1
|
Réthelyi JM, Vincze K, Schall D, Glennon J, Berkel S. The role of insulin/IGF1 signalling in neurodevelopmental and neuropsychiatric disorders - Evidence from human neuronal cell models. Neurosci Biobehav Rev 2023; 153:105330. [PMID: 37516219 DOI: 10.1016/j.neubiorev.2023.105330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 07/15/2023] [Accepted: 07/26/2023] [Indexed: 07/31/2023]
Abstract
Insulin and insulin-like growth factor 1 (IGF1) signalling play a central role in the development and maintenance of neurons in the brain, and human neurodevelopmental as well as neuropsychiatric disorders have been linked to impaired insulin and IGF1 signalling. This review focuses on the impairments of the insulin and IGF1 signalling cascade in the context of neurodevelopmental and neuropsychiatric disorders, based on evidence from human neuronal cell models. Clear evidence was obtained for impaired insulin and IGF1 receptor downstream signalling in neurodevelopmental disorders, while the evidence for its role in neuropsychiatric disorders was less substantial. Human neuronal model systems can greatly add to our knowledge about insulin/IGF1 signalling in the brain, its role in restoring dendritic maturity, and complement results from clinical studies and animal models. Moreover, they represent a useful model for the development of new therapeutic strategies. Further research is needed to systematically investigate the exact role of the insulin/IGF1 signalling cascades in neurodevelopmental and neuropsychiatric disorders, and to elucidate the respective therapeutic implications.
Collapse
Affiliation(s)
- János M Réthelyi
- Department of Psychiatry and Psychotherapy, Semmelweis University, Budapest, Hungary
| | - Katalin Vincze
- Department of Psychiatry and Psychotherapy, Semmelweis University, Budapest, Hungary; Doctoral School of Mental Health Sciences, Semmelweis University, Budapest, Hungary
| | - Dorothea Schall
- Institute of Human Genetics, Heidelberg University, Heidelberg, Germany
| | - Jeffrey Glennon
- Conway Institute of Biomedical and Biomolecular Research, School of Medicine, University College Dublin, Dublin, Ireland
| | - Simone Berkel
- Institute of Human Genetics, Heidelberg University, Heidelberg, Germany; Interdisciplinary Centre of Neurosciences (IZN), Heidelberg University, Germany.
| |
Collapse
|
2
|
Zheng Y, Li Q, Gong B, Xia Y, Lu X, Liu Y, Wu H, She S, Wu C. Negative-emotion-induced reduction in speech-in-noise recognition is associated with source-monitoring deficits and psychiatric symptoms in mandarin-speaking patients with schizophrenia. Compr Psychiatry 2023; 124:152395. [PMID: 37216805 DOI: 10.1016/j.comppsych.2023.152395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Revised: 05/03/2023] [Accepted: 05/15/2023] [Indexed: 05/24/2023] Open
Abstract
BACKGROUND Patients with schizophrenia (SCH) have deficits in source monitoring (SM), speech-in-noise recognition (SR), and auditory prosody recognition. This study aimed to test the covariation between SM and SR alteration induced by negative prosodies and their association with psychiatric symptoms in SCH. METHODS Fifty-four SCH patients and 59 healthy controls (HCs) underwent a speech SM task, an SR task, and the assessment of positive and negative syndrome scale (PANSS). We used the multivariate analyses of partial least squares (PLS) regression to explore the associations among SM (external/internal/new attribution error [AE] and response bias [RB]), SR alteration/release induced by four negative-emotion (sad, angry, fear, and disgust) prosodies of target speech, and psychiatric symptoms. RESULTS In SCH, but not HCs, a profile (linear combination) of SM (especially the external-source RB) was positively associated with a profile of SR reductions (induced especially by the angry prosody). Moreover, two SR reduction profiles (especially in the anger and sadness conditions) were related to two profiles of psychiatric symptoms (negative symptoms, lack of insight, and emotional disturbances). The two PLS components explained 50.4% of the total variances of the release-symptom association. CONCLUSION Compared to HCs, SCH is more likely to perceive the external-source speech as internal/new source speech. The SM-related SR reduction induced by the angry prosody was mainly associated with negative symptoms. These findings help understand the psychopathology of SCH and may provide a potential direction to improve negative symptoms via minimizing emotional SR reduction in schizophrenia.
Collapse
Affiliation(s)
- Yingjun Zheng
- The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou 510145, Guangdong, China
| | - Qiuhong Li
- Peking University School of Nursing, Beijing 100191, China
| | - Bingyan Gong
- Peking University School of Nursing, Beijing 100191, China
| | - Yu Xia
- The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou 510145, Guangdong, China
| | - Xiaohua Lu
- The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou 510145, Guangdong, China
| | - Yi Liu
- The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou 510145, Guangdong, China
| | - Huawang Wu
- The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou 510145, Guangdong, China
| | - Shenglin She
- The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou 510145, Guangdong, China.
| | - Chao Wu
- Peking University School of Nursing, Beijing 100191, China.
| |
Collapse
|
3
|
de Bartolomeis A, Vellucci L, Barone A, Manchia M, De Luca V, Iasevoli F, Correll CU. Clozapine's multiple cellular mechanisms: What do we know after more than fifty years? A systematic review and critical assessment of translational mechanisms relevant for innovative strategies in treatment-resistant schizophrenia. Pharmacol Ther 2022; 236:108236. [PMID: 35764175 DOI: 10.1016/j.pharmthera.2022.108236] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 06/21/2022] [Accepted: 06/21/2022] [Indexed: 12/21/2022]
Abstract
Almost fifty years after its first introduction into clinical care, clozapine remains the only evidence-based pharmacological option for treatment-resistant schizophrenia (TRS), which affects approximately 30% of patients with schizophrenia. Despite the long-time experience with clozapine, the specific mechanism of action (MOA) responsible for its superior efficacy among antipsychotics is still elusive, both at the receptor and intracellular signaling level. This systematic review is aimed at critically assessing the role and specific relevance of clozapine's multimodal actions, dissecting those mechanisms that under a translational perspective could shed light on molecular targets worth to be considered for further innovative antipsychotic development. In vivo and in vitro preclinical findings, supported by innovative techniques and methods, together with pharmacogenomic and in vivo functional studies, point to multiple and possibly overlapping MOAs. To better explore this crucial issue, the specific affinity for 5-HT2R, D1R, α2c, and muscarinic receptors, the relatively low occupancy at dopamine D2R, the interaction with receptor dimers, as well as the potential confounder effects resulting in biased ligand action, and lastly, the role of the moiety responsible for lipophilic and alkaline features of clozapine are highlighted. Finally, the role of transcription and protein changes at the synaptic level, and the possibility that clozapine can directly impact synaptic architecture are addressed. Although clozapine's exact MOAs that contribute to its unique efficacy and some of its severe adverse effects have not been fully understood, relevant information can be gleaned from recent mechanistic understandings that may help design much needed additional therapeutic strategies for TRS.
Collapse
Affiliation(s)
- Andrea de Bartolomeis
- Section of Psychiatry, Laboratory of Translational and Molecular Psychiatry and Unit of Treatment Resistant Psychosis, Department of Neuroscience, Reproductive Science and Dentistry, University Medical School of Naples "Federico II", Naples, Italy.
| | - Licia Vellucci
- Section of Psychiatry, Laboratory of Translational and Molecular Psychiatry and Unit of Treatment Resistant Psychosis, Department of Neuroscience, Reproductive Science and Dentistry, University Medical School of Naples "Federico II", Naples, Italy
| | - Annarita Barone
- Section of Psychiatry, Laboratory of Translational and Molecular Psychiatry and Unit of Treatment Resistant Psychosis, Department of Neuroscience, Reproductive Science and Dentistry, University Medical School of Naples "Federico II", Naples, Italy
| | - Mirko Manchia
- Section of Psychiatry, Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy; Department of Pharmacology, Dalhousie University, Halifax, Nova Scotia, Canada
| | | | - Felice Iasevoli
- Section of Psychiatry, Laboratory of Translational and Molecular Psychiatry and Unit of Treatment Resistant Psychosis, Department of Neuroscience, Reproductive Science and Dentistry, University Medical School of Naples "Federico II", Naples, Italy
| | - Christoph U Correll
- The Zucker Hillside Hospital, Department of Psychiatry, Northwell Health, Glen Oaks, NY, USA; Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Department of Psychiatry and Molecular Medicine, Hempstead, NY, USA; Charité Universitätsmedizin Berlin, Department of Child and Adolescent Psychiatry, Berlin, Germany
| |
Collapse
|
4
|
Chen M, Wang W, Song W, Qian W, Lin GN. Integrative Analysis Identified Key Schizophrenia Risk Factors from an Abnormal Behavior Mouse Gene Set. Life (Basel) 2021; 11:172. [PMID: 33672431 PMCID: PMC7927082 DOI: 10.3390/life11020172] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 02/10/2021] [Accepted: 02/20/2021] [Indexed: 01/12/2023] Open
Abstract
Schizophrenia (SCZ) is a severe chronic psychiatric illness with heterogeneous symptoms. However, the pathogenesis of SCZ is unclear, and the number of well-defined SCZ risk factors is limited. We hypothesized that an abnormal behavior (AB) gene set verified by mouse model experiments can be used to better understand SCZ risks. In this work, we carried out an integrative bioinformatics analysis to study two types of risk genes that are either differentially expressed (DEGs) in the case-control study data or carry reported SCZ genetic variants (MUTs). Next, we used RNA-Seq expression data from the hippocampus (HIPPO) and dorsolateral prefrontal cortex (DLPFC) to define the key genes affected by different types (DEGs and MUTs) in different brain regions (DLPFC and HIPPO): DLPFC-kDEG, DLPFC-kMUT, HIPPO-kDEG, and HIPPO-kMUT. The four hub genes (SHANK1, SHANK2, DLG4, and NLGN3) of the biological functionally enriched terms were strongly linked to SCZ via gene co-expression network analysis. Then, we observed that specific spatial expressions of DLPFC-kMUT and HIPPO-kMUT were convergent in the early stages and divergent in the later stages of development. In addition, all four types of key genes showed significantly larger average protein-protein interaction degrees than the background. Comparing the different cell types, the expression of four types of key genes showed specificity in different dimensions. Together, our results offer new insights into potential risk factors and help us understand the complexity and regional heterogeneity of SCZ.
Collapse
Affiliation(s)
- Miao Chen
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China; (M.C.); (W.W.); (W.S.); (W.Q.)
| | - Weidi Wang
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China; (M.C.); (W.W.); (W.S.); (W.Q.)
| | - Weicheng Song
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China; (M.C.); (W.W.); (W.S.); (W.Q.)
| | - Wei Qian
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China; (M.C.); (W.W.); (W.S.); (W.Q.)
| | - Guan Ning Lin
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China; (M.C.); (W.W.); (W.S.); (W.Q.)
- Engineering Research Center of Digital Medicine and Clinical Translational, Ministry of Education of China, Shanghai 200030, China
| |
Collapse
|
5
|
Wan L, Liu D, Xiao WB, Zhang BX, Yan XX, Luo ZH, Xiao B. Association of SHANK Family with Neuropsychiatric Disorders: An Update on Genetic and Animal Model Discoveries. Cell Mol Neurobiol 2021; 42:1623-1643. [PMID: 33595806 DOI: 10.1007/s10571-021-01054-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 02/02/2021] [Indexed: 12/14/2022]
Abstract
The Shank family proteins are enriched at the postsynaptic density (PSD) of excitatory glutamatergic synapses. They serve as synaptic scaffolding proteins and appear to play a critical role in the formation, maintenance and functioning of synapse. Increasing evidence from genetic association and animal model studies indicates a connection of SHANK genes defects with the development of neuropsychiatric disorders. In this review, we first update the current understanding of the SHANK family genes and their encoded protein products. We then denote the literature relating their alterations to the risk of neuropsychiatric diseases. We further review evidence from animal models that provided molecular insights into the biological as well as pathogenic roles of Shank proteins in synapses, and the potential relationship to the development of abnormal neurobehavioral phenotypes.
Collapse
Affiliation(s)
- Lily Wan
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Du Liu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.,Taikang Tongji Hospital, Wuhan, 430050, Hubei, China
| | - Wen-Biao Xiao
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Bo-Xin Zhang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Xiao-Xin Yan
- Department of Anatomy and Neurobiology, Central South University, Changsha, 410013, Hunan, China
| | - Zhao-Hui Luo
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
| | - Bo Xiao
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
| |
Collapse
|
6
|
Translating preclinical findings in clinically relevant new antipsychotic targets: focus on the glutamatergic postsynaptic density. Implications for treatment resistant schizophrenia. Neurosci Biobehav Rev 2019; 107:795-827. [DOI: 10.1016/j.neubiorev.2019.08.019] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2019] [Revised: 07/20/2019] [Accepted: 08/22/2019] [Indexed: 02/07/2023]
|
7
|
Qiu S, Li Y, Bai Y, Shi J, Cui H, Gu Y, Ren Y, Zhao Q, Zhang K, Lu M, Wang Y, Li Y, Zhong W, Zhu X, Liu Y, Cheng Y, Qiao Y, Liu Y. SHANK1 polymorphisms and SNP-SNP interactions among SHANK family: A possible cue for recognition to autism spectrum disorder in infant age. Autism Res 2019; 12:375-383. [PMID: 30629339 DOI: 10.1002/aur.2065] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2018] [Revised: 11/20/2018] [Accepted: 11/26/2018] [Indexed: 02/01/2023]
Abstract
Autism spectrum disorder (ASD) is a serious lifelong neurodevelopmental disorder. ASD is diagnosed for children at the age of two. ASD diagnosis, as early as possible, lays the foundation for treatment and much better prognosis. Notably, gene-based test is an inherent method to recognize the potential infants with ASD before the age of two. To investigate whether SHANK family contributes to ASD prediction, on the basis of our previous studies of SHANK2 and SHANK3, we further investigated associations between SHANK1 polymorphisms and ASD risk as well as SNP-SNP interactions among SHANK family. We enrolled 470 subjects (229 cases and 241 healthy controls) who were northeast Chinese Han. Four tag SNPs (rs73042561, rs3745521, rs4801846, and rs12461427) of SHANK1 were selected and genotyped. We used the SNPStats online analysis program to assess the associations between the four SNPs and ASD risk. The SNP-SNP interactions among SHANK family were analyzed using multifactor dimensionality reduction method. We found that the four SHANK1 SNPs were not associated with ASD risk in northeast Chinese Han population. There existed a strong synergistic interaction between rs11236697 [SHANK2] and rs74336682 [SHANK2], and moderate synergistic interactions (rs74336682 [SHANK2]-rs73042561 [SHANK1], rs11236697 [SHANK2]-rs77716438 [SHANK2], and rs11236697 [SHANK2]-rs75357229 [SHANK2]). These SHANK1 variants may not affect the susceptibility to ASD in Chinese Han population. SNP-SNP interactions in SHANK family may confer ASD risk. Autism Res 2019, 12: 375-383 © 2019 International Society for Autism Research, Wiley Periodicals, Inc. LAY SUMMARY: ASD is a serious lifelong neurodevelopmental disorder with strong genetic components. We investigated associations between SHANK1 polymorphisms and ASD risk as well as SNP-SNP interactions among SHANK family. Our results indicated that there exists no association between SHANK1 SNPs and ASD, and SNP-SNP interactions in SHANK family may confer ASD risk in the Northeast Han Chinese population. Future studies are needed to test more SHANK family SNPs in a large sample to demonstrate the associations.
Collapse
Affiliation(s)
- Shuang Qiu
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, Changchun, Jilin, China
| | - Yan Li
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, Changchun, Jilin, China
| | - Ye Bai
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, Changchun, Jilin, China
| | - Jikang Shi
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, Changchun, Jilin, China
| | - Heran Cui
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, Changchun, Jilin, China
| | - Yulu Gu
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, Changchun, Jilin, China
| | - Yaxuan Ren
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, Changchun, Jilin, China
| | - Qian Zhao
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, Changchun, Jilin, China
| | - Kaixin Zhang
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, Changchun, Jilin, China
| | - Meihan Lu
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, Changchun, Jilin, China
| | - Yihan Wang
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, Changchun, Jilin, China
| | - Yong Li
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, Changchun, Jilin, China
| | - Weijing Zhong
- Chunguang Rehabilitation Hospital, Changchun, Jilin, China
| | - Xiaojuan Zhu
- The Key Laboratory of Molecular Epigenetics of Ministry of Education, Institute of Cytology and Genetics, Northeast Normal University, Changchun, Jilin, China
| | - Yunkai Liu
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Yi Cheng
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Yichun Qiao
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, Changchun, Jilin, China
| | - Yawen Liu
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, Changchun, Jilin, China
| |
Collapse
|
8
|
Sungur AÖ, Redecker TM, Andres E, Dürichen W, Schwarting RKW, Del Rey A, Wöhr M. Reduced Efficacy of d-Amphetamine and 3,4-Methylenedioxymethamphetamine in Inducing Hyperactivity in Mice Lacking the Postsynaptic Scaffolding Protein SHANK1. Front Mol Neurosci 2018; 11:419. [PMID: 30505269 PMCID: PMC6250831 DOI: 10.3389/fnmol.2018.00419] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Accepted: 10/26/2018] [Indexed: 02/02/2023] Open
Abstract
Genetic defects in the three SH3 and multiple ankyrin repeat domains (SHANK) genes (SHANK1, SHANK2, and SHANK3) are associated with multiple major neuropsychiatric disorders, including autism spectrum disorder (ASD), schizophrenia (SCZ), and bipolar disorder (BPD). Psychostimulant-induced hyperactivity is a commonly applied paradigm to assess behavioral phenotypes related to BPD and considered to be the gold standard for modeling mania-like elevated drive in mouse models. Therefore, the goal of our present study was to test whether Shank1 plays a role in the behavioral effects of psychostimulants and whether this is associated with genotype-dependent neurochemical alterations. To this aim, male and female null mutant Shank1-/- mice were treated with d-amphetamine (AMPH; 2.5 mg/kg) and 3,4-methylenedioxymethamphetamine (MDMA, commonly known as ecstasy; 20 mg/kg), and psychostimulant-induced hyperactivity was compared to heterozygous Shank1+/- and wildtype Shank1+/+ littermate controls. Results show that Shank1-/- mice display reduced psychostimulant-induced hyperactivity, although psychostimulants robustly stimulated locomotor activity in littermate controls. Shank1 deletion effects emerged throughout development, were particularly prominent in adulthood, and seen in response to both psychostimulants, i.e., AMPH and MDMA. Specifically, while AMPH-induced hyperactivity was reduced but still detectable in Shank1-/- mice, MDMA-induced hyperactivity was robustly blocked and completely absent in Shank1-/- mice. Reduced efficacy of psychostimulants to stimulate hyperactivity in Shank1-/- mice might be associated with alterations in the neurochemical architecture in prefrontal cortex, nucleus accumbens, and hypothalamus. Our observation that psychostimulant-induced hyperactivity is reduced rather than enhanced in Shank1-/- mice clearly speaks against a behavioral phenotype with relevance to BPD. Lack of BPD-like phenotype is consistent with currently available human data linking mutations in SHANK2 and SHANK3 but not SHANK1 to BPD.
Collapse
Affiliation(s)
- A Özge Sungur
- Behavioral Neuroscience, Experimental and Biological Psychology, Philipps University of Marburg, Marburg, Germany.,Center for Mind, Brain and Behavior, Philipps University of Marburg, Marburg, Germany
| | - Tobias M Redecker
- Behavioral Neuroscience, Experimental and Biological Psychology, Philipps University of Marburg, Marburg, Germany
| | - Elena Andres
- Research Group Immunophysiology, Division of Neurophysiology, Institute of Physiology and Pathophysiology, Philipps University of Marburg, Marburg, Germany
| | - Wiebke Dürichen
- 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.,Center for Mind, Brain and Behavior, Philipps University of Marburg, Marburg, Germany
| | - Adriana Del Rey
- Research Group Immunophysiology, Division of Neurophysiology, Institute of Physiology and Pathophysiology, Philipps University of Marburg, Marburg, Germany
| | - Markus Wöhr
- Behavioral Neuroscience, Experimental and Biological Psychology, Philipps University of Marburg, Marburg, Germany.,Center for Mind, Brain and Behavior, Philipps University of Marburg, Marburg, Germany
| |
Collapse
|
9
|
Shen X, Yeung HT, Lai KO. Application of Human-Induced Pluripotent Stem Cells (hiPSCs) to Study Synaptopathy of Neurodevelopmental Disorders. Dev Neurobiol 2018; 79:20-35. [PMID: 30304570 DOI: 10.1002/dneu.22644] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 09/27/2018] [Accepted: 10/04/2018] [Indexed: 12/15/2022]
Abstract
Synapses are the basic structural and functional units for information processing and storage in the brain. Their diverse properties and functions ultimately underlie the complexity of human behavior. Proper development and maintenance of synapses are essential for normal functioning of the nervous system. Disruption in synaptogenesis and the consequent alteration in synaptic function have been strongly implicated to cause neurodevelopmental disorders such as autism spectrum disorders (ASDs) and schizophrenia (SCZ). The introduction of human-induced pluripotent stem cells (hiPSCs) provides a new path to elucidate disease mechanisms and potential therapies. In this review, we will discuss the advantages and limitations of using hiPSC-derived neurons to study synaptic disorders. Many mutations in genes encoding for proteins that regulate synaptogenesis have been identified in patients with ASDs and SCZ. We use Methyl-CpG binding protein 2 (MECP2), SH3 and multiple ankyrin repeat domains 3 (SHANK3) and Disrupted in schizophrenia 1 (DISC1) as examples to illustrate the promise of using hiPSCs as cellular models to elucidate the mechanisms underlying disease-related synaptopathy.
Collapse
Affiliation(s)
- Xuting Shen
- Faculty of Medicine, School of Biomedical Sciences, The University of Hong Kong, 21 Sassoon Road, Hong Kong, China
| | - Hoi Ting Yeung
- Faculty of Medicine, School of Biomedical Sciences, The University of Hong Kong, 21 Sassoon Road, Hong Kong, China
| | - Kwok-On Lai
- Faculty of Medicine, School of Biomedical Sciences, The University of Hong Kong, 21 Sassoon Road, Hong Kong, China.,State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, 21 Sassoon Road, Hong Kong, China
| |
Collapse
|
10
|
nArgBP2-SAPAP-SHANK, the core postsynaptic triad associated with psychiatric disorders. Exp Mol Med 2018; 50:1-9. [PMID: 29628500 PMCID: PMC5938024 DOI: 10.1038/s12276-017-0018-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Accepted: 11/29/2017] [Indexed: 11/23/2022] Open
Abstract
Despite the complex genetic architecture, a broad spectrum of psychiatric disorders can still be caused by mutation(s) in the same gene. These disorders are interrelated with overlapping causative mechanisms including variations in the interaction among the risk-associated proteins that may give rise to the specific spectrum of each disorder. Additionally, multiple lines of evidence implicate an imbalance between excitatory and inhibitory neuronal activity (E/I imbalance) as the shared key etiology. Thus, understanding the molecular mechanisms underlying E/I imbalance provides essential insight into the etiology of these disorders. One important class of candidate risk genes is the postsynaptic scaffolding proteins, such as nArgBP2, SAPAP, and SHANK that regulate the actin cytoskeleton in dendritic spines of excitatory synapses. This review will cover and discuss recent studies that examined how these proteins, especially nArgBP2, are associated with psychiatric disorders. Next, we propose a possibility that variations in the interaction among these proteins in a specific brain region might contribute to the onset of diverse phenotypes of psychiatric disorders. The assembly of scaffolding proteins, key regulators of many signaling pathways, found in the brain’s synapses underpin a diverse range of neuropsychiatric disorders. Sunghoe Chang and colleagues from Seoul National University, South Korea, review how these postsynaptic proteins regulate the cellular cytoskeleton in nerve cell protrusions to maintain the balance between excitatory and inhibitory inputs in the brain. They discuss how perturbations in three particular proteins can cause an imbalance in synaptic signals that leads to conditions such as bipolar disorder, schizophrenia and autism. The authors propose that these proteins form a “core scaffolding triad” and interact in different ways to cause different mental illnesses. Dysregulation of these proteins could explain how mutations in the same genes, depending on whether they boost or decrease gene expression, contribute to the onset of diverse psychiatric disorders.
Collapse
|
11
|
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: 2.6] [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
|
12
|
de Sena Cortabitarte A, Degenhardt F, Strohmaier J, Lang M, Weiss B, Roeth R, Giegling I, Heilmann-Heimbach S, Hofmann A, Rujescu D, Fischer C, Rietschel M, Nöthen MM, Rappold GA, Berkel S. Investigation of SHANK3 in schizophrenia. Am J Med Genet B Neuropsychiatr Genet 2017; 174:390-398. [PMID: 28371232 DOI: 10.1002/ajmg.b.32528] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Accepted: 01/03/2017] [Indexed: 12/21/2022]
Abstract
The postsynaptic scaffolding protein SHANK3 is essential for the normal function of glutamatergic synapses in the brain. Emerging evidence suggests that impaired plasticity of glutamatergic synapses contributes to the pathology of schizophrenia (SCZ). To investigate whether variants in the SHANK3 gene contribute to the etiology of SCZ, we sequenced SHANK3 in 500 affected individuals (cohort C1). In total, we identified 48 variants and compared them to European controls from the 1000 Genomes Project and the Exome Variant Server. Five variants showed significant differences in frequencies between patients and controls. We were able to follow three of them up in an independent cohort (C2) comprising 993 SCZ patients and 932 German controls. We could not confirm an association for three of these variants (rs140201628, rs1557620, and rs61729471). Two rare variants with predicted functional relevance were identified in further SCZ individuals of cohort C1: c.3032G>T (p.G1011V) and c.*27C>T. The latter variant was found in one additional SCZ individual and the p.G1011V variant was identified in two additional SCZ individuals from cohort C2. The p.G1011V variant was the most interesting variant in our study; together with previous studies this variant has been identified in 4 out of 1,524 SCZ patients and in 4 out of 2,147 individuals with autism spectrum disorder (ASD), but not in 2468 European Sanger-sequenced controls. Therefore, we consider this variant a promising candidate variant for follow-up studies in larger samples and functional investigations. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Ana de Sena Cortabitarte
- Department of Human Molecular Genetics, Institute of Human Genetics, University of Heidelberg, Heidelberg, Germany
| | - Franziska Degenhardt
- Department of Genomics, Life & Brain Center, University of Bonn, Bonn, Germany.,Institute of Human Genetics, University of Bonn, Bonn, Germany
| | - Jana Strohmaier
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health Mannheim, Faculty of Medicine Mannheim, University of Heidelberg, Mannheim, Germany
| | - Maren Lang
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health Mannheim, Faculty of Medicine Mannheim, University of Heidelberg, Mannheim, Germany
| | - Birgit Weiss
- Department of Human Molecular Genetics, Institute of Human Genetics, University of Heidelberg, Heidelberg, Germany
| | - Ralph Roeth
- Department of Human Molecular Genetics, Institute of Human Genetics, University of Heidelberg, Heidelberg, Germany
| | - Ina Giegling
- Department of Psychiatry, University Hospital Halle (Saale), Halle, Germany
| | | | - Andrea Hofmann
- Department of Genomics, Life & Brain Center, University of Bonn, Bonn, Germany
| | - Dan Rujescu
- Department of Psychiatry, University Hospital Halle (Saale), Halle, Germany
| | - Christine Fischer
- Institute of Human Genetics, University of Heidelberg, Heidelberg, Germany
| | - Marcella Rietschel
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health Mannheim, Faculty of Medicine Mannheim, University of Heidelberg, Mannheim, Germany.,Department of Psychiatry, University of Bonn, Bonn, Germany
| | - Markus M Nöthen
- Department of Genomics, Life & Brain Center, University of Bonn, Bonn, Germany.,Institute of Human Genetics, University of Bonn, Bonn, Germany
| | - Gudrun A Rappold
- Department of Human Molecular Genetics, Institute of Human Genetics, University of Heidelberg, Heidelberg, Germany.,Interdisciplinary Center for Neurosciences (IZN), University of Heidelberg, Heidelberg, Germany
| | - Simone Berkel
- Department of Human Molecular Genetics, Institute of Human Genetics, University of Heidelberg, Heidelberg, Germany
| |
Collapse
|
13
|
Sungur AÖ, Jochner MCE, Harb H, Kılıç A, Garn H, Schwarting RKW, Wöhr M. Aberrant cognitive phenotypes and altered hippocampal BDNF expression related to epigenetic modifications in mice lacking the post-synaptic scaffolding protein SHANK1: Implications for autism spectrum disorder. Hippocampus 2017; 27:906-919. [PMID: 28500650 DOI: 10.1002/hipo.22741] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Revised: 04/05/2017] [Accepted: 05/03/2017] [Indexed: 12/29/2022]
Abstract
Autism spectrum disorder (ASD) is a class of neurodevelopmental disorders characterized by persistent deficits in social communication/interaction, together with restricted/repetitive patterns of behavior. ASD is among the most heritable neuropsychiatric conditions, and while available evidence points to a complex set of genetic factors, the SHANK gene family has emerged as one of the most promising candidates. Here, we assessed ASD-related phenotypes with particular emphasis on social behavior and cognition in Shank1 mouse mutants in comparison to heterozygous and wildtype littermate controls across development in both sexes. While social approach behavior was evident in all experimental conditions and social recognition was only mildly affected by genotype, Shank1-/- null mutant mice were severely impaired in object recognition memory. This effect was particularly prominent in juveniles, not due to impairments in object discrimination, and replicated in independent mouse cohorts. At the neurobiological level, object recognition deficits were paralleled by increased brain-derived neurotrophic factor (BDNF) protein expression in the hippocampus of Shank1-/- mice; yet BDNF levels did not differ under baseline conditions. We therefore investigated changes in the epigenetic regulation of hippocampal BDNF expression and detected an enrichment of histone H3 acetylation at the Bdnf promoter1 in Shank1-/- mice, consistent with increased learning-associated BDNF. Together, our findings indicate that Shank1 deletions lead to an aberrant cognitive phenotype characterized by severe impairments in object recognition memory and increased hippocampal BDNF levels, possibly due to epigenetic modifications. This result supports the link between ASD and intellectual disability, and suggests epigenetic regulation as a potential therapeutic target.
Collapse
Affiliation(s)
- A Özge Sungur
- Behavioral Neuroscience, Experimental and Biological Psychology, Philipps-University of Marburg, Marburg, Germany
| | - Magdalena C E Jochner
- Behavioral Neuroscience, Experimental and Biological Psychology, Philipps-University of Marburg, Marburg, Germany
| | - Hani Harb
- Institute of Laboratory Medicine and Pathobiochemistry-Molecular Diagnostics, Philipps-University of Marburg, Marburg, Germany
| | - Ayşe Kılıç
- Institute of Laboratory Medicine and Pathobiochemistry-Molecular Diagnostics, Philipps-University of Marburg, Marburg, Germany
| | - Holger Garn
- Institute of Laboratory Medicine and Pathobiochemistry-Molecular Diagnostics, 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
|
14
|
Emerging Synaptic Molecules as Candidates in the Etiology of Neurological Disorders. Neural Plast 2017; 2017:8081758. [PMID: 28331639 PMCID: PMC5346360 DOI: 10.1155/2017/8081758] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Accepted: 02/06/2017] [Indexed: 01/06/2023] Open
Abstract
Synapses are complex structures that allow communication between neurons in the central nervous system. Studies conducted in vertebrate and invertebrate models have contributed to the knowledge of the function of synaptic proteins. The functional synapse requires numerous protein complexes with specialized functions that are regulated in space and time to allow synaptic plasticity. However, their interplay during neuronal development, learning, and memory is poorly understood. Accumulating evidence links synapse proteins to neurodevelopmental, neuropsychiatric, and neurodegenerative diseases. In this review, we describe the way in which several proteins that participate in cell adhesion, scaffolding, exocytosis, and neurotransmitter reception from presynaptic and postsynaptic compartments, mainly from excitatory synapses, have been associated with several synaptopathies, and we relate their functions to the disease phenotype.
Collapse
|
15
|
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: 74] [Impact Index Per Article: 9.3] [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
|
16
|
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
|
17
|
Peykov S, Berkel S, Schoen M, Weiss K, Degenhardt F, Strohmaier J, Weiss B, Proepper C, Schratt G, Nöthen MM, Boeckers TM, Rietschel M, Rappold GA. Identification and functional characterization of rare SHANK2 variants in schizophrenia. Mol Psychiatry 2015; 20:1489-98. [PMID: 25560758 PMCID: PMC4653611 DOI: 10.1038/mp.2014.172] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2014] [Revised: 10/30/2014] [Accepted: 11/12/2014] [Indexed: 12/20/2022]
Abstract
Recent genetic data on schizophrenia (SCZ) have suggested that proteins of the postsynaptic density of excitatory synapses have a role in its etiology. Mutations in the three SHANK genes encoding for postsynaptic scaffolding proteins have been shown to represent risk factors for autism spectrum disorders and other neurodevelopmental disorders. To address if SHANK2 variants are associated with SCZ, we sequenced SHANK2 in 481 patients and 659 unaffected individuals. We identified a significant increase in the number of rare (minor allele frequency<1%) SHANK2 missense variants in SCZ individuals (6.9%) compared with controls (3.9%, P=0.039). Four out of fifteen non-synonymous variants identified in the SCZ cohort (S610Y, R958S, P1119T and A1731S) were selected for functional analysis. Overexpression and knockdown-rescue experiments were carried out in cultured primary hippocampal neurons with a major focus on the analysis of morphological changes. Furthermore, the effect on actin polymerization in fibroblast cell lines was investigated. All four variants revealed functional impairment to various degrees, as a consequence of alterations in spine volume and clustering at synapses and an overall loss of presynaptic contacts. The A1731S variant was identified in four unrelated SCZ patients (0.83%) but not in any of the sequenced controls and public databases (P=4.6 × 10(-5)). Patients with the A1731S variant share an early prodromal phase with an insidious onset of psychiatric symptoms. A1731S overexpression strongly decreased the SHANK2-Bassoon-positive synapse number and diminished the F/G-actin ratio. Our results strongly suggest a causative role of rare SHANK2 variants in SCZ and underline the contribution of SHANK2 gene mutations in a variety of neuropsychiatric disorders.
Collapse
Affiliation(s)
- S Peykov
- Department of Human Molecular Genetics, Heidelberg University, Heidelberg, Germany
| | - S Berkel
- Department of Human Molecular Genetics, Heidelberg University, Heidelberg, Germany
| | - M Schoen
- Institute of Anatomy and Cell Biology, Ulm University, Ulm, Germany
| | - K Weiss
- Institute of Physiological Chemistry, Phillipps-University Marburg, Marburg, Germany
| | - F Degenhardt
- Institute of Human Genetics, Bonn University, Bonn, Germany
| | - J Strohmaier
- Department of Genomics, Life and Brain Center, University of Bonn, Bonn, Germany
| | - B Weiss
- Department of Human Molecular Genetics, Heidelberg University, Heidelberg, Germany
| | - C Proepper
- Institute of Anatomy and Cell Biology, Ulm University, Ulm, Germany
| | - G Schratt
- Institute of Physiological Chemistry, Phillipps-University Marburg, Marburg, Germany
| | - M M Nöthen
- Institute of Human Genetics, Bonn University, Bonn, Germany,Department of Genomics, Life and Brain Center, University of Bonn, Bonn, Germany
| | - T M Boeckers
- Institute of Anatomy and Cell Biology, Ulm University, Ulm, Germany
| | - M Rietschel
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Mannheim, Germany
| | - G A Rappold
- Department of Human Molecular Genetics, Heidelberg University, Heidelberg, Germany,Interdisciplinary Center of Neurosciences (IZN), Heidelberg University, Heidelberg, Germany,Department of Human Molecular Genetics, Institute of Human Genetics, Im Neuenheimer Feld 366, Heidelberg 69120, Germany. E-mail:
| |
Collapse
|
18
|
Sungur AÖ, Schwarting RK, Wöhr M. Early communication deficits in theShank1knockout mouse model for autism spectrum disorder: Developmental aspects and effects of social context. Autism Res 2015; 9:696-709. [DOI: 10.1002/aur.1564] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Revised: 07/07/2015] [Accepted: 08/21/2015] [Indexed: 11/05/2022]
Affiliation(s)
- A. Özge Sungur
- Behavioral Neuroscience, Experimental and Biological Psychology; Philipps-University of Marburg; Gutenbergstr. 18 D-35032 Germany
| | - Rainer K.W. Schwarting
- Behavioral Neuroscience, Experimental and Biological Psychology; Philipps-University of Marburg; Gutenbergstr. 18 D-35032 Germany
| | - Markus Wöhr
- Behavioral Neuroscience, Experimental and Biological Psychology; Philipps-University of Marburg; Gutenbergstr. 18 D-35032 Germany
| |
Collapse
|
19
|
Shifting towards a model of mGluR5 dysregulation in schizophrenia: Consequences for future schizophrenia treatment. Neuropharmacology 2015; 115:73-91. [PMID: 26349010 DOI: 10.1016/j.neuropharm.2015.08.003] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Revised: 08/02/2015] [Accepted: 08/03/2015] [Indexed: 12/22/2022]
Abstract
Metabotropic glutamate receptor subtype 5 (mGluR5), encoded by the GRM5 gene, represents a compelling novel drug target for the treatment of schizophrenia. mGluR5 is a postsynaptic G-protein coupled glutamate receptor strongly linked with several critical cellular processes that are reported to be disrupted in schizophrenia. Accordingly, mGluR5 positive allosteric modulators show encouraging therapeutic potential in preclinical schizophrenia models, particularly for the treatment of cognitive dysfunctions against which currently available therapeutics are largely ineffective. More work is required to support the progression of mGluR5-targeting drugs into the clinic for schizophrenia treatment, although some obstacles may be overcome by comprehensively understanding how mGluR5 itself is involved in the neurobiology of the disorder. Several processes that are necessary for the regulation of mGluR5 activity have been identified, but not examined, in the context of schizophrenia. These processes include protein-protein interactions, dimerisation, subcellular trafficking, the impact of genetic variability or mutations on protein function, as well as epigenetic, post-transcriptional and post-translational processes. It is essential to understand these aspects of mGluR5 to determine whether they are affected in schizophrenia pathology, and to assess the consequences of mGluR5 dysfunction for the future use of mGluR5-based drugs. Here, we summarise the known processes that regulate mGluR5 and those that have already been studied in schizophrenia, and discuss the consequences of this dysregulation for current mGluR5 pharmacological strategies. This article is part of the Special Issue entitled 'Metabotropic Glutamate Receptors, 5 years on'.
Collapse
|
20
|
Sala C, Vicidomini C, Bigi I, Mossa A, Verpelli C. Shank synaptic scaffold proteins: keys to understanding the pathogenesis of autism and other synaptic disorders. J Neurochem 2015; 135:849-58. [PMID: 26338675 DOI: 10.1111/jnc.13232] [Citation(s) in RCA: 117] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Revised: 06/24/2015] [Accepted: 06/25/2015] [Indexed: 02/01/2023]
Abstract
Shank/ProSAP proteins are essential to synaptic formation, development, and function. Mutations in the family of SHANK genes are strongly associated with autism spectrum disorders (ASD) and other neurodevelopmental and neuropsychiatric disorders, such as intellectual disability (ID), and schizophrenia. Thus, the term 'Shankopathies' identifies a number of neuronal diseases caused by alteration of Shank protein expression leading to abnormal synaptic development. With this review we want to summarize the major genetic, molecular, behavior and electrophysiological studies that provide new clues into the function of Shanks and pave the way for the discovery of new therapeutic drugs targeted to treat patients with SHANK mutations and also patients affected by other neurodevelopmental and neuropsychiatric disorders. Shank/ProSAP proteins are essential to synaptic formation, development, and function. Mutations in the family of SHANK genes are strongly associated with autism spectrum disorders (ASD) and other neurodevelopmental and neuropsychiatric disorders, such as intellectual disability (ID), and schizophrenia (SCZ). With this review we want to summarize the major genetic, molecular, behavior and electrophysiological studies that provide new clues into the function of Shanks and pave the way for the discovery of new therapeutic drugs targeted to treat patients with SHANK mutations.
Collapse
Affiliation(s)
- Carlo Sala
- CNR Neuroscience Institute and Department of Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Cinzia Vicidomini
- CNR Neuroscience Institute and Department of Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Ilaria Bigi
- CNR Neuroscience Institute and Department of Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Adele Mossa
- CNR Neuroscience Institute and Department of Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Chiara Verpelli
- CNR Neuroscience Institute and Department of Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| |
Collapse
|
21
|
Kang J, Park H, Kim E. IRSp53/BAIAP2 in dendritic spine development, NMDA receptor regulation, and psychiatric disorders. Neuropharmacology 2015; 100:27-39. [PMID: 26275848 DOI: 10.1016/j.neuropharm.2015.06.019] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Revised: 06/26/2015] [Accepted: 06/28/2015] [Indexed: 01/08/2023]
Abstract
IRSp53 (also known as BAIAP2) is a multi-domain scaffolding and adaptor protein that has been implicated in the regulation of membrane and actin dynamics at subcellular structures, including filopodia and lamellipodia. Accumulating evidence indicates that IRSp53 is an abundant component of the postsynaptic density at excitatory synapses and an important regulator of actin-rich dendritic spines. In addition, IRSp53 has been implicated in diverse psychiatric disorders, including autism spectrum disorders, schizophrenia, and attention deficit/hyperactivity disorder. Mice lacking IRSp53 display enhanced NMDA (N-methyl-d-aspartate) receptor function accompanied by social and cognitive deficits, which are reversed by pharmacological suppression of NMDA receptor function. These results suggest the hypothesis that defective actin/membrane modulation in IRSp53-deficient dendritic spines may lead to social and cognitive deficits through NMDA receptor dysfunction. This article is part of the Special Issue entitled 'Synaptopathy--from Biology to Therapy'.
Collapse
Affiliation(s)
- Jaeseung Kang
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 305-701, South Korea
| | - Haram Park
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 305-701, South Korea
| | - Eunjoon Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 305-701, South Korea; Center for Synaptic Brain Dysfunctions, Institute for Basic Science (IBS), Daejeon 305-701, South Korea.
| |
Collapse
|
22
|
Washbourne P. Synapse assembly and neurodevelopmental disorders. Neuropsychopharmacology 2015; 40:4-15. [PMID: 24990427 PMCID: PMC4262893 DOI: 10.1038/npp.2014.163] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2014] [Revised: 06/23/2014] [Accepted: 06/26/2014] [Indexed: 12/31/2022]
Abstract
In this review we examine the current understanding of how genetic deficits associated with neurodevelopmental disorders may impact synapse assembly. We then go on to discuss how the critical periods for these genetic deficits will shape the nature of future clinical interventions.
Collapse
Affiliation(s)
- Philip Washbourne
- Institute of Neuroscience, University of Oregon, Eugene, OR, USA,Institute of Neuroscience, University of Oregon, 1254 University of Oregon, Eugene, OR 97403, USA, Tel: +1 541 346 4138, Fax: +1 541 346 4548, E-mail:
| |
Collapse
|
23
|
Chow F, Gong Y, Lippa CF. The Potential Role of Insulin on the Shank-Postsynaptic Platform in Neurodegenerative Diseases Involving Cognition. Am J Alzheimers Dis Other Demen 2014; 29:303-10. [PMID: 24421411 PMCID: PMC10852640 DOI: 10.1177/1533317513518645] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Loss of synaptic function is critical in the pathogenesis of Alzheimer's disease (AD) and other central nervous system (CNS) degenerations. A promising candidate in the regulation of synaptic function is Shank, a protein that serves as a scaffold for excitatory synaptic receptors and proteins. Loss of Shank alters structure and function of the postsynaptic density (PSD). Shank proteins are associated with N-methyl-d-aspartate and α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor loss at the PSD in AD; mutations in Shank also lead to autism spectrum disorders (ASDs) and schizophrenia, both of which affect cognition, suggesting that Shank may play a common pathologic role in AD, ASD, and schizophrenia. Shank protein directly associates with insulin receptor substrate protein p53 in PSD. Insulin and insulin sensitizers have been used in clinical trials for these diseases; this suggests that insulin signals may alter protein homeostasis at the shank-postsynaptic platform in PSDs; insulin could improve the function of synapses in these diseases.
Collapse
Affiliation(s)
- Frances Chow
- Department of Neurology, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Yuesong Gong
- Department of Neurology, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Carol F Lippa
- Department of Neurology, Drexel University College of Medicine, Philadelphia, PA, USA
| |
Collapse
|
24
|
Iasevoli F, Tomasetti C, Buonaguro EF, de Bartolomeis A. The glutamatergic aspects of schizophrenia molecular pathophysiology: role of the postsynaptic density, and implications for treatment. Curr Neuropharmacol 2014; 12:219-38. [PMID: 24851087 PMCID: PMC4023453 DOI: 10.2174/1570159x12666140324183406] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2013] [Revised: 02/14/2014] [Accepted: 03/14/2014] [Indexed: 01/23/2023] Open
Abstract
Schizophrenia is one of the most debilitating psychiatric diseases with a lifetime prevalence of approximately
1%. Although the specific molecular underpinnings of schizophrenia are still unknown, evidence has long linked its
pathophysiology to postsynaptic abnormalities.
The postsynaptic density (PSD) is among the molecular structures suggested to be potentially involved in schizophrenia.
More specifically, the PSD is an electron-dense thickening of glutamatergic synapses, including ionotropic and
metabotropic glutamate receptors, cytoskeletal and scaffolding proteins, and adhesion and signaling molecules. Being
implicated in the postsynaptic signaling of multiple neurotransmitter systems, mostly dopamine and glutamate, the PSD
constitutes an ideal candidate for studying dopamine-glutamate disturbances in schizophrenia. Recent evidence suggests
that some PSD proteins, such as PSD-95, Shank, and Homer are implicated in severe behavioral disorders, including
schizophrenia. These findings, further corroborated by genetic and animal studies of schizophrenia, offer new insights for
the development of pharmacological strategies able to overcome the limitations in terms of efficacy and side effects of
current schizophrenia treatment. Indeed, PSD proteins are now being considered as potential molecular targets against this
devastating illness.
The current paper reviews the most recent hypotheses on the molecular mechanisms underlying schizophrenia
pathophysiology. First, we review glutamatergic dysfunctions in schizophrenia and we provide an update on postsynaptic
molecules involvement in schizophrenia pathophysiology by addressing both human and animal studies. Finally, the
possibility that PSD proteins may represent potential targets for new molecular interventions in psychosis will be
discussed.
Collapse
Affiliation(s)
- Felice Iasevoli
- Department of Neuroscience, Reproductive and Odontostomatological Sciences - University "Federico II", Naples, Italy
| | - Carmine Tomasetti
- Department of Neuroscience, Reproductive and Odontostomatological Sciences - University "Federico II", Naples, Italy
| | - Elisabetta F Buonaguro
- Department of Neuroscience, Reproductive and Odontostomatological Sciences - University "Federico II", Naples, Italy
| | - Andrea de Bartolomeis
- Department of Neuroscience, Reproductive and Odontostomatological Sciences - University "Federico II", Naples, Italy
| |
Collapse
|
25
|
Fuxe K, Borroto-Escuela DO, Tarakanov AO, Romero-Fernandez W, Ferraro L, Tanganelli S, Perez-Alea M, Di Palma M, Agnati LF. Dopamine D2 heteroreceptor complexes and their receptor-receptor interactions in ventral striatum: novel targets for antipsychotic drugs. PROGRESS IN BRAIN RESEARCH 2014; 211:113-39. [PMID: 24968778 DOI: 10.1016/b978-0-444-63425-2.00005-2] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
This review is focused on the D2 heteroreceptor complexes within the ventral striatum with their receptor-receptor interactions and relevance for the treatment of schizophrenia. A "guide-and-clasp" manner for receptor-receptor interactions is proposed where "adhesive guides" may be amino acid triplet homologies, which were determined for different kinds of D2 heteroreceptor complexes. The first putative D2 heteroreceptor complex to be discovered in relation to schizophrenia was the A2A-D2 heteroreceptor complex where antagonistic A2A-D2 receptor-receptor interactions were demonstrated after A2A agonist treatment in the ventral striatum. The A2A agonist CGS 21680 with atypical antipsychotic properties may at least in part act by increasing β-arrestin2 signaling over the D2 protomer in the A2A-D2 heteroreceptor complex in the ventral striatum. The antagonistic NTS1-D2 interactions in the NTS1-D2 heteroreceptor complex in the ventral striatum are proposed as one molecular mechanism for the potential antipsychotic effects of NT. Indications were obtained that the psychotic actions of the 5-HT2AR hallucinogens LSD and DOI can involve enhancement of D2R protomer signaling via a biased agonist action at the 5-HT2A protomer in the D2-5-HT2A heteroreceptor complex in the ventral striatum. Facilitatory allosteric D2likeR-OTR interactions in heteroreceptor complexes in nucleus accumbens may have a role in social and emotional behaviors. By blocking the D2 protomers of these heteroreceptor complexes, antipsychotics can fail to reduce the negative symptoms of schizophrenia. The discovery of different types of D2 heteroreceptor complexes gives an increased understanding of molecular mechanisms involved in causing schizophrenia and new strategies for its treatment and understanding the side effects of antipsychotics.
Collapse
Affiliation(s)
- Kjell Fuxe
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden.
| | | | - Alexander O Tarakanov
- Russian Academy of Sciences, St Petersburg Institute for Informatics and Automatation, St. Petersburg, Russia
| | | | - Luca Ferraro
- Department of Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy
| | - Sergio Tanganelli
- Department of Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy
| | - Mileidys Perez-Alea
- Department of Pathology, Hospital Universitari Vall d'Hebron, Barcelona, Spain
| | - Michael Di Palma
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden; Department of Earth, Life and Environmental Sciences, Section of Physiology, Campus Scientifico 'Enrico Mattei', Urbino, Italy
| | | |
Collapse
|
26
|
Grabrucker S, Proepper C, Mangus K, Eckert M, Chhabra R, Schmeisser MJ, Boeckers TM, Grabrucker AM. The PSD protein ProSAP2/Shank3 displays synapto-nuclear shuttling which is deregulated in a schizophrenia-associated mutation. Exp Neurol 2013; 253:126-37. [PMID: 24382453 DOI: 10.1016/j.expneurol.2013.12.015] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2013] [Revised: 12/12/2013] [Accepted: 12/20/2013] [Indexed: 12/11/2022]
Abstract
Recently, mutations in ProSAP2/Shank3 have been discovered as one of the genetic factors for schizophrenia (SCZ). Here, we show that the postsynaptic density protein ProSAP2/Shank3 undergoes activity dependent synapse-to-nucleus shuttling in hippocampal neurons. Our study shows that the de novo mutation (R1117X) in ProSAP2/Shank3 that was identified in a patient with SCZ leads to an accumulation of mutated ProSAP2/Shank3 within the nucleus independent of synaptic activity. Furthermore, we identified novel nuclear ProSAP2/Shank3 interaction partners. Nuclear localization of mutated ProSAP2/Shank3 alters transcription of several genes, among them already identified genetic risk factors for SCZ such as Synaptotagmin 1 and LRRTM1. Comparing the SCZ mutation of ProSAP2/Shank3 to the knockdown of ProSAP2/Shank3 we found some shared features such as reduced synaptic density in neuronal cultures. However, hippocampal neurons expressing the ProSAP2/Shank3 SCZ mutation furthermore show altered E/I ratio and reduced dendritic branching. Thus, we conclude that the uncoupling of ProSAP2/Shank3 nuclear shuttling from synaptic activity may represent a molecular mechanism that contributes to the pathology of SCZ in patients with mutations in ProSAP2/Shank3.
Collapse
Affiliation(s)
- Stefanie Grabrucker
- WG Molecular Analysis of Synaptopathies, Neurology Dept., Neurocenter of Ulm University, Albert-Einstein Allee 11, 89081 Ulm, Germany; Institute for Anatomy and Cell Biology, Ulm University, Albert-Einstein Allee 11, 89081 Ulm, Germany
| | - Christian Proepper
- Institute for Anatomy and Cell Biology, Ulm University, Albert-Einstein Allee 11, 89081 Ulm, Germany
| | - Katharina Mangus
- WG Molecular Analysis of Synaptopathies, Neurology Dept., Neurocenter of Ulm University, Albert-Einstein Allee 11, 89081 Ulm, Germany
| | - Matti Eckert
- WG Molecular Analysis of Synaptopathies, Neurology Dept., Neurocenter of Ulm University, Albert-Einstein Allee 11, 89081 Ulm, Germany
| | - Resham Chhabra
- WG Molecular Analysis of Synaptopathies, Neurology Dept., Neurocenter of Ulm University, Albert-Einstein Allee 11, 89081 Ulm, Germany
| | - Michael J Schmeisser
- Institute for Anatomy and Cell Biology, Ulm University, Albert-Einstein Allee 11, 89081 Ulm, Germany
| | - Tobias M Boeckers
- Institute for Anatomy and Cell Biology, Ulm University, Albert-Einstein Allee 11, 89081 Ulm, Germany
| | - Andreas M Grabrucker
- WG Molecular Analysis of Synaptopathies, Neurology Dept., Neurocenter of Ulm University, Albert-Einstein Allee 11, 89081 Ulm, Germany; Institute for Anatomy and Cell Biology, Ulm University, Albert-Einstein Allee 11, 89081 Ulm, Germany.
| |
Collapse
|
27
|
Carbonetto S. A blueprint for research on Shankopathies: a view from research on autism spectrum disorder. Dev Neurobiol 2013; 74:85-112. [PMID: 24218108 DOI: 10.1002/dneu.22150] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2013] [Accepted: 11/06/2013] [Indexed: 01/21/2023]
Abstract
Autism spectrum disorders (ASD) are associated with mutations in a host of genes including a number that function in synaptic transmission. Phelan McDermid syndrome involves mutations in SHANK3 which encodes a protein that forms a scaffold for glutamate receptors at the synapse. SHANK3 is one of the genes that underpins the synaptic hypothesis for ASD. We discuss this hypothesis with a view to the broader context of ASD and with special emphasis on highly penetrant genetic disorders including Shankopathies. We propose a blueprint for near and longer-term goals for fundamental and translational research on Shankopathies.
Collapse
Affiliation(s)
- Salvatore Carbonetto
- Centre for Research in Neuroscience, Department of Neurology, McGill University Health Centre, Montreal, Quebec, H3G1A4, Canada
| |
Collapse
|
28
|
Guilmatre A, Huguet G, Delorme R, Bourgeron T. The emerging role of SHANK genes in neuropsychiatric disorders. Dev Neurobiol 2013; 74:113-22. [PMID: 24124131 DOI: 10.1002/dneu.22128] [Citation(s) in RCA: 177] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2013] [Accepted: 08/28/2013] [Indexed: 11/09/2022]
Abstract
The genetic heterogeneity of neuropsychiatric disorders is high, but some pathways emerged, notably synaptic functioning. A large number of mutations have been described in genes such as neuroligins, neurexins, and SHANK that play a role in the formation and the maintenance of synapses. This review focuses on the disorders associated with mutations in SHANK3 and the other members of its family, SHANK1 and SHANK2. SHANKs are scaffolding proteins of the postsynaptic density of glutamatergic synapses. SHANK3 has been described in the Phelan-McDermid syndrome (PMS), but also in autism spectrum disorders (ASD) and schizophrenia associated to moderate to severe intellectual disability (ID) and poor language. The evolution of patients with PMS includes symptoms of bipolar disorder and regression. SHANK2 has been identified in patients with ASD with mild to severe ID. SHANK1 has been associated with high-functioning autism in male patients, while carrier females only display anxiety and shyness. Finally, based on neuropathological findings in animal models and patients, a possible role of SHANK in Alzheimer's disease is discussed. Altogether, this review describes the clinical trajectories associated with different mutations of the SHANK genes and provides information to further investigate the role of the SHANK genes in neuropsychiatric disorders.
Collapse
Affiliation(s)
- Audrey Guilmatre
- Human Genetics and Cognitive Functions Unit, Institut Pasteur, Paris, France; CNRS URA 2182 'Genes, Synapses and Cognition,' Institut Pasteur, Paris, France; Human Genetics and Cognitive Functions, University Paris Diderot, Sorbonne Paris Cité, Paris, France
| | | | | | | |
Collapse
|
29
|
de Bartolomeis A, Latte G, Tomasetti C, Iasevoli F. Glutamatergic postsynaptic density protein dysfunctions in synaptic plasticity and dendritic spines morphology: relevance to schizophrenia and other behavioral disorders pathophysiology, and implications for novel therapeutic approaches. Mol Neurobiol 2013; 49:484-511. [PMID: 23999870 DOI: 10.1007/s12035-013-8534-3] [Citation(s) in RCA: 101] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2013] [Accepted: 08/13/2013] [Indexed: 02/06/2023]
Abstract
Emerging researches point to a relevant role of postsynaptic density (PSD) proteins, such as PSD-95, Homer, Shank, and DISC-1, in the pathophysiology of schizophrenia and autism spectrum disorders. The PSD is a thickness, detectable at electronic microscopy, localized at the postsynaptic membrane of glutamatergic synapses, and made by scaffolding proteins, receptors, and effector proteins; it is considered a structural and functional crossroad where multiple neurotransmitter systems converge, including the dopaminergic, serotonergic, and glutamatergic ones, which are all implicated in the pathophysiology of psychosis. Decreased PSD-95 protein levels have been reported in postmortem brains of schizophrenia patients. Variants of Homer1, a key PSD protein for glutamate signaling, have been associated with schizophrenia symptoms severity and therapeutic response. Mutations in Shank gene have been recognized in autism spectrum disorder patients, as well as reported to be associated to behaviors reminiscent of schizophrenia symptoms when expressed in genetically engineered mice. Here, we provide a critical appraisal of PSD proteins role in the pathophysiology of schizophrenia and autism spectrum disorders. Then, we discuss how antipsychotics may affect PSD proteins in brain regions relevant to psychosis pathophysiology, possibly by controlling synaptic plasticity and dendritic spine rearrangements through the modulation of glutamate-related targets. We finally provide a framework that may explain how PSD proteins might be useful candidates to develop new therapeutic approaches for schizophrenia and related disorders in which there is a need for new biological treatments, especially against some symptom domains, such as negative symptoms, that are poorly affected by current antipsychotics.
Collapse
Affiliation(s)
- Andrea de Bartolomeis
- Laboratory of Molecular and Translational Psychiatry, Unit of Treatment Resistant Psychosis, Department of Neuroscience, Reproductive and Odontostomatologic Sciences, Section of Psychiatry, University School of Medicine "Federico II", Via Pansini 5, 80131, Naples, Italy,
| | | | | | | |
Collapse
|
30
|
Gao C, Tronson NC, Radulovic J. Modulation of behavior by scaffolding proteins of the post-synaptic density. Neurobiol Learn Mem 2013; 105:3-12. [PMID: 23701866 DOI: 10.1016/j.nlm.2013.04.014] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2013] [Revised: 04/05/2013] [Accepted: 04/09/2013] [Indexed: 12/23/2022]
Abstract
Scaffolding proteins of the neuronal post-synaptic density (PSD) are principal organizers of glutamatergic neurotransmission that bring together glutamate receptors and signaling molecules at discrete synaptic locations. Genetic alterations of individual PSD scaffolds therefore disrupt the function of entire multiprotein modules rather than a single glutamatergic mechanism, and thus induce a range of molecular and structural abnormalities in affected neurons. Despite such broad molecular consequences, knockout, knockdown, or knockin of glutamate receptor scaffolds typically affect a subset of specific behaviors and thereby mold and specialize the actions of the ubiquitous glutamatergic neurotransmitter system. Approaches designed to control the function of neuronal scaffolds may therefore have high potential to restore behavioral morbidities and comorbidities in patients with psychiatric disorders. Here we summarize a series of experiments with genetically modified mice revealing the roles of main N-methyl-d-aspartate (NMDA) and group I metabotropic glutamate (mGluR1/5) receptor scaffolds in behavior, discuss the clinical implications of the findings, and propose future research directions.
Collapse
Affiliation(s)
- Can Gao
- Jiangsu Key Laboratory of Anesthesiology, Xuzhou Medical College, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China.
| | | | | |
Collapse
|
31
|
Pal A, Das S. Chronic morphine exposure and its abstinence alters dendritic spine morphology and upregulates Shank1. Neurochem Int 2013; 62:956-64. [PMID: 23538264 DOI: 10.1016/j.neuint.2013.03.011] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2012] [Revised: 03/01/2013] [Accepted: 03/15/2013] [Indexed: 02/04/2023]
Abstract
Exposure to chronic drugs of abuse has been reported to produce significant changes in postsynaptic protein profile, dendritic spine morphology and synaptic transmission. In the present study we demonstrate alterations in dendritic spine morphology in the frontal cortex and nucleus accumbens of mice following chronic morphine treatment as well as during abstinence for two months. Such alterations were accompanied with significant upregulation of the postsynaptic protein Shank1 in synaptosomal enriched fractions. mRNA levels of Shank1 was also markedly increased during morphine treatment and during withdrawal. Studies of the different postsynaptic proteins at the protein and mRNA levels showed significant alterations in the morphine treated groups compared to that of saline treated controls. Taken together, these observations suggest that Shank1 may have an important role in the regulation of spine morphology induced by chronic morphine leading to addiction.
Collapse
Affiliation(s)
- Ayantika Pal
- Neurobiology Department, Cell Biology & Physiology Division, CSIR-Indian Institute of Chemical Biology, 4 Raja S. C. Mullick Road, Jadavpur, Kolkata 700032, India
| | | |
Collapse
|
32
|
Piers TM, Kim DH, Kim BC, Regan P, Whitcomb DJ, Cho K. Translational Concepts of mGluR5 in Synaptic Diseases of the Brain. Front Pharmacol 2012. [PMID: 23205012 PMCID: PMC3506921 DOI: 10.3389/fphar.2012.00199] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
The G-protein coupled receptor family of glutamate receptors, termed metabotropic glutamate receptors (mGluRs), are implicated in numerous cellular mechanisms ranging from neural development to the processing of cognitive, sensory, and motor information. Over the last decade, multiple mGluR-related signal cascades have been identified at excitatory synapses, indicating their potential roles in various forms of synaptic function and dysfunction. This review highlights recent studies investigating mGluR5, a subtype of group I mGluRs, and its association with a number of developmental, psychiatric, and senile synaptic disorders with respect to associated synaptic proteins, with an emphasis on translational pre-clinical studies targeting mGluR5 in a range of synaptic diseases of the brain.
Collapse
Affiliation(s)
- Thomas M Piers
- School of Clinical Sciences, Faculty of Medicine and Dentistry, University of Bristol Bristol, UK
| | | | | | | | | | | |
Collapse
|
33
|
Scaffolding proteins of the post-synaptic density contribute to synaptic plasticity by regulating receptor localization and distribution: relevance for neuropsychiatric diseases. Neurochem Res 2012; 38:1-22. [PMID: 22991141 DOI: 10.1007/s11064-012-0886-y] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2012] [Revised: 08/16/2012] [Accepted: 09/10/2012] [Indexed: 10/27/2022]
Abstract
Synaptic plasticity represents the long lasting activity-related strengthening or weakening of synaptic transmission, whose well-characterized types are the long term potentiation and depression. Despite this classical definition, however, the molecular mechanisms by which synaptic plasticity may occur appear to be extremely complex and various. The post-synaptic density (PSD) of glutamatergic synapses is a major site for synaptic plasticity processes and alterations of PSD members have been recently implicated in neuropsychiatric diseases where an impairment of synaptic plasticity has also been reported. Among PSD members, scaffolding proteins have been demonstrated to bridge surface receptors with their intracellular effectors and to regulate receptors distribution and localization both at surface membranes and within the PSD. This review will focus on the molecular physiology and pathophysiology of synaptic plasticity processes, which are tuned by scaffolding PSD proteins and their close related partners, through the modulation of receptor localization and distribution at post-synaptic sites. We suggest that, by regulating both the compartmentalization of receptors along surface membrane and their degradation as well as by modulating receptor trafficking into the PSD, postsynaptic scaffolding proteins may contribute to form distinct signaling micro-domains, whose efficacy in transmitting synaptic signals depends on the dynamic stability of the scaffold, which in turn is provided by relative amounts and post-translational modifications of scaffolding members. The putative relevance for neuropsychiatric diseases and possible pathophysiological mechanisms are discussed in the last part of this work.
Collapse
|
34
|
de Bartolomeis A, Tomasetti C. Calcium-Dependent Networks in Dopamine–Glutamate Interaction: The Role of Postsynaptic Scaffolding Proteins. Mol Neurobiol 2012; 46:275-96. [DOI: 10.1007/s12035-012-8293-6] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2012] [Accepted: 06/21/2012] [Indexed: 01/11/2023]
|
35
|
Falkai P, Möller HJ. Neurobiology of schizophrenia: from outcome to pathophysiological insights. Eur Arch Psychiatry Clin Neurosci 2012; 262:93-4. [PMID: 22271345 PMCID: PMC3297739 DOI: 10.1007/s00406-012-0294-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- P. Falkai
- Department of Psychiatry and Psychotherapy, University of Göttingen, Von-Siebold-Str. 5, 37075 Göttingen, Germany
| | - H. -J. Möller
- Department of Psychiatry, Ludwigs-Maximililans-University Munich, Nussbaumstr. 7, 80336 Munich, Germany
| |
Collapse
|