101
|
Klug JR, Yan X, Hoffman HA, Engelhardt MD, Osakada F, Callaway EM, Jin X. Asymmetric cortical projections to striatal direct and indirect pathways distinctly control actions. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.02.560589. [PMID: 37873164 PMCID: PMC10592949 DOI: 10.1101/2023.10.02.560589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
The striatal direct and indirect pathways constitute the core for basal ganglia function in action control. Although both striatal D1- and D2-spiny projection neurons (SPNs) receive excitatory inputs from the cerebral cortex, whether or not they share inputs from the same cortical neurons, and how pathway-specific corticostriatal projections control behavior remain largely unknown. Here using a new G-deleted rabies system in mice, we found that more than two-thirds of excitatory inputs to D2-SPNs also target D1-SPNs, while only one-third do so vice versa. Optogenetic stimulation of striatal D1- vs. D2-SPN-projecting cortical neurons differently regulate locomotion, reinforcement learning and sequence behavior, implying the functional dichotomy of pathway-specific corticostriatal subcircuits. These results reveal the partially segregated yet asymmetrically overlapping cortical projections on striatal D1- vs. D2-SPNs, and that the pathway-specific corticostriatal subcircuits distinctly control behavior. It has important implications in a wide range of neurological and psychiatric diseases affecting cortico-basal ganglia circuitry.
Collapse
Affiliation(s)
- Jason R. Klug
- Molecular Neurobiology Laboratory, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
- These authors contributed equally to this work
| | - Xunyi Yan
- Molecular Neurobiology Laboratory, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
- Center for Motor Control and Disease, Key Laboratory of Brain Functional Genomics, East China Normal University, 3663 North Zhongshan Road, Shanghai 200062, China
- These authors contributed equally to this work
| | - Hilary A. Hoffman
- Molecular Neurobiology Laboratory, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Max D. Engelhardt
- Molecular Neurobiology Laboratory, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Fumitaka Osakada
- Systems Neurobiology Laboratories, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Edward M. Callaway
- Systems Neurobiology Laboratories, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Xin Jin
- Molecular Neurobiology Laboratory, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
- Center for Motor Control and Disease, Key Laboratory of Brain Functional Genomics, East China Normal University, 3663 North Zhongshan Road, Shanghai 200062, China
- NYU–ECNU Institute of Brain and Cognitive Science, New York University Shanghai, 3663 North Zhongshan Road, Shanghai 200062, China
| |
Collapse
|
102
|
Réthelyi JM, Vincze K, Schall D, Glennon J, Berkel S. The role of insulin/IGF1 signalling in neurodevelopmental and neuropsychiatric disorders - Evidence from human neuronal cell models. Neurosci Biobehav Rev 2023; 153:105330. [PMID: 37516219 DOI: 10.1016/j.neubiorev.2023.105330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 07/15/2023] [Accepted: 07/26/2023] [Indexed: 07/31/2023]
Abstract
Insulin and insulin-like growth factor 1 (IGF1) signalling play a central role in the development and maintenance of neurons in the brain, and human neurodevelopmental as well as neuropsychiatric disorders have been linked to impaired insulin and IGF1 signalling. This review focuses on the impairments of the insulin and IGF1 signalling cascade in the context of neurodevelopmental and neuropsychiatric disorders, based on evidence from human neuronal cell models. Clear evidence was obtained for impaired insulin and IGF1 receptor downstream signalling in neurodevelopmental disorders, while the evidence for its role in neuropsychiatric disorders was less substantial. Human neuronal model systems can greatly add to our knowledge about insulin/IGF1 signalling in the brain, its role in restoring dendritic maturity, and complement results from clinical studies and animal models. Moreover, they represent a useful model for the development of new therapeutic strategies. Further research is needed to systematically investigate the exact role of the insulin/IGF1 signalling cascades in neurodevelopmental and neuropsychiatric disorders, and to elucidate the respective therapeutic implications.
Collapse
Affiliation(s)
- János M Réthelyi
- Department of Psychiatry and Psychotherapy, Semmelweis University, Budapest, Hungary
| | - Katalin Vincze
- Department of Psychiatry and Psychotherapy, Semmelweis University, Budapest, Hungary; Doctoral School of Mental Health Sciences, Semmelweis University, Budapest, Hungary
| | - Dorothea Schall
- Institute of Human Genetics, Heidelberg University, Heidelberg, Germany
| | - Jeffrey Glennon
- Conway Institute of Biomedical and Biomolecular Research, School of Medicine, University College Dublin, Dublin, Ireland
| | - Simone Berkel
- Institute of Human Genetics, Heidelberg University, Heidelberg, Germany; Interdisciplinary Centre of Neurosciences (IZN), Heidelberg University, Germany.
| |
Collapse
|
103
|
You M, Li S, Yan S, Yao D, Wang T, Wang Y. Exposure to nonylphenol in early life causes behavioural deficits related with autism spectrum disorders in rats. ENVIRONMENT INTERNATIONAL 2023; 180:108228. [PMID: 37802007 DOI: 10.1016/j.envint.2023.108228] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 08/31/2023] [Accepted: 09/20/2023] [Indexed: 10/08/2023]
Abstract
Early-life exposure to environmental endocrine disruptors (EDCs) is a potential risk factor for autism spectrum disorder (ASD). Exposure to nonylphenol (NP), a typical EDC, is known to cause some long-term behavioural abnormalities. Moreover, these abnormal behaviours are the most frequent psychiatric co-morbidities in ASD. However, the direct evidence for the link between NP exposure in early life and ASD-like behavioural phenotypes is still missing. In the present study, typical ASD-like behaviours induced by valproic acid treatment were considered as a positive behavioural control. We investigated impacts on social behaviours following early-life exposure to NP, and explored effects of this exposure on neuronal dendritic spines, mitochondria function, oxidative stress, and endoplasmic reticulum (ER) stress. Furthermore, primary cultured rat neurons were employed as in vitro model to evaluate changes in dendritic spine caused by exposure to NP, and oxidative stress and ER stress were specifically modulated to further explore their roles in these changes. Our results indicated rats exposed to NP in early life showed mild ASD-like behaviours. Moreover, we also found the activation of ER stress triggered by oxidative stress may contribute to dendritic spine decrease and synaptic dysfunction, which may underlie neurobehavioural abnormalities induced by early-life exposure to NP.
Collapse
Affiliation(s)
- Mingdan You
- Department of Occupational and Environmental Health, School of Public Health, China Medical University, Shenyang, Liaoning, People's Republic of China; School of Public Heath, Guizhou Medical University, Guiyang, Guizhou, People's Republic of China
| | - Siyao Li
- Department of Occupational and Environmental Health, School of Public Health, China Medical University, Shenyang, Liaoning, People's Republic of China; Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention (China Medical University), Ministry of Education, Shenyang, Liaoning, People's Republic of China
| | - Siyu Yan
- Department of Occupational and Environmental Health, School of Public Health, China Medical University, Shenyang, Liaoning, People's Republic of China; Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention (China Medical University), Ministry of Education, Shenyang, Liaoning, People's Republic of China
| | - Dianqi Yao
- Department of Occupational and Environmental Health, School of Public Health, China Medical University, Shenyang, Liaoning, People's Republic of China; Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention (China Medical University), Ministry of Education, Shenyang, Liaoning, People's Republic of China
| | - Tingyu Wang
- College of Medical Laboratory, Dalian Medical University, Dalian, Liaoning, People's Republic of China
| | - Yi Wang
- Department of Occupational and Environmental Health, School of Public Health, China Medical University, Shenyang, Liaoning, People's Republic of China; Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention (China Medical University), Ministry of Education, Shenyang, Liaoning, People's Republic of China.
| |
Collapse
|
104
|
Coulon A, Siedlecki-Wullich D, Najdek C, Gelle C, Ayral AM, Demiautte F, Lambert E, Vandeputte A, Brodin P, Mendes T, Lambert JC, Kilinc D, Dumont J, Chapuis J. High-Content Screening of Synaptic Density Modulators in Primary Neuronal Cultures. Curr Protoc 2023; 3:e904. [PMID: 37882787 DOI: 10.1002/cpz1.904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2023]
Abstract
The synapse, which represents the structural and functional basis of neuronal communication, is one of the first elements affected in several neurodegenerative diseases. To better understand the potential role of gene expression in synapse loss, we developed an original high-content screening (HCS) model capable of quantitatively assessing the impact of gene silencing on synaptic density. Our approach is based on a model of primary neuronal cultures (PNCs) from the neonatal rat hippocampus, whose mature synapses are visualized by the relative localization of the presynaptic protein Synaptophysin with the postsynaptic protein Homer1. The heterogeneity of PNCs and the small sizes of the synaptic structures pose technical challenges associated with the level of automation necessary for HCS studies. We overcame these technical challenges, automated the processes of image analysis and data analysis, and carried out tests under real-world conditions to demonstrate the robustness of the model developed. In this article, we describe the screening of a custom library of 198 shRNAs in PNCs in the 384-well plate format. © 2023 The Authors. Current Protocols published by Wiley Periodicals LLC. Basic Protocol 1: Culture of primary hippocampal rat neurons in 384-well plates Basic Protocol 2: Lentiviral shRNA transduction of primary neuronal culture in 384-well plates Basic Protocol 3: Immunostaining of the neuronal network and synaptic markers in 384-well plates Basic Protocol 4: Image acquisition using a high-throughput reader Basic Protocol 5: Image segmentation and analysis Basic Protocol 6: Synaptic density analysis.
Collapse
Affiliation(s)
- Audrey Coulon
- Université de Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE - Facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, Lille, France
| | - Dolores Siedlecki-Wullich
- Université de Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE - Facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, Lille, France
| | - Chloé Najdek
- Université de Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE - Facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, Lille, France
| | - Carla Gelle
- Université de Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE - Facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, Lille, France
| | - Anne-Marie Ayral
- Université de Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE - Facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, Lille, France
| | - Florie Demiautte
- Université de Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE - Facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, Lille, France
| | - Erwan Lambert
- Université de Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE - Facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, Lille, France
| | - Alexandre Vandeputte
- Université de Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, Center for Infection and Immunity of Lille (CIIL), Lille, France
| | - Priscille Brodin
- Université de Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, Center for Infection and Immunity of Lille (CIIL), Lille, France
| | - Tiago Mendes
- Université de Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE - Facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, Lille, France
| | - Jean-Charles Lambert
- Université de Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE - Facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, Lille, France
| | - Devrim Kilinc
- Université de Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE - Facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, Lille, France
| | - Julie Dumont
- Université de Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE - Facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, Lille, France
| | - Julien Chapuis
- Université de Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE - Facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, Lille, France
| |
Collapse
|
105
|
Yang Q, Perfitt TL, Quay J, Hu L, Lawson-Qureshi D, Colbran RJ. Clustering of Ca V 1.3 L-type calcium channels by Shank3. J Neurochem 2023; 167:16-37. [PMID: 37392026 PMCID: PMC10543641 DOI: 10.1111/jnc.15880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 05/19/2023] [Accepted: 05/28/2023] [Indexed: 07/02/2023]
Abstract
Clustering of L-type voltage-gated Ca2+ channels (LTCCs) in the plasma membrane is increasingly implicated in creating highly localized Ca2+ signaling nanodomains. For example, neuronal LTCC activation can increase phosphorylation of the nuclear CREB transcription factor by increasing Ca2+ concentrations within a nanodomain close to the channel, without requiring bulk Ca2+ increases in the cytosol or nucleus. However, the molecular basis for LTCC clustering is poorly understood. The postsynaptic scaffolding protein Shank3 specifically associates with one of the major neuronal LTCCs, the CaV 1.3 calcium channel, and is required for optimal LTCC-dependent excitation-transcription coupling. Here, we co-expressed CaV 1.3 α1 subunits with two distinct epitope-tags with or without Shank3 in HEK cells. Co-immunoprecipitation studies using the cell lysates revealed that Shank3 can assemble complexes containing multiple CaV 1.3 α1 subunits under basal conditions. Moreover, CaV 1.3 LTCC complex formation was facilitated by CaV β subunits (β3 and β2a), which also interact with Shank3. Shank3 interactions with CaV 1.3 LTCCs and multimeric CaV 1.3 LTCC complex assembly were disrupted following the addition of Ca2+ to cell lysates, perhaps simulating conditions within an activated CaV 1.3 LTCC nanodomain. In intact HEK293T cells, co-expression of Shank3 enhanced the intensity of membrane-localized CaV 1.3 LTCC clusters under basal conditions, but not after Ca2+ channel activation. Live cell imaging studies also revealed that Ca2+ influx through LTCCs disassociated Shank3 from CaV 1.3 LTCCs clusters and reduced the CaV 1.3 cluster intensity. Deletion of the Shank3 PDZ domain prevented both binding to CaV 1.3 and the changes in multimeric CaV 1.3 LTCC complex assembly in vitro and in HEK293 cells. Finally, we found that shRNA knock-down of Shank3 expression in cultured rat primary hippocampal neurons reduced the intensity of surface-localized CaV 1.3 LTCC clusters in dendrites. Taken together, our findings reveal a novel molecular mechanism contributing to neuronal LTCC clustering under basal conditions.
Collapse
Affiliation(s)
- Qian Yang
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN, USA 37232-0615
| | - Tyler L. Perfitt
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN, USA 37232-0615
- Current address: Rare Disease Research Unit, Pfizer Inc
| | - Juliana Quay
- Chemical and Physical Biology, Vanderbilt University School of Medicine, Nashville, TN, USA 37232-0615
| | - Lan Hu
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN, USA 37232-0615
| | - Dorian Lawson-Qureshi
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN, USA 37232-0615
| | - Roger J. Colbran
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN, USA 37232-0615
- Vanderbilt Brain Institute, Vanderbilt University School of Medicine, Nashville, TN, USA 37232-0615
- Vanderbilt-Kennedy Center for Research on Human Development, Vanderbilt University School of Medicine, Nashville, TN, USA 37232-0615
| |
Collapse
|
106
|
Maccallini C, Amoroso R. Neuronal Nitric Oxide Synthase and Post-Translational Modifications in the Development of Central Nervous System Diseases: Implications and Regulation. Molecules 2023; 28:6691. [PMID: 37764469 PMCID: PMC10538099 DOI: 10.3390/molecules28186691] [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: 08/26/2023] [Revised: 09/14/2023] [Accepted: 09/15/2023] [Indexed: 09/29/2023] Open
Abstract
In the Central Nervous System (CNS), Nitric Oxide (NO) is mainly biosynthesized by neuronal Nitric Oxide Synthase (nNOS). The dysregulated activation of nNOS in neurons is critical in the development of different conditions affecting the CNS. The excessive production of NO by nNOS is responsible for a number of proteins' post-translational modifications (PTMs), which can lead to aberrant biochemical pathways, impairing CNS functions. In this review, we briefly revise the main implications of dysregulated nNOS in the progression of the most prevalent CNS neurodegenerative disorders, i.e., Alzheimer's disease (AD) and Parkinson's disease, as well as in the development of neuronal disorders. Moreover, a specific focus on compounds able to modulate nNOS activity as promising therapeutics to tackle different neuronal diseases is presented.
Collapse
Affiliation(s)
- Cristina Maccallini
- Department of Pharmacy, University “G. d’Annunzio” of Chieti-Pescara, Via dei Vestini 31, 66100 Chieti, Italy;
| | | |
Collapse
|
107
|
Silverman JL, Fenton T, Haouchine O, Hallam E, Smith E, Jackson K, Rahbarian D, Canales C, Adhikari A, Nord A, Ben-Shalom R. Hyperexcitability and translational phenotypes in a preclinical model of SYNGAP1 mutations. RESEARCH SQUARE 2023:rs.3.rs-3246655. [PMID: 37790402 PMCID: PMC10543290 DOI: 10.21203/rs.3.rs-3246655/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/05/2023]
Abstract
SYNGAP1 is a critical gene for neuronal development, synaptic structure, and function. Although rare, the disruption of SYNGAP1 directly causes a genetically identifiable neurodevelopmental disorder (NDD) called SYNGAP1 -related intellectual disability. Without functional SynGAP1 protein, patients present with intellectual disability, motor impairments, and epilepsy. Previous work using mouse models with a variety of germline and conditional mutations has helped delineate SynGAP1's critical roles in neuronal structure and function, as well as key biochemical signaling pathways essential to synapse integrity. Homozygous loss of SYNGAP1 is embryonically lethal. Heterozygous mutations of SynGAP1 result in a broad range of phenotypes including increased locomotor activity, impaired working spatial memory, impaired cued fear memory, and increased stereotypic behavior. Our in vivo functional data, using the original germline mutation mouse line from the Huganir laboratory, corroborated robust hyperactivity and learning and memory deficits. Here, we describe impairments in the translational biomarker domain of sleep, characterized using neurophysiological data collected with wireless telemetric electroencephalography (EEG). We discovered Syngap1+/- mice exhibited elevated spike trains in both number and duration, in addition to elevated power, most notably in the delta power band. Primary neurons from Syngap1+/- mice displayed increased network firing activity, greater spikes per burst, and shorter inter-burst intervals between peaks using high density micro-electrode arrays (HD-MEA). This work is translational, innovative, and highly significant as it outlines functional impairments in Syngap1 mutant mice. Simultaneously, the work utilized untethered, wireless neurophysiology that can discover potential biomarkers of Syngap1 RI-D, for clinical trials, as it has done with other NDDs. Our work is substantial forward progress toward translational work for SynGAP1R-ID as it bridges in-vitro electrophysiological neuronal activity and function with in vivo neurophysiological brain activity and function. These data elucidate multiple quantitative, translational biomarkers in vivo and in vitro for the development of treatments for SYNGAP1-related intellectual disability.
Collapse
Affiliation(s)
- Jill L Silverman
- MIND Institute and Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
108
|
Ganesh S, Lam TT, Garcia-Milian R, D’Souza D, Nairn AC, Elgert K, Eitan E, Ranganathan M. Peripheral signature of altered synaptic integrity in young onset cannabis use disorder: A proteomic study of circulating extracellular vesicles. World J Biol Psychiatry 2023; 24:603-613. [PMID: 36994633 PMCID: PMC10471733 DOI: 10.1080/15622975.2023.2197039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Revised: 03/20/2023] [Accepted: 03/27/2023] [Indexed: 03/31/2023]
Abstract
BACKGROUND Rates of Cannabis Use Disorder (CUD) are highest amongst young adults. Paucity of brain tissue samples limits the ability to examine the molecular basis of cannabis related neuropathology. Proteomic studies of neuron-derived extracellular vesicles (NDEs) isolated from the biofluids may reveal markers of neuropathology in CUD. METHODS NDEs were extracted using ExoSORT, an immunoaffinity method to enrich NDEs from plasma samples from patients with young onset CUD and matched controls. Differential proteomic profiles were explored with Label Free Quantification (LFQ) mass spectrometry. Selected proteins were validated using orthogonal methods. RESULTS A total of 231 (±10) proteins were identified in NDE preparations from CUD and controls of which 28 were differentially abundant between groups. The difference in abundance of properdin (CFP gene) was statistically significant. SHANK1 (SHANK1 gene), an adapter protein at the post-synaptic density, was nominally depleted in the CUD NDE preparations. CONCLUSION In this pilot study, we noted a decrease in SHANK1 protein, involved in the structural and functional integrity of glutamatergic post-synapse, a potential peripheral signature of CUD neuropathology. The study shows that LFQ mass spectrometry proteomic analysis of NDEs derived from plasma may yield important insights into the synaptic pathology associated with CUD.
Collapse
Affiliation(s)
- Suhas Ganesh
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, 06520
| | - TuKiet T. Lam
- Department of Molecular Biophysics and Biochemistry, Yale School of Medicine, New Haven, CT, 06520
- Keck Mass Spectrometry & Proteomics Resource, Yale School of Medicine, New Haven, CT, 06510
| | - Rolando Garcia-Milian
- Bioinformatics Support Program, Cushing/Whitney Medical Library, Yale School of Medicine, New Haven, CT, 06510
| | - Deepak D’Souza
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, 06520
| | - Angus C. Nairn
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, 06520
| | | | | | - Mohini Ranganathan
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, 06520
| |
Collapse
|
109
|
Tian R, Li Y, Zhao H, Lyu W, Zhao J, Wang X, Lu H, Xu H, Ren W, Tan QQ, Shi Q, Wang GD, Zhang YP, Lai L, Mi J, Jiang YH, Zhang YQ. Modeling SHANK3-associated autism spectrum disorder in Beagle dogs via CRISPR/Cas9 gene editing. Mol Psychiatry 2023; 28:3739-3750. [PMID: 37848710 DOI: 10.1038/s41380-023-02276-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 08/30/2023] [Accepted: 09/14/2023] [Indexed: 10/19/2023]
Abstract
Despite intensive studies in modeling neuropsychiatric disorders especially autism spectrum disorder (ASD) in animals, many challenges remain. Genetic mutant mice have contributed substantially to the current understanding of the molecular and neural circuit mechanisms underlying ASD. However, the translational value of ASD mouse models in preclinical studies is limited to certain aspects of the disease due to the apparent differences in brain and behavior between rodents and humans. Non-human primates have been used to model ASD in recent years. However, a low reproduction rate due to a long reproductive cycle and a single birth per pregnancy, and an extremely high cost prohibit a wide use of them in preclinical studies. Canine model is an appealing alternative because of its complex and effective dog-human social interactions. In contrast to non-human primates, dog has comparable drug metabolism as humans and a high reproduction rate. In this study, we aimed to model ASD in experimental dogs by manipulating the Shank3 gene as SHANK3 mutations are one of most replicated genetic defects identified from ASD patients. Using CRISPR/Cas9 gene editing, we successfully generated and characterized multiple lines of Beagle Shank3 (bShank3) mutants that have been propagated for a few generations. We developed and validated a battery of behavioral assays that can be used in controlled experimental setting for mutant dogs. bShank3 mutants exhibited distinct and robust social behavior deficits including social withdrawal and reduced social interactions with humans, and heightened anxiety in different experimental settings (n = 27 for wild-type controls and n = 44 for mutants). We demonstrate the feasibility of producing a large number of mutant animals in a reasonable time frame. The robust and unique behavioral findings support the validity and value of a canine model to investigate the pathophysiology and develop treatments for ASD and potentially other psychiatric disorders.
Collapse
Affiliation(s)
- Rui Tian
- State Key Laboratory of Molecular Developmental Biology, and CAS Center for Excellence in Brain Science and Intelligence Technology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Yuan Li
- Beijing Sinogene Biotechnology Co. Ltd, Beijing, China
| | - Hui Zhao
- State Key Laboratory of Molecular Developmental Biology, and CAS Center for Excellence in Brain Science and Intelligence Technology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
- Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Wen Lyu
- State Key Laboratory of Molecular Developmental Biology, and CAS Center for Excellence in Brain Science and Intelligence Technology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Jianping Zhao
- Beijing Sinogene Biotechnology Co. Ltd, Beijing, China
| | - Xiaomin Wang
- Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Heng Lu
- Department of Life Science and Medicine, University of Science and Technology of China, Hefei, 230022, China
- State Key Laboratory of Genetic Resources and Evolution, and Center for Excellence in Animal Evolution and Genetics, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, China
| | - Huijuan Xu
- State Key Laboratory of Molecular Developmental Biology, and CAS Center for Excellence in Brain Science and Intelligence Technology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Wei Ren
- State Key Laboratory of Molecular Developmental Biology, and CAS Center for Excellence in Brain Science and Intelligence Technology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Qing-Quan Tan
- State Key Laboratory of Molecular Developmental Biology, and CAS Center for Excellence in Brain Science and Intelligence Technology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Qi Shi
- State Key Laboratory of Molecular Developmental Biology, and CAS Center for Excellence in Brain Science and Intelligence Technology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Guo-Dong Wang
- State Key Laboratory of Genetic Resources and Evolution, and Center for Excellence in Animal Evolution and Genetics, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, China
| | - Ya-Ping Zhang
- State Key Laboratory of Genetic Resources and Evolution, and Center for Excellence in Animal Evolution and Genetics, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, China
| | - Liangxue Lai
- Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Jidong Mi
- Beijing Sinogene Biotechnology Co. Ltd, Beijing, China
| | - Yong-Hui Jiang
- Department of Genetics and Neuroscience, Yale University School of Medicine, New Haven, CT, 06510, USA.
| | - Yong Q Zhang
- State Key Laboratory of Molecular Developmental Biology, and CAS Center for Excellence in Brain Science and Intelligence Technology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China.
- School of Life Sciences, Hubei University, Wuhan, China.
| |
Collapse
|
110
|
Zhang R, Jiang H, Liu Y, He G. Structure, function, and pathology of Neurexin-3. Genes Dis 2023; 10:1908-1919. [PMID: 37492720 PMCID: PMC10363586 DOI: 10.1016/j.gendis.2022.04.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 04/06/2022] [Accepted: 04/08/2022] [Indexed: 11/22/2022] Open
Abstract
Neurexin-3 is primarily localized in the presynaptic membrane and forms complexes with various ligands located in the postsynaptic membrane. Neurexin-3 has important roles in synapse development and synapse functions. Neurexin-3 mediates excitatory presynaptic differentiation by interacting with leucine-rich-repeat transmembrane neuronal proteins. Meanwhile, neurexin-3 modulates the expression of presynaptic α-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid receptors and γ-aminobutyric acid A receptors by interacting with neuroligins at excitatory and inhibitory synapses. Numerous studies have documented the potential contribution of neurexin-3 to neurodegenerative and neuropsychiatric disorders, such as Alzheimer's disease, addiction behaviors, and other diseases, which raises hopes that understanding the mechanisms of neurexin-3 may hold the key to developing new strategies for related illnesses. This review comprehensively covers the literature to provide current knowledge of the structure, function, and clinical role of neurexin-3.
Collapse
Affiliation(s)
- Rui Zhang
- Chongqing Key Laboratory of Neurobiology, Chongqing Medical University, Chongqing 400016, China
| | - HanXiao Jiang
- Department of Neurology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, China
| | - YuanJie Liu
- Chongqing Key Laboratory of Neurobiology, Chongqing Medical University, Chongqing 400016, China
- Department of Anatomy, Chongqing Medical University, Chongqing 400016, China
| | - GuiQiong He
- Chongqing Key Laboratory of Neurobiology, Chongqing Medical University, Chongqing 400016, China
- Department of Anatomy, Chongqing Medical University, Chongqing 400016, China
| |
Collapse
|
111
|
Gonçalves AM, Sousa N, Jacinto L, Monteiro P. The Shank3-InsG3680(+/+) mouse model of autism spectrum disorder displays auditory avoidance in a novel behavioral test. Front Behav Neurosci 2023; 17:1205507. [PMID: 37693284 PMCID: PMC10483143 DOI: 10.3389/fnbeh.2023.1205507] [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: 04/13/2023] [Accepted: 07/26/2023] [Indexed: 09/12/2023] Open
Abstract
Introduction Autism spectrum disorder (ASD) is characterized by deficits in communication and social interaction, restricted interests, repetitive behaviors, and sensory alterations, with auditory hypersensitivity being one of the most commonly reported sensory-perceptual abnormalities. Several candidate genes for involvement in this disorder have emerged from patient studies, including SHANK3, a gene that encodes a protein (SHANK3) in the postsynaptic density of excitatory synapses. Previous work has shown that mutant mice carrying a human ASD mutation in the Shank3 gene (InsG3680) exhibit repetitive behaviors and social interaction deficits, indicating important construct and face validity for this genotype as an animal model of ASD. Methods To further address whether these mice also present auditory sensory-perceptual alterations, we developed a novel behavioral test in which mice can choose between different soundscapes. Results Our results reveal that, in comparison to wild-type mice, Shank3 mutants display a strong behavioral preference toward silent regions of the arena. Discussion These data suggest that Shank3- mutant mice might express an auditory hypersensitivity phenotype, further adding to the face validity of this genotype as an animal model of ASD.
Collapse
Affiliation(s)
- Ana Margarida Gonçalves
- Life and Health Sciences Research Institute, School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B’s—PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Nuno Sousa
- Life and Health Sciences Research Institute, School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B’s—PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Luis Jacinto
- Experimental Biology Unit, Department of Biomedicine, Faculty of Medicine, University of Porto (FMUP), Porto, Portugal
| | - Patricia Monteiro
- Life and Health Sciences Research Institute, School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B’s—PT Government Associate Laboratory, Braga/Guimarães, Portugal
- Experimental Biology Unit, Department of Biomedicine, Faculty of Medicine, University of Porto (FMUP), Porto, Portugal
| |
Collapse
|
112
|
Dawson MS, Gordon-Fleet K, Yan L, Tardos V, He H, Mui K, Nawani S, Asgarian Z, Catani M, Fernandes C, Drescher U. Sexual dimorphism in the social behaviour of Cntnap2-null mice correlates with disrupted synaptic connectivity and increased microglial activity in the anterior cingulate cortex. Commun Biol 2023; 6:846. [PMID: 37582968 PMCID: PMC10427688 DOI: 10.1038/s42003-023-05215-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 08/04/2023] [Indexed: 08/17/2023] Open
Abstract
A biological understanding of the apparent sex bias in autism is lacking. Here we have identified Cntnap2 KO mice as a model system to help better understand this dimorphism. Using this model, we observed social deficits in juvenile male KO mice only. These male-specific social deficits correlated with reduced spine densities of Layer 2/3 and Layer 5 pyramidal neurons in the Anterior Cingulate Cortex, a forebrain region prominently associated with the control of social behaviour. Furthermore, in male KO mice, microglia showed an increased activated morphology and phagocytosis of synaptic structures compared to WT mice, whereas no differences were seen in female KO and WT mice. Our data suggest that sexually dimorphic microglial activity may be involved in the aetiology of ASD, disrupting the development of neural circuits that control social behaviour by overpruning synapses at a developmentally critical period.
Collapse
Affiliation(s)
- Matt S Dawson
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King's College London, London, SE1 1UL, UK
| | - Kevin Gordon-Fleet
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King's College London, London, SE1 1UL, UK
| | - Lingxin Yan
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King's College London, London, SE1 1UL, UK
| | - Vera Tardos
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King's College London, London, SE1 1UL, UK
| | - Huanying He
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King's College London, London, SE1 1UL, UK
| | - Kwong Mui
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King's College London, London, SE1 1UL, UK
| | - Smriti Nawani
- Social, Genetic & Developmental Psychiatry Centre, IoPPN, King's College London, London, SE1 1UL, UK
| | - Zeinab Asgarian
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King's College London, London, SE1 1UL, UK
- Molecular Therapeutics Lab, University College London, Research Department of Targeted Intervention, London, W1W 7TY, UK
| | - Marco Catani
- NatBrainLab, Departments of Neuroimaging Sciences and Forensic and Neurodevelopmental Sciences, IoPPN, King's College London, London, SE1 1UL, UK
| | - Cathy Fernandes
- Social, Genetic & Developmental Psychiatry Centre, IoPPN, King's College London, London, SE1 1UL, UK
- MRC Centre for Neurodevelopmental Disorders, IoPPN, King's College London, London, SE1 1UL, UK
| | - Uwe Drescher
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King's College London, London, SE1 1UL, UK.
- MRC Centre for Neurodevelopmental Disorders, IoPPN, King's College London, London, SE1 1UL, UK.
| |
Collapse
|
113
|
Fenton TA, Haouchine OY, Hallam EL, Smith EM, Jackson KC, Rahbarian D, Canales C, Adhikari A, Nord AS, Ben-Shalom R, Silverman JL. Hyperexcitability and translational phenotypes in a preclinical model of SYNGAP1 mutations. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.24.550093. [PMID: 37546838 PMCID: PMC10402099 DOI: 10.1101/2023.07.24.550093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
Abstract
SYNGAP1 is a critical gene for neuronal development, synaptic structure, and function. Although rare, the disruption of SYNGAP1 directly causes a genetically identifiable neurodevelopmental disorder (NDD) called SYNGAP1-related intellectual disability. Without functional SynGAP1 protein, patients present with intellectual disability, motor impairments, and epilepsy. Previous work using mouse models with a variety of germline and conditional mutations has helped delineate SynGAP1's critical roles in neuronal structure and function, as well as key biochemical signaling pathways essential to synapse integrity. Homozygous loss of SYNGAP1 is embryonically lethal. Heterozygous mutations of SynGAP1 result in a broad range of phenotypes including increased locomotor activity, impaired working spatial memory, impaired cued fear memory, and increased stereotypic behavior. Our in vivo functional data, using the original germline mutation mouse line from the Huganir laboratory, corroborated robust hyperactivity and learning and memory deficits. Here, we describe impairments in the translational biomarker domain of sleep, characterized using neurophysiological data collected with wireless telemetric electroencephalography (EEG). We discovered Syngap1 +/- mice exhibited elevated spike trains in both number and duration, in addition to elevated power, most notably in the delta power band. Primary neurons from Syngap1 +/- mice displayed increased network firing activity, greater spikes per burst, and shorter inter-burst intervals between peaks using high density micro-electrode arrays (HD-MEA). This work is translational, innovative, and highly significant as it outlines functional impairments in Syngap1 mutant mice. Simultaneously, the work utilized untethered, wireless neurophysiology that can discover potential biomarkers of Syngap1R-ID, for clinical trials, as it has done with other NDDs. Our work is substantial forward progress toward translational work for SynGAP1R-ID as it bridges in-vitro electrophysiological neuronal activity and function with in vivo neurophysiological brain activity and function. These data elucidate multiple quantitative, translational biomarkers in vivo and in vitro for the development of treatments for SYNGAP1-related intellectual disability.
Collapse
|
114
|
Hussein Y, Tripathi U, Choudhary A, Nayak R, Peles D, Rosh I, Rabinski T, Djamus J, Vatine GD, Spiegel R, Garin-Shkolnik T, Stern S. Early maturation and hyperexcitability is a shared phenotype of cortical neurons derived from different ASD-associated mutations. Transl Psychiatry 2023; 13:246. [PMID: 37414777 PMCID: PMC10326262 DOI: 10.1038/s41398-023-02535-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 06/18/2023] [Accepted: 06/21/2023] [Indexed: 07/08/2023] Open
Abstract
Autism Spectrum Disorder (ASD) is characterized mainly by social and sensory-motor abnormal and repetitive behavior patterns. Over hundreds of genes and thousands of genetic variants were reported to be highly penetrant and causative of ASD. Many of these mutations cause comorbidities such as epilepsy and intellectual disabilities (ID). In this study, we measured cortical neurons derived from induced pluripotent stem cells (iPSCs) of patients with four mutations in the genes GRIN2B, SHANK3, UBTF, as well as chromosomal duplication in the 7q11.23 region and compared them to neurons derived from a first-degree relative without the mutation. Using a whole-cell patch-clamp, we observed that the mutant cortical neurons demonstrated hyperexcitability and early maturation compared to control lines. These changes were characterized by increased sodium currents, increased amplitude and rate of excitatory postsynaptic currents (EPSCs), and more evoked action potentials in response to current stimulation in early-stage cell development (3-5 weeks post differentiation). These changes that appeared in all the different mutant lines, together with previously reported data, indicate that an early maturation and hyperexcitability may be a convergent phenotype of ASD cortical neurons.
Collapse
Affiliation(s)
- Yara Hussein
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel
| | - Utkarsh Tripathi
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel
| | - Ashwani Choudhary
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel
| | - Ritu Nayak
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel
| | - David Peles
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel
| | - Idan Rosh
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel
| | - Tatiana Rabinski
- The Department of Physiology and Cell Biology, Faculty of Health Sciences and the Regenerative Medicine and Stem Cell (RMSC) Research Center, Ben-Gurion University of the Negev, Beer Sheva, Israel
- The Regenerative Medicine and Stem Cell (RMSC) Research Center, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Jose Djamus
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel
| | - Gad David Vatine
- The Department of Physiology and Cell Biology, Faculty of Health Sciences and the Regenerative Medicine and Stem Cell (RMSC) Research Center, Ben-Gurion University of the Negev, Beer Sheva, Israel
- The Regenerative Medicine and Stem Cell (RMSC) Research Center, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Ronen Spiegel
- Center for Rare Diseases, Emek Medical Center, Afula, Israel
| | | | - Shani Stern
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel.
| |
Collapse
|
115
|
Yamada T, Watanabe T, Sasaki Y. Are sleep disturbances a cause or consequence of autism spectrum disorder? Psychiatry Clin Neurosci 2023; 77:377-385. [PMID: 36949621 PMCID: PMC10871071 DOI: 10.1111/pcn.13550] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 03/10/2023] [Accepted: 03/17/2023] [Indexed: 03/24/2023]
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder characterized by core symptoms such as atypical social communication, stereotyped behaviors, and restricted interests. One of the comorbid symptoms of individuals with ASD is sleep disturbance. There are two major hypotheses regarding the neural mechanism underlying ASD, i.e., the excitation/inhibition (E/I) imbalance and the altered neuroplasticity hypotheses. However, the pathology of ASD remains unclear due to inconsistent research results. This paper argues that sleep is a confounding factor, thus, must be considered when examining the pathology of ASD because sleep plays an important role in modulating the E/I balance and neuroplasticity in the human brain. Investigation of the E/I balance and neuroplasticity during sleep might enhance our understanding of the neural mechanisms of ASD. It may also lead to the development of neurobiologically informed interventions to supplement existing psychosocial therapies.
Collapse
Affiliation(s)
- Takashi Yamada
- Department of Cognitive, Linguistic, and Psychological Sciences, Brown University, Providence, 02912, USA
| | - Takeo Watanabe
- Department of Cognitive, Linguistic, and Psychological Sciences, Brown University, Providence, 02912, USA
| | - Yuka Sasaki
- Department of Cognitive, Linguistic, and Psychological Sciences, Brown University, Providence, 02912, USA
| |
Collapse
|
116
|
Kareklas K, Teles MC, Dreosti E, Oliveira RF. Autism-associated gene shank3 is necessary for social contagion in zebrafish. Mol Autism 2023; 14:23. [PMID: 37391856 PMCID: PMC10311831 DOI: 10.1186/s13229-023-00555-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 06/19/2023] [Indexed: 07/02/2023] Open
Abstract
BACKGROUND Animal models enable targeting autism-associated genes, such as the shank3 gene, to assess their impact on behavioural phenotypes. However, this is often limited to simple behaviours relevant for social interaction. Social contagion is a complex phenotype forming the basis of human empathic behaviour and involves attention to the behaviour of others for recognizing and sharing their emotional or affective state. Thus, it is a form of social communication, which constitutes the most common developmental impairment across autism spectrum disorders (ASD). METHODS Here we describe the development of a zebrafish model that identifies the neurocognitive mechanisms by which shank3 mutation drives deficits in social contagion. We used a CRISPR-Cas9 technique to generate mutations to the shank3a gene, a zebrafish paralogue found to present greater orthology and functional conservation relative to the human gene. Mutants were first compared to wild types during a two-phase protocol that involves the observation of two conflicting states, distress and neutral, and the later recall and discrimination of others when no longer presenting such differences. Then, the whole-brain expression of different neuroplasticity markers was compared between genotypes and their contribution to cluster-specific phenotypic variation was assessed. RESULTS The shank3 mutation markedly reduced social contagion via deficits in attention contributing to difficulties in recognising affective states. Also, the mutation changed the expression of neuronal plasticity genes. However, only downregulated neuroligins clustered with shank3a expression under a combined synaptogenesis component that contributed specifically to variation in attention. LIMITATIONS While zebrafish are extremely useful in identifying the role of shank3 mutations to composite social behaviour, they are unlikely to represent the full complexity of socio-cognitive and communication deficits presented by human ASD pathology. Moreover, zebrafish cannot represent the scaling up of these deficits to higher-order empathic and prosocial phenotypes seen in humans. CONCLUSIONS We demonstrate a causal link between the zebrafish orthologue of an ASD-associated gene and the attentional control of affect recognition and consequent social contagion. This models autistic affect-communication pathology in zebrafish and reveals a genetic attention-deficit mechanism, addressing the ongoing debate for such mechanisms accounting for emotion recognition difficulties in autistic individuals.
Collapse
Affiliation(s)
- Kyriacos Kareklas
- Instituto Gulbenkian de Ciência, R. Q.ta Grande 6, 2780-156, Oeiras, Portugal
| | - Magda C Teles
- Instituto Gulbenkian de Ciência, R. Q.ta Grande 6, 2780-156, Oeiras, Portugal
- ISPA - Instituto Universitário, Rua Jardim do Tabaco 34, 1149-041, Lisbon, Portugal
| | - Elena Dreosti
- Department of Cell and Developmental Biology, University College London, London, UK
| | - Rui F Oliveira
- Instituto Gulbenkian de Ciência, R. Q.ta Grande 6, 2780-156, Oeiras, Portugal.
- ISPA - Instituto Universitário, Rua Jardim do Tabaco 34, 1149-041, Lisbon, Portugal.
| |
Collapse
|
117
|
Landry O, François A, Oye Mintsa Mi-Mba MF, Traversy MT, Tremblay C, Emond V, Bennett DA, Gylys KH, Buxbaum JD, Calon F. Postsynaptic Protein Shank3a Deficiency Synergizes with Alzheimer's Disease Neuropathology to Impair Cognitive Performance in the 3xTg-AD Murine Model. J Neurosci 2023; 43:4941-4954. [PMID: 37253603 PMCID: PMC10312061 DOI: 10.1523/jneurosci.1945-22.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 04/17/2023] [Accepted: 05/04/2023] [Indexed: 06/01/2023] Open
Abstract
Synaptic loss is intrinsically linked to Alzheimer's disease (AD) neuropathology and symptoms, but its direct impact on clinical symptoms remains elusive. The postsynaptic protein Shank3 (SH3 and multiple ankyrin repeat domains) is of particular interest, as the loss of a single allele of the SHANK3 gene is sufficient to cause profound cognitive symptoms in children. We thus sought to determine whether a SHANK3 deficiency could contribute to the emergence or worsening of AD symptoms and neuropathology. We first found a 30%-50% postmortem loss of SHANK3a associated with cognitive decline in the parietal cortex of individuals with AD. To further probe the role of SHANK3 in AD, we crossed male and female 3xTg-AD mice modelling Aβ and tau pathologies with Shank3a-deficient mice (Shank3Δex4-9). We observed synergistic deleterious effects of Shank3a deficiency and AD neuropathology on object recognition memory at 9, 12, and 18 months of age and on anxious behavior at 9 and 12 months of age in hemizygous Shank3Δex4-9-3xTg-AD mice. In addition to the expected 50% loss of Shank3a, levels of other synaptic proteins, such as PSD-95, drebrin, and homer1, remained unchanged in the parietotemporal cortex of hemizygous Shank3Δex4-9 animals. However, Shank3a deficiency increased the levels of soluble Aβ42 and human tau at 18 months of age compared with 3xTg-AD mice with normal Shank3 expression. The results of this study in human brain samples and in transgenic mice are consistent with the hypothesis that Shank3 deficiency makes a key contribution to cognitive impairment in AD.SIGNIFICANCE STATEMENT Although the loss of several synaptic proteins has been described in Alzheimer's disease (AD), it remains unclear whether their reduction contributes to clinical symptoms. The results of this study in human samples show lower levels of SHANK3a in AD brain, correlating with cognitive decline. Data gathered in a novel transgenic mouse suggest that Shank3a deficiency synergizes with AD neuropathology to induce cognitive impairment, consistent with a causal role in AD. Therefore, treatment aiming at preserving Shank3 in the aging brain may be beneficial to prevent AD.
Collapse
Affiliation(s)
- Olivier Landry
- Faculté de pharmacie, Université Laval, Quebec G1V 0A6, Quebec, Canada
- Axe Neurosciences, Centre de recherche du CHU de Québec-Université Laval, Quebec G1V 4G2, Quebec, Canada
| | - Arnaud François
- Faculté de pharmacie, Université Laval, Quebec G1V 0A6, Quebec, Canada
- Axe Neurosciences, Centre de recherche du CHU de Québec-Université Laval, Quebec G1V 4G2, Quebec, Canada
| | - Méryl-Farelle Oye Mintsa Mi-Mba
- Faculté de pharmacie, Université Laval, Quebec G1V 0A6, Quebec, Canada
- Axe Neurosciences, Centre de recherche du CHU de Québec-Université Laval, Quebec G1V 4G2, Quebec, Canada
| | - Marie-Therese Traversy
- Faculté de pharmacie, Université Laval, Quebec G1V 0A6, Quebec, Canada
- Axe Neurosciences, Centre de recherche du CHU de Québec-Université Laval, Quebec G1V 4G2, Quebec, Canada
| | - Cyntia Tremblay
- Axe Neurosciences, Centre de recherche du CHU de Québec-Université Laval, Quebec G1V 4G2, Quebec, Canada
| | - Vincent Emond
- Axe Neurosciences, Centre de recherche du CHU de Québec-Université Laval, Quebec G1V 4G2, Quebec, Canada
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, Illinois 60612
| | - Karen H Gylys
- School of Nursing, University of California, Los Angeles, California 90095
| | - Joseph D Buxbaum
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York 10029, New York
| | - Frédéric Calon
- Faculté de pharmacie, Université Laval, Quebec G1V 0A6, Quebec, Canada
- Axe Neurosciences, Centre de recherche du CHU de Québec-Université Laval, Quebec G1V 4G2, Quebec, Canada
| |
Collapse
|
118
|
Molloy CJ, Cooke J, Gatford NJF, Rivera-Olvera A, Avazzadeh S, Homberg JR, Grandjean J, Fernandes C, Shen S, Loth E, Srivastava DP, Gallagher L. Bridging the translational gap: what can synaptopathies tell us about autism? Front Mol Neurosci 2023; 16:1191323. [PMID: 37441676 PMCID: PMC10333541 DOI: 10.3389/fnmol.2023.1191323] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 05/24/2023] [Indexed: 07/15/2023] Open
Abstract
Multiple molecular pathways and cellular processes have been implicated in the neurobiology of autism and other neurodevelopmental conditions. There is a current focus on synaptic gene conditions, or synaptopathies, which refer to clinical conditions associated with rare genetic variants disrupting genes involved in synaptic biology. Synaptopathies are commonly associated with autism and developmental delay and may be associated with a range of other neuropsychiatric outcomes. Altered synaptic biology is suggested by both preclinical and clinical studies in autism based on evidence of differences in early brain structural development and altered glutamatergic and GABAergic neurotransmission potentially perturbing excitatory and inhibitory balance. This review focusses on the NRXN-NLGN-SHANK pathway, which is implicated in the synaptic assembly, trans-synaptic signalling, and synaptic functioning. We provide an overview of the insights from preclinical molecular studies of the pathway. Concentrating on NRXN1 deletion and SHANK3 mutations, we discuss emerging understanding of cellular processes and electrophysiology from induced pluripotent stem cells (iPSC) models derived from individuals with synaptopathies, neuroimaging and behavioural findings in animal models of Nrxn1 and Shank3 synaptic gene conditions, and key findings regarding autism features, brain and behavioural phenotypes from human clinical studies of synaptopathies. The identification of molecular-based biomarkers from preclinical models aims to advance the development of targeted therapeutic treatments. However, it remains challenging to translate preclinical animal models and iPSC studies to interpret human brain development and autism features. We discuss the existing challenges in preclinical and clinical synaptopathy research, and potential solutions to align methodologies across preclinical and clinical research. Bridging the translational gap between preclinical and clinical studies will be necessary to understand biological mechanisms, to identify targeted therapies, and ultimately to progress towards personalised approaches for complex neurodevelopmental conditions such as autism.
Collapse
Affiliation(s)
- Ciara J. Molloy
- Department of Psychiatry, School of Medicine, Trinity College Dublin, Dublin, Ireland
| | - Jennifer Cooke
- Forensic and Neurodevelopmental Sciences, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kingdom
| | - Nicholas J. F. Gatford
- Kavli Institute for Nanoscience Discovery, Nuffield Department of Clinical Neurosciences, University of Oxford, Medical Sciences Division, Oxford, United Kingdom
| | - Alejandro Rivera-Olvera
- Donders Institute for Brain, Cognition, and Behaviour, Radboud University Medical Centre, Nijmegen, Netherlands
| | - Sahar Avazzadeh
- Physiology and Cellular Physiology Research Laboratory, CÚRAM SFI Centre for Research in Medical Devices, School of Medicine, Human Biology Building, University of Galway, Galway, Ireland
| | - Judith R. Homberg
- Donders Institute for Brain, Cognition, and Behaviour, Radboud University Medical Centre, Nijmegen, Netherlands
| | - Joanes Grandjean
- Physiology and Cellular Physiology Research Laboratory, CÚRAM SFI Centre for Research in Medical Devices, School of Medicine, Human Biology Building, University of Galway, Galway, Ireland
- Department of Medical Imaging, Radboud University Medical Centre, Nijmegen, Netherlands
| | - Cathy Fernandes
- Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kingdom
- MRC Centre for Neurodevelopmental Disorders, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kingdom
| | - Sanbing Shen
- Regenerative Medicine Institute, School of Medicine, University of Galway, Galway, Ireland
- FutureNeuro, The SFI Research Centre for Chronic and Rare Neurological Diseases, Royal College of Surgeons, Dublin, Ireland
| | - Eva Loth
- Forensic and Neurodevelopmental Sciences, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kingdom
| | - Deepak P. Srivastava
- MRC Centre for Neurodevelopmental Disorders, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kingdom
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kingdom
| | - Louise Gallagher
- Department of Psychiatry, School of Medicine, Trinity College Dublin, Dublin, Ireland
- The Hospital for SickKids, Toronto, ON, Canada
- The Peter Gilgan Centre for Research and Learning, SickKids Research Institute, Toronto, ON, Canada
- The Centre for Addiction and Mental Health, Toronto, ON, Canada
- Department of Psychiatry, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
119
|
Olde Heuvel F, Ouali Alami N, Aousji O, Pogatzki-Zahn E, Zahn PK, Wilhelm H, Deshpande D, Khatamsaz E, Catanese A, Woelfle S, Schön M, Jain S, Grabrucker S, Ludolph AC, Verpelli C, Michaelis J, Boeckers TM, Roselli F. Shank2 identifies a subset of glycinergic neurons involved in altered nociception in an autism model. Mol Autism 2023; 14:21. [PMID: 37316943 DOI: 10.1186/s13229-023-00552-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 05/26/2023] [Indexed: 06/16/2023] Open
Abstract
BACKGROUND Autism Spectrum Disorders (ASD) patients experience disturbed nociception in the form of either hyposensitivity to pain or allodynia. A substantial amount of processing of somatosensory and nociceptive stimulus takes place in the dorsal spinal cord. However, many of these circuits are not very well understood in the context of nociceptive processing in ASD. METHODS We have used a Shank2-/- mouse model, which displays a set of phenotypes reminiscent of ASD, and performed behavioural and microscopic analysis to investigate the role of dorsal horn circuitry in nociceptive processing of ASD. RESULTS We determined that Shank2-/- mice display increased sensitivity to formalin pain and thermal preference, but a sensory specific mechanical allodynia. We demonstrate that high levels of Shank2 expression identifies a subpopulation of neurons in murine and human dorsal spinal cord, composed mainly by glycinergic interneurons and that loss of Shank2 causes the decrease in NMDAR in excitatory synapses on these inhibitory interneurons. In fact, in the subacute phase of the formalin test, glycinergic interneurons are strongly activated in wild type (WT) mice but not in Shank2-/- mice. Consequently, nociception projection neurons in laminae I are activated in larger numbers in Shank2-/- mice. LIMITATIONS Our investigation is limited to male mice, in agreement with the higher representation of ASD in males; therefore, caution should be applied to extrapolate the findings to females. Furthermore, ASD is characterized by extensive genetic diversity and therefore the findings related to Shank2 mutant mice may not necessarily apply to patients with different gene mutations. Since nociceptive phenotypes in ASD range between hyper- and hypo-sensitivity, diverse mutations may affect the circuit in opposite ways. CONCLUSION Our findings prove that Shank2 expression identifies a new subset of inhibitory interneurons involved in reducing the transmission of nociceptive stimuli and whose unchecked activation is associated with pain hypersensitivity. We provide evidence that dysfunction in spinal cord pain processing may contribute to the nociceptive phenotypes in ASD.
Collapse
Affiliation(s)
| | - Najwa Ouali Alami
- Department of Neurology, Ulm University, Ulm, Germany
- International PhD Program, Ulm University, Ulm, Germany
| | | | - Esther Pogatzki-Zahn
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Münster, Münster, Germany
| | - Peter K Zahn
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Münster, Münster, Germany
- Clinic for Anesthesiology, Intensive Care and Pain Medicine, University Hospital Bergmannsheil Bochum, Bochum, Germany
| | - Hanna Wilhelm
- Department of Neurology, Ulm University, Ulm, Germany
| | | | | | - Alberto Catanese
- Institute of Anatomy and Cell Biology, Ulm University, Ulm, Germany
| | - Sarah Woelfle
- Institute of Anatomy and Cell Biology, Ulm University, Ulm, Germany
| | - Michael Schön
- Institute of Anatomy and Cell Biology, Ulm University, Ulm, Germany
| | - Sanjay Jain
- Department of Internal Medicine (Renal), Pathology and Immunology, Washington University School of Medicine, St. Louis, USA
| | | | - Albert C Ludolph
- Department of Neurology, Ulm University, Ulm, Germany
- German Center for Neurodegenerative Diseases (DZNE), Ulm, Germany
| | - Chiara Verpelli
- Institute of Neuroscience, National Science Council, Milan, Italy
| | | | - Tobias M Boeckers
- Institute of Anatomy and Cell Biology, Ulm University, Ulm, Germany.
- German Center for Neurodegenerative Diseases (DZNE), Ulm, Germany.
- Department of Anatomy and Cell Biology, Ulm University, Albert-Einstein Allee 11, 89081, Ulm, Germany.
| | - Francesco Roselli
- Department of Neurology, Ulm University, Ulm, Germany.
- German Center for Neurodegenerative Diseases (DZNE), Ulm, Germany.
- Center for Biomedical Research (ZBF), Helmholtzstraße 8/2, 89081, Ulm, Germany.
| |
Collapse
|
120
|
Wang D, Xie D, Zhang J, Cai B, Yang B, Zhou L, Huang X. Comprehensive analysis of the coding and non-coding RNA transcriptome expression profiles of hippocampus tissue in tx-J animal model of Wilson's disease. Sci Rep 2023; 13:9252. [PMID: 37286730 DOI: 10.1038/s41598-023-36503-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 06/05/2023] [Indexed: 06/09/2023] Open
Abstract
Wilson's disease (WD) is an autosomal recessive disorder with a genetic basis. The predominant non-motor symptom of WD is cognitive dysfunction, although the specific genetic regulatory mechanism remains unclear. Tx-J mice, with an 82% sequence homology of the ATP7B gene to the human gene, are considered the most suitable model for WD. This study employs deep sequencing to investigate the differences in RNA transcript profiles, both coding and non-coding, as well as the functional characteristics of the regulatory network involved in WD cognitive impairment. The cognitive function of tx-J mice was evaluated using the Water Maze Test (WMT). Long non-coding RNA (lncRNA), circular RNA (circRNA), and messenger RNA (mRNA) profiles were analyzed in the hippocampal tissue of tx-J mice to identify differentially expressed RNAs (DE-RNAs). Subsequently, the DE-RNAs were used to construct protein-protein interaction (PPI) networks, as well as DE-circRNAs and lncRNAs-associated competing endogenous RNA (ceRNA) expression networks, and coding-noncoding co-expression (CNC) networks. To elucidate their biological functions and pathways, the PPI and ceRNA networks were subjected to Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis. A total of 361 differentially expressed mRNAs (DE-mRNAs), comprising 193 up-regulated and 168 down-regulated mRNAs, 2627 differentially expressed long non-coding RNAs (DE-lncRNAs), consisting of 1270 up-regulated and 1357 down-regulated lncRNAs, and 99 differentially expressed circular RNAs (DE-circRNAs), consisting of 68 up-regulated and 31 down-regulated circRNAs, were observed in the tx-J mice group when compared to the control mice group. Gene Ontology (GO) and pathway analyses revealed that DE-mRNAs were enriched in cellular processes, calcium signaling pathways, and mRNA surveillance pathways. In contrast, the DE-circRNAs-associated competing endogenous RNA (ceRNA) network was enriched for covalent chromatin modification, histone modification, and axon guidance, whereas the DE-lncRNAs-associated ceRNA network was enriched for dendritic spine, regulation of cell morphogenesis involved in differentiation, and mRNA surveillance pathway. The study presented the expression profiles of lncRNA, circRNA, and mRNA in the hippocampal tissue of tx-J mice. Furthermore, the study constructed PPI, ceRNA, and CNC expression networks. The findings are significant in comprehending the function of regulatory genes in WD associated with cognitive impairment. These results also offer valuable information for the diagnosis and treatment of WD.
Collapse
Affiliation(s)
- Dan Wang
- College of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, No. 1 Qianjiang Road, Xinzhan District, Hefei, 230012, People's Republic of China
| | - Daojun Xie
- Encephalopathy Center, The First Affiliated Hospital of Anhui University of Chinese Medicine, No. 117 Meishan Road, Shushan District, Hefei, 230031, People's Republic of China.
| | - Juan Zhang
- Encephalopathy Center, The First Affiliated Hospital of Anhui University of Chinese Medicine, No. 117 Meishan Road, Shushan District, Hefei, 230031, People's Republic of China
| | - Biao Cai
- College of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, No. 1 Qianjiang Road, Xinzhan District, Hefei, 230012, People's Republic of China
| | - Bo Yang
- Encephalopathy Center, The First Affiliated Hospital of Anhui University of Chinese Medicine, No. 117 Meishan Road, Shushan District, Hefei, 230031, People's Republic of China
| | - Lei Zhou
- Encephalopathy Center, The First Affiliated Hospital of Anhui University of Chinese Medicine, No. 117 Meishan Road, Shushan District, Hefei, 230031, People's Republic of China
| | - Xiaofeng Huang
- Encephalopathy Center, The First Affiliated Hospital of Anhui University of Chinese Medicine, No. 117 Meishan Road, Shushan District, Hefei, 230031, People's Republic of China
| |
Collapse
|
121
|
Gao SQ, Chen JQ, Zhou HY, Luo L, Zhang BY, Li MT, He HY, Chen C, Guo Y. Thrombospondin1 mimics rapidly relieve depression via Shank3 dependent uncoupling between dopamine D1 and D2 receptors. iScience 2023; 26:106488. [PMID: 37091229 PMCID: PMC10119609 DOI: 10.1016/j.isci.2023.106488] [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: 12/10/2020] [Revised: 06/17/2022] [Accepted: 03/18/2023] [Indexed: 04/25/2023] Open
Abstract
Deficits in astrocyte function contribute to major depressive disorder (MDD) and suicide, but the therapeutic effect of directly reactivating astrocytes for depression remains unclear. Here, specific gains and losses of astrocytic cell functions in the medial prefrontal cortex (mPFC) bidirectionally regulate depression-like symptoms. Remarkably, recombinant human Thrombospondin-1 (rhTSP1), an astrocyte-secreted protein, exerted rapidly antidepressant-like actions through tyrosine hydroxylase (Th)/dopamine (DA)/dopamine D2 receptors (D2Rs) pathways, but not dopamine D1 receptors (D1Rs), which was dependent on SH3 and multiple ankyrin repeat domains 3 (Shank3) in the mPFC. TSP1 in the mPFC might have potential as a target for treating clinical depression.
Collapse
Affiliation(s)
- Shuang-Qi Gao
- Departments of Neurosurgery, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province 510630, China
- Corresponding author
| | - Jun-Quan Chen
- Departments of Neurosurgery, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province 510630, China
| | - Hai-Yun Zhou
- Department of Pharmacy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Lun Luo
- Departments of Neurosurgery, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province 510630, China
| | - Bao-Yu Zhang
- Departments of Neurosurgery, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province 510630, China
| | - Man-Ting Li
- Departments of Neurosurgery, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province 510630, China
| | - Hai-Yong He
- Departments of Neurosurgery, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province 510630, China
| | - Chuan Chen
- Departments of Neurosurgery, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province 510630, China
- Corresponding author
| | - Ying Guo
- Departments of Neurosurgery, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province 510630, China
- Corresponding author
| |
Collapse
|
122
|
Guerra M, Medici V, Weatheritt R, Corvino V, Palacios D, Geloso MC, Farini D, Sette C. Fetal exposure to valproic acid dysregulates the expression of autism-linked genes in the developing cerebellum. Transl Psychiatry 2023; 13:114. [PMID: 37019889 PMCID: PMC10076313 DOI: 10.1038/s41398-023-02391-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 02/27/2023] [Accepted: 03/01/2023] [Indexed: 04/07/2023] Open
Abstract
Autism spectrum disorder (ASD) includes a set of highly heritable neurodevelopmental syndromes characterized by social and communication impairment, repetitive behaviour, and intellectual disability. Although mutations in multiple genes have been associated to ASD, most patients lack detectable genetic alterations. For this reason, environmental factors are commonly thought to also contribute to ASD aetiology. Transcriptome analyses have revealed that autistic brains possess distinct gene expression signatures, whose elucidation can provide insights about the mechanisms underlying the effects of ASD-causing genetic and environmental factors. Herein, we have identified a coordinated and temporally regulated programme of gene expression in the post-natal development of cerebellum, a brain area whose defects are strongly associated with ASD. Notably, this cerebellar developmental programme is significantly enriched in ASD-linked genes. Clustering analyses highlighted six different patterns of gene expression modulated during cerebellar development, with most of them being enriched in functional processes that are frequently dysregulated in ASD. By using the valproic acid mouse model of ASD, we found that ASD-linked genes are dysregulated in the developing cerebellum of ASD-like mice, a defect that correlates with impaired social behaviour and altered cerebellar cortical morphology. Moreover, changes in transcript levels were reflected in aberrant protein expression, indicating the functional relevance of these alterations. Thus, our work uncovers a complex ASD-related transcriptional programme regulated during cerebellar development and highlight genes whose expression is dysregulated in this brain area of an ASD mouse model.
Collapse
Affiliation(s)
- Marika Guerra
- Department of Neuroscience, Section of Human Anatomy, Catholic University of the Sacred Hearth, Rome, Italy
- GSTeP-Organoids Research Core Facility, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Rome, Italy
| | - Vanessa Medici
- Department of Neuroscience, Section of Human Anatomy, Catholic University of the Sacred Hearth, Rome, Italy
| | - Robert Weatheritt
- Garvan Institute of Medical Research, EMBL Australia, Darlinghurst, NSW, Australia
| | - Valentina Corvino
- Department of Neuroscience, Section of Human Anatomy, Catholic University of the Sacred Hearth, Rome, Italy
| | - Daniela Palacios
- Department of Life Science and Public Health, Section of Biology, Catholic University of the Sacred Hearth, Rome, Italy
| | - Maria Concetta Geloso
- Department of Neuroscience, Section of Human Anatomy, Catholic University of the Sacred Hearth, Rome, Italy
- GSTeP-Organoids Research Core Facility, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Rome, Italy
| | - Donatella Farini
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | - Claudio Sette
- Department of Neuroscience, Section of Human Anatomy, Catholic University of the Sacred Hearth, Rome, Italy.
- GSTeP-Organoids Research Core Facility, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Rome, Italy.
| |
Collapse
|
123
|
Cope EC, Wang SH, Waters RC, Gore IR, Vasquez B, Laham BJ, Gould E. Activation of the CA2-ventral CA1 pathway reverses social discrimination dysfunction in Shank3B knockout mice. Nat Commun 2023; 14:1750. [PMID: 36991001 PMCID: PMC10060401 DOI: 10.1038/s41467-023-37248-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 03/07/2023] [Indexed: 03/30/2023] Open
Abstract
Mutation or deletion of the SHANK3 gene, which encodes a synaptic scaffolding protein, is linked to autism spectrum disorder and Phelan-McDermid syndrome, conditions associated with social memory impairments. Shank3B knockout mice also exhibit social memory deficits. The CA2 region of the hippocampus integrates numerous inputs and sends a major output to the ventral CA1 (vCA1). Despite finding few differences in excitatory afferents to the CA2 in Shank3B knockout mice, we found that activation of CA2 neurons as well as the CA2-vCA1 pathway restored social recognition function to wildtype levels. vCA1 neuronal oscillations have been linked to social memory, but we observed no differences in these measures between wildtype and Shank3B knockout mice. However, activation of the CA2 enhanced vCA1 theta power in Shank3B knockout mice, concurrent with behavioral improvements. These findings suggest that stimulating adult circuitry in a mouse model with neurodevelopmental impairments can invoke latent social memory function.
Collapse
Affiliation(s)
- Elise C Cope
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, 08544, USA
- Department of Neuroscience, University of Virginia School of Medicine, Charlottesville, VA, 22908, USA
| | - Samantha H Wang
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, 08544, USA
| | - Renée C Waters
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, 08544, USA
| | - Isha R Gore
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, 08544, USA
| | - Betsy Vasquez
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, 08544, USA
| | - Blake J Laham
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, 08544, USA
| | - Elizabeth Gould
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, 08544, USA.
| |
Collapse
|
124
|
Galgani A, Bartolini E, D’Amora M, Faraguna U, Giorgi FS. The Central Noradrenergic System in Neurodevelopmental Disorders: Merging Experimental and Clinical Evidence. Int J Mol Sci 2023; 24:5805. [PMID: 36982879 PMCID: PMC10055776 DOI: 10.3390/ijms24065805] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 03/15/2023] [Accepted: 03/16/2023] [Indexed: 03/30/2023] Open
Abstract
The aim of this article is to highlight the potential role of the locus-coeruleus-noradrenergic (LC-NA) system in neurodevelopmental disorders (NdDs). The LC is the main brain noradrenergic nucleus, key in the regulation of arousal, attention, and stress response, and its early maturation and sensitivity to perinatal damage make it an interesting target for translational research. Clinical data shows the involvement of the LC-NA system in several NdDs, suggesting a pathogenetic role in the development of such disorders. In this context, a new neuroimaging tool, LC Magnetic Resonance Imaging (MRI), has been developed to visualize the LC in vivo and assess its integrity, which could be a valuable tool for exploring morphological alterations in NdD in vivo in humans. New animal models may be used to test the contribution of the LC-NA system to the pathogenic pathways of NdD and to evaluate the efficacy of NA-targeting drugs. In this narrative review, we provide an overview of how the LC-NA system may represent a common pathophysiological and pathogenic mechanism in NdD and a reliable target for symptomatic and disease-modifying drugs. Further research is needed to fully understand the interplay between the LC-NA system and NdD.
Collapse
Affiliation(s)
- Alessandro Galgani
- Department of Translational Research and of New Surgical and Medical Technologies, University of Pisa, 56126 Pisa, Italy; (A.G.)
| | - Emanuele Bartolini
- Department of Developmental Neuroscience, IRCCS Fondazione Stella Maris, 56128 Pisa, Italy
- Tuscany PhD Programme in Neurosciences, 50121 Florence, Italy
| | - Marta D’Amora
- Department of Biology, University of Pisa, 56125 Pisa, Italy
- Istituto Italiano di Tecnologia, 16163 Genova, Italy
| | - Ugo Faraguna
- Department of Translational Research and of New Surgical and Medical Technologies, University of Pisa, 56126 Pisa, Italy; (A.G.)
- Department of Developmental Neuroscience, IRCCS Fondazione Stella Maris, 56128 Pisa, Italy
| | - Filippo Sean Giorgi
- Department of Translational Research and of New Surgical and Medical Technologies, University of Pisa, 56126 Pisa, Italy; (A.G.)
| |
Collapse
|
125
|
Ferhat AT, Verpy E, Biton A, Forget B, De Chaumont F, Mueller F, Le Sourd AM, Coqueran S, Schmitt J, Rochefort C, Rondi-Reig L, Leboucher A, Boland A, Fin B, Deleuze JF, Boeckers TM, Ey E, Bourgeron T. Excessive self-grooming, gene dysregulation and imbalance between the striosome and matrix compartments in the striatum of Shank3 mutant mice. Front Mol Neurosci 2023; 16:1139118. [PMID: 37008785 PMCID: PMC10061084 DOI: 10.3389/fnmol.2023.1139118] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 02/16/2023] [Indexed: 03/18/2023] Open
Abstract
Autism is characterized by atypical social communication and stereotyped behaviors. Mutations in the gene encoding the synaptic scaffolding protein SHANK3 are detected in 1-2% of patients with autism and intellectual disability, but the mechanisms underpinning the symptoms remain largely unknown. Here, we characterized the behavior of Shank3 Δ11/Δ11 mice from 3 to 12 months of age. We observed decreased locomotor activity, increased stereotyped self-grooming and modification of socio-sexual interaction compared to wild-type littermates. We then used RNAseq on four brain regions of the same animals to identify differentially expressed genes (DEGs). DEGs were identified mainly in the striatum and were associated with synaptic transmission (e.g., Grm2, Dlgap1), G-protein-signaling pathways (e.g., Gnal, Prkcg1, and Camk2g), as well as excitation/inhibition balance (e.g., Gad2). Downregulated and upregulated genes were enriched in the gene clusters of medium-sized spiny neurons expressing the dopamine 1 (D1-MSN) and the dopamine 2 receptor (D2-MSN), respectively. Several DEGs (Cnr1, Gnal, Gad2, and Drd4) were reported as striosome markers. By studying the distribution of the glutamate decarboxylase GAD65, encoded by Gad2, we showed that the striosome compartment of Shank3 Δ11/Δ11 mice was enlarged and displayed much higher expression of GAD65 compared to wild-type mice. Altogether, these results indicate altered gene expression in the striatum of Shank3-deficient mice and strongly suggest, for the first time, that the excessive self-grooming of these mice is related to an imbalance in the striatal striosome and matrix compartments.
Collapse
Affiliation(s)
- Allain-Thibeault Ferhat
- Génétique Humaine et Fonctions Cognitives, Institut Pasteur, CNRS UMR 3571, IUF, Université Paris Cité, Paris, France
- Department of Neuroscience, Columbia University Irving Medical Center, New York, NY, United States
- Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, United States
| | - Elisabeth Verpy
- Génétique Humaine et Fonctions Cognitives, Institut Pasteur, CNRS UMR 3571, IUF, Université Paris Cité, Paris, France
| | - Anne Biton
- Bioinformatics and Biostatistics Hub, Institut Pasteur, Université Paris Cité, Paris, France
| | - Benoît Forget
- Génétique Humaine et Fonctions Cognitives, Institut Pasteur, CNRS UMR 3571, IUF, Université Paris Cité, Paris, France
| | - Fabrice De Chaumont
- Génétique Humaine et Fonctions Cognitives, Institut Pasteur, CNRS UMR 3571, IUF, Université Paris Cité, Paris, France
| | - Florian Mueller
- Imagerie et Modélisation, Institut Pasteur, CNRS UMR 3691, Paris, France
| | - Anne-Marie Le Sourd
- Génétique Humaine et Fonctions Cognitives, Institut Pasteur, CNRS UMR 3571, IUF, Université Paris Cité, Paris, France
| | - Sabrina Coqueran
- Génétique Humaine et Fonctions Cognitives, Institut Pasteur, CNRS UMR 3571, IUF, Université Paris Cité, Paris, France
| | - Julien Schmitt
- Cerebellum Navigation and Memory Team, Institut de Biologie Paris Seine, Neurosciences Paris Seine, CNRS UMR 8246, Inserm UMR-S 1130, Sorbonne Université, Paris, France
| | - Christelle Rochefort
- Cerebellum Navigation and Memory Team, Institut de Biologie Paris Seine, Neurosciences Paris Seine, CNRS UMR 8246, Inserm UMR-S 1130, Sorbonne Université, Paris, France
| | - Laure Rondi-Reig
- Cerebellum Navigation and Memory Team, Institut de Biologie Paris Seine, Neurosciences Paris Seine, CNRS UMR 8246, Inserm UMR-S 1130, Sorbonne Université, Paris, France
| | - Aziliz Leboucher
- Génétique Humaine et Fonctions Cognitives, Institut Pasteur, CNRS UMR 3571, IUF, Université Paris Cité, Paris, France
| | - Anne Boland
- Centre National de Recherche en Génomique Humaine, CEA, Université Paris-Saclay, Evry, France
| | - Bertrand Fin
- Centre National de Recherche en Génomique Humaine, CEA, Université Paris-Saclay, Evry, France
| | - Jean-François Deleuze
- Centre National de Recherche en Génomique Humaine, CEA, Université Paris-Saclay, Evry, France
- Centre d’Étude du Polymorphisme Humain, Paris, France
| | - Tobias M. Boeckers
- Institute of Anatomy and Cell Biology, Ulm University, Ulm, Germany
- Deutsches Zentrum für Neurodegenerative Erkrankungen, Ulm, Germany
| | - Elodie Ey
- Génétique Humaine et Fonctions Cognitives, Institut Pasteur, CNRS UMR 3571, IUF, Université Paris Cité, Paris, France
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS UMR 7104, Inserm UMR-S 1258, Université de Strasbourg, Illkirch-Graffenstaden, France
| | - Thomas Bourgeron
- Génétique Humaine et Fonctions Cognitives, Institut Pasteur, CNRS UMR 3571, IUF, Université Paris Cité, Paris, France
| |
Collapse
|
126
|
Okuzono S, Fujii F, Matsushita Y, Setoyama D, Shinmyo Y, Taira R, Yonemoto K, Akamine S, Motomura Y, Sanefuji M, Sakurai T, Kawasaki H, Han K, Kato TA, Torisu H, Kang D, Nakabeppu Y, Sakai Y, Ohga S. Shank3a/b isoforms regulate the susceptibility to seizures and thalamocortical development in the early postnatal period of mice. Neurosci Res 2023:S0168-0102(23)00051-2. [PMID: 36871873 DOI: 10.1016/j.neures.2023.03.001] [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: 12/19/2022] [Revised: 02/19/2023] [Accepted: 03/01/2023] [Indexed: 03/07/2023]
Abstract
Epileptic seizures are distinct but frequent comorbidities in children with autism spectrum disorder (ASD). The hyperexcitability of cortical and subcortical neurons appears to be involved in both phenotypes. However, little information is available concerning which genes are involved and how they regulate the excitability of the thalamocortical network. In this study, we investigate whether an ASD-associated gene, SH3 and multiple ankyrin repeat domains 3 (Shank3), plays a unique role in the postnatal development of thalamocortical neurons. We herein report that Shank3a/b, the splicing isoforms of mouse Shank3, were uniquely expressed in the thalamic nuclei, peaking from two to four weeks after birth. Shank3a/b-knockout mice showed lower parvalbumin signals in the thalamic nuclei. Consistently, Shank3a/b-knockout mice were more susceptible to generalized seizures than wild-type mice after kainic acid treatments. Together, these data indicate that NT-Ank domain of Shank3a/b regulates molecular pathways that protect thalamocortical neurons from hyperexcitability during the early postnatal period of mice.
Collapse
Affiliation(s)
- Sayaka Okuzono
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan; Section of Pediatrics, Department of Medicine, Fukuoka Dental College, Fukuoka 814-0193, Japan
| | - Fumihiko Fujii
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Yuki Matsushita
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Daiki Setoyama
- Department of Clinical Chemistry and Laboratory Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Yohei Shinmyo
- Department of Medical Neuroscience, Graduate School of Medical Sciences, Kanazawa University, Kanazawa 920-8640, Japan
| | - Ryoji Taira
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Kousuke Yonemoto
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Satoshi Akamine
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Yoshitomo Motomura
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Masafumi Sanefuji
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Takeshi Sakurai
- Medical Innovation Center, Kyoto University Graduate School of Medicine, Kyoto 606-8507, Japan
| | - Hiroshi Kawasaki
- Department of Medical Neuroscience, Graduate School of Medical Sciences, Kanazawa University, Kanazawa 920-8640, Japan
| | - Kihoon Han
- Department of Neuroscience, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Takahiro A Kato
- Department of Neuropsychiatry, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Hiroyuki Torisu
- Section of Pediatrics, Department of Medicine, Fukuoka Dental College, Fukuoka 814-0193, Japan
| | - Dongchon Kang
- Department of Clinical Chemistry and Laboratory Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Yusaku Nakabeppu
- Division of Neurofunctional Genomics, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan
| | - Yasunari Sakai
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan.
| | - Shouichi Ohga
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| |
Collapse
|
127
|
Jaber M. Genetic and environmental mouse models of autism reproduce the spectrum of the disease. J Neural Transm (Vienna) 2023; 130:425-432. [PMID: 36318343 DOI: 10.1007/s00702-022-02555-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 10/17/2022] [Indexed: 03/23/2023]
Abstract
Genetic and environmental factors increase autism spectrum disorder (ASD) incidence, and this has led to the generation of corresponding animal models, with some showing strong construct and face validity. This short review focuses on results we have recently obtained with environmental and genetic mouse models of ASD and that are the valproic acid, the poly I:C and the Shank 3 models. This has allowed us to provide a comparative description of these widely used animal models providing an interesting perspective as to the pros and cons of each one of them, in our experimental settings. In these papers, we focused on motor and gait disorders which are currently not included in the diagnosis criteria, but which may provide new insights to ASD pathophysiology potentially leading to innovative therapies for a disease that currently has none. In all these models, we reported behavioral, cellular and molecular alterations related to the cerebellum. Motor and gait deficits were observed to various degrees in animal models and, when strongly present, they were correlated to the severity of social deficits as well as to the number of cerebellar Purkinje cells. Additionally, we also reported that, like in humans, males are more severely affected than females in these ASD models. These findings, along with an increasing body of literature, open new hopes in the ASD field pointing to brain regions, such the cerebellum, that are at the crossroads between cognitive, social and motor deficits. Targeting these brain regions and their underlying pathways and synaptic connections may prove of significant benefits.
Collapse
Affiliation(s)
- Mohamed Jaber
- Université de Poitiers, Inserm, Laboratoire de Neurosciences Expérimentales et Cliniques, Bâtiment B36, 1 Rue Georges Bonnet, BP 633, TSA 51106, 86073, Poitiers cedex9, France.
- Centre Hospitalier Universitaire de Poitiers, Poitiers, France.
| |
Collapse
|
128
|
Tu G, Guo Y, Xiao R, Tang L, Hu M, Liao B. Effects of Exercise Training on the Phosphoproteomics of the Medial Prefrontal Cortex in Rats With Autism Spectrum Disorder Induced by Valproic Acid. Neurorehabil Neural Repair 2023; 37:94-108. [PMID: 36860155 DOI: 10.1177/15459683231152814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/03/2023]
Abstract
BACKGROUND The key neural pathological characteristics of autism spectrum disorder (ASD) include abnormal synaptic plasticity of the medial prefrontal cortex (mPFC). Exercise therapy is widely used to rehabilitate children with ASD, but its neurobiological mechanism is unclear. METHODS To clarify whether the structural and molecular plasticity of synapses in the mPFC are related to improvement in ASD behavioral deficits after continuous exercise rehabilitation training, we applied phosphoproteomic, behavioral, morphological, and molecular biological methods to investigate the impact of exercise on the phosphoprotein expression profile and synaptic structure of the mPFC in valproic acid (VPA)-induced ASD rats. RESULTS Exercise training differentially regulated the density, morphology, and ultrastructure of synapses in mPFC subregions in the VPA-induced ASD rats. In total, 1031 phosphopeptides were upregulated and 782 phosphopeptides were downregulated in the mPFC in the ASD group. After exercise training, 323 phosphopeptides were upregulated, and 1098 phosphopeptides were downregulated in the ASDE group. Interestingly, 101 upregulated and 33 downregulated phosphoproteins in the ASD group were reversed after exercise training, and these phosphoproteins were mostly involved in synapses. Consistent with the phosphoproteomics data, the total and phosphorylated levels of the proteins MARK1 and MYH10 were upregulated in the ASD group and reversed after exercise training. CONCLUSIONS The differential structural plasticity of synapses in mPFC subregions may be the basic neural architecture of ASD behavioral abnormalities. The phosphoproteins involved in mPFC synapses, such as MARK1 and MYH10, may play important roles in the exercise rehabilitation effect on ASD-induced behavioral deficits and synaptic structural plasticity, which requires further investigation.
Collapse
Affiliation(s)
- Genghong Tu
- Department of Sports Medicine, Guangzhou Sport University, Guangzhou, Guangdong, P.R. China.,Guangdong Provincial Key Laboratory of Physical Activity and Health Promotion, Scientific Research Center, Guangzhou Sport University, Guangzhou, Guangdong, P.R. China
| | - Youli Guo
- Department of Pharmacy, Guangdong Provincial Corps Hospital of Chinese People's Armed Police Forces, Guangzhou, Guangdong, P.R. China
| | - Ruoshi Xiao
- Department of Sports Medicine, Guangzhou Sport University, Guangzhou, Guangdong, P.R. China.,Guangdong Provincial Key Laboratory of Physical Activity and Health Promotion, Scientific Research Center, Guangzhou Sport University, Guangzhou, Guangdong, P.R. China
| | - Lianying Tang
- Department of Sports Medicine, Guangzhou Sport University, Guangzhou, Guangdong, P.R. China.,Guangdong Provincial Key Laboratory of Physical Activity and Health Promotion, Scientific Research Center, Guangzhou Sport University, Guangzhou, Guangdong, P.R. China
| | - Min Hu
- Guangdong Provincial Key Laboratory of Physical Activity and Health Promotion, Scientific Research Center, Guangzhou Sport University, Guangzhou, Guangdong, P.R. China
| | - Bagen Liao
- Department of Sports Medicine, Guangzhou Sport University, Guangzhou, Guangdong, P.R. China.,Guangdong Provincial Key Laboratory of Physical Activity and Health Promotion, Scientific Research Center, Guangzhou Sport University, Guangzhou, Guangdong, P.R. China
| |
Collapse
|
129
|
Li K, Liang X, Xie X, Tian L, Yan J, Lin B, Liu H, Lai W, Liu X, Xi Z. Role of SHANK3 in concentrated ambient PM2. 5 exposure induced autism-like phenotype. Heliyon 2023; 9:e14328. [PMID: 36938421 PMCID: PMC10018567 DOI: 10.1016/j.heliyon.2023.e14328] [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: 05/09/2022] [Revised: 02/23/2023] [Accepted: 03/01/2023] [Indexed: 03/08/2023] Open
Abstract
Perinatal air pollution plays an important role in the development of autism. However, research on the pathogenic mechanism remains limited. In this study, the model of systemic inhalation of concentrated approximately 8-fold the level (mean concentration was 224 μg/m3) reported in ambient outdoor air of PM2.5 (particulate matters that are 2.5 μm or less in diameter)in early-postnatal male Sprague-Dawley (SD) rats was established. Through a series of autism-related behavioral tests, it was identified that young rats (postnatal day 1-day21, named PND1-PND21) exposed to PM2.5 exhibited typical autistic phenotypes, such as impaired language communication, abnormal repetitive and stereotyped behaviors, and impaired social skills. Moreover, synaptic abnormalities have been found in the brain tissues of young rats exposed to PM2.5. In terms of the molecular mechanism, we found that the levels of SH3 and multiple ankyrin repeat domains 3 (SHANK3) expression and key molecular proteins in the downstream signaling pathways were decreased in the brain tissues of the exposed rats. Finally, at the epigenetic level, SHANK3 methylation levels were increased in young rats exposed to PM2.5. In conclusion, the study revealed that PM2.5 exposure might induce the early postnatal autism through the SHANK3 signaling pathway by affecting the SHANK3 methylation levels and reducing the SHANK3 expression levels. The study could provide new ideas for autism etiology and a theoretical basis for the prevention and treatment of autism in children.
Collapse
Affiliation(s)
- Kang Li
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, 300050, China
| | - Xiaotian Liang
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, 300050, China
- Binzhou Medical College, Yantai, 264000, China
| | - Xiaoqian Xie
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, 300050, China
- Binzhou Medical College, Yantai, 264000, China
| | - Lei Tian
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, 300050, China
| | - Jun Yan
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, 300050, China
| | - Bencheng Lin
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, 300050, China
| | - Huanliang Liu
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, 300050, China
| | - Wenqin Lai
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, 300050, China
| | - Xiaohua Liu
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, 300050, China
- Corresponding author.
| | - Zhuge Xi
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, 300050, China
- Binzhou Medical College, Yantai, 264000, China
- Corresponding author. Tianjin Institute of Environmental and Operational Medicine, No. 1, Dali Road, Heping District, Tianjin, 300050, PR China.
| |
Collapse
|
130
|
Evaluation of Individuals with Non-Syndromic Global Developmental Delay and Intellectual Disability. CHILDREN 2023; 10:children10030414. [PMID: 36979972 PMCID: PMC10047567 DOI: 10.3390/children10030414] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 02/11/2023] [Accepted: 02/16/2023] [Indexed: 02/24/2023]
Abstract
Global Developmental Delay (GDD) and Intellectual Disability (ID) are two of the most common presentations encountered by physicians taking care of children. GDD/ID is classified into non-syndromic GDD/ID, where GDD/ID is the sole evident clinical feature, or syndromic GDD/ID, where there are additional clinical features or co-morbidities present. Careful evaluation of children with GDD and ID, starting with detailed history followed by a thorough examination, remain the cornerstone for etiologic diagnosis. However, when initial history and examination fail to identify a probable underlying etiology, further genetic testing is warranted. In recent years, genetic testing has been shown to be the single most important diagnostic modality for clinicians evaluating children with non-syndromic GDD/ID. In this review, we discuss different genetic testing currently available, review common underlying copy-number variants and molecular pathways, explore the recent evidence and recommendations for genetic evaluation and discuss an approach to the diagnosis and management of children with non-syndromic GDD and ID.
Collapse
|
131
|
Moffitt BA, Sarasua SM, Ivankovic D, Ward LD, Valentine K, Bennett WE, Rogers C, Phelan K, Boccuto L. Stratification of a Phelan-McDermid Syndrome Population Based on Their Response to Human Growth Hormone and Insulin-like Growth Factor. Genes (Basel) 2023; 14:490. [PMID: 36833418 PMCID: PMC9956088 DOI: 10.3390/genes14020490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 02/11/2023] [Accepted: 02/13/2023] [Indexed: 02/17/2023] Open
Abstract
Phelan-McDermid syndrome (PMS), caused by pathogenic variants in the SHANK3 gene or 22q13 deletions, is characterized by intellectual disability, autistic features, developmental delays, and neonatal hypotonia. Insulin-like growth factor 1 (IGF-1) and human growth hormone (hGH) have been shown to reverse neurobehavioral deficits in PMS. We assessed the metabolic profiling of 48 individuals with PMS and 50 controls and determined subpopulations by taking the top and bottom 25% of responders to hGH and IGF-1. A distinct metabolic profile for individuals with PMS showed a reduced ability to metabolize major energy sources and a higher metabolism of alternative energy sources. The analysis of the metabolic response to hGH or IGF-1 highlighted a major overlap between both high and low responders, validating the model and suggesting that the two growth factors share many target pathways. When we investigated the effect of hGH and IGF-1 on the metabolism of glucose, the correlation between the high-responder subgroups showed less similarity, whereas the low-responders were still relatively similar. Classification of individuals with PMS into subgroups based on responses to a compound can allow an investigation into pathogenic mechanisms, the identification of molecular biomarkers, an exploration of in vitro responses to candidate drugs, and eventually the selection of better candidates for clinical trials.
Collapse
Affiliation(s)
- Bridgette A. Moffitt
- School of Nursing, Healthcare Genetics Program, Clemson University, Clemson, SC 29634, USA
| | - Sara M. Sarasua
- School of Nursing, Healthcare Genetics Program, Clemson University, Clemson, SC 29634, USA
| | - Diana Ivankovic
- School of Nursing, Healthcare Genetics Program, Clemson University, Clemson, SC 29634, USA
| | - Linda D. Ward
- School of Nursing, Healthcare Genetics Program, Clemson University, Clemson, SC 29634, USA
| | - Kathleen Valentine
- School of Nursing, Healthcare Genetics Program, Clemson University, Clemson, SC 29634, USA
| | - William E. Bennett
- Division of Pediatric Gastroenterology, Hepatology, and Nutrition, Indiana University, Riley Hospital for Children, Indianapolis, IN 46202, USA
| | | | - Katy Phelan
- Genetics Laboratory, Florida Cancer Specialists & Research Institute, Fort Myers, FL 33916, USA
| | - Luigi Boccuto
- School of Nursing, Healthcare Genetics Program, Clemson University, Clemson, SC 29634, USA
| |
Collapse
|
132
|
Kamran M, Laighneach A, Bibi F, Donohoe G, Ahmed N, Rehman AU, Morris DW. Independent Associated SNPs at SORCS3 and Its Protein Interactors for Multiple Brain-Related Disorders and Traits. Genes (Basel) 2023; 14:482. [PMID: 36833409 PMCID: PMC9956385 DOI: 10.3390/genes14020482] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 01/26/2023] [Accepted: 02/06/2023] [Indexed: 02/16/2023] Open
Abstract
Sortilin-related vacuolar protein sorting 10 (VPS10) domain containing receptor 3 (SORCS3) is a neuron-specific transmembrane protein involved in the trafficking of proteins between intracellular vesicles and the plasma membrane. Genetic variation at SORCS3 is associated with multiple neuropsychiatric disorders and behavioural phenotypes. Here, we undertake a systematic search of published genome-wide association studies to identify and catalogue associations between SORCS3 and brain-related disorders and traits. We also generate a SORCS3 gene-set based on protein-protein interactions and investigate the contribution of this gene-set to the heritability of these phenotypes and its overlap with synaptic biology. Analysis of association signals at SORSC3 showed individual SNPs to be associated with multiple neuropsychiatric and neurodevelopmental brain-related disorders and traits that have an impact on the experience of feeling, emotion or mood or cognitive function, while multiple LD-independent SNPs were associated with the same phenotypes. Across these SNPs, alleles associated with the more favourable outcomes for each phenotype (e.g., decreased risk of neuropsychiatric illness) were associated with increased expression of the SORCS3 gene. The SORCS3 gene-set was enriched for heritability contributing to schizophrenia (SCZ), bipolar disorder (BPD), intelligence (IQ) and education attainment (EA). Eleven genes from the SORCS3 gene-set were associated with more than one of these phenotypes at the genome-wide level, with RBFOX1 associated with SCZ, IQ and EA. Functional annotation revealed that the SORCS3 gene-set is enriched for multiple ontologies related to the structure and function of synapses. Overall, we find many independent association signals at SORCS3 with brain-related disorders and traits, with the effect possibly mediated by reduced gene expression, resulting in a negative impact on synaptic function.
Collapse
Affiliation(s)
- Muhammad Kamran
- Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad 45320, Pakistan
- Centre for Neuroimaging, Cognition and Genomics (NICOG), School of Biological and Chemical Sciences and School of Psychology, University of Galway, H91 CF50 Galway, Ireland
| | - Aodán Laighneach
- Centre for Neuroimaging, Cognition and Genomics (NICOG), School of Biological and Chemical Sciences and School of Psychology, University of Galway, H91 CF50 Galway, Ireland
| | - Farhana Bibi
- Department of Biosciences, Grand Asian University, Sialkot 51040, Pakistan
| | - Gary Donohoe
- Centre for Neuroimaging, Cognition and Genomics (NICOG), School of Biological and Chemical Sciences and School of Psychology, University of Galway, H91 CF50 Galway, Ireland
| | - Naveed Ahmed
- Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad 45320, Pakistan
| | - Asim Ur Rehman
- Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad 45320, Pakistan
| | - Derek W. Morris
- Centre for Neuroimaging, Cognition and Genomics (NICOG), School of Biological and Chemical Sciences and School of Psychology, University of Galway, H91 CF50 Galway, Ireland
| |
Collapse
|
133
|
Liu P, Zhao Y, Xiong W, Pan Y, Zhu M, Zhu X. Degradation of Perineuronal Nets in the Cerebellar Interpositus Nucleus Ameliorated Social Deficits in Shank3-deficient Mice. Neuroscience 2023; 511:29-38. [PMID: 36587867 DOI: 10.1016/j.neuroscience.2022.12.024] [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: 07/22/2022] [Revised: 12/08/2022] [Accepted: 12/26/2022] [Indexed: 12/30/2022]
Abstract
Perineuronal nets (PNNs) are structures that contain extracellular matrix chondroitin sulfate proteoglycan and surround the soma and dendrites of various neuronal cell types. They are involved in synaptic plasticity and undertake important physiological functions. Altered expression of PNNs has been demonstrated in the brains of autism-related animal models. However, the underlying mechanism is still unknown. In this study, we demonstrated that the PNNs in the cerebellum are involved in modulating social and repetitive/inflexible behaviors in Shank3B-/- mice, an established animal model of autism spectrum disorder. First, we performed wisteria floribunda agglutinin staining of the whole brain of Shank3B-/- mice, and found wisteria floribunda agglutinin-positive PNNs are significantly increased in the cerebellar interpositus nucleus (IntP) in Shank3B-/- mice compared to control littermates. After degradation of PNNs in the IntP by chondroitinase ABC, the repetitive behaviors of Shank3B-/- mice were decreased, while their social behaviors were ameliorated. These results suggested that PNNs homeostasis is involved in the regulation of social behavior, revealing a potential therapeutic strategy targeting PNNs in the IntP for the treatment of autism spectrum disorder.
Collapse
Affiliation(s)
- Peng Liu
- School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Yulu Zhao
- School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Wenchao Xiong
- School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China.
| | - Yida Pan
- School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Minzhen Zhu
- School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Xinhong Zhu
- School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China; School of Psychology, Shenzhen University, Shenzhen 518060, China; Research Center for Brain Health, Pazhou Lab, Guangzhou 510330, China.
| |
Collapse
|
134
|
Huang M, Qi Q, Xu T. Targeting Shank3 deficiency and paresthesia in autism spectrum disorder: A brief review. Front Mol Neurosci 2023; 16:1128974. [PMID: 36846568 PMCID: PMC9948097 DOI: 10.3389/fnmol.2023.1128974] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 01/18/2023] [Indexed: 02/11/2023] Open
Abstract
Autism spectrum disorder (ASD) includes a group of multifactorial neurodevelopmental disorders characterized by impaired social communication, social interaction, and repetitive behaviors. Several studies have shown an association between cases of ASD and mutations in the genes of SH3 and multiple ankyrin repeat domain protein 3 (SHANK3). These genes encode many cell adhesion molecules, scaffold proteins, and proteins involved in synaptic transcription, protein synthesis, and degradation. They have a profound impact on all aspects of synaptic transmission and plasticity, including synapse formation and degeneration, suggesting that the pathogenesis of ASD may be partially attributable to synaptic dysfunction. In this review, we summarize the mechanism of synapses related to Shank3 in ASD. We also discuss the molecular, cellular, and functional studies of experimental models of ASD and current autism treatment methods targeting related proteins.
Collapse
Affiliation(s)
- Min Huang
- Department of Anesthesiology, Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China,Department of Anesthesiology, Suzhou Hospital of Anhui Medical University, Suzhou, China
| | - Qi Qi
- Department of Anesthesiology, Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China,Department of Anesthesiology, Suzhou Hospital of Anhui Medical University, Suzhou, China
| | - Tao Xu
- Department of Anesthesiology, Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China,Department of Anesthesiology, Suzhou Hospital of Anhui Medical University, Suzhou, China,*Correspondence: Tao Xu,
| |
Collapse
|
135
|
Transition from Animal-Based to Human Induced Pluripotent Stem Cells (iPSCs)-Based Models of Neurodevelopmental Disorders: Opportunities and Challenges. Cells 2023; 12:cells12040538. [PMID: 36831205 PMCID: PMC9954744 DOI: 10.3390/cells12040538] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 01/25/2023] [Accepted: 02/02/2023] [Indexed: 02/11/2023] Open
Abstract
Neurodevelopmental disorders (NDDs) arise from the disruption of highly coordinated mechanisms underlying brain development, which results in impaired sensory, motor and/or cognitive functions. Although rodent models have offered very relevant insights to the field, the translation of findings to clinics, particularly regarding therapeutic approaches for these diseases, remains challenging. Part of the explanation for this failure may be the genetic differences-some targets not being conserved between species-and, most importantly, the differences in regulation of gene expression. This prompts the use of human-derived models to study NDDS. The generation of human induced pluripotent stem cells (hIPSCs) added a new suitable alternative to overcome species limitations, allowing for the study of human neuronal development while maintaining the genetic background of the donor patient. Several hIPSC models of NDDs already proved their worth by mimicking several pathological phenotypes found in humans. In this review, we highlight the utility of hIPSCs to pave new paths for NDD research and development of new therapeutic tools, summarize the challenges and advances of hIPSC-culture and neuronal differentiation protocols and discuss the best way to take advantage of these models, illustrating this with examples of success for some NDDs.
Collapse
|
136
|
Transient hearing abnormalities precede social deficits in a mouse model of autism. Behav Brain Res 2023; 437:114149. [PMID: 36206820 DOI: 10.1016/j.bbr.2022.114149] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Revised: 09/30/2022] [Accepted: 10/03/2022] [Indexed: 11/13/2022]
Abstract
Hearing abnormalities are important symptoms of autism spectrum disorders (ASDs), a neurological and developmental disorder. However, the characteristics of hearing abnormalities associated with ASD during development have not been fully investigated. We found that in Shank3B knockout mice (a high-confidence mouse model of ASD), transient hearing abnormalities can be found in auditory brainstem response, auditory cortical activity, as well as acoustic startle response. More importantly, all hearing abnormalities at 4 weeks were most prominent and preceded the onset of social deficits at 6 weeks. These hearing abnormalities gradually recovered with age. In addition, analysis of ABR data at 4 weeks using Support Vector Machine (SVM) can faithfully predict the genotype of mice with an accuracy of 85.71%. These findings not only revealed hearing changes in Shank3B knockout autistic-like mice during development, but also suggested that hearing abnormalities could potentially be used as an early and effective indicator of ASD risk.
Collapse
|
137
|
Wang Y, Liu C, Deng J, Xu Q, Lin J, Li H, Hu M, Hu C, Li Q, Xu X. Behavioral and Sensory Deficits Associated with Dysfunction of GABAergic System in a Novel shank2-Deficient Zebrafish Model. Int J Mol Sci 2023; 24:2208. [PMID: 36768529 PMCID: PMC9916955 DOI: 10.3390/ijms24032208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 01/17/2023] [Accepted: 01/18/2023] [Indexed: 01/24/2023] Open
Abstract
Hyper-reactivity to sensory inputs is a common and debilitating symptom of autism spectrum disorder (ASD), but the underlying neural abnormalities remain unclear. Two of three patients in our clinical cohort screen harboring de novo SHANK2 mutations also exhibited high sensitivity to visual, auditory, and tactile stimuli, so we examined whether shank2 deficiencies contribute to sensory abnormalities and other ASD-like phenotypes by generating a stable shank2b-deficient zebrafish model (shank2b-/-). The adult shank2b-/- zebrafish demonstrated reduced social preference and kin preference as well as enhanced behavioral stereotypy, while larvae exhibited hyper-sensitivity to auditory noise and abnormal hyperactivity during dark-to-light transitions. This model thus recapitulated the core developmental and behavioral phenotypes of many previous genetic ASD models. Expression levels of γ-aminobutyric acid (GABA) receptor subunit mRNAs and proteins were also reduced in shank2b-/- zebrafish, and these animals exhibited greater sensitivity to drug-induced seizures. Our results suggest that GABAergic dysfunction is a major contributor to the sensory hyper-reactivity in ASD, and they underscore the need for interventions that target sensory-processing disruptions during early neural development to prevent disease progression.
Collapse
Affiliation(s)
- Yi Wang
- Division of Child Health Care, Children’s Hospital of Fudan University, National Children’s Medical Center, 399 Wanyuan Road, Shanghai 201102, China
| | - Chunxue Liu
- Division of Child Health Care, Children’s Hospital of Fudan University, National Children’s Medical Center, 399 Wanyuan Road, Shanghai 201102, China
| | - Jingxin Deng
- Division of Child Health Care, Children’s Hospital of Fudan University, National Children’s Medical Center, 399 Wanyuan Road, Shanghai 201102, China
| | - Qiong Xu
- Division of Child Health Care, Children’s Hospital of Fudan University, National Children’s Medical Center, 399 Wanyuan Road, Shanghai 201102, China
| | - Jia Lin
- Center for Translational Medicine, Institute of Pediatrics, Shanghai Key Laboratory of Birth Defect, Children’s Hopstial of Fudan University, National Children’s Medical Center, 399 Wangyuan Road, Shanghai 201102, China
| | - Huiping Li
- Division of Child Health Care, Children’s Hospital of Fudan University, National Children’s Medical Center, 399 Wanyuan Road, Shanghai 201102, China
| | - Meixin Hu
- Division of Child Health Care, Children’s Hospital of Fudan University, National Children’s Medical Center, 399 Wanyuan Road, Shanghai 201102, China
| | - Chunchun Hu
- Division of Child Health Care, Children’s Hospital of Fudan University, National Children’s Medical Center, 399 Wanyuan Road, Shanghai 201102, China
| | - Qiang Li
- Center for Translational Medicine, Institute of Pediatrics, Shanghai Key Laboratory of Birth Defect, Children’s Hopstial of Fudan University, National Children’s Medical Center, 399 Wangyuan Road, Shanghai 201102, China
| | - Xiu Xu
- Division of Child Health Care, Children’s Hospital of Fudan University, National Children’s Medical Center, 399 Wanyuan Road, Shanghai 201102, China
| |
Collapse
|
138
|
Agostini S, Bolognesi E, Mancuso R, Marventano I, Citterio LA, Guerini FR, Clerici M. miR-23a-3p and miR-181a-5p modulate SNAP-25 expression. PLoS One 2023; 18:e0279961. [PMID: 36649268 PMCID: PMC9844927 DOI: 10.1371/journal.pone.0279961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 12/14/2022] [Indexed: 01/18/2023] Open
Abstract
SNAP-25 protein is a key protein of the SNARE complex that is involved in synaptic vesicles fusion with plasma membranes and neurotransmitter release, playing a fundamental role in neural plasticity. Recently the concentration of three specific miRNAs-miR-27b-3p, miR-181a-5p and miR-23a-3p -was found to be associated with a specific SNAP-25 polymorphism (rs363050). in silico analysis showed that all the three miRNAs target SNAP-25, but the effect of the interaction between these miRNAs and the 3'UTR of SNAP-25 mRNA is currently unknown. For this reason, we verified in vitro whether miR-27b-3p, miR-181a-5p and miR-23a-3p modulate SNAP-25 gene and protein expression. Initial experiments using miRNAs-co-transfected Vero cells and SNAP-25 3'UTR luciferase reporter plasmids showed that miR-181a-5p (p≤0.01) and miR-23a-3p (p<0.05), but not miR-27b-3p, modulate the luciferase signal, indicating that these two miRNAs bind the SNAP-25 3'UTR. Results obtained using human oligodendroglial cell line (MO3.13) transfected with miR-181a-5p or miR-27b-3p confirmed that miR-181a-5p and miR-23a-3p regulate SNAP-25 gene and protein expression. Interestingly, the two miRNAs modulate in an opposite way SNAP-25, as miR-181a-5p significantly increases (p<0.0005), whereas miR-23a-3p decreases (p<0.0005) its expression. These results for the first time describe the ability of miR-181a-5p and miR-23a-3p to modulate SNAP-25 expression, suggesting their possible use as biomarkers or as therapeutical targets for diseases in which SNAP-25 expression is altered.
Collapse
Affiliation(s)
| | | | - Roberta Mancuso
- IRCCS Fondazione Don Carlo Gnocchi ONLUS, Milan, Italy
- * E-mail:
| | | | | | | | - Mario Clerici
- IRCCS Fondazione Don Carlo Gnocchi ONLUS, Milan, Italy
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| |
Collapse
|
139
|
Kim Y, Ko TH, Jin C, Zhang Y, Kang HR, Ma R, Li H, Choi JI, Han K. The emerging roles of Shank3 in cardiac function and dysfunction. Front Cell Dev Biol 2023; 11:1191369. [PMID: 37187620 PMCID: PMC10175600 DOI: 10.3389/fcell.2023.1191369] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 04/17/2023] [Indexed: 05/17/2023] Open
Abstract
Shank3 is a member of the Shank family proteins (Shank1-3), which are abundantly present in the postsynaptic density (PSD) of neuronal excitatory synapses. As a core scaffold in the PSD, Shank3 plays a critical role in organizing the macromolecular complex, ensuring proper synaptic development and function. Clinically, various mutations of the SHANK3 gene are causally associated with brain disorders such as autism spectrum disorders and schizophrenia. However, recent in vitro and in vivo functional studies and expression profiling in various tissues and cell types suggest that Shank3 also plays a role in cardiac function and dysfunction. For example, Shank3 interacts with phospholipase Cβ1b (PLCβ1b) in cardiomyocytes, regulating its localization to the sarcolemma and its role in mediating Gq-induced signaling. In addition, changes in cardiac morphology and function associated with myocardial infarction and aging have been investigated in a few Shank3 mutant mouse models. This review highlights these results and potential underlying mechanisms, and predicts additional molecular functions of Shank3 based on its protein interactors in the PSD, which are also highly expressed and function in the heart. Finally, we provide perspectives and possible directions for future studies to better understand the roles of Shank3 in the heart.
Collapse
Affiliation(s)
- Yoonhee Kim
- Department of Neuroscience, Korea University College of Medicine, Seoul, Republic of Korea
| | - Tae Hee Ko
- Division of Cardiology, Department of Internal Medicine, Korea University College of Medicine and Korea University Anam Hospital, Seoul, Republic of Korea
| | - Chunmei Jin
- Department of Neuroscience, Korea University College of Medicine, Seoul, Republic of Korea
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science, Daejeon, Republic of Korea
| | - Yinhua Zhang
- Department of Neuroscience, Korea University College of Medicine, Seoul, Republic of Korea
| | - Hyae Rim Kang
- Department of Neuroscience, Korea University College of Medicine, Seoul, Republic of Korea
- BK21 Graduate Program, Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Republic of Korea
| | - Ruiying Ma
- Department of Neuroscience, Korea University College of Medicine, Seoul, Republic of Korea
- BK21 Graduate Program, Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Republic of Korea
| | - Huiling Li
- Department of Neuroscience, Korea University College of Medicine, Seoul, Republic of Korea
- BK21 Graduate Program, Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Republic of Korea
| | - Jong-Il Choi
- Division of Cardiology, Department of Internal Medicine, Korea University College of Medicine and Korea University Anam Hospital, Seoul, Republic of Korea
- BK21 Graduate Program, Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Republic of Korea
- *Correspondence: Jong-Il Choi, ; Kihoon Han,
| | - Kihoon Han
- Department of Neuroscience, Korea University College of Medicine, Seoul, Republic of Korea
- BK21 Graduate Program, Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Republic of Korea
- *Correspondence: Jong-Il Choi, ; Kihoon Han,
| |
Collapse
|
140
|
Steinert JR, Amal H. The contribution of an imbalanced redox signalling to neurological and neurodegenerative conditions. Free Radic Biol Med 2023; 194:71-83. [PMID: 36435368 DOI: 10.1016/j.freeradbiomed.2022.11.035] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 11/17/2022] [Accepted: 11/22/2022] [Indexed: 11/25/2022]
Abstract
Nitric oxide and other redox active molecules such as oxygen free radicals provide essential signalling in diverse neuronal functions, but their excess production and insufficient scavenging induces cytotoxic redox stress which is associated with numerous neurodegenerative and neurological conditions. A further component of redox signalling is mediated by a homeostatic regulation of divalent metal ions, the imbalance of which contributes to neuronal dysfunction. Additional antioxidant molecules such as glutathione and enzymes such as super oxide dismutase are involved in maintaining a physiological redox status within neurons. When cellular processes are perturbed and generation of free radicals overwhelms the antioxidants capacity of the neurons, a resulting redox damage leads to neuronal dysfunction and cell death. Cellular sources for production of redox-active molecules may include NADPH oxidases, mitochondria, cytochrome P450 and nitric oxide (NO)-generating enzymes, such as endothelial, neuronal and inducible NO synthases. Several neurodegenerative and developmental neurological conditions are associated with an imbalanced redox state as a result of neuroinflammatory processes leading to nitrosative and oxidative stress. Ongoing research aims at understanding the causes and consequences of such imbalanced redox homeostasis and its role in neuronal dysfunction.
Collapse
Affiliation(s)
- Joern R Steinert
- Division of Physiology, Pharmacology and Neuroscience, University of Nottingham, School of Life Sciences, Nottingham, NG7 2NR, UK.
| | - Haitham Amal
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel.
| |
Collapse
|
141
|
Ahmad M, Stirmlinger N, Jan I, Stifel U, Lee S, Weingandt M, Kelp U, Bockmann J, Ignatius A, Böckers TM, Tuckermann J. Downregulation of the Autism Spectrum Disorder Gene Shank2 Decreases Bone Mass in Male Mice. JBMR Plus 2022; 7:e10711. [PMID: 36751416 PMCID: PMC9893268 DOI: 10.1002/jbm4.10711] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 11/25/2022] [Accepted: 11/30/2022] [Indexed: 12/05/2022] Open
Abstract
Mutations of the postsynaptic scaffold protein Shank2 lead to autism spectrum disorders (ASD). These patients frequently suffer from higher fracture risk. Here, we investigated whether Shank2 directly regulates bone mass. We show that Shank2 is expressed in bone and that Shank2 levels are increased during osteoblastogenesis. Knockdown of Shank2 by siRNA targeting the encoding regions for PDZ and SAM domain inhibits osteoblastogenesis of primary murine calvarial osteoblasts. Shank2 knockout mice (Shank2 -/-) have a decreased bone mass due to reduced osteoblastogenesis and bone formation, whereas bone resorption remains unaffected. Induced pluripotent stem cells (iPSCs)-derived osteoblasts from a loss-of-function Shank2 mutation in a patient showed a significantly reduced osteoblast differentiation potential. Moreover, silencing of known Shank2 interacting proteins revealed that a majority of them promote osteoblast differentiation. From this we conclude that Shank2 and interacting proteins known from the central nervous system are decisive regulators in osteoblast differentiation. © 2022 The Authors. JBMR Plus published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Mubashir Ahmad
- Institute of Comparative Molecular Endocrinology (CME)Ulm UniversityUlmGermany
| | | | - Irfana Jan
- Institute of Comparative Molecular Endocrinology (CME)Ulm UniversityUlmGermany
| | - Ulrich Stifel
- Institute of Comparative Molecular Endocrinology (CME)Ulm UniversityUlmGermany
| | - Sooyeon Lee
- Institute of Comparative Molecular Endocrinology (CME)Ulm UniversityUlmGermany
| | - Marcel Weingandt
- Institute of Comparative Molecular Endocrinology (CME)Ulm UniversityUlmGermany
| | - Ulrike Kelp
- Institute of Comparative Molecular Endocrinology (CME)Ulm UniversityUlmGermany
| | - Jürgen Bockmann
- Institute for Anatomy and Cell BiologyUlm UniversityUlmGermany
| | - Anita Ignatius
- Institute of Orthopaedic Research and BiomechanicsUlm UniversityUlmGermany
| | | | - Jan Tuckermann
- Institute of Comparative Molecular Endocrinology (CME)Ulm UniversityUlmGermany
| |
Collapse
|
142
|
Bouquier N, Sakkaki S, Raynaud F, Hemonnot-Girard AL, Seube V, Compan V, Bertaso F, Perroy J, Moutin E. The Shank3 Venus/Venus knock in mouse enables isoform-specific functional studies of Shank3a. Front Neurosci 2022; 16:1081010. [PMID: 36570823 PMCID: PMC9773256 DOI: 10.3389/fnins.2022.1081010] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 11/23/2022] [Indexed: 12/14/2022] Open
Abstract
Background Shank3 is a scaffolding protein essential for the organization and function of the glutamatergic postsynapse. Monogenic mutations in SHANK3 gene are among the leading genetic causes of Autism Spectrum Disorders (ASD). The multiplicity of Shank3 isoforms seems to generate as much functional diversity and yet, there are no tools to study endogenous Shank3 proteins in an isoform-specific manner. Methods In this study, we created a novel transgenic mouse line, the Shank3Venus/Venus knock in mouse, which allows to monitor the endogenous expression of the major Shank3 isoform in the brain, the full-length Shank3a isoform. Results We show that the endogenous Venus-Shank3a protein is localized in spines and is mainly expressed in the striatum, hippocampus and cortex of the developing and adult brain. We show that Shank3Venus/+ and Shank3Venus/Venus mice have no behavioral deficiency. We further crossed Shank3Venus/Venus mice with Shank3ΔC/ΔC mice, a model of ASD, to track the Venus-tagged wild-type copy of Shank3a in physiological (Shank3Venus/+) and pathological (Shank3Venus/ΔC) conditions. We report a developmental delay in brain expression of the Venus-Shank3a isoform in Shank3Venus/ΔC mice, compared to Shank3Venus/+ control mice. Conclusion Altogether, our results show that the Shank3Venus/Venus mouse line is a powerful tool to study endogenous Shank3a expression, in physiological conditions and in ASD.
Collapse
Affiliation(s)
- Nathalie Bouquier
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Sophie Sakkaki
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Fabrice Raynaud
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, Montpellier, France,PhyMedExp, Univ Montpellier, INSERM, CNRS, CHU de Montpellier, Montpellier, France
| | | | - Vincent Seube
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Vincent Compan
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Federica Bertaso
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Julie Perroy
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, Montpellier, France,*Correspondence: Julie Perroy,
| | - Enora Moutin
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, Montpellier, France,Enora Moutin,
| |
Collapse
|
143
|
Abstract
Interest in gene-based therapies for neurodevelopmental disorders is increasing exponentially, driven by the rise in recognition of underlying genetic etiology, progress in genomic technology, and recent proof of concept in several disorders. The current prioritization of one genetic disorder over another for development of therapies is driven by competing interests of pharmaceutical companies, advocacy groups, and academic scientists. Although these are all valid perspectives, a consolidated framework will facilitate more efficient and rational gene therapy development. Here we outline features of Mendelian neurodevelopmental disorders that warrant consideration when determining suitability for gene therapy. These features fit into four broad domains: genetics, preclinical validation, clinical considerations, and ethics. We propose a simple mnemonic, GENE TARGET, to remember these features and illustrate how they could be scored using a preliminary scoring rubric. In this suggested rubric, for a given disorder, scores for each feature may be added up to a composite GENE TARGET suitability (GTS) score. In addition to proposing a systematic method to evaluate and compare disorders, our framework helps identify gaps in the translational pipeline for a given disorder, which can inform prioritization of future research efforts.
Collapse
|
144
|
Xiao L, Jiang S, Wang Y, Gao C, Liu C, Huo X, Li W, Guo B, Wang C, Sun Y, Wang A, Feng Y, Wang F, Sun T. Continuous high-frequency deep brain stimulation of the anterior insula modulates autism-like behavior in a valproic acid-induced rat model. J Transl Med 2022; 20:570. [PMID: 36474209 PMCID: PMC9724311 DOI: 10.1186/s12967-022-03787-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 11/23/2022] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Until now, the treatment of patients with autism spectrum disorder (ASD) remain a difficult problem. The insula is involved in empathy and sensorimotor integration, which are often impaired in individuals with ASD. Deep brain stimulation, modulating neuronal activity in specific brain circuits, has recently been considered as a promising intervention for neuropsychiatric disorders. Valproic acid (VPA) is a potential teratogenic agent, and prenatal exposure can cause autism-like symptoms including repetitive behaviors and defective sociability. Herein, we investigated the effects of continuous high-frequency deep brain stimulation in the anterior insula of rats exposed to VPA and explored cognitive functions, behavior, and molecular proteins connected to autism spectrum disorder. METHODS VPA-exposed offspring were bilaterally implanted with electrodes in the anterior insula (Day 0) with a recovery period of 1 week. (Day 0-7). High-frequency deep brain stimulation was applied from days 11 to 29. Three behavioral tests, including three-chamber social interaction test, were performed on days 7, 13, 18, 25 and 36, and several rats were used for analysis of immediate early genes and proteomic after deep brain stimulation intervention. Meanwhile, animals were subjected to a 20 day spatial learning and cognitive rigidity test using IntelliCage on day 11. RESULTS Deep brain stimulation improved the sociability and social novelty preference at day 18 prior to those at day 13, and the improvement has reached the upper limit compared to day 25. As for repetitive/stereotypic-like behavior, self- grooming time were reduced at day 18 and reached the upper limit, and the numbers of burried marbles were reduced at day 13 prior to those at day 18 and day 25. The improvements of sociability and social novelty preference were persistent after the stimulation had ceased. Spatial learning ability and cognitive rigidity were unaffected. We identified 35 proteins in the anterior insula, some of which were intimately linked to autism, and their expression levels were reversed upon administration of deep brain stimulation. CONCLUSIONS Autism-like behavior was ameliorated and autism-related proteins were reversed in the insula by deep brain stimulation intervention, these findings reveal that the insula may be a potential target for DBS in the treatment of autism, which provide a theoretical basis for its clinical application., although future studies are still warranted.
Collapse
Affiliation(s)
- Lifei Xiao
- grid.412194.b0000 0004 1761 9803Ningxia Key Laboratory of Cerebrocranial Disease, Incubation Base of National Key Laboratory, Ningxia Medical University, Yinchuan, 750000 China ,grid.413385.80000 0004 1799 1445Department of Neurosurgery, General Hospital of Ningxia Medical University, Yinchuan, 750000 China
| | - Shucai Jiang
- grid.416966.a0000 0004 1758 1470Department of Neurosurgery, Weifang People’s Hospital, Weifang, 261000 China
| | - Yangyang Wang
- grid.412194.b0000 0004 1761 9803Ningxia Key Laboratory of Cerebrocranial Disease, Incubation Base of National Key Laboratory, Ningxia Medical University, Yinchuan, 750000 China
| | - Caibin Gao
- grid.413385.80000 0004 1799 1445Department of Neurosurgery, General Hospital of Ningxia Medical University, Yinchuan, 750000 China
| | - Cuicui Liu
- grid.477991.5Department of Otolaryngology and Head Surgery, The First People’s Hospital of Yinchuan, Yinchuan, 750000 China
| | - Xianhao Huo
- grid.412194.b0000 0004 1761 9803Ningxia Key Laboratory of Cerebrocranial Disease, Incubation Base of National Key Laboratory, Ningxia Medical University, Yinchuan, 750000 China ,grid.413385.80000 0004 1799 1445Department of Neurosurgery, General Hospital of Ningxia Medical University, Yinchuan, 750000 China
| | - Wenchao Li
- grid.412194.b0000 0004 1761 9803Ningxia Key Laboratory of Cerebrocranial Disease, Incubation Base of National Key Laboratory, Ningxia Medical University, Yinchuan, 750000 China
| | - Baorui Guo
- grid.440288.20000 0004 1758 0451Department of Neurosurgery, Shaanxi Provincial People’s Hospital, Xi’an, 710000 China
| | - Chaofan Wang
- grid.412194.b0000 0004 1761 9803Ningxia Key Laboratory of Cerebrocranial Disease, Incubation Base of National Key Laboratory, Ningxia Medical University, Yinchuan, 750000 China
| | - Yu Sun
- grid.412194.b0000 0004 1761 9803Ningxia Key Laboratory of Cerebrocranial Disease, Incubation Base of National Key Laboratory, Ningxia Medical University, Yinchuan, 750000 China
| | - Anni Wang
- grid.412194.b0000 0004 1761 9803Ningxia Key Laboratory of Cerebrocranial Disease, Incubation Base of National Key Laboratory, Ningxia Medical University, Yinchuan, 750000 China
| | - Yan Feng
- grid.412194.b0000 0004 1761 9803Ningxia Key Laboratory of Cerebrocranial Disease, Incubation Base of National Key Laboratory, Ningxia Medical University, Yinchuan, 750000 China
| | - Feng Wang
- grid.13402.340000 0004 1759 700XDepartment of Neurosurgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310000 China
| | - Tao Sun
- grid.412194.b0000 0004 1761 9803Ningxia Key Laboratory of Cerebrocranial Disease, Incubation Base of National Key Laboratory, Ningxia Medical University, Yinchuan, 750000 China ,grid.413385.80000 0004 1799 1445Department of Neurosurgery, General Hospital of Ningxia Medical University, Yinchuan, 750000 China
| |
Collapse
|
145
|
Abstract
Recent advances in genomics have revealed a wide spectrum of genetic variants associated with neurodevelopmental disorders at an unprecedented scale. An increasing number of studies have consistently identified mutations-both inherited and de novo-impacting the function of specific brain circuits. This suggests that, during brain development, alterations in distinct neural circuits, cell types, or broad regulatory pathways ultimately shaping synapses might be a dysfunctional process underlying these disorders. Here, we review findings from human studies and animal model research to provide a comprehensive description of synaptic and circuit mechanisms implicated in neurodevelopmental disorders. We discuss how specific synaptic connections might be commonly disrupted in different disorders and the alterations in cognition and behaviors emerging from imbalances in neuronal circuits. Moreover, we review new approaches that have been shown to restore or mitigate dysfunctional processes during specific critical windows of brain development. Considering the heterogeneity of neurodevelopmental disorders, we also highlight the recent progress in developing improved clinical biomarkers and strategies that will help to identify novel therapeutic compounds and opportunities for early intervention.
Collapse
Affiliation(s)
- David Exposito-Alonso
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
- MRC Centre for Neurodevelopmental Disorders, King's College London, London, United Kingdom;
- Current affiliation: Division of Genetics and Genomics, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA;
| | - Beatriz Rico
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
- MRC Centre for Neurodevelopmental Disorders, King's College London, London, United Kingdom;
| |
Collapse
|
146
|
Bai Y, Wang H, Li C. SAPAP Scaffold Proteins: From Synaptic Function to Neuropsychiatric Disorders. Cells 2022; 11:cells11233815. [PMID: 36497075 PMCID: PMC9740047 DOI: 10.3390/cells11233815] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 11/24/2022] [Accepted: 11/25/2022] [Indexed: 11/30/2022] Open
Abstract
Excitatory (glutamatergic) synaptic transmission underlies many aspects of brain activity and the genesis of normal human behavior. The postsynaptic scaffolding proteins SAP90/PSD-95-associated proteins (SAPAPs), which are abundant components of the postsynaptic density (PSD) at excitatory synapses, play critical roles in synaptic structure, formation, development, plasticity, and signaling. The convergence of human genetic data with recent in vitro and in vivo animal model data indicates that mutations in the genes encoding SAPAP1-4 are associated with neurological and psychiatric disorders, and that dysfunction of SAPAP scaffolding proteins may contribute to the pathogenesis of various neuropsychiatric disorders, such as schizophrenia, autism spectrum disorders, obsessive compulsive disorders, Alzheimer's disease, and bipolar disorder. Here, we review recent major genetic, epigenetic, molecular, behavioral, electrophysiological, and circuitry studies that have advanced our knowledge by clarifying the roles of SAPAP proteins at the synapses, providing new insights into the mechanistic links to neurodevelopmental and neuropsychiatric disorders.
Collapse
Affiliation(s)
- Yunxia Bai
- Key Laboratory of Brain Functional Genomics (STCSM & MOE), Affiliated Mental Health Center (ECNU), School of Psychology and Cognitive Science, East China Normal University, Shanghai 200062, China
- Shanghai Changning Mental Health Center, Shanghai 200335, China
| | - Huimin Wang
- Key Laboratory of Brain Functional Genomics (STCSM & MOE), Affiliated Mental Health Center (ECNU), School of Psychology and Cognitive Science, East China Normal University, Shanghai 200062, China
- Shanghai Changning Mental Health Center, Shanghai 200335, China
- NYU-ECNU Institute of Brain and Cognitive Science at NYU Shanghai, Shanghai 200062, China
| | - Chunxia Li
- Key Laboratory of Brain Functional Genomics (STCSM & MOE), Affiliated Mental Health Center (ECNU), School of Psychology and Cognitive Science, East China Normal University, Shanghai 200062, China
- Shanghai Changning Mental Health Center, Shanghai 200335, China
- Correspondence:
| |
Collapse
|
147
|
Wang Y, Xu Y, Guo W, Fang Y, Hu L, Wang R, Zhao R, Guo D, Qi B, Ren G, Ren J, Li Y, Zhang M. Ablation of Shank3 alleviates cardiac dysfunction in aging mice by promoting CaMKII activation and Parkin-mediated mitophagy. Redox Biol 2022; 58:102537. [PMID: 36436456 PMCID: PMC9709154 DOI: 10.1016/j.redox.2022.102537] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 11/08/2022] [Indexed: 11/19/2022] Open
Abstract
Compromised mitophagy and mitochondrial homeostasis are major contributors for the etiology of cardiac aging, although the precise underlying mechanisms remains elusive. Shank3, a heart-enriched protein, has recently been reported to regulate aging-related neurodegenerative diseases. This study aimed to examine the role of Shank3 in the pathogenesis of cardiac senescence and the possible mechanisms involved. Cardiac-specific conditional Shank3 knockout (Shank3CKO) mice were subjected to natural aging. Mitochondrial function and mitophagy activity were determined in vivo, in mouse hearts and in vitro, in cardiomyocytes. Here, we showed that cardiac Shank3 expression exhibited a gradual increase during the natural progression of the aging, accompanied by overtly decreased mitophagy activity and a decline in cardiac function. Ablation of Shank3 promoted mitophagy, reduced mitochondria-derived superoxide (H2O2 and O2•-) production and apoptosis, and protected against cardiac dysfunction in the aged heart. In an in vitro study, senescent cardiomyocytes treated with D-gal exhibited reduced mitophagy and significantly elevated Shank3 expression. Shank3 knock-down restored mitophagy, leading to increased mitochondrial membrane potential, decreased mitochondrial oxidative stress, and reduced apoptosis in senescent cardiomyocytes, whereas Shank3 overexpression mimicked D-gal-induced mitophagy inhibition and mitochondrial dysfunction in normally cultured cardiomyocytes. Mechanistically, the IP assay revealed that Shank3 directly binds to CaMKII, and this interaction was further increased in the aged heart. Enhanced Shank3/CaMKII binding impedes mitochondrial translocation of CaMKII, resulting in the inhibition of parkin-mediated mitophagy, which ultimately leads to mitochondrial dysfunction and cardiac damage in the aged heart. Our study identified Shank3 as a novel contributor to aging-related cardiac damage. Manipulating Shank3/CaMKII-induced mitophagy inhibition could thus be an optional strategy for therapeutic intervention in clinical aging-related cardiac dysfunctions.
Collapse
Affiliation(s)
- Ying Wang
- Department of Cardiology, Tangdu Hospital, Air Force Medical University, Xi’an, 710038, China
| | - Yuerong Xu
- Department of Orthodontics, School of Stomatology, Air Force Medical University, Xi’an, 710032, China
| | - Wangang Guo
- Department of Cardiology, Tangdu Hospital, Air Force Medical University, Xi’an, 710038, China
| | - Yexian Fang
- Department of Cardiology, Tangdu Hospital, Air Force Medical University, Xi’an, 710038, China
| | - Lang Hu
- Department of Cardiology, Tangdu Hospital, Air Force Medical University, Xi’an, 710038, China
| | - Runze Wang
- Department of Cardiology, Tangdu Hospital, Air Force Medical University, Xi’an, 710038, China
| | - Ran Zhao
- Department of Cardiology, Tangdu Hospital, Air Force Medical University, Xi’an, 710038, China
| | - Dong Guo
- Department of Cardiology, Tangdu Hospital, Air Force Medical University, Xi’an, 710038, China
| | - Bingchao Qi
- Department of Cardiology, Tangdu Hospital, Air Force Medical University, Xi’an, 710038, China
| | - Gaotong Ren
- Department of Cardiology, Tangdu Hospital, Air Force Medical University, Xi’an, 710038, China
| | - Jun Ren
- Department of Cardiology and Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China,Corresponding author. Department of Cardiology and Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China.
| | - Yan Li
- Department of Cardiology, Tangdu Hospital, Air Force Medical University, Xi’an, 710038, China,Corresponding author.
| | - Mingming Zhang
- Department of Cardiology, Tangdu Hospital, Air Force Medical University, Xi’an, 710038, China,Corresponding author.
| |
Collapse
|
148
|
Groves Kuhnle C, Grimes M, Suárez Casanova VM, Turrigiano GG, Van Hooser SD. Juvenile Shank3 KO Mice Adopt Distinct Hunting Strategies during Prey Capture Learning. eNeuro 2022; 9:ENEURO.0230-22.2022. [PMID: 36446569 PMCID: PMC9768843 DOI: 10.1523/eneuro.0230-22.2022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 10/11/2022] [Accepted: 10/23/2022] [Indexed: 12/02/2022] Open
Abstract
Mice are opportunistic omnivores that readily learn to hunt and eat insects such as crickets. The details of how mice learn these behaviors and how these behaviors may differ in strains with altered neuroplasticity are unclear. We quantified the behavior of juvenile wild-type (WT) and Shank3 knock-out (KO) mice as they learned to hunt crickets during the critical period for ocular dominance plasticity. This stage involves heightened cortical plasticity including homeostatic synaptic scaling, which requires Shank3, a glutamatergic synaptic protein that, when mutated, produces Phelan-McDermid syndrome and is often comorbid with autism spectrum disorder (ASD). Both strains showed interest in examining live and dead crickets and learned to hunt. Shank3 knock-out mice took longer to become proficient, and, after 5 d, did not achieve the efficiency of wild-type mice in either time-to-capture or distance-to-capture. Shank3 knock-out mice also exhibited different characteristics when pursuing crickets that could not be explained by a simple motor deficit. Although both genotypes moved at the same average speed when approaching a cricket, Shank3 KO mice paused more often, did not begin final accelerations toward crickets as early, and did not close the distance gap to the cricket as quickly as wild-type mice. These differences in Shank3 KO mice are reminiscent of some behavioral characteristics of individuals with ASD as they perform complex tasks, such as slower action initiation and completion. This paradigm will be useful for exploring the neural circuit mechanisms that underlie these learning and performance differences in monogenic ASD rodent models.
Collapse
Affiliation(s)
| | - Micaela Grimes
- Department of Biology, Brandeis University, Waltham, MA 02453
| | | | | | | |
Collapse
|
149
|
SHANK family on stem cell fate and development. Cell Death Dis 2022; 13:880. [PMID: 36257935 PMCID: PMC9579136 DOI: 10.1038/s41419-022-05325-3] [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: 07/25/2022] [Revised: 09/30/2022] [Accepted: 10/05/2022] [Indexed: 11/24/2022]
Abstract
SH3 and multiple ankyrin repeat domains protein (SHANK) 1, SHANK2, and SHANK3 encode a family of postsynaptic scaffolding proteins present at glutamatergic synapses and play a crucial role in synaptogenesis. In the past years, studies have provided a preliminary appreciation and understanding of the influence of the SHANK family in controlling stem cell fate. Here, we review the modulation of SHANK gene expression and their related signaling pathways, allowing for an in-depth understanding of the role of SHANK in stem cells. Besides, their role in governing stem cell self-renewal, proliferation, differentiation, apoptosis, and metabolism are explored in neural stem cells (NSCs), stem cells from apical papilla (SCAPs), and induced pluripotent stem cells (iPSCs). Moreover, iPSCs and embryonic stem cells (ESCs) have been utilized as model systems for analyzing their functions in terms of neuronal development. SHANK-mediated stem cell fate determination is an intricate and multifactorial process. This study aims to achieve a better understanding of the role of SHANK in these processes and their clinical applications, thereby advancing the field of stem cell therapy. This review unravels the regulatory role of the SHANK family in the fate of stem cells.
Collapse
|
150
|
Michetti C, Falace A, Benfenati F, Fassio A. Synaptic genes and neurodevelopmental disorders: From molecular mechanisms to developmental strategies of behavioral testing. Neurobiol Dis 2022; 173:105856. [PMID: 36070836 DOI: 10.1016/j.nbd.2022.105856] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 08/29/2022] [Accepted: 08/30/2022] [Indexed: 10/14/2022] Open
Abstract
Synaptopathies are a class of neurodevelopmental disorders caused by modification in genes coding for synaptic proteins. These proteins oversee the process of neurotransmission, mainly controlling the fusion and recycling of synaptic vesicles at the presynaptic terminal, the expression and localization of receptors at the postsynapse and the coupling between the pre- and the postsynaptic compartments. Murine models, with homozygous or heterozygous deletion for several synaptic genes or knock-in for specific pathogenic mutations, have been developed. They have proved to be extremely informative for understanding synaptic physiology, as well as for clarifying the patho-mechanisms leading to developmental delay, epilepsy and motor, cognitive and social impairments that are the most common clinical manifestations of neurodevelopmental disorders. However, the onset of these disorders emerges during infancy and adolescence while the behavioral phenotyping is often conducted in adult mice, missing important information about the impact of synaptic development and maturation on the manifestation of the behavioral phenotype. Here, we review the main achievements obtained by behavioral testing in murine models of synaptopathies and propose a battery of behavioral tests to improve classification, diagnosis and efficacy of potential therapeutic treatments. Our aim is to underlie the importance of studying behavioral development and better focusing on disease onset and phenotypes.
Collapse
Affiliation(s)
- Caterina Michetti
- Department of Experimental Medicine, University of Genoa, Genoa, Italy; Center for Synaptic Neuroscience, Istituto Italiano di Tecnologia, Genoa, Italy.
| | - Antonio Falace
- Pediatric Neurology, Neurogenetics and Neurobiology Unit and Laboratories, Children's Hospital A. Meyer-University of Florence, Florence, Italy
| | - Fabio Benfenati
- Center for Synaptic Neuroscience, Istituto Italiano di Tecnologia, Genoa, Italy; IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Anna Fassio
- Department of Experimental Medicine, University of Genoa, Genoa, Italy; IRCCS Ospedale Policlinico San Martino, Genoa, Italy.
| |
Collapse
|