101
|
Mueller-Buehl C, Reinhard J, Roll L, Bader V, Winklhofer KF, Faissner A. Brevican, Neurocan, Tenascin-C, and Tenascin-R Act as Important Regulators of the Interplay Between Perineuronal Nets, Synaptic Integrity, Inhibitory Interneurons, and Otx2. Front Cell Dev Biol 2022; 10:886527. [PMID: 35721494 PMCID: PMC9201762 DOI: 10.3389/fcell.2022.886527] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 04/11/2022] [Indexed: 11/13/2022] Open
Abstract
Fast-spiking parvalbumin interneurons are critical for the function of mature cortical inhibitory circuits. Most of these neurons are enwrapped by a specialized extracellular matrix (ECM) structure called perineuronal net (PNN), which can regulate their synaptic input. In this study, we investigated the relationship between PNNs, parvalbumin interneurons, and synaptic distribution on these cells in the adult primary visual cortex (V1) of quadruple knockout mice deficient for the ECM molecules brevican, neurocan, tenascin-C, and tenascin-R. We used super-resolution structured illumination microscopy (SIM) to analyze PNN structure and associated synapses. In addition, we examined parvalbumin and calretinin interneuron populations. We observed a reduction in the number of PNN-enwrapped cells and clear disorganization of the PNN structure in the quadruple knockout V1. This was accompanied by an imbalance of inhibitory and excitatory synapses with a reduction of inhibitory and an increase of excitatory synaptic elements along the PNNs. Furthermore, the number of parvalbumin interneurons was reduced in the quadruple knockout, while calretinin interneurons, which do not wear PNNs, did not display differences in number. Interestingly, we found the transcription factor Otx2 homeoprotein positive cell population also reduced. Otx2 is crucial for parvalbumin interneuron and PNN maturation, and a positive feedback loop between these parameters has been described. Collectively, these data indicate an important role of brevican, neurocan, tenascin-C, and tenascin-R in regulating the interplay between PNNs, inhibitory interneurons, synaptic distribution, and Otx2 in the V1.
Collapse
Affiliation(s)
- Cornelius Mueller-Buehl
- Department of Cell Morphology and Molecular Neurobiology, Faculty of Biology and Biotechnology, Ruhr University Bochum, Bochum, Germany
| | - Jacqueline Reinhard
- Department of Cell Morphology and Molecular Neurobiology, Faculty of Biology and Biotechnology, Ruhr University Bochum, Bochum, Germany
| | - Lars Roll
- Department of Cell Morphology and Molecular Neurobiology, Faculty of Biology and Biotechnology, Ruhr University Bochum, Bochum, Germany
| | - Verian Bader
- Department of Molecular Cell Biology, Institute of Biochemistry and Pathobiochemistry, Ruhr University Bochum, Bochum, Germany
- Department of Biochemistry of Neurodegenerative Diseases, Institute of Biochemistry and Pathobiochemistry, Ruhr University Bochum, Bochum, Germany
| | - Konstanze F. Winklhofer
- Department of Molecular Cell Biology, Institute of Biochemistry and Pathobiochemistry, Ruhr University Bochum, Bochum, Germany
- Cluster of Excellence RESOLV, Ruhr University Bochum, Bochum, Germany
| | - Andreas Faissner
- Department of Cell Morphology and Molecular Neurobiology, Faculty of Biology and Biotechnology, Ruhr University Bochum, Bochum, Germany
| |
Collapse
|
102
|
Genetic mutation of TRPV2 induces anxiety by decreasing GABA-B R2 expression in hippocampus. Biochem Biophys Res Commun 2022; 620:135-142. [DOI: 10.1016/j.bbrc.2022.06.079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 06/23/2022] [Indexed: 11/22/2022]
|
103
|
Chung C, Shin W, Kim E. Early and Late Corrections in Mouse Models of Autism Spectrum Disorder. Biol Psychiatry 2022; 91:934-944. [PMID: 34556257 DOI: 10.1016/j.biopsych.2021.07.021] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 06/18/2021] [Accepted: 07/21/2021] [Indexed: 12/18/2022]
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder characterized by social and repetitive symptoms. A key feature of ASD is early-life manifestations of symptoms, indicative of early pathophysiological mechanisms. In mouse models of ASD, increasing evidence indicates that there are early pathophysiological mechanisms that can be corrected early to prevent phenotypic defects in adults, overcoming the disadvantage of the short-lasting effects that characterize adult-initiated treatments. In addition, the results from gene restorations indicate that ASD-related phenotypes can be rescued in some cases even after the brain has fully matured. These results suggest that we need to consider both temporal and mechanistic aspects in studies of ASD models and carefully compare genetic and nongenetic corrections. Here, we summarize the early and late corrections in mouse models of ASD by genetic and pharmacological interventions and discuss how to better integrate these results to ensure efficient and long-lasting corrections for eventual clinical translation.
Collapse
Affiliation(s)
- Changuk Chung
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science, Daejeon, South Korea; Department of Neurosciences, University of California San Diego, La Jolla, California
| | - Wangyong Shin
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science, Daejeon, South Korea
| | - Eunjoon Kim
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science, Daejeon, South Korea; Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, South Korea.
| |
Collapse
|
104
|
Xie Y, Xu Z, Xia M, Liu J, Shou X, Cui Z, Liao X, He Y. Alterations in Connectome Dynamics in Autism Spectrum Disorder: A Harmonized Mega- and Meta-analysis Study Using the Autism Brain Imaging Data Exchange Dataset. Biol Psychiatry 2022; 91:945-955. [PMID: 35144804 DOI: 10.1016/j.biopsych.2021.12.004] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 11/22/2021] [Accepted: 12/07/2021] [Indexed: 12/24/2022]
Abstract
BACKGROUND Neuroimaging studies have reported functional connectome aberrancies in autism spectrum disorder (ASD). However, the time-varying patterns of connectome topology in individuals with ASD and the connection between these patterns and gene expression profiles remain unknown. METHODS To investigate case-control differences in dynamic connectome topology, we conducted mega- and meta-analyses of resting-state functional magnetic resonance imaging data of 939 participants (440 patients with ASD and 499 healthy control subjects, all males) from 18 independent sites, selected from the Autism Brain Imaging Data Exchange (ABIDE) dataset. Functional data were preprocessed and analyzed using harmonized protocols, and brain module dynamics was assessed using a multilayer network model. We further leveraged postmortem brain-wide gene expression data to identify transcriptomic signatures associated with ASD-related alterations in brain dynamics. RESULTS Compared with healthy control participants, individuals with ASD exhibited a higher global mean and lower standard deviation of whole-brain module dynamics, indicating an unstable and less regionally differentiated pattern. More specifically, individuals with ASD showed higher module switching, primarily in the medial prefrontal cortex, posterior cingulate gyrus, and angular gyrus, and lower switching in the visual regions. These alterations in brain dynamics were predictive of social impairments in individuals with ASD and were linked with expression profiles of genes primarily involved in the regulation of neurotransmitter transport and secretion as well as with previously identified autism-related genes. CONCLUSIONS This study is the first to identify consistent alterations in brain network dynamics in ASD and the transcriptomic signatures related to those alterations, furthering insights into the biological basis behind this disorder.
Collapse
Affiliation(s)
- Yapei Xie
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing, China; Beijing Key Laboratory of Brain Imaging and Connectomics, Beijing Normal University, Beijing, China; IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, China
| | - Zhilei Xu
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing, China; Beijing Key Laboratory of Brain Imaging and Connectomics, Beijing Normal University, Beijing, China; IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, China
| | - Mingrui Xia
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing, China; Beijing Key Laboratory of Brain Imaging and Connectomics, Beijing Normal University, Beijing, China; IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, China
| | - Jin Liu
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing, China; Beijing Key Laboratory of Brain Imaging and Connectomics, Beijing Normal University, Beijing, China; IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, China
| | - Xiaojing Shou
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing, China; Beijing Key Laboratory of Brain Imaging and Connectomics, Beijing Normal University, Beijing, China; IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, China
| | - Zaixu Cui
- Chinese Institute for Brain Research, Beijing, China
| | - Xuhong Liao
- Beijing Key Laboratory of Brain Imaging and Connectomics, Beijing Normal University, Beijing, China; School of Systems Science, Beijing Normal University, Beijing, China.
| | - Yong He
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing, China; Beijing Key Laboratory of Brain Imaging and Connectomics, Beijing Normal University, Beijing, China; IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, China; Chinese Institute for Brain Research, Beijing, China.
| |
Collapse
|
105
|
Transcranial Direct Current Stimulation as an Approach to Mitigate Neurodevelopmental Disorders Affecting Excitation/Inhibition Balance: Focus on Autism Spectrum Disorder, Schizophrenia, and Attention Deficit/Hyperactivity Disorder. J Clin Med 2022; 11:jcm11102839. [PMID: 35628965 PMCID: PMC9143428 DOI: 10.3390/jcm11102839] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 05/12/2022] [Accepted: 05/14/2022] [Indexed: 02/04/2023] Open
Abstract
Transcranial direct current stimulation (tDCS) has been proposed as a promising therapy for rehabilitation of neurodevelopmental disorders. In this review, we discuss studies on the impact of tDCS as a therapy for autism, schizophrenia, and attention deficit/hyperactivity disorder, as well as the tDCS' mechanism of action, and propose future paths of research to optimize tDCS treatment protocols. The mechanism underlying tDCS effects is the modulation of excitatory and/or inhibitory activity, making it a valuable tool for restoring the excitation/inhibition (E/I) balance which is disrupted in many neurodevelopmental disorders. Clinical studies have shown that tDCS therapy is well-tolerated by patients and seems to ameliorate behavior and cognitive functions. Alterations in early development of neuronal circuits lead to disruptions in brain activity in neurodevelopmental disorders. An increasing amount of research into the effects of tDCS on neuronal activity has provided a foundation for its use as a therapy for behavior and cognitive characteristics of neurodevelopmental disorders. Clinical studies show that tDCS appears to ameliorate behavioral and cognitive outcomes of patients with autism, schizophrenia, and attention deficit/hyperactivity disorder. More research is needed to understand the mechanisms of action of tDCS and to optimize treatment protocols.
Collapse
|
106
|
Noel JP, Shivkumar S, Dokka K, Haefner RM, Angelaki DE. Aberrant causal inference and presence of a compensatory mechanism in autism spectrum disorder. eLife 2022; 11:71866. [PMID: 35579424 PMCID: PMC9170250 DOI: 10.7554/elife.71866] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 05/15/2022] [Indexed: 12/02/2022] Open
Abstract
Autism spectrum disorder (ASD) is characterized by a panoply of social, communicative, and sensory anomalies. As such, a central goal of computational psychiatry is to ascribe the heterogenous phenotypes observed in ASD to a limited set of canonical computations that may have gone awry in the disorder. Here, we posit causal inference - the process of inferring a causal structure linking sensory signals to hidden world causes - as one such computation. We show that audio-visual integration is intact in ASD and in line with optimal models of cue combination, yet multisensory behavior is anomalous in ASD because this group operates under an internal model favoring integration (vs. segregation). Paradoxically, during explicit reports of common cause across spatial or temporal disparities, individuals with ASD were less and not more likely to report common cause, particularly at small cue disparities. Formal model fitting revealed differences in both the prior probability for common cause (p-common) and choice biases, which are dissociable in implicit but not explicit causal inference tasks. Together, this pattern of results suggests (i) different internal models in attributing world causes to sensory signals in ASD relative to neurotypical individuals given identical sensory cues, and (ii) the presence of an explicit compensatory mechanism in ASD, with these individuals putatively having learned to compensate for their bias to integrate in explicit reports.
Collapse
Affiliation(s)
- Jean-Paul Noel
- Center for Neural Science, New York University, New York City, United States
| | | | - Kalpana Dokka
- Department of Neuroscience, Baylor College of Medicine, Houston, United States
| | - Ralf M Haefner
- Brain and Cognitive Sciences, University of Rochester, Rochester, United States
| | - Dora E Angelaki
- Center for Neural Science, New York University, New York City, United States.,Department of Neuroscience, Baylor College of Medicine, Houston, United States
| |
Collapse
|
107
|
Manyukhina VO, Prokofyev AO, Galuta IA, Goiaeva DE, Obukhova TS, Schneiderman JF, Altukhov DI, Stroganova TA, Orekhova EV. Globally elevated excitation-inhibition ratio in children with autism spectrum disorder and below-average intelligence. Mol Autism 2022; 13:20. [PMID: 35550191 PMCID: PMC9102291 DOI: 10.1186/s13229-022-00498-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Accepted: 04/25/2022] [Indexed: 12/04/2022] Open
Abstract
Background Altered neuronal excitation–inhibition (E–I) balance is strongly implicated in ASD. However, it is not known whether the direction and degree of changes in the E–I ratio in individuals with ASD correlates with intellectual disability often associated with this developmental disorder. The spectral slope of the aperiodic 1/f activity reflects the E–I balance at the scale of large neuronal populations and may uncover its putative alternations in individuals with ASD with and without intellectual disability. Methods Herein, we used magnetoencephalography (MEG) to test whether the 1/f slope would differentiate ASD children with average and below–average (< 85) IQ. MEG was recorded at rest with eyes open/closed in 49 boys with ASD aged 6–15 years with IQ ranging from 54 to 128, and in 49 age-matched typically developing (TD) boys. The cortical source activity was estimated using the beamformer approach and individual brain models. We then extracted the 1/f slope by fitting a linear function to the log–log-scale power spectra in the high-frequency range. Results The global 1/f slope averaged over all cortical sources demonstrated high rank-order stability between the two conditions. Consistent with previous research, it was steeper in the eyes-closed than in the eyes-open condition and flattened with age. Regardless of condition, children with ASD and below-average IQ had flatter slopes than either TD or ASD children with average or above-average IQ. These group differences could not be explained by differences in signal-to-noise ratio or periodic (alpha and beta) activity. Limitations Further research is needed to find out whether the observed changes in E–I ratios are characteristic of children with below-average IQ of other diagnostic groups. Conclusions The atypically flattened spectral slope of aperiodic activity in children with ASD and below-average IQ suggests a shift of the global E–I balance toward hyper-excitation. The spectral slope can provide an accessible noninvasive biomarker of the E–I ratio for making objective judgments about treatment effectiveness in people with ASD and comorbid intellectual disability. Supplementary Information The online version contains supplementary material available at 10.1186/s13229-022-00498-2.
Collapse
Affiliation(s)
- Viktoriya O Manyukhina
- Center for Neurocognitive Research (MEG Center), Moscow State University of Psychology and Education, Moscow, Russian Federation.,Department of Psychology, National Research University Higher School of Economics, Moscow, Russian Federation
| | - Andrey O Prokofyev
- Center for Neurocognitive Research (MEG Center), Moscow State University of Psychology and Education, Moscow, Russian Federation
| | - Ilia A Galuta
- Center for Neurocognitive Research (MEG Center), Moscow State University of Psychology and Education, Moscow, Russian Federation
| | - Dzerassa E Goiaeva
- Center for Neurocognitive Research (MEG Center), Moscow State University of Psychology and Education, Moscow, Russian Federation
| | - Tatiana S Obukhova
- Center for Neurocognitive Research (MEG Center), Moscow State University of Psychology and Education, Moscow, Russian Federation
| | - Justin F Schneiderman
- MedTech West and the Institute of Neuroscience and Physiology, Sahlgrenska Academy, The University of Gothenburg, Gothenburg, Sweden
| | - Dmitrii I Altukhov
- Department of Psychology, National Research University Higher School of Economics, Moscow, Russian Federation
| | - Tatiana A Stroganova
- Center for Neurocognitive Research (MEG Center), Moscow State University of Psychology and Education, Moscow, Russian Federation
| | - Elena V Orekhova
- Center for Neurocognitive Research (MEG Center), Moscow State University of Psychology and Education, Moscow, Russian Federation.
| |
Collapse
|
108
|
Kim D, Jung H, Shirai Y, Kim H, Kim J, Lim D, Mori T, Lee H, Park D, Kim HY, Guo Q, Pang B, Qiu W, Cao X, Kouyama-Suzuki E, Uemura T, Kasem E, Fu Y, Kim S, Tokunaga A, Yoshizawa T, Suzuki T, Sakagami H, Lee KJ, Ko J, Tabuchi K, Um JW. IQSEC3 Deletion Impairs Fear Memory Through Upregulation of Ribosomal S6K1 Signaling in the Hippocampus. Biol Psychiatry 2022; 91:821-831. [PMID: 35219498 DOI: 10.1016/j.biopsych.2021.12.016] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 11/29/2021] [Accepted: 12/15/2021] [Indexed: 12/11/2022]
Abstract
BACKGROUND IQSEC3, a gephyrin-binding GABAergic (gamma-aminobutyric acidergic) synapse-specific guanine nucleotide exchange factor, was recently reported to regulate activity-dependent GABAergic synapse maturation, but the underlying signaling mechanisms remain incompletely understood. METHODS We generated mice with conditional knockout (cKO) of Iqsec3 to examine whether altered synaptic inhibition influences hippocampus-dependent fear memory formation. In addition, electrophysiological recordings, immunohistochemistry, and behavioral assays were used to address our question. RESULTS We found that Iqsec3-cKO induces a specific reduction in GABAergic synapse density, GABAergic synaptic transmission, and maintenance of long-term potentiation in the hippocampal CA1 region. In addition, Iqsec3-cKO mice exhibited impaired fear memory formation. Strikingly, Iqsec3-cKO caused abnormally enhanced activation of ribosomal P70-S6K1-mediated signaling in the hippocampus but not in the cortex. Furthermore, inhibiting upregulated S6K1 signaling by expressing dominant-negative S6K1 in the hippocampal CA1 of Iqsec3-cKO mice completely rescued impaired fear learning and inhibitory synapse density but not deficits in long-term potentiation maintenance. Finally, upregulated S6K1 signaling was rescued by IQSEC3 wild-type, but not by an ARF-GEF (adenosine diphosphate ribosylation factor-guanine nucleotide exchange factor) inactive IQSEC3 mutant. CONCLUSIONS Our results suggest that IQSEC3-mediated balanced synaptic inhibition in hippocampal CA1 is critical for the proper formation of hippocampus-dependent fear memory.
Collapse
Affiliation(s)
- Dongwook Kim
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology, Daegu, South Korea
| | - Hyeji Jung
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology, Daegu, South Korea
| | - Yoshinori Shirai
- Department of Molecular and Cellular Physiology, Institute of Medicine, Academic Assembly, Shinshu University, Nagano, Japan
| | - Hyeonho Kim
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology, Daegu, South Korea
| | - Jinhu Kim
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology, Daegu, South Korea
| | - Dongseok Lim
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology, Daegu, South Korea
| | - Takuma Mori
- Department of Molecular and Cellular Physiology, Institute of Medicine, Academic Assembly, Shinshu University, Nagano, Japan; Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, Nagano, Japan
| | - Hyojeong Lee
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology, Daegu, South Korea
| | - Dongseok Park
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology, Daegu, South Korea
| | - Hee Young Kim
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology, Daegu, South Korea
| | - Qi Guo
- Department of Molecular and Cellular Physiology, Institute of Medicine, Academic Assembly, Shinshu University, Nagano, Japan
| | - Bo Pang
- Department of Molecular and Cellular Physiology, Institute of Medicine, Academic Assembly, Shinshu University, Nagano, Japan
| | - Wen Qiu
- Department of Molecular and Cellular Physiology, Institute of Medicine, Academic Assembly, Shinshu University, Nagano, Japan
| | - Xueshan Cao
- Department of Molecular and Cellular Physiology, Institute of Medicine, Academic Assembly, Shinshu University, Nagano, Japan
| | - Emi Kouyama-Suzuki
- Department of Molecular and Cellular Physiology, Institute of Medicine, Academic Assembly, Shinshu University, Nagano, Japan
| | - Takeshi Uemura
- Department of Molecular and Cellular Physiology, Institute of Medicine, Academic Assembly, Shinshu University, Nagano, Japan
| | - Enas Kasem
- Department of Molecular and Cellular Physiology, Institute of Medicine, Academic Assembly, Shinshu University, Nagano, Japan; Department of Zoology, Faculty of Science, Kafrelsheikh University, Kafr el-Sheikh, Egypt
| | - Yu Fu
- Department of Molecular and Cellular Physiology, Institute of Medicine, Academic Assembly, Shinshu University, Nagano, Japan
| | - Seungjoon Kim
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology, Daegu, South Korea
| | - Akinori Tokunaga
- Division of Laboratory Animal Resources, Life Science Research Laboratory, University of Fukui, Fukui, Japan
| | - Takahiro Yoshizawa
- Research Center for Supports to Advanced Science, Shinshu University, Nagano, Japan
| | - Tatsuo Suzuki
- Department of Molecular and Cellular Physiology, Institute of Medicine, Academic Assembly, Shinshu University, Nagano, Japan
| | - Hiroyuki Sakagami
- Department of Anatomy, Kitasato University School of Medicine, Kanagawa, Japan
| | - Kea Joo Lee
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology, Daegu, South Korea; Neural Circuits Research Group, Korea Brain Research Institute, Daegu, South Korea
| | - Jaewon Ko
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology, Daegu, South Korea
| | - Katsuhiko Tabuchi
- Department of Molecular and Cellular Physiology, Institute of Medicine, Academic Assembly, Shinshu University, Nagano, Japan; Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, Nagano, Japan.
| | - Ji Won Um
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology, Daegu, South Korea.
| |
Collapse
|
109
|
Balasco L, Pagani M, Pangrazzi L, Chelini G, Viscido F, Chama AGC, Galbusera A, Provenzano G, Gozzi A, Bozzi Y. Somatosensory cortex hyperconnectivity and impaired whisker-dependent responses in Cntnap2 -/- mice. Neurobiol Dis 2022; 169:105742. [PMID: 35483565 DOI: 10.1016/j.nbd.2022.105742] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 04/16/2022] [Accepted: 04/21/2022] [Indexed: 11/16/2022] Open
Abstract
Sensory abnormalities are a common feature in autism spectrum disorders (ASDs). Tactile responsiveness is altered in autistic individuals, with hypo-responsiveness being associated with the severity of ASD core symptoms. Similarly, sensory abnormalities have been described in mice lacking ASD-associated genes. Loss-of-function mutations in CNTNAP2 result in cortical dysplasia-focal epilepsy syndrome (CDFE) and autism. Likewise, Cntnap2-/- mice show epilepsy and deficits relevant with core symptoms of human ASDs, and are considered a reliable model to study ASDs. Altered synaptic transmission and synchronicity found in the cerebral cortex of Cntnap2-/- mice would suggest a network dysfunction. Here, we investigated the neural substrates of whisker-dependent responses in Cntnap2+/+ and Cntnap2-/- adult mice. When compared to controls, Cntnap2-/- mice showed focal hyper-connectivity within the primary somatosensory cortex (S1), in the absence of altered connectivity between S1 and other somatosensory areas. This data suggests the presence of impaired somatosensory processing in these mutants. Accordingly, Cntnap2-/- mice displayed impaired whisker-dependent discrimination in the textured novel object recognition test (tNORT) and increased c-fos mRNA induction within S1 following whisker stimulation. S1 functional hyperconnectivity might underlie the aberrant whisker-dependent responses observed in Cntnap2-/- mice, indicating that Cntnap2 mice are a reliable model to investigate sensory abnormalities that characterize ASDs.
Collapse
Affiliation(s)
- Luigi Balasco
- Center for Mind/Brain Sciences - CIMeC, University of Trento, Piazza della Manifattura 1, 38068 Rovereto, TN, Italy
| | - Marco Pagani
- Functional Neuroimaging Laboratory, Center for Neuroscience and Cognitive Systems, Istituto Italiano di Tecnologia, Corso Bettini 31, 38068 Rovereto, Italy
| | - Luca Pangrazzi
- Center for Mind/Brain Sciences - CIMeC, University of Trento, Piazza della Manifattura 1, 38068 Rovereto, TN, Italy
| | - Gabriele Chelini
- Center for Mind/Brain Sciences - CIMeC, University of Trento, Piazza della Manifattura 1, 38068 Rovereto, TN, Italy
| | - Francesca Viscido
- Center for Mind/Brain Sciences - CIMeC, University of Trento, Piazza della Manifattura 1, 38068 Rovereto, TN, Italy
| | | | - Alberto Galbusera
- Functional Neuroimaging Laboratory, Center for Neuroscience and Cognitive Systems, Istituto Italiano di Tecnologia, Corso Bettini 31, 38068 Rovereto, Italy
| | - Giovanni Provenzano
- Department of Cellular, Computational, and Integrative Biology (CIBIO), University of Trento, via Sommarive 9, 38123 Trento, Italy
| | - Alessandro Gozzi
- Functional Neuroimaging Laboratory, Center for Neuroscience and Cognitive Systems, Istituto Italiano di Tecnologia, Corso Bettini 31, 38068 Rovereto, Italy
| | - Yuri Bozzi
- Center for Mind/Brain Sciences - CIMeC, University of Trento, Piazza della Manifattura 1, 38068 Rovereto, TN, Italy; CNR Neuroscience Institute, via Moruzzi 1, 56124 Pisa, Italy.
| |
Collapse
|
110
|
Molecular Alterations of the Endocannabinoid System in Psychiatric Disorders. Int J Mol Sci 2022; 23:ijms23094764. [PMID: 35563156 PMCID: PMC9104141 DOI: 10.3390/ijms23094764] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 04/22/2022] [Accepted: 04/23/2022] [Indexed: 02/07/2023] Open
Abstract
The therapeutic benefits of the current medications for patients with psychiatric disorders contrast with a great variety of adverse effects. The endocannabinoid system (ECS) components have gained high interest as potential new targets for treating psychiatry diseases because of their neuromodulator role, which is essential to understanding the regulation of many brain functions. This article reviewed the molecular alterations in ECS occurring in different psychiatric conditions. The methods used to identify alterations in the ECS were also described. We used a translational approach. The animal models reproducing some behavioral and/or neurochemical aspects of psychiatric disorders and the molecular alterations in clinical studies in post-mortem brain tissue or peripheral tissues were analyzed. This article reviewed the most relevant ECS changes in prevalent psychiatric diseases such as mood disorders, schizophrenia, autism, attentional deficit, eating disorders (ED), and addiction. The review concludes that clinical research studies are urgently needed for two different purposes: (1) To identify alterations of the ECS components potentially useful as new biomarkers relating to a specific disease or condition, and (2) to design new therapeutic targets based on the specific alterations found to improve the pharmacological treatment in psychiatry.
Collapse
|
111
|
Qi C, Chen A, Mao H, Hu E, Ge J, Ma G, Ren K, Xue Q, Wang W, Wu S. Excitatory and Inhibitory Synaptic Imbalance Caused by Brain-Derived Neurotrophic Factor Deficits During Development in a Valproic Acid Mouse Model of Autism. Front Mol Neurosci 2022; 15:860275. [PMID: 35465089 PMCID: PMC9019547 DOI: 10.3389/fnmol.2022.860275] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Accepted: 03/11/2022] [Indexed: 11/13/2022] Open
Abstract
Environmental factors, such as medication during pregnancy, are one of the major causes of autism spectrum disorder (ASD). Valproic acid (VPA) intake during pregnancy has been reported to dramatically elevate autism risk in offspring. Recently, researchers have proposed that VPA exposure could induce excitatory or inhibitory synaptic dysfunction. However, it remains to be determined whether and how alterations in the excitatory/inhibitory (E/I) balance contribute to VPA-induced ASD in a mouse model. In the present study, we explored changes in the E/I balance during different developmental periods in a VPA mouse model. We found that typical markers of pre- and postsynaptic excitatory and inhibitory function involved in E/I balance markedly decreased during development, reflecting difficulties in the development of synaptic plasticity in VPA-exposed mice. The expression of brain-derived neurotrophic factor (BDNF), a neurotrophin that promotes the formation and maturation of glutamatergic and GABAergic synapses during postnatal development, was severely reduced in the VPA-exposed group. Treatment with exogenous BDNF during the critical E/I imbalance period rescued synaptic functions and autism-like behaviors, such as social defects. With these results, we experimentally showed that social dysfunction in the VPA mouse model of autism might be caused by E/I imbalance stemming from BDNF deficits during the developmental stage.
Collapse
Affiliation(s)
- Chuchu Qi
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi’an, China
| | - Andi Chen
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi’an, China
| | - Honghui Mao
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi’an, China
| | - Erling Hu
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi’an, China
| | - Junye Ge
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi’an, China
- MOE Key Laboratory of Modern Teaching Technology, Center for Teacher Professional Ability Development, Shaanxi Normal University, Xi’an, China
| | - Guaiguai Ma
- Department of Physiology, Medical College of Yan’an University, Yan’an, China
| | - Keke Ren
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi’an, China
| | - Qian Xue
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi’an, China
| | - Wenting Wang
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi’an, China
- *Correspondence: Wenting Wang,
| | - Shengxi Wu
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi’an, China
- Shengxi Wu,
| |
Collapse
|
112
|
Siper PM, Rowe MA, Guillory SB, Rouhandeh AA, George-Jones JL, Tavassoli T, Lurie S, Zweifach J, Weissman J, Foss-Feig J, Halpern D, Trelles MP, Mulhern MS, Brittenham C, Gordon J, Zemon V, Buxbaum JD, Kolevzon A. Visual Evoked Potential Abnormalities in Phelan-McDermid Syndrome. J Am Acad Child Adolesc Psychiatry 2022; 61:565-574.e1. [PMID: 34303785 PMCID: PMC8782912 DOI: 10.1016/j.jaac.2021.07.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 06/28/2021] [Accepted: 07/15/2021] [Indexed: 10/20/2022]
Abstract
OBJECTIVE The current study used visual evoked potentials (VEPs) to examine excitatory and inhibitory postsynaptic activity in children with Phelan-McDermid syndrome (PMS) and the association with genetic factors. PMS is caused by haploinsufficiency of SHANK3 on chromosome 22 and represents a common single-gene cause of autism spectrum disorder (ASD) and intellectual disability. METHOD Transient VEPs were obtained from 175 children, including 31 with PMS, 79 with idiopathic ASD, 45 typically developing controls, and 20 unaffected siblings of children with PMS. Stimuli included standard and short-duration contrast-reversing checkerboard conditions, and the reliability between these 2 conditions was assessed. Test-retest reliability and correlations with deletion size were explored in the group with PMS. RESULTS Children with PMS and, to a lesser extent, those with idiopathic ASD displayed significantly smaller amplitudes and decreased beta and gamma band activity relative to TD controls and PMS siblings. Across groups, high intraclass correlation coefficients were obtained between standard and short-duration conditions. In children with PMS, test-retest reliability was strong. Deletion size was significantly correlated with P60-N75 amplitude for both conditions. CONCLUSION Children with PMS displayed distinct transient VEP waveform abnormalities in both time and frequency domains that might reflect underlying glutamatergic deficits that were associated with deletion size. A similar response pattern was observed in a subset of children with idiopathic ASD. VEPs offer a noninvasive measure of excitatory and inhibitory neurotransmission that holds promise for stratification and surrogate endpoints in ongoing clinical trials in PMS and ASD.
Collapse
|
113
|
Montanari M, Martella G, Bonsi P, Meringolo M. Autism Spectrum Disorder: Focus on Glutamatergic Neurotransmission. Int J Mol Sci 2022; 23:ijms23073861. [PMID: 35409220 PMCID: PMC8998955 DOI: 10.3390/ijms23073861] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 03/24/2022] [Accepted: 03/29/2022] [Indexed: 12/16/2022] Open
Abstract
Disturbances in the glutamatergic system have been increasingly documented in several neuropsychiatric disorders, including autism spectrum disorder (ASD). Glutamate-centered theories of ASD are based on evidence from patient samples and postmortem studies, as well as from studies documenting abnormalities in glutamatergic gene expression and metabolic pathways, including changes in the gut microbiota glutamate metabolism in patients with ASD. In addition, preclinical studies on animal models have demonstrated glutamatergic neurotransmission deficits and altered expression of glutamate synaptic proteins. At present, there are no approved glutamatergic drugs for ASD, but several ongoing clinical trials are currently focusing on evaluating in autistic patients glutamatergic pharmaceuticals already approved for other conditions. In this review, we provide an overview of the literature concerning the role of glutamatergic neurotransmission in the pathophysiology of ASD and as a potential target for novel treatments.
Collapse
Affiliation(s)
- Martina Montanari
- Laboratory of Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, 00179 Rome, Italy; (M.M.); (G.M.)
- Department of Systems Neuroscience, University Tor Vergata, 00133 Rome, Italy
| | - Giuseppina Martella
- Laboratory of Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, 00179 Rome, Italy; (M.M.); (G.M.)
| | - Paola Bonsi
- Laboratory of Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, 00179 Rome, Italy; (M.M.); (G.M.)
- Correspondence: (P.B.); (M.M.)
| | - Maria Meringolo
- Laboratory of Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, 00179 Rome, Italy; (M.M.); (G.M.)
- Correspondence: (P.B.); (M.M.)
| |
Collapse
|
114
|
Jiang S, Xiao L, Sun Y, He M, Gao C, Zhu C, Chang H, Ding J, Li W, Wang Y, Sun T, Wang F. The GABAB receptor agonist STX209 reverses the autism‑like behaviour in an animal model of autism induced by prenatal exposure to valproic acid. Mol Med Rep 2022; 25:154. [PMID: 35244195 PMCID: PMC8941376 DOI: 10.3892/mmr.2022.12670] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 02/16/2022] [Indexed: 11/06/2022] Open
Abstract
Autism spectrum disorder (ASD) is a lifelong neurodevelopmental condition characterized by impaired social interaction, compromised communication, and restrictive or stereotyped behaviours and interests. Due to the complex pathophysiology of ASD, there are currently no available medical therapies for improving the associated social deficits. Consequently, the present study investigated the effects of STX209, a selective γ‑aminobutyric acid type B receptor (GABABR2) agonist, on an environmental rodent model of autism. The mouse model of autism induced by prenatal exposure to valproic acid (VPA) was used to assess the therapeutic potential of STX209 on autism‑like behaviour in the present study. This study investigated the effects of STX209 on VPA model mice via behavioral testing and revealed a significant reversal of core/associated autism‑like behavior, including sociability and preference for social novelty, novelty recognition, locomotion and exploration activity and marble‑burying deficit. This may be associated with STX209 correcting dendritic arborization, spine density and GABABR2 expression in hippocampus of VPA model mice. However, expression of glutamic acid decarboxylase 65/67 in the hippocampus were not altered by STX209. The present results demonstrated that STX209 administration ameliorated autism‑like symptoms in mice exposed to VPA prenatally, suggesting that autism‑like symptoms in children with a history of prenatal VPA exposure may also benefit from treatment with the GABABR2 agonist STX209.
Collapse
Affiliation(s)
- Shucai Jiang
- Ningxia Key Laboratory of Craniocerebral Disease, Incubation Base of National Key Laboratory, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Lifei Xiao
- Ningxia Key Laboratory of Craniocerebral Disease, Incubation Base of National Key Laboratory, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Yu Sun
- Ningxia Key Laboratory of Craniocerebral Disease, Incubation Base of National Key Laboratory, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Maotao He
- Department of Diagnostic Pathology, School of Basic Medical Sciences, Weifang Medical University, Weifang, Shandong 261053, P.R. China
| | - Caibin Gao
- Ningxia Key Laboratory of Craniocerebral Disease, Incubation Base of National Key Laboratory, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Changliang Zhu
- Ningxia Key Laboratory of Craniocerebral Disease, Incubation Base of National Key Laboratory, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Haigang Chang
- Ningxia Key Laboratory of Craniocerebral Disease, Incubation Base of National Key Laboratory, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Jiangwei Ding
- Ningxia Key Laboratory of Craniocerebral Disease, Incubation Base of National Key Laboratory, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Wenchao Li
- Ningxia Key Laboratory of Craniocerebral Disease, Incubation Base of National Key Laboratory, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Yangyang Wang
- Ningxia Key Laboratory of Craniocerebral Disease, Incubation Base of National Key Laboratory, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Tao Sun
- Ningxia Key Laboratory of Craniocerebral Disease, Incubation Base of National Key Laboratory, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Feng Wang
- Department of Neurosurgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, P.R. China
| |
Collapse
|
115
|
Zosen D, Austdal LPE, Bjørnstad S, Lumor JS, Paulsen RE. Antiepileptic drugs lamotrigine and valproate differentially affect neuronal maturation in the developing chick embryo, yet with PAX6 as a potential common mediator. Neurotoxicol Teratol 2022; 90:107057. [DOI: 10.1016/j.ntt.2021.107057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 12/14/2021] [Accepted: 12/14/2021] [Indexed: 10/19/2022]
|
116
|
Park G, Jeon SJ, Ko IO, Park JH, Lee KC, Kim MS, Shin CY, Kim H, Lee YS. Decreased in vivo glutamate/GABA ratio correlates with the social behavior deficit in a mouse model of autism spectrum disorder. Mol Brain 2022; 15:19. [PMID: 35183218 PMCID: PMC8858545 DOI: 10.1186/s13041-022-00904-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 02/08/2022] [Indexed: 12/29/2022] Open
Abstract
To diagnose autism spectrum disorder (ASD), researchers have sought biomarkers whose alterations correlate with the susceptibility to ASD. However, biomarkers closely related to the pathophysiology of ASD are lacking. Even though excitation/inhibition (E/I) imbalance has been suggested as an underlying mechanism of ASD, few studies have investigated the actual ratio of glutamate (Glu) to γ-aminobutyric acid (GABA) concentration in vivo. Moreover, there are controversies in the directions of E/I ratio alterations even in extensively studied ASD animal models. Here, using proton magnetic resonance spectroscopy (1H-MRS) at 9.4T, we found significant differences in the levels of different metabolites or their ratios in the prefrontal cortex and hippocampus of Cntnap2−/− mice compared to their wild-type littermates. The Glu/GABA ratio, N-acetylaspartate (NAA)/total creatine (tCr) ratio, and tCr level in the prefrontal cortex were significantly different in Cntnap2−/− mice compared to those in wild-type mice, and they significantly correlated with the sociability of mice. Moreover, receiver operating characteristic (ROC) analyses indicated high specificity and selectivity of these metabolites in discriminating genotypes. These results suggest that the lowered Glu/GABA ratio in the prefrontal cortex along with the changes in the other metabolites might contribute to the social behavior deficit in Cntnap2−/− mice. Our results also demonstrate the utility of 1H-MRS in investigating the underlying mechanisms or the diagnosis of ASD.
Collapse
|
117
|
Cooke J, Molloy CJ, Cáceres ASJ, Dinneen T, Bourgeron T, Murphy D, Gallagher L, Loth E. The Synaptic Gene Study: Design and Methodology to Identify Neurocognitive Markers in Phelan-McDermid Syndrome and NRXN1 Deletions. Front Neurosci 2022; 16:806990. [PMID: 35250452 PMCID: PMC8894872 DOI: 10.3389/fnins.2022.806990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 01/26/2022] [Indexed: 11/26/2022] Open
Abstract
Synaptic gene conditions, i.e., “synaptopathies,” involve disruption to genes expressed at the synapse and account for between 0.5 and 2% of autism cases. They provide a unique entry point to understanding the molecular and biological mechanisms underpinning autism-related phenotypes. Phelan-McDermid Syndrome (PMS, also known as 22q13 deletion syndrome) and NRXN1 deletions (NRXN1ds) are two synaptopathies associated with autism and related neurodevelopmental disorders (NDDs). PMS often incorporates disruption to the SHANK3 gene, implicated in excitatory postsynaptic scaffolding, whereas the NRXN1 gene encodes neurexin-1, a presynaptic cell adhesion protein; both are implicated in trans-synaptic signaling in the brain. Around 70% of individuals with PMS and 43–70% of those with NRXN1ds receive a diagnosis of autism, suggesting that alterations in synaptic development may play a crucial role in explaining the aetiology of autism. However, a substantial amount of heterogeneity exists between conditions. Most individuals with PMS have moderate to profound intellectual disability (ID), while those with NRXN1ds have no ID to severe ID. Speech abnormalities are common to both, although appear more severe in PMS. Very little is currently known about the neurocognitive underpinnings of phenotypic presentations in PMS and NRXN1ds. The Synaptic Gene (SynaG) study adopts a gene-first approach and comprehensively assesses these two syndromic forms of autism. The study compliments preclinical efforts within AIMS-2-TRIALS focused on SHANK3 and NRXN1. The aims of the study are to (1) establish the frequency of autism diagnosis and features in individuals with PMS and NRXN1ds, (2) to compare the clinical profile of PMS, NRXN1ds, and individuals with ‘idiopathic’ autism (iASD), (3) to identify mechanistic biomarkers that may account for autistic features and/or heterogeneity in clinical profiles, and (4) investigate the impact of second or multiple genetic hits on heterogeneity in clinical profiles. In the current paper we describe our methodology for phenotyping the sample and our planned comparisons, with information on the necessary adaptations made during the global COVID-19 pandemic. We also describe the demographics of the data collected thus far, including 25 PMS, 36 NRXN1ds, 33 iASD, and 52 NTD participants, and present an interim analysis of autistic features and adaptive functioning.
Collapse
Affiliation(s)
- Jennifer Cooke
- Forensic and Neurodevelopmental Sciences, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, United Kingdom
- *Correspondence: Jennifer Cooke,
| | - Ciara J. Molloy
- Department of Psychiatry, School of Medicine, Trinity College Dublin, Dublin, Ireland
| | - Antonia San José Cáceres
- Forensic and Neurodevelopmental Sciences, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, United Kingdom
- Fundación para la Investigación Biomédica del Hospital General Universitario Gregorio Marañón, Madrid, Spain
- Biomedical Research Networking Center for Mental Health Network (CIBERSAM), Madrid, Spain
| | - Thomas Dinneen
- Department of Psychiatry, School of Medicine, Trinity College Dublin, Dublin, Ireland
| | | | - Declan Murphy
- Forensic and Neurodevelopmental Sciences, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, United Kingdom
| | - Louise Gallagher
- Department of Psychiatry, School of Medicine, Trinity College Dublin, Dublin, Ireland
| | - Eva Loth
- Forensic and Neurodevelopmental Sciences, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, United Kingdom
- Eva Loth,
| |
Collapse
|
118
|
Sun C, Zhao Z, Cheng L, Tian R, Zhao W, Du J, Zhang Y, Wang C. Effect of Transcranial Direct Current Stimulation on the Mismatch Negativity Features of Deviated Stimuli in Children With Autism Spectrum Disorder. Front Neurosci 2022; 16:721987. [PMID: 35221894 PMCID: PMC8863939 DOI: 10.3389/fnins.2022.721987] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 01/19/2022] [Indexed: 11/13/2022] Open
Abstract
Autism spectrum disorder (ASD) is a devastating mental disorder in children. Currently, there is no effective treatment for ASD. Transcranial direct current stimulation (tDCS), which is a non-invasive brain stimulation neuromodulation technology, is a promising method for the treatment of ASD. However, the manner in which tDCS changes the electrophysiological process in the brain is still unclear. In this study, we used tDCS to stimulate the dorsolateral prefrontal cortex area of children with ASD (one group received anode tDCS, and the other received sham tDCS) and investigated the changes in evoked EEG signals and behavioral abilities before and after anode and sham stimulations. In addition to tDCS, all patients received conventional rehabilitation treatment. Results show that although conventional treatment can effectively improve the behavioral ability of children with ASD, the use of anode tDCS with conventional rehabilitation can boost this improvement, thus leading to increased treatment efficacy. By analyzing the electroencephalography pre- and post-treatment, we noticed a decrease in the mismatch negativity (MMN) latency and an increase in the MMN amplitude in both groups, these features are considered similar to MMN features from healthy children. However, no statistical difference between the two groups was observed after 4 weeks of treatment. In addition, the MMN features correlate well with the aberrant behavior checklist (ABC) scale, particularly the amplitude of MMN, thus suggesting the feasibility of using MMN features to assess the behavioral ability of children with ASD.
Collapse
Affiliation(s)
- Changcheng Sun
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China
- Department of Rehabilitation Medical, Tianjin Union Medical Centre, Tianjin, China
| | - Zhuoyue Zhao
- Department of Rehabilitation Medical, Tianjin Union Medical Centre, Tianjin, China
| | - Longlong Cheng
- China Electronics Cloud Brain (Tianjin) Technology Co., Ltd., Tianjin, China
| | - Rong Tian
- Department of Rehabilitation Medical, Tianjin Union Medical Centre, Tianjin, China
| | - Wenchang Zhao
- Department of Rehabilitation Medical, Tianjin Union Medical Centre, Tianjin, China
| | - Jingang Du
- Department of Rehabilitation Medical, Tianjin Union Medical Centre, Tianjin, China
| | - Ying Zhang
- Department of Rehabilitation Medical, Tianjin Union Medical Centre, Tianjin, China
| | - Chunfang Wang
- Department of Rehabilitation Medical, Tianjin Union Medical Centre, Tianjin, China
| |
Collapse
|
119
|
Sun YY, Chen WJ, Huang ZP, Yang G, Wu ML, Xu DE, Yang WL, Luo YC, Xiao ZC, Xu RX, Ma QH. TRIM32 Deficiency Impairs the Generation of Pyramidal Neurons in Developing Cerebral Cortex. Cells 2022; 11:cells11030449. [PMID: 35159260 PMCID: PMC8834167 DOI: 10.3390/cells11030449] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 01/20/2022] [Accepted: 01/25/2022] [Indexed: 02/01/2023] Open
Abstract
Excitatory-inhibitory imbalance (E/I) is a fundamental mechanism underlying autism spectrum disorders (ASD). TRIM32 is a risk gene genetically associated with ASD. The absence of TRIM32 causes impaired generation of inhibitory GABAergic interneurons, neural network hyperexcitability, and autism-like behavior in mice, emphasizing the role of TRIM32 in maintaining E/I balance, but despite the description of TRIM32 in regulating proliferation and differentiation of cultured mouse neural progenitor cells (NPCs), the role of TRIM32 in cerebral cortical development, particularly in the production of excitatory pyramidal neurons, remains unknown. The present study observed that TRIM32 deficiency resulted in decreased numbers of distinct layer-specific cortical neurons and decreased radial glial cell (RGC) and intermediate progenitor cell (IPC) pool size. We further demonstrated that TRIM32 deficiency impairs self-renewal of RGCs and IPCs as indicated by decreased proliferation and mitosis. A TRIM32 deficiency also affects or influences the formation of cortical neurons. As a result, TRIM32-deficient mice showed smaller brain size. At the molecular level, RNAseq analysis indicated reduced Notch signalling in TRIM32-deficient mice. Therefore, the present study indicates a role for TRIM32 in pyramidal neuron generation. Impaired generation of excitatory pyramidal neurons may explain the hyperexcitability observed in TRIM32-deficient mice.
Collapse
Affiliation(s)
- Yan-Yun Sun
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou 215123, China; (Y.-Y.S.); (Z.-P.H.); (M.-L.W.)
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, Institute of Neuroscience, Soochow University, Suzhou 215123, China
| | - Wen-Jin Chen
- Department of Neurosurgery, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu 610072, China;
| | - Ze-Ping Huang
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou 215123, China; (Y.-Y.S.); (Z.-P.H.); (M.-L.W.)
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, Institute of Neuroscience, Soochow University, Suzhou 215123, China
| | - Gang Yang
- Lab Center, Medical College of Soochow University, Suzhou 215123, China;
| | - Ming-Lei Wu
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou 215123, China; (Y.-Y.S.); (Z.-P.H.); (M.-L.W.)
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, Institute of Neuroscience, Soochow University, Suzhou 215123, China
| | - De-En Xu
- Wuxi No. 2 People’s Hospital, Wuxi 214001, China;
| | - Wu-Lin Yang
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China;
- Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei 230031, China
| | - Yong-Chun Luo
- Department of Neurosurgery, First Medical Center of Chinese PLA General Hospital, Beijing 100028, China;
| | - Zhi-Cheng Xiao
- Department of Anatomy and Developmental Biology, Monash University, Clayton 3800, Australia;
| | - Ru-Xiang Xu
- Department of Neurosurgery, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu 610072, China;
- Correspondence: (Q.-H.M.); (R.-X.X.)
| | - Quan-Hong Ma
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou 215123, China; (Y.-Y.S.); (Z.-P.H.); (M.-L.W.)
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, Institute of Neuroscience, Soochow University, Suzhou 215123, China
- Correspondence: (Q.-H.M.); (R.-X.X.)
| |
Collapse
|
120
|
Gu J, Guo M, Yin X, Huang C, Qian L, Zhou L, Wang Z, Wang L, Shi L, Ji G. A systematic comparison of neurotoxicity of bisphenol A and its derivatives in zebrafish. THE SCIENCE OF THE TOTAL ENVIRONMENT 2022; 805:150210. [PMID: 34534871 DOI: 10.1016/j.scitotenv.2021.150210] [Citation(s) in RCA: 63] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 08/31/2021] [Accepted: 09/03/2021] [Indexed: 06/13/2023]
Abstract
As more and more countries have prohibited the manufacture and sale of plastic products with bisphenol A (BPA), a number of bisphenol analogues (BPs), including BPS, BPF and BPAF, have gradually been used as its primary substitutes. Ideally, substitutes used to replace chemicals with environmental risks should be inert, so it makes sense that the risk of the similar chemical substitutes (BPS, BPF, and BPAF) should be assessed before they used. Therefore, in the present study, the neurotoxicity of four BPs at environmentally relevant concentration (200 μg/L) were systematically compared using zebrafish as a model. Our results showed that the four BPs (BPA, BPS, BPF and BPAF) exhibited no obvious effect on the hatchability, survival rate and body length of zebrafish larvae, noteworthily a significant inhibitory effect on spontaneous movement at 24 hpf was observed in the BPA, BPF and BPAF treatment groups. Behavioral tests showed that BPAF, BPF and BPA exposure significantly reduced the locomotor activity of the larvae. Additionally, BPAF treatment adversely affected motor neuron axon length in transgenic lines hb9-GFP zebrafish and decreased central nervous system (CNS) neurogenesis in transgenic lines HuC-GFP zebrafish. Intriguingly, BPAF displayed the strongest effects on the levels and metabolism of neurotransmitters, followed by BPF and BPA, while BPS showed the weakest effects on neurotransmitters. In conclusion, our study deciphered that environmentally relevant concentrations of BPs exposure exhibited differential degrees of neurotoxicity, which ranked as below: BPAF > BPF ≈ BPA > BPS. The possible mechanisms can be partially ascribed to the dramatical changes of multiple neurotransmitters and the inhibitory effects on neuronal development. These results suggest that BPAF and BPF should be carefully considered as alternatives to BPA.
Collapse
Affiliation(s)
- Jie Gu
- Nanjing Institute of Environmental Sciences, Ministry of Ecology and Environment, Nanjing 210042, China
| | - Min Guo
- Nanjing Institute of Environmental Sciences, Ministry of Ecology and Environment, Nanjing 210042, China
| | - Xiaogang Yin
- Co-Innovation Center for Efficient Processing and Utilization of Forest Resources, College of Chemical Engineering, Nanjing Forestry University, Longpan Road 159, Nanjing 210037, China
| | - Caoxing Huang
- Co-Innovation Center for Efficient Processing and Utilization of Forest Resources, College of Chemical Engineering, Nanjing Forestry University, Longpan Road 159, Nanjing 210037, China
| | - Lingling Qian
- Co-Innovation Center for Efficient Processing and Utilization of Forest Resources, College of Chemical Engineering, Nanjing Forestry University, Longpan Road 159, Nanjing 210037, China
| | - Linjun Zhou
- Nanjing Institute of Environmental Sciences, Ministry of Ecology and Environment, Nanjing 210042, China
| | - Zhen Wang
- Nanjing Institute of Environmental Sciences, Ministry of Ecology and Environment, Nanjing 210042, China
| | - Lei Wang
- Nanjing Institute of Environmental Sciences, Ministry of Ecology and Environment, Nanjing 210042, China
| | - Lili Shi
- Nanjing Institute of Environmental Sciences, Ministry of Ecology and Environment, Nanjing 210042, China
| | - Guixiang Ji
- Nanjing Institute of Environmental Sciences, Ministry of Ecology and Environment, Nanjing 210042, China.
| |
Collapse
|
121
|
Cherubini E, Di Cristo G, Avoli M. Dysregulation of GABAergic Signaling in Neurodevelomental Disorders: Targeting Cation-Chloride Co-transporters to Re-establish a Proper E/I Balance. Front Cell Neurosci 2022; 15:813441. [PMID: 35069119 PMCID: PMC8766311 DOI: 10.3389/fncel.2021.813441] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 11/30/2021] [Indexed: 01/01/2023] Open
Abstract
The construction of the brain relies on a series of well-defined genetically and experience- or activity -dependent mechanisms which allow to adapt to the external environment. Disruption of these processes leads to neurological and psychiatric disorders, which in many cases are manifest already early in postnatal life. GABA, the main inhibitory neurotransmitter in the adult brain is one of the major players in the early assembly and formation of neuronal circuits. In the prenatal and immediate postnatal period GABA, acting on GABAA receptors, depolarizes and excites targeted cells via an outwardly directed flux of chloride. In this way it activates NMDA receptors and voltage-dependent calcium channels contributing, through intracellular calcium rise, to shape neuronal activity and to establish, through the formation of new synapses and elimination of others, adult neuronal circuits. The direction of GABAA-mediated neurotransmission (depolarizing or hyperpolarizing) depends on the intracellular levels of chloride [Cl−]i, which in turn are maintained by the activity of the cation-chloride importer and exporter KCC2 and NKCC1, respectively. Thus, the premature hyperpolarizing action of GABA or its persistent depolarizing effect beyond the postnatal period, leads to behavioral deficits associated with morphological alterations and an excitatory (E)/inhibitory (I) imbalance in selective brain areas. The aim of this review is to summarize recent data concerning the functional role of GABAergic transmission in building up and refining neuronal circuits early in development and its dysfunction in neurodevelopmental disorders such as Autism Spectrum Disorders (ASDs), schizophrenia and epilepsy. In particular, we focus on novel information concerning the mechanisms by which alterations in cation-chloride co-transporters (CCC) generate behavioral and cognitive impairment in these diseases. We discuss also the possibility to re-establish a proper GABAA-mediated neurotransmission and excitatory (E)/inhibitory (I) balance within selective brain areas acting on CCC.
Collapse
Affiliation(s)
- Enrico Cherubini
- European Brain Research Institute (EBRI)-Rita Levi-Montalcini, Roma, Italy
- *Correspondence: Enrico Cherubini
| | - Graziella Di Cristo
- Neurosciences Department, Université de Montréal and CHU Sainte-Justine Research Center, Montreal, QC, Canada
| | - Massimo Avoli
- Montreal Neurological Institute-Hospital and Departments of Neurology and Neurosurgery and of Physiology, McGill University, Montreal, QC, Canada
| |
Collapse
|
122
|
Neurobiology of ARID1B haploinsufficiency related to neurodevelopmental and psychiatric disorders. Mol Psychiatry 2022; 27:476-489. [PMID: 33686214 PMCID: PMC8423853 DOI: 10.1038/s41380-021-01060-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 02/04/2021] [Accepted: 02/18/2021] [Indexed: 01/31/2023]
Abstract
ARID1B haploinsufficiency is a frequent cause of intellectual disability (ID) and autism spectrum disorder (ASD), and also leads to emotional disturbances. In this review, we examine past and present clinical and preclinical research into the neurobiological function of ARID1B. The presentation of ARID1B-related disorders (ARID1B-RD) is highly heterogeneous, including varying degrees of ID, ASD, and physical features. Recent research includes the development of suitable clinical readiness assessments for the treatment of ARID1B-RD, as well as similar neurodevelopmental disorders. Recently developed mouse models of Arid1b haploinsufficiency successfully mirror many of the behavioral phenotypes of ASD and ID. These animal models have helped to solidify the molecular mechanisms by which ARID1B regulates brain development and function, including epigenetic regulation of the Pvalb gene and promotion of Wnt/β-catenin signaling in neural progenitors in the ventral telencephalon. Finally, preclinical studies have identified the use of a positive allosteric modulator of the GABAA receptor as an effective treatment for some Arid1b haploinsufficiency-related behavioral phenotypes, and there is potential for the refinement of this therapy in order to translate it into clinical use.
Collapse
|
123
|
Yan Z, Rein B. Mechanisms of synaptic transmission dysregulation in the prefrontal cortex: pathophysiological implications. Mol Psychiatry 2022; 27:445-465. [PMID: 33875802 PMCID: PMC8523584 DOI: 10.1038/s41380-021-01092-3] [Citation(s) in RCA: 113] [Impact Index Per Article: 56.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Revised: 03/13/2021] [Accepted: 03/29/2021] [Indexed: 02/02/2023]
Abstract
The prefrontal cortex (PFC) serves as the chief executive officer of the brain, controlling the highest level cognitive and emotional processes. Its local circuits among glutamatergic principal neurons and GABAergic interneurons, as well as its long-range connections with other brain regions, have been functionally linked to specific behaviors, ranging from working memory to reward seeking. The efficacy of synaptic signaling in the PFC network is profundedly influenced by monoaminergic inputs via the activation of dopamine, adrenergic, or serotonin receptors. Stress hormones and neuropeptides also exert complex effects on the synaptic structure and function of PFC neurons. Dysregulation of PFC synaptic transmission is strongly linked to social deficits, affective disturbance, and memory loss in brain disorders, including autism, schizophrenia, depression, and Alzheimer's disease. Critical neural circuits, biological pathways, and molecular players that go awry in these mental illnesses have been revealed by integrated electrophysiological, optogenetic, biochemical, and transcriptomic studies of PFC. Novel epigenetic mechanism-based strategies are proposed as potential avenues of therapeutic intervention for PFC-involved diseases. This review provides an overview of PFC network organization and synaptic modulation, as well as the mechanisms linking PFC dysfunction to the pathophysiology of neurodevelopmental, neuropsychiatric, and neurodegenerative diseases. Insights from the preclinical studies offer the potential for discovering new medical treatments for human patients with these brain disorders.
Collapse
Affiliation(s)
- Zhen Yan
- Department of Physiology and Biophysics, State University of New York at Buffalo, Jacobs School of Medicine and Biomedical Sciences, Buffalo, NY, USA.
| | | |
Collapse
|
124
|
Jiang S, He M, Xiao L, Sun Y, Ding J, Li W, Guo B, Wang L, Wang Y, Gao C, Sun T, Wang F. Prenatal GABAB Receptor Agonist Administration Corrects the Inheritance of Autism-Like Core Behaviors in Offspring of Mice Prenatally Exposed to Valproic Acid. Front Psychiatry 2022; 13:835993. [PMID: 35492716 PMCID: PMC9051083 DOI: 10.3389/fpsyt.2022.835993] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 03/07/2022] [Indexed: 12/21/2022] Open
Abstract
This study was performed to evaluate the effects of prenatal baclofen (a GABAB receptor agonist) treatment on the inheritance of autism-like behaviors in valproic acid (VPA)-exposed mice. VPA model mice (first generation, F1) that were prenatally exposed to VPA exhibited robust core autism-like behaviors, and we found that oral administration of baclofen to F1 mice corrected their autism-like behavioral phenotypes at an early age. Based on a previous epigenetics study, we mated the F1 male offspring with litter females to produce the second generation (F2). The F2 male mice showed obvious inheritance of autism-like phenotypes from F1 mice, implying the heritability of autism symptoms in patients with prenatal VPA exposure. Furthermore, we found prenatal baclofen administration was associated with beneficial effects on the autism-like phenotype in F2 male mice. This may have involved corrections in the density of total/mature dendritic spines in the hippocampus (HC) and medial prefrontal cortex (mPFC), normalizing synaptic plasticity. In this research, GABAB receptor agonist administration corrected the core autism-like behaviors of F1 mice and protected against the inheritance of neurodevelopmental disorders in the offspring of F1 mice, suggesting the potential of early intervention with GABAB receptor agonists in the treatment of neurodevelopmental disorders.
Collapse
Affiliation(s)
- Shucai Jiang
- Ningxia Key Laboratory of Craniocerebral Disease, Incubation Base of National Key Laboratory, Ningxia Medical University, Yinchuan, China
| | - Maotao He
- School of Basic Medical Sciences, Weifang Medical University, Weifang, China
| | - Lifei Xiao
- Ningxia Key Laboratory of Craniocerebral Disease, Incubation Base of National Key Laboratory, Ningxia Medical University, Yinchuan, China
| | - Yu Sun
- Ningxia Key Laboratory of Craniocerebral Disease, Incubation Base of National Key Laboratory, Ningxia Medical University, Yinchuan, China
| | - Jiangwei Ding
- Ningxia Key Laboratory of Craniocerebral Disease, Incubation Base of National Key Laboratory, Ningxia Medical University, Yinchuan, China
| | - Wenchao Li
- Ningxia Key Laboratory of Craniocerebral Disease, Incubation Base of National Key Laboratory, Ningxia Medical University, Yinchuan, China
| | - Baorui Guo
- Ningxia Key Laboratory of Craniocerebral Disease, Incubation Base of National Key Laboratory, Ningxia Medical University, Yinchuan, China
| | - Lei Wang
- Ningxia Key Laboratory of Craniocerebral Disease, Incubation Base of National Key Laboratory, Ningxia Medical University, Yinchuan, China
| | - Yangyang Wang
- Ningxia Key Laboratory of Craniocerebral Disease, Incubation Base of National Key Laboratory, Ningxia Medical University, Yinchuan, China
| | - Caibin Gao
- Ningxia Key Laboratory of Craniocerebral Disease, Incubation Base of National Key Laboratory, Ningxia Medical University, Yinchuan, China
| | - Tao Sun
- Ningxia Key Laboratory of Craniocerebral Disease, Incubation Base of National Key Laboratory, Ningxia Medical University, Yinchuan, China
| | - Feng Wang
- Ningxia Key Laboratory of Craniocerebral Disease, Incubation Base of National Key Laboratory, Ningxia Medical University, Yinchuan, China.,The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
125
|
Canitano R, Palumbi R. Excitation/Inhibition Modulators in Autism Spectrum Disorder: Current Clinical Research. Front Neurosci 2021; 15:753274. [PMID: 34916897 PMCID: PMC8669810 DOI: 10.3389/fnins.2021.753274] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 11/08/2021] [Indexed: 11/13/2022] Open
Abstract
Autism spectrum disorder (ASD) is a group of neurodevelopmental disorders characterized by social and communication abnormalities. Heterogeneity in the expression and severity of the core and associated symptoms poses difficulties in classification and the overall clinical approach. Synaptic abnormalities have been observed in preclinical ASD models. They are thought to play a major role in clinical functional abnormalities and might be modified by targeted interventions. An imbalance in excitatory to inhibitory neurotransmission (E/I imbalance), through altered glutamatergic and GABAergic neurotransmission, respectively, is thought to be implicated in the pathogenesis of ASD. Glutamatergic and GABAergic agents have been tested in clinical trials with encouraging results as to efficacy and tolerability. Further studies are needed to confirm the role of E/I modulators in the treatment of ASD and on the safety and efficacy of the current agents.
Collapse
Affiliation(s)
- Roberto Canitano
- Division of Child and Adolescent Neuropsychiatry, University Hospital of Siena, Siena, Italy
| | - Roberto Palumbi
- Division of Child and Adolescent Neuropsychiatry, Basic Medical Sciences, Neuroscience and Sense Organs Department, University Hospital of Bari, Bari, Italy
| |
Collapse
|
126
|
Cui J, Park J, Ju X, Lee Y, Hong B, Ahn J, Kim YH, Ko Y, Yoon SH, Lim C, Lee SY, Huh SO, Heo JY, Chung W. General Anesthesia During Neurodevelopment Reduces Autistic Behavior in Adult BTBR Mice, a Murine Model of Autism. Front Cell Neurosci 2021; 15:772047. [PMID: 34912193 PMCID: PMC8667765 DOI: 10.3389/fncel.2021.772047] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 10/20/2021] [Indexed: 12/30/2022] Open
Abstract
Preclinical studies suggest that repeated exposure to anesthetics during a critical period of neurodevelopment induces long-term changes in synaptic transmission, plasticity, and behavior. Such changes are of great concern, as similar changes have also been identified in animal models of neurodevelopmental disorders (NDDs) such as autism. Because of overlapping synaptic changes, it is also possible that anesthetic exposures have a more significant effect in individuals diagnosed with NDDs. Thus, we evaluated the effects of early, multiple anesthetic exposures in BTBR mice, an inbred strain that displays autistic behavior. We discovered that three cycles of sevoflurane anesthesia (2.5%, 1 h) with 2-h intervals between each exposure in late postnatal BTBR mice did not aggravate, but instead improved pathophysiological mechanisms involved with autistic behavior. Sevoflurane exposures restored E/I balance (by increasing inhibitory synaptic transmission), and increased mitochondrial respiration and BDNF signaling in BTBR mice. Most importantly, such changes were associated with reduced autistic behavior in BTBR mice, as sociability was increased in the three-chamber test and repetitive behavior was reduced in the self-grooming test. Our results suggest that anesthetic exposures during neurodevelopment may affect individuals diagnosed with NDDs differently.
Collapse
Affiliation(s)
- Jianchen Cui
- Department of Medical Science, Chungnam National University School of Medicine, Daejeon, South Korea.,Department of Biochemistry, Chungnam National University School of Medicine, Daejeon, South Korea.,Infection Control Convergence Research Center, Chungnam National University School of Medicine, Daejeon, South Korea.,Department of Anesthesiology, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, China.,Brain Korea 21 FOUR Project for Medical Science, Chungnam National University, Daejeon, South Korea
| | - Jiho Park
- Department of Anesthesiology and Pain Medicine, Chungnam National University Hospital, Daejeon, South Korea
| | - Xianshu Ju
- Department of Medical Science, Chungnam National University School of Medicine, Daejeon, South Korea.,Department of Biochemistry, Chungnam National University School of Medicine, Daejeon, South Korea.,Infection Control Convergence Research Center, Chungnam National University School of Medicine, Daejeon, South Korea
| | - Yulim Lee
- Department of Medical Science, Chungnam National University School of Medicine, Daejeon, South Korea.,Department of Biochemistry, Chungnam National University School of Medicine, Daejeon, South Korea.,Infection Control Convergence Research Center, Chungnam National University School of Medicine, Daejeon, South Korea.,Brain Korea 21 FOUR Project for Medical Science, Chungnam National University, Daejeon, South Korea
| | - Boohwi Hong
- Department of Anesthesiology and Pain Medicine, Chungnam National University Hospital, Daejeon, South Korea.,Department of Anesthesiology and Pain Medicine, Chungnam National University School of Medicine, Daejeon, South Korea
| | - Jeonghoon Ahn
- Department of Anesthesiology and Pain Medicine, Chungnam National University Hospital, Daejeon, South Korea
| | - Yoon Hee Kim
- Department of Anesthesiology and Pain Medicine, Chungnam National University Hospital, Daejeon, South Korea.,Department of Anesthesiology and Pain Medicine, Chungnam National University School of Medicine, Daejeon, South Korea
| | - Youngkwon Ko
- Department of Anesthesiology and Pain Medicine, Chungnam National University Hospital, Daejeon, South Korea.,Department of Anesthesiology and Pain Medicine, Chungnam National University School of Medicine, Daejeon, South Korea
| | - Seok-Hwa Yoon
- Department of Anesthesiology and Pain Medicine, Chungnam National University Hospital, Daejeon, South Korea.,Department of Anesthesiology and Pain Medicine, Chungnam National University School of Medicine, Daejeon, South Korea
| | - Chaeseong Lim
- Department of Anesthesiology and Pain Medicine, Chungnam National University Hospital, Daejeon, South Korea.,Department of Anesthesiology and Pain Medicine, Chungnam National University School of Medicine, Daejeon, South Korea
| | - Sun Yeul Lee
- Department of Anesthesiology and Pain Medicine, Chungnam National University Hospital, Daejeon, South Korea.,Department of Anesthesiology and Pain Medicine, Chungnam National University School of Medicine, Daejeon, South Korea
| | - Sung-Oh Huh
- Department of Pharmacology, College of Medicine, Institute of Natural Medicine, Hallym University, Chuncheon, South Korea
| | - Jun Young Heo
- Department of Medical Science, Chungnam National University School of Medicine, Daejeon, South Korea.,Department of Biochemistry, Chungnam National University School of Medicine, Daejeon, South Korea.,Brain Korea 21 FOUR Project for Medical Science, Chungnam National University, Daejeon, South Korea
| | - Woosuk Chung
- Department of Medical Science, Chungnam National University School of Medicine, Daejeon, South Korea.,Brain Korea 21 FOUR Project for Medical Science, Chungnam National University, Daejeon, South Korea.,Department of Anesthesiology and Pain Medicine, Chungnam National University Hospital, Daejeon, South Korea.,Department of Anesthesiology and Pain Medicine, Chungnam National University School of Medicine, Daejeon, South Korea
| |
Collapse
|
127
|
Lutz AK, Pérez Arévalo A, Ioannidis V, Stirmlinger N, Demestre M, Delorme R, Bourgeron T, Boeckers TM. SHANK2 Mutations Result in Dysregulation of the ERK1/2 Pathway in Human Induced Pluripotent Stem Cells-Derived Neurons and Shank2(-/-) Mice. Front Mol Neurosci 2021; 14:773571. [PMID: 34899182 PMCID: PMC8662699 DOI: 10.3389/fnmol.2021.773571] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Accepted: 10/07/2021] [Indexed: 12/28/2022] Open
Abstract
SHANK2 (ProSAP1) is a postsynaptic scaffolding protein of excitatory synapses in the central nervous system and implicated in the development of autism spectrum disorders (ASD). Patients with mutations in SHANK2 show autism-like behaviors, developmental delay, and intellectual disability. We generated human induced pluripotent stem cells (hiPSC) from a patient carrying a heterozygous deletion of SHANK2 and from the unaffected parents. In patient hiPSCs and derived neurons SHANK2 mRNA and protein expression was reduced. During neuronal maturation, a reduction in growth cone size and a transient increase in neuronal soma size were observed. Neuronal proliferation was increased, and apoptosis was decreased in young and mature neurons. Additionally, mature patient hiPSC-derived neurons showed dysregulated excitatory signaling and a decrease of a broad range of signaling molecules of the ERK-MAP kinase pathway. These findings could be confirmed in brain samples from Shank2(−/−) mice, which also showed decreased mGluR5 and phospho-ERK1/2 expression. Our study broadens the current knowledge of SHANK2-related ASD. We highlight the importance of excitatory-inhibitory balance and mGluR5 dysregulation with disturbed downstream ERK1/2 signaling in ASD, which provides possible future therapeutic strategies for SHANK2-related ASD.
Collapse
Affiliation(s)
- Anne-Kathrin Lutz
- Institute for Anatomy and Cell Biology, Ulm University, Ulm, Germany
| | | | | | | | - Maria Demestre
- Institute for Anatomy and Cell Biology, Ulm University, Ulm, Germany
| | | | - Thomas Bourgeron
- Génétique Humaine et Fonctions Cognitives, Institut Pasteur, Université Paris Diderot, Paris, France
| | - Tobias M Boeckers
- Institute for Anatomy and Cell Biology, Ulm University, Ulm, Germany.,Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Ulm Site, Ulm, Germany
| |
Collapse
|
128
|
Rais M, Lovelace JW, Shuai XS, Woodard W, Bishay S, Estrada L, Sharma AR, Nguy A, Kulinich A, Pirbhoy PS, Palacios AR, Nelson DL, Razak KA, Ethell IM. Functional consequences of postnatal interventions in a mouse model of Fragile X syndrome. Neurobiol Dis 2021; 162:105577. [PMID: 34871737 DOI: 10.1016/j.nbd.2021.105577] [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: 07/03/2021] [Revised: 10/22/2021] [Accepted: 12/02/2021] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND Fragile X syndrome (FXS) is a leading genetic cause of autism and intellectual disability with cortical hyperexcitability and sensory hypersensitivity attributed to loss and hypofunction of inhibitory parvalbumin-expressing (PV) cells. Our studies provide novel insights into the role of excitatory neurons in abnormal development of PV cells during a postnatal period of inhibitory circuit refinement. METHODS To achieve Fragile X mental retardation gene (Fmr1) deletion and re-expression in excitatory neurons during the postnatal day (P)14-P21 period, we generated CreCaMKIIa/Fmr1Flox/y (cOFF) and CreCaMKIIa/Fmr1FloxNeo/y (cON) mice, respectively. Cortical phenotypes were evaluated in adult mice using biochemical, cellular, clinically relevant electroencephalogram (EEG) and behavioral tests. RESULTS We found that similar to global Fmr1 KO mice, the density of PV-expressing cells, their activation, and sound-evoked gamma synchronization were impaired in cOFF mice, but the phenotypes were improved in cON mice. cOFF mice also showed enhanced cortical gelatinase activity and baseline EEG gamma power, which were reduced in cON mice. In addition, TrkB phosphorylation and PV levels were lower in cOFF mice, which also showed increased locomotor activity and anxiety-like behaviors. Remarkably, when FMRP levels were restored in only excitatory neurons during the P14-P21 period, TrkB phosphorylation and mouse behaviors were also improved. CONCLUSIONS These results indicate that postnatal deletion or re-expression of FMRP in excitatory neurons is sufficient to elicit or ameliorate structural and functional cortical deficits, and abnormal behaviors in mice, informing future studies about appropriate treatment windows and providing fundamental insights into the cellular mechanisms of cortical circuit dysfunction in FXS.
Collapse
Affiliation(s)
- Maham Rais
- Division of Biomedical Sciences and Biomedical Sciences Graduate Program, School of Medicine, University of California Riverside, Riverside, CA 92521, USA
| | - Jonathan W Lovelace
- Department of Psychology, University of California Riverside, Riverside, CA 92521, USA
| | - Xinghao S Shuai
- Division of Biomedical Sciences and Biomedical Sciences Graduate Program, School of Medicine, University of California Riverside, Riverside, CA 92521, USA
| | - Walker Woodard
- Division of Biomedical Sciences and Biomedical Sciences Graduate Program, School of Medicine, University of California Riverside, Riverside, CA 92521, USA
| | - Steven Bishay
- Division of Biomedical Sciences and Biomedical Sciences Graduate Program, School of Medicine, University of California Riverside, Riverside, CA 92521, USA
| | - Leo Estrada
- Division of Biomedical Sciences and Biomedical Sciences Graduate Program, School of Medicine, University of California Riverside, Riverside, CA 92521, USA
| | - Ashwin R Sharma
- Division of Biomedical Sciences and Biomedical Sciences Graduate Program, School of Medicine, University of California Riverside, Riverside, CA 92521, USA
| | - Austin Nguy
- Division of Biomedical Sciences and Biomedical Sciences Graduate Program, School of Medicine, University of California Riverside, Riverside, CA 92521, USA
| | - Anna Kulinich
- Division of Biomedical Sciences and Biomedical Sciences Graduate Program, School of Medicine, University of California Riverside, Riverside, CA 92521, USA
| | - Patricia S Pirbhoy
- Division of Biomedical Sciences and Biomedical Sciences Graduate Program, School of Medicine, University of California Riverside, Riverside, CA 92521, USA
| | - Arnold R Palacios
- Division of Biomedical Sciences and Biomedical Sciences Graduate Program, School of Medicine, University of California Riverside, Riverside, CA 92521, USA
| | | | - Khaleel A Razak
- Department of Psychology, University of California Riverside, Riverside, CA 92521, USA; Neuroscience Graduate Program, University of California Riverside, Riverside, CA 92521, USA
| | - Iryna M Ethell
- Division of Biomedical Sciences and Biomedical Sciences Graduate Program, School of Medicine, University of California Riverside, Riverside, CA 92521, USA; Neuroscience Graduate Program, University of California Riverside, Riverside, CA 92521, USA.
| |
Collapse
|
129
|
Reversing frontal disinhibition rescues behavioural deficits in models of CACNA1A-associated neurodevelopment disorders. Mol Psychiatry 2021; 26:7225-7246. [PMID: 34127816 DOI: 10.1038/s41380-021-01175-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 04/27/2021] [Accepted: 05/12/2021] [Indexed: 12/20/2022]
Abstract
CACNA1A deletions cause epilepsy, ataxia, and a range of neurocognitive deficits, including inattention, impulsivity, intellectual deficiency and autism. To investigate the underlying mechanisms, we generated mice carrying a targeted Cacna1a deletion restricted to parvalbumin-expressing (PV) neurons (PVCre;Cacna1ac/+) or to cortical pyramidal cells (PC) (Emx1Cre;Cacna1ac/+). GABA release from PV-expressing GABAergic interneurons (PV-INs) is reduced in PVCre;Cacna1ac/+ mutants, resulting in impulsivity, cognitive rigidity and inattention. By contrast, the deletion of Cacna1a in PCs does not impact cortical excitability or behaviour in Emx1Cre;Cacna1ac/+ mutants. A targeted Cacna1a deletion in the orbitofrontal cortex (OFC) results in reversal learning deficits while a medial prefrontal cortex (mPFC) deletion impairs selective attention. These deficits can be rescued by the selective chemogenetic activation of cortical PV-INs in the OFC or mPFC of PVCre;Cacna1ac/+ mutants. Thus, Cacna1a haploinsufficiency disrupts perisomatic inhibition in frontal cortical circuits, leading to a range of potentially reversible neurocognitive deficits.
Collapse
|
130
|
Leithead AB, Tasker JG, Harony‐Nicolas H. The interplay between glutamatergic circuits and oxytocin neurons in the hypothalamus and its relevance to neurodevelopmental disorders. J Neuroendocrinol 2021; 33:e13061. [PMID: 34786775 PMCID: PMC8951898 DOI: 10.1111/jne.13061] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 10/14/2021] [Accepted: 10/30/2021] [Indexed: 11/27/2022]
Abstract
Oxytocin (OXT) neurons of the hypothalamus are at the center of several physiological functions, including milk ejection, uterus contraction, and maternal and social behavior. In lactating females, OXT neurons show a pattern of burst firing and inter-neuron synchronization during suckling that leads to pulsatile release of surges of OXT into the bloodstream to stimulate milk ejection. This pattern of firing and population synchronization may be facilitated in part by hypothalamic glutamatergic circuits, as has been observed in vitro using brain slices obtained from male rats and neonates. However, it remains unknown how hypothalamic glutamatergic circuits influence OXT cell activity outside the context of lactation. In this review, we summarize the in vivo and in vitro studies that describe the synchronized burst firing pattern of OXT neurons and the implication of hypothalamic glutamate in this pattern of firing. We also make note of the few studies that have traced glutamatergic afferents to the hypothalamic paraventricular and supraoptic nuclei. Finally, we discuss the genetic findings implicating several glutamatergic genes in neurodevelopmental disorders, including autism spectrum disorder, thus underscoring the need for future studies to investigate the impact of these mutations on hypothalamic glutamatergic circuits and the OXT system.
Collapse
Affiliation(s)
- Amanda B. Leithead
- Department of PsychiatryIcahn School of Medicine at Mount SinaiNew YorkNYUSA
- Seaver Autism Center for Research and TreatmentNew YorkNYUSA
- Department of NeuroscienceIcahn School of Medicine at Mount SinaiNew YorkNYUSA
- Friedman Brain Institute at the Icahn School of Medicine at Mount SinaiNew YorkNYUSA
| | - Jeffrey G. Tasker
- Neurobiology DivisionDepartment of Cell and Molecular BiologyTulane UniversityNew OrleansLAUSA
| | - Hala Harony‐Nicolas
- Department of PsychiatryIcahn School of Medicine at Mount SinaiNew YorkNYUSA
- Seaver Autism Center for Research and TreatmentNew YorkNYUSA
- Department of NeuroscienceIcahn School of Medicine at Mount SinaiNew YorkNYUSA
- Friedman Brain Institute at the Icahn School of Medicine at Mount SinaiNew YorkNYUSA
- Mindich Child Health and Development Institute at the Icahn School of Medicine at Mount SinaiNew YorkNYUSA
| |
Collapse
|
131
|
Naderi M, Puar P, JavadiEsfahani R, Kwong RWM. Early developmental exposure to bisphenol A and bisphenol S disrupts socio-cognitive function, isotocin equilibrium, and excitation-inhibition balance in developing zebrafish. Neurotoxicology 2021; 88:144-154. [PMID: 34808222 DOI: 10.1016/j.neuro.2021.11.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 10/26/2021] [Accepted: 11/16/2021] [Indexed: 12/01/2022]
Abstract
Dysregulation of the oxytocinergic system and excitation/inhibition (E/I) balance in synaptic transmission and neural circuits are common hallmarks of various neurodevelopmental disorders. Several experimental and epidemiological studies have shown that perinatal exposure to endocrine-disrupting chemicals bisphenol A (BPA) and bisphenol S (BPS) may contribute to a range of childhood neurodevelopmental disorders. However, the effects of BPA and BPS on social-cognitive development and the associated mechanisms remain largely unknown. In this study, we explored the impacts of early developmental exposure (2hpf-5dpf) to environmentally relevant concentrations of BPA, and its analog BPS (0.001, 0.01, and 0.1 μM), on anxiety, social behaviors, and memory performance in 21 dpf zebrafish larvae. Our results revealed that early-life exposure to low concentrations of BPA and BPS elevated anxiety-like behavior, while fish exposed to higher concentrations of these chemicals displayed social deficits and impaired object recognition memory. Additionally, we found that co-exposure with an aromatase inhibitor antagonized BPA- and BPS-induced effects on anxiety levels and social behaviors, while the co-exposure to an estrogen receptor antagonist restored recognition memory in zebrafish larvae. These results indicate that BPA and BPS may affect social-cognitive function through distinct mechanisms. On the other hand, exposure to low BPA/BPS concentrations increased both the mRNA and protein levels of isotocin (zebrafish oxytocin) in the zebrafish brain, whereas a reduction in its mRNA level was observed at higher concentrations. Further, alterations in the transcript abundance of chloride transporters, and molecular markers of gamma-aminobutyric acid (GABA) and glutamatergic systems, were observed in the zebrafish brain, suggesting possible E/I imbalance following BPA or BPS exposure. Collectively, the results of this study demonstrate that early-life exposure to low concentrations of the environmental contaminants BPA and BPS can interfere with the isotocinergic signaling pathway and disrupts the establishment of E/I balance in the developing brain, subsequently leading to the onset of a suite of behavioral deficits and neurodevelopmental disorders.
Collapse
Affiliation(s)
- Mohammad Naderi
- Department of Biology, York University, Toronto, ON, M3J 1P3, Canada.
| | - Pankaj Puar
- Department of Biology, York University, Toronto, ON, M3J 1P3, Canada
| | | | - Raymond W M Kwong
- Department of Biology, York University, Toronto, ON, M3J 1P3, Canada
| |
Collapse
|
132
|
Significance of GABA A Receptor for Cognitive Function and Hippocampal Pathology. Int J Mol Sci 2021; 22:ijms222212456. [PMID: 34830337 PMCID: PMC8623595 DOI: 10.3390/ijms222212456] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 11/08/2021] [Accepted: 11/08/2021] [Indexed: 02/05/2023] Open
Abstract
The hippocampus is a primary area for contextual memory, known to process spatiotemporal information within a specific episode. Long-term strengthening of glutamatergic transmission is a mechanism of contextual learning in the dorsal cornu ammonis 1 (CA1) area of the hippocampus. CA1-specific immobilization or blockade of α-amino-3-hydroxyl-5-methyl-4-isoxazole-propionate (AMPA) receptor delivery can impair learning performance, indicating a causal relationship between learning and receptor delivery into the synapse. Moreover, contextual learning also strengthens GABAA (gamma-aminobutyric acid) receptor-mediated inhibitory synapses onto CA1 neurons. Recently we revealed that strengthening of GABAA receptor-mediated inhibitory synapses preceded excitatory synaptic plasticity after contextual learning, resulting in a reduced synaptic excitatory/inhibitory (E/I) input balance that returned to pretraining levels within 10 min. The faster plasticity at inhibitory synapses may allow encoding a contextual memory and prevent cognitive dysfunction in various hippocampal pathologies. In this review, we focus on the dynamic changes of GABAA receptor mediated-synaptic currents after contextual learning and the intracellular mechanism underlying rapid inhibitory synaptic plasticity. In addition, we discuss that several pathologies, such as Alzheimer’s disease, autism spectrum disorders and epilepsy are characterized by alterations in GABAA receptor trafficking, synaptic E/I imbalance and neuronal excitability.
Collapse
|
133
|
Deficiency of autism risk factor ASH1L in prefrontal cortex induces epigenetic aberrations and seizures. Nat Commun 2021; 12:6589. [PMID: 34782621 PMCID: PMC8593046 DOI: 10.1038/s41467-021-26972-8] [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: 06/10/2021] [Accepted: 10/29/2021] [Indexed: 12/30/2022] Open
Abstract
ASH1L, a histone methyltransferase, is identified as a top-ranking risk factor for autism spectrum disorder (ASD), however, little is known about the biological mechanisms underlying the link of ASH1L haploinsufficiency to ASD. Here we show that ASH1L expression and H3K4me3 level are significantly decreased in the prefrontal cortex (PFC) of postmortem tissues from ASD patients. Knockdown of Ash1L in PFC of juvenile mice induces the downregulation of risk genes associated with ASD, intellectual disability (ID) and epilepsy. These downregulated genes are enriched in excitatory and inhibitory synaptic function and have decreased H3K4me3 occupancy at their promoters. Furthermore, Ash1L deficiency in PFC causes the diminished GABAergic inhibition, enhanced glutamatergic transmission, and elevated PFC pyramidal neuronal excitability, which is associated with severe seizures and early mortality. Chemogenetic inhibition of PFC pyramidal neuronal activity, combined with the administration of GABA enhancer diazepam, rescues PFC synaptic imbalance and seizures, but not autistic social deficits or anxiety-like behaviors. These results have revealed the critical role of ASH1L in regulating synaptic gene expression and seizures, which provides insights into treatment strategies for ASH1L-associated brain diseases.
Collapse
|
134
|
The potential roles of excitatory-inhibitory imbalances and the repressor element-1 silencing transcription factor in aging and aging-associated diseases. Mol Cell Neurosci 2021; 117:103683. [PMID: 34775008 DOI: 10.1016/j.mcn.2021.103683] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 11/02/2021] [Accepted: 11/06/2021] [Indexed: 12/28/2022] Open
Abstract
Disruptions to the central excitatory-inhibitory (E/I) balance are thought to be related to aging and underlie a host of neural pathologies, including Alzheimer's disease. Aging may induce an increase in excitatory signaling, causing an E/I imbalance, which has been linked to shorter lifespans in mice, flies, and worms. In humans, extended longevity correlates to greater repression of genes involved in excitatory neurotransmission. The repressor element-1 silencing transcription factor (REST) is a master regulator in neural cells and is believed to be upregulated with senescent stimuli, whereupon it counters hyperexcitability, insulin/insulin-like signaling pathway activity, oxidative stress, and neurodegeneration. This review examines the putative mechanisms that distort the E/I balance with aging and neurodegeneration, and the putative roles of REST in maintaining neuronal homeostasis.
Collapse
|
135
|
Paterno R, Marafiga JR, Ramsay H, Li T, Salvati KA, Baraban SC. Hippocampal gamma and sharp-wave ripple oscillations are altered in a Cntnap2 mouse model of autism spectrum disorder. Cell Rep 2021; 37:109970. [PMID: 34758298 PMCID: PMC8783641 DOI: 10.1016/j.celrep.2021.109970] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 09/02/2021] [Accepted: 10/19/2021] [Indexed: 01/02/2023] Open
Abstract
Impaired synaptic neurotransmission may underly circuit alterations contributing to behavioral autism spectrum disorder (ASD) phenotypes. A critical component of impairments reported in somatosensory and prefrontal cortex of ASD mouse models are parvalbumin (PV)-expressing fast-spiking interneurons. However, it remains unknown whether PV interneurons mediating hippocampal networks crucial to navigation and memory processing are similarly impaired. Using PV-labeled transgenic mice, a battery of behavioral assays, in vitro patch-clamp electrophysiology, and in vivo 32-channel silicon probe local field potential recordings, we address this question in a Cntnap2-null mutant mouse model representing a human ASD risk factor gene. Cntnap2-/- mice show a reduction in hippocampal PV interneuron density, reduced inhibitory input to CA1 pyramidal cells, deficits in spatial discrimination ability, and frequency-dependent circuit changes within the hippocampus, including alterations in gamma oscillations, sharp-wave ripples, and theta-gamma modulation. Our findings highlight hippocampal involvement in ASD and implicate interneurons as a potential therapeutical target.
Collapse
Affiliation(s)
- Rosalia Paterno
- Department of Neurological Surgery and Weill Institute of Neuroscience, University of California, San Francisco, CA 94143, USA.
| | - Joseane Righes Marafiga
- Neurophysiology and Neurochemistry of Neuronal Excitability and Synaptic Plasticity Laboratory, Graduate Program in Biological Sciences: Biochemistry, Department of Biochemistry, ICBS, Universidade Federal do Rio Grande do Sul, Porto Alegre, 90035-003, Brazil
| | - Harrison Ramsay
- Department of Neurological Surgery and Weill Institute of Neuroscience, University of California, San Francisco, CA 94143, USA
| | - Tina Li
- Department of Neurological Surgery and Weill Institute of Neuroscience, University of California, San Francisco, CA 94143, USA
| | - Kathryn A Salvati
- Department of Neurological Surgery and Weill Institute of Neuroscience, University of California, San Francisco, CA 94143, USA
| | - Scott C Baraban
- Department of Neurological Surgery and Weill Institute of Neuroscience, University of California, San Francisco, CA 94143, USA
| |
Collapse
|
136
|
Adil KJ, Gonzales EL, Remonde CG, Boo KJ, Jeon SJ, Shin CY. Autism-Like Behavioral Phenotypes in Mice Treated with Systemic N-Methyl-D-Aspartate. Biomol Ther (Seoul) 2021; 30:232-237. [PMID: 34702791 PMCID: PMC9047488 DOI: 10.4062/biomolther.2021.133] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 09/15/2021] [Accepted: 09/17/2021] [Indexed: 11/05/2022] Open
Abstract
Autism spectrum disorder (ASD) having core characteristics of social interaction problems and repetitive behaviors and interests affects individuals at varying degrees and comorbidities, making it difficult to determine the precise etiology underlying the symptoms. Given its heterogeneity, ASD is difficult to treat and the development of therapeutics is slow due to the scarcity of animal models that are easy to produce and screen with. Based on the theory of excitation/inhibition imbalance in the brain with ASD which involves glutamatergic and/or GABAergic neurotransmission, a pharmacologic agent to modulate these receptors might be a good starting point for modeling. N-methyl-D-aspartic acid (NMDA) is an amino acid derivative acting as a specific agonist at the NMDA receptor and therefore imitates the action of the neurotransmitter glutamate on that receptor. In contrast to glutamate, NMDA selectively binds to and regulates the NMDA receptor, but not other glutamate receptors such as AMPA and kainite receptors. Given this role, we aimed to determine whether NMDA administration could result in autistic-like behavior in adolescent mice. Both male and female mice were treated with saline or NMDA (50 and 75 mg/kg) and were tested on various behavior experiments. Interestingly, acute NMDA-treated mice showed social deficits and repetitive behavior similar to ASD phenotypes. These results support the excitation/inhibition imbalance theory of ASD and that NMDA injection can be used as a pharmacologic model of ASD-like behaviors.
Collapse
Affiliation(s)
- Keremkleroo Jym Adil
- School of Medicine and Center for Neuroscience Research, Konkuk University, Seoul 05029, Republic of Korea
| | - Edson Luck Gonzales
- School of Medicine and Center for Neuroscience Research, Konkuk University, Seoul 05029, Republic of Korea
| | - Chilly Gay Remonde
- School of Medicine and Center for Neuroscience Research, Konkuk University, Seoul 05029, Republic of Korea
| | - Kyung-Jun Boo
- School of Medicine and Center for Neuroscience Research, Konkuk University, Seoul 05029, Republic of Korea
| | - Se Jin Jeon
- School of Medicine and Center for Neuroscience Research, Konkuk University, Seoul 05029, Republic of Korea
| | - Chan Young Shin
- School of Medicine and Center for Neuroscience Research, Konkuk University, Seoul 05029, Republic of Korea
| |
Collapse
|
137
|
Nomura T. Interneuron Dysfunction and Inhibitory Deficits in Autism and Fragile X Syndrome. Cells 2021; 10:cells10102610. [PMID: 34685590 PMCID: PMC8534049 DOI: 10.3390/cells10102610] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 09/27/2021] [Accepted: 09/28/2021] [Indexed: 01/18/2023] Open
Abstract
The alteration of excitatory–inhibitory (E–I) balance has been implicated in various neurological and psychiatric diseases, including autism spectrum disorder (ASD). Fragile X syndrome (FXS) is a single-gene disorder that is the most common known cause of ASD. Understanding the molecular and physiological features of FXS is thought to enhance our knowledge of the pathophysiology of ASD. Accumulated evidence implicates deficits in the inhibitory circuits in FXS that tips E–I balance toward excitation. Deficits in interneurons, the main source of an inhibitory neurotransmitter, gamma-aminobutyric acid (GABA), have been reported in FXS, including a reduced number of cells, reduction in intrinsic cellular excitability, or weaker synaptic connectivity. Manipulating the interneuron activity ameliorated the symptoms in the FXS mouse model, which makes it reasonable to conceptualize FXS as an interneuronopathy. While it is still poorly understood how the developmental profiles of the inhibitory circuit go awry in FXS, recent works have uncovered several developmental alterations in the functional properties of interneurons. Correcting disrupted E–I balance by potentiating the inhibitory circuit by targeting interneurons may have a therapeutic potential in FXS. I will review the recent evidence about the inhibitory alterations and interneuron dysfunction in ASD and FXS and will discuss the future directions of this field.
Collapse
Affiliation(s)
- Toshihiro Nomura
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| |
Collapse
|
138
|
Mizusaki BEP, O'Donnell C. Neural circuit function redundancy in brain disorders. Curr Opin Neurobiol 2021; 70:74-80. [PMID: 34416675 PMCID: PMC8694099 DOI: 10.1016/j.conb.2021.07.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 06/17/2021] [Accepted: 07/14/2021] [Indexed: 02/07/2023]
Abstract
Redundancy is a ubiquitous property of the nervous system. This means that vastly different configurations of cellular and synaptic components can enable the same neural circuit functions. However, until recently, very little brain disorder research has considered the implications of this characteristic when designing experiments or interpreting data. Here, we first summarise the evidence for redundancy in healthy brains, explaining redundancy and three related sub-concepts: sloppiness, dependencies and multiple solutions. We then lay out key implications for brain disorder research, covering recent examples of redundancy effects in experimental studies on psychiatric disorders. Finally, we give predictions for future experiments based on these concepts.
Collapse
Affiliation(s)
- Beatriz E P Mizusaki
- Computational Neuroscience Unit, School of Computer Science, Electrical and Electronic Engineering, and Engineering Mathematics, University of Bristol, BS8 1UB, United Kingdom
| | - Cian O'Donnell
- Computational Neuroscience Unit, School of Computer Science, Electrical and Electronic Engineering, and Engineering Mathematics, University of Bristol, BS8 1UB, United Kingdom.
| |
Collapse
|
139
|
Dong H, Wang B, Feng J, Yue X, Jia F. Correlation Between Serum Concentrations of Menaquinone-4 and Developmental Quotients in Children With Autism Spectrum Disorder. Front Nutr 2021; 8:748513. [PMID: 34660670 PMCID: PMC8514626 DOI: 10.3389/fnut.2021.748513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 09/06/2021] [Indexed: 11/13/2022] Open
Abstract
Objective: The vitamin K family has a wide range of effects in the body, including the central nervous system. Menaquinone-4 (MK-4), a form of vitamin K2, is converted from phylloquinone (PK), which is the main source of dietary vitamin K and is the main form of vitamin K in the brain. We conducted this study to investigate the serum concentration of MK-4 and the correlations between MK-4 and developmental quotients in children with autism spectrum disorder (ASD). Methods: We selected 731 children with ASD who were diagnosed for the first time. During the same period, 332 neurotypical children who underwent regular physical examinations in our outpatient department were selected as the TD group. We investigated the general situation of children, including gender and age. Children in ASD group were assessed for autistic symptoms and development quotients, including Autism Behavior Checklist (ABC), Childhood Autism Rating Scale (CARS), ADOS-2, and Griffiths Development Scales-Chinese Language Edition (GDS-C). Both groups of children were tested for serum menaquinone-4. We compared serum menaquinone-4 levels of ASD group and TD group. We then conducted a correlation analysis between the level of menaquinone-4 and the developmental quotient of children with ASD. Results: The results of this study indicate that the serum concentration of MK-4 in children with ASD is lower than that in children with typical development (t = -2.702, P = 0.007). The serum concentration of MK-4 is related to the developmental quotients of several subscales in ASD children, and this correlation is more obvious in males. Conclusion: we conclude that MK-4 is present in lower concentrations in children with ASD, which may affect cognition and developmental quotients. The role of MK-4 in ASD needs to be further explored.
Collapse
Affiliation(s)
| | | | | | | | - Feiyong Jia
- Department of Developmental and Behavioral Pediatrics, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
140
|
High Behavioral Variability Mediated by Altered Neuronal Excitability in auts2 Mutant Zebrafish. eNeuro 2021; 8:ENEURO.0493-20.2021. [PMID: 34544758 PMCID: PMC8503961 DOI: 10.1523/eneuro.0493-20.2021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 08/25/2021] [Accepted: 08/27/2021] [Indexed: 01/28/2023] Open
Abstract
Autism spectrum disorders (ASDs) are characterized by abnormal behavioral traits arising from neural circuit dysfunction. While a number of genes have been implicated in ASDs, in most cases, a clear understanding of how mutations in these genes lead to circuit dysfunction and behavioral abnormality is absent. The autism susceptibility candidate 2 (AUTS2) gene is one such gene, associated with ASDs, intellectual disability and a range of other neurodevelopmental conditions. However, the role of AUTS2 in neural development and circuit function is not at all known. Here, we undertook functional analysis of Auts2a, the main homolog of AUTS2 in zebrafish, in the context of the escape behavior. Escape behavior in wild-type zebrafish is critical for survival and is therefore, reliable, rapid, and has well-defined kinematic properties. auts2a mutant zebrafish are viable, have normal gross morphology and can generate escape behavior with normal kinematics. However, the behavior is unreliable and delayed, with high trial-to-trial variability in the latency. Using calcium imaging we probed the activity of Mauthner neurons during otic vesicle (OV) stimulation and observed lower probability of activation and reduced calcium transients in the mutants. With direct activation of Mauthner by antidromic stimulation, the threshold for activation in mutants was higher than that in wild-type, even when inhibition was blocked. Taken together, these results point to reduced excitability of Mauthner neurons in auts2a mutant larvae leading to unreliable escape responses. Our results show a novel role for Auts2a in regulating neural excitability and reliability of behavior.
Collapse
|
141
|
Kissoondoyal A, Rai-Bhogal R, Crawford DA. Abnormal dendritic morphology in the cerebellum of cyclooxygenase-2 - knockin mice. Eur J Neurosci 2021; 54:6355-6373. [PMID: 34510613 DOI: 10.1111/ejn.15454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Accepted: 09/02/2021] [Indexed: 11/28/2022]
Abstract
Prostaglandin E2 (PGE2) is a bioactive signalling molecule metabolized from the phospholipid membranes by the enzymatic activity of cycloxygenase-2 (COX-2). In the developing brain, COX-2 constitutively regulates the production of PGE2, which is important in neuronal development. However, abnormal COX-2/PGE2 signalling has been linked to neurodevelopmental disorders including autism spectrum disorders (ASDs). We have previously demonstrated that COX-2- -KI mice show autism-related behaviours including social deficits, repetitive behaviours and anxious behaviours. COX-2-deficient mice also have deficits in pathways involved in synaptic transmission and dendritic spine formation. In this study, we use a Golgi-COX staining method to examine sex-dependent differences in dendritic and dendritic spine morphology in neurons of COX-2- -KI mice cerebellum compared with wild-type (WT) matched controls at postnatal day 25 (P25). We show that COX-2- -KI mice have increased dendritic arborization closer to the cell soma and increased dendritic looping. We also observed a sex-dependent effect of the COX-2- -KI on dendritic thickness, dendritic spine density, dendritic spine morphology, and the expression of β-actin and the actin-binding protein spinophilin. Our findings show that changes in COX-2/PGE2 signalling lead to impaired morphology of dendrites and dendritic spines in a sex-dependant manner and may contribute the pathology of the cerebellum seen in individuals with ASD. This study provides further evidence that the COX-2- -KI mouse model can be used to study a subset of ASD pathologies.
Collapse
Affiliation(s)
- Ashby Kissoondoyal
- School of Kinesiology and Health Science, York University, Toronto, Ontario, Canada.,Neuroscience Graduate Diploma Program, York University, Toronto, Ontario, Canada
| | - Ravneet Rai-Bhogal
- Neuroscience Graduate Diploma Program, York University, Toronto, Ontario, Canada.,Department of Biology, York University, Toronto, Ontario, Canada
| | - Dorota A Crawford
- School of Kinesiology and Health Science, York University, Toronto, Ontario, Canada.,Neuroscience Graduate Diploma Program, York University, Toronto, Ontario, Canada.,Department of Biology, York University, Toronto, Ontario, Canada
| |
Collapse
|
142
|
Soorya LV, Fogg L, Ocampo E, Printen M, Youngkin S, Halpern D, Kolevzon A, Lee S, Grodberg D, Anagnostou E. Neurocognitive Outcomes from Memantine: A Pilot, Double-Blind, Placebo-Controlled Trial in Children with Autism Spectrum Disorder. J Child Adolesc Psychopharmacol 2021; 31:475-484. [PMID: 34543081 DOI: 10.1089/cap.2021.0010] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Objective: Studies interrogating therapeutics which alter the excitation-inhibition balance in the treatment of autism spectrum disorder (ASD) have reported mixed results on social and behavioral outcomes. Methods: The aim of this randomized, double-blind placebo-controlled pilot trial was to evaluate neurocognitive effects of memantine over a 24-week trial. Twenty-three children ages 6-12 years old with ASD were randomized to memantine or placebo. Primary outcomes included measures of apraxia and expressive language with evaluations at midpoint (week 12) and endpoint (week 24). Secondary outcomes included memory and adaptive behavior measures. Exploratory outcomes included changes in overall cognitive functioning and behavior (e.g., Aberrant Behavior Checklist). Results: Results suggest that memantine was well-tolerated. Dropout rates were high across groups with only 14 participants completing the 6-month trial. Memantine was not associated with improvements in apraxia and expressive language. Treatment with memantine was associated with improvements in verbal recognition memory as measured by the Narrative Memory-Recognition (NEPSY-II) (F = 5.05, p = .03). In addition, exploratory analyses of changes in Intelligence quotient (IQ) suggest improvements on verbal IQ (d = 1.8). Conclusions: Results suggest future studies of memantine in ASD may benefit from shifting treatment targets from social and behavioral outcomes to exploration of effects of memantine on cognition, potentially as an adjunct to learning and educational interventions. ClinicalTrials.gov: NCT01372449.
Collapse
Affiliation(s)
- Latha Valluripalli Soorya
- Department of Psychiatry and Behavioral Sciences, Rush University Medical Center, Chicago, Illinois, USA
| | - Louis Fogg
- Department of Nursing, Rush University Medical Center, Chicago, Illinois, USA
| | - Edith Ocampo
- Department of Psychiatry and Behavioral Sciences, Rush University Medical Center, Chicago, Illinois, USA
| | - Madison Printen
- Department of Psychiatry and Behavioral Sciences, Rush University Medical Center, Chicago, Illinois, USA
| | - Sarah Youngkin
- Department of Psychiatry and Behavioral Sciences, Rush University Medical Center, Chicago, Illinois, USA
| | - Danielle Halpern
- Department of Psychiatry, Icahn School of Medicine, New York, New York, USA
| | - Alexander Kolevzon
- Department of Psychiatry, Icahn School of Medicine, New York, New York, USA
| | - Soo Lee
- Department of Psychiatry and Behavioral Sciences, Rush University Medical Center, Chicago, Illinois, USA
| | - David Grodberg
- Child Study Center, Yale University, New Haven, Connecticut, USA
| | | |
Collapse
|
143
|
Román V, Adham N, Foley AG, Hanratty L, Farkas B, Lendvai B, Kiss B. Cariprazine alleviates core behavioral deficits in the prenatal valproic acid exposure model of autism spectrum disorder. Psychopharmacology (Berl) 2021; 238:2381-2392. [PMID: 34264367 DOI: 10.1007/s00213-021-05851-6/figures/5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 04/12/2021] [Indexed: 05/20/2023]
Abstract
RATIONALE Autism spectrum disorder (ASD) is a neurodevelopmental condition characterized by deficits in social communication and interaction and restricted, repetitive behaviors. The unmet medical need in ASD is considerable since there is no approved pharmacotherapy for the treatment of these deficits in social communication, interaction, and behavior. Cariprazine, a dopamine D3-preferring D3/D2 receptor partial agonist, is already approved for the treatment of schizophrenia and bipolar I disorder in adults; investigation in patients with ASD is warranted. OBJECTIVES The aim of this study was to investigate the effects of cariprazine, compared with risperidone and aripiprazole, in the rat prenatal valporic acid (VPA) exposure model on behavioral endpoints representing the core and associated symptoms of ASD. METHODS To induce the ASD model, time-mated Wistar rat dams were treated with VPA during pregnancy. Male offspring were assigned to groups and studied in a behavioral test battery at different ages, employing social play, open field, social approach-avoidance, and social recognition memory tests. Animals were dosed orally, once a day for 8 days, with test compounds (cariprazine, risperidone, aripiprazole) or vehicle before behavioral assessment. RESULTS Cariprazine showed dose-dependent efficacy on all behavioral endpoints. In the social play paradigm, only cariprazine was effective. On the remaining behavioral endpoints, including the reversal of hyperactivity, risperidone and aripiprazole displayed similar efficacy to cariprazine. CONCLUSIONS In the present study, cariprazine effectively reversed core behavioral deficits and hyperactivity present in juvenile and young adult autistic-like rats. These findings indicate that cariprazine may be useful in the treatment of ASD symptoms.
Collapse
Affiliation(s)
| | | | - Andrew G Foley
- Berand Neuropharmacology Limited, NovaUCD, Belfield Innovation Park, University College Dublin, Dublin, Ireland
| | - Lynsey Hanratty
- Berand Neuropharmacology Limited, NovaUCD, Belfield Innovation Park, University College Dublin, Dublin, Ireland
| | | | | | - Béla Kiss
- Gedeon Richter Plc, Budapest, Hungary
| |
Collapse
|
144
|
Clipperton-Allen AE, Zhang A, Cohen OS, Page DT. Environmental Enrichment Rescues Social Behavioral Deficits and Synaptic Abnormalities in Pten Haploinsufficient Mice. Genes (Basel) 2021; 12:1366. [PMID: 34573348 PMCID: PMC8468545 DOI: 10.3390/genes12091366] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 08/20/2021] [Accepted: 08/26/2021] [Indexed: 01/16/2023] Open
Abstract
Pten germline haploinsufficient (Pten+/-) mice, which model macrocephaly/autism syndrome, show social and repetitive behavior deficits, early brain overgrowth, and cortical-subcortical hyperconnectivity. Previous work indicated that altered neuronal connectivity may be a substrate for behavioral deficits. We hypothesized that exposing Pten+/- mice to environmental enrichment after brain overgrowth has occurred may facilitate adaptation to abnormal "hard-wired" connectivity through enhancing synaptic plasticity. Thus, we reared Pten+/- mice and their wild-type littermates from weaning under either standard (4-5 mice per standard-sized cage, containing only bedding and nestlet) or enriched (9-10 mice per large-sized cage, containing objects for exploration and a running wheel, plus bedding and nestlet) conditions. Adult mice were tested on social and non-social assays in which Pten+/- mice display deficits. Environmental enrichment rescued sex-specific deficits in social behavior in Pten+/- mice and partially rescued increased repetitive behavior in Pten+/- males. We found that Pten+/- mice show increased excitatory and decreased inhibitory pre-synaptic proteins; this phenotype was also rescued by environmental enrichment. Together, our results indicate that environmental enrichment can rescue social behavioral deficits in Pten+/- mice, possibly through normalizing the excitatory synaptic protein abundance.
Collapse
Affiliation(s)
| | | | | | - Damon Theron Page
- Department of Neuroscience, The Scripps Research Institute, Jupiter, FL 33458, USA; (A.E.C.-A.); (A.Z.); (O.S.C.)
| |
Collapse
|
145
|
Lee E, Lee S, Shin JJ, Choi W, Chung C, Lee S, Kim J, Ha S, Kim R, Yoo T, Yoo YE, Kim J, Noh YW, Rhim I, Lee SY, Kim W, Lee T, Shin H, Cho IJ, Deisseroth K, Kim SJ, Park JM, Jung MW, Paik SB, Kim E. Excitatory synapses and gap junctions cooperate to improve Pv neuronal burst firing and cortical social cognition in Shank2-mutant mice. Nat Commun 2021; 12:5116. [PMID: 34433814 PMCID: PMC8387434 DOI: 10.1038/s41467-021-25356-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Accepted: 08/05/2021] [Indexed: 02/07/2023] Open
Abstract
NMDA receptor (NMDAR) and GABA neuronal dysfunctions are observed in animal models of autism spectrum disorders, but how these dysfunctions impair social cognition and behavior remains unclear. We report here that NMDARs in cortical parvalbumin (Pv)-positive interneurons cooperate with gap junctions to promote high-frequency (>80 Hz) Pv neuronal burst firing and social cognition. Shank2–/– mice, displaying improved sociability upon NMDAR activation, show impaired cortical social representation and inhibitory neuronal burst firing. Cortical Shank2–/– Pv neurons show decreased NMDAR activity, which suppresses the cooperation between NMDARs and gap junctions (GJs) for normal burst firing. Shank2–/– Pv neurons show compensatory increases in GJ activity that are not sufficient for social rescue. However, optogenetic boosting of Pv neuronal bursts, requiring GJs, rescues cortical social cognition in Shank2–/– mice, similar to the NMDAR-dependent social rescue. Therefore, NMDARs and gap junctions cooperate to promote cortical Pv neuronal bursts and social cognition. How NMDAR and GABA neuronal dysfunctions result in impaired social behaviour is unclear. Here, the authors show that NMDARs and gap junctions in cortical PV interneurons modulate burst firing, affecting social behaviour.
Collapse
Affiliation(s)
- Eunee Lee
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science (IBS), Daejeon, Korea.,Department of Anatomy, College of Medicine, Yonsei University, Seoul, Korea
| | - Seungjoon Lee
- Department of Biological Sciences, KAIST, Daejeon, Korea
| | - Jae Jin Shin
- Department of Brain and Cognitive Science, College of Natural Science, Seoul National University, Seoul, Korea.,Center for Cognition and Sociality, Institute for Basic Science (IBS), Daejeon, Korea
| | - Woochul Choi
- Program of Brain and Cognitive Engineering, Department of Bio and Brain Engineering, Korea Advanced Institute for Science and Technology (KAIST), Daejeon, Korea
| | - Changuk Chung
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science (IBS), Daejeon, Korea
| | - Suho Lee
- Department of Biological Sciences, KAIST, Daejeon, Korea
| | - Jihye Kim
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science (IBS), Daejeon, Korea
| | - Seungmin Ha
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science (IBS), Daejeon, Korea
| | - Ryunhee Kim
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science (IBS), Daejeon, Korea
| | - Taesun Yoo
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science (IBS), Daejeon, Korea
| | - Ye-Eun Yoo
- Department of Biological Sciences, KAIST, Daejeon, Korea
| | - Jisoo Kim
- Department of Biological Sciences, KAIST, Daejeon, Korea
| | - Young Woo Noh
- Department of Biological Sciences, KAIST, Daejeon, Korea
| | - Issac Rhim
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science (IBS), Daejeon, Korea
| | - Soo Yeon Lee
- Department of Biological Sciences, KAIST, Daejeon, Korea
| | - Woohyun Kim
- Department of Biological Sciences, KAIST, Daejeon, Korea
| | - Taekyung Lee
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science (IBS), Daejeon, Korea
| | - Hyogeun Shin
- Center for BioMicrosystems, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, Korea
| | - Il-Joo Cho
- Center for BioMicrosystems, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, Korea
| | - Karl Deisseroth
- Department of Bioengineering, Department of Psychiatry and Behavioral Sciences, Howard Hughes Medical Institute, Stanford University, Stanford, CA, USA
| | - Sang Jeong Kim
- Department of Physiology, College of Medicine, Seoul National University, Seoul, Korea
| | - Joo Min Park
- Center for Cognition and Sociality, Institute for Basic Science (IBS), Daejeon, Korea.
| | - Min Whan Jung
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science (IBS), Daejeon, Korea. .,Department of Biological Sciences, KAIST, Daejeon, Korea.
| | - Se-Bum Paik
- Program of Brain and Cognitive Engineering, Department of Bio and Brain Engineering, Korea Advanced Institute for Science and Technology (KAIST), Daejeon, Korea.
| | - Eunjoon Kim
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science (IBS), Daejeon, Korea. .,Department of Biological Sciences, KAIST, Daejeon, Korea.
| |
Collapse
|
146
|
Deemyad T, Puig S, Papale AE, Qi H, LaRocca GM, Aravind D, LaNoce E, Urban NN. Lateralized Decrease of Parvalbumin+ Cells in the Somatosensory Cortex of ASD Models Is Correlated with Unilateral Tactile Hypersensitivity. Cereb Cortex 2021; 32:554-568. [PMID: 34347040 DOI: 10.1093/cercor/bhab233] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 05/31/2021] [Accepted: 06/21/2021] [Indexed: 12/27/2022] Open
Abstract
Inhibitory control of excitatory networks contributes to cortical functions. Increasing evidence indicates that parvalbumin (PV+)-expressing basket cells (BCs) are a major player in maintaining the balance between excitation (E) and inhibition (I). Disruption of E/I balance in cortical networks is believed to be a hallmark of autism spectrum disorder (ASD). Here, we report a lateralized decrease in the number of PV+ BCs in L2/3 of the somatosensory cortex in the dominant hemisphere of Shank3-/- and Cntnap2-/- mouse models of ASD. The dominant hemisphere was identified during a reaching task to establish each animal's dominant forepaw. Double labeling with anti-PV antibody and a biotinylated lectin (Vicia villosa lectin [VVA]) showed that the number of BCs was not different but rather, some BCs did not express PV (PV-), resulting in an elevated number of PV- VVA+ BCs. Finally, we showed that dominant hindpaws had higher mechanical sensitivity when compared with the other hindpaws. This mechanical hypersensitivity in the dominant paw strongly correlated with the decrease in the number of PV+ interneurons and reduced PV expression in the corresponding cortex. Together, these results suggest that the hypersensitivity in ASD patients could be due to decreased inhibitory inputs to the dominant somatosensory cortex.
Collapse
Affiliation(s)
- Tara Deemyad
- Department of Neurobiology, University of Pittsburgh, Pittsburgh, PA 15213, USA.,Department of Psychiatry, Translational Neuroscience Program, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - Stephanie Puig
- Department of Psychiatry, Translational Neuroscience Program, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - Andrew E Papale
- Department of Neurobiology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Hang Qi
- Department of Neurobiology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Gregory M LaRocca
- Department of Neurobiology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Deepthi Aravind
- Department of Neurobiology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Emma LaNoce
- Department of Neurobiology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Nathaniel N Urban
- Department of Neurobiology, University of Pittsburgh, Pittsburgh, PA 15213, USA.,Center for Neuroscience at the University of Pittsburgh, University of Pittsburgh, Pittsburgh, PA 15213, USA
| |
Collapse
|
147
|
He JL, Oeltzschner G, Mikkelsen M, Deronda A, Harris AD, Crocetti D, Wodka EL, Mostofsky SH, Edden RAE, Puts NAJ. Region-specific elevations of glutamate + glutamine correlate with the sensory symptoms of autism spectrum disorders. Transl Psychiatry 2021; 11:411. [PMID: 34326312 PMCID: PMC8322079 DOI: 10.1038/s41398-021-01525-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 06/04/2021] [Accepted: 06/10/2021] [Indexed: 02/07/2023] Open
Abstract
Individuals on the autism spectrum are often reported as being hyper- and/or hyporeactive to sensory input. These sensory symptoms were one of the key observations that led to the development of the altered excitation-inhibition (E-I) model of autism, which posits that an increase ratio of excitatory to inhibitory signaling may explain certain phenotypical expressions of autism spectrum disorders (ASD). While there has been strong support for the altered E-I model of autism, much of the evidence has come from animal models. With regard to in-vivo human studies, evidence for altered E-I balance in ASD come from studies adopting magnetic resonance spectroscopy (MRS). Spectral-edited MRS can be used to provide measures of the levels of GABA + (GABA + macromolecules) and Glx (glutamate + glutamine) in specific brain regions as proxy markers of inhibition and excitation respectively. In the current study, we found region-specific elevations of Glx in the primary sensorimotor cortex (SM1) in ASD. There were no group differences of GABA+ in either the SM1 or thalamus. Higher levels of Glx were associated with more parent reported difficulties of sensory hyper- and hyporeactivity, as well as reduced feed-forward inhibition during tactile perception in children with ASD. Critically, the finding of elevated Glx provides strong empirical support for increased excitation in ASD. Our results also provide a clear link between Glx and the sensory symptoms of ASD at both behavioral and perceptual levels.
Collapse
Affiliation(s)
- Jason L He
- Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
- F. M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD, USA
- Department of Forensic and Neurodevelopmental Sciences, Sackler Institute for Translational Neurodevelopment, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, UK
| | - Georg Oeltzschner
- Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
- F. M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD, USA
| | - Mark Mikkelsen
- Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
- F. M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD, USA
| | - Alyssa Deronda
- Center for Neurodevelopmental and Imaging Research, Kennedy Krieger Institute, Baltimore, MD, USA
| | - Ashley D Harris
- Department of Radiology, University of Calgary, Calgary, Canada
| | - Deana Crocetti
- Center for Neurodevelopmental and Imaging Research, Kennedy Krieger Institute, Baltimore, MD, USA
| | - Ericka L Wodka
- Center for Autism and Related Disorders, Kennedy Krieger Institute, Baltimore, MD, USA
| | - Stewart H Mostofsky
- Center for Neurodevelopmental and Imaging Research, Kennedy Krieger Institute, Baltimore, MD, USA
- Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Psychiatry and Behavioral Sciences, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Richard A E Edden
- Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
- F. M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD, USA
| | - Nicolaas A J Puts
- Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- F. M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD, USA.
- Department of Forensic and Neurodevelopmental Sciences, Sackler Institute for Translational Neurodevelopment, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, UK.
- MRC Centre for Neurodevelopmental Disorders, King's College London, London, UK.
| |
Collapse
|
148
|
Yang Y, Wang B, Zhong Z, Chen H, Ding W, Hoi MPM. Clonazepam attenuates neurobehavioral abnormalities in offspring exposed to maternal immune activation by enhancing GABAergic neurotransmission. Biochem Pharmacol 2021; 192:114711. [PMID: 34324871 DOI: 10.1016/j.bcp.2021.114711] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 07/23/2021] [Accepted: 07/23/2021] [Indexed: 10/20/2022]
Abstract
Ample evidence indicates that maternal immune activation (MIA) during gestation is linked to an increased risk for neurodevelopmental and psychiatric disorders, such as autism spectrum disorder (ASD), anxiety and depression, in offspring. However, the underlying mechanism for such a link remains largely elusive. Here, we performed RNA sequencing (RNA-seq) to examine the transcriptional profiles changes in mice in response to MIA and identified that the expression of Scn1a gene, encoding the pore-forming α-subunit of the brain voltage-gated sodium channel type-1 (NaV1.1) primarily in fast-spiking inhibitory interneurons, was significantly decreased in the medial prefrontal cortex (mPFC) of juvenile offspring after MIA. Moreover, diminished excitatory drive onto interneurons causes reduction of spontaneous gamma-aminobutyric acid (GABA)ergic neurotransmission in the mPFC of MIA offspring, leading to hyperactivity in this brain region. Remarkably, treatment with low-dose benzodiazepines clonazepam, an agonist of GABAA receptors, completely prevented the behavioral abnormalities, including stereotypies, social deficits, anxiety- and depression-like behavior, via increasing inhibitory neurotransmission as well as decreasing neural activity in the mPFC of MIA offspring. Our results demonstrate that decreased expression of NaV1.1 in the mPFC leads to abnormalities in maternal inflammation-related behaviors and provides a potential therapeutic strategy for the abnormal behavioral phenotypes observed in the offspring exposed to MIA.
Collapse
Affiliation(s)
- Youjun Yang
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macao 999078, China; School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Baojia Wang
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Zhanqion Zhong
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Hanbin Chen
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macao 999078, China
| | - Weijun Ding
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Maggie Pui Man Hoi
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macao 999078, China.
| |
Collapse
|
149
|
Mao M, Zhou Z, Sun M, Wang C, Sun J. The dysfunction of parvalbumin interneurons mediated by microglia contributes to cognitive impairment induced by lipopolysaccharide challenge. Neurosci Lett 2021; 762:136133. [PMID: 34311051 DOI: 10.1016/j.neulet.2021.136133] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Revised: 07/15/2021] [Accepted: 07/20/2021] [Indexed: 11/27/2022]
Abstract
BACKGROUND The mechanisms underlying cognitive impairments induced by systemic inflammation remain unclear. Increasing evidence has suggested that parvalbumin (PV) interneurons play an important role in regulating cognitive behaviors and its dysfunction is implicated in many neurological disorders. Thus, the present study was aimed to detect whether the destruction of PV interneurons mediates cognitive impairment associated with systemic inflammation. METHODS Male wild-type C57BL/6J mice (12-14 weeks old) received lipopolysaccharide (LPS 2 mg/kg i.p.) injection to establish the systemic inflammation model. For the suppression of microglial activation, minocycline (50 mg/kg i.p.) was applied. Animal behavior tests were conducted on day 3 post-LPS injection including the open field test, fear conditioning test and Y maze test. The PV expression in hippocampus was detected by Western blot and immunofluorescence. The number of perisomatic boutons around the NeuN-positive cells and microglia in hippocampus was detected by immunofluorescence. RESULTS LPS induced hippocampus-dependent memory and working memory impairment, coinciding with decreased PV expression, reduced perisomatic boutons around the NeuN-positive cells and activated microglia in the hippocampus. Notably, the treatment of minocycline suppressed the microglial activation and rescued the PV expression as well as the perisomatic boutons around the NeuN-positive cells in the hippocampus, contributing to improved cognitive function. CONCLUSION Our study suggests that the dysfunction of parvalbumin interneurons mediated by microglia plays a key role in LPS-induced cognitive impairments, which may serve a therapeutic strategy for cognitive disorders associated with systemic inflammation.
Collapse
Affiliation(s)
- Meng Mao
- Department of Anesthesiology, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Zhenhui Zhou
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Menghan Sun
- Department of Anesthesiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, China
| | - Chaoran Wang
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jie Sun
- Department of Anesthesiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, China.
| |
Collapse
|
150
|
Pretel J, Torres JJ, Marro J. EEGs Disclose Significant Brain Activity Correlated with Synaptic Fickleness. BIOLOGY 2021; 10:647. [PMID: 34356502 PMCID: PMC8301300 DOI: 10.3390/biology10070647] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Revised: 07/04/2021] [Accepted: 07/08/2021] [Indexed: 11/17/2022]
Abstract
We here study a network of synaptic relations mingling excitatory and inhibitory neuron nodes that displays oscillations quite similar to electroencephalogram (EEG) brain waves, and identify abrupt variations brought about by swift synaptic mediations. We thus conclude that corresponding changes in EEG series surely come from the slowdown of the activity in neuron populations due to synaptic restrictions. The latter happens to generate an imbalance between excitation and inhibition causing a quick explosive increase of excitatory activity, which turns out to be a (first-order) transition among dynamic mental phases. Moreover, near this phase transition, our model system exhibits waves with a strong component in the so-called delta-theta domain that coexist with fast oscillations. These findings provide a simple explanation for the observed delta-gamma and theta-gamma modulation in actual brains, and open a serious and versatile path to understand deeply large amounts of apparently erratic, easily accessible brain data.
Collapse
|