101
|
Taylor SE, Taylor RD, Price J, Andreae LC. Single-molecule fluorescence in-situ hybridization reveals that human SHANK3 mRNA expression varies during development and in autism-associated SHANK3 heterozygosity. Stem Cell Res Ther 2018; 9:206. [PMID: 30064494 PMCID: PMC6069870 DOI: 10.1186/s13287-018-0957-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Revised: 06/13/2018] [Accepted: 07/16/2018] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Deletions and mutations in the SHANK3 gene are strongly associated with autism spectrum disorder and underlie the autism-associated disorder Phelan-McDermid syndrome. SHANK3 is a scaffolding protein found at the post-synaptic membrane of excitatory neurons. METHODS Single-molecule fluorescence in-situ hybridization (smFISH) allows the visualization of single mRNA transcripts in vitro. Here we perform and quantify smFISH in human inducible pluripotent stem cell (hiPSC)-derived cortical neurons, targeting the SHANK3 transcript. RESULTS Both smFISH and conventional immunofluorescence staining demonstrated a developmental increase in SHANK3 mRNA and protein, respectively, in control human cortical neurons. Analysis of single SHANK3 mRNA molecules in neurons derived from an autistic individual heterozygous for SHANK3 indicated that while the number of SHANK3 mRNA transcripts remained comparable with control levels in the cell soma, there was a 50% reduction within neuronal processes, suggesting that local, dendritic targeting of SHANK3 mRNA may be specifically affected in SHANK3 haploinsufficiency. CONCLUSION Human SHANK3 mRNA shows developmentally regulated dendritic localization in hiPSC-derived neurons, which is reduced in neurons generated from a haploinsufficient individual with autism. Although further replication is needed, given the importance of local mRNA translation in synaptic function, this could represent an important early abnormality.
Collapse
Affiliation(s)
- Samuel E Taylor
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, SE1 1UL, UK.,MRC Centre for Neurodevelopmental Disorders, King's College London, London, UK
| | - Ruth D Taylor
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, SE1 1UL, UK.,MRC Centre for Neurodevelopmental Disorders, King's College London, London, UK
| | - Jack Price
- MRC Centre for Neurodevelopmental Disorders, King's College London, London, UK.,Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, SE5 8AF, UK
| | - Laura C Andreae
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, SE1 1UL, UK. .,MRC Centre for Neurodevelopmental Disorders, King's College London, London, UK.
| |
Collapse
|
102
|
Collins SM, Galvez R. Neocortical SHANK1 regulation of forebrain dependent associative learning. Neurobiol Learn Mem 2018; 155:173-179. [PMID: 30053575 DOI: 10.1016/j.nlm.2018.07.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Revised: 07/20/2018] [Accepted: 07/23/2018] [Indexed: 11/18/2022]
Abstract
Learning-induced neocortical synaptic plasticity is a well-established mechanism mediating memory consolidation. Classic learning paradigms elicit synaptic changes in various brain regions including the neocortex. Work from our laboratory has further suggested synaptic remodeling in primary somatosensory cortex (S1) during forebrain-dependent associative learning. While this process of synaptic remodeling is largely believed to contribute to memory consolidation, the underlying processes mediating this plasticity are poorly understood. Interestingly, abnormal expression of the synaptic scaffolding protein SHANK1 has been linked with aberrant synaptic plasticity and learning impairments, suggesting that it plays a critical role in these processes. However, a direct analysis of the role for SHANK1 during learning in the neocortex, the most likely site for memory storage, has never been adequately explored. To directly examine SHANK1's potential role during learning and memory, the following study set out to both examine neocortical SHANK1 expression during a learning event and determine the consequences of reducing neocortical SHANK1 expression on learning. The current study found that SHANK1 expression is transiently increased during periods of learning-induced dendritic spine plasticity in the neocortex. Furthermore, shRNA-mediated neocortical SHANK1 knockdown significantly impairs acquisition for the forebrain-dependent associative learning task (whisker-trace-eyeblink conditioning). Consistent with these findings, SHANK1 has been implicated in various neurological disorders. Collectively, these findings suggest a role for SHANK1 in neocortical learning-induced dendritic spine plasticity underlying learning and normal cognition; thus, providing potential insight into neurological mechanisms mediating abnormalities of impaired cognition.
Collapse
Affiliation(s)
- Sean M Collins
- Psychology Department, University of Illinois at Urbana-Champaign, 405 N Mathews Ave, Urbana, IL 61801, United States; Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, 405 N Mathews Ave, Urbana, IL 61801, United States
| | - Roberto Galvez
- Psychology Department, University of Illinois at Urbana-Champaign, 405 N Mathews Ave, Urbana, IL 61801, United States; Neuroscience Program, University of Illinois at Urbana-Champaign, 405 N Mathews Ave, Urbana, IL 61801, United States; Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, 405 N Mathews Ave, Urbana, IL 61801, United States; Carle Illinois College of Medicine, University of Illinois at Urbana-Champaign, 405 N Mathews Ave, Urbana, IL 61801, United States.
| |
Collapse
|
103
|
Kennedy MB. The Protein Biochemistry of the Postsynaptic Density in Glutamatergic Synapses Mediates Learning in Neural Networks. Biochemistry 2018; 57:4005-4009. [PMID: 29913061 PMCID: PMC7879948 DOI: 10.1021/acs.biochem.8b00496] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The strength of each excitatory synapse in the central nervous system is regulated by its prior activity in a process called synaptic plasticity. The initiation of synaptic plasticity occurs when calcium ions enter the postsynaptic compartment and encounter a subcellular structure called the postsynaptic density (PSD). The PSD is attached to the postsynaptic membrane just underneath the concentrated plaque of neurotransmitter receptors. It is comprised of a core set of 30-60 proteins, approximately 20 of which are scaffold proteins. The rest include protein kinases and phosphatases, some of which respond to calcium ion; small GTPases and their regulators; chaperones; ubiquitins; and proteases. The assembly of the PSD involves competitive binding among a variety of specific protein binding sites to form a dynamic network. A biochemical challenge for the future is to understand how the dynamic regulation of the structure, composition, and activity of the PSD mediates synaptic plasticity and how mutations in PSD proteins lead to mental and neurodegenerative diseases.
Collapse
Affiliation(s)
- Mary B. Kennedy
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California 91125, United States
| |
Collapse
|
104
|
Lu ZA, Mu W, Osborne LM, Cordner ZA. Eighteen-year-old man with autism, obsessive compulsive disorder and a SHANK2 variant presents with severe anorexia that responds to high-dose fluoxetine. BMJ Case Rep 2018; 2018:bcr-2018-225119. [PMID: 29991577 DOI: 10.1136/bcr-2018-225119] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
The SHANK2 gene codes for a protein involved in organising the postsynaptic density and disruptions have been associated with autism spectrum disorders (ASDs). ASDs are frequently comorbid with intellectual disability and anxiety disorders and emerging evidence suggests potentially common aetiologies. Here, we report the case of an 18-year-old man with ASD who presented with severe anorexia due to fear of food contamination, food avoidance and stereotypies attributable to underlying obsessive compulsive disorder (OCD). The patient was found to be heterozygous for c.2518C>T (p.Pro840Ser), a likely damaging coding variant in the proline rich region of SHANK2 Interestingly, the patient's disordered eating behaviour began to improve only after high-dose fluoxetine was initiated to target OCD symptoms. Overall, this case highlights the utility of molecular genetic testing in clinical psychiatry and provides an example of how genetic information can inform clinicians in the treatment of complex neuropsychiatric syndromes.
Collapse
Affiliation(s)
- Zhen A Lu
- Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Weiyi Mu
- Institute of Genetic Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Lauren M Osborne
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, Maryland, USA.,Department of Gynecology and Obstetrics, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Zachary A Cordner
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
105
|
Lee S, Lee E, Kim R, Kim J, Lee S, Park H, Yang E, Kim H, Kim E. Shank2 Deletion in Parvalbumin Neurons Leads to Moderate Hyperactivity, Enhanced Self-Grooming and Suppressed Seizure Susceptibility in Mice. Front Mol Neurosci 2018; 11:209. [PMID: 29970987 PMCID: PMC6018407 DOI: 10.3389/fnmol.2018.00209] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 05/28/2018] [Indexed: 01/05/2023] Open
Abstract
Shank2 is an abundant postsynaptic scaffolding protein implicated in neurodevelopmental and psychiatric disorders, including autism spectrum disorders (ASD). Deletion of Shank2 in mice has been shown to induce social deficits, repetitive behaviors, and hyperactivity, but the identity of the cell types that contribute to these phenotypes has remained unclear. Here, we report a conditional mouse line with a Shank2 deletion restricted to parvalbumin (PV)-positive neurons (Pv-Cre;Shank2fl/fl mice). These mice display moderate hyperactivity in both novel and familiar environments and enhanced self-grooming in novel, but not familiar, environments. In contrast, they showed normal levels of social interaction, anxiety-like behavior, and learning and memory. Basal brain rhythms in Pv-Cre;Shank2fl/fl mice, measured by electroencephalography, were normal, but susceptibility to pentylenetetrazole (PTZ)-induced seizures was decreased. These results suggest that Shank2 deletion in PV-positive neurons leads to hyperactivity, enhanced self-grooming and suppressed brain excitation.
Collapse
Affiliation(s)
- Seungjoon Lee
- Department of Biological Sciences, Korea Advanced Institute for Science and Technology (KAIST), Daejeon, South Korea
| | - Eunee Lee
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science (IBS), Daejeon, South Korea
| | - Ryunhee Kim
- Department of Biological Sciences, Korea Advanced Institute for Science and Technology (KAIST), Daejeon, South Korea
| | - Jihye Kim
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science (IBS), Daejeon, South Korea
| | - Suho Lee
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science (IBS), Daejeon, South Korea
| | - Haram Park
- Department of Biological Sciences, Korea Advanced Institute for Science and Technology (KAIST), Daejeon, South Korea
| | - Esther Yang
- Department of Anatomy, College of Medicine, Korea University, Seoul, South Korea
| | - Hyun Kim
- Department of Anatomy, College of Medicine, Korea University, Seoul, South Korea
| | - Eunjoon Kim
- Department of Biological Sciences, Korea Advanced Institute for Science and Technology (KAIST), Daejeon, South Korea.,Center for Synaptic Brain Dysfunctions, Institute for Basic Science (IBS), Daejeon, South Korea
| |
Collapse
|
106
|
Abstract
The formation of correct synaptic structures and neuronal connections is paramount for normal brain development and a functioning adult brain. The integrin family of cell adhesion receptors and their ligands play essential roles in the control of several processes regulating neuronal connectivity - including neurite outgrowth, the formation and maintenance of synapses, and synaptic plasticity - that are affected in neurodevelopmental disorders, such as autism spectrum disorders (ASDs) and schizophrenia. Many ASD- and schizophrenia-associated genes are linked to alterations in the genetic code of integrins and associated signalling pathways. In non-neuronal cells, crosstalk between integrin-mediated adhesions and the actin cytoskeleton, and the regulation of integrin activity (affinity for extracellular ligands) are widely studied in healthy and pathological settings. In contrast, the roles of integrin-linked pathways in the central nervous system remains less well defined. In this Review, we will provide an overview of the known pathways that are regulated by integrin-ECM interaction in developing neurons and in adult brain. We will also describe recent advances in the identification of mechanisms that regulate integrin activity in neurons, and highlight the interesting emerging links between integrins and neurodevelopment.
Collapse
Affiliation(s)
- Johanna Lilja
- Turku Centre for Biotechnology, University of Turku, FIN-20520 Turku, Finland
| | - Johanna Ivaska
- Turku Centre for Biotechnology, University of Turku, FIN-20520 Turku, Finland .,Department of Biochemistry, University of Turku, FIN-20500 Turku, Finland
| |
Collapse
|
107
|
Joensuu M, Lanoue V, Hotulainen P. Dendritic spine actin cytoskeleton in autism spectrum disorder. Prog Neuropsychopharmacol Biol Psychiatry 2018; 84:362-381. [PMID: 28870634 DOI: 10.1016/j.pnpbp.2017.08.023] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 08/21/2017] [Accepted: 08/30/2017] [Indexed: 01/01/2023]
Abstract
Dendritic spines are small actin-rich protrusions from neuronal dendrites that form the postsynaptic part of most excitatory synapses. Changes in the shape and size of dendritic spines correlate with the functional changes in excitatory synapses and are heavily dependent on the remodeling of the underlying actin cytoskeleton. Recent evidence implicates synapses at dendritic spines as important substrates of pathogenesis in neuropsychiatric disorders, including autism spectrum disorder (ASD). Although synaptic perturbations are not the only alterations relevant for these diseases, understanding the molecular underpinnings of the spine and synapse pathology may provide insight into their etiologies and could reveal new drug targets. In this review, we will discuss recent findings of defective actin regulation in dendritic spines associated with ASD.
Collapse
Affiliation(s)
- Merja Joensuu
- Minerva Foundation Institute for Medical Research, 00290 Helsinki, Finland; Clem Jones Centre for Ageing Dementia Research, The University of Queensland, Brisbane, Queensland 4072, Australia; Queensland Brain Institute, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Vanessa Lanoue
- Clem Jones Centre for Ageing Dementia Research, The University of Queensland, Brisbane, Queensland 4072, Australia; Queensland Brain Institute, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Pirta Hotulainen
- Minerva Foundation Institute for Medical Research, 00290 Helsinki, Finland.
| |
Collapse
|
108
|
Moretto E, Murru L, Martano G, Sassone J, Passafaro M. Glutamatergic synapses in neurodevelopmental disorders. Prog Neuropsychopharmacol Biol Psychiatry 2018; 84:328-342. [PMID: 28935587 DOI: 10.1016/j.pnpbp.2017.09.014] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Revised: 08/28/2017] [Accepted: 09/16/2017] [Indexed: 12/22/2022]
Abstract
Neurodevelopmental disorders (NDDs) are a group of diseases whose symptoms arise during childhood or adolescence and that impact several higher cognitive functions such as learning, sociability and mood. Accruing evidence suggests that a shared pathogenic mechanism underlying these diseases is the dysfunction of glutamatergic synapses. We summarize present knowledge on autism spectrum disorders (ASD), intellectual disability (ID), Down syndrome (DS), Rett syndrome (RS) and attention-deficit hyperactivity disorder (ADHD), highlighting the involvement of glutamatergic synapses and receptors in these disorders. The most commonly shared defects involve α-amino-3-hydroxy-5-methyl- 4-isoxazole propionic acid receptors (AMPARs), N-methyl-d-aspartate receptors (NMDARs) and metabotropic glutamate receptors (mGluRs), whose functions are strongly linked to synaptic plasticity, affecting both cell-autonomous features as well as circuit formation. Moreover, the major scaffolding proteins and, thus, the general structure of the synapse are often deregulated in neurodevelopmental disorders, which is not surprising considering their crucial role in the regulation of glutamate receptor positioning and functioning. This convergence of defects supports the definition of neurodevelopmental disorders as a continuum of pathological manifestations, suggesting that glutamatergic synapses could be a therapeutic target to ameliorate patient symptomatology.
Collapse
Affiliation(s)
- Edoardo Moretto
- CNR, Institute of Neuroscience, Via Vanvitelli 32, 20129 Milan, Italy
| | - Luca Murru
- CNR, Institute of Neuroscience, Via Vanvitelli 32, 20129 Milan, Italy
| | - Giuseppe Martano
- CNR, Institute of Neuroscience, Via Vanvitelli 32, 20129 Milan, Italy
| | - Jenny Sassone
- San Raffaele Scientific Institute, Vita-Salute University, Milan, Italy
| | - Maria Passafaro
- CNR, Institute of Neuroscience, Via Vanvitelli 32, 20129 Milan, Italy.
| |
Collapse
|
109
|
Mossa A, Giona F, Pagano J, Sala C, Verpelli C. SHANK genes in autism: Defining therapeutic targets. Prog Neuropsychopharmacol Biol Psychiatry 2018; 84:416-423. [PMID: 29175319 DOI: 10.1016/j.pnpbp.2017.11.019] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Revised: 11/14/2017] [Accepted: 11/18/2017] [Indexed: 01/16/2023]
Affiliation(s)
- Adele Mossa
- CNR Neuroscience Institute, Department of Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Federica Giona
- CNR Neuroscience Institute, Department of Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Jessica Pagano
- CNR Neuroscience Institute, Department of Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Carlo Sala
- CNR Neuroscience Institute, Department of Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Chiara Verpelli
- CNR Neuroscience Institute, Department of Biotechnology and Translational Medicine, University of Milan, Milan, Italy.
| |
Collapse
|
110
|
Ali Rodriguez R, Joya C, Hines RM. Common Ribs of Inhibitory Synaptic Dysfunction in the Umbrella of Neurodevelopmental Disorders. Front Mol Neurosci 2018; 11:132. [PMID: 29740280 PMCID: PMC5928253 DOI: 10.3389/fnmol.2018.00132] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Accepted: 04/03/2018] [Indexed: 01/06/2023] Open
Abstract
The term neurodevelopmental disorder (NDD) is an umbrella term used to group together a heterogeneous class of disorders characterized by disruption in cognition, emotion, and behavior, early in the developmental timescale. These disorders are heterogeneous, yet they share common behavioral symptomatology as well as overlapping genetic contributors, including proteins involved in the formation, specialization, and function of synaptic connections. Advances may arise from bridging the current knowledge on synapse related factors indicated from both human studies in NDD populations, and in animal models. Mounting evidence has shown a link to inhibitory synapse formation, specialization, and function among Autism, Angelman, Rett and Dravet syndromes. Inhibitory signaling is diverse, with numerous subtypes of inhibitory interneurons, phasic and tonic modes of inhibition, and the molecular and subcellular diversity of GABAA receptors. We discuss common ribs of inhibitory synapse dysfunction in the umbrella of NDD, highlighting alterations in the developmental switch to inhibitory GABA, dysregulation of neuronal activity patterns by parvalbumin-positive interneurons, and impaired tonic inhibition. Increasing our basic understanding of inhibitory synapses, and their role in NDDs is likely to produce significant therapeutic advances in behavioral symptom alleviation for interrelated NDDs.
Collapse
Affiliation(s)
- Rachel Ali Rodriguez
- Neuroscience Emphasis, Department of Psychology, University of Nevada, Las Vegas, Las Vegas, NV, United States
| | - Christina Joya
- Neuroscience Emphasis, Department of Psychology, University of Nevada, Las Vegas, Las Vegas, NV, United States
| | - Rochelle M Hines
- Neuroscience Emphasis, Department of Psychology, University of Nevada, Las Vegas, Las Vegas, NV, United States
| |
Collapse
|
111
|
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: 30] [Impact Index Per Article: 4.3] [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
|
112
|
Zhu M, Idikuda VK, Wang J, Wei F, Kumar V, Shah N, Waite CB, Liu Q, Zhou L. Shank3-deficient thalamocortical neurons show HCN channelopathy and alterations in intrinsic electrical properties. J Physiol 2018; 596:1259-1276. [PMID: 29327340 DOI: 10.1113/jp275147] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2017] [Accepted: 01/04/2018] [Indexed: 01/11/2023] Open
Abstract
KEY POINTS Shank3 increases the HCN channel surface expression in heterologous expression systems. Shank3Δ13-16 deficiency causes significant reduction in HCN2 expression and Ih current amplitude in thalamocortical (TC) neurons. Shank3Δ13-16 - but not Shank3Δ4-9 -deficient TC neurons share changes in basic electrical properties which are comparable to those of HCN2-/- TC neurons. HCN channelopathy may critically mediate events downstream from Shank3 deficiency. ABSTRACT SHANK3 is a scaffolding protein that is highly enriched in excitatory synapses. Mutations in the SHANK3 gene have been linked to neuropsychiatric disorders especially the autism spectrum disorders. SHANK3 deficiency is known to cause impairments in synaptic transmission, but its effects on basic neuronal electrical properties that are more localized to the soma and proximal dendrites remain unclear. Here we confirmed that in heterologous expression systems two different mouse Shank3 isoforms, Shank3A and Shank3C, significantly increase the surface expression of the mouse hyperpolarization-activated, cyclic-nucleotide-gated (HCN) channel. In Shank3Δ13-16 knockout mice, which lack exons 13-16 in the Shank3 gene (both Shank3A and Shank3C are removed) and display a severe behavioural phenotype, the expression of HCN2 is reduced to an undetectable level. The thalamocortical (TC) neurons from the ventrobasal (VB) complex of Shank3Δ13-16 mice demonstrate reduced Ih current amplitude and correspondingly increased input resistance, negatively shifted resting membrane potential, and abnormal spike firing in both tonic and burst modes. Impressively, these changes closely resemble those of HCN2-/- TC neurons but not of the TC neurons from Shank3Δ4-9 mice, which lack exons 4-9 in the Shank3 gene (Shank3C still exists) and demonstrate moderate behavioural phenotypes. Additionally, Shank3 deficiency increases the ratio of excitatory/inhibitory balance in VB neurons but has a limited impact on the electrical properties of connected thalamic reticular (RTN) neurons. These results provide new understanding about the role of HCN channelopathy in mediating detrimental effects downstream from Shank3 deficiency.
Collapse
Affiliation(s)
- Mengye Zhu
- Department of Physiology and Biophysics, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA.,Department of Pain Clinic, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Vinay Kumar Idikuda
- Department of Physiology and Biophysics, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Jianbing Wang
- Department of Physiology and Biophysics, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA.,Department of Anesthesiology, Jiangxi Cancer Hospital, Nanchang, Jiangxi, China
| | - Fusheng Wei
- Department of Physiology and Biophysics, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA.,Department of Anesthesiology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Virang Kumar
- Department of Physiology and Biophysics, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Nikhil Shah
- Department of Physiology and Biophysics, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Christopher B Waite
- Department of Physiology and Biophysics, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Qinglian Liu
- Department of Physiology and Biophysics, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Lei Zhou
- Department of Physiology and Biophysics, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA
| |
Collapse
|
113
|
Zhu W, Li J, Chen S, Zhang J, Vetrini F, Braxton A, Eng CM, Yang Y, Xia F, Keller KL, Okinaka-Hu L, Lee C, Holder JL, Bi W. Two de novo novel mutations in one SHANK3 allele in a patient with autism and moderate intellectual disability. Am J Med Genet A 2018; 176:973-979. [PMID: 29423971 DOI: 10.1002/ajmg.a.38622] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 11/30/2017] [Accepted: 01/06/2018] [Indexed: 11/10/2022]
Abstract
SHANK3 encodes for a scaffolding protein that links neurotransmitter receptors to the cytoskeleton and is enriched in postsynaptic densities of excitatory synapses. Deletions or mutations in one copy of the SHANK3 gene cause Phelan-McDermid syndrome, also called 22q13.3 deletion syndrome, a neurodevelopmental disorder with common features including global developmental delay, absent to severely impaired language, autistic behavior, and minor dysmorphic features. By whole exome sequencing, we identified two de novo novel variants including one frameshift pathogenic variant and one missense variant of unknown significance in a 14-year-old boy with delayed motor milestones, delayed language acquisition, autism, intellectual disability, ataxia, progressively worsening spasticity of the lower extremities, dysmorphic features, short stature, microcephaly, failure to thrive, chronic constipation, intrauterine growth restriction, and bilateral inguinal hernias. Both changes are within the CpG island in exon 21, separated by a 375 bp sequence. Next generation sequencing of PCR products revealed that the two variants are most frequently associated with each other. Sanger sequencing of the cloned PCR products further confirmed that both changes were on a single allele. The clinical presentation in this individual is consistent with other patients with a truncating mutation in exon 21, suggesting that the missense change contributes none or minimally to the phenotypes. This is the first report of two de novo mutations in one SHANK3 allele.
Collapse
Affiliation(s)
- Wenmiao Zhu
- Baylor Genetics Laboratories, Houston, Texas
| | - Jianli Li
- Baylor Genetics Laboratories, Houston, Texas
| | - Stella Chen
- Baylor Genetics Laboratories, Houston, Texas
| | - Jinglan Zhang
- Baylor Genetics Laboratories, Houston, Texas.,Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas
| | | | | | - Christine M Eng
- Baylor Genetics Laboratories, Houston, Texas.,Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas
| | - Yaping Yang
- Baylor Genetics Laboratories, Houston, Texas.,Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas
| | - Fan Xia
- Baylor Genetics Laboratories, Houston, Texas.,Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas
| | - Kory L Keller
- Department of Molecular and Medical Genetics, Oregon Health & Science University, Portland, Oregon
| | - Leila Okinaka-Hu
- Department of Molecular and Medical Genetics, Oregon Health & Science University, Portland, Oregon
| | - Chung Lee
- Division of Medical Genetics, Department of Pediatrics, Stanford University, Stanford, California
| | - J Lloyd Holder
- Departments of Pediatrics, Baylor College of Medicine, Houston, Texas.,Division of Neurology and Developmental Neuroscience, Baylor College of Medicine, Houston, Texas.,Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, Texas
| | - Weimin Bi
- Baylor Genetics Laboratories, Houston, Texas.,Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas
| |
Collapse
|
114
|
Zeng HL, Yu FL, Zhang Z, Yang Q, Jin S, He X, Chen X, Shen Y, Cheng L, Guo L, Xu F. Quantitative proteomics study of host response to virulent and attenuated pseudorabies virus infection in mouse brain. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2018; 1866:307-315. [DOI: 10.1016/j.bbapap.2017.11.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Revised: 11/16/2017] [Accepted: 11/21/2017] [Indexed: 12/22/2022]
|
115
|
Stephens KE, Chen Z, Sivanesan E, Raja SN, Linderoth B, Taverna SD, Guan Y. RNA-seq of spinal cord from nerve-injured rats after spinal cord stimulation. Mol Pain 2018; 14:1744806918817429. [PMID: 30451078 PMCID: PMC6293371 DOI: 10.1177/1744806918817429] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Revised: 10/28/2018] [Accepted: 11/12/2018] [Indexed: 12/12/2022] Open
Abstract
Spinal cord stimulation has become an important modality in pain treatment especially for neuropathic pain conditions refractory to pharmacotherapy. However, the molecular control of inhibitory and excitatory mechanisms observed after spinal cord stimulation are poorly understood. Here, we used RNA-seq to identify differences in the expression of genes and gene networks in spinal cord tissue from nerve-injured rats with and without repetitive conventional spinal cord stimulation treatment. Five weeks after chronic constrictive injury to the left sciatic nerve, male and female rats were randomized to receive repetitive spinal cord stimulation or no treatment. Rats receiving spinal cord stimulation underwent epidural placement of a miniature stimulating electrode and received seven sessions of spinal cord stimulation (50 Hz, 80% motor threshold, 0.2 ms, constant current bipolar stimulation, 120 min/session) over four consecutive days. Within 2 h after the last spinal cord stimulation treatment, the L4-L6 spinal segments ipsilateral to the side of nerve injury were harvested and used to generate libraries for RNA-seq. Our RNA-seq data suggest further increases of many existing upregulated immune responses in chronic constrictive injury rats after repetitive spinal cord stimulation, including transcription of cell surface receptors and activation of non-neuronal cells. We also demonstrate that repetitive spinal cord stimulation represses transcription of several key synaptic signaling genes that encode scaffold proteins in the post-synaptic density. Our transcriptional studies suggest a potential relationship between specific genes and the therapeutic effects observed in patients undergoing conventional spinal cord stimulation after nerve injury. Furthermore, our results may help identify new therapeutic targets for improving the efficacy of conventional spinal cord stimulation and other chronic pain treatments.
Collapse
Affiliation(s)
- Kimberly E Stephens
- Department of Pharmacology and Molecular Sciences,
School of Medicine, Johns Hopkins University, Baltimore, MA, USA
- Center for Epigenetics, School of Medicine, Johns
Hopkins University, Baltimore, MA, USA
- Department of Anesthesia and Critical Care Medicine,
School of Medicine, Johns Hopkins University, Baltimore, MA, USA
| | - Zhiyong Chen
- Department of Anesthesia and Critical Care Medicine,
School of Medicine, Johns Hopkins University, Baltimore, MA, USA
- Institute of Basic Medical Sciences, Department of
Human Anatomy, Histology and Embryology, Neuroscience Center, Chinese
Academy of Medical Sciences, School of Basic Medicine, Peking Union
Medical College, Beijing, China
| | - Eellan Sivanesan
- Department of Anesthesia and Critical Care Medicine,
School of Medicine, Johns Hopkins University, Baltimore, MA, USA
| | - Srinivasa N Raja
- Department of Anesthesia and Critical Care Medicine,
School of Medicine, Johns Hopkins University, Baltimore, MA, USA
| | - Bengt Linderoth
- Division of Functional Neurosurgery, Department of
Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Sean D Taverna
- Department of Pharmacology and Molecular Sciences,
School of Medicine, Johns Hopkins University, Baltimore, MA, USA
- Center for Epigenetics, School of Medicine, Johns
Hopkins University, Baltimore, MA, USA
| | - Yun Guan
- Department of Anesthesia and Critical Care Medicine,
School of Medicine, Johns Hopkins University, Baltimore, MA, USA
- School of Medicine, Department of Neurological
Surgery, Johns Hopkins University, Baltimore, MA, USA
| |
Collapse
|
116
|
Kathuria A, Nowosiad P, Jagasia R, Aigner S, Taylor RD, Andreae LC, Gatford NJF, Lucchesi W, Srivastava DP, Price J. Stem cell-derived neurons from autistic individuals with SHANK3 mutation show morphogenetic abnormalities during early development. Mol Psychiatry 2018; 23:735-746. [PMID: 28948968 PMCID: PMC5822449 DOI: 10.1038/mp.2017.185] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Revised: 07/04/2017] [Accepted: 07/19/2017] [Indexed: 01/02/2023]
Abstract
Shank3 is a structural protein found predominantly at the postsynaptic density. Mutations in the SHANK3 gene have been associated with risk for autism spectrum disorder (ASD). We generated induced pluripotent stem cells (iPSCs) from control individuals and from human donors with ASD carrying microdeletions of SHANK3. In addition, we used Zinc finger nucleases to generate isogenic SHANK3 knockout human embryonic stem (ES) cell lines. We differentiated pluripotent cells into either cortical or olfactory placodal neurons. We show that patient-derived placodal neurons make fewer synapses than control cells. Moreover, patient-derived cells display a developmental phenotype: young postmitotic neurons have smaller cell bodies, more extensively branched neurites, and reduced motility compared with controls. These phenotypes were mimicked by SHANK3-edited ES cells and rescued by transduction with a Shank3 expression construct. This developmental phenotype is not observed in the same iPSC lines differentiated into cortical neurons. Therefore, we suggest that SHANK3 has a critical role in neuronal morphogenesis in placodal neurons and that early defects are associated with ASD-associated mutations.
Collapse
Affiliation(s)
- A Kathuria
- Cells & Behavior Unit, Department of Basic and Clinical Neuroscience, Institute of Psychiatry Psychology & Neuroscience, King’s College London, London, UK
| | - P Nowosiad
- Cells & Behavior Unit, Department of Basic and Clinical Neuroscience, Institute of Psychiatry Psychology & Neuroscience, King’s College London, London, UK
| | - R Jagasia
- CNS Discovery/F-Hoffmann-La Roche Ltd, Basel, Switzerland
| | - S Aigner
- Department of Cellular and Molecular Medicine School of Medicine University of California, San Diego, CA, USA
| | - R D Taylor
- Developmental Neurobiology/New Hunt’s House Guy’s Campus, King’s College London, London, UK,MRC Centre for Neurodevelopmental Disorders, King's College London, London, UK
| | - L C Andreae
- Developmental Neurobiology/New Hunt’s House Guy’s Campus, King’s College London, London, UK,MRC Centre for Neurodevelopmental Disorders, King's College London, London, UK
| | - N J F Gatford
- Cells & Behavior Unit, Department of Basic and Clinical Neuroscience, Institute of Psychiatry Psychology & Neuroscience, King’s College London, London, UK
| | - W Lucchesi
- School of Biological Sciences, Royal Holloway, University of London, Egham, UK
| | - D P Srivastava
- Cells & Behavior Unit, Department of Basic and Clinical Neuroscience, Institute of Psychiatry Psychology & Neuroscience, King’s College London, London, UK,MRC Centre for Neurodevelopmental Disorders, King's College London, London, UK
| | - J Price
- Cells & Behavior Unit, Department of Basic and Clinical Neuroscience, Institute of Psychiatry Psychology & Neuroscience, King’s College London, London, UK,MRC Centre for Neurodevelopmental Disorders, King's College London, London, UK,Division of Advanced Therapies, National Institute for Biological Standards and Control, Hertfordshire, UK,Cells & Behavior Unit, Department of Basic and Clinical Neuroscience, Institute of Psychiatry Psychology & Neuroscience, King’s College London, London SE5 9RT, UK. E-mail:
| |
Collapse
|
117
|
Collins SM, Belagodu AP, Reed SL, Galvez R. SHANK1 is differentially expressed during development in CA1 hippocampal neurons and astrocytes. Dev Neurobiol 2017; 78:363-373. [PMID: 29218848 DOI: 10.1002/dneu.22564] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Revised: 10/19/2017] [Accepted: 11/30/2017] [Indexed: 01/25/2023]
Abstract
Recent studies have strongly suggested a role for the synaptic scaffolding protein SHANK1 in normal synaptic structure and signaling. Global SHANK1 knockout (SHANK1-/-) mice demonstrate reduced dendritic spine density, an immature dendritic spine phenotype and impairments in various cognitive tasks. SHANK1 overexpression is associated with increased dendritic spine size and impairments in fear conditioning. These studies suggest proper regulation of SHANK1 is crucial for appropriate synaptic structure and cognition. However, little is known regarding SHANK1's developmental expression in brain regions critical for learning. The current study quantified cell specific developmental expression of SHANK1 in the hippocampus, a brain region critically involved in various learning paradigms shown to be disrupted by SHANK1 dysregulation. Consistent with prior studies, SHANK1 was found to be strongly co-expressed with dendritic markers, with significant increased co-expression at postnatal day (P) 15, an age associated with increased synaptogenesis in the hippocampus. Interestingly, SHANK1 was also found to be expressed in astrocytes and microglia. To our knowledge, this is the first demonstration of glial SHANK1 localization; therefore, these findings were further examined via a glial purified primary cell culture fraction using magnetic cell sorting. This additional analysis further demonstrated that SHANK1 was expressed in glial cells, supporting our immunofluorescence co-expression findings. Developmentally, astroglial SHANK1 co-expression was found to be significantly elevated at P5 with a reduction into adulthood, while SHANK1 microglial co-expression did not significantly change across development. These data collectively implicate a more global role for SHANK1 in mediating normal cellular signaling in the brain. © 2017 Wiley Periodicals, Inc. Develop Neurobiol 78: 363-373, 2018.
Collapse
Affiliation(s)
- Sean M Collins
- Psychology Department, Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, 405 N Mathews Ave, Urbana, Illinois, 61801
| | - Amogh P Belagodu
- Neuroscience Program, Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, 405 N Mathews Ave, Urbana, Illinois, 61801
| | - Samantha L Reed
- Psychology Department, Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, 405 N Mathews Ave, Urbana, Illinois, 61801
| | - Roberto Galvez
- Psychology Department, Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, 405 N Mathews Ave, Urbana, Illinois, 61801.,Neuroscience Program, Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, 405 N Mathews Ave, Urbana, Illinois, 61801
| |
Collapse
|
118
|
Trifonova EA, Khlebodarova TM, Gruntenko NE. Molecular mechanisms of autism as a form of synaptic dysfunction. ACTA ACUST UNITED AC 2017. [DOI: 10.1134/s2079059717080020] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
119
|
Anderson GR, Maxeiner S, Sando R, Tsetsenis T, Malenka RC, Südhof TC. Postsynaptic adhesion GPCR latrophilin-2 mediates target recognition in entorhinal-hippocampal synapse assembly. J Cell Biol 2017; 216:3831-3846. [PMID: 28972101 PMCID: PMC5674891 DOI: 10.1083/jcb.201703042] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Revised: 07/24/2017] [Accepted: 08/17/2017] [Indexed: 12/15/2022] Open
Abstract
Synapse assembly likely requires postsynaptic target recognition by incoming presynaptic afferents. Using newly generated conditional knock-in and knockout mice, we show in this study that latrophilin-2 (Lphn2), a cell-adhesion G protein-coupled receptor and presumptive α-latrotoxin receptor, controls the numbers of a specific subset of synapses in CA1-region hippocampal neurons, suggesting that Lphn2 acts as a synaptic target-recognition molecule. In cultured hippocampal neurons, Lphn2 maintained synapse numbers via a postsynaptic instead of a presynaptic mechanism, which was surprising given its presumptive role as an α-latrotoxin receptor. In CA1-region neurons in vivo, Lphn2 was specifically targeted to dendritic spines in the stratum lacunosum-moleculare, which form synapses with presynaptic entorhinal cortex afferents. In this study, postsynaptic deletion of Lphn2 selectively decreased spine numbers and impaired synaptic inputs from entorhinal but not Schaffer-collateral afferents. Behaviorally, loss of Lphn2 from the CA1 region increased spatial memory retention but decreased learning of sequential spatial memory tasks. Thus, Lphn2 appears to control synapse numbers in the entorhinal cortex/CA1 region circuit by acting as a domain-specific postsynaptic target-recognition molecule.
Collapse
MESH Headings
- Animals
- Behavior, Animal
- CA1 Region, Hippocampal/metabolism
- CA1 Region, Hippocampal/pathology
- CA1 Region, Hippocampal/physiopathology
- Cells, Cultured
- Dendritic Spines/metabolism
- Dendritic Spines/pathology
- Entorhinal Cortex/metabolism
- Entorhinal Cortex/pathology
- Entorhinal Cortex/physiopathology
- Fear
- Genotype
- Maze Learning
- Memory
- Mice, Mutant Strains
- Motor Activity
- Neurons/metabolism
- Neurons/pathology
- Phenotype
- Presynaptic Terminals/metabolism
- Presynaptic Terminals/pathology
- Receptors, G-Protein-Coupled/genetics
- Receptors, G-Protein-Coupled/metabolism
- Receptors, Peptide/genetics
- Receptors, Peptide/metabolism
- Rotarod Performance Test
- Smell
- Synaptic Membranes/metabolism
- Synaptic Membranes/pathology
- Synaptic Potentials
- Time Factors
- Transfection
Collapse
Affiliation(s)
- Garret R Anderson
- Department of Molecular and Cellular Physiology, Howard Hughes Medical Institute, Stanford University Medical School, Stanford, CA
- Department of Psychiatry and Behavioral Science, Nancy Pritzker Laboratory, Stanford University Medical School, Stanford, CA
| | - Stephan Maxeiner
- Department of Molecular and Cellular Physiology, Howard Hughes Medical Institute, Stanford University Medical School, Stanford, CA
| | - Richard Sando
- Department of Molecular and Cellular Physiology, Howard Hughes Medical Institute, Stanford University Medical School, Stanford, CA
| | - Theodoros Tsetsenis
- Department of Molecular and Cellular Physiology, Howard Hughes Medical Institute, Stanford University Medical School, Stanford, CA
| | - Robert C Malenka
- Department of Psychiatry and Behavioral Science, Nancy Pritzker Laboratory, Stanford University Medical School, Stanford, CA
| | - Thomas C Südhof
- Department of Molecular and Cellular Physiology, Howard Hughes Medical Institute, Stanford University Medical School, Stanford, CA
| |
Collapse
|
120
|
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
|
121
|
Yoon SY, Kwon SG, Kim YH, Yeo JH, Ko HG, Roh DH, Kaang BK, Beitz AJ, Lee JH, Oh SB. A critical role of spinal Shank2 proteins in NMDA-induced pain hypersensitivity. Mol Pain 2017; 13:1744806916688902. [PMID: 28326932 PMCID: PMC5302174 DOI: 10.1177/1744806916688902] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Background Self-injurious behaviors (SIBs) are devastating traits in autism spectrum disorder (ASD). Although deficits in pain sensation might be one of the contributing factors underlying the development of SIBs, the mechanisms have yet to be addressed. Recently, the Shank2 synaptic protein has been considered to be a key component in ASD, and mutations of SHANK2 gene induce the dysfunction of N-methyl-D-aspartate (NMDA) receptors, suggesting a link between Shank2 and NMDA receptors in ASD. Given that spinal NMDA receptors play a pivotal role in pain hypersensitivity, we investigated the possible role of Shank2 in nociceptive hypersensitivity by examining changes in spontaneous pain following intrathecal NMDA injection in Shank2−/− (Shank2 knock-out, KO) mice. Results Intrathecal NMDA injection evoked spontaneous nociceptive behaviors. These NMDA-induced nociceptive responses were significantly reduced in Shank2 KO mice. We also observed a significant decrease of NMDA currents in the spinal dorsal horn of Shank2 KO mice. Subsequently, we examined whether mitogen-activated protein kinase or AKT signaling is involved in this reduced pain behavior in Shank2 KO mice because the NMDA receptor is closely related to these signaling molecules. Western blotting and immunohistochemistry revealed that spinally administered NMDA increased the expression of a phosphorylated form of extracellular signal-regulated kinase (p-ERK) which was significantly reduced in Shank2 KO mice. However, p38, JNK, or AKT were not changed by NMDA administration. The ERK inhibitor, PD98059, decreased NMDA-induced spontaneous pain behaviors in a dose-dependent manner in wild-type mice. Moreover, it was found that the NMDA-induced increase in p-ERK was primarily colocalized with Shank2 proteins in the spinal cord dorsal horn. Conclusion Shank2 protein is involved in spinal NMDA receptor-mediated pain, and mutations of Shank2 may suppress NMDA-ERK signaling in spinal pain transmission. This study provides new clues into the mechanisms underlying pain deficits associated with SIB and deserves further study in patients with ASD.
Collapse
Affiliation(s)
- Seo-Yeon Yoon
- 1 Department of Brain and Cognitive Sciences, College of Natural Sciences, Pain Cognitive Function Research Center, Dental Research Institute, Seoul National University, Seoul, Republic of Korea.,2 Department of Neurobiology and Physiology School of Dentistry, Seoul National University, Seoul, Republic of Korea
| | - Soon-Gu Kwon
- 3 Department of Oral Physiology and Research Center for Tooth and Periodontal Tissue Regeneration, School of Dentistry, Kyung Hee University, Seoul, Republic of Korea
| | - Yong Ho Kim
- 1 Department of Brain and Cognitive Sciences, College of Natural Sciences, Pain Cognitive Function Research Center, Dental Research Institute, Seoul National University, Seoul, Republic of Korea.,2 Department of Neurobiology and Physiology School of Dentistry, Seoul National University, Seoul, Republic of Korea.,4 Department of Physiology, College of Medicine, Gachon University, Incheon, Republic of Korea
| | - Ji-Hee Yeo
- 3 Department of Oral Physiology and Research Center for Tooth and Periodontal Tissue Regeneration, School of Dentistry, Kyung Hee University, Seoul, Republic of Korea
| | - Hyoung-Gon Ko
- 5 Department of Biological Sciences, College of Natural Sciences, Seoul National University, Seoul, Republic of Korea
| | - Dae-Hyun Roh
- 3 Department of Oral Physiology and Research Center for Tooth and Periodontal Tissue Regeneration, School of Dentistry, Kyung Hee University, Seoul, Republic of Korea
| | - Bong-Kiun Kaang
- 5 Department of Biological Sciences, College of Natural Sciences, Seoul National University, Seoul, Republic of Korea.,6 Center for Neuron and Disease, Frontier Institutes of Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Alvin J Beitz
- 7 Department of Veterinary and Biomedical Sciences, College of Veterinary Medicine, University of Minnesota, St Paul, MN, USA
| | - Jang-Hern Lee
- 8 Department of Veterinary Physiology, BK21 PLUS Program for Creative Veterinary Science Research, Seoul National University, Seoul, Republic of Korea.,9 Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Seog Bae Oh
- 1 Department of Brain and Cognitive Sciences, College of Natural Sciences, Pain Cognitive Function Research Center, Dental Research Institute, Seoul National University, Seoul, Republic of Korea.,2 Department of Neurobiology and Physiology School of Dentistry, Seoul National University, Seoul, Republic of Korea
| |
Collapse
|
122
|
Sungur AÖ, Schwarting RKW, Wöhr M. Behavioral phenotypes and neurobiological mechanisms in the Shank1 mouse model for autism spectrum disorder: A translational perspective. Behav Brain Res 2017; 352:46-61. [PMID: 28963042 DOI: 10.1016/j.bbr.2017.09.038] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2017] [Revised: 09/11/2017] [Accepted: 09/25/2017] [Indexed: 11/27/2022]
Abstract
Autism spectrum disorder (ASD) is a heterogeneous group of neurodevelopmental disorders, characterized by early-onset deficits in social behavior and communication across multiple contexts, together with restricted, repetitive patterns of behavior, interests, or activities. ASD is among the most heritable neuropsychiatric conditions with heritability estimates higher than 80%, and while available evidence points to a complex set of genetic factors, the SHANK (also known as ProSAP) gene family has emerged as one of the most promising candidates. Several genetic Shank mouse models for ASD were generated, including Shank1 knockout mice. Behavioral studies focusing on the Shank1 knockout mouse model for ASD included assays for detecting ASD-relevant behavioral phenotypes in the following domains: (I) social behavior, (II) communication, and (III) repetitive and stereotyped patterns of behavior. In addition, assays for detecting behavioral phenotypes with relevance to comorbidities in ASD were performed, including but not limited to (IV) cognitive functioning. Here, we summarize and discuss behavioral and neuronal findings obtained in the Shank1 knockout mouse model for ASD. We identify open research questions by comparing such findings with the symptoms present in humans diagnosed with ASD and carrying SHANK1 deletions. We conclude by discussing the implications of the behavioral and neuronal phenotypes displayed by the Shank1 knockout mouse model for the development of future pharmacological interventions in ASD.
Collapse
Affiliation(s)
- A Özge Sungur
- Behavioral Neuroscience, Experimental and Biological Psychology, Philipps-University of Marburg, Marburg, Germany
| | - Rainer K W Schwarting
- Behavioral Neuroscience, Experimental and Biological Psychology, Philipps-University of Marburg, Marburg, Germany
| | - Markus Wöhr
- Behavioral Neuroscience, Experimental and Biological Psychology, Philipps-University of Marburg, Marburg, Germany.
| |
Collapse
|
123
|
Rasmussen AH, Rasmussen HB, Silahtaroglu A. The DLGAP family: neuronal expression, function and role in brain disorders. Mol Brain 2017; 10:43. [PMID: 28870203 PMCID: PMC5583998 DOI: 10.1186/s13041-017-0324-9] [Citation(s) in RCA: 124] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Accepted: 08/24/2017] [Indexed: 11/10/2022] Open
Abstract
The neurotransmitter glutamate facilitates neuronal signalling at excitatory synapses. Glutamate is released from the presynaptic membrane into the synaptic cleft. Across the synaptic cleft glutamate binds to both ion channels and metabotropic glutamate receptors at the postsynapse, which expedite downstream signalling in the neuron. The postsynaptic density, a highly specialized matrix, which is attached to the postsynaptic membrane, controls this downstream signalling. The postsynaptic density also resets the synapse after each synaptic firing. It is composed of numerous proteins including a family of Discs large associated protein 1, 2, 3 and 4 (DLGAP1-4) that act as scaffold proteins in the postsynaptic density. They link the glutamate receptors in the postsynaptic membrane to other glutamate receptors, to signalling proteins and to components of the cytoskeleton. With the central localisation in the postsynapse, the DLGAP family seems to play a vital role in synaptic scaling by regulating the turnover of both ionotropic and metabotropic glutamate receptors in response to synaptic activity. DLGAP family has been directly linked to a variety of psychological and neurological disorders. In this review we focus on the direct and indirect role of DLGAP family on schizophrenia as well as other brain diseases.
Collapse
Affiliation(s)
- Andreas H Rasmussen
- Department of Cellular and Molecular Medicine, Faculty of Medical and Health Sciences, University of Copenhagen, DK-2200, Copenhagen, Denmark
| | - Hanne B Rasmussen
- Department of Biomedical Sciences, Faculty of Medical and Health Sciences, University of Copenhagen, DK-2200, Copenhagen, Denmark
| | - Asli Silahtaroglu
- Department of Cellular and Molecular Medicine, Faculty of Medical and Health Sciences, University of Copenhagen, DK-2200, Copenhagen, Denmark.
| |
Collapse
|
124
|
Zhang Y, Gao B, Xiong Y, Zheng F, Xu X, Yang Y, Hu Y, Wang X. Expression of SHANK3 in the Temporal Neocortex of Patients with Intractable Temporal Epilepsy and Epilepsy Rat Models. Cell Mol Neurobiol 2017; 37:857-867. [PMID: 27592227 PMCID: PMC11482059 DOI: 10.1007/s10571-016-0423-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2016] [Accepted: 08/27/2016] [Indexed: 12/16/2022]
Abstract
SH3 and multiple ankyrin (ANK) repeat domain 3 (SHANK3) is a synaptic scaffolding protein enriched in the postsynaptic density of excitatory synapses. SHANK3 plays an important role in the formation and maturation of excitatory synapses. In the brain, SHANK3 directly or indirectly interacts with various synaptic molecules including N-methyl-D-aspartate receptor, the metabotropic glutamate receptor (mGluR), and α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor. Previous studies have shown that Autism spectrum disorder is a result of mutations of the main SHANK3 isoforms, which may be due to deficit in excitatory synaptic transmission and plasticity. Recently, accumulating evidence has demonstrated that overexpression of SHANK3 could induce seizures in vivo. However, little is known about the role of SHANK3 in refractory temporal lobe epilepsy (TLE). Therefore, we investigated the expression pattern of SHANK3 in patients with intractable temporal lobe epilepsy and in pilocarpine-induced models of epilepsy. Immunofluorescence, immunohistochemistry, and western blot analysis were used to locate and determine the expression of SHANK3 in the temporal neocortex of patients with epilepsy, and in the hippocampus and temporal lobe cortex of rats in a pilocarpine-induced epilepsy model. Double-labeled immunofluorescence showed that SHANK3 was mainly expressed in neurons. Western blot analysis confirmed that SHANK3 expression was increased in the neocortex of TLE patients and rats. These results indicate that SHANK3 participates in the pathology of epilepsy.
Collapse
Affiliation(s)
- Yanke Zhang
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Baobing Gao
- Department of Neurology, Chongqing General Hospital, Chongqing, China
| | - Yan Xiong
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Fangshuo Zheng
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Xin Xu
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Yong Yang
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Yida Hu
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Xuefeng Wang
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
- Center of Epilepsy, Beijing Institute for Brain Disorders, Beijing, 100871, China.
- Chongqing Key Laboratory of Neurology, 1 You Yi Road, Chongqing, 400016, China.
| |
Collapse
|
125
|
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
|
126
|
Ponna SK, Myllykoski M, Boeckers TM, Kursula P. Structure of an unconventional SH3 domain from the postsynaptic density protein Shank3 at ultrahigh resolution. Biochem Biophys Res Commun 2017. [PMID: 28647360 DOI: 10.1016/j.bbrc.2017.06.121] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
The Shank family comprises three large multi-domain proteins playing central roles as protein scaffolds in the neuronal postsynaptic density. The Shank proteins are closely linked to neuropsychiatric diseases, such as autism spectrum disorders. One characteristic domain in the Shank family is the SH3 domain, assumed to play a role in protein-protein interactions; however, no specific ligand binding to any Shank SH3 domain has been described. We solved the crystal structure of the SH3 domain from Shank3 at sub-atomic resolution. While the structure presents the canonical SH3 domain fold, the binding site for proline-rich peptides is not conserved. In line with this, no binding of Pro-rich sequences by the Shank3 SH3 domain was observed. Sequence comparisons indicate that all Shank isoforms have similarly lost the classical Pro-rich peptide binding site from the SH3 domain. Whether the corresponding site in the Shank SH3 domains has evolved to bind a non-poly-Pro target sequence is currently not known. Our work provides an intriguing example of the evolution of a well-characterized protein-protein interaction domain within the context of multi-domain protein scaffolds, allowing the conservation of structural features, but losing canonical functional sites. The data are further discussed in light of known mutations in the SH3 domain or its vicinity in the different Shank isoforms.
Collapse
Affiliation(s)
- Srinivas Kumar Ponna
- Faculty of Biochemistry and Molecular Medicine & Biocenter Oulu, University of Oulu, Finland
| | - Matti Myllykoski
- Faculty of Biochemistry and Molecular Medicine & Biocenter Oulu, University of Oulu, Finland
| | | | - Petri Kursula
- Faculty of Biochemistry and Molecular Medicine & Biocenter Oulu, University of Oulu, Finland; Department of Biomedicine, University of Bergen, Norway.
| |
Collapse
|
127
|
Lee Y, Kang H, Lee B, Zhang Y, Kim Y, Kim S, Kim WK, Han K. Integrative Analysis of Brain Region-specific Shank3 Interactomes for Understanding the Heterogeneity of Neuronal Pathophysiology Related to SHANK3 Mutations. Front Mol Neurosci 2017; 10:110. [PMID: 28469556 PMCID: PMC5395616 DOI: 10.3389/fnmol.2017.00110] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Accepted: 04/04/2017] [Indexed: 01/08/2023] Open
Abstract
Recent molecular genetic studies have identified 100s of risk genes for various neurodevelopmental and neuropsychiatric disorders. As the number of risk genes increases, it is becoming clear that different mutations of a single gene could cause different types of disorders. One of the best examples of such a gene is SHANK3, which encodes a core scaffold protein of the neuronal excitatory post-synapse. Deletions, duplications, and point mutations of SHANK3 are associated with autism spectrum disorders, intellectual disability, schizophrenia, bipolar disorder, and attention deficit hyperactivity disorder. Nevertheless, how the different mutations of SHANK3 can lead to such phenotypic diversity remains largely unknown. In this study, we investigated whether Shank3 could form protein complexes in a brain region-specific manner, which might contribute to the heterogeneity of neuronal pathophysiology caused by SHANK3 mutations. To test this, we generated a medial prefrontal cortex (mPFC) Shank3 in vivo interactome consisting of 211 proteins, and compared this protein list with a Shank3 interactome previously generated from mixed hippocampal and striatal (HP+STR) tissues. Unexpectedly, we found that only 47 proteins (about 20%) were common between the two interactomes, while 164 and 208 proteins were specifically identified in the mPFC and HP+STR interactomes, respectively. Each of the mPFC- and HP+STR-specific Shank3 interactomes represents a highly interconnected network. Upon comparing the brain region-enriched proteomes, we found that the large difference between the mPFC and HP+STR Shank3 interactomes could not be explained by differential protein expression profiles among the brain regions. Importantly, bioinformatic pathway analysis revealed that the representative biological functions of the mPFC- and HP+STR-specific Shank3 interactomes were different, suggesting that these interactors could mediate the brain region-specific functions of Shank3. Meanwhile, the same analysis on the common Shank3 interactors, including Homer and GKAP/SAPAP proteins, suggested that they could mainly function as scaffolding proteins at the post-synaptic density. Lastly, we found that the mPFC- and HP+STR-specific Shank3 interactomes contained a significant number of proteins associated with neurodevelopmental and neuropsychiatric disorders. These results suggest that Shank3 can form protein complexes in a brain region-specific manner, which might contribute to the pathophysiological and phenotypic diversity of disorders related to SHANK3 mutations.
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
| | - Hyojin Kang
- HPC-enabled Convergence Technology Research Division, Korea Institute of Science and Technology InformationDaejeon, 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
| | - Won-Ki Kim
- Department of Neuroscience, College of Medicine, Korea UniversitySeoul, South Korea
- Department of Biomedical Sciences, College of Medicine, Korea UniversitySeoul, 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
|
128
|
Wang W, Li C, Chen Q, van der Goes MS, Hawrot J, Yao AY, Gao X, Lu C, Zang Y, Zhang Q, Lyman K, Wang D, Guo B, Wu S, Gerfen CR, Fu Z, Feng G. Striatopallidal dysfunction underlies repetitive behavior in Shank3-deficient model of autism. J Clin Invest 2017; 127:1978-1990. [PMID: 28414301 DOI: 10.1172/jci87997] [Citation(s) in RCA: 119] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Accepted: 02/16/2017] [Indexed: 11/17/2022] Open
Abstract
The postsynaptic scaffolding protein SH3 and multiple ankyrin repeat domains 3 (SHANK3) is critical for the development and function of glutamatergic synapses. Disruption of the SHANK3-encoding gene has been strongly implicated as a monogenic cause of autism, and Shank3 mutant mice show repetitive grooming and social interaction deficits. Although basal ganglia dysfunction has been proposed to underlie repetitive behaviors, few studies have provided direct evidence to support this notion and the exact cellular mechanisms remain largely unknown. Here, we utilized the Shank3B mutant mouse model of autism to investigate how Shank3 mutation may differentially affect striatonigral (direct pathway) and striatopallidal (indirect pathway) medium spiny neurons (MSNs) and its relevance to repetitive grooming behavior in Shank3B mutant mice. We found that Shank3 deletion preferentially affects synapses onto striatopallidal MSNs. Striatopallidal MSNs showed profound defects, including alterations in synaptic transmission, synaptic plasticity, and spine density. Importantly, the repetitive grooming behavior was rescued by selectively enhancing the striatopallidal MSN activity via a Gq-coupled human M3 muscarinic receptor (hM3Dq), a type of designer receptors exclusively activated by designer drugs (DREADD). Our findings directly demonstrate the existence of distinct changes between 2 striatal pathways in a mouse model of autism and indicate that the indirect striatal pathway disruption might play a causative role in repetitive behavior of Shank3B mutant mice.
Collapse
|
129
|
Lee B, Zhang Y, Kim Y, Kim S, Lee Y, Han K. Age-dependent decrease of GAD65/67 mRNAs but normal densities of GABAergic interneurons in the brain regions of Shank3-overexpressing manic mouse model. Neurosci Lett 2017; 649:48-54. [PMID: 28400125 DOI: 10.1016/j.neulet.2017.04.016] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Revised: 03/31/2017] [Accepted: 04/07/2017] [Indexed: 10/19/2022]
Abstract
Dysfunction of inhibitory GABAergic interneurons is considered a major pathophysiological feature of various neurodevelopmental and neuropsychiatric disorders. The variants of SHANK3 gene, encoding a core scaffold protein of the excitatory postsynapse, have been associated with numerous brain disorders. It has been suggested that abnormalities of GABAergic interneurons could contribute to the SHANK3-related disorders, but the limitation of these studies is that they used mainly Shank3 knock-out mice. Notably, Shank3-overexpressing transgenic mice, modeling human hyperkinetic disorders, also show reduced inhibitory synaptic transmission, abnormal electroencephalography, and spontaneous seizures. However, it has not been investigated whether these phenotypes of Shank3 transgenic mice are associated with GABAergic interneuron dysfunction, or solely due to the cell-autonomous postsynaptic changes of principal neurons. To address this issue, we investigated the densities of parvalbumin- and somatostatin-positive interneurons, and the mRNA and protein levels of GAD65/67 GABA-synthesizing enzymes in the medial prefrontal cortex, striatum, and hippocampus of adult Shank3 transgenic mice. We found no significant difference in the measurements performed on wild-type versus Shank3 transgenic mice, except for the decreased GAD65 or GAD67 mRNAs in these brain regions. Interestingly, only GAD65 mRNA was decreased in the hippocampus, but not mPFC and striatum, of juvenile Shank3 transgenic mice which, unlike the adult mice, did not show behavioral hyperactivity. Together, our results suggest age-dependent decrease of GAD65/67 mRNAs but normal densities of certain GABAergic interneurons in the Shank3 transgenic mice.
Collapse
Affiliation(s)
- Bokyoung Lee
- Department of Neuroscience, College of Medicine, Korea University, Seoul 02841, South Korea
| | - Yinhua Zhang
- Department of Neuroscience, College of Medicine, Korea University, 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, Seoul 02841, South Korea
| | - Shinhyun Kim
- Department of Neuroscience, College of Medicine, Korea University, 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, 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, Seoul 02841, South Korea; Department of Biomedical Sciences, College of Medicine, Korea University, Seoul 02841, South Korea.
| |
Collapse
|
130
|
Sanders AR, Drigalenko EI, Duan J, Moy W, Freda J, Göring HHH, Gejman PV. Transcriptome sequencing study implicates immune-related genes differentially expressed in schizophrenia: new data and a meta-analysis. Transl Psychiatry 2017; 7:e1093. [PMID: 28418402 PMCID: PMC5416689 DOI: 10.1038/tp.2017.47] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Revised: 01/16/2017] [Accepted: 02/01/2017] [Indexed: 12/17/2022] Open
Abstract
We undertook an RNA sequencing (RNAseq)-based transcriptomic profiling study on lymphoblastoid cell lines of a European ancestry sample of 529 schizophrenia cases and 660 controls, and found 1058 genes to be differentially expressed by affection status. These differentially expressed genes were enriched for involvement in immunity, especially the 697 genes with higher expression in cases. Comparing the current RNAseq transcriptomic profiling to our previous findings in an array-based study of 268 schizophrenia cases and 446 controls showed a highly significant positive correlation over all genes. Fifteen (18%) of the 84 genes with significant (false discovery rate<0.05) expression differences between cases and controls in the previous study and analyzed here again were differentially expressed by affection status here at a genome-wide significance level (Bonferroni P<0.05 adjusted for 8141 analyzed genes in total, or P<~6.1 × 10-6), all with the same direction of effect, thus providing corroborative evidence despite each sample of fully independent subjects being studied by different technological approaches. Meta-analysis of the RNAseq and array data sets (797 cases and 1106 controls) showed 169 additional genes (besides those found in the primary RNAseq-based analysis) to be differentially expressed, and provided further evidence of immune gene enrichment. In addition to strengthening our previous array-based gene expression differences in schizophrenia cases versus controls and providing transcriptomic support for some genes implicated by other approaches for schizophrenia, our study detected new genes differentially expressed in schizophrenia. We highlight RNAseq-based differential expression of various genes involved in neurodevelopment and/or neuronal function, and discuss caveats of the approach.
Collapse
Affiliation(s)
- A R Sanders
- Department of Psychiatry and Behavioral Sciences, NorthShore University HealthSystem, Evanston, IL, USA,Department of Psychiatry and Behavioral Sciences, University of Chicago, Chicago, IL, USA,Department of Psychiatry and Behavioral Sciences, NorthShore University HealthSystem, 1001 University Place, Evanston, IL 60201, USA. E-mail:
| | - E I Drigalenko
- Department of Genetics, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - J Duan
- Department of Psychiatry and Behavioral Sciences, NorthShore University HealthSystem, Evanston, IL, USA,Department of Psychiatry and Behavioral Sciences, University of Chicago, Chicago, IL, USA
| | - W Moy
- Department of Psychiatry and Behavioral Sciences, NorthShore University HealthSystem, Evanston, IL, USA
| | - J Freda
- Department of Psychiatry and Behavioral Sciences, NorthShore University HealthSystem, Evanston, IL, USA
| | - H H H Göring
- South Texas Diabetes and Obesity Institute, University of Texas Rio Grande Valley School of Medicine, San Antonio, TX, USA
| | - P V Gejman
- Department of Psychiatry and Behavioral Sciences, NorthShore University HealthSystem, Evanston, IL, USA,Department of Psychiatry and Behavioral Sciences, University of Chicago, Chicago, IL, USA
| |
Collapse
|
131
|
SHANK proteins limit integrin activation by directly interacting with Rap1 and R-Ras. Nat Cell Biol 2017; 19:292-305. [PMID: 28263956 PMCID: PMC5386136 DOI: 10.1038/ncb3487] [Citation(s) in RCA: 98] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Accepted: 02/06/2017] [Indexed: 12/17/2022]
Abstract
SHANK3, a synaptic scaffold protein and actin regulator, is widely
expressed outside of the central nervous system with predominantly unknown
function. Solving the structure of the SHANK3 N-terminal region revealed that
the SPN-domain is an unexpected Ras-association domain with high affinity for
GTP-bound Ras and Rap G-proteins. The role of Rap1 in integrin activation is
well established but the mechanisms to antagonize it remain largely unknown.
Here, we show that SHANK1 and SHANK3 act as integrin activation inhibitors by
sequestering active Rap1 and R-Ras via the SPN-domain and thus limiting their
bioavailability at the plasma membrane. Consistently, SHANK3
silencing triggers increased plasma membrane Rap1 activity, cell spreading,
migration and invasion. Autism-related mutations within the SHANK3 SPN-domain
(R12C and L68P) disrupt G-protein interaction and fail to counteract integrin
activation along the Rap1/RIAM/talin axis in cancer cells and neurons.
Altogether, we establish SHANKs as critical regulators of G-protein signalling
and integrin-dependent processes.
Collapse
|
132
|
Abstract
Several large-scale genomic studies have supported an association between cases of autism spectrum disorder and mutations in the genes SH3 and multiple ankyrin repeat domains protein 1 (SHANK1), SHANK2 and SHANK3, which encode a family of postsynaptic scaffolding proteins that are present at glutamatergic synapses in the CNS. An evaluation of human genetic data, as well as of in vitro and in vivo animal model data, may allow us to understand how disruption of SHANK scaffolding proteins affects the structure and function of neural circuits and alters behaviour.
Collapse
|
133
|
Bariselli S, Bellone C. VTA DA neuron excitatory synapses in Shank3 Δex 4-9 mouse line. Synapse 2017; 71. [PMID: 28002633 DOI: 10.1002/syn.21955] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 11/18/2016] [Accepted: 12/12/2016] [Indexed: 01/22/2023]
Abstract
Several mutations within SHANK3 gene have been identified in Autism Spectrum Disorder patients and several studies have now started to show that those mutations could impact different brain circuits leading to the heterogeneity of the disease. Here we show that, compared to a mouse model lacking SHANK3 proline-rich containing isoforms, in a mouse model lacking SHANK3 ANK(yrin)-domain containing isoforms, the excitatory synaptic transmission within the Ventral Tegmental Area is not affected. We discuss about the possibility that different domains of SHANK3 are involved in regulating the synapses in a circuit-specific manner resulting in different behavioral and synaptic phenotypes.
Collapse
Affiliation(s)
| | - Camilla Bellone
- Department of Basic Neurosciences, University of Geneva, Geneva, Switzerland
| |
Collapse
|
134
|
Peter S, De Zeeuw CI, Boeckers TM, Schmeisser MJ. Cerebellar and Striatal Pathologies in Mouse Models of Autism Spectrum Disorder. ADVANCES IN ANATOMY, EMBRYOLOGY, AND CELL BIOLOGY 2017; 224:103-119. [PMID: 28551753 DOI: 10.1007/978-3-319-52498-6_6] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Autism spectrum disorder (ASD) is a complex neurodevelopmental condition with a strong genetic component. To date, several hundred different genetic mutations have been identified to play a role in its aetiology. The heterogeneity of genetic abnormalities combined with the different brain regions where aberrations are found makes the search for causative mechanisms a daunting task. Even within a limited number of brain regions, a myriad of different neural circuit dysfunctions may lead to ASD. Here, we review mouse models that incorporate mutations of ASD risk genes causing pathologies in the cerebellum and striatum and highlight the vulnerability of related circuit dysfunctions within these brain regions in ASD pathophysiology.
Collapse
Affiliation(s)
- Saša Peter
- Netherlands Institute for Neuroscience, Amsterdam, The Netherlands. .,Department of Neuroscience, Erasmus Medical Center, Rotterdam, The Netherlands.
| | - Chris I De Zeeuw
- Netherlands Institute for Neuroscience, Amsterdam, The Netherlands.,Department of Neuroscience, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Tobias M Boeckers
- Institute for Anatomy and Cell Biology, Ulm University, Ulm, Germany
| | - Michael J Schmeisser
- Institute for Anatomy and Cell Biology, Ulm University, Ulm, Germany. .,Division of Neuroanatomy, Institute of Anatomy, Otto-von-Guericke University, Magdeburg, Germany. .,Leibniz Institute for Neurobiology, Magdeburg, Germany.
| |
Collapse
|
135
|
Homann OR, Misura K, Lamas E, Sandrock RW, Nelson P, McDonough SI, DeLisi LE. Whole-genome sequencing in multiplex families with psychoses reveals mutations in the SHANK2 and SMARCA1 genes segregating with illness. Mol Psychiatry 2016; 21:1690-1695. [PMID: 27001614 PMCID: PMC5033653 DOI: 10.1038/mp.2016.24] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2015] [Revised: 01/16/2016] [Accepted: 01/20/2016] [Indexed: 12/30/2022]
Abstract
A current focus in psychiatric genetics is detection of multiple common risk alleles through very large genome-wide association study analyses. Yet families do exist, albeit rare, that have multiple affected members who are presumed to have a similar inherited cause to their illnesses. We hypothesized that within some of these families there may be rare highly penetrant mutations that segregate with illness. In this exploratory study, the genomes of 90 individuals across nine families were sequenced. Each family included a minimum of three available relatives affected with a psychotic illness and three available unaffected relatives. Twenty-six variants were identified that are private to a family, alter protein sequence, and are transmitted to all sequenced affected individuals within the family. In one family, seven siblings with schizophrenia spectrum disorders each carry a novel private missense variant within the SHANK2 gene. This variant lies within the consensus SH3 protein-binding motif by which SHANK2 may interact with post-synaptic glutamate receptors. In another family, four affected siblings and their unaffected mother each carry a novel private missense variant in the SMARCA1 gene on the X chromosome. Both variants represent candidates that may be causal for psychotic disorders when considered in the context of their transmission pattern and known gene and disease biology.
Collapse
Affiliation(s)
| | | | | | | | - Paul Nelson
- The BVARI Foundation, VA Boston Healthcare System
| | | | - Lynn E DeLisi
- The BVARI Foundation, VA Boston Healthcare System, VA Boston Healthcare System, Boston and Brockton, Ma, Department of Psychiatry, Harvard Medical School,Corresponding Author Address: Building 2, Rm 204, 940 Belmont Avenue, Brockton, Massachusetts, 02301 USA, Phone: 774-826-3155;
| |
Collapse
|
136
|
Han Q, Kim YH, Wang X, Liu D, Zhang ZJ, Bey AL, Lay M, Chang W, Berta T, Zhang Y, Jiang YH, Ji RR. SHANK3 Deficiency Impairs Heat Hyperalgesia and TRPV1 Signaling in Primary Sensory Neurons. Neuron 2016; 92:1279-1293. [PMID: 27916453 DOI: 10.1016/j.neuron.2016.11.007] [Citation(s) in RCA: 114] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Revised: 09/04/2016] [Accepted: 10/27/2016] [Indexed: 12/21/2022]
Abstract
Abnormal pain sensitivity is commonly associated with autism spectrum disorders (ASDs) and affects the life quality of ASD individuals. SHANK3 deficiency was implicated in ASD and pain dysregulation. Here, we report functional expression of SHANK3 in mouse dorsal root ganglion (DRG) sensory neurons and spinal cord presynaptic terminals. Homozygous and heterozygous Shank3 complete knockout (Δe4-22) results in impaired heat hyperalgesia in inflammatory and neuropathic pain. Specific deletion of Shank3 in Nav1.8-expressing sensory neurons also impairs heat hyperalgesia in homozygous and heterozygous mice. SHANK3 interacts with transient receptor potential subtype V1 (TRPV1) via Proline-rich region and regulates TRPV1 surface expression. Furthermore, capsaicin-induced spontaneous pain, inward currents in DRG neurons, and synaptic currents in spinal cord neurons are all reduced after Shank3 haploinsufficiency. Finally, partial knockdown of SHANK3 expression in human DRG neurons abrogates TRPV1 function. Our findings reveal a peripheral mechanism of SHANK3, which may underlie pain deficits in SHANK3-related ASDs.
Collapse
Affiliation(s)
- Qingjian Han
- Department of Anesthesiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Yong Ho Kim
- Department of Anesthesiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Xiaoming Wang
- Department of Pediatrics, Duke University Medical Center, Durham, NC 27710, USA
| | - Di Liu
- Department of Anesthesiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Zhi-Jun Zhang
- Department of Anesthesiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Alexandra L Bey
- Department of Neurobiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Mark Lay
- Department of Anesthesiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Wonseok Chang
- Department of Anesthesiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Temugin Berta
- Department of Anesthesiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Yan Zhang
- Department of Anesthesiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Yong-Hui Jiang
- Department of Pediatrics, Duke University Medical Center, Durham, NC 27710, USA; Department of Neurobiology, Duke University Medical Center, Durham, NC 27710, USA.
| | - Ru-Rong Ji
- Department of Anesthesiology, Duke University Medical Center, Durham, NC 27710, USA; Department of Neurobiology, Duke University Medical Center, Durham, NC 27710, USA.
| |
Collapse
|
137
|
Lin YC, Frei JA, Kilander MBC, Shen W, Blatt GJ. A Subset of Autism-Associated Genes Regulate the Structural Stability of Neurons. Front Cell Neurosci 2016; 10:263. [PMID: 27909399 PMCID: PMC5112273 DOI: 10.3389/fncel.2016.00263] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Accepted: 10/28/2016] [Indexed: 12/15/2022] Open
Abstract
Autism spectrum disorder (ASD) comprises a range of neurological conditions that affect individuals’ ability to communicate and interact with others. People with ASD often exhibit marked qualitative difficulties in social interaction, communication, and behavior. Alterations in neurite arborization and dendritic spine morphology, including size, shape, and number, are hallmarks of almost all neurological conditions, including ASD. As experimental evidence emerges in recent years, it becomes clear that although there is broad heterogeneity of identified autism risk genes, many of them converge into similar cellular pathways, including those regulating neurite outgrowth, synapse formation and spine stability, and synaptic plasticity. These mechanisms together regulate the structural stability of neurons and are vulnerable targets in ASD. In this review, we discuss the current understanding of those autism risk genes that affect the structural connectivity of neurons. We sub-categorize them into (1) cytoskeletal regulators, e.g., motors and small RhoGTPase regulators; (2) adhesion molecules, e.g., cadherins, NCAM, and neurexin superfamily; (3) cell surface receptors, e.g., glutamatergic receptors and receptor tyrosine kinases; (4) signaling molecules, e.g., protein kinases and phosphatases; and (5) synaptic proteins, e.g., vesicle and scaffolding proteins. Although the roles of some of these genes in maintaining neuronal structural stability are well studied, how mutations contribute to the autism phenotype is still largely unknown. Investigating whether and how the neuronal structure and function are affected when these genes are mutated will provide insights toward developing effective interventions aimed at improving the lives of people with autism and their families.
Collapse
Affiliation(s)
- Yu-Chih Lin
- Laboratory of Neuronal Connectivity, Program in Neuroscience, Hussman Institute for Autism, Baltimore MD, USA
| | - Jeannine A Frei
- Laboratory of Neuronal Connectivity, Program in Neuroscience, Hussman Institute for Autism, Baltimore MD, USA
| | - Michaela B C Kilander
- Laboratory of Neuronal Connectivity, Program in Neuroscience, Hussman Institute for Autism, Baltimore MD, USA
| | - Wenjuan Shen
- Laboratory of Neuronal Connectivity, Program in Neuroscience, Hussman Institute for Autism, Baltimore MD, USA
| | - Gene J Blatt
- Laboratory of Autism Neurocircuitry, Program in Neuroscience, Hussman Institute for Autism, Baltimore MD, USA
| |
Collapse
|
138
|
Determining the Roles of Inositol Trisphosphate Receptors in Neurodegeneration: Interdisciplinary Perspectives on a Complex Topic. Mol Neurobiol 2016; 54:6870-6884. [PMID: 27771899 DOI: 10.1007/s12035-016-0205-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Accepted: 10/11/2016] [Indexed: 02/06/2023]
Abstract
It is well known that calcium (Ca2+) is involved in the triggering of neuronal death. Ca2+ cytosolic levels are regulated by Ca2+ release from internal stores located in organelles, such as the endoplasmic reticulum. Indeed, Ca2+ transit from distinct cell compartments follows complex dynamics that are mediated by specific receptors, notably inositol trisphosphate receptors (IP3Rs). Ca2+ release by IP3Rs plays essential roles in several neurological disorders; however, details of these processes are poorly understood. Moreover, recent studies have shown that subcellular location, molecular identity, and density of IP3Rs profoundly affect Ca2+ transit in neurons. Therefore, regulation of IP3R gene products in specific cellular vicinities seems to be crucial in a wide range of cellular processes from neuroprotection to neurodegeneration. In this regard, microRNAs seem to govern not only IP3Rs translation levels but also subcellular accumulation. Combining new data from molecular cell biology with mathematical modelling, we were able to summarize the state of the art on this topic. In addition to presenting how Ca2+ dynamics mediated by IP3R activation follow a stochastic regimen, we integrated a theoretical approach in an easy-to-apply, cell biology-coherent fashion. Following the presented premises and in contrast to previously tested hypotheses, Ca2+ released by IP3Rs may play different roles in specific neurological diseases, including Alzheimer's disease and Parkinson's disease.
Collapse
|
139
|
Actin-Dependent Alterations of Dendritic Spine Morphology in Shankopathies. Neural Plast 2016; 2016:8051861. [PMID: 27795858 PMCID: PMC5067329 DOI: 10.1155/2016/8051861] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Accepted: 08/30/2016] [Indexed: 12/11/2022] Open
Abstract
Shank proteins (Shank1, Shank2, and Shank3) act as scaffolding molecules in the postsynaptic density of many excitatory neurons. Mutations in SHANK genes, in particular SHANK2 and SHANK3, lead to autism spectrum disorders (ASD) in both human and mouse models. Shank3 proteins are made of several domains-the Shank/ProSAP N-terminal (SPN) domain, ankyrin repeats, SH3 domain, PDZ domain, a proline-rich region, and the sterile alpha motif (SAM) domain. Via various binding partners of these domains, Shank3 is able to bind and interact with a wide range of proteins including modulators of small GTPases such as RICH2, a RhoGAP protein, and βPIX, a RhoGEF protein for Rac1 and Cdc42, actin binding proteins and actin modulators. Dysregulation of all isoforms of Shank proteins, but especially Shank3, leads to alterations in spine morphogenesis, shape, and activity of the synapse via altering actin dynamics. Therefore, here, we highlight the role of Shank proteins as modulators of small GTPases and, ultimately, actin dynamics, as found in multiple in vitro and in vivo models. The failure to mediate this regulatory role might present a shared mechanism in the pathophysiology of autism-associated mutations, which leads to dysregulation of spine morphogenesis and synaptic signaling.
Collapse
|
140
|
Liu CX, Peng XL, Hu CC, Li CY, Li Q, Xu X. Developmental profiling of ASD-related shank3 transcripts and their differential regulation by valproic acid in zebrafish. Dev Genes Evol 2016; 226:389-400. [PMID: 27562614 PMCID: PMC5099374 DOI: 10.1007/s00427-016-0561-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Accepted: 08/15/2016] [Indexed: 01/15/2023]
Abstract
SHANK3 is a scaffolding protein that binds to various synaptic proteins at the postsynaptic density (PSD) of excitatory glutamatergic synapses. SHANK3 is not only strongly implicated in autism spectrum disorders (ASD) but also plays a critical role in human Phelan-McDermid syndrome (22q13.3 deletion syndrome). Accumulated experimental evidence demonstrates that the zebrafish model system is useful for studying the functions of ASD-related gene during early development. However, many basic features of shank3 transcript expression in zebrafish remain poorly understood. Here, we investigated temporal, spatial, and isoform-specific expression patterns of shank3 during zebrafish development on the basis of previous researches and the differential effects of each shank3 transcript expression after exposure to valproic acid (VPA), an ASD-associated drug. At first, we observed that both shank3a and shank3b were barely expressed at very early ages (before 24 h post-fertilization (hpf)), whereas their expression levels were increased and mainly enriched in the nervous system after 24 hpf. Secondly, all of the six shank3 transcripts gradually increased during the first 7 hpf and then decreased. Subsequently, they exhibited a second increasing peak between 1 month post-fertilization (mpf) and adulthood. Thirdly, VPA treatment affected the isoform-specific expression of zebrafish shank3. In particular, the mRNA expression levels of those isoforms that contain a SAM domain were significantly increased, whereas the mRNA expression level of those which contained an ANK domain but without a SAM domain was decreased. To conclude, our findings support the molecular diversity of shank3 in zebrafish and provide a molecular framework to understand the isoform-specific function of shank3 in zebrafish.
Collapse
Affiliation(s)
- Chun-Xue Liu
- Division of Child Health Care, Children's Hospital of Fudan University, 399 Wanyuan Road, Shanghai, 201102, China
| | - Xiao-Lan Peng
- Center for Translational Medicine, Institute of Pediatrics, Shanghai Key Laboratory of Birth Defect, Children's Hospital of Fudan University, 399 Wanyuan Road, Shanghai, 201102, China
| | - Chun-Chun Hu
- Division of Child Health Care, Children's Hospital of Fudan University, 399 Wanyuan Road, Shanghai, 201102, China
| | - Chun-Yang Li
- Division of Child Health Care, Children's Hospital of Fudan University, 399 Wanyuan Road, Shanghai, 201102, China
| | - Qiang Li
- Center for Translational Medicine, Institute of Pediatrics, Shanghai Key Laboratory of Birth Defect, Children's Hospital of Fudan University, 399 Wanyuan Road, Shanghai, 201102, China.
| | - Xiu Xu
- Division of Child Health Care, Children's Hospital of Fudan University, 399 Wanyuan Road, Shanghai, 201102, China.
| |
Collapse
|
141
|
Vester A, Caudle WM. The Synapse as a Central Target for Neurodevelopmental Susceptibility to Pesticides. TOXICS 2016; 4:toxics4030018. [PMID: 29051423 PMCID: PMC5606656 DOI: 10.3390/toxics4030018] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Revised: 08/07/2016] [Accepted: 08/17/2016] [Indexed: 12/12/2022]
Abstract
The developmental period of the nervous system is carefully orchestrated and highly vulnerable to alterations. One crucial factor of a properly-functioning nervous system is the synapse, as synaptic signaling is critical for the formation and maturation of neural circuits. Studies show that genetic and environmental impacts can affect diverse components of synaptic function. Importantly, synaptic dysfunction is known to be associated with neurologic and psychiatric disorders, as well as more subtle cognitive, psychomotor, and sensory defects. Given the importance of the synapse in numerous domains, we wanted to delineate the effects of pesticide exposure on synaptic function. In this review, we summarize current epidemiologic and molecular studies that demonstrate organochlorine, organophosphate, and pyrethroid pesticide exposures target the developing synapse. We postulate that the synapse plays a central role in synaptic vulnerability to pesticide exposure during neurodevelopment, and the synapse is a worthy candidate for investigating more subtle effects of chronic pesticide exposure in future studies.
Collapse
Affiliation(s)
- Aimee Vester
- Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA 30322, USA.
| | - W Michael Caudle
- Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA 30322, USA.
- Center for Neurodegenerative Disease, School of Medicine, Emory University, Atlanta, GA 30322, USA.
| |
Collapse
|
142
|
Abstract
Autism spectrum disorders (ASD) are highly heterogeneous pediatric developmental disorders with estimated heritability more than 70%. Although the genetic factors in ASD are mainly unknown, a large number of gene mutations have been found, especially in genes involved in neurogenesis. The Neurexin-Neuroligin-Shank (NRXN-NLGN-SHANK) pathway plays a key role in the formation, maturation and maintenance of synapses, consistent with the hypothesis of neurodevelopmental abnormality in ASD. Presynaptic NRXNs interact with postsynaptic NLGNs in excitatory glutamatergic synapses. SHANK proteins function as core components of the postsynaptic density (PSD) by interacting with multiple proteins. Recently, deletions and point mutations of the SHANK1 gene have been detected in ASD individuals, indicating the involvement of SHANK1 in ASD. This review focuses on the function of SHANK1 protein, Shank1 mouse models, and the molecular genetics of the SHANK1 gene in human ASD.
Collapse
Affiliation(s)
- XiaoHong Gong
- MOE Key Laboratory of Contemporary Anthropology and State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, 200433, China.
| | - HongYan Wang
- MOE Key Laboratory of Contemporary Anthropology and State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, 200433, China.
| |
Collapse
|
143
|
Zatkova M, Bakos J, Hodosy J, Ostatnikova D. Synapse alterations in autism: Review of animal model findings. Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub 2016; 160:201-10. [DOI: 10.5507/bp.2015.066] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Accepted: 12/04/2015] [Indexed: 12/30/2022] Open
|
144
|
Reijntjes DO, Pyott SJ. The afferent signaling complex: Regulation of type I spiral ganglion neuron responses in the auditory periphery. Hear Res 2016; 336:1-16. [DOI: 10.1016/j.heares.2016.03.011] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Revised: 02/12/2016] [Accepted: 03/07/2016] [Indexed: 12/19/2022]
|
145
|
Reim D, Weis TM, Halbedl S, Delling JP, Grabrucker AM, Boeckers TM, Schmeisser MJ. The Shank3 Interaction Partner ProSAPiP1 Regulates Postsynaptic SPAR Levels and the Maturation of Dendritic Spines in Hippocampal Neurons. Front Synaptic Neurosci 2016; 8:13. [PMID: 27252646 PMCID: PMC4877498 DOI: 10.3389/fnsyn.2016.00013] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Accepted: 05/10/2016] [Indexed: 11/13/2022] Open
Abstract
The postsynaptic density or PSD is a submembranous compartment containing a wide array of proteins that contribute to both morphology and function of excitatory glutamatergic synapses. In this study, we have analyzed functional aspects of the Fezzin ProSAP-interacting protein 1 (ProSAPiP1), an interaction partner of the well-known PSD proteins Shank3 and SPAR. Using lentiviral-mediated overexpression and knockdown of ProSAPiP1, we found that this protein is dispensable for the formation of both pre- and postsynaptic specializations per se. We further show that ProSAPiP1 regulates SPAR levels at the PSD and the maturation of dendritic spines. In line with previous findings on the ProSAPiP1 homolog PSD-Zip70, we conclude that Fezzins essentially contribute to the maturation of excitatory spine synapses.
Collapse
Affiliation(s)
- Dominik Reim
- Institute for Anatomy and Cell Biology, Ulm UniversityUlm, Germany; International Graduate School in Molecular Medicine, Ulm UniversityUlm, Germany
| | - Tobias M Weis
- Institute for Anatomy and Cell Biology, Ulm UniversityUlm, Germany; International Graduate School in Molecular Medicine, Ulm UniversityUlm, Germany
| | - Sonja Halbedl
- Institute for Anatomy and Cell Biology, Ulm UniversityUlm, Germany; International Graduate School in Molecular Medicine, Ulm UniversityUlm, Germany
| | - Jan Philipp Delling
- Institute for Anatomy and Cell Biology, Ulm UniversityUlm, Germany; International Graduate School in Molecular Medicine, Ulm UniversityUlm, Germany
| | - Andreas M Grabrucker
- Institute for Anatomy and Cell Biology, Ulm UniversityUlm, Germany; WG Molecular Analysis of Synaptopathies, Department of Neurology, Neurocenter of Ulm UniversityUlm, Germany
| | | | - Michael J Schmeisser
- Institute for Anatomy and Cell Biology, Ulm UniversityUlm, Germany; Department of Neurology, Ulm UniversityUlm, Germany
| |
Collapse
|
146
|
Copping NA, Berg EL, Foley GM, Schaffler MD, Onaga BL, Buscher N, Silverman JL, Yang M. Touchscreen learning deficits and normal social approach behavior in the Shank3B model of Phelan-McDermid Syndrome and autism. Neuroscience 2016; 345:155-165. [PMID: 27189882 DOI: 10.1016/j.neuroscience.2016.05.016] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Revised: 05/02/2016] [Accepted: 05/05/2016] [Indexed: 11/24/2022]
Abstract
SHANK3 is a synaptic scaffolding protein localized in the postsynaptic density and has a crucial role in synaptogenesis and neural physiology. Deletions and point mutations in SHANK3 cause Phelan-McDermid Syndrome (PMS), and have also been implicated in autism spectrum disorder (ASD) and intellectual disabilities, leading to the hypothesis that reduced SHANK3 expression impairs basic brain functions that are important for social communication and cognition. Several mouse models of Shank3 deletions have been generated, varying in the specific domain deleted. Here we report impairments in cognitive function in mice heterozygous for exon 13-16 (coding for the PDZ domain) deletion. The touchscreen pairwise discrimination task was chosen by virtue of its: (a) conceptual and technical similarities to the Cambridge Neuropsychological Test Automated Battery (CANTAB) and NIH Toolbox Cognition Battery used for testing cognitive functions in humans, (b) minimal demand on motor abilities, and (c) capability to measure many aspects of learning and memory and complex cognitive functions, including cognitive flexibility. The similarity between our mouse tasks and human cognitive assays means a high translational validity in future intervention studies using preclinical models. Our study revealed that Shank3B heterozygous mice (+/-) were slower to reach criterion in the pairwise visual discrimination task, and exhibited trends toward making more errors (first trial errors) and more correction errors than wildtype mice (+/+). Open field activity was normal in +/-, ruling out hypo- or hyperactivity as potential confounds in the touchscreen test. Sociability in the three chamber test was also normal in both +/+ and +/-. These results indicate a deficit in discrimination learning in the Shank3B model of PMS and ASD, suggesting that this mouse model is a useful preclinical tool for studying neurobiological mechanisms behind cognitive impairments in PMS and ASD. The current findings are the starting point for our future research in which we will investigate multiple domains of cognition and explore pharmacological interventions.
Collapse
Affiliation(s)
- Nycole A Copping
- MIND Institute, Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, CA 95817, United States
| | - Elizabeth L Berg
- MIND Institute, Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, CA 95817, United States
| | - Gillian M Foley
- MIND Institute, Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, CA 95817, United States
| | - Melanie D Schaffler
- MIND Institute, Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, CA 95817, United States
| | - Beth L Onaga
- MIND Institute, Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, CA 95817, United States
| | - Nathalie Buscher
- MIND Institute, Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, CA 95817, United States
| | - Jill L Silverman
- MIND Institute, Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, CA 95817, United States
| | - Mu Yang
- MIND Institute, Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, CA 95817, United States.
| |
Collapse
|
147
|
Zinc Stabilizes Shank3 at the Postsynaptic Density of Hippocampal Synapses. PLoS One 2016; 11:e0153979. [PMID: 27144302 PMCID: PMC4856407 DOI: 10.1371/journal.pone.0153979] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Accepted: 04/06/2016] [Indexed: 12/18/2022] Open
Abstract
Shank3 is a postsynaptic density (PSD) scaffold protein of the Shank family. Here we use pre-embedding immunogold electron microscopy to investigate factors influencing the distribution of Shank3 at the PSD. In dissociated rat hippocampal cultures under basal conditions, label for Shank3 was concentrated in a broad layer of the PSD, ~20–80 nm from the postsynaptic membrane. Upon depolarization with high K+ (90 mM, 2 min), or application of NMDA (50 μM, 2 min), both the labeling intensity at the PSD and the median distance of label from the postsynaptic membrane increased significantly, indicating that Shank3 molecules are preferentially recruited to the distal layer of the PSD. Incubation in medium supplemented with zinc (50 μM ZnCl2, 1 hr) also significantly increased labeling intensity for Shank3 at the PSD, but this addition of Shank3 was not preferential to the distal layer. When cells were incubated with zinc and then treated with NMDA, labeling intensity of Shank3 became higher than with either treatment alone and manifested a preference for the distal layer of the PSD. Without zinc supplementation, NMDA-induced accumulation of Shank3 at the PSD was transient, reversing within 30 min after return to control medium. However, when zinc was included in culture media throughout the experiment, the NMDA-induced accumulation of Shank3 was largely retained, including Shank3 molecules recruited to the distal layer of the PSD. These results demonstrate that activity induces accumulation of Shank3 at the PSD and that zinc stabilizes PSD-associated Shank3, possibly through strengthening of Shank-Shank association.
Collapse
|
148
|
Heise C, Schroeder JC, Schoen M, Halbedl S, Reim D, Woelfle S, Kreutz MR, Schmeisser MJ, Boeckers TM. Selective Localization of Shanks to VGLUT1-Positive Excitatory Synapses in the Mouse Hippocampus. Front Cell Neurosci 2016; 10:106. [PMID: 27199660 PMCID: PMC4844616 DOI: 10.3389/fncel.2016.00106] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2016] [Accepted: 04/11/2016] [Indexed: 12/13/2022] Open
Abstract
Members of the Shank family of multidomain proteins (Shank1, Shank2, and Shank3) are core components of the postsynaptic density (PSD) of excitatory synapses. At synaptic sites Shanks serve as scaffolding molecules that cluster neurotransmitter receptors as well as cell adhesion molecules attaching them to the actin cytoskeleton. In this study we investigated the synapse specific localization of Shank1-3 and focused on well-defined synaptic contacts within the hippocampal formation. We found that all three family members are present only at VGLUT1-positive synapses, which is particularly visible at mossy fiber contacts. No costaining was found at VGLUT2-positive contacts indicating that the molecular organization of VGLUT2-associated PSDs diverges from classical VGLUT1-positive excitatory contacts in the hippocampus. In light of SHANK mutations in neuropsychiatric disorders, this study indicates which glutamatergic networks within the hippocampus will be primarily affected by shankopathies.
Collapse
Affiliation(s)
- Christopher Heise
- Institute for Anatomy and Cell Biology, Ulm UniversityUlm, Germany; RG Neuroplasticity, Leibniz Institute for NeurobiologyMagdeburg, Germany
| | - Jan C Schroeder
- Institute for Anatomy and Cell Biology, Ulm University Ulm, Germany
| | - Michael Schoen
- Institute for Anatomy and Cell Biology, Ulm University Ulm, Germany
| | - Sonja Halbedl
- Institute for Anatomy and Cell Biology, Ulm University Ulm, Germany
| | - Dominik Reim
- Institute for Anatomy and Cell Biology, Ulm University Ulm, Germany
| | - Sarah Woelfle
- Institute for Anatomy and Cell Biology, Ulm University Ulm, Germany
| | - Michael R Kreutz
- RG Neuroplasticity, Leibniz Institute for Neurobiology Magdeburg, Germany
| | - Michael J Schmeisser
- Institute for Anatomy and Cell Biology, Ulm UniversityUlm, Germany; Department of Neurology, Ulm UniversityUlm, Germany
| | | |
Collapse
|
149
|
Yi F, Danko T, Botelho SC, Patzke C, Pak C, Wernig M, Südhof TC. Autism-associated SHANK3 haploinsufficiency causes Ih channelopathy in human neurons. Science 2016; 352:aaf2669. [PMID: 26966193 DOI: 10.1126/science.aaf2669] [Citation(s) in RCA: 233] [Impact Index Per Article: 25.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Accepted: 02/26/2016] [Indexed: 12/13/2022]
Abstract
Heterozygous SHANK3 mutations are associated with idiopathic autism and Phelan-McDermid syndrome. SHANK3 is a ubiquitously expressed scaffolding protein that is enriched in postsynaptic excitatory synapses. Here, we used engineered conditional mutations in human neurons and found that heterozygous and homozygous SHANK3 mutations severely and specifically impaired hyperpolarization-activated cation (Ih) channels. SHANK3 mutations caused alterations in neuronal morphology and synaptic connectivity; chronic pharmacological blockage of Ih channels reproduced these phenotypes, suggesting that they may be secondary to Ih-channel impairment. Moreover, mouse Shank3-deficient neurons also exhibited severe decreases in Ih currents. SHANK3 protein interacted with hyperpolarization-activated cyclic nucleotide-gated channel proteins (HCN proteins) that form Ih channels, indicating that SHANK3 functions to organize HCN channels. Our data suggest that SHANK3 mutations predispose to autism, at least partially, by inducing an Ih channelopathy that may be amenable to pharmacological intervention.
Collapse
Affiliation(s)
- Fei Yi
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, 265 Campus Drive, Stanford, CA 94305, USA
| | - Tamas Danko
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, 265 Campus Drive, Stanford, CA 94305, USA. Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, 265 Campus Drive, Stanford, CA 94305, USA. Department of Pathology, Stanford University School of Medicine, 265 Campus Drive, Stanford, CA 94305, USA
| | - Salome Calado Botelho
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, 265 Campus Drive, Stanford, CA 94305, USA
| | - Christopher Patzke
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, 265 Campus Drive, Stanford, CA 94305, USA
| | - ChangHui Pak
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, 265 Campus Drive, Stanford, CA 94305, USA
| | - Marius Wernig
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, 265 Campus Drive, Stanford, CA 94305, USA. Department of Pathology, Stanford University School of Medicine, 265 Campus Drive, Stanford, CA 94305, USA
| | - Thomas C Südhof
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, 265 Campus Drive, Stanford, CA 94305, USA. Howard Hughes Medical Institute, Stanford University School of Medicine, 265 Campus Drive, Stanford, CA 94305, USA.
| |
Collapse
|
150
|
He Y, Wang P, Wei P, Feng H, Ren Y, Yang J, Rao Y, Shi J, Tian J. Effects of curcumin on synapses in APPswe/PS1dE9 mice. Int J Immunopathol Pharmacol 2016; 29:217-25. [PMID: 26957323 DOI: 10.1177/0394632016638099] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Accepted: 02/16/2016] [Indexed: 12/13/2022] Open
Abstract
Significant losses of synapses have been demonstrated in studies of Alzheimer's disease (AD), but structural and functional changes in synapses that depend on alterations of the postsynaptic density (PSD) area occur prior to synaptic loss and play a crucial role in the pathology of AD. Evidence suggests that curcumin can ameliorate the learning and memory deficits of AD. To investigate the effects of curcumin on synapses, APPswe/PS1dE9 double transgenic mice (an AD model) were used, and the ultra-structures of synapses and synapse-associated proteins were observed. Six months after administration, few abnormal synapses were observed upon electron microscopy in the hippocampal CA1 areas of the APPswe/PS1dE9 double transgenic mice. The treatment of the mice with curcumin resulted in improvements in the quantity and structure of the synapses. Immunohistochemistry and western blot analyses revealed that the expressions of PSD95 and Shank1 were reduced in the hippocampal CA1 areas of the APPswe/PS1dE9 double transgenic mice, but curcumin treatment increased the expressions of these proteins. Our findings suggest that curcumin improved the structure and function of the synapses by regulating the synapse-related proteins PSD95 and Shank1.
Collapse
Affiliation(s)
- Yingkun He
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine (BUCM), Beijing, PR China Key Laboratory of Pharmacology of Dongzhimen Hospital (BUCM), State Administration of Traditional Chinese Medicine, Beijing, PR China
| | - Pengwen Wang
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine (BUCM), Beijing, PR China Key Laboratory of Pharmacology of Dongzhimen Hospital (BUCM), State Administration of Traditional Chinese Medicine, Beijing, PR China
| | - Peng Wei
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine (BUCM), Beijing, PR China Jiaozuo Hospital of Traditional Chinese Medicine, Jiaozuo City, Henan Province, PR China
| | - Huili Feng
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine (BUCM), Beijing, PR China Key Laboratory of Pharmacology of Dongzhimen Hospital (BUCM), State Administration of Traditional Chinese Medicine, Beijing, PR China
| | - Ying Ren
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine (BUCM), Beijing, PR China Key Laboratory of Pharmacology of Dongzhimen Hospital (BUCM), State Administration of Traditional Chinese Medicine, Beijing, PR China
| | - Jinduo Yang
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine (BUCM), Beijing, PR China Key Laboratory of Pharmacology of Dongzhimen Hospital (BUCM), State Administration of Traditional Chinese Medicine, Beijing, PR China
| | - Yingxue Rao
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine (BUCM), Beijing, PR China University of Washington, Seattle, WA, USA
| | - Jing Shi
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine (BUCM), Beijing, PR China Beijing University of Chinese Medicine, BUCM Neurology Center, Dongzhimen Hospital, Beijing, PR China
| | - Jinzhou Tian
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine (BUCM), Beijing, PR China Beijing University of Chinese Medicine, BUCM Neurology Center, Dongzhimen Hospital, Beijing, PR China
| |
Collapse
|