun Ahn
- Division of Child and Adolescent Psychiatry, Department of Psychiatry, Severance Hospital, Institute of Behavioral Science in Medicine, Yonsei University College of Medicine, Seoul, South Korea
| | - Seung-Tae Lee
- Department of Laboratory Medicine, Yonsei University College of Medicine, Seoul, South Korea
| | - Jong Rak Choi
- Department of Laboratory Medicine, Yonsei University College of Medicine, Seoul, South Korea
| | - Keun-Ah Cheon
- Division of Child and Adolescent Psychiatry, Department of Psychiatry, Severance Hospital, Institute of Behavioral Science in Medicine, Yonsei University College of Medicine, Seoul, South Korea
| |
Collapse
|
31
|
Ahring PK, Liao VWY, Gardella E, Johannesen KM, Krey I, Selmer KK, Stadheim BF, Davis H, Peinhardt C, Koko M, Coorg RK, Syrbe S, Bertsche A, Santiago-Sim T, Diemer T, Fenger CD, Platzer K, Eichler EE, Lerche H, Lemke JR, Chebib M, Møller RS. Gain-of-function variants in GABRD reveal a novel pathway for neurodevelopmental disorders and epilepsy. Brain 2021; 145:1299-1309. [PMID: 34633442 DOI: 10.1093/brain/awab391] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 09/15/2021] [Accepted: 09/20/2021] [Indexed: 11/14/2022] Open
Abstract
A potential link between GABRD encoding the δ subunit of extrasynaptic GABAA receptors and neurodevelopmental disorders has largely been disregarded due to conflicting conclusions from early studies. However, we identified seven heterozygous missense GABRD variants in 10 patients with neurodevelopmental disorders and generalized epilepsy. One variant occurred in two sibs of healthy parents with presumed somatic mosaicism, another segregated with the disease in three affected family members, and the remaining five occurred de novo in sporadic patients. Electrophysiological measurements were used to determine the functional consequence of the seven missense δ subunit variants in receptor combinations of α1β3δ and α4β2δ GABAA receptors. This was accompanied by analysis of electro-clinical phenotypes of the affected individuals. We determined that five of the seven variants caused altered function of the resulting α1β3δ and α4β2δ GABAA receptors. Surprisingly, four of the five variants led to gain-of-function effects whereas one led to a loss-of-function effect. The stark differences between the gain-of-function and loss-of function effects were mirrored by the clinical phenotypes. Six patients with gain-of-function variants shared common phenotypes: neurodevelopmental disorders with generalized epilepsy, behavioral issues, and various degrees of intellectual disability. Six patients with gain-of-function variants shared common phenotypes: neurodevelopmental disorders with behavioral issues, various degrees of intellectual disability, generalized epilepsy with atypical absences and generalized myoclonic and/or bilateral tonic-clonic seizures. The EEG showed qualitative analogies among the different gain-of-function variant carriers consisting of focal slowing in the occipital regions often preceding irregular generalized epileptiform discharges, with frontal predominance. In contrast, the one patient carrying a loss-of-function variant had normal intelligence, no seizure history but has a diagnosis of autism spectrum disorder and suffering from elevated internalizing psychiatric symptoms. We hypothesize that increase in tonic GABA-evoked current levels mediated by δ-containing extrasynaptic GABAA receptors lead to abnormal neurotransmission, which represent a novel mechanism for severe neurodevelopmental disorders. In support of this, the electro-clinical findings for the gain-of-function GABRD variants resemble the phenotypic spectrum reported in patients with missense SLC6A1 (GABA uptake transporter) variants. This also indicates that the phenomenon of extrasynaptic receptor over-activity is observed in a broader range of patients with neurodevelopmental disorders, since SLC6A1 loss-of-function variants also lead to overactive extrasynaptic δ-containing GABAA receptors. These findings have implications when selecting potential treatment options, since a substantial portion of available anti-seizure medication act by enhancing GABAergic function either directly or indirectly, which could exacerbate symptoms in patients with gain-of-function GABRD variants.
Collapse
Affiliation(s)
- Philip K Ahring
- Brain and Mind Centre, School of Pharmacy, Faculty of Medicine and Health, The University of Sydney; Sydney, New South Wales, Australia
| | - Vivian W Y Liao
- Brain and Mind Centre, School of Pharmacy, Faculty of Medicine and Health, The University of Sydney; Sydney, New South Wales, Australia
| | - Elena Gardella
- Department of Epilepsy Genetics and Personalized Treatment, The Danish Epilepsy Centre; Dianalund, Denmark.,Department of Regional Health Research, University of Southern Denmark; Odense, Denmark
| | - Katrine M Johannesen
- Department of Epilepsy Genetics and Personalized Treatment, The Danish Epilepsy Centre; Dianalund, Denmark.,Department of Regional Health Research, University of Southern Denmark; Odense, Denmark
| | - Ilona Krey
- Institute of Human Genetics, University of Leipzig Medical Center; Leipzig, Germany
| | - Kaja K Selmer
- National Centre for Rare Epilepsy-Related Disorders, Oslo University Hospital; Oslo, Norway.,Department of Medical Genetics, Oslo University Hospital; Oslo, Norway
| | - Barbro F Stadheim
- Department of Medical Genetics, Oslo University Hospital; Oslo, Norway
| | - Hannah Davis
- Division of Medical Genetics, Department of Human Genetics, Emory University School of Medicine; Atlanta, GA, USA
| | - Charlotte Peinhardt
- Division of Medical Genetics, Department of Human Genetics, Emory University School of Medicine; Atlanta, GA, USA
| | - Mahmoud Koko
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain Research, University of Tübingen; Tübingen, Germany
| | - Rohini K Coorg
- Department of Pediatrics and Neurology, Neurophysiology and Epilepsy, Baylor College of Medicine; Houston, Texas, USA
| | - Steffen Syrbe
- Division of Paediatric Epileptology, Centre for Paediatric and Adolescent Medicine, University Hospital Heidelberg; Heidelberg, Germany
| | - Astrid Bertsche
- University Hospital for Children and Adolescents, Neuropaediatrics; Rostock, Germany.,University Hospital for Children and Adolescents, Center for Pediatric Research; Leipzig, Germany
| | | | - Tue Diemer
- Department of Clinical Genetics, Aalborg University Hospital; Aalborg, Denmark
| | - Christina D Fenger
- Department of Epilepsy Genetics and Personalized Treatment, The Danish Epilepsy Centre; Dianalund, Denmark.,Department of Regional Health Research, University of Southern Denmark; Odense, Denmark
| | - Konrad Platzer
- Institute of Human Genetics, University of Leipzig Medical Center; Leipzig, Germany
| | - Evan E Eichler
- Department of Genome Sciences, University of Washington School of Medicine; Seattle, WA, USA.,Howard Hughes Medical Institute, University of Washington; Seattle, WA 98195, USA
| | - Holger Lerche
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain Research, University of Tübingen; Tübingen, Germany
| | - Johannes R Lemke
- Institute of Human Genetics, University of Leipzig Medical Center; Leipzig, Germany
| | - Mary Chebib
- Brain and Mind Centre, School of Pharmacy, Faculty of Medicine and Health, The University of Sydney; Sydney, New South Wales, Australia
| | - Rikke S Møller
- Department of Epilepsy Genetics and Personalized Treatment, The Danish Epilepsy Centre; Dianalund, Denmark.,Department of Regional Health Research, University of Southern Denmark; Odense, Denmark
| |
Collapse
|
32
|
Rodriguez-Gomez DA, Garcia-Guaqueta DP, Charry-Sánchez JD, Sarquis-Buitrago E, Blanco M, Velez-van-Meerbeke A, Talero-Gutiérrez C. A systematic review of common genetic variation and biological pathways in autism spectrum disorder. BMC Neurosci 2021; 22:60. [PMID: 34627165 PMCID: PMC8501721 DOI: 10.1186/s12868-021-00662-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 09/16/2021] [Indexed: 01/21/2023] Open
Abstract
Background Autism spectrum disorder (ASD) is a complex neurodevelopmental condition characterized by persistent deficits in social communication and interaction. Common genetic variation appears to play a key role in the development of this condition. In this systematic review, we describe the relationship between genetic variations and autism. We created a gene dataset of the genes involved in the pathogenesis of autism and performed an over-representation analysis to evaluate the biological functions and molecular pathways that may explain the associations between these variants and the development of ASD. Results 177 studies and a gene set composed of 139 were included in this qualitative systematic review. Enriched pathways in the over-representation analysis using the KEGG pathway database were mostly associated with neurotransmitter receptors and their subunits. Major over-represented biological processes were social behavior, vocalization behavior, learning and memory. The enriched cellular component of the proteins encoded by the genes identified in this systematic review were the postsynaptic membrane and the cell junction. Conclusions Among the biological processes that were examined, genes involved in synaptic integrity, neurotransmitter metabolism, and cell adhesion molecules were significantly involved in the development of autism. Supplementary Information The online version contains supplementary material available at 10.1186/s12868-021-00662-z.
Collapse
Affiliation(s)
- Diego Alejandro Rodriguez-Gomez
- Neuroscience Research Group (NeURos), NeuroVitae Center for Neuroscience, School of Medicine and Health Sciences, Universidad del Rosario, Carrera 24 No. 63C-69, 111221, Bogotá D.C., Colombia
| | - Danna Paola Garcia-Guaqueta
- Neuroscience Research Group (NeURos), NeuroVitae Center for Neuroscience, School of Medicine and Health Sciences, Universidad del Rosario, Carrera 24 No. 63C-69, 111221, Bogotá D.C., Colombia
| | - Jesús David Charry-Sánchez
- Neuroscience Research Group (NeURos), NeuroVitae Center for Neuroscience, School of Medicine and Health Sciences, Universidad del Rosario, Carrera 24 No. 63C-69, 111221, Bogotá D.C., Colombia
| | - Elias Sarquis-Buitrago
- Neuroscience Research Group (NeURos), NeuroVitae Center for Neuroscience, School of Medicine and Health Sciences, Universidad del Rosario, Carrera 24 No. 63C-69, 111221, Bogotá D.C., Colombia
| | - Mariana Blanco
- Neuroscience Research Group (NeURos), NeuroVitae Center for Neuroscience, School of Medicine and Health Sciences, Universidad del Rosario, Carrera 24 No. 63C-69, 111221, Bogotá D.C., Colombia
| | - Alberto Velez-van-Meerbeke
- Neuroscience Research Group (NeURos), NeuroVitae Center for Neuroscience, School of Medicine and Health Sciences, Universidad del Rosario, Carrera 24 No. 63C-69, 111221, Bogotá D.C., Colombia.,NeuroVitae Center for Neuroscience, School of Medicine and Health Sciences, Universidad del Rosario, Carrera 24 No. 63C-69, 111221, Bogotá D.C., Colombia
| | - Claudia Talero-Gutiérrez
- Neuroscience Research Group (NeURos), NeuroVitae Center for Neuroscience, School of Medicine and Health Sciences, Universidad del Rosario, Carrera 24 No. 63C-69, 111221, Bogotá D.C., Colombia. .,NeuroVitae Center for Neuroscience, School of Medicine and Health Sciences, Universidad del Rosario, Carrera 24 No. 63C-69, 111221, Bogotá D.C., Colombia.
| |
Collapse
|
33
|
Hou SJ, Tsai SJ, Kuo PH, Lin WY, Liu YL, Yang AC, Lin E, Lan TH. An association study in the Taiwan Biobank elicits the GABAA receptor genes GABRB3, GABRA5, and GABRG3 as candidate loci for sleep duration in the Taiwanese population. BMC Med Genomics 2021; 14:223. [PMID: 34530807 PMCID: PMC8447520 DOI: 10.1186/s12920-021-01083-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 09/07/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Gamma-aminobutyric acid type A (GABAA) receptors mainly mediate the effects of gamma-aminobutyric acid, which is the primary inhibitory neurotransmitter in the central nervous system. Abundant evidence suggests that GABAA receptors play a key role in sleep-regulating processes. No genetic association study has explored the relationships between GABAA receptor genes and sleep duration, sleep quality, and sleep timing in humans. METHODS We determined the association between single-nucleotide polymorphisms (SNPs) in the GABAA receptor genes GABRA1, GABRA2, GABRB3, GABRA5, and GABRG3 and sleep duration, sleep quality, and sleep timing in the Taiwan Biobank with a sample of 10,127 Taiwanese subjects. There were 10,142 subjects in the original study cohort. We excluded 15 subjects with a medication history of sedative-hypnotics. RESULTS Our data revealed an association of the GABRB3-GABRA5-GABRG3 gene cluster with sleep duration, which has not been previously identified: rs79333046 (beta = - 0.07; P = 1.21 × 10-3) in GABRB3, rs189790076 (beta = 0.92; P = 1.04 × 10-3) in GABRA5, and rs147619342 (beta = - 0.72; P = 3.97 × 10-3) in GABRG3. The association between rs189790076 in GABRA5 and sleep duration remained significant after Bonferroni correction. A variant (rs12438141) in GABRB3 was also found to act as a potential expression quantitative trait locus. Additionally, we discovered interactions between variants in the GABRB3-GABRA5-GABRG3 gene cluster and lifestyle factors, such as tea and coffee consumption, smoking, and physical activity, that influenced sleep duration, although some interactions became nonsignificant after Bonferroni correction. We also found interactions among GABRB3, GABRA5, and GABRG3 that affected sleep duration. Furthermore, we identified an association of rs7165524 (beta = - 0.06; P = 2.20 × 10-3) in GABRA5 with sleep quality and an association of rs79465949 (beta = - 0.12; P = 3.95 × 10-3) in GABRB3 with sleep timing, although these associations became nonsignificant after Bonferroni correction. However, we detected no evidence of an association of individual SNPs in GABRA1 and GABRA2. CONCLUSIONS Our results indicate that rs189790076 in GABRA5 and gene-gene interactions among GABRB3, GABRA5, and GABRG3 may contribute to sleep duration in the Taiwanese population.
Collapse
Affiliation(s)
- Sheue-Jane Hou
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Department of Psychiatry, Cheng Hsin General Hospital, Taipei, Taiwan
| | - Shih-Jen Tsai
- Department of Psychiatry, Taipei Veterans General Hospital, Taipei, Taiwan
- Division of Psychiatry, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Po-Hsiu Kuo
- Department of Public Health, Institute of Epidemiology and Preventive Medicine, National Taiwan University, Taipei, Taiwan
- Department of Psychiatry, National Taiwan University Hospital, Taipei, Taiwan
| | - Wan-Yu Lin
- Department of Public Health, Institute of Epidemiology and Preventive Medicine, National Taiwan University, Taipei, Taiwan
| | - Yu-Li Liu
- Center for Neuropsychiatric Research, National Health Research Institutes, Miaoli County, Taiwan
| | - Albert C Yang
- Division of Interdisciplinary Medicine and Biotechnology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA
- Institute of Brain Science, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Eugene Lin
- Department of Biostatistics, University of Washington, 3980 15th Avenue NE, Box 351617, Seattle, WA, 98195, USA.
- Department of Electrical and Computer Engineering, University of Washington, Seattle, WA, 98195, USA.
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan.
| | - Tsuo-Hung Lan
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan.
- Tsaotun Psychiatric Center, Ministry of Health and Welfare, Nantou, Taiwan.
| |
Collapse
|
34
|
Belelli D, Hales TG, Lambert JJ, Luscher B, Olsen R, Peters JA, Rudolph U, Sieghart W. GABA A receptors in GtoPdb v.2021.3. IUPHAR/BPS GUIDE TO PHARMACOLOGY CITE 2021; 2021. [PMID: 35005623 DOI: 10.2218/gtopdb/f72/2021.3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The GABAA receptor is a ligand-gated ion channel of the Cys-loop family that includes the nicotinic acetylcholine, 5-HT3 and strychnine-sensitive glycine receptors. GABAA receptor-mediated inhibition within the CNS occurs by fast synaptic transmission, sustained tonic inhibition and temporally intermediate events that have been termed 'GABAA, slow' [45]. GABAA receptors exist as pentamers of 4TM subunits that form an intrinsic anion selective channel. Sequences of six α, three β, three γ, one δ, three ρ, one ε, one π and one θ GABAA receptor subunits have been reported in mammals [278, 235, 236, 283]. The π-subunit is restricted to reproductive tissue. Alternatively spliced versions of many subunits exist (e.g. α4- and α6- (both not functional) α5-, β2-, β3- and γ2), along with RNA editing of the α3 subunit [71]. The three ρ-subunits, (ρ1-3) function as either homo- or hetero-oligomeric assemblies [359, 50]. Receptors formed from ρ-subunits, because of their distinctive pharmacology that includes insensitivity to bicuculline, benzodiazepines and barbiturates, have sometimes been termed GABAC receptors [359], but they are classified as GABA A receptors by NC-IUPHAR on the basis of structural and functional criteria [16, 235, 236]. Many GABAA receptor subtypes contain α-, β- and γ-subunits with the likely stoichiometry 2α.2β.1γ [168, 235]. It is thought that the majority of GABAA receptors harbour a single type of α- and β - subunit variant. The α1β2γ2 hetero-oligomer constitutes the largest population of GABAA receptors in the CNS, followed by the α2β3γ2 and α3β3γ2 isoforms. Receptors that incorporate the α4- α5-or α 6-subunit, or the β1-, γ1-, γ3-, δ-, ε- and θ-subunits, are less numerous, but they may nonetheless serve important functions. For example, extrasynaptically located receptors that contain α6- and δ-subunits in cerebellar granule cells, or an α4- and δ-subunit in dentate gyrus granule cells and thalamic neurones, mediate a tonic current that is important for neuronal excitability in response to ambient concentrations of GABA [209, 272, 83, 19, 288]. GABA binding occurs at the β+/α- subunit interface and the homologous γ+/α- subunits interface creates the benzodiazepine site. A second site for benzodiazepine binding has recently been postulated to occur at the α+/β- interface ([254]; reviewed by [282]). The particular α-and γ-subunit isoforms exhibit marked effects on recognition and/or efficacy at the benzodiazepine site. Thus, receptors incorporating either α4- or α6-subunits are not recognised by 'classical' benzodiazepines, such as flunitrazepam (but see [356]). The trafficking, cell surface expression, internalisation and function of GABAA receptors and their subunits are discussed in detail in several recent reviews [52, 140, 188, 316] but one point worthy of note is that receptors incorporating the γ2 subunit (except when associated with α5) cluster at the postsynaptic membrane (but may distribute dynamically between synaptic and extrasynaptic locations), whereas as those incorporating the δ subunit appear to be exclusively extrasynaptic. NC-IUPHAR [16, 235, 3, 2] class the GABAA receptors according to their subunit structure, pharmacology and receptor function. Currently, eleven native GABAA receptors are classed as conclusively identified (i.e., α1β2γ2, α1βγ2, α3βγ2, α4βγ2, α4β2δ, α4β3δ, α5βγ2, α6βγ2, α6β2δ, α6β3δ and ρ) with further receptor isoforms occurring with high probability, or only tentatively [235, 236]. It is beyond the scope of this Guide to discuss the pharmacology of individual GABAA receptor isoforms in detail; such information can be gleaned in the reviews [16, 95, 168, 173, 143, 278, 216, 235, 236] and [9, 10]. Agents that discriminate between α-subunit isoforms are noted in the table and additional agents that demonstrate selectivity between receptor isoforms, for example via β-subunit selectivity, are indicated in the text below. The distinctive agonist and antagonist pharmacology of ρ receptors is summarised in the table and additional aspects are reviewed in [359, 50, 145, 223]. Several high-resolution cryo-electron microscopy structures have been described in which the full-length human α1β3γ2L GABAA receptor in lipid nanodiscs is bound to the channel-blocker picrotoxin, the competitive antagonist bicuculline, the agonist GABA (γ-aminobutyric acid), and the classical benzodiazepines alprazolam and diazepam [198].
Collapse
|
35
|
Ghit A, Assal D, Al-Shami AS, Hussein DEE. GABA A receptors: structure, function, pharmacology, and related disorders. J Genet Eng Biotechnol 2021; 19:123. [PMID: 34417930 PMCID: PMC8380214 DOI: 10.1186/s43141-021-00224-0] [Citation(s) in RCA: 182] [Impact Index Per Article: 45.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Accepted: 08/08/2021] [Indexed: 02/03/2023]
Abstract
Background γ-Aminobutyric acid sub-type A receptors (GABAARs) are the most prominent inhibitory neurotransmitter receptors in the CNS. They are a family of ligand-gated ion channel with significant physiological and therapeutic implications. Main body GABAARs are heteropentamers formed from a selection of 19 subunits: six α (alpha1-6), three β (beta1-3), three γ (gamma1-3), three ρ (rho1-3), and one each of the δ (delta), ε (epsilon), π (pi), and θ (theta) which result in the production of a considerable number of receptor isoforms. Each isoform exhibits distinct pharmacological and physiological properties. However, the majority of GABAARs are composed of two α subunits, two β subunits, and one γ subunit arranged as γ2β2α1β2α1 counterclockwise around the center. The mature receptor has a central chloride ion channel gated by GABA neurotransmitter and modulated by a variety of different drugs. Changes in GABA synthesis or release may have a significant effect on normal brain function. Furthermore, The molecular interactions and pharmacological effects caused by drugs are extremely complex. This is due to the structural heterogeneity of the receptors, and the existence of multiple allosteric binding sites as well as a wide range of ligands that can bind to them. Notably, dysfunction of the GABAergic system contributes to the development of several diseases. Therefore, understanding the relationship between GABAA receptor deficits and CNS disorders thus has a significant impact on the discovery of disease pathogenesis and drug development. Conclusion To date, few reviews have discussed GABAA receptors in detail. Accordingly, this review aims to summarize the current understanding of the structural, physiological, and pharmacological properties of GABAARs, as well as shedding light on the most common associated disorders.
Collapse
Affiliation(s)
- Amr Ghit
- Department of Biology and Biotechnology, University of Pavia, Pavia, Italy. .,Department of Biotechnology, Institute of Graduate Studies and Research (IGSR), Alexandria University, Alexandria, Egypt.
| | - Dina Assal
- Department of Biotechnology, American University in Cairo (AUC), Cairo, Egypt
| | - Ahmed S Al-Shami
- Department of Biotechnology, Institute of Graduate Studies and Research (IGSR), Alexandria University, Alexandria, Egypt.,Department of Zoology, Faculty of Science, Alexandria University, Alexandria, Egypt
| | - Diaa Eldin E Hussein
- Animal Health Research Institute (AHRI), Agricultural Research Center (ARC), Port of Alexandria, Alexandria, Egypt
| |
Collapse
|
36
|
Nelson AD, Bender KJ. Dendritic Integration Dysfunction in Neurodevelopmental Disorders. Dev Neurosci 2021; 43:201-221. [PMID: 34139699 PMCID: PMC8440332 DOI: 10.1159/000516657] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 04/13/2021] [Indexed: 11/19/2022] Open
Abstract
Neurodevelopmental disorders (NDDs) that affect cognition, social interaction, and learning, including autism spectrum disorder (ASD) and intellectual disability (ID), have a strong genetic component. Our current understanding of risk genes highlights two main groups of dysfunction: those in genes that act as chromatin modifiers and those in genes that encode for proteins localized at or near synapses. Understanding how dysfunction in these genes contributes to phenotypes observed in ASD and ID remains a major question in neuroscience. In this review, we highlight emerging evidence suggesting that dysfunction in dendrites - regions of neurons that receive synaptic input - may be key to understanding features of neuronal processing affected in these disorders. Dendritic integration plays a fundamental role in sensory processing, cognition, and conscious perception, processes hypothesized to be impaired in NDDs. Many high-confidence ASD genes function within dendrites where they control synaptic integration and dendritic excitability. Further, increasing evidence demonstrates that several ASD/ID genes, including chromatin modifiers and transcription factors, regulate the expression or scaffolding of dendritic ion channels, receptors, and synaptic proteins. Therefore, we discuss how dysfunction of subsets of NDD-associated genes in dendrites leads to defects in dendritic integration and excitability and may be one core phenotype in ASD and ID.
Collapse
Affiliation(s)
- Andrew D Nelson
- Department of Neurology, University of California, San Francisco, San Francisco, California, USA
| | - Kevin J Bender
- Department of Neurology, University of California, San Francisco, San Francisco, California, USA
| |
Collapse
|
37
|
Tkatchenko TV, Tkatchenko AV. Genome-wide analysis of retinal transcriptome reveals common genetic network underlying perception of contrast and optical defocus detection. BMC Med Genomics 2021; 14:153. [PMID: 34107987 PMCID: PMC8190860 DOI: 10.1186/s12920-021-01005-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 06/04/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Refractive eye development is regulated by optical defocus in a process of emmetropization. Excessive exposure to negative optical defocus often leads to the development of myopia. However, it is still largely unknown how optical defocus is detected by the retina. METHODS Here, we used genome-wide RNA-sequencing to conduct analysis of the retinal gene expression network underlying contrast perception and refractive eye development. RESULTS We report that the genetic network subserving contrast perception plays an important role in optical defocus detection and emmetropization. Our results demonstrate an interaction between contrast perception, the retinal circadian clock pathway and the signaling pathway underlying optical defocus detection. We also observe that the relative majority of genes causing human myopia are involved in the processing of optical defocus. CONCLUSIONS Together, our results support the hypothesis that optical defocus is perceived by the retina using contrast as a proxy and provide new insights into molecular signaling underlying refractive eye development.
Collapse
Affiliation(s)
| | - Andrei V. Tkatchenko
- Department of Ophthalmology, Columbia University, New York, NY USA
- Department of Pathology and Cell Biology, Columbia University, New York, NY USA
- Edward S. Harkness Eye Institute, Research Annex Room 415, 635 W. 165th Street, New York, NY 10032 USA
| |
Collapse
|
38
|
Park BY, Hong SJ, Valk SL, Paquola C, Benkarim O, Bethlehem RAI, Di Martino A, Milham MP, Gozzi A, Yeo BTT, Smallwood J, Bernhardt BC. Differences in subcortico-cortical interactions identified from connectome and microcircuit models in autism. Nat Commun 2021; 12:2225. [PMID: 33850128 PMCID: PMC8044226 DOI: 10.1038/s41467-021-21732-0] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Accepted: 02/05/2021] [Indexed: 01/14/2023] Open
Abstract
The pathophysiology of autism has been suggested to involve a combination of both macroscale connectome miswiring and microcircuit anomalies. Here, we combine connectome-wide manifold learning with biophysical simulation models to understand associations between global network perturbations and microcircuit dysfunctions in autism. We studied neuroimaging and phenotypic data in 47 individuals with autism and 37 typically developing controls obtained from the Autism Brain Imaging Data Exchange initiative. Our analysis establishes significant differences in structural connectome organization in individuals with autism relative to controls, with strong between-group effects in low-level somatosensory regions and moderate effects in high-level association cortices. Computational models reveal that the degree of macroscale anomalies is related to atypical increases of recurrent excitation/inhibition, as well as subcortical inputs into cortical microcircuits, especially in sensory and motor areas. Transcriptomic association analysis based on postmortem datasets identifies genes expressed in cortical and thalamic areas from childhood to young adulthood. Finally, supervised machine learning finds that the macroscale perturbations are associated with symptom severity scores on the Autism Diagnostic Observation Schedule. Together, our analyses suggest that atypical subcortico-cortical interactions are associated with both microcircuit and macroscale connectome differences in autism.
Collapse
Affiliation(s)
- Bo-Yong Park
- McConnell Brain Imaging Centre, Montreal Neurological Institute and Hospital, McGill University, Montreal, QC, Canada.
- Department of Data Science, Inha University, Incheon, South Korea.
| | - Seok-Jun Hong
- McConnell Brain Imaging Centre, Montreal Neurological Institute and Hospital, McGill University, Montreal, QC, Canada
- Center for the Developing Brain, Child Mind Institute, New York City, NY, USA
- Center for Neuroscience Imaging Research, Institute for Basic Science, Sungkyunkwan University, Suwon, South Korea
- Department of Biomedical Engineering, Sungkyunkwan University, Suwon, South Korea
| | - Sofie L Valk
- Forschungszentrum, Julich, Germany
- Max Planck Institute for Cognitive and Brain Sciences, Leipzig, Germany
| | - Casey Paquola
- McConnell Brain Imaging Centre, Montreal Neurological Institute and Hospital, McGill University, Montreal, QC, Canada
| | - Oualid Benkarim
- McConnell Brain Imaging Centre, Montreal Neurological Institute and Hospital, McGill University, Montreal, QC, Canada
| | - Richard A I Bethlehem
- Autism Research Centre, Department of Psychiatry, University of Cambridge, Cambridge, UK
- Brain Mapping Unit, Department of Psychiatry, University of Cambridge, Cambridge, UK
| | - Adriana Di Martino
- Center for the Developing Brain, Child Mind Institute, New York City, NY, USA
| | - Michael P Milham
- Center for the Developing Brain, Child Mind Institute, New York City, NY, USA
| | - Alessandro Gozzi
- Istituto Italiano di Tecnologia, Centre for Neuroscience and Cognitive Systems @ UNITN, Rovereto, Italy
| | - B T Thomas Yeo
- Department of Electrical and Computer Engineering, National University of Singapore, Singapore, Singapore
- Centre for Sleep and Cognition (CSC) & Centre for Translational Magnetic Resonance Research (TMR), National University of Singapore, Singapore, Singapore
- N.1 Institute for Health & Institute for Digital Medicine (WisDM), National University of Singapore, Singapore, Singapore
- Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, USA
- Integrative Sciences and Engineering Programme (ISEP), National University of Singapore, Singapore, Singapore
| | - Jonathan Smallwood
- Department of Psychology, York Neuroimaging Centre, University of York, York, UK
- Department of Psychology, Queen's University, Kingston, ON, Canada
| | - Boris C Bernhardt
- McConnell Brain Imaging Centre, Montreal Neurological Institute and Hospital, McGill University, Montreal, QC, Canada.
| |
Collapse
|
39
|
Yong Z, Dou Y, Gao Y, Xu X, Xiao Y, Zhu H, Li S, Yuan B. Prenatal, perinatal, and postnatal factors associated with autism spectrum disorder cases in Xuzhou, China. Transl Pediatr 2021; 10:635-646. [PMID: 33880333 PMCID: PMC8041613 DOI: 10.21037/tp-21-54] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND The aim of the present study was to explore the prenatal, perinatal, and postnatal risk factors in children with autism spectrum disorder (ASD) from Xuzhou, China by comparing them with healthy children. METHODS Children with ASD who received rehabilitation training at special education schools and rehabilitation institutions in Xuzhou were selected as the ASD group, and healthy children during the same period were selected as the healthy non-ASD group. A questionnaire based on the possible causes and susceptibility factors of ASD in children was issued and given to all children in this study. RESULTS The findings of the present study revealed a higher prevalence of prenatal, perinatal, and postnatal factors in children with ASD compared with healthy children. There were significantly more males than females in the ASD group, and the proportion of boys to girls was 5.81:1 (P<0.05). Logistic regression analysis suggested that the risk factors of male children developing ASD were feeding difficulties, poor living environment during pregnancy, maternal exposure to cigarette smoking during pregnancy, and perinatal hypoxia. Factors associated with ASD risk among were identified, such as living environment during pregnancy, delivery method, feeding difficulties, and epilepsy (P<0.05). Feeding difficulties and living in the countryside during pregnancy might be risk factors for ASD in girls according to the logistic regression analysis. CONCLUSIONS This survey confirmed the high prevalence of prenatal, perinatal, and postnatal factors in children with ASD. Some of these factors may be effective entry points for the prevention and treatment of ASD.
Collapse
Affiliation(s)
- Zenghua Yong
- Children's Neurological Rehabilitation Center, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Yunlong Dou
- Children's Neurological Rehabilitation Center, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Yuchen Gao
- Children's Neurological Rehabilitation Center, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Xuena Xu
- Children's Neurological Rehabilitation Center, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Yanli Xiao
- Children's Neurological Rehabilitation Center, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Hongjuan Zhu
- Children's Neurological Rehabilitation Center, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Shengli Li
- Department of Medical Records, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Baoqiang Yuan
- Children's Neurological Rehabilitation Center, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
40
|
Yang JQ, Yang CH, Yin BQ. Combined the GABA-A and GABA-B receptor agonists attenuates autistic behaviors in a prenatal valproic acid-induced mouse model of autism. Behav Brain Res 2021; 403:113094. [PMID: 33359845 DOI: 10.1016/j.bbr.2020.113094] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Revised: 12/08/2020] [Accepted: 12/19/2020] [Indexed: 01/18/2023]
Abstract
Autism spectrum disorder (ASD) is an immensely challenging developmental disorder characterized primarily by two core behavioral symptoms of social communication deficits and restricted/repetitive behaviors. Investigating the etiological process and identifying an appropriate therapeutic target remain as formidable challenges to overcome ASD due to numerous risk factors and complex symptoms associated with the disorder. Among the various mechanisms that contribute to ASD, the maintenance of excitation and inhibition balance emerged as a key factor to regulate proper functioning of neuronal circuitry. In this study, we employed prenatally exposed to valproic acid (VPA) to establish a validated ASD mouse model and found impaired inhibitory gamma-aminobutyric acid (GABAergic) neurotransmission through a presynaptic mechanism in these model mice, which was accompanied with decreased GABA release and GABA-A and GABA-B receptor subunits expression. And acute administration of individual GABA-A or GABA-B receptor agonists partially reversed autistic-like behaviors in the model mice. Furthermore, acute administration of the combined GABA-A and GABA-B receptor agonists palliated sociability deficits, anxiety and repetitive behaviors in the animal model of autistic-like behaviors, demonstrating the therapeutic potential of above cocktail in the treatment of ASD.
Collapse
Affiliation(s)
- Jian-Quan Yang
- Department of Children Rehabilitation, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Chao-Hua Yang
- Department of Children Rehabilitation, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Bao-Qi Yin
- Department of Children Rehabilitation, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
| |
Collapse
|
41
|
Adak P, Sinha S, Banerjee N. An Association Study of Gamma-Aminobutyric Acid Type A Receptor Variants and Susceptibility to Autism Spectrum Disorders. J Autism Dev Disord 2021; 51:4043-4053. [PMID: 33442857 DOI: 10.1007/s10803-020-04865-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/29/2020] [Indexed: 10/22/2022]
Abstract
In this pilot study, we aim to identify the role of few genetic variants of GABA-receptor type A subunits GABRB3 (rs4906902, rs7171660), GABRG3 (rs208129, rs140679), GABRA5 (rs 140681) in the aetiology of autism spectrum disorders in a population of West Bengal. 192 ASD probands, their parents and 184 ethnically-matched healthy controls were recruited for the study. The rs4906902G and the rs140679T conferred significant risk towards ASD. rs7171660 and rs140679 had transmission bias in the family. Neither alleles of rs 208129 and rs 140681 showed significant over-representation in either groups. All these variants were associated with at least one deficit in ASD-associated phenotypes like 'relating to people', 'Imitation', 'emotional response', 'body use', 'taste, smell, touch response' and 'activity levels'.
Collapse
Affiliation(s)
- Pallabi Adak
- Manovikas Biomedical Research and Diagnostic Centre, Manovikas Kendra, 482 Madudah, Plot I-24, Sector J, E.M. Bypass, Kolkata, West Bengal, 700107, India
| | - Swagata Sinha
- Out Patient Department, Manovikas Kendra, Kolkata, West Bengal, 700107, India
| | - Nilanjana Banerjee
- Manovikas Biomedical Research and Diagnostic Centre, Manovikas Kendra, 482 Madudah, Plot I-24, Sector J, E.M. Bypass, Kolkata, West Bengal, 700107, India.
| |
Collapse
|
42
|
Chang MY, Doppee D, Yu F, Perez C, Coleman AL, Pineles SL. Prevalence of Ophthalmologic Diagnoses in Children With Autism Spectrum Disorder Using the Optum Dataset: APopulation-Based Study. Am J Ophthalmol 2021; 221:147-153. [PMID: 32896499 DOI: 10.1016/j.ajo.2020.08.048] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 08/25/2020] [Accepted: 08/30/2020] [Indexed: 10/23/2022]
Abstract
PURPOSE Autism spectrum disorder (ASD) affects an estimated 1.85% of children in the United States and is increasing in prevalence. Any relationship between ophthalmologic disorders and ASD is poorly understood. The purpose of this study was to calculate the prevalence of ophthalmologic disorders in children with ASD. DESIGN Population-based retrospective cohort study. METHODS Setting: claims data from Optum Labs Data Warehouse, a longitudinal real-world data asset with de-identified administrative claims and electronic health records data. StudyPopulation: children ≤18 years of age at the time of first claim between 2007 and 2013. OBSERVATION diagnosis of pervasive developmental disorder (PDD) or autistic disorder (AD) according to International Classification of Disease-9th edition (ICD-9) codes. MainOutcomeMeasurements: prevalence of an ophthalmologic diagnosis (amblyopia, strabismus, optic neuropathy, nystagmus, or retinopathy of prematurity) by ICD-9 codes in typically developing (TD) controls and children with PDD and AD. Adjusted odds ratios (OR) for each diagnosis were calculated using multivariate logistic regression models. RESULTS Claims from more than 10 million children were included. The prevalence of any ophthalmologic diagnosis considered in this study was 3.5% in TD controls, 12.5% in children with PDD (adjusted OR, 3.22; 95% confidence interval [CI, ], 3.16-3.29; P < .001), and 13.5% in children with AD (adjusted OR, 3.23; 95% CI, 3.15-3.31; P < .001). CONCLUSIONS Population-based data suggested an increased risk of ophthalmologic diagnoses in children with ASD. Future research is necessary to further clarify the relationship between ophthalmologic disorders and autistic symptoms and severity.
Collapse
|
43
|
Koumangoye R, Bastarache L, Delpire E. NKCC1: Newly Found as a Human Disease-Causing Ion Transporter. FUNCTION 2020; 2:zqaa028. [PMID: 33345190 PMCID: PMC7727275 DOI: 10.1093/function/zqaa028] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 10/28/2020] [Accepted: 10/30/2020] [Indexed: 01/06/2023] Open
Abstract
Among the electroneutral Na+-dependent chloride transporters, NKCC1 had until now evaded identification as a protein causing human diseases. The closely related SLC12A transporters, NKCC2 and NCC have been identified some 25 years ago as responsible for Bartter and Gitelman syndromes: two renal-dependent salt wasting disorders. Absence of disease was most surprising since the NKCC1 knockout mouse was shown in 1999 to be viable, albeit with a wide range of deleterious phenotypes. Here we summarize the work of the past 5 years that introduced us to clinical cases involving NKCC1. The most striking cases are of 3 children with inherited mutations, who have complete absence of NKCC1 expression. These cases establish that lack of NKCC1 causes deafness; CFTR-like secretory defects with mucus accumulation in lung and intestine; severe xerostomia, hypotonia, dysmorphic facial features, and severe neurodevelopmental disorder. Another intriguing case is of a patient with a dominant deleterious SLC12A2 allele. This de novo mutation introduced a premature stop codon leading to a truncated protein. This mutant transporter seems to exert dominant-negative effect on wild-type transporter only in epithelial cells. The patient who suffers from lung, bladder, intestine, pancreas, and multiple endocrine abnormalities has, however, normal hearing and cognition. Finally, new reports substantiate the haploinsufficiency prediction of the SLC12A2 gene. Cases with single allele mutations in SLC12A2 have been linked to hearing loss and neurodevelopmental disorders.
Collapse
Affiliation(s)
- Rainelli Koumangoye
- Department of Anesthesiology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Lisa Bastarache
- Department of Biomedical Informatics, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Eric Delpire
- Department of Anesthesiology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA,Corresponding author. E-mail:
| |
Collapse
|
44
|
Heidari Nia M, Sargazi S, Saravani R, Mirinejad S, Jahantigh D, Shakiba M. Relationship between GABRB2 gene polymorphisms and schizophrenia susceptibility: a case-control study and in silico analyses. Int J Neurosci 2020; 132:633-642. [PMID: 32988247 DOI: 10.1080/00207454.2020.1830087] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
PURPOSE Converging evidence has recently established the significance of γ-aminobutyric acid neurotransmitter (GABA) system in the development of schizophrenia (SCZ). We aimed to determine the association of two markers of the GABAA receptor β2 subunit gene (GABRB2), rs12187676 G/C and rs1816072 T/C, with the risk of SCZ in Iranian population. MATERIALS AND METHODS In this case-control study, 190 patients with SCZ and 200 healthy controls were recruited from December 2018 to February 2020. Genotyping was done using the Tetra-ARMS-PCR technique. In silico analyses were performed to determine the potential effects of the variants. RESULTS The C allele and genotypes of codominant CC vs.TT and CT vs.TT, dominant TT vs. TC + CC, recessive TT + TC vs. CC of rs1816072 polymorphism, as well as codominant CC vs. GG and recessive GG + GC vs. CC genetic models of rs12187676 polymorphism were significantly associated with SCZ susceptibility. Compared to the TC/GC model, we have found that the TC/CC combination significantly increased the risk of SCZ by 4.32 fold while the TT/GG combination conferred a protective role against SCZ. Haplotypes analysis indicated that GABRB2 polymorphisms are in weak linkage disequilibrium with each other (LD = 0.1). However, bioinformatics analyses predicted that these polymorphisms do not have significant effects on the secondary structure and the splicing of GABRB2-mRNA. CONCLUSIONS We found that intronic GABRB2 polymorphisms were associated with SCZ risk in a sample of the Iranian population. These findings provided proof of concept for the involvement of the GABAergic neurotransmission system in SCZ development. These observations should be validated across other ethnicities and clinical subtypes.
Collapse
Affiliation(s)
- Milad Heidari Nia
- Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Saman Sargazi
- Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Ramin Saravani
- Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran.,Department of Clinical Biochemistry, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Shekoufeh Mirinejad
- Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Danial Jahantigh
- Department of Biology, Faculty of Science, University of Sistan and Baluchestan, Zahedan, Iran
| | - Mansoor Shakiba
- Department of Psychiatry, Zahedan University of Medical Sciences, Zahedan, Iran
| |
Collapse
|
45
|
Pandey S, Shukla T, Mishra A. The Spectrum of Repetitive Behaviors Associated With Subacute Sclerosing Panencephalitis. Mov Disord 2020; 36:497-503. [PMID: 32986918 DOI: 10.1002/mds.28323] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 07/30/2020] [Accepted: 09/03/2020] [Indexed: 11/06/2022] Open
Abstract
BACKGROUND Repetitive behaviors refer to a broad class of responses ranging from stereotypic body movements to impulsive/compulsive behaviors. They may be associated with neurological disorders. METHODS This is a case series of six subacute sclerosing panencephalitis (SSPE) patients who presented with a wide spectrum of repetitive motor behaviors and vocalizations. RESULTS Repetitive motor behaviors involved the upper limbs in all patients and lower limbs in 3 patients. The repetitive movements in the upper limbs were clapping, finger-clicking, hand rubbing, flailing, and dystonic posturing. In the lower limbs, the repetitive movements were rubbing with the heel, pelvic thrusting with flexion extension of the leg, and foot tapping. The spectrum of vocalizations included palilalia, whistling, grunting with spitting, and pathological crying. Repetitive behaviors were the presenting features in 2 patients. CONCLUSIONS This case series expands the spectrum of repetitive behaviors seen in neurological disorders associated with brain infections. © 2020 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Sanjay Pandey
- Department of Neurology, Govind Ballabh Pant Postgraduate Institute of Medical Education and Research, New Delhi, India
| | - Tanvi Shukla
- Department of Neurology, Govind Ballabh Pant Postgraduate Institute of Medical Education and Research, New Delhi, India
| | - Anumeha Mishra
- Department of Neurology, Govind Ballabh Pant Postgraduate Institute of Medical Education and Research, New Delhi, India
| |
Collapse
|
46
|
Donegan JJ, Lodge DJ. Stem Cells for Improving the Treatment of Neurodevelopmental Disorders. Stem Cells Dev 2020; 29:1118-1130. [PMID: 32008442 PMCID: PMC7469694 DOI: 10.1089/scd.2019.0265] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Accepted: 01/16/2020] [Indexed: 12/11/2022] Open
Abstract
Treatment options for neurodevelopmental disorders such as schizophrenia and autism are currently limited. Antipsychotics used to treat schizophrenia are not effective for all patients, do not target all symptoms of the disease, and have serious adverse side effects. There are currently no FDA-approved drugs to treat the core symptoms of autism. In an effort to develop new and more effective treatment strategies, stem cell technologies have been used to reprogram adult somatic cells into induced pluripotent stem cells, which can be differentiated into neuronal cells and even three-dimensional brain organoids. This new technology has the potential to elucidate the complex mechanisms that underlie neurodevelopmental disorders, offer more relevant platforms for drug discovery and personalized medicine, and may even be used to treat the disease.
Collapse
Affiliation(s)
- Jennifer J. Donegan
- Department of Pharmacology, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
- Center for Biomedical Neuroscience, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Daniel J. Lodge
- Department of Pharmacology, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
- Center for Biomedical Neuroscience, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| |
Collapse
|
47
|
Precenzano F, Parisi L, Lanzara V, Vetri L, Operto FF, Pastorino GMG, Ruberto M, Messina G, Risoleo MC, Santoro C, Bitetti I, Marotta R. Electroencephalographic Abnormalities in Autism Spectrum Disorder: Characteristics and Therapeutic Implications. ACTA ACUST UNITED AC 2020; 56:medicina56090419. [PMID: 32825169 PMCID: PMC7559692 DOI: 10.3390/medicina56090419] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 08/16/2020] [Accepted: 08/17/2020] [Indexed: 12/03/2022]
Abstract
A large body of literature reports the higher prevalence of epilepsy in subjects with Autism Spectrum Disorder (ASD) compared to the general population. Similarly, several studies report an increased rate of Subclinical Electroencephalographic Abnormalities (SEAs) in seizure-free patients with ASD rather than healthy controls, although with varying percentages. SEAs include both several epileptiform discharges and different non-epileptiform electroencephalographic abnormalities. They are more frequently associated with lower intellectual functioning, more serious dysfunctional behaviors, and they are often sign of severer forms of autism. However, SEAs clinical implications remain controversial, and they could represent an epiphenomenon of the neurochemical alterations of autism etiology. This paper provides an overview of the major research findings with two main purposes: to better delineate the state-of-the-art about EEG abnormalities in ASD and to find evidence for or against appropriateness of SEAs pharmacological treatment in ASD.
Collapse
Affiliation(s)
- Francesco Precenzano
- Epilepsy and EEG lab for Developmental Age; Clinic of Child and Adolescent Neuropsychiatry, Department of Mental Health, Physical and Preventive Medicine, University of Campania “Luigi Vanvitelli”, 80138 Napoli, Italy; (F.P.); (V.L.); (M.C.R.); (C.S.); (I.B.)
- Inter-University Group for Study and Research on Neurodevelopmental Disorders in Children and Adolescents; (L.P.); (G.M.G.P.)
| | - Lucia Parisi
- Inter-University Group for Study and Research on Neurodevelopmental Disorders in Children and Adolescents; (L.P.); (G.M.G.P.)
- Department of Psychology, Educational Science and Human Movement, University of Palermo, 90127 Palermo, Italy
| | - Valentina Lanzara
- Epilepsy and EEG lab for Developmental Age; Clinic of Child and Adolescent Neuropsychiatry, Department of Mental Health, Physical and Preventive Medicine, University of Campania “Luigi Vanvitelli”, 80138 Napoli, Italy; (F.P.); (V.L.); (M.C.R.); (C.S.); (I.B.)
- Inter-University Group for Study and Research on Neurodevelopmental Disorders in Children and Adolescents; (L.P.); (G.M.G.P.)
| | - Luigi Vetri
- Department of Sciences for Health Promotion and Mother and Child Care “G. D’Alessandro”, University of Palermo, 90127 Palermo, Italy
- Correspondence: or ; Tel.: +39-328-643-4126
| | - Francesca Felicia Operto
- Child and Adolescent Neuropsychiatry Unit, Department of Medicine, Surgery and Dentistry, University of Salerno, 84084 Fisciano, Italy;
| | - Grazia Maria Giovanna Pastorino
- Inter-University Group for Study and Research on Neurodevelopmental Disorders in Children and Adolescents; (L.P.); (G.M.G.P.)
- Child and Adolescent Neuropsychiatry Unit, Department of Medicine, Surgery and Dentistry, University of Salerno, 84084 Fisciano, Italy;
| | - Maria Ruberto
- Centro Pro Juventute Minerva SRL, 80131 Napoli, Italy;
| | - Giovanni Messina
- Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy;
| | - Maria Cristina Risoleo
- Epilepsy and EEG lab for Developmental Age; Clinic of Child and Adolescent Neuropsychiatry, Department of Mental Health, Physical and Preventive Medicine, University of Campania “Luigi Vanvitelli”, 80138 Napoli, Italy; (F.P.); (V.L.); (M.C.R.); (C.S.); (I.B.)
- Department of Medical and Surgical Science, University “Magna Graecia”, 88100 Catanzaro, Italy;
| | - Claudia Santoro
- Epilepsy and EEG lab for Developmental Age; Clinic of Child and Adolescent Neuropsychiatry, Department of Mental Health, Physical and Preventive Medicine, University of Campania “Luigi Vanvitelli”, 80138 Napoli, Italy; (F.P.); (V.L.); (M.C.R.); (C.S.); (I.B.)
| | - Ilaria Bitetti
- Epilepsy and EEG lab for Developmental Age; Clinic of Child and Adolescent Neuropsychiatry, Department of Mental Health, Physical and Preventive Medicine, University of Campania “Luigi Vanvitelli”, 80138 Napoli, Italy; (F.P.); (V.L.); (M.C.R.); (C.S.); (I.B.)
| | - Rosa Marotta
- Department of Medical and Surgical Science, University “Magna Graecia”, 88100 Catanzaro, Italy;
| |
Collapse
|
48
|
G2G: A web-server for the prediction of human synthetic lethal interactions. Comput Struct Biotechnol J 2020; 18:1028-1031. [PMID: 32419903 PMCID: PMC7215103 DOI: 10.1016/j.csbj.2020.04.012] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 04/18/2020] [Accepted: 04/19/2020] [Indexed: 12/04/2022] Open
Abstract
Genetic interactions (GIs) are fundamental to our understanding of biological processes in the cell. While GIs have been systematically mapped in yeast, there is scarce information about them in humans. Recently, we have suggested a state-of-the-art hierarchical method that leverages gene ontology information for predicting GIs in yeast. Here, we adapt this method and apply it for the first time to predict GIs in human. We introduce a web service called G2G for this task that is available at http://bnet.cs.tau.ac.il/g2g/.
Collapse
|
49
|
GABA ARα2 is Decreased in the Axon Initial Segment of Pyramidal Cells in Specific Areas of the Prefrontal Cortex in Autism. Neuroscience 2020; 437:76-86. [PMID: 32335215 DOI: 10.1016/j.neuroscience.2020.04.025] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 04/13/2020] [Accepted: 04/16/2020] [Indexed: 12/20/2022]
Abstract
Some forms of Autism Spectrum Disorder, a neurodevelopmental syndrome characterized by impaired communication and social skills as well as repetitive behaviors, are purportedly associated with dysregulation of the excitation/inhibition balance in the cerebral cortex. Through human postmortem tissue analysis, we previously found a significant decrease in the number of a gamma-aminobutyric acid (GABA)ergic interneuron subtype, the chandelier (Ch) cell, in the prefrontal cortex of subjects with autism. Ch cells exclusively target the axon initial segment (AIS) of excitatory pyramidal (Pyr) neurons, and a single Ch cell forms synapses on hundreds of Pyr cells, indicating a possible role in maintaining electrical balance. Thus, we herein investigated this crucial link between Ch and Pyr cells in the anatomy of autism neuropathology by examining GABA receptor protein expression in the Pyr cell AIS in subjects with autism. We collected tissue from the prefrontal cortex (Brodmann Areas (BA) 9, 46, and 47) of 20 subjects with autism and 20 age- and sex-matched control subjects. Immunohistochemical staining with antibodies against the GABAA receptor subunit α2 (GABAARα2) - the subunit most prevalent in the Pyr cell AIS - revealed a significantly decreased GABAARα2 protein in the Pyr cell AIS in supragranular layers of prefrontal cortical areas BA9 and BA47 in autism. Downregulated GABAARα2 protein in the Pyr cell AIS may result from decreased GABA synthesis in the prefrontal cortex of subjects with autism, and thereby contribute to an excitation/inhibition imbalance. Our findings support the potential for GABA receptor agonists asa therapeutic tool for autism.
Collapse
|
50
|
Kolodny T, Schallmo MP, Gerdts J, Edden RAE, Bernier RA, Murray SO. Concentrations of Cortical GABA and Glutamate in Young Adults With Autism Spectrum Disorder. Autism Res 2020; 13:1111-1129. [PMID: 32297709 DOI: 10.1002/aur.2300] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2020] [Revised: 03/02/2020] [Accepted: 03/18/2020] [Indexed: 12/19/2022]
Abstract
The balance of excitation and inhibition in neural circuits is hypothesized to be increased in autism spectrum disorder, possibly mediated by altered signaling of the inhibitory neurotransmitter γ-aminobutyric acid (GABA), yet empirical evidence in humans is inconsistent. We used edited magnetic resonance spectroscopy (MRS) to quantify signals associated with both GABA and the excitatory neurotransmitter glutamate in multiple regions of the sensory and sensorimotor cortex, including primary visual, auditory, and motor areas in adult individuals with autism and in neurotypical controls. Despite the strong a priori hypothesis of reduced GABA in autism spectrum disorder, we found no group differences in neurometabolite concentrations in any of the examined regions and no correlations of MRS measure with psychophysical visual sensitivity or autism symptomatology. We demonstrate high data quality that is comparable across groups, with a relatively large sample of well-characterized participants, and use Bayesian statistics to corroborate the lack of any group differences. We conclude that levels of GABA and Glx (glutamate, glutamine, and glutathione) in the sensory and sensorimotor cortex, as measured with MRS at 3T, are comparable in adults with autism and neurotypical individuals. Autism Res 2020, 13: 1111-1129. © 2020 International Society for Autism Research, Wiley Periodicals, Inc. LAY SUMMARY: γ-Aminobutyric acid (GABA) and glutamate are the main inhibitory and excitatory neurotransmitters in the human brain, respectively, and their balanced interaction is necessary for neural function. Previous research suggests that the GABA and glutamate systems might be altered in autism. In this study, we used magnetic resonance spectroscopy to measure concentrations of these neurotransmitters in the sensory areas in the brains of young adults with autism. In contradiction to the common hypothesis of reduced GABA in autism, we demonstrate that concentrations of both GABA and glutamate, in all the brain regions examined, are comparable in individuals with autism and in neurotypical adults. © 2020 International Society for Autism Research, Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Tamar Kolodny
- Department of Psychology, University of Washington, Seattle, Washington, USA
| | - Michael-Paul Schallmo
- Department of Psychology, University of Washington, Seattle, Washington, USA.,Department of Psychiatry and Behavioral Science, University of Minnesota, Minneapolis, Minnesota, USA
| | - Jennifer Gerdts
- Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, Washington, USA
| | - Richard A E Edden
- Department of Radiology and Radiological Sciences, Johns Hopkins University, Baltimore, Maryland, USA
| | - Raphael A Bernier
- Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, Washington, USA
| | - Scott O Murray
- Department of Psychology, University of Washington, Seattle, Washington, USA
| |
Collapse
|