1
|
Kim Y, Ma R, Zhang Y, Kang HR, Kim US, Han K. Cell-autonomous reduction of CYFIP2 changes dendrite length, dendritic protrusion morphology, and inhibitory synapse density in the hippocampal CA1 pyramidal neurons of 17-month-old mice. Anim Cells Syst (Seoul) 2024; 28:294-302. [PMID: 38832126 PMCID: PMC11146249 DOI: 10.1080/19768354.2024.2360740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 05/22/2024] [Indexed: 06/05/2024] Open
Abstract
The cytoplasmic FMR1-interacting protein 2 (CYFIP2) have diverse molecular functions in neurons, including the regulation of actin polymerization, mRNA translation, and mitochondrial morphology and function. Mutations in the CYFIP2 gene are associated with early-onset epilepsy and neurodevelopmental disorders, while decreases in its protein levels are linked to Alzheimer's disease (AD). Notably, previous research has revealed AD-like phenotypes, such as dendritic spine loss, in the hippocampal CA1 pyramidal neurons of 12-month-old Cyfip2 heterozygous mice but not of age-matched CA1 pyramidal neuron-specific Cyfip2 conditional knock-out (cKO) mice. This study aims to investigate whether dendritic spine loss in Cyfip2 cKO mice is merely delayed compared to Cyfip2 heterozygous mice, and to explore further neuronal phenotypes regulated by CYFIP2 in aged mice. We characterized dendrite and dendritic protrusion morphologies, along with excitatory/inhibitory synapse densities in CA1 pyramidal neurons of 17-month-old Cyfip2 cKO mice. Overall dendritic branching was normal, with a reduction in the length of basal, not apical, dendrites in CA1 pyramidal neurons of Cyfip2 cKO mice. Furthermore, while dendritic protrusion density remained normal, alterations were observed in the length of mushroom spines and the head volume of stubby spines in basal, not apical, dendrites of Cyfip2 cKO mice. Although excitatory synapse density remained unchanged, inhibitory synapse density increased in apical, not basal, dendrites of Cyfip2 cKO mice. Consequently, a cell-autonomous reduction of CYFIP2 appears insufficient to induce dendritic spine loss in CA1 pyramidal neurons of aged mice. However, CYFIP2 is required to maintain normal dendritic length, dendritic protrusion morphology, and inhibitory synapse density.
Collapse
Affiliation(s)
- Yoonhee Kim
- Department of Neuroscience, 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
| | - 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
| | - U Suk Kim
- Department of Neuroscience, Korea University College of Medicine, Seoul, Republic of Korea
| | - 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
| |
Collapse
|
2
|
Biosca-Brull J, Guardia-Escote L, Basaure P, Cabré M, Blanco J, Pérez-Fernández C, Sánchez-Santed F, Domingo JL, Colomina MT. Exposure to chlorpyrifos during pregnancy differentially affects social behavior and GABA signaling elements in an APOE- and sex-dependent manner in a transgenic mouse model. ENVIRONMENTAL RESEARCH 2023; 224:115461. [PMID: 36796608 DOI: 10.1016/j.envres.2023.115461] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 02/06/2023] [Accepted: 02/08/2023] [Indexed: 06/18/2023]
Abstract
The massive use of chlorpyrifos (CPF) has been associated with an increased prevalence of neurodevelopmental disorders. Some previous studies have shown that prenatal, but not postnatal, CPF exposure causes social behavior deficits in mice depending on sex while others have found that in transgenic mice models carrying the human apolipoprotein E (APOE) ε3 and ε4 allele confer different vulnerabilities to either behavioral or metabolic disorders after CPF exposure. This study aims to evaluate, in both sexes, how prenatal CPF exposure and APOE genotype impact on social behavior and its relation to changes in GABAergic and glutamatergic systems. For this purpose, apoE3 and apoE4 transgenic mice were exposed through the diet to 0 or 1 mg/kg/day of CPF, between gestational day 12 and 18. A three-chamber test was used to assess social behavior on postnatal day (PND) 45. Then, mice were sacrificed, and hippocampal samples were analyzed to study the gene expression of GABAergic and glutamatergic elements. Results showed that prenatal exposure to CPF impaired social novelty preference and increased the expression of GABA-A α1 subunit in females of both genotypes. In addition, the expression of GAD1, the ionic cotransporter KCC2 and the GABA-A α2 and α5 subunits were increased in apoE3 mice, whereas CPF treatment only accentuated the expression of GAD1 and KCC2. Nevertheless, future research is needed to evaluate whether the influences detected in the GABAergic system are present and functionally relevant in adults and old mice.
Collapse
Affiliation(s)
- Judit Biosca-Brull
- Universitat Rovira i Virgili, Research Group in Neurobehavior and Health (NEUROLAB), Tarragona, Spain; Universitat Rovira i Virgili, Department of Psychology and Research Center for Behavior Assessment (CRAMC), Tarragona, Spain; Universitat Rovira i Virgili, Laboratory of Toxicology and Environmental Health, School of Medicine, Reus, Spain.
| | - Laia Guardia-Escote
- Universitat Rovira i Virgili, Research Group in Neurobehavior and Health (NEUROLAB), Tarragona, Spain; Universitat Rovira i Virgili, Department of Psychology and Research Center for Behavior Assessment (CRAMC), Tarragona, Spain
| | - Pia Basaure
- Universitat Rovira i Virgili, Research Group in Neurobehavior and Health (NEUROLAB), Tarragona, Spain
| | - Maria Cabré
- Universitat Rovira i Virgili, Research Group in Neurobehavior and Health (NEUROLAB), Tarragona, Spain; Universitat Rovira i Virgili, Department of Biochemistry and Biotechnology, Tarragona, Spain
| | - Jordi Blanco
- Universitat Rovira i Virgili, Research Group in Neurobehavior and Health (NEUROLAB), Tarragona, Spain; Universitat Rovira i Virgili, Laboratory of Toxicology and Environmental Health, School of Medicine, Reus, Spain; Universitat Rovira i Virgili, Department of Basic Medical Sciences, Reus, Spain
| | - Cristian Pérez-Fernández
- Department of Psychology, Health Research Center (CEINSA), Almeria University, 04120, Almeria, Spain
| | - Fernando Sánchez-Santed
- Department of Psychology, Health Research Center (CEINSA), Almeria University, 04120, Almeria, Spain
| | - José L Domingo
- Universitat Rovira i Virgili, Laboratory of Toxicology and Environmental Health, School of Medicine, Reus, Spain
| | - Maria Teresa Colomina
- Universitat Rovira i Virgili, Research Group in Neurobehavior and Health (NEUROLAB), Tarragona, Spain; Universitat Rovira i Virgili, Department of Psychology and Research Center for Behavior Assessment (CRAMC), Tarragona, Spain; Universitat Rovira i Virgili, Laboratory of Toxicology and Environmental Health, School of Medicine, Reus, Spain.
| |
Collapse
|
3
|
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
|
4
|
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
|
5
|
Ma R, Zhang Y, Li H, Kang HR, Kim Y, Han K. Cell-autonomous reduction of CYFIP2 is insufficient to induce Alzheimer's disease-like pathologies in the hippocampal CA1 pyramidal neurons of aged mice. Anim Cells Syst (Seoul) 2023; 27:93-101. [PMID: 36999135 PMCID: PMC10044167 DOI: 10.1080/19768354.2023.2192263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/01/2023] Open
Abstract
Cytoplasmic FMR1-interacting protein 2 (CYFIP2) is an evolutionarily conserved multifunctional protein that regulates the neuronal actin cytoskeleton, mRNA translation and transport, and mitochondrial morphology and function. Supporting its critical roles in proper neuronal development and function, human genetic studies have repeatedly identified variants of the CYFIP2 gene in individuals diagnosed with neurodevelopmental disorders. Notably, a few recent studies have also suggested a mechanistic link between reduced CYFIP2 level and Alzheimer's disease (AD). Specifically, in the hippocampus of 12-month-old Cyfip2 heterozygous mice, several AD-like pathologies were identified, including increased levels of Tau phosphorylation and gliosis, and loss of dendritic spines in CA1 pyramidal neurons. However, detailed pathogenic mechanisms, such as cell types and their circuits where the pathologies originate, remain unknown for AD-like pathologies caused by CYFIP2 reduction. In this study, we aimed to address this issue by examining whether the cell-autonomous reduction of CYFIP2 in CA1 excitatory pyramidal neurons is sufficient to induce AD-like phenotypes in the hippocampus. We performed immunohistochemical, morphological, and biochemical analyses in 12-month-old Cyfip2 conditional knock-out mice, which have postnatally reduced CYFIP2 expression level in CA1, but not in CA3, excitatory pyramidal neurons of the hippocampus. Unexpectedly, we could not find any significant AD-like phenotype, suggesting that the CA1 excitatory neuron-specific reduction of CYFIP2 level is insufficient to lead to AD-like pathologies in the hippocampus. Therefore, we propose that CYFIP2 reduction in other neurons and/or their synaptic connections with CA1 pyramidal neurons may be critically involved in the hippocampal AD-like phenotypes of Cyfip2 heterozygous mice.
Collapse
Affiliation(s)
- 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
| | - Yinhua Zhang
- Department of Neuroscience, 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
| | - 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
| | - Yoonhee Kim
- Department of Neuroscience, Korea University College of Medicine, Seoul, Republic of Korea
| | - 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
- Kihoon Han
| |
Collapse
|
6
|
Biosca-Brull J, Guardia-Escote L, Blanco J, Basaure P, Cabré M, Sánchez-Santed F, Domingo JL, Colomina MT. Prenatal, but not postnatal exposure to chlorpyrifos affects social behavior of mice and the excitatory-inhibitory balance in a sex-dependent manner. Food Chem Toxicol 2022; 169:113423. [PMID: 36113784 DOI: 10.1016/j.fct.2022.113423] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Accepted: 08/26/2022] [Indexed: 11/17/2022]
Abstract
The balance between excitatory and inhibitory neurotransmitters is essential for proper brain development. An imbalance between these two systems has been associated with neurodevelopmental disorders. On the other hand, literature also associates the massive use of pesticides with the increase of these disorders, with a particular focus on chlorpyrifos (CPF) a world-wide used organophosphate pesticide. This study was aimed at assessing social autistic-like behaviors on mice pre or postnatally exposed to CPF (0 or 1 mg/kg/day), in both sexes. In prenatal exposure, C57BL/6J pregnant mice were exposed to CPF through the diet, between gestational days (GD) 12 and 18, while a positive control group for some autistic behaviors was exposed to valproic acid (VPA) on GD 12 and 13. To assess postnatal exposure, C57BL/6J mice were orally exposed to the vehicle (corn oil) or CPF, from postnatal days (PND) 10-15. Social behavior and gene expression analysis were assessed on PND 45. Results showed social alterations only in males prenatally treated. GABA system was upregulated in CPF-treated females, whereas an increase in both systems was observed in both treated males. These findings suggest that males are more sensitive to prenatal CPF exposure, favoring the sex bias observed in ASD.
Collapse
Affiliation(s)
- Judit Biosca-Brull
- Universitat Rovira i Virgili, Research Group in Neurobehavior and Health (NEUROLAB), Tarragona, Spain; Universitat Rovira i Virgili, Department of Psychology and Research Center for Behavior Assessment (CRAMC), Tarragona, Spain; Universitat Rovira i Virgili, Laboratory of Toxicology and Environmental Health (TECNATOX), Reus, Spain.
| | - Laia Guardia-Escote
- Universitat Rovira i Virgili, Research Group in Neurobehavior and Health (NEUROLAB), Tarragona, Spain; Universitat Rovira i Virgili, Department of Psychology and Research Center for Behavior Assessment (CRAMC), Tarragona, Spain
| | - Jordi Blanco
- Universitat Rovira i Virgili, Research Group in Neurobehavior and Health (NEUROLAB), Tarragona, Spain; Universitat Rovira i Virgili, Laboratory of Toxicology and Environmental Health (TECNATOX), Reus, Spain; Universitat Rovira i Virgili, Department of Basic Medical Sciences, Reus, Spain
| | - Pia Basaure
- Universitat Rovira i Virgili, Research Group in Neurobehavior and Health (NEUROLAB), Tarragona, Spain
| | - Maria Cabré
- Universitat Rovira i Virgili, Research Group in Neurobehavior and Health (NEUROLAB), Tarragona, Spain; Universitat Rovira i Virgili, Department of Biochemistry and Biotechnology, Tarragona, Spain
| | - Fernando Sánchez-Santed
- Department of Psychology, Health Research Center (CEINSA), Almeria University, 04120, Almeria, Spain
| | - José L Domingo
- Universitat Rovira i Virgili, Laboratory of Toxicology and Environmental Health (TECNATOX), Reus, Spain
| | - Maria Teresa Colomina
- Universitat Rovira i Virgili, Research Group in Neurobehavior and Health (NEUROLAB), Tarragona, Spain; Universitat Rovira i Virgili, Department of Psychology and Research Center for Behavior Assessment (CRAMC), Tarragona, Spain; Universitat Rovira i Virgili, Laboratory of Toxicology and Environmental Health (TECNATOX), Reus, Spain.
| |
Collapse
|
7
|
Abu Bakar N, Wan Ibrahim WN, Che Abdullah CA, Ramlan NF, Shaari K, Shohaimi S, Mediani A, Nasruddin NS, Kim CH, Mohd Faudzi SM. Embryonic Arsenic Exposure Triggers Long-Term Behavioral Impairment with Metabolite Alterations in Zebrafish. TOXICS 2022; 10:493. [PMID: 36136458 PMCID: PMC9502072 DOI: 10.3390/toxics10090493] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 08/19/2022] [Accepted: 08/21/2022] [Indexed: 05/10/2023]
Abstract
Arsenic trioxide (As2O3) is a ubiquitous heavy metal in the environment. Exposure to this toxin at low concentrations is unremarkable in developing organisms. Nevertheless, understanding the underlying mechanism of its long-term adverse effects remains a challenge. In this study, embryos were initially exposed to As2O3 from gastrulation to hatching under semi-static conditions. Results showed dose-dependent increased mortality, with exposure to 30-40 µM As2O3 significantly reducing tail-coiling and heart rate at early larval stages. Surviving larvae after 30 µM As2O3 exposure showed deficits in motor behavior without impairment of anxiety-like responses at 6 dpf and a slight impairment in color preference behavior at 11 dpf, which was later evident in adulthood. As2O3 also altered locomotor function, with a loss of directional and color preference in adult zebrafish, which correlated with changes in transcriptional regulation of adsl, shank3a, and tsc1b genes. During these processes, As2O3 mainly induced metabolic changes in lipids, particularly arachidonic acid, docosahexaenoic acid, prostaglandin, and sphinganine-1-phosphate in the post-hatching period of zebrafish. Overall, this study provides new insight into the potential mechanism of arsenic toxicity leading to long-term learning impairment in zebrafish and may benefit future risk assessments of other environmental toxins of concern.
Collapse
Affiliation(s)
- Noraini Abu Bakar
- Department of Biology, Faculty of Science, Universiti Putra Malaysia, Serdang 43400, Malaysia
| | - Wan Norhamidah Wan Ibrahim
- Department of Biology, Faculty of Science, Universiti Putra Malaysia, Serdang 43400, Malaysia
- Natural Medicines and Products Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia, Serdang 43400, Malaysia
| | - Che Azurahanim Che Abdullah
- Department of Physics, Faculty of Science, Universiti Putra Malaysia, Serdang 43400, Malaysia
- The Institute of Advanced Technology (ITMA), Universiti Putra Malaysia, Serdang 43400, Malaysia
| | - Nurul Farhana Ramlan
- Department of Biology, Faculty of Science, Universiti Putra Malaysia, Serdang 43400, Malaysia
| | - Khozirah Shaari
- Natural Medicines and Products Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia, Serdang 43400, Malaysia
| | - Shamarina Shohaimi
- Department of Biology, Faculty of Science, Universiti Putra Malaysia, Serdang 43400, Malaysia
| | - Ahmed Mediani
- Institute of Systems Biology, Universiti Kebangsaan Malaysia (UKM), Bangi 43600, Malaysia
| | - Nurrul Shaqinah Nasruddin
- Centre for Craniofacial Diagnostics, Faculty of Dentistry, Universiti Kebangsaan Malaysia (UKM), Kuala Lumpur 50300, Malaysia
| | - Cheol-Hee Kim
- Department of Biology, Chungnam National University, Daejeon 34134, Korea
| | - Siti Munirah Mohd Faudzi
- Natural Medicines and Products Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia, Serdang 43400, Malaysia
- Department of Chemistry, Faculty of Science, Universiti Putra Malaysia, Serdang 43400, Malaysia
| |
Collapse
|
8
|
Ma R, Pang K, Kang H, Zhang Y, Bang G, Park S, Hwang E, Ryu JR, Kwon Y, Kang HR, Jin C, Kim Y, Kim SY, Kwon SK, Kim D, Sun W, Kim JY, Han K. Protein interactome and cell-type expression analyses reveal that cytoplasmic FMR1-interacting protein 1 (CYFIP1), but not CYFIP2, associates with astrocytic focal adhesion. J Neurochem 2022; 162:190-206. [PMID: 35567753 DOI: 10.1111/jnc.15622] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 02/24/2022] [Accepted: 05/11/2022] [Indexed: 11/28/2022]
Abstract
The two members of the cytoplasmic FMR1-interacting protein family, CYFIP1 and CYFIP2, are evolutionarily conserved multifunctional proteins whose defects are associated with distinct types of brain disorders. Even with high sequence homology between CYFIP1 and CYFIP2, several lines of evidence indicate their different functions in the brain; however, the underlying mechanisms remain largely unknown. Here, we performed reciprocal immunoprecipitation experiments using CYFIP1-2×Myc and CYFIP2-3×Flag knock-in mice and found that CYFIP1 and CYFIP2 are not significantly co-immunoprecipitated with each other in the knock-in brains compared to negative control wild-type brains. Moreover, CYFIP1 and CYFIP2 showed different size distributions by size-exclusion chromatography of wild-type mouse brains. Specifically, mass spectrometry-based analysis of CYFIP1-2×Myc knock-in brains identified 131 proteins in the CYFIP1 interactome. Comparison of the CYFIP1 interactome with the previously identified brain region- and age-matched CYFIP2 interactome, consisting of 140 proteins, revealed only eight common proteins. Investigations using single-cell RNA-sequencing databases suggested non-neuronal cell- and neuron-enriched expression of Cyfip1 and Cyfip2, respectively. At the protein level, CYFIP1 was detected in both neurons and astrocytes, while CYFIP2 was detected only in neurons, suggesting the predominant expression of CYFIP1 in astrocytes. Bioinformatic characterization of the CYFIP1 interactome, and co-expression analysis of Cyfip1 with astrocytic genes, commonly linked CYFIP1 with focal adhesion proteins. Immunocytochemical analysis and proximity ligation assay suggested partial co-localization of CYFIP1 and focal adhesion proteins in cultured astrocytes. Together, these results suggest a CYFIP1-specific association with astrocytic focal adhesion, which may contribute to the different brain functions and dysfunctions of CYFIP1 and CYFIP2.
Collapse
Affiliation(s)
- Ruiying Ma
- Department of Neuroscience, Korea University College of Medicine, Seoul, 02841, Republic of Korea.,BK21 Graduate Program, Department of Biomedical Sciences, Korea University College of Medicine, Seoul, 02841, Republic of Korea
| | - Kaifang Pang
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas, 77030, USA.,Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, Texas, 77030, USA
| | - Hyojin Kang
- Division of National Supercomputing, Korea Institute of Science and Technology Information (KISTI), Daejeon, 34141, Republic of Korea
| | - Yinhua Zhang
- Department of Neuroscience, Korea University College of Medicine, Seoul, 02841, Republic of Korea.,BK21 Graduate Program, Department of Biomedical Sciences, Korea University College of Medicine, Seoul, 02841, Republic of Korea
| | - Geul Bang
- Research Center for Bioconvergence Analysis, Korea Basic Science Institute (KBSI), Ochang 28119, Republic of Korea.,Korea University College of Pharmacy, Sejong, 30019, Republic of Korea
| | - Sangwoo Park
- Research Center for Bioconvergence Analysis, Korea Basic Science Institute (KBSI), Ochang 28119, Republic of Korea
| | - Eunha Hwang
- Center for Research Equipment, Korea Basic Science Institute (KBSI), Ochang 28119, Republic of Korea
| | - Jae Ryun Ryu
- Department of Anatomy, Korea University College of Medicine, Seoul, 02841, Republic of Korea
| | - Yujin Kwon
- Therapeutics & Biotechnology Division, Drug discovery platform research center, Korea Research Institute of Chemical Technology (KRICT), Daejeon, 34114, Republic of Korea
| | - Hyae Rim Kang
- Department of Neuroscience, Korea University College of Medicine, Seoul, 02841, Republic of Korea.,BK21 Graduate Program, Department of Biomedical Sciences, Korea University College of Medicine, Seoul, 02841, Republic of Korea
| | - Chunmei Jin
- Department of Neuroscience, Korea University College of Medicine, Seoul, 02841, Republic of Korea.,BK21 Graduate Program, Department of Biomedical Sciences, Korea University College of Medicine, Seoul, 02841, Republic of Korea
| | - Yoonhee Kim
- Department of Neuroscience, Korea University College of Medicine, Seoul, 02841, Republic of Korea
| | - Su Yeon Kim
- Department of Neuroscience, Korea University College of Medicine, Seoul, 02841, Republic of Korea.,Korea Institute of Science and Technology (KIST), Brain Science Institute, Seoul, 02792, Republic of Korea
| | - Seok-Kyu Kwon
- Korea Institute of Science and Technology (KIST), Brain Science Institute, Seoul, 02792, Republic of Korea
| | - Doyoun Kim
- Therapeutics & Biotechnology Division, Drug discovery platform research center, Korea Research Institute of Chemical Technology (KRICT), Daejeon, 34114, Republic of Korea.,Medicinal Chemistry and Pharmacology, Korea University of Science and Technology (UST), Daejeon, 34113, Republic of Korea
| | - Woong Sun
- BK21 Graduate Program, Department of Biomedical Sciences, Korea University College of Medicine, Seoul, 02841, Republic of Korea.,Department of Anatomy, Korea University College of Medicine, Seoul, 02841, Republic of Korea
| | - Jin Young Kim
- Research Center for Bioconvergence Analysis, Korea Basic Science Institute (KBSI), Ochang 28119, Republic of Korea
| | - Kihoon Han
- Department of Neuroscience, Korea University College of Medicine, Seoul, 02841, Republic of Korea.,BK21 Graduate Program, Department of Biomedical Sciences, Korea University College of Medicine, Seoul, 02841, Republic of Korea
| |
Collapse
|
9
|
Jiang S, Xiao L, Sun Y, He M, Gao C, Zhu C, Chang H, Ding J, Li W, Wang Y, Sun T, Wang F. The GABAB receptor agonist STX209 reverses the autism‑like behaviour in an animal model of autism induced by prenatal exposure to valproic acid. Mol Med Rep 2022; 25:154. [PMID: 35244195 PMCID: PMC8941376 DOI: 10.3892/mmr.2022.12670] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 02/16/2022] [Indexed: 11/06/2022] Open
Abstract
Autism spectrum disorder (ASD) is a lifelong neurodevelopmental condition characterized by impaired social interaction, compromised communication, and restrictive or stereotyped behaviours and interests. Due to the complex pathophysiology of ASD, there are currently no available medical therapies for improving the associated social deficits. Consequently, the present study investigated the effects of STX209, a selective γ‑aminobutyric acid type B receptor (GABABR2) agonist, on an environmental rodent model of autism. The mouse model of autism induced by prenatal exposure to valproic acid (VPA) was used to assess the therapeutic potential of STX209 on autism‑like behaviour in the present study. This study investigated the effects of STX209 on VPA model mice via behavioral testing and revealed a significant reversal of core/associated autism‑like behavior, including sociability and preference for social novelty, novelty recognition, locomotion and exploration activity and marble‑burying deficit. This may be associated with STX209 correcting dendritic arborization, spine density and GABABR2 expression in hippocampus of VPA model mice. However, expression of glutamic acid decarboxylase 65/67 in the hippocampus were not altered by STX209. The present results demonstrated that STX209 administration ameliorated autism‑like symptoms in mice exposed to VPA prenatally, suggesting that autism‑like symptoms in children with a history of prenatal VPA exposure may also benefit from treatment with the GABABR2 agonist STX209.
Collapse
Affiliation(s)
- Shucai Jiang
- Ningxia Key Laboratory of Craniocerebral Disease, Incubation Base of National Key Laboratory, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Lifei Xiao
- Ningxia Key Laboratory of Craniocerebral Disease, Incubation Base of National Key Laboratory, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Yu Sun
- Ningxia Key Laboratory of Craniocerebral Disease, Incubation Base of National Key Laboratory, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Maotao He
- Department of Diagnostic Pathology, School of Basic Medical Sciences, Weifang Medical University, Weifang, Shandong 261053, P.R. China
| | - Caibin Gao
- Ningxia Key Laboratory of Craniocerebral Disease, Incubation Base of National Key Laboratory, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Changliang Zhu
- Ningxia Key Laboratory of Craniocerebral Disease, Incubation Base of National Key Laboratory, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Haigang Chang
- Ningxia Key Laboratory of Craniocerebral Disease, Incubation Base of National Key Laboratory, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Jiangwei Ding
- Ningxia Key Laboratory of Craniocerebral Disease, Incubation Base of National Key Laboratory, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Wenchao Li
- Ningxia Key Laboratory of Craniocerebral Disease, Incubation Base of National Key Laboratory, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Yangyang Wang
- Ningxia Key Laboratory of Craniocerebral Disease, Incubation Base of National Key Laboratory, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Tao Sun
- Ningxia Key Laboratory of Craniocerebral Disease, Incubation Base of National Key Laboratory, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Feng Wang
- Ningxia Key Laboratory of Craniocerebral Disease, Incubation Base of National Key Laboratory, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
- Department of Neurosurgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, P.R. China
| |
Collapse
|
10
|
Montanari M, Martella G, Bonsi P, Meringolo M. Autism Spectrum Disorder: Focus on Glutamatergic Neurotransmission. Int J Mol Sci 2022; 23:ijms23073861. [PMID: 35409220 PMCID: PMC8998955 DOI: 10.3390/ijms23073861] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 03/24/2022] [Accepted: 03/29/2022] [Indexed: 12/16/2022] Open
Abstract
Disturbances in the glutamatergic system have been increasingly documented in several neuropsychiatric disorders, including autism spectrum disorder (ASD). Glutamate-centered theories of ASD are based on evidence from patient samples and postmortem studies, as well as from studies documenting abnormalities in glutamatergic gene expression and metabolic pathways, including changes in the gut microbiota glutamate metabolism in patients with ASD. In addition, preclinical studies on animal models have demonstrated glutamatergic neurotransmission deficits and altered expression of glutamate synaptic proteins. At present, there are no approved glutamatergic drugs for ASD, but several ongoing clinical trials are currently focusing on evaluating in autistic patients glutamatergic pharmaceuticals already approved for other conditions. In this review, we provide an overview of the literature concerning the role of glutamatergic neurotransmission in the pathophysiology of ASD and as a potential target for novel treatments.
Collapse
Affiliation(s)
- Martina Montanari
- Laboratory of Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, 00179 Rome, Italy; (M.M.); (G.M.)
- Department of Systems Neuroscience, University Tor Vergata, 00133 Rome, Italy
| | - Giuseppina Martella
- Laboratory of Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, 00179 Rome, Italy; (M.M.); (G.M.)
| | - Paola Bonsi
- Laboratory of Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, 00179 Rome, Italy; (M.M.); (G.M.)
- Correspondence: (P.B.); (M.M.)
| | - Maria Meringolo
- Laboratory of Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, 00179 Rome, Italy; (M.M.); (G.M.)
- Correspondence: (P.B.); (M.M.)
| |
Collapse
|
11
|
Shank3 Deficiency is Associated With Altered Profile of Neurotransmission Markers in Pups and Adult Mice. Neurochem Res 2021; 46:3342-3355. [PMID: 34453663 DOI: 10.1007/s11064-021-03435-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 08/04/2021] [Accepted: 08/20/2021] [Indexed: 12/26/2022]
Abstract
Alterations in the balance between excitation and inhibition, especially in the brain's critical developmental periods, are considered an integral part of the pathophysiology of autism. However, the precise mechanisms have not yet been established. SH3 and multiple Ankyrin repeat domains 3 (Shank3) deficient mice represent a well-established transgenic model of a neurodevelopmental disorder with autistic symptomatology. In this study, we characterize the consequences of Shank3 deficiency according to (1) expression of specific markers of different neuronal populations in pups and adult mice and (2) social behaviour and anxiety in adult mice. Our research found enhanced expression of serotonin transporter and choline acetyltransferase in the hippocampus and hypothalamus in Shank3-deficient pups. We demonstrated marked brain region differences in expression of excitatory glutamatergic markers in pups and adult Shank3 deficient mice. We also observed reduced expression of inhibitory GABAergic markers and GABA receptor subunits in several brain areas in both pups and adult Shank3 deficient mice. Further analysis of dopaminergic brain areas (nucleus accumbens, ventral tegmental area) revealed lower expression levels of GABAergic markers in adult Shank3 deficient mice. Adult Shank3- deficient mice exhibited excessive repetitive behaviour, a higher level of anxiety, and lower locomotor activity. Our data support the theory of an imbalance between excitatory and inhibitory neurotransmission in conditions of abnormal SHANK3 protein. We therefore suggest that autism-like conditions are accompanied by reduced expression of GABAergic markers in the brain during early development as well as in the adult age, which could be associated with long-lasting behavioural abnormalities.
Collapse
|
12
|
Jin C, Kang H, Yoo T, Ryu JR, Yoo YE, Ma R, Zhang Y, Kang HR, Kim Y, Seong H, Bang G, Park S, Kwon SK, Sun W, Kim H, Kim JY, Kim E, Han K. The Neomycin Resistance Cassette in the Targeted Allele of Shank3B Knock-Out Mice Has Potential Off-Target Effects to Produce an Unusual Shank3 Isoform. Front Mol Neurosci 2021; 13:614435. [PMID: 33505245 PMCID: PMC7831789 DOI: 10.3389/fnmol.2020.614435] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 12/02/2020] [Indexed: 01/20/2023] Open
Abstract
Variants of the SH3 and multiple ankyrin repeat domains 3 (SHANK3), which encodes postsynaptic scaffolds, are associated with brain disorders. The targeted alleles in a few Shank3 knock-out (KO) lines contain a neomycin resistance (Neo) cassette, which may perturb the normal expression of neighboring genes; however, this has not been investigated in detail. We previously reported an unexpected increase in the mRNA expression of Shank3 exons 1–12 in the brains of Shank3B KO mice generated by replacing Shank3 exons 13–16 with the Neo cassette. In this study, we confirmed that the increased Shank3 mRNA in Shank3B KO brains produced an unusual ∼60 kDa Shank3 isoform (Shank3-N), which did not properly localize to the synaptic compartment. Functionally, Shank3-N overexpression altered the dendritic spine morphology in cultured neurons. Importantly, Shank3-N expression in Shank3B KO mice was not a compensatory response to a reduction of full-length Shank3 because expression was still detected in the brain after normalizing the level of full-length Shank3. Moreover, in another Shank3 KO line (Shank3 gKO) with a similar Shank3 exonal deletion as that in Shank3B KO mice but without a Neo cassette, the mRNA expression levels of Shank3 exons 1–12 were lower than those of wild-type mice and Shank3-N was not detected in the brain. In addition, the expression levels of genes neighboring Shank3 on chromosome 15 were altered in the striatum of Shank3B KO but not Shank3 gKO mice. These results suggest that the Neo cassette has potential off-target effects in Shank3B KO mice.
Collapse
Affiliation(s)
- Chunmei Jin
- Department of Neuroscience, College of Medicine, Korea University, Seoul, South Korea.,Department of Biomedical Sciences, College of Medicine, Korea University, Seoul, South Korea
| | - Hyojin Kang
- Division of National Supercomputing, Korea Institute of Science and Technology Information, Daejeon, South Korea
| | - Taesun Yoo
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science, Daejeon, South Korea
| | - Jae Ryun Ryu
- Department of Anatomy, College of Medicine, Korea University, Seoul, South Korea
| | - Ye-Eun Yoo
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, South Korea
| | - Ruiying Ma
- Department of Neuroscience, College of Medicine, Korea University, Seoul, South Korea.,Department of Biomedical Sciences, College of Medicine, Korea University, Seoul, South Korea
| | - Yinhua Zhang
- Department of Neuroscience, College of Medicine, Korea University, Seoul, South Korea.,Department of Biomedical Sciences, College of Medicine, Korea University, Seoul, South Korea
| | - Hyae Rim Kang
- Department of Neuroscience, College of Medicine, Korea University, Seoul, South Korea.,Department of Biomedical Sciences, College of Medicine, Korea University, Seoul, South Korea
| | - Yoonhee Kim
- Department of Neuroscience, College of Medicine, Korea University, Seoul, South Korea
| | - Hyunyoung Seong
- Department of Neuroscience, College of Medicine, Korea University, Seoul, South Korea
| | - Geul Bang
- Research Center for Bioconvergence Analysis, Korea Basic Science Institute, Ochang, South Korea.,College of Pharmacy, Korea University, Sejong, South Korea
| | - Sangwoo Park
- Research Center for Bioconvergence Analysis, Korea Basic Science Institute, Ochang, South Korea
| | - Seok-Kyu Kwon
- Center for Functional Connectomics, Korea Institute of Science and Technology, Brain Science Institute, Seoul, South Korea
| | - Woong Sun
- Department of Biomedical Sciences, College of Medicine, Korea University, Seoul, South Korea.,Department of Anatomy, College of Medicine, Korea University, Seoul, South Korea
| | - Hyunkyung Kim
- Department of Biomedical Sciences, College of Medicine, Korea University, Seoul, South Korea.,Department of Biochemistry and Molecular Biology, College of Medicine, Korea University, Seoul, South Korea
| | - Jin Young Kim
- Research Center for Bioconvergence Analysis, Korea Basic Science Institute, Ochang, South Korea
| | - Eunjoon Kim
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science, Daejeon, South Korea.,Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, South Korea
| | - Kihoon Han
- Department of Neuroscience, College of Medicine, Korea University, Seoul, South Korea.,Department of Biomedical Sciences, College of Medicine, Korea University, Seoul, South Korea
| |
Collapse
|
13
|
Lee SH, Zhang Y, Park J, Kim B, Kim Y, Lee SH, Kim GH, Huh YH, Lee B, Kim Y, Lee Y, Kim JY, Kang H, Choi SY, Jang S, Li Y, Kim S, Jin C, Pang K, Kim E, Lee Y, Kim H, Kim E, Choi JH, Kim J, Lee KJ, Choi SY, Han K. Haploinsufficiency of Cyfip2 Causes Lithium-Responsive Prefrontal Dysfunction. Ann Neurol 2020; 88:526-543. [PMID: 32562430 DOI: 10.1002/ana.25827] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 05/22/2020] [Accepted: 06/14/2020] [Indexed: 12/17/2022]
Abstract
OBJECTIVE Genetic variants of the cytoplasmic FMR1-interacting protein 2 (CYFIP2) encoding an actin-regulatory protein are associated with brain disorders, including intellectual disability and epilepsy. However, specific in vivo neuronal defects and potential treatments for CYFIP2-associated brain disorders remain largely unknown. Here, we characterized Cyfip2 heterozygous (Cyfip2+/- ) mice to understand their neurobehavioral phenotypes and the underlying pathological mechanisms. Furthermore, we examined a potential treatment for such phenotypes of the Cyfip2+/- mice and specified a neuronal function mediating its efficacy. METHODS We performed behavioral analyses of Cyfip2+/- mice. We combined molecular, ultrastructural, and in vitro and in vivo electrophysiological analyses of Cyfip2+/- prefrontal neurons. We also selectively reduced CYFIP2 in the prefrontal cortex (PFC) of mice with virus injections. RESULTS Adult Cyfip2+/- mice exhibited lithium-responsive abnormal behaviors. We found increased filamentous actin, enlarged dendritic spines, and enhanced excitatory synaptic transmission and excitability in the adult Cyfip2+/- PFC that was restricted to layer 5 (L5) neurons. Consistently, adult Cyfip2+/- mice showed increased seizure susceptibility and auditory steady-state responses from the cortical electroencephalographic recordings. Among the identified prefrontal defects, lithium selectively normalized the hyperexcitability of Cyfip2+/- L5 neurons. RNA sequencing revealed reduced expression of potassium channel genes in the adult Cyfip2+/- PFC. Virus-mediated reduction of CYFIP2 in the PFC was sufficient to induce L5 hyperexcitability and lithium-responsive abnormal behavior. INTERPRETATION These results suggest that L5-specific prefrontal dysfunction, especially hyperexcitability, underlies both the pathophysiology and the lithium-mediated amelioration of neurobehavioral phenotypes in adult Cyfip2+/- mice, which can be implicated in CYFIP2-associated brain disorders. ANN NEUROL 2020;88:526-543.
Collapse
Affiliation(s)
- Seung-Hyun Lee
- Department of Physiology, Dental Research Institute, Seoul National University School of Dentistry
| | - Yinhua Zhang
- Department of Neuroscience, College of Medicine, Korea University.,Department of Biomedical Sciences, College of Medicine, Korea University
| | - Jina Park
- Center for Neuroscience, Korea Institute of Science and Technology, Seoul
| | - Bowon Kim
- Center for Neuroscience, Korea Institute of Science and Technology, Seoul
| | - Yangsik Kim
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science, Daejeon.,Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon
| | - Sang Hoon Lee
- Neural Circuits Research Group, Korea Brain Research Institute, Daegu
| | - Gyu Hyun Kim
- Neural Circuits Research Group, Korea Brain Research Institute, Daegu
| | - Yang Hoon Huh
- Center for Electron Microscopy Research, Korea Basic Science Institute, Chungcheongbuk-do
| | - Bokyoung Lee
- Department of Neuroscience, College of Medicine, Korea University
| | - Yoonhee Kim
- Department of Neuroscience, College of Medicine, Korea University.,Department of Biomedical Sciences, College of Medicine, Korea University
| | - Yeunkum Lee
- Department of Neuroscience, College of Medicine, Korea University.,Department of Biomedical Sciences, College of Medicine, Korea University
| | - Jin Yong Kim
- Department of Biomedical Sciences, College of Medicine, Korea University.,Department of Anatomy, College of Medicine, Korea University, Seoul
| | - Hyojin Kang
- Division of National Supercomputing, Korea Institute of Science and Technology Information, Daejeon, South Korea
| | - Su-Yeon Choi
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science, Daejeon.,Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon
| | - Seil Jang
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science, Daejeon.,Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon
| | - Yan Li
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science, Daejeon.,Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon
| | - Shinhyun Kim
- Department of Neuroscience, College of Medicine, Korea University.,Department of Biomedical Sciences, College of Medicine, Korea University
| | - Chunmei Jin
- Department of Neuroscience, College of Medicine, Korea University.,Department of Biomedical Sciences, College of Medicine, Korea University
| | - Kaifang Pang
- Department of Pediatrics, Baylor College of Medicine.,Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX
| | - Eunjeong Kim
- Department of Life Sciences, Pohang University of Science and Technology, Pohang
| | - Yoontae Lee
- Department of Life Sciences, Pohang University of Science and Technology, Pohang
| | - Hyun Kim
- Department of Biomedical Sciences, College of Medicine, Korea University.,Department of Anatomy, College of Medicine, Korea University, Seoul
| | - Eunjoon Kim
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science, Daejeon.,Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon
| | - Jee Hyun Choi
- Center for Neuroscience, Korea Institute of Science and Technology, Seoul
| | - Jeongjin Kim
- Center for Neuroscience, Korea Institute of Science and Technology, Seoul
| | - Kea Joo Lee
- Neural Circuits Research Group, Korea Brain Research Institute, Daegu.,Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science & Technology, Daegu, South Korea
| | - Se-Young Choi
- Department of Physiology, Dental Research Institute, Seoul National University School of Dentistry
| | - Kihoon Han
- Department of Neuroscience, College of Medicine, Korea University.,Department of Biomedical Sciences, College of Medicine, Korea University
| |
Collapse
|
14
|
Jin C, Kang HR, Kang H, Zhang Y, Lee Y, Kim Y, Han K. Unexpected Compensatory Increase in Shank3 Transcripts in Shank3 Knock-Out Mice Having Partial Deletions of Exons. Front Mol Neurosci 2019; 12:228. [PMID: 31607862 PMCID: PMC6761322 DOI: 10.3389/fnmol.2019.00228] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Accepted: 09/04/2019] [Indexed: 12/18/2022] Open
Abstract
Genetic variants of the SH3 and multiple ankyrin repeat domains 3 (SHANK3) gene, which encodes excitatory postsynaptic core scaffolds cause numerous brain disorders. Several lines of Shank3 knock-out (KO) mice with deletions of different Shank3 exons have previously been generated and characterized. The different Shank3 KO mouse lines have both common and line-specific phenotypes. Shank3 isoform diversity is considered a mechanism underlying phenotypic heterogeneity, and compensatory changes through regulation of Shank3 expression may contribute to this heterogeneity. However, whether such compensatory changes occur in Shank3 KO mouse lines has not been investigated in detail. Using previously reported RNA-sequencing analyses, we identified an unexpected increase in Shank3 transcripts in two different Shank3 mutant mouse lines (Shank3B and Shank3ΔC) having partial deletions of Shank3 exons. We validated an increase in Shank3 transcripts in the hippocampus, cortex, and striatum, but not in the cerebellum, of Shank3B heterozygous (HET) and KO mice, using qRT-PCR analyses. In particular, expression of the N-terminal exons 1–12, but not the more C-terminal exons 19–22, was observed to increase in Shank3B mice with deletion of exons 13–16. This suggests a selective compensatory activation of upstream Shank3 promoters. Furthermore, using domain-specific Shank3 antibodies, we confirmed that the increased Shank3 transcripts in Shank3B KO mice produced a small Shank3 isoform that was not detected in wild-type mice. Taken together, our results illustrate another layer of complexity in the regulation of Shank3 expression in the brain, which may also contribute to the phenotypic heterogeneity of different Shank3 KO mouse lines.
Collapse
Affiliation(s)
- Chunmei Jin
- Department of Neuroscience, College of Medicine, Korea University, Seoul, South Korea.,Department of Biomedical Sciences, College of Medicine, Korea University, Seoul, South Korea
| | - Hyae Rim Kang
- Department of Neuroscience, College of Medicine, Korea University, Seoul, South Korea.,Department of Biomedical Sciences, College of Medicine, Korea University, Seoul, South Korea
| | - Hyojin Kang
- Division of National Supercomputing, Korea Institute of Science & Technology Information (KISTI), Daejeon, South Korea
| | - Yinhua Zhang
- Department of Neuroscience, College of Medicine, Korea University, Seoul, South Korea.,Department of Biomedical Sciences, College of Medicine, Korea University, Seoul, South Korea
| | - Yeunkum Lee
- Department of Neuroscience, College of Medicine, Korea University, Seoul, South Korea.,Department of Biomedical Sciences, College of Medicine, Korea University, Seoul, South Korea
| | - Yoonhee Kim
- Department of Neuroscience, College of Medicine, Korea University, Seoul, South Korea.,Department of Biomedical Sciences, College of Medicine, Korea University, Seoul, South Korea
| | - Kihoon Han
- Department of Neuroscience, College of Medicine, Korea University, Seoul, South Korea.,Department of Biomedical Sciences, College of Medicine, Korea University, Seoul, South Korea
| |
Collapse
|
15
|
Jin C, Kim S, Kang H, Yun KN, Lee Y, Zhang Y, Kim Y, Kim JY, Han K. Shank3 regulates striatal synaptic abundance of Cyld, a deubiquitinase specific for Lys63-linked polyubiquitin chains. J Neurochem 2019; 150:776-786. [PMID: 31215654 DOI: 10.1111/jnc.14796] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 05/16/2019] [Accepted: 06/11/2019] [Indexed: 12/21/2022]
Abstract
The SH3 and multiple ankyrin repeat domains 3 (Shank3) proteins are core organizers of the postsynaptic density in neuronal excitatory synapses, and their defects cause various neurodevelopmental and neuropsychiatric disorders. Mechanistically, Shank3 directly and indirectly interacts with hundreds of synaptic proteins with diverse functions and potentially exerts its regulatory roles in synaptic development and function via these interactors. However, Shank3-dependent regulation of synaptic abundance has been validated in vivo for only a few Shank3 interactors. Here, using a quantitative proteomic analysis, we identified 136 proteins with altered synaptic abundance in the striatum of Shank3-overexpressing transgenic (TG) mice. By comparing these proteins with those found in a previous analysis of the postsynaptic density of Shank3 knock-out (KO) striatum, we identified and confirmed that cylindromatosis-associated deubiquitinase (Cyld), a deubiquitinase specific for Lys63-linked polyubiquitin chains, was up- and down-regulated in Shank3 TG and KO striatal synapses, respectively. Consistently, we found that the synaptic levels of Lys63-linked polyubiquitin chains were down- and up-regulated in the Shank3 TG and KO striata, respectively. Furthermore, by isolating and analyzing the synaptic Cyld complex, we generated a Cyld interactome consisting of 103 proteins, which may include Cyld substrates. Bioinformatic analyses suggested associations of the Cyld interactome with a few brain disorders and synaptic functions. Taken together, these results suggest that Shank3 regulates the synaptic abundance of Cyld in the mouse striatum and, thereby, potentially modulates the Lys63-linked polyubiquitination of striatal synaptic proteins.
Collapse
Affiliation(s)
- Chunmei Jin
- Departments of Neuroscience, College of Medicine, Korea University, Seoul, South Korea.,Biomedical Sciences, College of Medicine, Korea University, Seoul, South Korea
| | - Shinhyun Kim
- Departments of Neuroscience, College of Medicine, Korea University, Seoul, South Korea.,Biomedical Sciences, College of Medicine, Korea University, Seoul, South Korea
| | - Hyojin Kang
- Division of National Supercomputing, KISTI, Daejeon, South Korea
| | - Ki Na Yun
- Biomedical Omics Group, Korea Basic Science Institute, Ochang, Korea
| | - Yeunkum Lee
- Departments of Neuroscience, College of Medicine, Korea University, Seoul, South Korea.,Biomedical Sciences, College of Medicine, Korea University, Seoul, South Korea
| | - Yinhua Zhang
- Departments of Neuroscience, College of Medicine, Korea University, Seoul, South Korea.,Biomedical Sciences, College of Medicine, Korea University, Seoul, South Korea
| | - Yoonhee Kim
- Departments of Neuroscience, College of Medicine, Korea University, Seoul, South Korea
| | - Jin Young Kim
- Biomedical Omics Group, Korea Basic Science Institute, Ochang, Korea
| | - Kihoon Han
- Departments of Neuroscience, College of Medicine, Korea University, Seoul, South Korea.,Biomedical Sciences, College of Medicine, Korea University, Seoul, South Korea
| |
Collapse
|
16
|
Lee Y, Kang H, Jin C, Zhang Y, Kim Y, Han K. Transcriptome analyses suggest minimal effects of Shank3 dosage on directional gene expression changes in the mouse striatum. Anim Cells Syst (Seoul) 2019; 23:270-274. [PMID: 31489248 PMCID: PMC6711111 DOI: 10.1080/19768354.2019.1595142] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 03/01/2019] [Accepted: 03/03/2019] [Indexed: 12/22/2022] Open
Abstract
Both deletions and duplications of the SH3 and multiple ankyrin repeat domains 3 (SHANK3) gene, encoding excitatory postsynaptic scaffolds, are causally associated with various brain disorders, suggesting that proper Shank3 dosage is critical for normal brain development and function. In addition to its well-established synaptic functions, recent studies have suggested that Shank3 can also affect gene expression in the nucleus. However, it has not been investigated whether there are a group of genes whose directional expression is regulated in a Shank3 dosage-dependent manner (i.e. showing opposite changes in expression following Shank3 reduction and overexpression). This is an important issue to be examined for better understanding why neuronal development and function are sensitive to Shank3 dosage, and how much transcriptional changes contribute to neuronal phenotypes affected by Shank3 dosage. To examine this, we performed transcriptome analyses on the striatum of Shank3 heterozygous and knock-out mice, which identified three and 17 differentially expressed genes, respectively. We then compared the results to those of our previous striatal transcriptome analysis of Shank3 overexpressing mice and identified 31 candidate genes showing directional expression changes in a Shank3 dosage-dependent manner. However, overall, their Shank3 dosage-dependent fold changes were very subtle (average of absolute log2(fold change) was 0.139). Meanwhile, the gene set enrichment analyses of the striatal transcriptome suggested that Shank3 dosage may affect anchoring junction-related functions. Taken together, these results suggest that Shank3 dosage minimally affects directional gene expression changes in the mouse striatum.
Collapse
Affiliation(s)
- Yeunkum Lee
- Department of Neuroscience, College of Medicine, Korea University, Seoul, South Korea.,Department of Biomedical Sciences, College of Medicine, Korea University, Seoul, South Korea
| | - Hyojin Kang
- Division of National Supercomputing, KISTI, Daejeon, South Korea
| | - Chunmei Jin
- Department of Neuroscience, College of Medicine, Korea University, Seoul, South Korea.,Department of Biomedical Sciences, College of Medicine, Korea University, Seoul, South Korea
| | - Yinhua Zhang
- Department of Neuroscience, College of Medicine, Korea University, Seoul, South Korea.,Department of Biomedical Sciences, College of Medicine, Korea University, Seoul, South Korea
| | - Yoonhee Kim
- Department of Neuroscience, College of Medicine, Korea University, Seoul, South Korea.,Department of Biomedical Sciences, College of Medicine, Korea University, Seoul, South Korea
| | - Kihoon Han
- Department of Neuroscience, College of Medicine, Korea University, Seoul, South Korea.,Department of Biomedical Sciences, College of Medicine, Korea University, Seoul, South Korea
| |
Collapse
|
17
|
Jin C, Zhang Y, Kim S, Kim Y, Lee Y, Han K. Spontaneous seizure and partial lethality of juvenile Shank3-overexpressing mice in C57BL/6 J background. Mol Brain 2018; 11:57. [PMID: 30305163 PMCID: PMC6180595 DOI: 10.1186/s13041-018-0403-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Accepted: 10/02/2018] [Indexed: 11/28/2022] Open
Abstract
The SH3 and multiple ankyrin repeat domains 3 (SHANK3) gene encodes core scaffolds in neuronal excitatory postsynapses. SHANK3 duplications have been identified in patients with hyperkinetic disorders and early-onset generalized tonic-clonic seizures. Consistently, Shank3 transgenic (TG) mice, which mildly overexpress Shank3 proteins exhibit hyperkinetic behavior and spontaneous seizures. However, the seizure phenotype of Shank3 TG mice has only been investigated in adults of the seizure-sensitive strain FVB/N. Therefore, it remains unknown if spontaneous seizures occur in Shank3 TG mice from the early postnatal stages onward, or even in seizure-resistant strains. Clinically, generalized tonic-clonic seizures are the critical risk factor for epilepsy-associated mortality. However, the potential association between Shank3 overexpression and mortality, at least in mice, has not been investigated in detail. In the present study, we backcrossed Shank3 TG mice in seizure-resistant C57BL/6 J strain and monitored their home-cage activities at 3 weeks of age. Of the 15 Shank3 TG mice monitored, two exhibited spontaneous tonic-clonic seizures, and one died immediately after the seizure event. Based on this observation, we determined the survival rate of the Shank3 TG mice from 3 to 12 weeks of age. We found that approximately 40–45% of the Shank3 TG mice, both males and females, died before reaching 12 weeks of age. Notably, 53% and 70% of the total deaths in male and female Shank3 TG mice, respectively, occurred in the juvenile stages. These results suggest spontaneous seizure and partial lethality of juvenile Shank3 TG mice in seizure-resistant background, further supporting the validity of this model.
Collapse
Affiliation(s)
- Chunmei Jin
- Department of Neuroscience, College of Medicine, Korea University, 73, Inchon-ro, Seongbuk-gu, Seoul, 02841, South Korea.,Department of Biomedical Sciences, College of Medicine, Korea University, Seoul, 02841, South Korea
| | - Yinhua Zhang
- Department of Neuroscience, College of Medicine, Korea University, 73, Inchon-ro, Seongbuk-gu, Seoul, 02841, South Korea.,Department of Biomedical Sciences, College of Medicine, Korea University, Seoul, 02841, South Korea
| | - Shinhyun Kim
- Department of Neuroscience, College of Medicine, Korea University, 73, Inchon-ro, Seongbuk-gu, Seoul, 02841, South Korea.,Department of Biomedical Sciences, College of Medicine, Korea University, Seoul, 02841, South Korea
| | - Yoonhee Kim
- Department of Neuroscience, College of Medicine, Korea University, 73, Inchon-ro, Seongbuk-gu, Seoul, 02841, South Korea.,Department of Biomedical Sciences, College of Medicine, Korea University, Seoul, 02841, South Korea
| | - Yeunkum Lee
- Department of Neuroscience, College of Medicine, Korea University, 73, Inchon-ro, Seongbuk-gu, Seoul, 02841, South Korea.,Department of Biomedical Sciences, College of Medicine, Korea University, Seoul, 02841, South Korea
| | - Kihoon Han
- Department of Neuroscience, College of Medicine, Korea University, 73, Inchon-ro, Seongbuk-gu, Seoul, 02841, South Korea. .,Department of Biomedical Sciences, College of Medicine, Korea University, Seoul, 02841, South Korea.
| |
Collapse
|
18
|
Jin C, Kang H, Ryu JR, Kim S, Zhang Y, Lee Y, Kim Y, Han K. Integrative Brain Transcriptome Analysis Reveals Region-Specific and Broad Molecular Changes in Shank3-Overexpressing Mice. Front Mol Neurosci 2018; 11:250. [PMID: 30233305 PMCID: PMC6127286 DOI: 10.3389/fnmol.2018.00250] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Accepted: 07/02/2018] [Indexed: 01/04/2023] Open
Abstract
Variants of the SH3 and multiple ankyrin repeat domain 3 (SHANK3) gene, encoding excitatory postsynaptic core scaffolding proteins, are causally associated with numerous neurodevelopmental and neuropsychiatric disorders, including autism spectrum disorder (ASD), bipolar disorder, intellectual disability, and schizophrenia (SCZ). Although detailed synaptic changes of various Shank3 mutant mice have been well characterized, broader downstream molecular changes, including direct and indirect changes, remain largely unknown. To address this issue, we performed a transcriptome analysis of the medial prefrontal cortex (mPFC) of adult Shank3-overexpressing transgenic (TG) mice, using an RNA-sequencing approach. We also re-analyzed previously reported RNA-sequencing results of the striatum of adult Shank3 TG mice and of the prefrontal cortex of juvenile Shank3+/ΔC mice with a 50–70% reduction of Shank3 proteins. We found that several myelin-related genes were significantly downregulated specifically in the mPFC, but not in the striatum or hippocampus, of adult Shank3 TG mice by comparing the differentially expressed genes (DEGs) of the analyses side by side. Moreover, we also found nine common DEGs between the mPFC and striatum of Shank3 TG mice, among which we further characterized ASD- and SCZ-associated G protein-coupled receptor 85 (Gpr85), encoding an orphan Gpr interacting with PSD-95. Unlike the mPFC-specific decrease of myelin-related genes, we found that the mRNA levels of Gpr85 increased in multiple brain regions of adult Shank3 TG mice, whereas the mRNA levels of its family members, Gpr27 and Gpr173, decreased in the cortex and striatum. Intriguingly, in cultured neurons, the mRNA levels of Gpr27, Gpr85, and Gpr173 were modulated by the neuronal activity. Furthermore, exogenously expressed GPR85 was co-localized with PSD-95 and Shank3 in cultured neurons and negatively regulated the number of excitatory synapses, suggesting its potential role in homeostatic regulation of excitatory synapses in Shank3 TG neurons. Finally, we performed a gene set enrichment analysis of the RNA-sequencing results, which suggested that Shank3 could affect the directional expression pattern of numerous ribosome-related genes in a dosage-dependent manner. To sum up, these results reveal previously unidentified brain region-specific and broad molecular changes in Shank3-overexpressing mice, further elucidating the complexity of the molecular pathophysiology of SHANK3-associated brain disorders.
Collapse
Affiliation(s)
- Chunmei Jin
- Department of Neuroscience, College of Medicine, Korea University, Seoul, South Korea.,Department of Biomedical Sciences, College of Medicine, Korea University, Seoul, South Korea
| | - Hyojin Kang
- Supercomputing Center, Korea Institute of Science and Technology Information, Daejeon, South Korea
| | - Jae Ryun Ryu
- Department of Biomedical Sciences, College of Medicine, Korea University, Seoul, South Korea.,Department of Anatomy, College of Medicine, Korea University, Seoul, South Korea
| | - Shinhyun Kim
- Department of Neuroscience, College of Medicine, Korea University, Seoul, South Korea.,Department of Biomedical Sciences, College of Medicine, Korea University, Seoul, South Korea
| | - Yinhua Zhang
- Department of Neuroscience, College of Medicine, Korea University, Seoul, South Korea.,Department of Biomedical Sciences, College of Medicine, Korea University, Seoul, South Korea
| | - Yeunkum Lee
- Department of Neuroscience, College of Medicine, Korea University, Seoul, South Korea.,Department of Biomedical Sciences, College of Medicine, Korea University, Seoul, South Korea
| | - Yoonhee Kim
- Department of Neuroscience, College of Medicine, Korea University, Seoul, South Korea.,Department of Biomedical Sciences, College of Medicine, Korea University, Seoul, South Korea
| | - Kihoon Han
- Department of Neuroscience, College of Medicine, Korea University, Seoul, South Korea.,Department of Biomedical Sciences, College of Medicine, Korea University, Seoul, South Korea
| |
Collapse
|
19
|
Excitatory and inhibitory synaptic dysfunction in mania: an emerging hypothesis from animal model studies. Exp Mol Med 2018; 50:1-11. [PMID: 29628501 PMCID: PMC5938027 DOI: 10.1038/s12276-018-0028-y] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Accepted: 11/29/2017] [Indexed: 12/26/2022] Open
Abstract
Bipolar disorder (BD) is a common psychiatric disorder characterized by recurrent mood swings between depression and mania, and is associated with high treatment costs. The existence of manic episodes is the defining feature of BD, during which period, patients experience extreme elevation in activity, energy, and mood, with changes in sleep patterns that together severely impair their ability to function in daily life. Despite some limitations in recapitulating the complex features of human disease, several rodent models of mania have been generated and characterized, which have provided important insights toward understanding its underlying pathogenic mechanisms. Among the mechanisms, neuronal excitatory and inhibitory (E/I) synaptic dysfunction in some brain regions, including the frontal cortex, hippocampus, and striatum, is an emerging hypothesis explaining mania. In this review, we highlight recent studies of rodent manic models having impairments in the E/I synaptic development and function. We also summarize the molecular and functional changes of E/I synapses by some mood stabilizers that may contribute to the therapeutic efficacy of drugs. Furthermore, we discuss potential future directions in the study of this emerging hypothesis to better connect the outcomes of basic research to the treatment of patients with this devastating mental illness. Studies in rodents offer insights into bipolar disorder that may help understanding and treatment of this common and debilitating condition. Kihoon Han and colleagues at Korea University in Seoul review research using mice and rats to model the episodes of mania in patients with bipolar disorder. The research supports an emerging hypothesis implicating specific problems with nervous transmission in the brain in the onset of mania. The hypothesis suggests that the transmission of signals between particular nerve cells whose normal function is either to excite or to inhibit other nerve cells may be involved. It also indicates regions of the brain most involved in manic episodes. Changes at the affected nerve junctions—called synapses—brought about by mood-stabilizing drugs are examined. The hypothesis suggests new approaches to treatment options for researchers to explore.
Collapse
|
20
|
Lee Y, Ryu JR, Kang H, Kim Y, Kim S, Zhang Y, Jin C, Cho HM, Kim WK, Sun W, Han K. Characterization of the zinc-induced Shank3 interactome of mouse synaptosome. Biochem Biophys Res Commun 2017; 494:581-586. [PMID: 29111324 DOI: 10.1016/j.bbrc.2017.10.143] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Accepted: 10/26/2017] [Indexed: 12/27/2022]
Abstract
Variants of the SHANK3 gene, which encodes a core scaffold protein of the postsynaptic density of excitatory synapses, have been causally associated with numerous brain disorders. Shank3 proteins directly bind zinc ions through their C-terminal sterile α motif domain, which enhances the multimerization and synaptic localization of Shank3, to regulate excitatory synaptic strength. However, no studies have explored whether zinc affects the protein interactions of Shank3, which might contribute to the synaptic changes observed after zinc application. To examine this, we first purified Shank3 protein complexes from mouse brain synaptosomal lysates that were incubated with different concentrations of ZnCl2, and analyzed them with mass spectrometry. We used strict criteria to identify 71 proteins that specifically interacted with Shank3 when extra ZnCl2 was added to the lysate. To characterize the zinc-induced Shank3 interactome, we performed various bioinformatic analyses that revealed significant associations of the interactome with subcellular compartments, including mitochondria, and brain disorders, such as bipolar disorder and schizophrenia. Together, our results showing that zinc affected the Shank3 protein interactions of in vitro mouse synaptosomes provided an additional link between zinc and core synaptic proteins that have been implicated in multiple brain disorders.
Collapse
Affiliation(s)
- Yeunkum Lee
- Department of Neuroscience, College of Medicine, Korea University, Seoul, South Korea; Department of Biomedical Sciences, College of Medicine, Korea University, Seoul, South Korea
| | - Jae Ryun Ryu
- Department of Biomedical Sciences, College of Medicine, Korea University, Seoul, South Korea; Department of Anatomy, College of Medicine, Korea University, Seoul, South Korea
| | - Hyojin Kang
- HPC-enabled Convergence Technology Research Division, Korea Institute of Science and Technology Information, Daejeon, South Korea
| | - Yoonhee Kim
- Department of Neuroscience, College of Medicine, Korea University, Seoul, South Korea; Department of Biomedical Sciences, College of Medicine, Korea University, Seoul, South Korea
| | - Shinhyun Kim
- Department of Neuroscience, College of Medicine, Korea University, Seoul, South Korea; Department of Biomedical Sciences, College of Medicine, Korea University, Seoul, South Korea
| | - Yinhua Zhang
- Department of Neuroscience, College of Medicine, Korea University, Seoul, South Korea; Department of Biomedical Sciences, College of Medicine, Korea University, Seoul, South Korea
| | - Chunmei Jin
- Department of Neuroscience, College of Medicine, Korea University, Seoul, South Korea; Department of Biomedical Sciences, College of Medicine, Korea University, Seoul, South Korea
| | - Hyo Min Cho
- Department of Biomedical Sciences, College of Medicine, Korea University, Seoul, South Korea; Department of Anatomy, College of Medicine, Korea University, Seoul, South Korea
| | - Won-Ki Kim
- Department of Neuroscience, College of Medicine, Korea University, Seoul, South Korea; Department of Biomedical Sciences, College of Medicine, Korea University, Seoul, South Korea
| | - Woong Sun
- Department of Biomedical Sciences, College of Medicine, Korea University, Seoul, South Korea; Department of Anatomy, College of Medicine, Korea University, Seoul, South Korea
| | - Kihoon Han
- Department of Neuroscience, College of Medicine, Korea University, Seoul, South Korea; Department of Biomedical Sciences, College of Medicine, Korea University, Seoul, South Korea.
| |
Collapse
|
21
|
Lee Y, Kim SG, Lee B, Zhang Y, Kim Y, Kim S, Kim E, Kang H, Han K. Striatal Transcriptome and Interactome Analysis of Shank3-overexpressing Mice Reveals the Connectivity between Shank3 and mTORC1 Signaling. Front Mol Neurosci 2017; 10:201. [PMID: 28701918 PMCID: PMC5487420 DOI: 10.3389/fnmol.2017.00201] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Accepted: 06/08/2017] [Indexed: 11/13/2022] Open
Abstract
Mania causes symptoms of hyperactivity, impulsivity, elevated mood, reduced anxiety and decreased need for sleep, which suggests that the dysfunction of the striatum, a critical component of the brain motor and reward system, can be causally associated with mania. However, detailed molecular pathophysiology underlying the striatal dysfunction in mania remains largely unknown. In this study, we aimed to identify the molecular pathways showing alterations in the striatum of SH3 and multiple ankyrin repeat domains 3 (Shank3)-overexpressing transgenic (TG) mice that display manic-like behaviors. The results of transcriptome analysis suggested that mammalian target of rapamycin complex 1 (mTORC1) signaling may be the primary molecular signature altered in the Shank3 TG striatum. Indeed, we found that striatal mTORC1 activity, as measured by mTOR S2448 phosphorylation, was significantly decreased in the Shank3 TG mice compared to wild-type (WT) mice. To elucidate the potential underlying mechanism, we re-analyzed previously reported protein interactomes, and detected a high connectivity between Shank3 and several upstream regulators of mTORC1, such as tuberous sclerosis 1 (TSC1), TSC2 and Ras homolog enriched in striatum (Rhes), via 94 common interactors that we denominated “Shank3-mTORC1 interactome”. We noticed that, among the 94 common interactors, 11 proteins were related to actin filaments, the level of which was increased in the dorsal striatum of Shank3 TG mice. Furthermore, we could co-immunoprecipitate Shank3, Rhes and Wiskott-Aldrich syndrome protein family verprolin-homologous protein 1 (WAVE1) proteins from the striatal lysate of Shank3 TG mice. By comparing with the gene sets of psychiatric disorders, we also observed that the 94 proteins of Shank3-mTORC1 interactome were significantly associated with bipolar disorder (BD). Altogether, our results suggest a protein interaction-mediated connectivity between Shank3 and certain upstream regulators of mTORC1 that might contribute to the abnormal striatal mTORC1 activity and to the manic-like behaviors of Shank3 TG mice.
Collapse
Affiliation(s)
- Yeunkum Lee
- Department of Neuroscience, College of Medicine, Korea UniversitySeoul, South Korea.,Department of Biomedical Sciences, College of Medicine, Korea UniversitySeoul, South Korea
| | - Sun Gyun Kim
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science (IBS)Daejeon, South Korea
| | - Bokyoung Lee
- Department of Neuroscience, College of Medicine, Korea UniversitySeoul, South Korea
| | - Yinhua Zhang
- Department of Neuroscience, College of Medicine, Korea UniversitySeoul, South Korea.,Department of Biomedical Sciences, College of Medicine, Korea UniversitySeoul, South Korea
| | - Yoonhee Kim
- Department of Neuroscience, College of Medicine, Korea UniversitySeoul, South Korea
| | - Shinhyun Kim
- Department of Neuroscience, College of Medicine, Korea UniversitySeoul, South Korea.,Department of Biomedical Sciences, College of Medicine, Korea UniversitySeoul, South Korea
| | - Eunjoon Kim
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science (IBS)Daejeon, South Korea.,Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST)Daejeon, South Korea
| | - Hyojin Kang
- HPC-enabled Convergence Technology Research Division, Korea Institute of Science and Technology InformationDaejeon, South Korea
| | - Kihoon Han
- Department of Neuroscience, College of Medicine, Korea UniversitySeoul, South Korea.,Department of Biomedical Sciences, College of Medicine, Korea UniversitySeoul, South Korea
| |
Collapse
|