101
|
Abstract
Epilepsy and autism frequently co-occur. Epilepsy confers an increased risk of autism and autism confers an increased risk of epilepsy. Specific epilepsy syndromes, intellectual disability, and female gender present a particular risk of autism in individuals with epilepsy. Epilepsy and autism are likely to share common etiologies, which predispose individuals to either or both conditions. Genetic factors, metabolic disorders, mitochondrial disorders, and immune dysfunction all can be implicated.
Collapse
Affiliation(s)
- Frank M C Besag
- East London NHS Foundation Trust, 5-7 Rush Court, Bedford MK40 3JT, UK; University College London, London, UK; King's College London, London, UK.
| | - Michael J Vasey
- East London NHS Foundation Trust, 5-7 Rush Court, Bedford MK40 3JT, UK
| |
Collapse
|
102
|
Supernumerary neurons within the cerebral cortical subplate in autism spectrum disorders. Brain Res 2021; 1760:147350. [PMID: 33607045 DOI: 10.1016/j.brainres.2021.147350] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 02/01/2021] [Accepted: 02/02/2021] [Indexed: 12/11/2022]
Abstract
Autism spectrum disorders (ASDs) involve alterations to cortical connectivity that manifest as reduced coordinated activity between cortical regions. The neurons of the cortical subplate are a major contributor to establishing thalamocortical, corticothalamic and corticocortical long-range connections and only a subset of this cell population survives into adulthood. Previous reports of an indistinct gray-white matter boundary in subjects with ASD suggest that the adjacent subplate may also show organizational abnormalities. Frozen human postmortem tissue samples from the parietal lobe (BA7) were used to evaluate white-matter neuron densities adjacent to layer VI with an antibody to NeuN. In addition, fixed postmortem tissue samples from frontal (BA9), parietal (BA7) and temporal lobe (BA21) locations, were stained with a Golgi-Kopsch procedure, and used to examine the morphology of these neuronal profiles. Relative to control cases, ASD subjects showed a large average density increase of NeuN-positive profiles of 44.7 percent. The morphologies of these neurons were consistent with subplate cells of the fusiform, polymorphic and pyramidal cell types. Lower ratios of fusiform to other cell types are found early in development and although adult ASD subjects showed consistently lower ratios, these differences were not significant. The increased number of retained subplate profiles, along with cell type ratios redolent of earlier developmental stages, suggests either an abnormal initial population or a partial failure of the apoptosis seen in neurotypical development. These results indicate abnormalities within a neuron population that plays multiple roles in the developing and mature cerebral cortex, including the establishment of long-range cortical connections.
Collapse
|
103
|
Prentzell MT, Rehbein U, Cadena Sandoval M, De Meulemeester AS, Baumeister R, Brohée L, Berdel B, Bockwoldt M, Carroll B, Chowdhury SR, von Deimling A, Demetriades C, Figlia G, de Araujo MEG, Heberle AM, Heiland I, Holzwarth B, Huber LA, Jaworski J, Kedra M, Kern K, Kopach A, Korolchuk VI, van 't Land-Kuper I, Macias M, Nellist M, Palm W, Pusch S, Ramos Pittol JM, Reil M, Reintjes A, Reuter F, Sampson JR, Scheldeman C, Siekierska A, Stefan E, Teleman AA, Thomas LE, Torres-Quesada O, Trump S, West HD, de Witte P, Woltering S, Yordanov TE, Zmorzynska J, Opitz CA, Thedieck K. G3BPs tether the TSC complex to lysosomes and suppress mTORC1 signaling. Cell 2021; 184:655-674.e27. [PMID: 33497611 PMCID: PMC7868890 DOI: 10.1016/j.cell.2020.12.024] [Citation(s) in RCA: 64] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 11/03/2020] [Accepted: 12/14/2020] [Indexed: 12/22/2022]
Abstract
Ras GTPase-activating protein-binding proteins 1 and 2 (G3BP1 and G3BP2, respectively) are widely recognized as core components of stress granules (SGs). We report that G3BPs reside at the cytoplasmic surface of lysosomes. They act in a non-redundant manner to anchor the tuberous sclerosis complex (TSC) protein complex to lysosomes and suppress activation of the metabolic master regulator mechanistic target of rapamycin complex 1 (mTORC1) by amino acids and insulin. Like the TSC complex, G3BP1 deficiency elicits phenotypes related to mTORC1 hyperactivity. In the context of tumors, low G3BP1 levels enhance mTORC1-driven breast cancer cell motility and correlate with adverse outcomes in patients. Furthermore, G3bp1 inhibition in zebrafish disturbs neuronal development and function, leading to white matter heterotopia and neuronal hyperactivity. Thus, G3BPs are not only core components of SGs but also a key element of lysosomal TSC-mTORC1 signaling.
Collapse
Affiliation(s)
- Mirja Tamara Prentzell
- Brain Cancer Metabolism Group, German Consortium of Translational Cancer Research (DKTK) & German Cancer Research Center (DKFZ), Heidelberg 69120, Germany; Department of Pediatrics, Section Systems Medicine of Metabolism and Signaling, University of Groningen, University Medical Center Groningen, Groningen 9700 RB, The Netherlands; Department of Bioinformatics and Molecular Genetics (Faculty of Biology), University of Freiburg, Freiburg 79104, Germany; Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, Freiburg 79104, Germany
| | - Ulrike Rehbein
- Department of Pediatrics, Section Systems Medicine of Metabolism and Signaling, University of Groningen, University Medical Center Groningen, Groningen 9700 RB, The Netherlands; Department for Neuroscience, School of Medicine and Health Sciences, Carl von Ossietzky University Oldenburg, Oldenburg 26129, Germany; Institute of Biochemistry and Center for Molecular Biosciences Innsbruck, University of Innsbruck, Innsbruck 6020, Austria
| | - Marti Cadena Sandoval
- Department of Pediatrics, Section Systems Medicine of Metabolism and Signaling, University of Groningen, University Medical Center Groningen, Groningen 9700 RB, The Netherlands; Institute of Biochemistry and Center for Molecular Biosciences Innsbruck, University of Innsbruck, Innsbruck 6020, Austria
| | - Ann-Sofie De Meulemeester
- Laboratory for Molecular Biodiscovery, Department of Pharmaceutical and Pharmacological Sciences, University of Leuven, Leuven BE-3000, Belgium
| | - Ralf Baumeister
- Department of Bioinformatics and Molecular Genetics (Faculty of Biology), University of Freiburg, Freiburg 79104, Germany; Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, Freiburg 79104, Germany; Signalling Research Centres BIOSS and CIBSS & ZBMZ Center for Biochemistry and Molecular Cell Research (Faculty of Medicine), University of Freiburg, Freiburg 79104, Germany
| | - Laura Brohée
- Cell Growth Control in Health and Age-Related Disease Group, Max Planck Institute for Biology of Ageing (MPI-AGE), Cologne 50931, Germany
| | - Bianca Berdel
- Brain Cancer Metabolism Group, German Consortium of Translational Cancer Research (DKTK) & German Cancer Research Center (DKFZ), Heidelberg 69120, Germany
| | - Mathias Bockwoldt
- Department of Arctic and Marine Biology, UiT The Arctic University of Norway, Tromsø 9037, Norway
| | - Bernadette Carroll
- School of Biochemistry, Biomedical Sciences Building, University Walk, Bristol BS8 1TD, UK
| | - Suvagata Roy Chowdhury
- Cell Signaling and Metabolism Group, German Cancer Research Center (DKFZ), Heidelberg 69120, Germany
| | - Andreas von Deimling
- German Consortium of Translational Cancer Research (DKTK), Clinical Cooperation Unit Neuropathology, German Cancer Research Center (DKFZ), Heidelberg 69120, Germany; Department of Neuropathology, Institute of Pathology, Heidelberg University, Heidelberg 69120, Germany
| | - Constantinos Demetriades
- Cell Growth Control in Health and Age-Related Disease Group, Max Planck Institute for Biology of Ageing (MPI-AGE), Cologne 50931, Germany; CECAD Cluster of Excellence, University of Cologne, Cologne 50931, Germany
| | - Gianluca Figlia
- Signal Transduction in Cancer and Metabolism, German Cancer Research Center (DKFZ), Heidelberg 69120, Germany; Heidelberg University, Heidelberg 69120, Germany
| | | | - Alexander M Heberle
- Department of Pediatrics, Section Systems Medicine of Metabolism and Signaling, University of Groningen, University Medical Center Groningen, Groningen 9700 RB, The Netherlands; Institute of Biochemistry and Center for Molecular Biosciences Innsbruck, University of Innsbruck, Innsbruck 6020, Austria
| | - Ines Heiland
- Department of Arctic and Marine Biology, UiT The Arctic University of Norway, Tromsø 9037, Norway
| | - Birgit Holzwarth
- Department of Bioinformatics and Molecular Genetics (Faculty of Biology), University of Freiburg, Freiburg 79104, Germany
| | - Lukas A Huber
- Institute of Cell Biology, Biocenter, Medical University of Innsbruck, Innsbruck 6020, Austria; Austrian Drug Screening Institute (ADSI), Innsbruck 6020, Austria
| | - Jacek Jaworski
- Laboratory of Molecular and Cellular Neurobiology, International Institute of Molecular and Cell Biology in Warsaw, Warsaw 02-109, Poland
| | - Magdalena Kedra
- Laboratory of Molecular and Cellular Neurobiology, International Institute of Molecular and Cell Biology in Warsaw, Warsaw 02-109, Poland
| | - Katharina Kern
- Brain Cancer Metabolism Group, German Consortium of Translational Cancer Research (DKTK) & German Cancer Research Center (DKFZ), Heidelberg 69120, Germany
| | - Andrii Kopach
- Laboratory of Molecular and Cellular Neurobiology, International Institute of Molecular and Cell Biology in Warsaw, Warsaw 02-109, Poland
| | - Viktor I Korolchuk
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Ineke van 't Land-Kuper
- Department of Pediatrics, Section Systems Medicine of Metabolism and Signaling, University of Groningen, University Medical Center Groningen, Groningen 9700 RB, The Netherlands; Department for Neuroscience, School of Medicine and Health Sciences, Carl von Ossietzky University Oldenburg, Oldenburg 26129, Germany
| | - Matylda Macias
- Laboratory of Molecular and Cellular Neurobiology, International Institute of Molecular and Cell Biology in Warsaw, Warsaw 02-109, Poland
| | - Mark Nellist
- Department of Clinical Genetics, Erasmus Medical Center, Rotterdam 3015 GD, The Netherlands
| | - Wilhelm Palm
- Cell Signaling and Metabolism Group, German Cancer Research Center (DKFZ), Heidelberg 69120, Germany
| | - Stefan Pusch
- German Consortium of Translational Cancer Research (DKTK), Clinical Cooperation Unit Neuropathology, German Cancer Research Center (DKFZ), Heidelberg 69120, Germany; Department of Neuropathology, Institute of Pathology, Heidelberg University, Heidelberg 69120, Germany
| | - Jose Miguel Ramos Pittol
- Institute of Biochemistry and Center for Molecular Biosciences Innsbruck, University of Innsbruck, Innsbruck 6020, Austria
| | - Michèle Reil
- Brain Cancer Metabolism Group, German Consortium of Translational Cancer Research (DKTK) & German Cancer Research Center (DKFZ), Heidelberg 69120, Germany
| | - Anja Reintjes
- Institute of Biochemistry and Center for Molecular Biosciences Innsbruck, University of Innsbruck, Innsbruck 6020, Austria
| | - Friederike Reuter
- Brain Cancer Metabolism Group, German Consortium of Translational Cancer Research (DKTK) & German Cancer Research Center (DKFZ), Heidelberg 69120, Germany
| | - Julian R Sampson
- Institute of Medical Genetics, Division of Cancer and Genetics, Cardiff University Medical School, Cardiff CF14 4AY, UK
| | - Chloë Scheldeman
- Laboratory for Molecular Biodiscovery, Department of Pharmaceutical and Pharmacological Sciences, University of Leuven, Leuven BE-3000, Belgium; Neurogenetics Research Group, VUB, Brussels 1090, Belgium
| | - Aleksandra Siekierska
- Laboratory for Molecular Biodiscovery, Department of Pharmaceutical and Pharmacological Sciences, University of Leuven, Leuven BE-3000, Belgium
| | - Eduard Stefan
- Institute of Biochemistry and Center for Molecular Biosciences Innsbruck, University of Innsbruck, Innsbruck 6020, Austria
| | - Aurelio A Teleman
- Signal Transduction in Cancer and Metabolism, German Cancer Research Center (DKFZ), Heidelberg 69120, Germany; Heidelberg University, Heidelberg 69120, Germany
| | - Laura E Thomas
- Institute of Life Science, Swansea University, Swansea SA2 8PP, UK
| | - Omar Torres-Quesada
- Institute of Biochemistry and Center for Molecular Biosciences Innsbruck, University of Innsbruck, Innsbruck 6020, Austria
| | - Saskia Trump
- Molecular Epidemiology Unit, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin 13353, Germany
| | - Hannah D West
- Institute of Medical Genetics, Division of Cancer and Genetics, Cardiff University Medical School, Cardiff CF14 4AY, UK
| | - Peter de Witte
- Laboratory for Molecular Biodiscovery, Department of Pharmaceutical and Pharmacological Sciences, University of Leuven, Leuven BE-3000, Belgium
| | - Sandra Woltering
- Brain Cancer Metabolism Group, German Consortium of Translational Cancer Research (DKTK) & German Cancer Research Center (DKFZ), Heidelberg 69120, Germany
| | - Teodor E Yordanov
- Institute of Cell Biology, Biocenter, Medical University of Innsbruck, Innsbruck 6020, Austria; Division of Cell and Developmental Biology, Institute for Molecular Bioscience, University of Queensland, St Lucia QLD 4072, Australia
| | - Justyna Zmorzynska
- Laboratory of Molecular and Cellular Neurobiology, International Institute of Molecular and Cell Biology in Warsaw, Warsaw 02-109, Poland
| | - Christiane A Opitz
- Brain Cancer Metabolism Group, German Consortium of Translational Cancer Research (DKTK) & German Cancer Research Center (DKFZ), Heidelberg 69120, Germany; Department of Neurology, University Hospital Heidelberg and National Center for Tumor Diseases, Heidelberg 69120, Germany.
| | - Kathrin Thedieck
- Department of Pediatrics, Section Systems Medicine of Metabolism and Signaling, University of Groningen, University Medical Center Groningen, Groningen 9700 RB, The Netherlands; Department for Neuroscience, School of Medicine and Health Sciences, Carl von Ossietzky University Oldenburg, Oldenburg 26129, Germany; Institute of Biochemistry and Center for Molecular Biosciences Innsbruck, University of Innsbruck, Innsbruck 6020, Austria.
| |
Collapse
|
104
|
Williams ZJ, He JL, Cascio CJ, Woynaroski TG. A review of decreased sound tolerance in autism: Definitions, phenomenology, and potential mechanisms. Neurosci Biobehav Rev 2021; 121:1-17. [PMID: 33285160 PMCID: PMC7855558 DOI: 10.1016/j.neubiorev.2020.11.030] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 11/11/2020] [Accepted: 11/12/2020] [Indexed: 12/23/2022]
Abstract
Atypical behavioral responses to environmental sounds are common in autistic children and adults, with 50-70 % of this population exhibiting decreased sound tolerance (DST) at some point in their lives. This symptom is a source of significant distress and impairment across the lifespan, contributing to anxiety, challenging behaviors, reduced community participation, and school/workplace difficulties. However, relatively little is known about its phenomenology or neurocognitive underpinnings. The present article synthesizes a large body of literature on the phenomenology and pathophysiology of DST-related conditions to generate a comprehensive theoretical account of DST in autism. Notably, we argue against conceptualizing DST as a unified construct, suggesting that it be separated into three phenomenologically distinct conditions: hyperacusis (the perception of everyday sounds as excessively loud or painful), misophonia (an acquired aversive reaction to specific sounds), and phonophobia (a specific phobia of sound), each responsible for a portion of observed DST behaviors. We further elaborate our framework by proposing preliminary neurocognitive models of hyperacusis, misophonia, and phonophobia that incorporate neurophysiologic findings from studies of autism.
Collapse
Affiliation(s)
- Zachary J Williams
- Medical Scientist Training Program, Vanderbilt University School of Medicine, 221 Eskind Biomedical Library and Learning Center, 2209 Garland Ave., Nashville, TN, 37240, United States; Department of Hearing and Speech Sciences, Vanderbilt University Medical Center, 1215 21st Avenue South, Medical Center East, Room 8310, Nashville, TN, 37232, United States; Vanderbilt Brain Institute, Vanderbilt University, 7203 Medical Research Building III, 465 21st Avenue South, Nashville, TN, 37232, United States; Frist Center for Autism and Innovation, Vanderbilt University, 2414 Highland Avenue, Suite 115, Nashville, TN, 37212, United States.
| | - Jason L He
- Department of Forensic and Neurodevelopmental Sciences, Sackler Institute for Translational Neurodevelopment, Institute of Psychiatry, Psychology and Neuroscience, King's College London, Strand Building, Strand Campus, Strand, London, WC2R 2LS, London, United Kingdom.
| | - Carissa J Cascio
- Vanderbilt Brain Institute, Vanderbilt University, 7203 Medical Research Building III, 465 21st Avenue South, Nashville, TN, 37232, United States; Frist Center for Autism and Innovation, Vanderbilt University, 2414 Highland Avenue, Suite 115, Nashville, TN, 37212, United States; Department of Psychiatry and Behavioral Sciences, Vanderbilt University Medical Center, 2254 Village at Vanderbilt, 1500 21st Ave South, Nashville, TN, 37212, United States; Vanderbilt Kennedy Center, Vanderbilt University Medical Center, 110 Magnolia Cir, Nashville, TN, 37203, United States.
| | - Tiffany G Woynaroski
- Department of Hearing and Speech Sciences, Vanderbilt University Medical Center, 1215 21st Avenue South, Medical Center East, Room 8310, Nashville, TN, 37232, United States; Vanderbilt Brain Institute, Vanderbilt University, 7203 Medical Research Building III, 465 21st Avenue South, Nashville, TN, 37232, United States; Frist Center for Autism and Innovation, Vanderbilt University, 2414 Highland Avenue, Suite 115, Nashville, TN, 37212, United States; Vanderbilt Kennedy Center, Vanderbilt University Medical Center, 110 Magnolia Cir, Nashville, TN, 37203, United States.
| |
Collapse
|
105
|
Norouzi Ofogh S, Rasoolijazi H, Shahsavand Ananloo E, Shahrivar Z, Joghataei MT, Sadeghi B, Bozorgmehr A, Alizadeh F. Alteration of TRIM33 Expression at Transcriptional and Translational Levels is Correlated with Autism Symptoms. J Mol Neurosci 2021; 71:1368-1377. [PMID: 33481220 DOI: 10.1007/s12031-020-01783-6] [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: 10/17/2020] [Accepted: 12/16/2020] [Indexed: 10/22/2022]
Abstract
As a complex neurodevelopmental disorder, autism affects children in three major cognitive domains including social interactions, language learning and repetitive stereotyped behaviors. Abnormal regulation of cell proliferation in the brain during the embryonic period via the TGF-β signaling pathway and TRIM33 gene that encodes a protein with a corepressor and regulatory role in this pathway has been considered as an etiology for autism. Here, we investigated the association of a variation of TRIM33 with autism symptoms at levels of mRNA and protein expression. We used Autism Diagnostic Interview-Revised (ADI-R) and Childhood Autism Rating Scale (CARS) as behavioral diagnostic tools. Normal and autistic children were genotyped for a TRIM33 polymorphism (rs11102807), and then expression was assessed at transcriptional and translational levels. Results demonstrated that the frequency of the homozygous A allele (AA genotype of rs11102807) was significantly higher in children with autism (P < 0.001), whereas carriers of the G allele were mostly among healthy individuals. Children homozygous for the rs11102807 A allele were associated with an increase in CARS and ADI-R scores, indicating a significant correlation with autism symptoms. TRIM33 gene expression at both mRNA (P < 0.01) and protein (P < 0.001) levels was significantly higher in controls compared to autistic children. A remarkable association between higher TRIM33 gene expression at the transcriptional level and lower scores for both CARS and ADI-R was observed in non-autistic children. It seems that rs11102807 modulates the function and expression of the TRIM33 gene, implying that the A allele may increase the risk of autism in children by reducing gene expression and altering the TGF-β signaling pathway.
Collapse
Affiliation(s)
- Sattar Norouzi Ofogh
- Department of Neuroscience, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Homa Rasoolijazi
- Department of Neuroscience, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences (IUMS), Tehran, Iran. .,Cellular and Molecular Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran. .,Department of Anatomy, School of Medicine, Iran University of Medical Sciences (IUMS), Tehran, Iran.
| | - Esmaeil Shahsavand Ananloo
- Department of Genomic Psychiatry and Behavioral Genomics (DGPBG), Roozbeh Hospital, School of Medicine, Tehran University of Medical Sciences (TUMS), Tehran, Iran. .,Department of Psychosomatic, Imam Khomeini Hospital Complex (IKHC), School of Medicine, Tehran University of Medical Sciences (TUMS), Tehran, Iran.
| | - Zahra Shahrivar
- Department of Psychiatry, Roozbeh Hospital, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Mohammad Taghi Joghataei
- Cellular and Molecular Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran.,Department of Anatomy, School of Medicine, Iran University of Medical Sciences (IUMS), Tehran, Iran.,Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Bahman Sadeghi
- Department of Biochemistry, Institute of Biochemistry and Biophysics (IBB), University of Tehran, Tehran, Iran
| | - Ali Bozorgmehr
- Iran Psychiatric Hospital, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Fatemeh Alizadeh
- Department of Genomic Psychiatry and Behavioral Genomics (DGPBG), Roozbeh Hospital, School of Medicine, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| |
Collapse
|
106
|
Peng J, Zhou Y, Wang K. Multiplex gene and phenotype network to characterize shared genetic pathways of epilepsy and autism. Sci Rep 2021; 11:952. [PMID: 33441621 PMCID: PMC7806931 DOI: 10.1038/s41598-020-78654-y] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 11/25/2020] [Indexed: 01/29/2023] Open
Abstract
It is well established that epilepsy and autism spectrum disorder (ASD) commonly co-occur; however, the underlying biological mechanisms of the co-occurence from their genetic susceptibility are not well understood. Our aim in this study is to characterize genetic modules of subgroups of epilepsy and autism genes that have similar phenotypic manifestations and biological functions. We first integrate a large number of expert-compiled and well-established epilepsy- and ASD-associated genes in a multiplex network, where one layer is connected through protein-protein interaction (PPI) and the other layer through gene-phenotype associations. We identify two modules in the multiplex network, which are significantly enriched in genes associated with both epilepsy and autism as well as genes highly expressed in brain tissues. We find that the first module, which represents the Gene Ontology category of ion transmembrane transport, is more epilepsy-focused, while the second module, representing synaptic signaling, is more ASD-focused. However, because of their enrichment in common genes and association with both epilepsy and ASD phenotypes, these modules point to genetic etiologies and biological processes shared between specific subtypes of epilepsy and ASD. Finally, we use our analysis to prioritize new candidate genes for epilepsy (i.e. ANK2, CACNA1E, CACNA2D3, GRIA2, DLG4) for further validation. The analytical approaches in our study can be applied to similar studies in the future to investigate the genetic connections between different human diseases.
Collapse
Affiliation(s)
- Jacqueline Peng
- grid.25879.310000 0004 1936 8972School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA 19104 USA ,grid.239552.a0000 0001 0680 8770Raymond G. Perelman Center for Cellular and Molecular Therapeutics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104 USA
| | - Yunyun Zhou
- grid.239552.a0000 0001 0680 8770Raymond G. Perelman Center for Cellular and Molecular Therapeutics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104 USA
| | - Kai Wang
- grid.239552.a0000 0001 0680 8770Raymond G. Perelman Center for Cellular and Molecular Therapeutics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104 USA ,grid.25879.310000 0004 1936 8972Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104 USA
| |
Collapse
|
107
|
Neuron-Glia Interactions in Tuberous Sclerosis Complex Affect the Synaptic Balance in 2D and Organoid Cultures. Cells 2021; 10:cells10010134. [PMID: 33445520 PMCID: PMC7826837 DOI: 10.3390/cells10010134] [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: 11/14/2020] [Revised: 01/05/2021] [Accepted: 01/08/2021] [Indexed: 02/06/2023] Open
Abstract
Tuberous sclerosis complex (TSC) is a genetic disease affecting the brain. Neurological symptoms like epilepsy and neurodevelopmental issues cause a significant burden on patients. Both neurons and glial cells are affected by TSC mutations. Previous studies have shown changes in the excitation/inhibition balance (E/I balance) in TSC. Astrocytes are known to be important for neuronal development, and astrocytic dysfunction can cause changes in the E/I balance. We hypothesized that astrocytes affect the synaptic balance in TSC. TSC patient-derived stem cells were differentiated into astrocytes, which showed increased proliferation compared to control astrocytes. RNA sequencing revealed changes in gene expression, which were related to epidermal growth factor (EGF) signaling and enriched for genes that coded for secreted or transmembrane proteins. Control neurons were cultured in astrocyte-conditioned medium (ACM) of TSC and control astrocytes. After culture in TSC ACM, neurons showed an altered synaptic balance, with an increase in the percentage of VGAT+ synapses. These findings were confirmed in organoids, presenting a spontaneous 3D organization of neurons and glial cells. To conclude, this study shows that TSC astrocytes are affected and secrete factors that alter the synaptic balance. As an altered E/I balance may underlie many of the neurological TSC symptoms, astrocytes may provide new therapeutic targets.
Collapse
|
108
|
Wang HT, Zhu ZA, Li YY, Lou SS, Yang G, Feng X, Xu W, Huang ZL, Cheng X, Xiong ZQ. CDKL5 deficiency in forebrain glutamatergic neurons results in recurrent spontaneous seizures. Epilepsia 2021; 62:517-528. [PMID: 33400301 DOI: 10.1111/epi.16805] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 12/13/2020] [Accepted: 12/13/2020] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Mutations of the cyclin-dependent kinase-like 5 (CDKL5) gene cause severe neurodevelopmental disorders characterized by intractable epilepsy, intellectual disability, and autism. Multiple mouse models generated for mechanistic studies have exhibited phenotypes similar to some human pathological features, but none of the models has developed one of the major symptoms affecting CDKL5 deficiency disorder (CDD) patients: intractable recurrent seizures. As disrupted neuronal excitation/inhibition balance is closely associated with the activity of glutamatergic and γ-aminobutyric acidergic (GABAergic) neurons, our aim was to study the effect of the loss of CDKL5 in different types of neurons on epilepsy. METHODS Using the Cre-LoxP system, we generated conditional knockout (cKO) mouse lines allowing CDKL5 deficiency in glutamatergic or GABAergic neurons. We employed noninvasive video recording and in vivo electrophysiological approaches to study seizure activity in these Cdkl5 cKO mice. Furthermore, we conducted Timm staining to confirm a morphological alteration, mossy fiber sprouting, which occurs with limbic epilepsy in both human and mouse brains. Finally, we performed whole-cell patch clamp in dentate granule cells to investigate cell-intrinsic properties and synaptic excitatory activity. RESULTS We demonstrate that Emx1- or CamK2α-derived Cdkl5 cKO mice manifest high-frequency spontaneous seizure activities recapitulating the epilepsy of CDD patients, which ultimately led to sudden death in mice. However, Cdkl5 deficiency in GABAergic neurons does not generate such seizures. The seizures were accompanied by typical epileptic features including higher amplitude spikes for epileptiform discharges and abnormal hippocampal mossy fiber sprouting. We also found an increase in spontaneous and miniature excitatory postsynaptic current frequencies but no change in amplitudes in the dentate granule cells of Emx1-cKO mice, indicating enhanced excitatory synaptic activity. SIGNIFICANCE Our study demonstrates that Cdkl5 cKO mice, serving as an animal model to study recurrent spontaneous seizures, have potential value for the pathological study of CDD-related seizures and for therapeutic innovation.
Collapse
Affiliation(s)
- Hong-Tao Wang
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China.,School of Life Science and Technology, Xinxiang Medical University, Xinxiang, China
| | - Zi-Ai Zhu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Yi-Yan Li
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Sen-Sen Lou
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Guang Yang
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Xue Feng
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China.,School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Wei Xu
- Department of Pharmacology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Zhi-Li Huang
- Department of Pharmacology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Xuewen Cheng
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Zhi-Qi Xiong
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China.,Shanghai Center for Brain Science and Brain-Inspired Intelligence Technology, Shanghai, China
| |
Collapse
|
109
|
Milovanovic M, Grujicic R. Electroencephalography in Assessment of Autism Spectrum Disorders: A Review. Front Psychiatry 2021; 12:686021. [PMID: 34658944 PMCID: PMC8511396 DOI: 10.3389/fpsyt.2021.686021] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 08/30/2021] [Indexed: 01/01/2023] Open
Abstract
Electroencephalography (EEG) can further out our understanding of autistic spectrum disorders (ASD) neurophysiology. Epilepsy and ASD comorbidity range between 5 and 46%, but its temporal relationship, causal mechanisms and interplay with intellectual disability are still unknown. Epileptiform discharges with or without seizures go as high as 60%, and associate with epileptic encephalopathies, conceptual term suggesting that epileptic activity can lead to cognitive and behavioral impairment beyond the underlying pathology. Seizures and ASD may be the result of similar mechanisms, such as abnormalities in GABAergic fibers or GABA receptor function. Epilepsy and ASD are caused by a number of genetic disorders and variations that induce such dysregulation. Similarly, initial epilepsy may influence synaptic plasticity and cortical connection, predisposing a growing brain to cognitive delays and behavioral abnormalities. The quantitative EEG techniques could be a useful tool in detecting and possibly measuring dysfunctions in specific brain regions and neuronal regulation in ASD. Power spectra analysis reveals a U-shaped pattern of power abnormalities, with excess power in the low and high frequency bands. These might be the consequence of a complicated network of neurochemical changes affecting the inhibitory GABAergic interneurons and their regulation of excitatory activity in pyramidal cells. EEG coherence studies of functional connectivity found general local over-connectivity and long-range under-connectivity between different brain areas. GABAergic interneuron growth and connections are presumably impaired in the prefrontal and temporal cortices in ASD, which is important for excitatory/inhibitory balance. Recent advances in quantitative EEG data analysis and well-known epilepsy ASD co-morbidity consistently indicate a role of aberrant GABAergic transmission that has consequences on neuronal organization and connectivity especially in the frontal cortex.
Collapse
Affiliation(s)
- Maja Milovanovic
- Department for Epilepsy and Clinical Neurophysiology, Institute of Mental Health, Belgrade, Serbia.,Faculty for Special Education and Rehabilitation, University of Belgrade, Belgrade, Serbia
| | - Roberto Grujicic
- Clinical Department for Children and Adolescents, Institute of Mental Health, Belgrade, Serbia
| |
Collapse
|
110
|
Gao J, Shen W. Xenopus in revealing developmental toxicity and modeling human diseases. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2021; 268:115809. [PMID: 33096388 DOI: 10.1016/j.envpol.2020.115809] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Revised: 10/01/2020] [Accepted: 10/09/2020] [Indexed: 06/11/2023]
Abstract
The Xenopus model offers many advantages for investigation of the molecular, cellular, and behavioral mechanisms underlying embryo development. Moreover, Xenopus oocytes and embryos have been extensively used to study developmental toxicity and human diseases in response to various environmental chemicals. This review first summarizes recent advances in using Xenopus as a vertebrate model to study distinct types of tissue/organ development following exposure to environmental toxicants, chemical reagents, and pharmaceutical drugs. Then, the successful use of Xenopus as a model for diseases, including fetal alcohol spectrum disorders, autism, epilepsy, and cardiovascular disease, is reviewed. The potential application of Xenopus in genetic and chemical screening to protect against embryo deficits induced by chemical toxicants and related diseases is also discussed.
Collapse
Affiliation(s)
- Juanmei Gao
- Zhejiang Key Laboratory of Organ Development and Regeneration, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China; College of Life and Sciences, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Wanhua Shen
- Zhejiang Key Laboratory of Organ Development and Regeneration, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China.
| |
Collapse
|
111
|
Althaus AL, Ackley MA, Belfort GM, Gee SM, Dai J, Nguyen DP, Kazdoba TM, Modgil A, Davies PA, Moss SJ, Salituro FG, Hoffmann E, Hammond RS, Robichaud AJ, Quirk MC, Doherty JJ. Preclinical characterization of zuranolone (SAGE-217), a selective neuroactive steroid GABA A receptor positive allosteric modulator. Neuropharmacology 2020; 181:108333. [PMID: 32976892 PMCID: PMC8265595 DOI: 10.1016/j.neuropharm.2020.108333] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 08/26/2020] [Accepted: 09/18/2020] [Indexed: 01/04/2023]
Abstract
Zuranolone (SAGE-217) is a novel, synthetic, clinical stage neuroactive steroid GABAA receptor positive allosteric modulator designed with the pharmacokinetic properties to support oral daily dosing. In vitro, zuranolone enhanced GABAA receptor current at nine unique human recombinant receptor subtypes, including representative receptors for both synaptic (γ subunit-containing) and extrasynaptic (δ subunit-containing) configurations. At a representative synaptic subunit configuration, α1β2γ2, zuranolone potentiated GABA currents synergistically with the benzodiazepine diazepam, consistent with the non-competitive activity and distinct binding sites of the two classes of compounds at synaptic receptors. In a brain slice preparation, zuranolone produced a sustained increase in GABA currents consistent with metabotropic trafficking of GABAA receptors to the cell surface. In vivo, zuranolone exhibited potent activity, indicating its ability to modulate GABAA receptors in the central nervous system after oral dosing by protecting against chemo-convulsant seizures in a mouse model and enhancing electroencephalogram β-frequency power in rats. Together, these data establish zuranolone as a potent and efficacious neuroactive steroid GABAA receptor positive allosteric modulator with drug-like properties and CNS exposure in preclinical models. Recent clinical data support the therapeutic promise of neuroactive steroid GABAA receptor positive modulators for treating mood disorders; brexanolone is the first therapeutic approved specifically for the treatment of postpartum depression. Zuranolone is currently under clinical investigation for the treatment of major depressive episodes in major depressive disorder, postpartum depression, and bipolar depression.
Collapse
Affiliation(s)
- Alison L Althaus
- Research and Nonclinical Development, Sage Therapeutics, Inc., Cambridge, MA, USA.
| | - Michael A Ackley
- Research and Nonclinical Development, Sage Therapeutics, Inc., Cambridge, MA, USA
| | - Gabriel M Belfort
- Research and Nonclinical Development, Sage Therapeutics, Inc., Cambridge, MA, USA
| | - Steven M Gee
- Research and Nonclinical Development, Sage Therapeutics, Inc., Cambridge, MA, USA
| | - Jing Dai
- Research and Nonclinical Development, Sage Therapeutics, Inc., Cambridge, MA, USA
| | - David P Nguyen
- Research and Nonclinical Development, Sage Therapeutics, Inc., Cambridge, MA, USA
| | - Tatiana M Kazdoba
- Research and Nonclinical Development, Sage Therapeutics, Inc., Cambridge, MA, USA
| | - Amit Modgil
- Department of Neuroscience, Tufts University, Boston, MA, USA
| | - Paul A Davies
- Department of Neuroscience, Tufts University, Boston, MA, USA
| | - Stephen J Moss
- Department of Neuroscience, Tufts University, Boston, MA, USA
| | - Francesco G Salituro
- Research and Nonclinical Development, Sage Therapeutics, Inc., Cambridge, MA, USA
| | - Ethan Hoffmann
- Research and Nonclinical Development, Sage Therapeutics, Inc., Cambridge, MA, USA
| | - Rebecca S Hammond
- Research and Nonclinical Development, Sage Therapeutics, Inc., Cambridge, MA, USA
| | - Albert J Robichaud
- Research and Nonclinical Development, Sage Therapeutics, Inc., Cambridge, MA, USA
| | - Michael C Quirk
- Research and Nonclinical Development, Sage Therapeutics, Inc., Cambridge, MA, USA
| | - James J Doherty
- Research and Nonclinical Development, Sage Therapeutics, Inc., Cambridge, MA, USA
| |
Collapse
|
112
|
An Unbiased Drug Screen for Seizure Suppressors in Duplication 15q Syndrome Reveals 5-HT 1A and Dopamine Pathway Activation as Potential Therapies. Biol Psychiatry 2020; 88:698-709. [PMID: 32507391 PMCID: PMC7554174 DOI: 10.1016/j.biopsych.2020.03.021] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 03/06/2020] [Accepted: 04/02/2020] [Indexed: 12/27/2022]
Abstract
BACKGROUND Duplication 15q (Dup15q) syndrome is a rare neurogenetic disorder characterized by autism and pharmacoresistant epilepsy. Most individuals with isodicentric duplications have been on multiple medications to control seizures. We recently developed a model of Dup15q in Drosophila by elevating levels of fly Dube3a in glial cells using repo-GAL4, not neurons. In contrast to other Dup15q models, these flies develop seizures that worsen with age. METHODS We screened repo>Dube3a flies for approved compounds that can suppress seizures. Flies 3 to 5 days old were exposed to compounds in the fly food during development. Flies were tested using a bang sensitivity assay for seizure recovery time. At least 40 animals were tested per experiment, with separate testing for male and female flies. Studies of K+ content in glial cells of the fly brain were also performed using a fluorescent K+ indicator. RESULTS We identified 17 of 1280 compounds in the Prestwick Chemical Library that could suppress seizures. Eight compounds were validated in secondary screening. Four of these compounds regulated either serotonergic or dopaminergic signaling, and subsequent experiments confirmed that seizure suppression occurred primarily through stimulation of serotonin receptor 5-HT1A. Additional studies of K+ levels showed that Dube3a regulation of the Na+/K+ exchanger ATPα (adenosine triphosphatase α) in glia may be modulated by serotonin/dopamine signaling, causing seizure suppression. CONCLUSIONS Based on these pharmacological and genetic studies, we present an argument for the use of 5-HT1A agonists in the treatment of Dup15q epilepsy.
Collapse
|
113
|
Jeong JH, Lee SH, Kho AR, Hong DK, Kang DH, Kang BS, Park MK, Choi BY, Choi HC, Lim MS, Suh SW. The Transient Receptor Potential Melastatin 7 (TRPM7) Inhibitors Suppress Seizure-Induced Neuron Death by Inhibiting Zinc Neurotoxicity. Int J Mol Sci 2020; 21:ijms21217897. [PMID: 33114331 PMCID: PMC7663745 DOI: 10.3390/ijms21217897] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 10/22/2020] [Accepted: 10/23/2020] [Indexed: 12/25/2022] Open
Abstract
Transient receptor potential melastatin 7 (TRPM7) is an ion channel that mediates monovalent cations out of cells, as well as the entry of divalent cations, such as zinc, magnesium, and calcium, into the cell. It has been reported that inhibitors of TRPM7 are neuroprotective in various neurological diseases. Previous studies in our lab suggested that seizure-induced neuronal death may be caused by the excessive release of vesicular zinc and the subsequent accumulation of zinc in the neurons. However, no studies have evaluated the effects of carvacrol and 2-aminoethoxydiphenyl borate (2-APB), both inhibitors of TRPM7, on the accumulation of intracellular zinc in dying neurons following seizure. Here, we investigated the therapeutic efficacy of carvacrol and 2-APB against pilocarpine-induced seizure. Carvacrol (50 mg/kg) was injected once per day for 3 or 7 days after seizure. 2-APB (2 mg/kg) was also injected once per day for 3 days after seizure. We found that inhibitors of TRPM7 reduced seizure-induced TRPM7 overexpression, intracellular zinc accumulation, and reactive oxygen species production. Moreover, there was a suppression of oxidative stress, glial activation, and the blood–brain barrier breakdown. In addition, inhibitors of TRPM7 remarkably decreased apoptotic neuron death following seizure. Taken together, the present study demonstrates that TRPM7-mediated zinc translocation is involved in neuron death after seizure. The present study suggests that inhibitors of TRPM7 may have high therapeutic potential to reduce seizure-induced neuron death.
Collapse
Affiliation(s)
- Jeong Hyun Jeong
- Department of Physiology, Hallym University, College of Medicine, Chuncheon 24252, Korea; (J.H.J.); (S.H.L.); (A.R.K.); (D.K.H.); (D.H.K.); (B.S.K.); (M.K.P.)
| | - Song Hee Lee
- Department of Physiology, Hallym University, College of Medicine, Chuncheon 24252, Korea; (J.H.J.); (S.H.L.); (A.R.K.); (D.K.H.); (D.H.K.); (B.S.K.); (M.K.P.)
| | - A Ra Kho
- Department of Physiology, Hallym University, College of Medicine, Chuncheon 24252, Korea; (J.H.J.); (S.H.L.); (A.R.K.); (D.K.H.); (D.H.K.); (B.S.K.); (M.K.P.)
| | - Dae Ki Hong
- Department of Physiology, Hallym University, College of Medicine, Chuncheon 24252, Korea; (J.H.J.); (S.H.L.); (A.R.K.); (D.K.H.); (D.H.K.); (B.S.K.); (M.K.P.)
| | - Dong Hyeon Kang
- Department of Physiology, Hallym University, College of Medicine, Chuncheon 24252, Korea; (J.H.J.); (S.H.L.); (A.R.K.); (D.K.H.); (D.H.K.); (B.S.K.); (M.K.P.)
| | - Beom Seok Kang
- Department of Physiology, Hallym University, College of Medicine, Chuncheon 24252, Korea; (J.H.J.); (S.H.L.); (A.R.K.); (D.K.H.); (D.H.K.); (B.S.K.); (M.K.P.)
| | - Min Kyu Park
- Department of Physiology, Hallym University, College of Medicine, Chuncheon 24252, Korea; (J.H.J.); (S.H.L.); (A.R.K.); (D.K.H.); (D.H.K.); (B.S.K.); (M.K.P.)
| | - Bo Young Choi
- Department of Physiology, Hallym University, College of Medicine, Chuncheon 24252, Korea; (J.H.J.); (S.H.L.); (A.R.K.); (D.K.H.); (D.H.K.); (B.S.K.); (M.K.P.)
- Correspondence: (B.Y.C.); (H.C.C.); (M.-S.L.); (S.W.S.); Tel.: +82-10-8573-6364 (S.W.S.)
| | - Hui Chul Choi
- Department of Neurology, Hallym University, College of Medicine, Chuncheon 24252, Korea
- Correspondence: (B.Y.C.); (H.C.C.); (M.-S.L.); (S.W.S.); Tel.: +82-10-8573-6364 (S.W.S.)
| | - Man-Sup Lim
- Department of Medical Education, Hallym University, College of Medicine, Chuncheon 24252, Korea
- Correspondence: (B.Y.C.); (H.C.C.); (M.-S.L.); (S.W.S.); Tel.: +82-10-8573-6364 (S.W.S.)
| | - Sang Won Suh
- Department of Physiology, Hallym University, College of Medicine, Chuncheon 24252, Korea; (J.H.J.); (S.H.L.); (A.R.K.); (D.K.H.); (D.H.K.); (B.S.K.); (M.K.P.)
- Correspondence: (B.Y.C.); (H.C.C.); (M.-S.L.); (S.W.S.); Tel.: +82-10-8573-6364 (S.W.S.)
| |
Collapse
|
114
|
SNX27-Mediated Recycling of Neuroligin-2 Regulates Inhibitory Signaling. Cell Rep 2020; 29:2599-2607.e6. [PMID: 31775031 PMCID: PMC6899438 DOI: 10.1016/j.celrep.2019.10.096] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 08/22/2019] [Accepted: 10/24/2019] [Indexed: 01/11/2023] Open
Abstract
GABAA receptors mediate fast inhibitory transmission in the brain, and their number can be rapidly up- or downregulated to alter synaptic strength. Neuroligin-2 plays a critical role in the stabilization of synaptic GABAA receptors and the development and maintenance of inhibitory synapses. To date, little is known about how the amount of neuroligin-2 at the synapse is regulated and whether neuroligin-2 trafficking affects inhibitory signaling. Here, we show that neuroligin-2, when internalized to endosomes, co-localizes with SNX27, a brain-enriched cargo-adaptor protein that facilitates membrane protein recycling. Direct interaction between the PDZ domain of SNX27 and PDZ-binding motif in neuroligin-2 enables membrane retrieval of neuroligin-2, thus enhancing synaptic neuroligin-2 clusters. Furthermore, SNX27 knockdown has the opposite effect. SNX27-mediated up- and downregulation of neuroligin-2 surface levels affects inhibitory synapse composition and signaling strength. Taken together, we show a role for SNX27-mediated recycling of neuroligin-2 in maintenance and signaling of the GABAergic synapse.
Collapse
|
115
|
Schizophrenic Psychosis Symptoms in a Background of Mild-To-Moderate Carnitine Palmitoyltransferase II Deficiency: A Case Report. REPORTS 2020. [DOI: 10.3390/reports3040031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Schizophrenia is a multifaceted mental illness characterized by cognitive and neurobehavioral abnormalities. Carnitine palmitoyltransferase II (CPT II) deficiency is a metabolic disorder resulting in impaired transport of long-chain fatty acids from the cytosol to the mitochondrial inner membrane, where fatty acid β-oxidation takes place. Here, we present an interesting clinical case of an adolescent male that presented with psychosis and a history of mild-to-moderate CPT II deficiency. To identify germline genetic variation that may contribute to the phenotypes observed, we performed whole-exome sequencing on DNA from the proband, unaffected fraternal twin, and biological parents. The proband was identified to be homozygous for the p.Val368Ile and heterozygous for the p.Met647Val variant in CPT2. Each of these variants are benign on their own; however, their combined effect is unclear. Further, variation was identified in the dopamine β-hydroxylase (DBH) gene (c.339+2T>C), which may contribute to decreased activity of DBH; however, based on the patient’s presentation, severe DBH deficiency is unlikely. In conclusion, the variants identified in this study do not clearly explain the observed patient phenotypes, indicating that the complex phenotypes are likely caused by an interplay of genetic and environmental factors that warrant further investigation.
Collapse
|
116
|
Affiliation(s)
- Yuri Bozzi
- Center for Mind/Brain Sciences (CIMeC), University of Trento, Italy; CNR Neuroscience Institute, Pisa, Italy
| | - Michela Fagiolini
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, United States
| |
Collapse
|
117
|
Neurobiological Mechanisms of Autism Spectrum Disorder and Epilepsy, Insights from Animal Models. Neuroscience 2020; 445:69-82. [DOI: 10.1016/j.neuroscience.2020.02.043] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 01/22/2020] [Accepted: 02/21/2020] [Indexed: 02/09/2023]
|
118
|
Trobiani L, Meringolo M, Diamanti T, Bourne Y, Marchot P, Martella G, Dini L, Pisani A, De Jaco A, Bonsi P. The neuroligins and the synaptic pathway in Autism Spectrum Disorder. Neurosci Biobehav Rev 2020; 119:37-51. [PMID: 32991906 DOI: 10.1016/j.neubiorev.2020.09.017] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 09/11/2020] [Accepted: 09/19/2020] [Indexed: 12/13/2022]
Abstract
The genetics underlying autism spectrum disorder (ASD) is complex and heterogeneous, and de novo variants are found in genes converging in functional biological processes. Neuronal communication, including trans-synaptic signaling involving two families of cell-adhesion proteins, the presynaptic neurexins and the postsynaptic neuroligins, is one of the most recurrently affected pathways in ASD. Given the role of these proteins in determining synaptic function, abnormal synaptic plasticity and failure to establish proper synaptic contacts might represent mechanisms underlying risk of ASD. More than 30 mutations have been found in the neuroligin genes. Most of the resulting residue substitutions map in the extracellular, cholinesterase-like domain of the protein, and impair protein folding and trafficking. Conversely, the stalk and intracellular domains are less affected. Accordingly, several genetic animal models of ASD have been generated, showing behavioral and synaptic alterations. The aim of this review is to discuss the current knowledge on ASD-linked mutations in the neuroligin proteins and their effect on synaptic function, in various brain areas and circuits.
Collapse
Affiliation(s)
- Laura Trobiani
- Dept. Biology and Biotechnology, Sapienza University of Rome, P.le Aldo Moro 5, 00185 Rome, Italy
| | - Maria Meringolo
- Lab. Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, Via del Fosso di Fiorano 64, 00143 Rome, Italy; Dept. Systems Medicine, University Tor Vergata, Via Montpellier 1, 00133 Rome, Italy
| | - Tamara Diamanti
- Dept. Biology and Biotechnology, Sapienza University of Rome, P.le Aldo Moro 5, 00185 Rome, Italy
| | - Yves Bourne
- Lab. "Architecture et Fonction des Macromolécules Biologiques", CNRS/Aix Marseille Univ, Faculté des Sciences - Campus Luminy, 163 Avenue de Luminy, 13288 Marseille cedex 09, France
| | - Pascale Marchot
- Lab. "Architecture et Fonction des Macromolécules Biologiques", CNRS/Aix Marseille Univ, Faculté des Sciences - Campus Luminy, 163 Avenue de Luminy, 13288 Marseille cedex 09, France
| | - Giuseppina Martella
- Lab. Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, Via del Fosso di Fiorano 64, 00143 Rome, Italy; Dept. Systems Medicine, University Tor Vergata, Via Montpellier 1, 00133 Rome, Italy
| | - Luciana Dini
- Dept. Biology and Biotechnology, Sapienza University of Rome, P.le Aldo Moro 5, 00185 Rome, Italy
| | - Antonio Pisani
- Lab. Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, Via del Fosso di Fiorano 64, 00143 Rome, Italy; Dept. Systems Medicine, University Tor Vergata, Via Montpellier 1, 00133 Rome, Italy
| | - Antonella De Jaco
- Dept. Biology and Biotechnology, Sapienza University of Rome, P.le Aldo Moro 5, 00185 Rome, Italy.
| | - Paola Bonsi
- Lab. Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, Via del Fosso di Fiorano 64, 00143 Rome, Italy.
| |
Collapse
|
119
|
Jęśko H, Cieślik M, Gromadzka G, Adamczyk A. Dysfunctional proteins in neuropsychiatric disorders: From neurodegeneration to autism spectrum disorders. Neurochem Int 2020; 141:104853. [PMID: 32980494 DOI: 10.1016/j.neuint.2020.104853] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 09/05/2020] [Accepted: 09/22/2020] [Indexed: 02/06/2023]
Abstract
Despite fundamental differences in disease course and outcomes, neurodevelopmental (autism spectrum disorders - ASD) and neurodegenerative disorders (Alzheimer's disease - AD and Parkinson's disease - PD) present surprising, common traits in their molecular pathomechanisms. Uncontrolled oligomerization and aggregation of amyloid β (Aβ), microtubule-associated protein (MAP) tau, or α-synuclein (α-syn) contribute to synaptic impairment and the ensuing neuronal death in both AD and PD. Likewise, the pathogenesis of ASD may be attributed, at least in part, to synaptic dysfunction; attention has also been recently paid to irregularities in the metabolism and function of the Aβ precursor protein (APP), tau, or α-syn. Commonly affected elements include signaling pathways that regulate cellular metabolism and survival such as insulin/insulin-like growth factor (IGF) - PI3 kinase - Akt - mammalian target of rapamycin (mTOR), and a number of key synaptic proteins critically involved in neuronal communication. Understanding how these shared pathomechanism elements operate in different conditions may help identify common targets and therapeutic approaches.
Collapse
Affiliation(s)
- Henryk Jęśko
- Department of Cellular Signalling, M. Mossakowski Medical Research Centre, Polish Academy of Sciences, 5 Pawińskiego Str., 02-106, Warsaw, Poland.
| | - Magdalena Cieślik
- Department of Cellular Signalling, M. Mossakowski Medical Research Centre, Polish Academy of Sciences, 5 Pawińskiego Str., 02-106, Warsaw, Poland.
| | - Grażyna Gromadzka
- Cardinal Stefan Wyszynski University, Faculty of Medicine. Collegium Medicum, Wóycickiego 1/3, 01-938, Warsaw, Poland.
| | - Agata Adamczyk
- Department of Cellular Signalling, M. Mossakowski Medical Research Centre, Polish Academy of Sciences, 5 Pawińskiego Str., 02-106, Warsaw, Poland.
| |
Collapse
|
120
|
Suzuki K, Elegheert J, Song I, Sasakura H, Senkov O, Matsuda K, Kakegawa W, Clayton AJ, Chang VT, Ferrer-Ferrer M, Miura E, Kaushik R, Ikeno M, Morioka Y, Takeuchi Y, Shimada T, Otsuka S, Stoyanov S, Watanabe M, Takeuchi K, Dityatev A, Aricescu AR, Yuzaki M. A synthetic synaptic organizer protein restores glutamatergic neuronal circuits. Science 2020; 369:369/6507/eabb4853. [PMID: 32855309 PMCID: PMC7116145 DOI: 10.1126/science.abb4853] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 07/24/2020] [Indexed: 12/18/2022]
Abstract
Neuronal synapses undergo structural and functional changes throughout life, which are essential for nervous system physiology. However, these changes may also perturb the excitatory-inhibitory neurotransmission balance and trigger neuropsychiatric and neurological disorders. Molecular tools to restore this balance are highly desirable. Here, we designed and characterized CPTX, a synthetic synaptic organizer combining structural elements from cerebellin-1 and neuronal pentraxin-1. CPTX can interact with presynaptic neurexins and postsynaptic AMPA-type ionotropic glutamate receptors and induced the formation of excitatory synapses both in vitro and in vivo. CPTX restored synaptic functions, motor coordination, spatial and contextual memories, and locomotion in mouse models for cerebellar ataxia, Alzheimer's disease, and spinal cord injury, respectively. Thus, CPTX represents a prototype for structure-guided biologics that can efficiently repair or remodel neuronal circuits.
Collapse
Affiliation(s)
- Kunimichi Suzuki
- Department of Physiology, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Jonathan Elegheert
- Division of Structural Biology, University of Oxford, Oxford OX3 7BN, UK
| | - Inseon Song
- Molecular Neuroplasticity, German Center for Neurodegenerative Diseases (DZNE), 39120 Magdeburg, Germany
| | - Hiroyuki Sasakura
- Department of Medical Cell Biology, School of Medicine, Aichi Medical University, Aichi, Japan
| | - Oleg Senkov
- Molecular Neuroplasticity, German Center for Neurodegenerative Diseases (DZNE), 39120 Magdeburg, Germany
| | - Keiko Matsuda
- Department of Physiology, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Wataru Kakegawa
- Department of Physiology, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Amber J Clayton
- Division of Structural Biology, University of Oxford, Oxford OX3 7BN, UK
| | - Veronica T Chang
- Division of Structural Biology, University of Oxford, Oxford OX3 7BN, UK
- Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge CB2 0QH, UK
| | - Maura Ferrer-Ferrer
- Molecular Neuroplasticity, German Center for Neurodegenerative Diseases (DZNE), 39120 Magdeburg, Germany
| | - Eriko Miura
- Department of Physiology, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Rahul Kaushik
- Molecular Neuroplasticity, German Center for Neurodegenerative Diseases (DZNE), 39120 Magdeburg, Germany
- Center for Behavioral Brain Sciences (CBBS), 39106 Magdeburg, Germany
| | - Masashi Ikeno
- Department of Medical Cell Biology, School of Medicine, Aichi Medical University, Aichi, Japan
| | - Yuki Morioka
- Department of Medical Cell Biology, School of Medicine, Aichi Medical University, Aichi, Japan
| | - Yuka Takeuchi
- Department of Medical Cell Biology, School of Medicine, Aichi Medical University, Aichi, Japan
| | - Tatsuya Shimada
- Department of Physiology, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Shintaro Otsuka
- Department of Physiology, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Stoyan Stoyanov
- Molecular Neuroplasticity, German Center for Neurodegenerative Diseases (DZNE), 39120 Magdeburg, Germany
| | - Masahiko Watanabe
- Department of Anatomy, Hokkaido University Graduate School of Medicine, Sapporo 060-8638, Japan
| | - Kosei Takeuchi
- Department of Medical Cell Biology, School of Medicine, Aichi Medical University, Aichi, Japan
| | - Alexander Dityatev
- Molecular Neuroplasticity, German Center for Neurodegenerative Diseases (DZNE), 39120 Magdeburg, Germany.
- Center for Behavioral Brain Sciences (CBBS), 39106 Magdeburg, Germany
- Medical Faculty, Otto von Guericke University, 39120 Magdeburg, Germany
| | - A Radu Aricescu
- Division of Structural Biology, University of Oxford, Oxford OX3 7BN, UK.
- Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge CB2 0QH, UK
| | - Michisuke Yuzaki
- Department of Physiology, Keio University School of Medicine, Tokyo 160-8582, Japan.
| |
Collapse
|
121
|
Cannabinoids for People with ASD: A Systematic Review of Published and Ongoing Studies. Brain Sci 2020; 10:brainsci10090572. [PMID: 32825313 PMCID: PMC7563787 DOI: 10.3390/brainsci10090572] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 08/15/2020] [Accepted: 08/17/2020] [Indexed: 12/22/2022] Open
Abstract
The etiopathogenesis of autism spectrum disorder (ASD) remains largely unclear. Among other biological hypotheses, researchers have evidenced an imbalance in the endocannabinoid (eCB) system, which regulates some functions typically impaired in ASD, such as emotional responses and social interaction. Additionally, cannabidiol (CBD), the non-intoxicating component of Cannabis sativa, was recently approved for treatment-resistant epilepsy. Epilepsy represents a common medical condition in people with ASD. Additionally, the two conditions share some neuropathological mechanisms, particularly GABAergic dysfunctions. Hence, it was hypothesized that cannabinoids could be useful in improving ASD symptoms. Our systematic review was conducted according to the PRISMA guidelines and aimed to summarize the literature regarding the use of cannabinoids in ASD. After searching in Web of KnowledgeTM, PsycINFO, and Embase, we included ten studies (eight papers and two abstracts). Four ongoing trials were retrieved in ClinicalTrials.gov. The findings were promising, as cannabinoids appeared to improve some ASD-associated symptoms, such as problem behaviors, sleep problems, and hyperactivity, with limited cardiac and metabolic side effects. Conversely, the knowledge of their effects on ASD core symptoms is scarce. Interestingly, cannabinoids generally allowed to reduce the number of prescribed medications and decreased the frequency of seizures in patients with comorbid epilepsy. Mechanisms of action could be linked to the excitatory/inhibitory imbalance found in people with ASD. However, further trials with better characterization and homogenization of samples, and well-defined outcomes should be implemented.
Collapse
|
122
|
Perets N, Oron O, Herman S, Elliott E, Offen D. Exosomes derived from mesenchymal stem cells improved core symptoms of genetically modified mouse model of autism Shank3B. Mol Autism 2020; 11:65. [PMID: 32807217 PMCID: PMC7433169 DOI: 10.1186/s13229-020-00366-x] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Accepted: 07/22/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Partial or an entire deletion of SHANK3 are considered as major drivers in the Phelan-McDermid syndrome, in which 75% of patients are diagnosed with autism spectrum disorder (ASD). During the recent years, there was an increasing interest in stem cell therapy in ASD, and specifically, mesenchymal stem cells (MSC). Moreover, it has been suggested that the therapeutic effect of the MSC is mediated mainly via the secretion of small extracellular vesicle that contains important molecular information of the cell and are used for cell-to-cell communication. Within the fraction of the extracellular vesicles, exosomes were highlighted as the most effective ones to convey the therapeutic effect. METHODS Exosomes derived from MSC (MSC-exo) were purified, characterized, and given via intranasal administration to Shank3B KO mice (in the concentration of 107 particles/ml). Three weeks post treatment, the mice were tested for behavioral scoring, and their results were compared with saline-treated control and their wild-type littermates. RESULTS Intranasal treatment with MSC-exo improves the social behavior deficit in multiple paradigms, increases vocalization, and reduces repetitive behaviors. We also observed an increase of GABARB1 in the prefrontal cortex. CONCLUSIONS Herein, we hypothesized that MSC-exo would have a direct beneficial effect on the behavioral autistic-like phenotype of the genetically modified Shank3B KO mouse model of autism. Taken together, our data indicate that intranasal treatment with MSC-exo improves the core ASD-like deficits of this mouse model of autism and therefore has the potential to treat ASD patients carrying the Shank3 mutation.
Collapse
Affiliation(s)
- N Perets
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel.
| | - O Oron
- Faculty of Medicine, Bar Ilan University, Tzfat, Israel
| | - S Herman
- Sacklar School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - E Elliott
- Faculty of Medicine, Bar Ilan University, Tzfat, Israel
| | - D Offen
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel.,Sacklar School of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
123
|
Trakoshis S, Martínez-Cañada P, Rocchi F, Canella C, You W, Chakrabarti B, Ruigrok ANV, Bullmore ET, Suckling J, Markicevic M, Zerbi V, Baron-Cohen S, Gozzi A, Lai MC, Panzeri S, Lombardo MV. Intrinsic excitation-inhibition imbalance affects medial prefrontal cortex differently in autistic men versus women. eLife 2020; 9:e55684. [PMID: 32746967 PMCID: PMC7402681 DOI: 10.7554/elife.55684] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Accepted: 06/29/2020] [Indexed: 12/22/2022] Open
Abstract
Excitation-inhibition (E:I) imbalance is theorized as an important pathophysiological mechanism in autism. Autism affects males more frequently than females and sex-related mechanisms (e.g., X-linked genes, androgen hormones) can influence E:I balance. This suggests that E:I imbalance may affect autism differently in males versus females. With a combination of in-silico modeling and in-vivo chemogenetic manipulations in mice, we first show that a time-series metric estimated from fMRI BOLD signal, the Hurst exponent (H), can be an index for underlying change in the synaptic E:I ratio. In autism we find that H is reduced, indicating increased excitation, in the medial prefrontal cortex (MPFC) of autistic males but not females. Increasingly intact MPFC H is also associated with heightened ability to behaviorally camouflage social-communicative difficulties, but only in autistic females. This work suggests that H in BOLD can index synaptic E:I ratio and that E:I imbalance affects autistic males and females differently.
Collapse
Affiliation(s)
- Stavros Trakoshis
- Laboratory for Autism and Neurodevelopmental Disorders, Center for Neuroscience and Cognitive Systems @UniTn, Istituto Italiano di TecnologiaRoveretoItaly
- Department of Psychology, University of CyprusNicosiaCyprus
| | - Pablo Martínez-Cañada
- Neural Computation Laboratory, Center for Neuroscience and Cognitive Systems @UniTn, Istituto Italiano di TecnologiaRoveretoItaly
- Optical Approaches to Brain Function Laboratory, Department of Neuroscience and Brain Technologies, Istituto Italiano di TecnologiaGenovaItaly
| | - Federico Rocchi
- Functional Neuroimaging Laboratory, Center for Neuroscience and Cognitive Systems @UniTn, Istituto Italiano di TecnologiaRoveretoItaly
| | - Carola Canella
- Functional Neuroimaging Laboratory, Center for Neuroscience and Cognitive Systems @UniTn, Istituto Italiano di TecnologiaRoveretoItaly
| | - Wonsang You
- Artificial Intelligence and Image Processing Laboratory, Department of Information and Communications Engineering, Sun Moon UniversityAsanRepublic of Korea
| | - Bhismadev Chakrabarti
- Autism Research Centre, Department of Psychiatry, University of CambridgeCambridgeUnited Kingdom
- Centre for Integrative Neuroscience and Neurodynamics, School of Psychology and Clinical Language Sciences, University of ReadingReadingUnited Kingdom
| | - Amber NV Ruigrok
- Autism Research Centre, Department of Psychiatry, University of CambridgeCambridgeUnited Kingdom
| | - Edward T Bullmore
- Cambridgeshire and Peterborough National Health Service Foundation TrustCambridgeUnited Kingdom
- Brain Mapping Unit, Department of Psychiatry, University of CambridgeCambridgeUnited Kingdom
| | - John Suckling
- Brain Mapping Unit, Department of Psychiatry, University of CambridgeCambridgeUnited Kingdom
| | - Marija Markicevic
- Neural Control of Movement Lab, D-HEST, ETH ZurichZurichSwitzerland
- Neuroscience Center Zurich, University and ETH ZurichZurichSwitzerland
| | - Valerio Zerbi
- Neural Control of Movement Lab, D-HEST, ETH ZurichZurichSwitzerland
- Neuroscience Center Zurich, University and ETH ZurichZurichSwitzerland
| | | | - Simon Baron-Cohen
- Autism Research Centre, Department of Psychiatry, University of CambridgeCambridgeUnited Kingdom
- Cambridgeshire and Peterborough National Health Service Foundation TrustCambridgeUnited Kingdom
| | - Alessandro Gozzi
- Functional Neuroimaging Laboratory, Center for Neuroscience and Cognitive Systems @UniTn, Istituto Italiano di TecnologiaRoveretoItaly
| | - Meng-Chuan Lai
- Autism Research Centre, Department of Psychiatry, University of CambridgeCambridgeUnited Kingdom
- The Margaret and Wallace McCain Centre for Child, Youth & Family Mental Health, Azrieli Adult Neurodevelopmental Centre, and Campbell Family Mental Health Research Institute, Centre for Addiction and Mental HealthTorontoCanada
- Department of Psychiatry and Autism Research Unit, The Hospital for Sick ChildrenTorontoCanada
- Department of Psychiatry, Faculty of Medicine, University of TorontoTorontoCanada
- Department of Psychiatry, National Taiwan University Hospital and College of MedicineTaipeiTaiwan
| | - Stefano Panzeri
- Neural Computation Laboratory, Center for Neuroscience and Cognitive Systems @UniTn, Istituto Italiano di TecnologiaRoveretoItaly
| | - Michael V Lombardo
- Laboratory for Autism and Neurodevelopmental Disorders, Center for Neuroscience and Cognitive Systems @UniTn, Istituto Italiano di TecnologiaRoveretoItaly
- Autism Research Centre, Department of Psychiatry, University of CambridgeCambridgeUnited Kingdom
| |
Collapse
|
124
|
Deng D, Masri S, Yao L, Ma X, Cao X, Yang S, Bao S, Zhou Q. Increasing endogenous activity of NMDARs on GABAergic neurons increases inhibition, alters sensory processing and prevents noise-induced tinnitus. Sci Rep 2020; 10:11969. [PMID: 32686710 PMCID: PMC7371882 DOI: 10.1038/s41598-020-68652-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2019] [Accepted: 06/15/2020] [Indexed: 01/04/2023] Open
Abstract
Selective enhancement of GABAergic inhibition is thought to impact many vital brain functions and interferes with the genesis and/or progression of numerous brain disorders. Here, we show that selectively increasing NMDA receptor activity in inhibitory neurons using an NMDAR positive allosteric modulator (PAM) elevates spiking activity of inhibitory neurons in vitro and in vivo. In vivo infusion of PAM increases spontaneous and sound-evoked spiking in inhibitory and decreases spiking in excitatory neurons, and increases signal-to-noise ratio in the primary auditory cortex. In addition, PAM infusion prior to noise trauma prevents the occurrence of tinnitus and reduction in GABAergic inhibition. These results reveal that selectively enhancing endogenous NMDAR activity on the GABAergic neurons can effectively enhance inhibitory activity and alter excitatory-inhibitory balance, and may be useful for preventing diseases that involve reduced inhibition as the major cause.
Collapse
Affiliation(s)
- Di Deng
- School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, China
| | - Samer Masri
- Department of Physiology, University of Arizona, Tucson, AZ, 85724, USA
| | - Lulu Yao
- School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, China
- South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xiaoyan Ma
- School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, China
| | - Xuebing Cao
- School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, China
| | - Sungchil Yang
- Department of Biomedical Sciences, City University of Hong Kong, Kowloon, Hong Kong
| | - Shaowen Bao
- Department of Physiology, University of Arizona, Tucson, AZ, 85724, USA
| | - Qiang Zhou
- School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, China.
- State Key Laboratory of Chemical Oncogenomics, Peking University Shenzhen Graduate School, Shenzhen, China.
| |
Collapse
|
125
|
Jaudon F, Thalhammer A, Cingolani LA. Integrin adhesion in brain assembly: From molecular structure to neuropsychiatric disorders. Eur J Neurosci 2020; 53:3831-3850. [PMID: 32531845 DOI: 10.1111/ejn.14859] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Revised: 05/21/2020] [Accepted: 06/02/2020] [Indexed: 02/07/2023]
Abstract
Integrins are extracellular matrix receptors that mediate biochemical and mechanical bi-directional signals between the extracellular and intracellular environment of a cell thanks to allosteric conformational changes. In the brain, they are found in both neurons and glial cells, where they play essential roles in several aspects of brain development and function, such as cell migration, axon guidance, synaptogenesis, synaptic plasticity and neuro-inflammation. Although there are many successful examples of how regulating integrin adhesion and signaling can be used for therapeutic purposes, for example for halting tumor progression, this is not the case for the brain, where the growing evidence of the importance of integrins for brain pathophysiology has not translated yet into medical applications. Here, we review recent literature showing how alterations in integrin structure, expression and signaling may be involved in the etiology of autism spectrum disorder, epilepsy, schizophrenia, addiction, depression and Alzheimer's disease. We focus on common mechanisms and recurrent signaling pathways, trying to bridge studies on the genetics and molecular structure of integrins with those on synaptic physiology and brain pathology. Further, we discuss integrin-targeting strategies and their potential benefits for therapeutic purposes in neuropsychiatric disorders.
Collapse
Affiliation(s)
- Fanny Jaudon
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia (IIT), Genoa, Italy.,IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Agnes Thalhammer
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia (IIT), Genoa, Italy.,IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Lorenzo A Cingolani
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia (IIT), Genoa, Italy.,Department of Life Sciences, University of Trieste, Trieste, Italy
| |
Collapse
|
126
|
Abstract
Epilepsy and autism frequently co-occur. Epilepsy confers an increased risk of autism and autism confers an increased risk of epilepsy. Specific epilepsy syndromes, intellectual disability, and female gender present a particular risk of autism in individuals with epilepsy. Epilepsy and autism are likely to share common etiologies, which predispose individuals to either or both conditions. Genetic factors, metabolic disorders, mitochondrial disorders, and immune dysfunction all can be implicated.
Collapse
Affiliation(s)
- Frank M C Besag
- East London NHS Foundation Trust, 5-7 Rush Court, Bedford MK40 3JT, UK; University College London, London, UK; King's College London, London, UK.
| | - Michael J Vasey
- East London NHS Foundation Trust, 5-7 Rush Court, Bedford MK40 3JT, UK
| |
Collapse
|
127
|
Pacheva I, Ivanov I. Targeted Biomedical Treatment for Autism Spectrum Disorders. Curr Pharm Des 2020; 25:4430-4453. [PMID: 31801452 DOI: 10.2174/1381612825666191205091312] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Accepted: 12/02/2019] [Indexed: 12/29/2022]
Abstract
BACKGROUND A diagnosis of autism spectrum disorders (ASD) represents presentations with impairment in communication and behaviour that vary considerably in their clinical manifestations and etiology as well as in their likely pathophysiology. A growing body of data indicates that the deleterious effect of oxidative stress, mitochondrial dysfunction, immune dysregulation and neuroinflammation, as well as their interconnections are important aspects of the pathophysiology of ASD. Glutathione deficiency decreases the mitochondrial protection against oxidants and tumor necrosis factor (TNF)-α; immune dysregulation and inflammation inhibit mitochondrial function through TNF-α; autoantibodies against the folate receptors underpin cerebral folate deficiency, resulting in disturbed methylation, and mitochondrial dysfunction. Such pathophysiological processes can arise from environmental and epigenetic factors as well as their combined interactions, such as environmental toxicant exposures in individuals with (epi)genetically impaired detoxification. The emerging evidence on biochemical alterations in ASD is forming the basis for treatments aimed to target its biological underpinnings, which is of some importance, given the uncertain and slow effects of the various educational interventions most commonly used. METHODS Literature-based review of the biomedical treatment options for ASD that are derived from established pathophysiological processes. RESULTS Most proposed biomedical treatments show significant clinical utility only in ASD subgroups, with specified pre-treatment biomarkers that are ameliorated by the specified treatment. For example, folinic acid supplementation has positive effects in ASD patients with identified folate receptor autoantibodies, whilst the clinical utility of methylcobalamine is apparent in ASD patients with impaired methylation capacity. Mitochondrial modulating cofactors should be considered when mitochondrial dysfunction is evident, although further research is required to identify the most appropriate single or combined treatment. Multivitamins/multiminerals formulas, as well as biotin, seem appropriate following the identification of metabolic abnormalities, with doses tapered to individual requirements. A promising area, requiring further investigations, is the utilization of antipurinergic therapies, such as low dose suramin. CONCLUSION The assessment and identification of relevant physiological alterations and targeted intervention are more likely to produce positive treatment outcomes. As such, current evidence indicates the utility of an approach based on personalized and evidence-based medicine, rather than treatment targeted to all that may not always be beneficial (primum non nocere).
Collapse
Affiliation(s)
- Iliyana Pacheva
- Department of Pediatrics and Medical Genetics, Medical University - Plovdiv, Plovdiv 4002, Bulgaria
| | - Ivan Ivanov
- Department of Pediatrics and Medical Genetics, Medical University - Plovdiv, Plovdiv 4002, Bulgaria
| |
Collapse
|
128
|
Carroll L, Braeutigam S, Dawes JM, Krsnik Z, Kostovic I, Coutinho E, Dewing JM, Horton CA, Gomez-Nicola D, Menassa DA. Autism Spectrum Disorders: Multiple Routes to, and Multiple Consequences of, Abnormal Synaptic Function and Connectivity. Neuroscientist 2020; 27:10-29. [PMID: 32441222 PMCID: PMC7804368 DOI: 10.1177/1073858420921378] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Autism spectrum disorders (ASDs) are a heterogeneous group of
neurodevelopmental disorders of genetic and environmental etiologies.
Some ASD cases are syndromic: associated with clinically defined
patterns of somatic abnormalities and a neurobehavioral phenotype
(e.g., Fragile X syndrome). Many cases, however, are idiopathic or
non-syndromic. Such disorders present themselves during the early
postnatal period when language, speech, and personality start to
develop. ASDs manifest by deficits in social communication and
interaction, restricted and repetitive patterns of behavior across
multiple contexts, sensory abnormalities across multiple modalities
and comorbidities, such as epilepsy among many others. ASDs are
disorders of connectivity, as synaptic dysfunction is common to both
syndromic and idiopathic forms. While multiple theories have been
proposed, particularly in idiopathic ASDs, none address why certain
brain areas (e.g., frontotemporal) appear more vulnerable than others
or identify factors that may affect phenotypic specificity. In this
hypothesis article, we identify possible routes leading to, and the
consequences of, altered connectivity and review the evidence of
central and peripheral synaptic dysfunction in ASDs. We postulate that
phenotypic specificity could arise from aberrant experience-dependent
plasticity mechanisms in frontal brain areas and peripheral sensory
networks and propose why the vulnerability of these areas could be
part of a model to unify preexisting pathophysiological theories.
Collapse
Affiliation(s)
- Liam Carroll
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, Oxfordshire, UK
| | - Sven Braeutigam
- Oxford Centre for Human Brain Activity, Wellcome Centre for Integrative Neuroimaging, Department of Psychiatry, University of Oxford, Oxford, Oxfordshire, UK
| | - John M Dawes
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, Oxfordshire, UK
| | - Zeljka Krsnik
- Croatian Institute for Brain Research, Centre of Research Excellence for Basic, Clinical and Translational Neuroscience, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Ivica Kostovic
- Croatian Institute for Brain Research, Centre of Research Excellence for Basic, Clinical and Translational Neuroscience, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Ester Coutinho
- Maurice Wohl Clinical Neuroscience Institute, King's College London, London, UK
| | - Jennifer M Dewing
- Faculty of Medicine, University of Southampton, Southampton, Hampshire, UK
| | - Christopher A Horton
- Sir William Dunn School of Pathology, University of Oxford, Oxford, Oxfordshire, UK
| | - Diego Gomez-Nicola
- Biological Sciences, Faculty of Environmental and Life Sciences, University of Southampton, Southampton, UK
| | - David A Menassa
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, Oxfordshire, UK.,Biological Sciences, Faculty of Environmental and Life Sciences, University of Southampton, Southampton, UK
| |
Collapse
|
129
|
Lee J, Ha S, Lee ST, Park SG, Shin S, Choi JR, Cheon KA. Next-Generation Sequencing in Korean Children With Autism Spectrum Disorder and Comorbid Epilepsy. Front Pharmacol 2020; 11:585. [PMID: 32477112 PMCID: PMC7240034 DOI: 10.3389/fphar.2020.00585] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Accepted: 04/16/2020] [Indexed: 12/23/2022] Open
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder characterized by impairments in social communication and restricted and repetitive behaviors and interests. Identifying the genetic background may be one of the key features for the future diagnosis and treatment of ASD. With the tremendous development in genetic diagnosis techniques, next-generation sequencing (NGS) can be used to analyze multiple genes simultaneously with a single test in laboratory and clinical settings and is well suited for investigating autism genetics. According to previous studies, there are two types of genetic variants in ASD, rare variants and common variants, and both are important in explaining pathogenesis. In this study, NGS data from 137 participants with ASD were reviewed retrospectively with consideration for comorbid epilepsy. Diagnostic yield was 17.51% (24/137), and pathogenic/likely pathogenic variants were seen more frequently in female participants. Fourteen participants were diagnosed with comorbid epilepsy, six of them had pathogenic/likely pathogenic variants (43%). Genes with variants of unknown significance (VOUS) which have one or more evidence of pathogenicity following the American College of Medical Genetics (ACMG) criteria were also reviewed in both ASD and ASD with comorbid epilepsy groups. We found that most frequently found VOUS genes have previously been reported as genes related to ASD or other developmental disorders. These results suggest that when interpreting the NGS results in the clinical setting, careful observation of VOUS with some pathological evidence might contribute to the discovery of genetic pathogenesis of neurodevelopmental disorders such as ASD and epilepsy.
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
| | - Seung-Tae Lee
- Department of Laboratory Medicine, Yonsei University College of Medicine, Seoul, South Korea
| | - Sung-Gyun Park
- Department of Laboratory Medicine, Yonsei University College of Medicine, Seoul, South Korea
| | - Saeam Shin
- 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
|
130
|
Culotta L, Penzes P. Exploring the mechanisms underlying excitation/inhibition imbalance in human iPSC-derived models of ASD. Mol Autism 2020; 11:32. [PMID: 32393347 PMCID: PMC7216514 DOI: 10.1186/s13229-020-00339-0] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Accepted: 04/21/2020] [Indexed: 12/14/2022] Open
Abstract
Autism spectrum disorder (ASD) is a range of neurodevelopmental disorders characterized by impaired social interaction and communication, and repetitive or restricted behaviors. ASD subjects exhibit complex genetic and clinical heterogeneity, thus hindering the discovery of pathophysiological mechanisms. Considering that several ASD-risk genes encode proteins involved in the regulation of synaptic plasticity, neuronal excitability, and neuronal connectivity, one hypothesis that has emerged is that ASD arises from a disruption of the neuronal network activity due to perturbation of the synaptic excitation and inhibition (E/I) balance. The development of induced pluripotent stem cell (iPSC) technology and recent advances in neuronal differentiation techniques provide a unique opportunity to model complex neuronal connectivity and to test the E/I hypothesis of ASD in human-based models. Here, we aim to review the latest advances in studying the different cellular and molecular mechanisms contributing to E/I balance using iPSC-based in vitro models of ASD.
Collapse
Affiliation(s)
- Lorenza Culotta
- Department of Physiology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.,Center for Autism and Neurodevelopment, Northwestern University, Chicago, IL, USA
| | - Peter Penzes
- Department of Physiology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA. .,Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL, USA. .,Center for Autism and Neurodevelopment, Northwestern University, Chicago, IL, USA.
| |
Collapse
|
131
|
Brunet A, Stuart-Lopez G, Burg T, Scekic-Zahirovic J, Rouaux C. Cortical Circuit Dysfunction as a Potential Driver of Amyotrophic Lateral Sclerosis. Front Neurosci 2020; 14:363. [PMID: 32410944 PMCID: PMC7201269 DOI: 10.3389/fnins.2020.00363] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Accepted: 03/25/2020] [Indexed: 12/11/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a devastating neurodegenerative disease that affects selected cortical and spinal neuronal populations, leading to progressive paralysis and death. A growing body of evidences suggests that the disease may originate in the cerebral cortex and propagate in a corticofugal manner. In particular, transcranial magnetic stimulation studies revealed that ALS patients present with early cortical hyperexcitability arising from a combination of increased excitability and decreased inhibition. Here, we discuss the possibility that initial cortical circuit dysfunction might act as the main driver of ALS onset and progression, and review recent functional, imaging and transcriptomic studies conducted on ALS patients, along with electrophysiological, pathological and transcriptomic studies on animal and cellular models of the disease, in order to evaluate the potential cellular and molecular origins of cortical hyperexcitability in ALS.
Collapse
Affiliation(s)
| | | | | | | | - Caroline Rouaux
- INSERM UMR_S 1118, Mécanismes Centraux et Périphériques de la Neurodégénérescence, Faculté de Médecine, Université de Strasbourg, Strasbourg, France
| |
Collapse
|
132
|
Altered Expression of GABAergic Markers in the Forebrain of Young and Adult Engrailed-2 Knockout Mice. Genes (Basel) 2020; 11:genes11040384. [PMID: 32244845 PMCID: PMC7231099 DOI: 10.3390/genes11040384] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Revised: 03/25/2020] [Accepted: 03/30/2020] [Indexed: 12/23/2022] Open
Abstract
Impaired function of GABAergic interneurons, and the subsequent alteration of excitation/inhibition balance, is thought to contribute to autism spectrum disorders (ASD). Altered numbers of GABAergic interneurons and reduced expression of GABA receptors has been detected in the brain of ASD subjects and mouse models of ASD. We previously showed a reduced expression of GABAergic interneuron markers parvalbumin (PV) and somatostatin (SST) in the forebrain of adult mice lacking the Engrailed2 gene (En2-/- mice). Here, we extended this analysis to postnatal day (P) 30 by using in situ hybridization, immunohistochemistry, and quantitative RT-PCR to study the expression of GABAergic interneuron markers in the hippocampus and somatosensory cortex of En2-/- and wild type (WT) mice. In addition, GABA receptor subunit mRNA expression was investigated by quantitative RT-PCR in the same brain regions of P30 and adult En2-/- and WT mice. As observed in adult animals, PV and SST expression was decreased in En2-/- forebrain of P30 mice. The expression of GABA receptor subunits (including the ASD-relevant Gabrb3) was also altered in young and adult En2-/- forebrain. Our results suggest that GABAergic neurotransmission deficits are already evident at P30, confirming that neurodevelopmental defects of GABAergic interneurons occur in the En2 mouse model of ASD.
Collapse
|
133
|
De Fusco A, Cerullo MS, Marte A, Michetti C, Romei A, Castroflorio E, Baulac S, Benfenati F. Acute knockdown of Depdc5 leads to synaptic defects in mTOR-related epileptogenesis. Neurobiol Dis 2020; 139:104822. [PMID: 32113911 DOI: 10.1016/j.nbd.2020.104822] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 02/02/2020] [Accepted: 02/26/2020] [Indexed: 12/17/2022] Open
Abstract
DEP-domain containing 5 (DEPDC5) is part of the GATOR1 complex that functions as key inhibitor of the mechanistic target of rapamycin complex 1 (mTORC1). Loss-of-function mutations in DEPDC5 leading to mTOR hyperactivation have been identified as the most common cause of either lesional or non-lesional focal epilepsy. However, the precise mechanisms by which DEPDC5 loss-of-function triggers neuronal and network hyperexcitability are still unclear. In this study, we investigated the cellular mechanisms of hyperexcitability by comparing the constitutive heterozygous Depdc5 knockout mouse versus different levels of acute Depdc5 deletion (≈40% and ≈80% neuronal knockdown of Depdc5 protein) by RNA interference in primary cortical cultures. While heterozygous Depdc5+/- neurons have only a subtle phenotype, acutely knocked-down neurons exhibit a strong dose-dependent phenotype characterized by mTOR hyperactivation, increased soma size, dendritic arborization, excitatory synaptic transmission and intrinsic excitability. The robust synaptic phenotype resulting from the acute knockdown Depdc5 deficiency highlights the importance of the temporal dynamics of Depdc5 knockdown in triggering the phenotypic changes, reminiscent of the somatic second-hit mechanism in patients with focal cortical dysplasia. These findings uncover a novel synaptic phenotype that is causally linked to Depdc5 knockdown, highlighting the developmental role of Depdc5. Interestingly, the synaptic defect appears to affect only excitatory synapses, while inhibitory synapses develop normally. The increased frequency and amplitude of mEPSCs, paralleled by increased density of excitatory synapses and expression of glutamate receptors, may generate an excitation/inhibition imbalance that triggers epileptogenesis.
Collapse
Affiliation(s)
- Antonio De Fusco
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Largo Rosanna Benzi 10, 16132 Genova, Italy; Department of Experimental Medicine, University of Genova, Viale Benedetto XV, 3, 16132 Genova, Italy
| | - Maria Sabina Cerullo
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Largo Rosanna Benzi 10, 16132 Genova, Italy; Department of Experimental Medicine, University of Genova, Viale Benedetto XV, 3, 16132 Genova, Italy
| | - Antonella Marte
- Department of Experimental Medicine, University of Genova, Viale Benedetto XV, 3, 16132 Genova, Italy; IRCSS, Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genova, Italy
| | - Caterina Michetti
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Largo Rosanna Benzi 10, 16132 Genova, Italy; IRCSS, Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genova, Italy
| | - Alessandra Romei
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Largo Rosanna Benzi 10, 16132 Genova, Italy; Department of Experimental Medicine, University of Genova, Viale Benedetto XV, 3, 16132 Genova, Italy
| | - Enrico Castroflorio
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Largo Rosanna Benzi 10, 16132 Genova, Italy
| | - Stephanie Baulac
- Sorbonne Université, UPMC Univ Paris 06, UMR S 1127, INSERM, U1127, CNRS, UMR 7225, Institut du Cerveau et de la Moelle épinière (ICM), Hôpital Pitié-Salpêtrière, F-75013 Paris, France
| | - Fabio Benfenati
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Largo Rosanna Benzi 10, 16132 Genova, Italy; IRCSS, Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genova, Italy.
| |
Collapse
|
134
|
Cukier HN, Griswold AJ, Hofmann NK, Gomez L, Whitehead PL, Abramson RK, Gilbert JR, Cuccaro ML, Dykxhoorn DM, Pericak-Vance MA. Three Brothers With Autism Carry a Stop-Gain Mutation in the HPA-Axis Gene NR3C2. Autism Res 2020; 13:523-531. [PMID: 32064789 DOI: 10.1002/aur.2269] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 11/20/2019] [Accepted: 01/06/2020] [Indexed: 12/30/2022]
Abstract
Whole exome sequencing and copy-number variant analysis was performed on a family with three brothers diagnosed with autism. Each of the siblings shares an alteration in the nuclear receptor subfamily 3 group C member 2 (NR3C2) gene that is predicted to result in a stop-gain mutation (p.Q919X) in the mineralocorticoid receptor (MR) protein. This variant was maternally inherited and provides further evidence for a connection between the NR3C2 and autism. Interestingly, the NR3C2 gene encodes the MR protein, a steroid hormone-regulated transcription factor that acts in the hypothalamic-pituitary-adrenal axis and has been connected to stress and anxiety, both of which are features often seen in individuals with autism. Autism Res 2020, 13: 523-531. © 2020 International Society for Autism Research, Wiley Periodicals, Inc. LAY SUMMARY: Given the complexity of the genetics underlying autism, each gene contributes to risk in a relatively small number of individuals, typically less than 1% of all autism cases. Whole exome sequencing of three brothers with autism identified a rare variant in the nuclear receptor subfamily 3 group C member 2 gene that is predicted to strongly interfere with its normal function. This gene encodes the mineralocorticoid receptor protein, which plays a role in how the body responds to stress and anxiety, features that are often elevated in people diagnosed with autism. This study adds further support to the relevance of this gene as a risk factor for autism.
Collapse
Affiliation(s)
- Holly N Cukier
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, Florida.,Department of Neurology, University of Miami Miller School of Medicine, Miami, Florida.,Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Miami, Florida
| | - Anthony J Griswold
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, Florida.,Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Miami, Florida
| | - Natalia K Hofmann
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, Florida
| | - Lissette Gomez
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, Florida
| | - Patrice L Whitehead
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, Florida
| | - Ruth K Abramson
- University of South Carolina School of Medicine, Columbia, South Carolina
| | - John R Gilbert
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, Florida.,Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Miami, Florida
| | - Michael L Cuccaro
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, Florida.,Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Miami, Florida
| | - Derek M Dykxhoorn
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, Florida.,Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Miami, Florida
| | - Margaret A Pericak-Vance
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, Florida.,Department of Neurology, University of Miami Miller School of Medicine, Miami, Florida.,Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Miami, Florida
| |
Collapse
|
135
|
Jaureguiberry MS, Venturino A. Nutritional and environmental contributions to Autism Spectrum Disorders: Focus on nutrigenomics as complementary therapy. INT J VITAM NUTR RES 2020; 92:248-266. [PMID: 32065556 DOI: 10.1024/0300-9831/a000630] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The prevalence of autism spectrum disorders (ASD) has risen sharply in the last 30 years, posing a major public health concern and a big emotional and financial challenge for families. While the underlying causes remain to be fully elucidated, evidence shows moderate genetic heritability contribution, but heavy environmental influence. Over the last decades, modern lifestyle has deeply changed our eating, rest, and exercise habits, while exposure to air, water, and food chemical pollution has increased due to indiscriminate use of pesticides, food additives, adjuvants, and antibiotics. The result is a drastic change in the quality of our energy source input, and an overload for antioxidant and detoxification pathways that compromises normal metabolism and homeostasis. Current research shows high prevalence of food selectivity and/or food allergy among children with autism, resulting in essential micronutrient deficits that may trigger or aggravate physical and cognitive symptoms. Nutrigenomics is an emerging discipline that focuses on genotype-micronutrient interaction, and a useful approach to tailor low risk, personalized interventions through diet and micronutrient supplementation. Here, we review available literature addressing the role of micronutrients in the symptomatology of ASD, the metabolic pathways involved, and their therapeutic relevance. Personalized and supervised supplementation according to individual needs is suggested as a complement of traditional therapies to improve outcome both for children with autism and their families.
Collapse
Affiliation(s)
- María S Jaureguiberry
- Centro de Investigaciones en Toxicología Ambiental y Agrobiotecnología del Comahue-CITAAC, Universidad Nacional del Comahue-CONICET, Neuquén, Argentina
| | - Andrés Venturino
- Centro de Investigaciones en Toxicología Ambiental y Agrobiotecnología del Comahue-CITAAC, Universidad Nacional del Comahue-CONICET, Neuquén, Argentina
| |
Collapse
|
136
|
Balasco L, Provenzano G, Bozzi Y. Sensory Abnormalities in Autism Spectrum Disorders: A Focus on the Tactile Domain, From Genetic Mouse Models to the Clinic. Front Psychiatry 2020; 10:1016. [PMID: 32047448 PMCID: PMC6997554 DOI: 10.3389/fpsyt.2019.01016] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Accepted: 12/20/2019] [Indexed: 12/20/2022] Open
Abstract
Sensory abnormalities are commonly recognized as diagnostic criteria in autism spectrum disorder (ASD), as reported in the last edition of the Diagnostic and Statistical Manual of Mental Disorder (DSM-V). About 90% of ASD individuals have atypical sensory experiences, described as both hyper- and hypo-reactivity, with abnormal responses to tactile stimulation representing a very frequent finding. In this review, we will address the neurobiological bases of sensory processing in ASD, with a specific focus of tactile sensitivity. In the first part, we will review the most relevant sensory abnormalities detected in ASD, and then focus on tactile processing deficits through the discussion of recent clinical and experimental studies. In the search for the neurobiological bases of ASD, several mouse models have been generated with knockout and humanized knockin mutations in many ASD-associated genes. Here, we will therefore give a brief overview of the anatomical structure of the mouse somatosensory system, and describe the somatosensory abnormalities so far reported in different mouse models of ASD. Understanding the neurobiological bases of sensory processing in ASD mouse models may represent an opportunity for a better comprehension of the mechanisms underlying sensory abnormalities, and for the development of novel effective therapeutic strategies.
Collapse
Affiliation(s)
- Luigi Balasco
- Center for Mind/Brain Sciences (CIMeC), University of Trento, Rovereto, Italy
| | - Giovanni Provenzano
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Trento, Italy
| | - Yuri Bozzi
- Center for Mind/Brain Sciences (CIMeC), University of Trento, Rovereto, Italy
- CNR Neuroscience Institute, Pisa, Italy
| |
Collapse
|
137
|
Excitation/inhibition imbalance and impaired neurogenesis in neurodevelopmental and neurodegenerative disorders. Rev Neurosci 2019; 30:807-820. [DOI: 10.1515/revneuro-2019-0014] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Accepted: 04/05/2019] [Indexed: 12/31/2022]
Abstract
AbstractThe excitation/inhibition (E/I) balance controls the synaptic inputs to prevent the inappropriate responses of neurons to input strength, and is required to restore the initial pattern of network activity. Various neurotransmitters affect synaptic plasticity within neural networks via the modulation of neuronal E/I balance in the developing and adult brain. Less is known about the role of E/I balance in the control of the development of the neural stem and progenitor cells in the course of neurogenesis and gliogenesis. Recent findings suggest that neural stem and progenitor cells appear to be the target for the action of GABA within the neurogenic or oligovascular niches. The same might be true for the role of neuropeptides (i.e. oxytocin) in neurogenic niches. This review covers current understanding of the role of E/I balance in the regulation of neuroplasticity associated with social behavior in normal brain, and in neurodevelopmental and neurodegenerative diseases. Further studies are required to decipher the GABA-mediated regulation of postnatal neurogenesis and synaptic integration of newly-born neurons as a potential target for the treatment of brain diseases.
Collapse
|
138
|
Foxe JJ, Molholm S, Baudouin SJ, Wallace MT. Explorations and perspectives on the neurobiological bases of autism spectrum disorder. Eur J Neurosci 2019; 47:488-496. [PMID: 29575230 DOI: 10.1111/ejn.13902] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- John J Foxe
- Department of Neuroscience, The Ernest J. Del Monte Institute for Neuroscience, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA.,The Cognitive Neurophysiology Laboratory, Departments of Pediatrics and Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Sophie Molholm
- Department of Neuroscience, The Ernest J. Del Monte Institute for Neuroscience, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA.,The Cognitive Neurophysiology Laboratory, Departments of Pediatrics and Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
| | | | - Mark T Wallace
- Center for Integrative and Cognitive Neuroscience, Vanderbilt University, Nashville, TN, USA
| |
Collapse
|
139
|
Prenatal valproate in rodents as a tool to understand the neural underpinnings of social dysfunctions in autism spectrum disorder. Neuropharmacology 2019; 159:107477. [DOI: 10.1016/j.neuropharm.2018.12.024] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Revised: 12/11/2018] [Accepted: 12/20/2018] [Indexed: 12/22/2022]
|
140
|
Fleury-Teixeira P, Caixeta FV, Ramires da Silva LC, Brasil-Neto JP, Malcher-Lopes R. Effects of CBD-Enriched Cannabis sativa Extract on Autism Spectrum Disorder Symptoms: An Observational Study of 18 Participants Undergoing Compassionate Use. Front Neurol 2019; 10:1145. [PMID: 31736860 PMCID: PMC6834767 DOI: 10.3389/fneur.2019.01145] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Accepted: 10/14/2019] [Indexed: 12/17/2022] Open
Abstract
Autism Spectrum Disorders comprise conditions that may affect cognitive development, motor skills, social interaction, communication, and behavior. This set of functional deficits often results in lack of independence for the diagnosed individuals, and severe distress for patients, families, and caregivers. There is a mounting body of evidence indicating the effectiveness of pure cannabidiol (CBD) and CBD-enriched Cannabis sativa extract (CE) for the treatment of autistic symptoms in refractory epilepsy patients. There is also increasing data support for the hypothesis that non-epileptic autism shares underlying etiological mechanisms with epilepsy. Here we report an observational study with a cohort of 18 autistic patients undergoing treatment with compassionate use of standardized CBD-enriched CE (with a CBD to THC ratio of 75/1). Among the 15 patients who adhered to the treatment (10 non-epileptic and five epileptic) only one patient showed lack of improvement in autistic symptoms. Due to adverse effects, three patients discontinued CE use before 1 month. After 6-9 months of treatment, most patients, including epileptic and non-epileptic, showed some level of improvement in more than one of the eight symptom categories evaluated: Attention Deficit/Hyperactivity Disorder; Behavioral Disorders; Motor Deficits; Autonomy Deficits; Communication and Social Interaction Deficits; Cognitive Deficits; Sleep Disorders and Seizures, with very infrequent and mild adverse effects. The strongest improvements were reported for Seizures, Attention Deficit/Hyperactivity Disorder, Sleep Disorders, and Communication and Social Interaction Deficits. This was especially true for the 10 non-epileptic patients, nine of which presented improvement equal to or above 30% in at least one of the eight categories, six presented improvement of 30% or more in at least two categories and four presented improvement equal to or above 30% in at least four symptom categories. Ten out of the 15 patients were using other medicines, and nine of these were able to keep the improvements even after reducing or withdrawing other medications. The results reported here are very promising and indicate that CBD-enriched CE may ameliorate multiple ASD symptoms even in non-epileptic patients, with substantial increase in life quality for both ASD patients and caretakers.
Collapse
Affiliation(s)
| | | | - Leandro Cruz Ramires da Silva
- Clinical Hospital, Federal University of Minas Gerais, Belo Horizonte, Brazil.,Associação Brasileira de Pacientes de Cannabis Medicinal, Belo Horizonte, Brazil
| | | | | |
Collapse
|
141
|
Rivell A, Mattson MP. Intergenerational Metabolic Syndrome and Neuronal Network Hyperexcitability in Autism. Trends Neurosci 2019; 42:709-726. [PMID: 31495451 PMCID: PMC6779523 DOI: 10.1016/j.tins.2019.08.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 07/17/2019] [Accepted: 08/06/2019] [Indexed: 12/13/2022]
Abstract
We review evidence that suggests a role for excessive consumption of energy-dense foods, particularly fructose, and consequent obesity and insulin resistance (metabolic syndrome) in the recent increase in prevalence of autism spectrum disorders (ASD). Maternal insulin resistance, obesity, and diabetes may predispose offspring to ASD by mechanisms involving chronic activation of anabolic cellular pathways and a lack of metabolic switching to ketosis resulting in a deficit in GABAergic signaling and neuronal network hyperexcitability. Metabolic reprogramming by epigenetic DNA and chromatin modifications may contribute to alterations in gene expression that result in ASD. These mechanistic insights suggest that interventions that improve metabolic health such as intermittent fasting and exercise may ameliorate developmental neuronal network abnormalities and consequent behavioral manifestations in ASD.
Collapse
Affiliation(s)
- Aileen Rivell
- Laboratory of Neurosciences, National Institute on Aging Intramural Research Program, Baltimore, MD, USA
| | - Mark P Mattson
- Laboratory of Neurosciences, National Institute on Aging Intramural Research Program, Baltimore, MD, USA; Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
142
|
Pan YH, Wu N, Yuan XB. Toward a Better Understanding of Neuronal Migration Deficits in Autism Spectrum Disorders. Front Cell Dev Biol 2019; 7:205. [PMID: 31620440 PMCID: PMC6763556 DOI: 10.3389/fcell.2019.00205] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2019] [Accepted: 09/06/2019] [Indexed: 11/13/2022] Open
Abstract
Newborn neurons in developing brains actively migrate from germinal zones to designated regions before being wired into functional circuits. The motility and trajectory of migrating neurons are regulated by both extracellular factors and intracellular signaling cascades. Defects in the molecular machinery of neuronal migration lead to mis-localization of affected neurons and are considered as an important etiology of multiple developmental disorders including epilepsy, dyslexia, schizophrenia (SCZ), and autism spectrum disorders (ASD). However, the mechanisms that link neuronal migration deficits to the development of these diseases remain elusive. This review focuses on neuronal migration deficits in ASD. From a translational perspective, we discuss (1) whether neuronal migration deficits are general neuropathological characteristics of ASD; (2) how the phenotypic heterogeneity of neuronal migration disorders is generated; (3) how neuronal migration deficits lead to functional defects of brain circuits; and (4) how therapeutic intervention of neuronal migration deficits can be a potential treatment for ASD.
Collapse
Affiliation(s)
- Yi-Hsuan Pan
- Key Laboratory of Brain Functional Genomics (Ministry of Education and Shanghai), Institute of Brain Functional Genomics, School of Life Sciences and the Collaborative Innovation Center for Brain Science, East China Normal University, Shanghai, China
| | - Nan Wu
- Key Laboratory of Brain Functional Genomics (Ministry of Education and Shanghai), Institute of Brain Functional Genomics, School of Life Sciences and the Collaborative Innovation Center for Brain Science, East China Normal University, Shanghai, China
| | - Xiao-Bing Yuan
- Key Laboratory of Brain Functional Genomics (Ministry of Education and Shanghai), Institute of Brain Functional Genomics, School of Life Sciences and the Collaborative Innovation Center for Brain Science, East China Normal University, Shanghai, China.,Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, United States
| |
Collapse
|
143
|
Cognitive and behavioral effects of brief seizures in mice. Epilepsy Behav 2019; 98:249-257. [PMID: 31398689 DOI: 10.1016/j.yebeh.2019.07.015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 07/03/2019] [Accepted: 07/04/2019] [Indexed: 11/22/2022]
Abstract
Comorbidities associated with epilepsy greatly reduce patients' quality of life. Since antiepilepsy drugs show limited success in ameliorating cognitive and behavioral symptoms, there is a need to better understand the mechanisms underlying epilepsy-related cognitive and behavioral impairments. Most prior research addressing this problem has focused on chronic epilepsy, wherein many factors can simultaneously impact cognition and behavior. The purpose of the present study was to develop a testing paradigm using mice that can provide new insight into how short-term biological changes underlying acute seizures impact cognition and behavior. In Experiment 1, naïve C57BL/6J mice were subjected to either three brief, generalized electroconvulsive seizure (ECS) or three sham treatments equally spaced over the course of 30 min. Over the next 2 h, mice were tested in a novel object recognition paradigm. Follow-up studies examined locomotor activity immediately before and after (Experiment 2), immediately after (Experiment 3), and 45 min after (Experiment 4) a set of three ECS or sham treatments. Whereas results demonstrated that there was no statistically significant difference in recognition memory acquisition between ECS and sham-treated mice, measures of anxiety-like behavior were increased and novel object interest was decreased in ECS-treated mice compared with that in sham. Interestingly, ECS also produced a delayed inhibitory effect on locomotion, decreasing open-field activity 45-min posttreatment compared to sham. We conclude that a small cluster of brief seizures can have acute, behaviorally relevant effects in mice, and that greater emphasis should be placed on events that take place before chronic epilepsy is established in order to better understand epilepsy-related cognitive and behavioral impairments. Future research would benefit from using the paradigms defined above to study the effects of individual seizures on mouse cognition and behavior.
Collapse
|
144
|
Kim Y, Vadodaria KC, Lenkei Z, Kato T, Gage FH, Marchetto MC, Santos R. Mitochondria, Metabolism, and Redox Mechanisms in Psychiatric Disorders. Antioxid Redox Signal 2019; 31:275-317. [PMID: 30585734 PMCID: PMC6602118 DOI: 10.1089/ars.2018.7606] [Citation(s) in RCA: 105] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Revised: 12/21/2018] [Accepted: 12/23/2018] [Indexed: 12/17/2022]
Abstract
Significance: Our current knowledge of the pathophysiology and molecular mechanisms causing psychiatric disorders is modest, but genetic susceptibility and environmental factors are central to the etiology of these conditions. Autism, schizophrenia, bipolar disorder and major depressive disorder show genetic gene risk overlap and share symptoms and metabolic comorbidities. The identification of such common features may provide insights into the development of these disorders. Recent Advances: Multiple pieces of evidence suggest that brain energy metabolism, mitochondrial functions and redox balance are impaired to various degrees in psychiatric disorders. Since mitochondrial metabolism and redox signaling can integrate genetic and environmental environmental factors affecting the brain, it is possible that they are implicated in the etiology and progression of psychiatric disorders. Critical Issue: Evidence for direct links between cellular mitochondrial dysfunction and disease features are missing. Future Directions: A better understanding of the mitochondrial biology and its intracellular connections to the nuclear genome, the endoplasmic reticulum and signaling pathways, as well as its role in intercellular communication in the organism, is still needed. This review focuses on the findings that implicate mitochondrial dysfunction, the resultant metabolic changes and oxidative stress as important etiological factors in the context of psychiatric disorders. We also propose a model where specific pathophysiologies of psychiatric disorders depend on circuit-specific impairments of mitochondrial dysfunction and redox signaling at specific developmental stages.
Collapse
Affiliation(s)
- Yeni Kim
- Department of Child and Adolescent Psychiatry, National Center for Mental Health, Seoul, South Korea
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, California
| | - Krishna C. Vadodaria
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, California
| | - Zsolt Lenkei
- Laboratory of Dynamic of Neuronal Structure in Health and Disease, Institute of Psychiatry and Neuroscience of Paris (UMR_S1266 INSERM, University Paris Descartes), Paris, France
| | - Tadafumi Kato
- Laboratory for Molecular Dynamics of Mental Disorders, RIKEN Center for Brain Science, Wako, Japan
| | - Fred H. Gage
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, California
| | - Maria C. Marchetto
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, California
| | - Renata Santos
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, California
- Laboratory of Dynamic of Neuronal Structure in Health and Disease, Institute of Psychiatry and Neuroscience of Paris (UMR_S1266 INSERM, University Paris Descartes), Paris, France
| |
Collapse
|
145
|
Mold M, Cottle J, Exley C. Aluminium in Brain Tissue in Epilepsy: A Case Report from Camelford. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2019; 16:ijerph16122129. [PMID: 31208130 PMCID: PMC6616903 DOI: 10.3390/ijerph16122129] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 06/14/2019] [Accepted: 06/14/2019] [Indexed: 11/16/2022]
Abstract
(1) Introduction: Human exposure to aluminium is a burgeoning problem. In 1988, the population of the Cornish town of Camelford was exposed to exceedingly high levels of aluminium in their potable water supply. Herein we provide evidence that aluminium played a role in the death of a Camelford resident following development of late-onset epilepsy. (2) Case summary: We have measured the aluminium content of brain tissue in this individual and demonstrated significant accumulations of aluminium in the hippocampus (4.35 (2.80) µg/g dry wt.) and the occipital lobe (2.22 (2.23) µg/g dry wt., mean, SD, n = 5), the latter being associated with abnormal calcifications. Aluminium-specific fluorescence microscopy confirmed the presence of aluminium in both of these tissues and made the consistent observation of aluminium-loaded glial cells in close proximity to aluminium-rich cell/neuronal debris. These observations support an inflammatory component in this case of late-onset epilepsy. Congo red failed to identify any amyloid deposits in any tissue while thioflavin S showed extensive extracellular and intracellular tau pathologies. (3) Discussion: We present the first data showing aluminium in brain tissue in epilepsy and suggest, in light of complementary evidence from scientific literature, the first evidence that aluminium played a role in the advent of this case of late-onset adult epilepsy.
Collapse
Affiliation(s)
- Matthew Mold
- The Birchall Centre, Lennard-Jones Laboratories, Keele University, Staffordshire ST5 5BG, UK.
| | - Jason Cottle
- School of Medicine, David Weatherly Building, Keele University, Staffordshire ST5 5BG, UK.
| | - Christopher Exley
- The Birchall Centre, Lennard-Jones Laboratories, Keele University, Staffordshire ST5 5BG, UK.
| |
Collapse
|
146
|
Loussouarn A, Dozières-Puyravel B, Auvin S. Autistic spectrum disorder and epilepsy: diagnostic challenges. Expert Rev Neurother 2019; 19:579-585. [PMID: 31081698 DOI: 10.1080/14737175.2019.1617699] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Introduction: Epilepsy is more frequent in individuals with Autism Spectrum Disorder (ASD) than in the general population; however, its diagnosis is frequently challenging. Areas Covered: We report the current diagnostic criteria for both ASD and epilepsy. We describe the incidence, prevalence, and risk factors for epilepsy in patients with ASD. We then focus on the electro-clinical approach, including the clinical evaluation of cognitive regression. Expert Opinion: A diagnosis of epilepsy should be made based on the International League Against Epilepsy (ILAE) definition. A diagnosis of epilepsy should be established based on a single seizure with electroencephalography (EEG) abnormalities. Considering the high prevalence of EEG abnormalities in children with ASD without epilepsy, EEG should only be performed at epilepsy onset, and more precisely when a clinical interview has confirmed that repetitive paroxysmal events could be seizures. There are still many gaps in our understanding of epilepsy in patients with ASD. It would be of interest to further understand the links, if any, between EEG abnormalities and ASD phenotype. The identification of epilepsy syndromes in ASD would help analyze the possible underlying etiologies, for the administration of more appropriate antiepileptic drugs (AED), and to explain the prognosis to caregivers.
Collapse
Affiliation(s)
- Anna Loussouarn
- a Department of Pediatric Neurology , AP-HP, Robert Debré Hospital , Paris , France
| | | | - Stéphane Auvin
- a Department of Pediatric Neurology , AP-HP, Robert Debré Hospital , Paris , France.,b University Paris Diderot, Sorbonne Paris Cité, INSERM UMR1141 , Paris , France
| |
Collapse
|
147
|
Xiong J, Chen S, Pang N, Deng X, Yang L, He F, Wu L, Chen C, Yin F, Peng J. Neurological Diseases With Autism Spectrum Disorder: Role of ASD Risk Genes. Front Neurosci 2019; 13:349. [PMID: 31031587 PMCID: PMC6470315 DOI: 10.3389/fnins.2019.00349] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Accepted: 03/26/2019] [Indexed: 02/06/2023] Open
Abstract
Autism spectrum disorder (ASD) is frequently comorbid with other neurological disorders such as intellectual disability (ID) or global development delay (GDD) and epilepsy. The pathogenesis of ASD is complex. So far, studies have identified more than 1000 ASD risk genes. Most of them were also reported to relate with other neurological diseases, and only several of them have been confirmed as pathogenic genes for autism. Little is known about the roles of these risk genes in neurological diseases with ASD. In the present study, we recruited a cohort of 158 neurological disorder probands with 163 variants of 48 ASD risk genes. Of these, 50 individuals (31.6%) were diagnosed with ASD. In the ASD patient subset, we identified several rarely reported candidate genes including DOLK, USH2A, and HUWE1. In a comparison of patients with neurological disorders with and without ASD, we found that ID/GDD was frequently comorbid with ASD whereas epilepsy was more common in the non-ASD group. Statistical analyses of all possible risk factors implicated that variants in synaptic genes, especially non-voltage-gated ion channel genes and in transcriptional and chromosome genes were related to ASD, but none of the investigated environmental factors was. Our results are useful for the future diagnosis and prognosis of patients with neurological disorders and emphasize the utility of genetic screening.
Collapse
Affiliation(s)
- Juan Xiong
- Departmen of Pediatrics, Xiangya Hospital, Central South University, Changsha, China.,Hunan Intellectual and Developmental Disabilities Research Center, Changsha, China
| | - Shimeng Chen
- Departmen of Pediatrics, Xiangya Hospital, Central South University, Changsha, China.,Hunan Intellectual and Developmental Disabilities Research Center, Changsha, China
| | - Nan Pang
- Departmen of Pediatrics, Xiangya Hospital, Central South University, Changsha, China.,Hunan Intellectual and Developmental Disabilities Research Center, Changsha, China
| | - Xiaolu Deng
- Departmen of Pediatrics, Xiangya Hospital, Central South University, Changsha, China.,Hunan Intellectual and Developmental Disabilities Research Center, Changsha, China
| | - Lifen Yang
- Departmen of Pediatrics, Xiangya Hospital, Central South University, Changsha, China.,Hunan Intellectual and Developmental Disabilities Research Center, Changsha, China
| | - Fang He
- Departmen of Pediatrics, Xiangya Hospital, Central South University, Changsha, China.,Hunan Intellectual and Developmental Disabilities Research Center, Changsha, China
| | - Liwen Wu
- Departmen of Pediatrics, Xiangya Hospital, Central South University, Changsha, China.,Hunan Intellectual and Developmental Disabilities Research Center, Changsha, China
| | - Chen Chen
- Departmen of Pediatrics, Xiangya Hospital, Central South University, Changsha, China.,Hunan Intellectual and Developmental Disabilities Research Center, Changsha, China
| | - Fei Yin
- Departmen of Pediatrics, Xiangya Hospital, Central South University, Changsha, China.,Hunan Intellectual and Developmental Disabilities Research Center, Changsha, China
| | - Jing Peng
- Departmen of Pediatrics, Xiangya Hospital, Central South University, Changsha, China.,Hunan Intellectual and Developmental Disabilities Research Center, Changsha, China
| |
Collapse
|
148
|
Carter CJ. Autism genes and the leukocyte transcriptome in autistic toddlers relate to pathogen interactomes, infection and the immune system. A role for excess neurotrophic sAPPα and reduced antimicrobial Aβ. Neurochem Int 2019; 126:36-58. [PMID: 30862493 DOI: 10.1016/j.neuint.2019.03.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2019] [Revised: 02/22/2019] [Accepted: 03/06/2019] [Indexed: 12/20/2022]
Abstract
Prenatal and early childhood infections have been implicated in autism. Many autism susceptibility genes (206 Autworks genes) are localised in the immune system and are related to immune/infection pathways. They are enriched in the host/pathogen interactomes of 18 separate microbes (bacteria/viruses and fungi) and to the genes regulated by bacterial toxins, mycotoxins and Toll-like receptor ligands. This enrichment was also observed for misregulated genes from a microarray study of leukocytes from autistic toddlers. The upregulated genes from this leukocyte study also matched the expression profiles in response to numerous infectious agents from the Broad Institute molecular signatures database. They also matched genes related to sudden infant death syndrome and autism comorbid conditions (autoimmune disease, systemic lupus erythematosus, diabetes, epilepsy and cardiomyopathy) as well as to estrogen and thyrotropin responses and to those upregulated by different types of stressors including oxidative stress, hypoxia, endoplasmic reticulum stress, ultraviolet radiation or 2,4-dinitrofluorobenzene, a hapten used to develop allergic skin reactions in animal models. The oxidative/integrated stress response is also upregulated in the autism brain and may contribute to myelination problems. There was also a marked similarity between the expression signatures of autism and Alzheimer's disease, and 44 shared autism/Alzheimer's disease genes are almost exclusively expressed in the blood-brain barrier. However, in contrast to Alzheimer's disease, levels of the antimicrobial peptide beta-amyloid are decreased and the levels of the neurotrophic/myelinotrophic soluble APP alpha are increased in autism, together with an increased activity of α-secretase. sAPPα induces an increase in glutamatergic and a decrease in GABA-ergic synapses creating and excitatory/inhibitory imbalance that has also been observed in autism. A literature survey showed that multiple autism genes converge on APP processing and that many are able to increase sAPPalpha at the expense of beta-amyloid production. A genetically programmed tilt of this axis towards an overproduction of neurotrophic/gliotrophic sAPPalpha and underproduction of antimicrobial beta-amyloid may explain the brain overgrowth and myelination dysfunction, as well as the involvement of pathogens in autism.
Collapse
Affiliation(s)
- C J Carter
- PolygenicPathways, 41C Marina, Saint Leonard's on Sea, TN38 0BU, East Sussex, UK.
| |
Collapse
|
149
|
Chelini G, Zerbi V, Cimino L, Grigoli A, Markicevic M, Libera F, Robbiati S, Gadler M, Bronzoni S, Miorelli S, Galbusera A, Gozzi A, Casarosa S, Provenzano G, Bozzi Y. Aberrant Somatosensory Processing and Connectivity in Mice Lacking Engrailed-2. J Neurosci 2019; 39:1525-1538. [PMID: 30593497 PMCID: PMC6381254 DOI: 10.1523/jneurosci.0612-18.2018] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Revised: 12/12/2018] [Accepted: 12/14/2018] [Indexed: 11/21/2022] Open
Abstract
Overreactivity and defensive behaviors in response to tactile stimuli are common symptoms in autism spectrum disorder (ASD) patients. Similarly, somatosensory hypersensitivity has also been described in mice lacking ASD-associated genes such as Fmr1 (fragile X mental retardation protein 1). Fmr1 knock-out mice also show reduced functional connectivity between sensory cortical areas, which may represent an endogenous biomarker for their hypersensitivity. Here, we measured whole-brain functional connectivity in Engrailed-2 knock-out (En2-/-) adult mice, which show a lower expression of Fmr1 and anatomical defects common to Fmr1 knock-outs. MRI-based resting-state functional connectivity in adult En2-/- mice revealed significantly reduced synchronization in somatosensory-auditory/associative cortices and dorsal thalamus, suggesting the presence of aberrant somatosensory processing in these mutants. Accordingly, when tested in the whisker nuisance test, En2-/- but not WT mice of both sexes showed fear behavior in response to repeated whisker stimulation. En2-/- mice undergoing this test exhibited decreased c-Fos-positive neurons (a marker of neuronal activity) in layer IV of the primary somatosensory cortex and increased immunoreactive cells in the basolateral amygdala compared with WT littermates. Conversely, when tested in a sensory maze, En2-/- and WT mice spent a comparable time in whisker-guided exploration, indicating that whisker-mediated behaviors are otherwise preserved in En2 mutants. Therefore, fearful responses to somatosensory stimuli in En2-/- mice are accompanied by reduced basal connectivity of sensory regions, reduced activation of somatosensory cortex, and increased activation of the basolateral amygdala, suggesting that impaired somatosensory processing is a common feature in mice lacking ASD-related genes.SIGNIFICANCE STATEMENT Overreactivity to tactile stimuli is a common symptom in autism spectrum disorder (ASD) patients. Recent studies performed in mice bearing ASD-related mutations confirmed these findings. Here, we evaluated the behavioral response to whisker stimulation in mice lacking the ASD-related gene Engrailed-2 (En2-/- mice). Compared with WT controls, En2-/- mice showed reduced functional connectivity in the somatosensory cortex, which was paralleled by fear behavior, reduced activation of somatosensory cortex, and increased activation of the basolateral amygdala in response to repeated whisker stimulation. These results suggest that impaired somatosensory signal processing is a common feature in mice harboring ASD-related mutations.
Collapse
Affiliation(s)
- Gabriele Chelini
- Center for Integrative Biology (CIBIO), University of Trento, 38123 Trento, Italy
| | - Valerio Zerbi
- Neural Control of Movement Laboratory, Department of Health Sciences and Technology, Eidgenössische Technische Hochschule (ETH) Zürich, 8057 Zurich, Switzerland
| | - Luca Cimino
- Center for Integrative Biology (CIBIO), University of Trento, 38123 Trento, Italy
| | - Andrea Grigoli
- Center for Integrative Biology (CIBIO), University of Trento, 38123 Trento, Italy
| | - Marija Markicevic
- Neural Control of Movement Laboratory, Department of Health Sciences and Technology, Eidgenössische Technische Hochschule (ETH) Zürich, 8057 Zurich, Switzerland
| | - Francesco Libera
- Center for Integrative Biology (CIBIO), University of Trento, 38123 Trento, Italy
- Center for Mind/Brain Sciences (CIMeC), University of Trento, 38068 Rovereto, Italy
| | - Sergio Robbiati
- Model Organisms Facility, Center for Integrative Biology, University of Trento, 38123 Trento, Italy
| | - Mattia Gadler
- Center for Integrative Biology (CIBIO), University of Trento, 38123 Trento, Italy
| | - Silvia Bronzoni
- Center for Integrative Biology (CIBIO), University of Trento, 38123 Trento, Italy
- Center for Mind/Brain Sciences (CIMeC), University of Trento, 38068 Rovereto, Italy
| | - Silvia Miorelli
- Center for Integrative Biology (CIBIO), University of Trento, 38123 Trento, Italy
| | - Alberto Galbusera
- Functional Neuroimaging Laboratory, Center for Neuroscience and Cognitive Systems, Istituto Italiano di Tecnologia, 38068 Rovereto, Italy, and
| | - Alessandro Gozzi
- Functional Neuroimaging Laboratory, Center for Neuroscience and Cognitive Systems, Istituto Italiano di Tecnologia, 38068 Rovereto, Italy, and
| | - Simona Casarosa
- Center for Integrative Biology (CIBIO), University of Trento, 38123 Trento, Italy
- CNR Neuroscience Institute, 56124 Pisa, Italy
| | - Giovanni Provenzano
- Center for Integrative Biology (CIBIO), University of Trento, 38123 Trento, Italy,
| | - Yuri Bozzi
- Center for Mind/Brain Sciences (CIMeC), University of Trento, 38068 Rovereto, Italy,
- CNR Neuroscience Institute, 56124 Pisa, Italy
| |
Collapse
|
150
|
Finisguerra A, Borgatti R, Urgesi C. Non-invasive Brain Stimulation for the Rehabilitation of Children and Adolescents With Neurodevelopmental Disorders: A Systematic Review. Front Psychol 2019; 10:135. [PMID: 30787895 PMCID: PMC6373438 DOI: 10.3389/fpsyg.2019.00135] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Accepted: 01/15/2019] [Indexed: 12/18/2022] Open
Abstract
In the last years, there has been a growing interest in the application of different non-invasive brain stimulation techniques to induce neuroplasticity and to modulate cognition and behavior in adults. Very recently, different attempts have been made to induce functional plastic changes also in pediatric populations. Importantly, not only sensorimotor processing, but also higher-level functions have been addressed, with the aim to boost rehabilitation in different neurodevelopmental disorders. However, efficacy and safety of using these techniques in pediatric population is still debated. The current article aims to review the non-invasive brain stimulation studies conducted in pediatric populations using Transcranial Magnetic Stimulation or transcranial Direct Current Stimulation. Specifically, the available proofs concerning the efficacy and safety of these techniques on Autism Spectrum Disorder, Attention-deficit/hyperactivity disorder, Dyslexia, Tourette syndrome, and tic disorders are systematically reviewed and discussed. The article also aims to provide an overview about other possible applications of these and other stimulation techniques for rehabilitative purposes in children and adolescents.
Collapse
Affiliation(s)
| | - Renato Borgatti
- Child Neuropsychiatry and Neurorehabilitation Unit, Scientific Institute, IRCCS E. Medea, Bosisio Parini, Italy
| | - Cosimo Urgesi
- Scientific Institute, IRCCS E. Medea, Pasian di Prato, Udine, Italy.,Child Neuropsychiatry and Neurorehabilitation Unit, Scientific Institute, IRCCS E. Medea, Bosisio Parini, Italy.,Laboratory of Cognitive Neuroscience, Department of Languages, Literatures, Communication, Education and Society, University of Udine, Udine, Italy
| |
Collapse
|