1
|
Chen W, Liu Y, Pu J, Gui S, Wang D, Zhong X, Tao W, Chen X, Chen X, Chen Y, Zhao L, Wu Q, Chen X, Zhang Y, Xie A, Xie P. Comparative transcriptional analyses of the striatum in the chronic social defeat stress model in C57BL/6J male mice and the gut microbiota-dysbiosis model in Kumming mice. Neuroscience 2024; 562:217-226. [PMID: 39489477 DOI: 10.1016/j.neuroscience.2024.10.057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 10/11/2024] [Accepted: 10/31/2024] [Indexed: 11/05/2024]
Abstract
Depression is a complex disorder with multiple contributing factors, and chronic stress has previously been recognized as a major causative factor, while gut microbes have also been found to be involved in depression recently. However, gene expression in depression models with different etiologies is unclear. Here, we compared the transcriptomes of the striatum in chronic social defeat stress (CSDS) model of C57BL/6J male mice and fecal microbiota transplant (FMT) model of Kumming male mice. We found that the proportion of shared differentially expressed genes (DEGs) between the two models was only 24 %. The specific DEGs of FMT model were enriched in immune and inflammatory, and are associated with changes in vascular and ciliated ependymal cells. The specific DEGs of CSDS model were enriched in neuron and synapse. The results of network analysis suggested the expression patterns and biological function of depressive-like behaviors-related modules in the two models are different. Further, the alternative splicing events of CSDS are more than FMT. Our results suggested models of depression induced by different etiologies differ significantly in gene expression and biological function. Our study also suggested us to pay attention to the characteristics of models of depression of different etiologies and provided a more comprehensive understanding of the heterogeneity of depression.
Collapse
Affiliation(s)
- Weiyi Chen
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; The Jin Feng Laboratory, Chongqing 401329, China
| | - Yiyun Liu
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; The Jin Feng Laboratory, Chongqing 401329, China
| | - Juncai Pu
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; The Jin Feng Laboratory, Chongqing 401329, China
| | - Siwen Gui
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; The Jin Feng Laboratory, Chongqing 401329, China
| | - Dongfang Wang
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; The Jin Feng Laboratory, Chongqing 401329, China
| | - Xiaogang Zhong
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; The Jin Feng Laboratory, Chongqing 401329, China
| | - Wei Tao
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; The Jin Feng Laboratory, Chongqing 401329, China
| | - Xiaopeng Chen
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; The Jin Feng Laboratory, Chongqing 401329, China
| | - Xiang Chen
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; The Jin Feng Laboratory, Chongqing 401329, China
| | - Yue Chen
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; The Jin Feng Laboratory, Chongqing 401329, China
| | - Libo Zhao
- Department of Neurology, Yongchuan Hospital of Chongqing Medical University 402160 Chongqing, China
| | - Qingyuan Wu
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Department of Neurology, Chongqing University Three Gorges Hospital, Chongqing 404000, China
| | - Xiangyu Chen
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; The Jin Feng Laboratory, Chongqing 401329, China
| | - Yingying Zhang
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao 266000, Shan-dong, China
| | - Anmu Xie
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao 266000, Shan-dong, China.
| | - Peng Xie
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; The Jin Feng Laboratory, Chongqing 401329, China.
| |
Collapse
|
2
|
Haetzel LM, Iafrati J, Cording KR, Farhan M, Noveir SD, Rumbaugh G, Bateup HS. Haploinsufficiency of Syngap1 in Striatal Indirect Pathway Neurons Alters Motor and Goal-Directed Behaviors in Mice. J Neurosci 2024; 44:e1264232024. [PMID: 39358043 PMCID: PMC11604145 DOI: 10.1523/jneurosci.1264-23.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 09/15/2024] [Accepted: 09/18/2024] [Indexed: 10/04/2024] Open
Abstract
SYNGAP1 is a high-confidence autism spectrum disorder (ASD) risk gene, and mutations in SYNGAP1 lead to a neurodevelopmental disorder (NDD) that presents with epilepsy, ASD, motor developmental delay, and intellectual disability. SYNGAP1 codes for Ras/Rap GTP-ase activating protein SynGAP (SynGAP). SynGAP is located in the postsynaptic density of glutamatergic synapses and regulates glutamate receptor trafficking in an activity-dependent manner. In addition to forebrain glutamatergic neurons, Syngap1 is highly expressed in the striatum, although the functions of SynGAP in the striatum have not been extensively studied. Here we show that Syngap1 is expressed in both direct and indirect pathway striatal projection neurons (dSPNs and iSPNs) in mice of both sexes. In a mouse model of Syngap1 haploinsufficiency, dendritic spine density, morphology, and intrinsic excitability are altered primarily in iSPNs, but not dSPNs. At the behavioral level, SynGAP reduction alters striatal-dependent motor learning and goal-directed behavior. Several behavioral phenotypes are reproduced by iSPN-specific Syngap1 reduction and, in turn, prevented by iSPN-specific Syngap1 rescue. These results establish the importance of SynGAP to striatal neuron function and pinpoint the indirect pathway as a key circuit in the neurobiology of SYNGAP1-related NDD.
Collapse
Affiliation(s)
- Laura M Haetzel
- Helen Wills Neuroscience Institute, University of California, Berkeley, California 94720
- Departments of Molecular and Cell Biology, University of California, Berkeley, California 94720
| | - Jillian Iafrati
- Departments of Molecular and Cell Biology, University of California, Berkeley, California 94720
| | - Katherine R Cording
- Helen Wills Neuroscience Institute, University of California, Berkeley, California 94720
- Departments of Molecular and Cell Biology, University of California, Berkeley, California 94720
- Neuroscience, University of California, Berkeley, California 94720
| | - Mahmoud Farhan
- Departments of Molecular and Cell Biology, University of California, Berkeley, California 94720
| | - Sasan D Noveir
- Departments of Molecular and Cell Biology, University of California, Berkeley, California 94720
| | - Gavin Rumbaugh
- Departments of Neuroscience and Molecular Medicine, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, Florida 33458
- Skaggs Graduate School of Chemical and Biological Sciences, Jupiter, Florida 33458
| | - Helen S Bateup
- Helen Wills Neuroscience Institute, University of California, Berkeley, California 94720
- Departments of Molecular and Cell Biology, University of California, Berkeley, California 94720
- Neuroscience, University of California, Berkeley, California 94720
- Weill Neurohub Investigator, University of California, Berkeley, California 94720
| |
Collapse
|
3
|
Bačová Z, Jurkovičová-Tarabová B, Havránek T, Mihalj D, Borbélyová V, Pirnik Z, Mravec B, Ostatníková D, Bakoš J. Shank3 deficiency alters midbrain GABAergic neuron morphology, GABAergic markers and synaptic activity in primary striatal neurons. Mol Brain 2024; 17:71. [PMID: 39334399 PMCID: PMC11430545 DOI: 10.1186/s13041-024-01145-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 09/12/2024] [Indexed: 09/30/2024] Open
Abstract
Abnormalities in gamma-aminobutyric acid (GABA)ergic neurotransmission play a role in the pathogenesis of autism, although the mechanisms responsible for alterations in specific brain regions remain unclear. Deficits in social motivation and interactions are core symptoms of autism, likely due to defects in dopaminergic neural pathways. Therefore, investigating the morphology and functional roles of GABAergic neurons within dopaminergic projection areas could elucidate the underlying etiology of autism. The aim of this study was to (1) compare the morphology and arborization of glutamate decarboxylase (GAD)-positive neurons from the midbrain tegmentum; (2) evaluate synaptic activity in primary neurons from the striatum; and (3) assess GABAergic postsynaptic puncta in the ventral striatum of wild-type (WT) and Shank3-deficient mice. We found a significant decrease in the number of short neurites in GAD positive primary neurons from the midbrain tegmentum in Shank3-deficient mice. The application of a specific blocker of GABAA receptors (GABAAR) revealed significantly increased frequency of spontaneous postsynaptic currents (sPSCs) in Shank3-deficient striatal neurons compared to their WT counterparts. The mean absolute amplitude of the events was significantly higher in striatal neurons from Shank3-deficient compared to WT mice. We also observed a significant reduction in gephyrin/GABAAR γ2 colocalization in the striatum of adult male Shank3-deficient mice. The gene expression of collybistin was significantly lower in the nucleus accumbens while gephyrin and GABAAR γ2 were lower in the ventral tegmental area (VTA) in male Shank3-deficient compared to WT mice. In conclusion, Shank3 deficiency leads to alterations in GABAergic neurons and impaired GABAergic function in dopaminergic brain areas. These changes may underlie autistic symptoms, and potential interventions modulating GABAergic activity in dopaminergic pathways may represent new treatment modality.
Collapse
Affiliation(s)
- Zuzana Bačová
- Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, Dubravska cesta 9, Bratislava, 845 05, Slovakia
| | - Bohumila Jurkovičová-Tarabová
- Institute of Molecular Physiology and Genetics, Center of Biosciences, Slovak Academy of Sciences, Bratislava, Slovakia
- Department of Biology, Faculty of Education, Trnava University, Trnava, Slovakia
| | - Tomáš Havránek
- Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, Dubravska cesta 9, Bratislava, 845 05, Slovakia
- Institute of Anatomy, Faculty of Medicine, Comenius University in Bratislava, Bratislava, Slovakia
| | - Denisa Mihalj
- Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, Dubravska cesta 9, Bratislava, 845 05, Slovakia
| | - Veronika Borbélyová
- Institute of Molecular Biomedicine, Faculty of Medicine, Comenius University in Bratislava, Bratislava, Slovakia
| | - Zdenko Pirnik
- Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, Dubravska cesta 9, Bratislava, 845 05, Slovakia
- Institute of Physiology, Faculty of Medicine, Comenius University in Bratislava, Sasinkova 2, Bratislava, 813 72, Slovakia
| | - Boris Mravec
- Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, Dubravska cesta 9, Bratislava, 845 05, Slovakia
- Institute of Physiology, Faculty of Medicine, Comenius University in Bratislava, Sasinkova 2, Bratislava, 813 72, Slovakia
| | - Daniela Ostatníková
- Institute of Physiology, Faculty of Medicine, Comenius University in Bratislava, Sasinkova 2, Bratislava, 813 72, Slovakia
| | - Ján Bakoš
- Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, Dubravska cesta 9, Bratislava, 845 05, Slovakia.
- Institute of Physiology, Faculty of Medicine, Comenius University in Bratislava, Sasinkova 2, Bratislava, 813 72, Slovakia.
| |
Collapse
|
4
|
Almeida AFSD, Silva TDD, Moraes ÍAPD, Menezes LDCD, Dias ED, Araújo LVD, Monteiro CBDM, Dawes H, Simcsik AO, Alberissi CADO, Silva VYHD, Brunherotti MAA, Tonello MGM. Virtual reality as a telerehabilitation strategy for people with autism spectrum disorder during the COVID-19 quarantine scenario: physical activity, motor performance and enjoyment. Disabil Rehabil Assist Technol 2024; 19:2046-2056. [PMID: 37695272 PMCID: PMC11229792 DOI: 10.1080/17483107.2023.2249031] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Accepted: 08/09/2023] [Indexed: 09/12/2023]
Abstract
PURPOSE People with autism spectrum disorder could benefit from physical activity during the pandemic and COVID-19 restrictions, mainly to maintain adequate physical activity. We aimed to evaluate the feasibility, enjoyment, and potential effect of telerehabilitation using a serious game named 'MoveHero'. MATERIALS AND METHODS Registered in Clinical Trials (NCT04402034). We adopted a remotely run Telerehabilitation research design with 44 participants recruited: 22 People with ASD people and 22 non-ASD individuals. RESULTS All participants safely participated, 100% adherence to sessions, ∼60% enjoying the task, and significantly improved performance, with better performance for the NA group at most practice moments. CONCLUSIONS Our findings support both how to implement a gaming intervention and the need to investigate the efficacy of serious games to motivate moderate intensity physical activity in people with ASD.
Collapse
Affiliation(s)
| | - Talita Dias da Silva
- Grupo de Pesquisa e Aplicações Tecnológicas em Reabilitaçaõ (PATER) da Escola de Artes, Ciências e Humanidades da Universidade de São Paulo (EACH-USP), São, Brazilulo
- Departamento de Medicina (Cardiologia), Escola Paulista de Medicina da Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
- Faculdade de Medicina, Universidade Cidade de São Paulo (UNICID). R. Butantã, São Paulo, Brazil
| | - Íbis Ariana Peña de Moraes
- Grupo de Pesquisa e Aplicações Tecnológicas em Reabilitaçaõ (PATER) da Escola de Artes, Ciências e Humanidades da Universidade de São Paulo (EACH-USP), São, Brazilulo
- Faculdade de Medicina, Universidade Cidade de São Paulo (UNICID). R. Butantã, São Paulo, Brazil
- Faculdade de Medicina, Universidade de São Paulo (USP), Cidade Universitária, São Paulo, Brazil
- NIHR Exeter Biomedical Research Centre, College of Medicine and Health, University of Exeter, Exeter, UK
| | - Lilian Del Ciello de Menezes
- Departamento de Medicina (Cardiologia), Escola Paulista de Medicina da Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| | - Eduardo Dati Dias
- Grupo de Pesquisa e Aplicações Tecnológicas em Reabilitaçaõ (PATER) da Escola de Artes, Ciências e Humanidades da Universidade de São Paulo (EACH-USP), São, Brazilulo
- Faculdade de Medicina, Universidade de São Paulo (USP), Cidade Universitária, São Paulo, Brazil
| | - Luciano Vieira de Araújo
- Grupo de Pesquisa e Aplicações Tecnológicas em Reabilitaçaõ (PATER) da Escola de Artes, Ciências e Humanidades da Universidade de São Paulo (EACH-USP), São, Brazilulo
| | - Carlos Bandeira de Mello Monteiro
- Grupo de Pesquisa e Aplicações Tecnológicas em Reabilitaçaõ (PATER) da Escola de Artes, Ciências e Humanidades da Universidade de São Paulo (EACH-USP), São, Brazilulo
- Faculdade de Medicina, Universidade de São Paulo (USP), Cidade Universitária, São Paulo, Brazil
| | - Helen Dawes
- NIHR Exeter Biomedical Research Centre, College of Medicine and Health, University of Exeter, Exeter, UK
| | - Amanda Orasmo Simcsik
- Grupo de Pesquisa e Aplicações Tecnológicas em Reabilitaçaõ (PATER) da Escola de Artes, Ciências e Humanidades da Universidade de São Paulo (EACH-USP), São, Brazilulo
| | - Camila Aparecida de Oliveira Alberissi
- Grupo de Pesquisa e Aplicações Tecnológicas em Reabilitaçaõ (PATER) da Escola de Artes, Ciências e Humanidades da Universidade de São Paulo (EACH-USP), São, Brazilulo
| | - Victoria Yanara Hernandes da Silva
- Grupo de Pesquisa e Aplicações Tecnológicas em Reabilitaçaõ (PATER) da Escola de Artes, Ciências e Humanidades da Universidade de São Paulo (EACH-USP), São, Brazilulo
- Departamento de Medicina (Cardiologia), Escola Paulista de Medicina da Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| | | | | |
Collapse
|
5
|
Cording KR, Tu EM, Wang H, Agopyan-Miu AHCW, Bateup HS. Cntnap2 loss drives striatal neuron hyperexcitability and behavioral inflexibility. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.09.593387. [PMID: 38766169 PMCID: PMC11100810 DOI: 10.1101/2024.05.09.593387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder characterized by two major diagnostic criteria - persistent deficits in social communication and interaction, and the presence of restricted, repetitive patterns of behavior (RRBs). Evidence from both human and animal model studies of ASD suggest that alteration of striatal circuits, which mediate motor learning, action selection, and habit formation, may contribute to the manifestation of RRBs. CNTNAP2 is a syndromic ASD risk gene, and loss of function of Cntnap2 in mice is associated with RRBs. How loss of Cntnap2 impacts striatal neuron function is largely unknown. In this study, we utilized Cntnap2-/- mice to test whether altered striatal neuron activity contributes to aberrant motor behaviors relevant to ASD. We find that Cntnap2-/- mice exhibit increased cortical drive of striatal projection neurons (SPNs), with the most pronounced effects in direct pathway SPNs. This enhanced drive is likely due to increased intrinsic excitability of SPNs, which make them more responsive to cortical inputs. We also find that Cntnap2-/- mice exhibit spontaneous repetitive behaviors, increased motor routine learning, and cognitive inflexibility. Increased corticostriatal drive, in particular of the direct pathway, may contribute to the acquisition of repetitive, inflexible behaviors in Cntnap2 mice.
Collapse
Affiliation(s)
- Katherine R. Cording
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA USA
| | - Emilie M. Tu
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA USA
| | - Hongli Wang
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA USA
| | | | - Helen S. Bateup
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA USA
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA USA
| |
Collapse
|
6
|
Khandelwal N, Kulkarni A, Ahmed NI, Harper M, Konopka G, Gibson JR. FOXP1 regulates the development of excitatory synaptic inputs onto striatal neurons and induces phenotypic reversal with reinstatement. SCIENCE ADVANCES 2024; 10:eadm7039. [PMID: 38701209 PMCID: PMC11068015 DOI: 10.1126/sciadv.adm7039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 04/01/2024] [Indexed: 05/05/2024]
Abstract
Long-range glutamatergic inputs originating from the cortex and thalamus are indispensable for striatal development, providing the foundation for motor and cognitive functions. Despite their significance, transcriptional regulation governing these inputs remains largely unknown. We investigated the role of a transcription factor encoded by a high-risk autism-associated gene, FOXP1, in sculpting glutamatergic inputs onto spiny projection neurons (SPNs) within the striatum. We find a neuron subtype-specific role of FOXP1 in strengthening and maturing glutamatergic inputs onto dopamine receptor 2-expressing SPNs (D2 SPNs). We also find that FOXP1 promotes synaptically driven excitability in these neurons. Using single-nuclei RNA sequencing, we identify candidate genes that mediate these cell-autonomous processes through postnatal FOXP1 function at the post-synapse. Last, we demonstrate that postnatal FOXP1 reinstatement rescues electrophysiological deficits, cell type-specific gene expression changes, and behavioral phenotypes. Together, this study enhances our understanding of striatal circuit development and provides proof of concept for a therapeutic approach for FOXP1 syndrome and other neurodevelopmental disorders.
Collapse
Affiliation(s)
- Nitin Khandelwal
- Department of Neuroscience and Peter O’Donnell Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Ashwinikumar Kulkarni
- Department of Neuroscience and Peter O’Donnell Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Newaz I. Ahmed
- Department of Neuroscience and Peter O’Donnell Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Matthew Harper
- Department of Neuroscience and Peter O’Donnell Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | | | | |
Collapse
|
7
|
Pan YD, Zhang Y, Zheng WY, Zhu MZ, Li HY, Ouyang WJ, Wen QQ, Zhu XH. Intermittent Hypobaric Hypoxia Ameliorates Autistic-Like Phenotypes in Mice. J Neurosci 2024; 44:e1665232023. [PMID: 38124211 PMCID: PMC10869151 DOI: 10.1523/jneurosci.1665-23.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 11/27/2023] [Accepted: 12/13/2023] [Indexed: 12/23/2023] Open
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental condition characterized by persistent deficits in social communication and stereotyped behaviors. Although major advances in basic research on autism have been achieved in the past decade, and behavioral interventions can mitigate the difficulties that individuals with autism experience, little is known about the many fundamental issues of the interventions, and no specific medication has demonstrated efficiency for the core symptoms of ASD. Intermittent hypobaric hypoxia (IHH) is characterized by repeated exposure to lowered atmospheric pressure and oxygen levels, which triggers multiple physiological adaptations in the body. Here, using two mouse models of ASD, male Shank3B -/- and Fmr1 -/y mice, we found that IHH training at an altitude of 5,000 m for 4 h per day, for 14 consecutive days, ameliorated autistic-like behaviors. Moreover, IHH training enhanced hypoxia inducible factor (HIF) 1α in the dorsal raphe nucleus (DRN) and activated the DRN serotonergic neurons. Infusion of cobalt chloride into the DRN, to mimic IHH in increasing HIF1α expression or genetically knockdown PHD2 to upregulate HIF1α expression in the DRN serotonergic neurons, alleviated autistic-like behaviors in Shank3B -/- mice. In contrast, downregulation of HIF1α in DRN serotonergic neurons induced compulsive behaviors. Furthermore, upregulating HIF1α in DRN serotonergic neurons increased the firing rates of these neurons, whereas downregulation of HIF1α in DRN serotonergic neurons decreased their firing rates. These findings suggest that IHH activated DRN serotonergic neurons via upregulation of HIF1α, and thus ameliorated autistic-like phenotypes, providing a novel therapeutic option for ASD.
Collapse
Affiliation(s)
- Yi-da Pan
- School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
- Research Center for Brain Health, Pazhou Lab, Guangzhou 510330, China
| | - Yuan Zhang
- School of Psychology, Shenzhen University, Shenzhen 518060, China
| | - Wen-Ying Zheng
- Research Center for Brain Health, Pazhou Lab, Guangzhou 510330, China
- School of Psychology, Shenzhen University, Shenzhen 518060, China
| | - Min-Zhen Zhu
- Research Center for Brain Health, Pazhou Lab, Guangzhou 510330, China
| | - Huan-Yu Li
- Research Center for Brain Health, Pazhou Lab, Guangzhou 510330, China
| | - Wen-Jie Ouyang
- Research Center for Brain Health, Pazhou Lab, Guangzhou 510330, China
- School of Psychology, Shenzhen University, Shenzhen 518060, China
| | - Qin-Qing Wen
- Research Center for Brain Health, Pazhou Lab, Guangzhou 510330, China
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China
| | - Xin-Hong Zhu
- School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
- Research Center for Brain Health, Pazhou Lab, Guangzhou 510330, China
- School of Psychology, Shenzhen University, Shenzhen 518060, China
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China
| |
Collapse
|
8
|
Liloia D, Manuello J, Costa T, Keller R, Nani A, Cauda F. Atypical local brain connectivity in pediatric autism spectrum disorder? A coordinate-based meta-analysis of regional homogeneity studies. Eur Arch Psychiatry Clin Neurosci 2024; 274:3-18. [PMID: 36599959 PMCID: PMC10787009 DOI: 10.1007/s00406-022-01541-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 12/16/2022] [Indexed: 01/05/2023]
Abstract
Despite decades of massive neuroimaging research, the comprehensive characterization of short-range functional connectivity in autism spectrum disorder (ASD) remains a major challenge for scientific advances and clinical translation. From the theoretical point of view, it has been suggested a generalized local over-connectivity that would characterize ASD. This stance is known as the general local over-connectivity theory. However, there is little empirical evidence supporting such hypothesis, especially with regard to pediatric individuals with ASD (age [Formula: see text] 18 years old). To explore this issue, we performed a coordinate-based meta-analysis of regional homogeneity studies to identify significant changes of local connectivity. Our analyses revealed local functional under-connectivity patterns in the bilateral posterior cingulate cortex and superior frontal gyrus (key components of the default mode network) and in the bilateral paracentral lobule (a part of the sensorimotor network). We also performed a functional association analysis of the identified areas, whose dysfunction is clinically consistent with the well-known deficits affecting individuals with ASD. Importantly, we did not find relevant clusters of local hyper-connectivity, which is contrary to the hypothesis that ASD may be characterized by generalized local over-connectivity. If confirmed, our result will provide a valuable insight into the understanding of the complex ASD pathophysiology.
Collapse
Affiliation(s)
- Donato Liloia
- GCS-fMRI Research Group, Koelliker Hospital and Department of Psychology, University of Turin, Via Giuseppe Verdi 10, 10124, Turin, Italy
- Functional Neuroimaging and Complex Neural Systems (FOCUS) Laboratory, Department of Psychology, University of Turin, Turin, Italy
| | - Jordi Manuello
- GCS-fMRI Research Group, Koelliker Hospital and Department of Psychology, University of Turin, Via Giuseppe Verdi 10, 10124, Turin, Italy
- Functional Neuroimaging and Complex Neural Systems (FOCUS) Laboratory, Department of Psychology, University of Turin, Turin, Italy
| | - Tommaso Costa
- GCS-fMRI Research Group, Koelliker Hospital and Department of Psychology, University of Turin, Via Giuseppe Verdi 10, 10124, Turin, Italy.
- Functional Neuroimaging and Complex Neural Systems (FOCUS) Laboratory, Department of Psychology, University of Turin, Turin, Italy.
- Neuroscience Institute of Turin (NIT), Turin, Italy.
| | - Roberto Keller
- Adult Autism Center, DSM Local Health Unit, ASL TO, Turin, Italy
| | - Andrea Nani
- Functional Neuroimaging and Complex Neural Systems (FOCUS) Laboratory, Department of Psychology, University of Turin, Turin, Italy
| | - Franco Cauda
- GCS-fMRI Research Group, Koelliker Hospital and Department of Psychology, University of Turin, Via Giuseppe Verdi 10, 10124, Turin, Italy
- Functional Neuroimaging and Complex Neural Systems (FOCUS) Laboratory, Department of Psychology, University of Turin, Turin, Italy
- Neuroscience Institute of Turin (NIT), Turin, Italy
| |
Collapse
|
9
|
Soghomonian JJ. The cortico-striatal circuitry in autism-spectrum disorders: a balancing act. Front Cell Neurosci 2024; 17:1329095. [PMID: 38273975 PMCID: PMC10808402 DOI: 10.3389/fncel.2023.1329095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 12/18/2023] [Indexed: 01/27/2024] Open
Abstract
The basal ganglia are major targets of cortical inputs and, in turn, modulate cortical function via their projections to the motor and prefrontal cortices. The role of the basal ganglia in motor control and reward is well documented and there is also extensive evidence that they play a key role in social and repetitive behaviors. The basal ganglia influence the activity of the cerebral cortex via two major projections from the striatum to the output nuclei, the globus pallidus internus and the substantia nigra, pars reticulata. This modulation involves a direct projection known as the direct pathway and an indirect projection via the globus pallidus externus and the subthalamic nucleus, known as the indirect pathway. This review discusses the respective contribution of the direct and indirect pathways to social and repetitive behaviors in neurotypical conditions and in autism spectrum disorders.
Collapse
|
10
|
Chen H, Mao Q, Zhang Y, Shi M, Geng W, Ma Y, Chen Y, Yin X. Disrupted Effective Connectivity within the Fronto-Thalamic Circuit in Pontine Infarction: A Spectral Dynamic Causal Modeling Study. Brain Sci 2024; 14:45. [PMID: 38248260 PMCID: PMC10813776 DOI: 10.3390/brainsci14010045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Revised: 12/13/2023] [Accepted: 12/19/2023] [Indexed: 01/23/2024] Open
Abstract
This study aims to investigate alterations in effective connectivity (EC) within the fronto-thalamic circuit and their associations with motor and cognitive declines in pontine infarction (PI). A total of 33 right PI patients (RPIs), 38 left PI patients (LPIs), and 67 healthy controls (HCs) were recruited. The spectral dynamic causal modeling (spDCM) approach was used for EC analysis within the fronto-thalamic circuit, including the thalamus, caudate, supplementary motor area (SMA), medial prefrontal cortex (mPFC), and anterior cingulate cortex (ACC). The EC differences between different sides of the patients and HCs were assessed, and their correlations with motor and cognitive dysfunctions were analyzed. The LPIs showed increased EC from the mPFC to the R-SMA and decreased EC from the L-thalamus to the ACC, the L-SMA to the R-SMA, the R-caudate to the R-thalamus, and the R-thalamus to the ACC. For RPIs, the EC of the R-caudate to the mPFC, the L-thalamus and L-caudate to the L-SMA, and the L-caudate to the ACC increased obviously, while a lower EC strength was shown from the L-thalamus to the mPFC, the LSMA to the R-caudate, and the R-SMA to the L-thalamus. The EC from the R-caudate to the mPFC was negatively correlated with the MoCA score for RPIs, and the EC from the R-caudate to the R-thalamus was negatively correlated with the FMA score for LPIs. The results demonstrated EC within the fronto-thalamic circuit in PI-related functional impairments and reveal its potential as a novel imaging marker.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Xindao Yin
- Department of Radiology, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China; (H.C.); (Q.M.); (Y.Z.); (M.S.); (W.G.); (Y.M.); (Y.C.)
| |
Collapse
|
11
|
Cording KR, Bateup HS. Altered motor learning and coordination in mouse models of autism spectrum disorder. Front Cell Neurosci 2023; 17:1270489. [PMID: 38026686 PMCID: PMC10663323 DOI: 10.3389/fncel.2023.1270489] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 09/25/2023] [Indexed: 12/01/2023] Open
Abstract
Autism spectrum disorder (ASD) is a complex neurodevelopmental disorder with increasing prevalence. Over 1,000 risk genes have now been implicated in ASD, suggesting diverse etiology. However, the diagnostic criteria for the disorder still comprise two major behavioral domains - deficits in social communication and interaction, and the presence of restricted and repetitive patterns of behavior (RRBs). The RRBs associated with ASD include both stereotyped repetitive movements and other motor manifestations including changes in gait, balance, coordination, and motor skill learning. In recent years, the striatum, the primary input center of the basal ganglia, has been implicated in these ASD-associated motor behaviors, due to the striatum's role in action selection, motor learning, and habit formation. Numerous mouse models with mutations in ASD risk genes have been developed and shown to have alterations in ASD-relevant behaviors. One commonly used assay, the accelerating rotarod, allows for assessment of both basic motor coordination and motor skill learning. In this corticostriatal-dependent task, mice walk on a rotating rod that gradually increases in speed. In the extended version of this task, mice engage striatal-dependent learning mechanisms to optimize their motor routine and stay on the rod for longer periods. This review summarizes the findings of studies examining rotarod performance across a range of ASD mouse models, and the resulting implications for the involvement of striatal circuits in ASD-related motor behaviors. While performance in this task is not uniform across mouse models, there is a cohort of models that show increased rotarod performance. A growing number of studies suggest that this increased propensity to learn a fixed motor routine may reflect a common enhancement of corticostriatal drive across a subset of mice with mutations in ASD-risk genes.
Collapse
Affiliation(s)
- Katherine R. Cording
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA, United States
| | - Helen S. Bateup
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA, United States
- Molecular and Cell Biology Department, University of California, Berkeley, Berkeley, CA, United States
- Chan Zuckerberg Biohub, San Francisco, CA, United States
| |
Collapse
|
12
|
Khandelwal N, Kulkarni A, Ahmed NI, Harper M, Konopka G, Gibson J. FOXP1 regulates the development of excitatory synaptic inputs onto striatal neurons and induces phenotypic reversal with reinstatement. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.23.563675. [PMID: 37961477 PMCID: PMC10634768 DOI: 10.1101/2023.10.23.563675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Long-range glutamatergic inputs from the cortex and thalamus are critical for motor and cognitive processing in the striatum. Transcription factors that orchestrate the development of these inputs are largely unknown. We investigated the role of a transcription factor and high-risk autism-associated gene, FOXP1, in the development of glutamatergic inputs onto spiny projection neurons (SPNs) in the striatum. We find that FOXP1 robustly drives the strengthening and maturation of glutamatergic input onto dopamine receptor 2-expressing SPNs (D2 SPNs) but has a comparatively milder effect on D1 SPNs. This process is cell-autonomous and is likely mediated through postnatal FOXP1 function at the postsynapse. We identified postsynaptic FOXP1-regulated transcripts as potential candidates for mediating these effects. Postnatal reinstatement of FOXP1 rescues electrophysiological deficits, reverses gene expression alterations resulting from embryonic deletion, and mitigates behavioral phenotypes. These results provide support for a possible therapeutic approach for individuals with FOXP1 syndrome.
Collapse
|
13
|
Mesías RE, Zaki Y, Guevara CA, Friedman LG, Hussein A, Therrien K, Magee AR, Tzavaras N, Del Valle P, Baxter MG, Huntley GW, Benson DL. Development and cadherin-mediated control of prefrontal corticostriatal projections in mice. iScience 2023; 26:108002. [PMID: 37854688 PMCID: PMC10579443 DOI: 10.1016/j.isci.2023.108002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 08/07/2023] [Accepted: 09/18/2023] [Indexed: 10/20/2023] Open
Abstract
Action-outcome associations depend on prefrontal cortex (PFC) projections to the dorsal striatum. To assess how these projections form, we measured PFC axon patterning, synapse formation, and functional maturation in the postnatally developing mouse striatum. Using Hotspot analysis, we show that PFC axons form an adult-like pattern of clustered terminations in the first postnatal week that remains largely stable thereafter. PFC-striatal synaptic strength is adult-like by P21, while excitatory synapse density increases until adulthood. We then tested how the targeted deletion of a candidate adhesion/guidance protein, Cadherin-8 (Cdh8), from corticostriatal neurons regulates pathway development. Mutant mice showed diminished PFC axon targeting and reduced spontaneous glutamatergic synaptic activity in the dorsal striatum. They also exhibited impaired behavioral performance in action-outcome learning. The data show that PFC-striatal axons form striatal territories through an early, directed growth model and they highlight essential contributions of Cdh8 to the anatomical and functional features critical for the formation of action-outcome associations.
Collapse
Affiliation(s)
- Roxana E. Mesías
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Yosif Zaki
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Christopher A. Guevara
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Lauren G. Friedman
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Ayan Hussein
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Karen Therrien
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Alexandra R. Magee
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Nikolaos Tzavaras
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Pamela Del Valle
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Mark G. Baxter
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Section on Comparative Medicine, Department of Pathology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | - George W. Huntley
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Deanna L. Benson
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
14
|
van der Westhuizen C, Botha TL, Finger-Baier K, Brouwer GD, Wolmarans DW. Contingency learning in zebrafish exposed to apomorphine- and levetiracetam. Behav Pharmacol 2023; 34:424-436. [PMID: 37578419 DOI: 10.1097/fbp.0000000000000750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/15/2023]
Abstract
Cognitive rigidity (CR) refers to inadequate executive adaptation in the face of changing circumstances. Increased CR is associated with a number of psychiatric disorders, for example, obsessive-compulsive disorder, and improving cognitive functioning by targeting CR in these conditions, may be fruitful. Levetiracetam (LEV), clinically used to treat epilepsy, may have pro-cognitive effects by restoring balance to neuronal signalling. To explore this possibility, we applied apomorphine (APO) exposure in an attempt to induce rigid cue-directed responses following a cue (visual pattern)-reward (social conspecifics) contingency learning phase and to assess the effects of LEV on such behaviours. Briefly, zebrafish were divided into four different 39-day-long exposure groups ( n = 9-10) as follows: control (CTRL), APO (100 µg/L), LEV (750 µg/L) and APO + LEV (100 µg/L + 750 µg/L). The main findings of this experiment were that 1) all four exposure groups performed similarly with respect to reward- and cue-directed learning over the first two study phases, 2) compared to the CTRL group, all drug interventions, but notably the APO + LEV combination, lowered the degree of reward-directed behaviour during a dissociated presentation of the cue and reward, and 3) temporal and spatial factors influenced the manner in which zebrafish responded to the presentation of the reward. Future studies are needed to explore the relevance of these findings for our understanding of the potential cognitive effects of LEV.
Collapse
Affiliation(s)
| | - Tarryn L Botha
- Water Research Group, Unit for Environmental Sciences and Management, North-West University, Potchefstroom
- Department of Zoology, University of Johannesburg, Johannesburg, South Africa
| | - Karin Finger-Baier
- Max Planck Institute of Neurobiology, now: Max Planck Institute for Biological Intelligence, Martinsried, Germany
| | - Geoffrey de Brouwer
- Centre of Excellence for Pharmaceutical Sciences, Faculty of Health, North-West University
| | - De Wet Wolmarans
- Centre of Excellence for Pharmaceutical Sciences, Faculty of Health, North-West University
| |
Collapse
|
15
|
Hogg EKJ, Findlay GM. Functions of SRPK, CLK and DYRK kinases in stem cells, development, and human developmental disorders. FEBS Lett 2023; 597:2375-2415. [PMID: 37607329 PMCID: PMC10952393 DOI: 10.1002/1873-3468.14723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 07/08/2023] [Accepted: 07/18/2023] [Indexed: 08/24/2023]
Abstract
Human developmental disorders encompass a wide range of debilitating physical conditions and intellectual disabilities. Perturbation of protein kinase signalling underlies the development of some of these disorders. For example, disrupted SRPK signalling is associated with intellectual disabilities, and the gene dosage of DYRKs can dictate the pathology of disorders including Down's syndrome. Here, we review the emerging roles of the CMGC kinase families SRPK, CLK, DYRK, and sub-family HIPK during embryonic development and in developmental disorders. In particular, SRPK, CLK, and DYRK kinase families have key roles in developmental signalling and stem cell regulation, and can co-ordinate neuronal development and function. Genetic studies in model organisms reveal critical phenotypes including embryonic lethality, sterility, musculoskeletal errors, and most notably, altered neurological behaviours arising from defects of the neuroectoderm and altered neuronal signalling. Further unpicking the mechanisms of specific kinases using human stem cell models of neuronal differentiation and function will improve our understanding of human developmental disorders and may provide avenues for therapeutic strategies.
Collapse
Affiliation(s)
- Elizabeth K. J. Hogg
- The MRC Protein Phosphorylation and Ubiquitylation Unit, School of Life SciencesUniversity of DundeeUK
| | - Greg M. Findlay
- The MRC Protein Phosphorylation and Ubiquitylation Unit, School of Life SciencesUniversity of DundeeUK
| |
Collapse
|
16
|
Patil O, Kaple M. Sensory Processing Differences in Individuals With Autism Spectrum Disorder: A Narrative Review of Underlying Mechanisms and Sensory-Based Interventions. Cureus 2023; 15:e48020. [PMID: 38034138 PMCID: PMC10687592 DOI: 10.7759/cureus.48020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 10/31/2023] [Indexed: 12/02/2023] Open
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental condition characterized by difficulties with social interaction and restricted, repetitive patterns of behavior. Altered sensory processing and perception are considered characteristics of ASD. Sensory processing differences (SPDs) are commonly observed in individuals with ASD, leading to atypical responses to sensory stimuli. SPDs refer to the way in which individuals receive, process, and respond to sensory information from the environment. People with SPDs may be hypersensitive (over-reactive) or hyposensitive (under-reactive) to sensory input, or they may experience fragmented or distorted perceptions. These differences can make it difficult for individuals with SPDs to filter out irrelevant sensory information, and to integrate sensory information from different sources. This study intends to investigate the underlying mechanisms contributing to SPDs in individuals with autism and determine the effectiveness of sensory-based therapies in addressing these challenges. The literature suggests that altered neural pathways, sensory gating dysfunction, and atypical sensory modulation contribute to SPDs in individuals with ASD. Assistive technology, environmental changes, and sensory-based interventions like sensory integration therapy (SIT) have all shown promise in improving sensory functioning and reducing associated behavioral issues. However, further research is needed to improve our understanding of sensory processing in autism and to optimize interventions for individuals with ASD.
Collapse
Affiliation(s)
- Om Patil
- Medicine, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Meghali Kaple
- Biochemistry, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| |
Collapse
|
17
|
Demler VF, Sterner EF, Wilson M, Zimmer C, Knolle F. Association between increased anterior cingulate glutamate and psychotic-like experiences, but not autistic traits in healthy volunteers. Sci Rep 2023; 13:12792. [PMID: 37550354 PMCID: PMC10406950 DOI: 10.1038/s41598-023-39881-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 08/01/2023] [Indexed: 08/09/2023] Open
Abstract
Despite many differences, autism spectrum disorder and schizophrenia spectrum disorder share environmental risk factors, genetic predispositions as well as neuronal abnormalities, and show similar cognitive deficits in working memory, perspective taking, or response inhibition. These shared abnormalities are already present in subclinical traits of these disorders. The literature proposes that changes in the inhibitory GABAergic and the excitatory glutamatergic system could explain underlying neuronal commonalities and differences. Using magnetic resonance spectroscopy (1H-MRS), we investigated the associations between glutamate concentrations in the anterior cingulate cortex (ACC), the left/right putamen, and left/right dorsolateral prefrontal cortex and psychotic-like experiences (Schizotypal Personality Questionnaire) and autistic traits (Autism Spectrum Quotient) in 53 healthy individuals (26 women). To investigate the contributions of glutamate concentrations in different cortical regions to symptom expression and their interactions, we used linear regression analyses. We found that only glutamate concentration in the ACC predicted psychotic-like experiences, but not autistic traits. Supporting this finding, a binomial logistic regression predicting median-split high and low risk groups for psychotic-like experiences revealed ACC glutamate levels as a significant predictor for group membership. Taken together, this study provides evidence that glutamate levels in the ACC are specifically linked to the expression of psychotic-like experiences, and may be a potential candidate in identifying early risk individuals prone to developing psychotic-like experiences.
Collapse
Affiliation(s)
- Verena F Demler
- Department of Diagnostic and Interventional Neuroradiology, Klinikum rechts der Isar, Technical University of Munich, Ismaninger Straße 22, 81675, Munich, Germany
| | - Elisabeth F Sterner
- Department of Diagnostic and Interventional Neuroradiology, Klinikum rechts der Isar, Technical University of Munich, Ismaninger Straße 22, 81675, Munich, Germany
| | - Martin Wilson
- Centre for Human Brain Health and School of Psychology, University of Birmingham, Birmingham, UK
| | - Claus Zimmer
- Department of Diagnostic and Interventional Neuroradiology, Klinikum rechts der Isar, Technical University of Munich, Ismaninger Straße 22, 81675, Munich, Germany
| | - Franziska Knolle
- Department of Diagnostic and Interventional Neuroradiology, Klinikum rechts der Isar, Technical University of Munich, Ismaninger Straße 22, 81675, Munich, Germany.
- Department of Psychiatry, University of Cambridge, Cambridge, UK.
| |
Collapse
|
18
|
Badke D’Andrea C, Marek S, Van AN, Miller RL, Earl EA, Stewart SB, Dosenbach NUF, Schlaggar BL, Laumann TO, Fair DA, Gordon EM, Greene DJ. Thalamo-cortical and cerebello-cortical functional connectivity in development. Cereb Cortex 2023; 33:9250-9262. [PMID: 37293735 PMCID: PMC10492576 DOI: 10.1093/cercor/bhad198] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 05/09/2023] [Accepted: 05/17/2023] [Indexed: 06/10/2023] Open
Abstract
The thalamus is a critical relay center for neural pathways involving sensory, motor, and cognitive functions, including cortico-striato-thalamo-cortical and cortico-ponto-cerebello-thalamo-cortical loops. Despite the importance of these circuits, their development has been understudied. One way to investigate these pathways in human development in vivo is with functional connectivity MRI, yet few studies have examined thalamo-cortical and cerebello-cortical functional connectivity in development. Here, we used resting-state functional connectivity to measure functional connectivity in the thalamus and cerebellum with previously defined cortical functional networks in 2 separate data sets of children (7-12 years old) and adults (19-40 years old). In both data sets, we found stronger functional connectivity between the ventral thalamus and the somatomotor face cortical functional network in children compared with adults, extending previous cortico-striatal functional connectivity findings. In addition, there was more cortical network integration (i.e. strongest functional connectivity with multiple networks) in the thalamus in children than in adults. We found no developmental differences in cerebello-cortical functional connectivity. Together, these results suggest different maturation patterns in cortico-striato-thalamo-cortical and cortico-ponto-cerebellar-thalamo-cortical pathways.
Collapse
Affiliation(s)
- Carolina Badke D’Andrea
- Department of Cognitive Science, University of California San Diego, La Jolla, CA 92093, United States
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110, United States
- Department of Radiology, Washington University School of Medicine, St. Louis, MO 63110, United States
| | - Scott Marek
- Department of Radiology, Washington University School of Medicine, St. Louis, MO 63110, United States
| | - Andrew N Van
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, United States
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO 63130, United States
| | - Ryland L Miller
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110, United States
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, United States
| | - Eric A Earl
- Data Science and Sharing Team, National Institute of Mental Health, NIH, DHHS, Bethesda, MD 20899, United States
| | - Stephanie B Stewart
- Department of Psychiatry, University of Colorado School of Medicine, Aurora, CO 80045, United States
| | - Nico U F Dosenbach
- Department of Radiology, Washington University School of Medicine, St. Louis, MO 63110, United States
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, United States
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO 63130, United States
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63110, United States
- Program in Occupational Therapy, Washington University School of Medicine, St. Louis, MO 63110, United States
| | | | - Timothy O Laumann
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110, United States
| | - Damien A Fair
- Institute of Child Development, College of Education and Human Development, University of Minnesota, Minneapolis, MN 55455, United States
- Department of Pediatrics, University of Minnesota Medical School, Minneapolis, MN 55455, United States
- Masonic Institute for the Developing Brain, University of Minnesota, Minneapolis, MN 55455, United States
| | - Evan M Gordon
- Department of Radiology, Washington University School of Medicine, St. Louis, MO 63110, United States
| | - Deanna J Greene
- Department of Cognitive Science, University of California San Diego, La Jolla, CA 92093, United States
| |
Collapse
|
19
|
Araujo MO, Tamplain P, Duarte NAC, Comodo ACM, Ferreira GOA, Queiróga A, Oliveira CS, Collange-Grecco LA. Transcranial direct current stimulation to facilitate neurofunctional rehabilitation in children with autism spectrum disorder: a protocol for a randomized, sham-controlled, double-blind clinical trial. Front Neurol 2023; 14:1196585. [PMID: 37396775 PMCID: PMC10310925 DOI: 10.3389/fneur.2023.1196585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 05/22/2023] [Indexed: 07/04/2023] Open
Abstract
Background Anodal transcranial direct current stimulation (tDCS) over the primary motor cortex and cerebellum is gaining prominence in the literature due to its potential to favor learning and motor performance. If administered during motor training, tDCS is capable of increasing the effect of training. Considering the motor impairment presented by children with Autism Spectrum Disorders (ASD), atDCS applied during motor training may contribute to the rehabilitation of these children. However, it is necessary to examine and compare the effects of atDCS over the motor cortex and the cerebellum on the motor skills of children with ASD. This information may benefit future clinical indications of tDCS for rehabilitation of children with ASD. The aim of the proposed study is to determine whether anodal tDCS over the primary motor cortex and cerebellum can enhance the effects of gait training and postural control on motor skills, mobility, functional balance, cortical excitability, cognitive aspects and behavioral aspects in children with ASD. Our hypothesis is the active tDCS combined with motor training will enhance the performance of the participants in comparison to sham tDCS. Methods and design A randomized, sham-controlled, double-blind clinical trial will be conducted involving 30 children with ASD that will be recruited to receive ten sessions of sham or ten sessions of active anodal tDCS (1 mA, 20 min) over the primary motor cortex or cerebellun combined with motor training. The participants will be assessed before as well as one, four and eight weeks after the interventions. The primary outcome will be gross and fine motor skills. The secondary outcomes will be mobility, functional balance, motor cortical excitability, cognitive aspects and behavioral aspects. Discussion Although abnormalities in gait and balance are not primary characteristics of ASD, such abnormalities compromise independence and global functioning during the execution of routine activities of childhood. If demonstrated that anodal tDCS administered over areas of the brain involved in motor control, such as the primary motor cortex and cerebellum, can enhance the effects of gait and balance training in only ten sessions in two consecutive weeks, the clinical applicability of this stimulation modality will be expanded as well as more scientifically founded.Clinical trial registration February 16, 2023 (https://ensaiosclinicos.gov.br/rg/RBR-3bskhwf).
Collapse
Affiliation(s)
- Marcela O. Araujo
- Human Movement and Rehabilitation, Post Graduate Program, Evangelic University of Goias, Anápolis, Brazil
- Children's Rehabilitation Department, Follow Kids Child Neurorehabilitation Clinic, Rio de Janeiro, Brazil
| | - Priscila Tamplain
- Department of Kinesiology, University of Texas at Arlington, Arlington, TX, United States
| | - Natália A. C. Duarte
- Human Movement and Rehabilitation, Post Graduate Program, Evangelic University of Goias, Anápolis, Brazil
| | - Andréa C. M. Comodo
- Children's Rehabilitation Department, Follow Kids Child Neurorehabilitation Clinic, Rio de Janeiro, Brazil
| | - Giselle O. A. Ferreira
- Children's Rehabilitation Department, Follow Kids Child Neurorehabilitation Clinic, Rio de Janeiro, Brazil
| | - Amanda Queiróga
- Department of Child Neurofunctional Physiotherapy, Center of Pediatric Neurostimulation, São Paulo, Brazil
| | - Claudia S. Oliveira
- Human Movement and Rehabilitation, Post Graduate Program, Evangelic University of Goias, Anápolis, Brazil
| | - Luanda A. Collange-Grecco
- Human Movement and Rehabilitation, Post Graduate Program, Evangelic University of Goias, Anápolis, Brazil
- Department of Child Neurofunctional Physiotherapy, Center of Pediatric Neurostimulation, São Paulo, Brazil
| |
Collapse
|
20
|
Mesías RE, Zaki Y, Guevara CA, Friedman LG, Hussein A, Therrien K, Magee AR, Tzavaras N, Valle PD, Baxter MG, Huntley GW, Benson DL. Development of prefrontal corticostriatal connectivity in mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.14.532475. [PMID: 36993639 PMCID: PMC10054964 DOI: 10.1101/2023.03.14.532475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Rational decision making is grounded in learning to associate actions with outcomes, a process that depends on projections from prefrontal cortex to dorsomedial striatum. Symptoms associated with a variety of human pathological conditions ranging from schizophrenia and autism to Huntington's and Parkinson's disease point toward functional deficits in this projection, but its development is not well understood, making it difficult to investigate how perturbations in development of this circuitry could contribute to pathophysiology. We applied a novel strategy based on Hotspot Analysis to assess the developmental progression of anatomical positioning of prefrontal cortex to striatal projections. Corticostriatal axonal territories established at P7 expand in concert with striatal growth but remain largely unchanged in positioning through adulthood, indicating they are generated by directed, targeted growth and not modified extensively by postnatal experience. Consistent with these findings, corticostriatal synaptogenesis increased steadily from P7 to P56, with no evidence for widescale pruning. As corticostriatal synapse density increased over late postnatal ages, the strength of evoked PFC input onto dorsomedial striatal projection neurons also increased, but spontaneous glutamatergic synaptic activity was stable. Based on its pattern of expression, we asked whether the adhesion protein, Cdh8, influenced this progression. In mice lacking Cdh8 in PFC corticostriatal projection neurons, axon terminal fields in dorsal striatum shifted ventrally. Corticostriatal synaptogenesis was unimpeded, but spontaneous EPSC frequency declined and mice failed to learn to associate an action with an outcome. Collectively these findings show that corticostriatal axons grow to their target zone and are restrained from an early age, do not undergo postnatal synapse pruning as the most dominant models predict, and that a relatively modest shift in terminal arbor positioning and synapse function has an outsized, negative impact on corticostriatal-dependent behavior.
Collapse
|
21
|
Crawley JN. Twenty years of discoveries emerging from mouse models of autism. Neurosci Biobehav Rev 2023; 146:105053. [PMID: 36682425 DOI: 10.1016/j.neubiorev.2023.105053] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 01/12/2023] [Accepted: 01/17/2023] [Indexed: 01/21/2023]
Abstract
More than 100 single gene mutations and copy number variants convey risk for autism spectrum disorder. To understand the extent to which each mutation contributes to the trajectory of individual symptoms of autism, molecular genetics laboratories have introduced analogous mutations into the genomes of laboratory mice and other species. Over the past twenty years, behavioral neuroscientists discovered the consequences of mutations in many risk genes for autism in animal models, using assays with face validity to the diagnostic and associated behavioral symptoms of people with autism. Identified behavioral phenotypes complement electrophysiological, neuroanatomical, and biochemical outcome measures in mutant mouse models of autism. This review describes the history of phenotyping assays in genetic mouse models, to evaluate social and repetitive behaviors relevant to the primary diagnostic criteria for autism. Robust phenotypes are currently employed in translational investigations to discover effective therapeutic interventions, representing the future direction of an intensely challenging research field.
Collapse
|
22
|
Huang M, Qi Q, Xu T. Targeting Shank3 deficiency and paresthesia in autism spectrum disorder: A brief review. Front Mol Neurosci 2023; 16:1128974. [PMID: 36846568 PMCID: PMC9948097 DOI: 10.3389/fnmol.2023.1128974] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 01/18/2023] [Indexed: 02/11/2023] Open
Abstract
Autism spectrum disorder (ASD) includes a group of multifactorial neurodevelopmental disorders characterized by impaired social communication, social interaction, and repetitive behaviors. Several studies have shown an association between cases of ASD and mutations in the genes of SH3 and multiple ankyrin repeat domain protein 3 (SHANK3). These genes encode many cell adhesion molecules, scaffold proteins, and proteins involved in synaptic transcription, protein synthesis, and degradation. They have a profound impact on all aspects of synaptic transmission and plasticity, including synapse formation and degeneration, suggesting that the pathogenesis of ASD may be partially attributable to synaptic dysfunction. In this review, we summarize the mechanism of synapses related to Shank3 in ASD. We also discuss the molecular, cellular, and functional studies of experimental models of ASD and current autism treatment methods targeting related proteins.
Collapse
Affiliation(s)
- Min Huang
- Department of Anesthesiology, Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China,Department of Anesthesiology, Suzhou Hospital of Anhui Medical University, Suzhou, China
| | - Qi Qi
- Department of Anesthesiology, Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China,Department of Anesthesiology, Suzhou Hospital of Anhui Medical University, Suzhou, China
| | - Tao Xu
- Department of Anesthesiology, Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China,Department of Anesthesiology, Suzhou Hospital of Anhui Medical University, Suzhou, China,*Correspondence: Tao Xu,
| |
Collapse
|
23
|
Wu D, Zhu J, You L, Wang J, Zhang S, Liu Z, Xu Q, Yuan X, Yang L, Wang W, Tong M, Hong Q, Chi X. NRXN1 depletion in the medial prefrontal cortex induces anxiety-like behaviors and abnormal social phenotypes along with impaired neurite outgrowth in rat. J Neurodev Disord 2023; 15:6. [PMID: 36737720 PMCID: PMC9896742 DOI: 10.1186/s11689-022-09471-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 12/07/2022] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Neurodevelopmental disorders (NDDs) are a group of disorders induced by abnormal brain developmental processes. The prefrontal cortex (PFC) plays an essential role in executive function, and its role in NDDs has been reported. NDDs are associated with high-risk gene mutations and share partially overlapping genetic abnormalities. METHODS Neurexins (NRXNs) are related to autism spectrum disorder (ASD) and attention-deficit hyperactivity disorder (ADHD). NRXN1, an essential susceptibility gene for NDDs, has been reported to be associated with NDDs. However, little is known about its key role in NDDs. RESULTS NRXN1 downregulation in the medial PFC induced anxiety-like behaviors and abnormal social phenotypes with impaired neurite outgrowth in Sh-NRXN1 in prefrontal neurons. Moreover, tandem mass tag (TMT)-based proteomic analysis of rat brain samples showed that NRXN1 downregulation led to significant proteome alterations, including pathways related to the extracellular matrix, cell membrane, and morphologic change. Furthermore, full-automatic immunoblotting analysis verified the differently expressed proteins related to cell morphology and membrane structure. CONCLUSIONS Our results confirmed the association of NRXN1 with abnormal behaviors in NDDs and provided richer insights into specific prefrontal knockdown in adolescence, potentially expanding the NRXN1 interactome and contributing to human health.
Collapse
Affiliation(s)
- Di Wu
- Department of Child Healthcare, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, China.,The Fourth Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jiansheng Zhu
- Department of Child Healthcare, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, China
| | - Lianghui You
- Department of Child Healthcare, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, China
| | - Jingyu Wang
- Department of Child Healthcare, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, China
| | - Sufen Zhang
- Department of Child Healthcare, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, China
| | - Zhonghui Liu
- Department of Child Healthcare, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, China
| | - Qu Xu
- Department of Child Healthcare, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, China
| | - Xiaojie Yuan
- Department of Child Healthcare, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, China
| | - Lei Yang
- Department of Child Healthcare, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, China
| | - Wei Wang
- The Fourth Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Meiling Tong
- Department of Child Healthcare, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, China
| | - Qin Hong
- Department of Child Healthcare, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, China.
| | - Xia Chi
- Department of Child Healthcare, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, China.
| |
Collapse
|
24
|
Zhang T, Rao Q, Lin K, He Y, Cai J, Yang M, Xu Y, Hou L, Lin Y, Liu H. CYP2C19-rs4986893 confers risk to major depressive disorder and bipolar disorder in the Han Chinese population whereas ABCB1-rs1045642 acts as a protective factor. BMC Psychiatry 2023; 23:69. [PMID: 36698099 PMCID: PMC9875448 DOI: 10.1186/s12888-022-04514-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 12/30/2022] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND Genetic risks may predispose individuals to major mood disorders differently. This study investigated the gene polymorphisms of previously reported candidate genes for major depressive disorder (MDD) and bipolar disorder (BPD) in the Han Chinese population. METHODS Twenty loci of 13 candidate genes were detected by MALDI-TOF mass spectrometry in 439 patients with MDD, 600 patients with BPD, and 464 healthy controls. The distribution of genotypes in alleles, Hardy-Weinberg equilibrium, and genetic association were analyzed using the PLINK software. The linkage of disequilibrium and haplotype analyses were performed using the Haploview software. RESULTS Out of the 20 loci analyzed, CYP2C19-rs4986893, ABCB1-rs1045642, and SCN2A-rs17183814 passed Bonferroni correction; their statistical powers were > 55%. The minor allele frequencies (MAF) of CYP2C19-rs4986893 in the MDD group (0.0547) and BPD group (0.0533) were higher than that of the control group (0.0259, P < 0.05), leading to the odds ratios (ORs) of MDD (2.178) and BPD (2.122), respectively. In contrast, the lower MAFs of ABCB1-rs1045642 were observed in both MDD (0.3599, OR = 0.726) and BPD (0.3700, OR = 0.758) groups than controls (0.4364, P < 0.05). The MDD group had a higher MAF of SCN2A-rs17183814 than controls (0.1743 vs. 0.1207, OR = 1.538, P < 0.05). Moreover, a G-A haplotype composed by CYP2C19-rs4986893 and -rs4244285 was associated with BPD (OR = 1.361, P < 0.01), and the A-G haplotype increased the risks to both MDD (OR = 2.306, P < 0.01) and BPD (OR = 2.332, P < 0.001). The CYP2C19 intermediate metabolizer and poor metabolizer (IM&PM) status was related to the raised risk of both MDD (OR = 1.547, P < 0.01) and BPD (OR = 1.808, P < 0.001). CONCLUSION Our data indicate that the impaired CYP2C19 metabolism caused by the haplotypes integrated by CYP2C19 alleles might confer the risk to MDD and BPD, whereas the ABCB1-rs1045642 T allele serves as a protective factor.
Collapse
Affiliation(s)
- Ting Zhang
- grid.410737.60000 0000 8653 1072Clinical Laboratory, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China
| | - Qingmin Rao
- grid.410737.60000 0000 8653 1072Clinical Laboratory, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China
| | - Kangguang Lin
- grid.410737.60000 0000 8653 1072Affective Disorder Department, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yongyin He
- grid.410737.60000 0000 8653 1072Clinical Laboratory, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jintai Cai
- grid.410737.60000 0000 8653 1072Clinical Laboratory, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China
| | - Mengxin Yang
- grid.410737.60000 0000 8653 1072Clinical Laboratory, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China
| | - Ying Xu
- grid.410737.60000 0000 8653 1072Clinical Laboratory, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China
| | - Le Hou
- grid.410737.60000 0000 8653 1072Department of Neurology, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yulong Lin
- Clinical Laboratory, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China.
| | - Haiying Liu
- Clinical Laboratory, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
25
|
Wang L, Wang B, Wu C, Wang J, Sun M. Autism Spectrum Disorder: Neurodevelopmental Risk Factors, Biological Mechanism, and Precision Therapy. Int J Mol Sci 2023; 24:ijms24031819. [PMID: 36768153 PMCID: PMC9915249 DOI: 10.3390/ijms24031819] [Citation(s) in RCA: 38] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 01/12/2023] [Accepted: 01/13/2023] [Indexed: 01/19/2023] Open
Abstract
Autism spectrum disorder (ASD) is a heterogeneous, behaviorally defined neurodevelopmental disorder. Over the past two decades, the prevalence of autism spectrum disorders has progressively increased, however, no clear diagnostic markers and specifically targeted medications for autism have emerged. As a result, neurobehavioral abnormalities, neurobiological alterations in ASD, and the development of novel ASD pharmacological therapy necessitate multidisciplinary collaboration. In this review, we discuss the development of multiple animal models of ASD to contribute to the disease mechanisms of ASD, as well as new studies from multiple disciplines to assess the behavioral pathology of ASD. In addition, we summarize and highlight the mechanistic advances regarding gene transcription, RNA and non-coding RNA translation, abnormal synaptic signaling pathways, epigenetic post-translational modifications, brain-gut axis, immune inflammation and neural loop abnormalities in autism to provide a theoretical basis for the next step of precision therapy. Furthermore, we review existing autism therapy tactics and limits and present challenges and opportunities for translating multidisciplinary knowledge of ASD into clinical practice.
Collapse
|
26
|
de Souza MM, Cenci AR, Teixeira KF, Machado V, Mendes Schuler MCG, Gonçalves AE, Paula Dalmagro A, André Cazarin C, Gomes Ferreira LL, de Oliveira AS, Andricopulo AD. DYRK1A Inhibitors and Perspectives for the Treatment of Alzheimer's Disease. Curr Med Chem 2023; 30:669-688. [PMID: 35726411 DOI: 10.2174/0929867329666220620162018] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 03/22/2022] [Accepted: 04/19/2022] [Indexed: 02/08/2023]
Abstract
BACKGROUND Alzheimer's disease (AD) is a chronic neurodegenerative disease and the most common form of dementia, especially in the elderly. Due to the increase in life expectancy, in recent years, there has been an excessive growth in the number of people affected by this disease, causing serious problems for health systems. In recent years, research has been intensified to find new therapeutic approaches that prevent the progression of the disease. In this sense, recent studies indicate that the dual-specificity tyrosine phosphorylation regulated kinase 1A (DYRK1A) gene, which is located on chromosome 21q22.2 and overexpressed in Down syndrome (DS), may play a significant role in developmental brain disorders and early onset neurodegeneration, neuronal loss and dementia in DS and AD. Inhibiting DYRK1A may serve to stop the phenotypic effects of its overexpression and, therefore, is a potential treatment strategy for the prevention of ageassociated neurodegeneration, including Alzheimer-type pathology. OBJECTIVE In this review, we investigate the contribution of DYRK1A inhibitors as potential anti-AD agents. METHODS A search in the literature to compile an in vitro dataset including IC50 values involving DYRK1A was performed from 2014 to the present day. In addition, we carried out structure-activity relationship studies based on in vitro and in silico data. RESULTS molecular modeling and enzyme kinetics studies indicate that DYRK1A may contribute to AD pathology through its proteolytic process, reducing its kinase specificity. CONCLUSION further evaluation of DYRK1A inhibitors may contribute to new therapeutic approaches for AD.
Collapse
Affiliation(s)
- Márcia Maria de Souza
- School of Health Sciences, Graduate Program in Pharmaceutical Sciences, UNIVALI, Rua Uruguai, 458 F6 lab 206 Campus I, Centro, Itajai, SC, 88302-202, Brazil
| | - Arthur Ribeiro Cenci
- Department of Exact Sciences and Education, Federal University of Santa Catarina, R. João Pessoa, 2750 - Velha, 89036-002, Blumenau, SC, Brazil
| | - Kerolain Faoro Teixeira
- Department of Exact Sciences and Education, Federal University of Santa Catarina, R. João Pessoa, 2750 - Velha, 89036-002, Blumenau, SC, Brazil
| | - Valkiria Machado
- Department of Exact Sciences and Education, Federal University of Santa Catarina, R. João Pessoa, 2750 - Velha, 89036-002, Blumenau, SC, Brazil
| | | | - Ana Elisa Gonçalves
- School of Health Sciences, Graduate Program in Pharmaceutical Sciences, UNIVALI, Rua Uruguai, 458 F6 lab 206 Campus I, Centro, Itajai, SC, 88302-202, Brazil
| | - Ana Paula Dalmagro
- School of Health Sciences, Graduate Program in Pharmaceutical Sciences, UNIVALI, Rua Uruguai, 458 F6 lab 206 Campus I, Centro, Itajai, SC, 88302-202, Brazil
| | - Camila André Cazarin
- School of Health Sciences, Graduate Program in Pharmaceutical Sciences, UNIVALI, Rua Uruguai, 458 F6 lab 206 Campus I, Centro, Itajai, SC, 88302-202, Brazil
| | - Leonardo Luiz Gomes Ferreira
- Laboratory of Medicinal and Computational Chemistry, Center for Research and Innovation in Biodiversity and Drug Discovery, Institute of Physics of São Carlos, University of São Paulo, São Carlos-SP, Brazil
| | - Aldo Sena de Oliveira
- Laboratory of Medicinal and Computational Chemistry, Center for Research and Innovation in Biodiversity and Drug Discovery, Institute of Physics of São Carlos, University of São Paulo, São Carlos-SP, Brazil
| | - Adriano Defini Andricopulo
- Laboratory of Medicinal and Computational Chemistry, Center for Research and Innovation in Biodiversity and Drug Discovery, Institute of Physics of São Carlos, University of São Paulo, São Carlos-SP, Brazil
| |
Collapse
|
27
|
Scott DN, Frank MJ. Adaptive control of synaptic plasticity integrates micro- and macroscopic network function. Neuropsychopharmacology 2023; 48:121-144. [PMID: 36038780 PMCID: PMC9700774 DOI: 10.1038/s41386-022-01374-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 06/23/2022] [Accepted: 06/24/2022] [Indexed: 11/09/2022]
Abstract
Synaptic plasticity configures interactions between neurons and is therefore likely to be a primary driver of behavioral learning and development. How this microscopic-macroscopic interaction occurs is poorly understood, as researchers frequently examine models within particular ranges of abstraction and scale. Computational neuroscience and machine learning models offer theoretically powerful analyses of plasticity in neural networks, but results are often siloed and only coarsely linked to biology. In this review, we examine connections between these areas, asking how network computations change as a function of diverse features of plasticity and vice versa. We review how plasticity can be controlled at synapses by calcium dynamics and neuromodulatory signals, the manifestation of these changes in networks, and their impacts in specialized circuits. We conclude that metaplasticity-defined broadly as the adaptive control of plasticity-forges connections across scales by governing what groups of synapses can and can't learn about, when, and to what ends. The metaplasticity we discuss acts by co-opting Hebbian mechanisms, shifting network properties, and routing activity within and across brain systems. Asking how these operations can go awry should also be useful for understanding pathology, which we address in the context of autism, schizophrenia and Parkinson's disease.
Collapse
Affiliation(s)
- Daniel N Scott
- Cognitive Linguistic, and Psychological Sciences, Brown University, Providence, RI, USA.
- Carney Institute for Brain Science, Brown University, Providence, RI, USA.
| | - Michael J Frank
- Cognitive Linguistic, and Psychological Sciences, Brown University, Providence, RI, USA.
- Carney Institute for Brain Science, Brown University, Providence, RI, USA.
| |
Collapse
|
28
|
Wei D, Hua XY, Zheng MX, Wu JJ, Xu JG. Effectiveness of robot-assisted virtual reality mirror therapy for upper limb motor dysfunction after stroke: study protocol for a single-center randomized controlled clinical trial. BMC Neurol 2022; 22:307. [PMID: 35996106 PMCID: PMC9396805 DOI: 10.1186/s12883-022-02836-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 08/14/2022] [Indexed: 11/29/2022] Open
Abstract
Background Upper limb motor dysfunction is a common sequela of stroke, and its clinical efficacy needs to be improved. This protocol describes a trial to verify the clinical efficacy of robot-assisted virtual reality mirror therapy (RAVRMT) in improving upper limb motor dysfunction in stroke patients, and to explore the central mechanism by using functional magnetic resonance imaging (fMRI). Methods This trial will be a single-center, assessor-blinded, randomized controlled clinical study. Thirty-two eligible patients will be randomly divided into 2 groups according to the ratio of 1:1, namely virtual reality mirror therapy (VRMT) group and robot-assisted virtual reality mirror therapy (RAVRMT) group. The interventions will be performed once a day for 4 weeks. Primary outcome is Fugl–Meyer motor function assessment-Upper Extremity (FMA-UE), secondary outcomes are the Montreal Cognitive Assessment (MoCA), activities of daily living (ADL), quality of life (QOL), the pain visual analogue scale (VAS-pain) and fMRI. Adverse events will be recorded, and severe adverse events will be used as criteria to discontinue the intervention. Discussion Combined application of robot-assisted therapy and virtual reality mirror therapy could theoretically activate mirror neuron system and reward circuits to a greater extent, but further high-quality research is needed. The results of this trial will determine whether RAVRMT could better improve upper limb motor dysfunction after stroke and explore its central mechanism using fMRI. Trial registration This trial was prospectively registered at ClinicalTrials.gov (ChiCTR2200061721; 01 July 2022).
Collapse
Affiliation(s)
- Dong Wei
- Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Xu-Yun Hua
- Shanghai University of Traditional Chinese Medicine Yueyang Hospital of Integrated Traditional Chinese Medicine and Western Medicine, Shanghai, 200437, China
| | - Mou-Xiong Zheng
- Shanghai University of Traditional Chinese Medicine Yueyang Hospital of Integrated Traditional Chinese Medicine and Western Medicine, Shanghai, 200437, China
| | - Jia-Jia Wu
- Shanghai University of Traditional Chinese Medicine Yueyang Hospital of Integrated Traditional Chinese Medicine and Western Medicine, Shanghai, 200437, China
| | - Jian-Guang Xu
- Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| |
Collapse
|
29
|
Yoo T, Joshi S, Prajapati S, Cho YS, Kim J, Park PH, Bae YC, Kim E, Kim SY. A Deficiency of the Psychiatric Risk Gene DLG2/PSD-93 Causes Excitatory Synaptic Deficits in the Dorsolateral Striatum. Front Mol Neurosci 2022; 15:938590. [PMID: 35966008 PMCID: PMC9370999 DOI: 10.3389/fnmol.2022.938590] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Accepted: 06/20/2022] [Indexed: 11/13/2022] Open
Abstract
Genetic variations resulting in the loss of function of the discs large homologs (DLG2)/postsynaptic density protein-93 (PSD-93) gene have been implicated in the increased risk for schizophrenia, intellectual disability, and autism spectrum disorders (ASDs). Previously, we have reported that mice lacking exon 14 of the Dlg2 gene (Dlg2–/– mice) display autistic-like behaviors, including social deficits and increased repetitive behaviors, as well as suppressed spontaneous excitatory postsynaptic currents in the striatum. However, the neural substrate underpinning such aberrant synaptic network activity remains unclear. Here, we found that the corticostriatal synaptic transmission was significantly impaired in Dlg2–/– mice, which did not seem attributed to defects in presynaptic releases of cortical neurons, but to the reduced number of functional synapses in the striatum, as manifested in the suppressed frequency of miniature excitatory postsynaptic currents in spiny projection neurons (SPNs). Using transmission electron microscopy, we found that both the density of postsynaptic densities and the fraction of perforated synapses were significantly decreased in the Dlg2–/– dorsolateral striatum. The density of dendritic spines was significantly reduced in striatal SPNs, but notably, not in the cortical pyramidal neurons of Dlg2–/– mice. Furthermore, a DLG2/PSD-93 deficiency resulted in the compensatory increases of DLG4/PSD-95 and decreases in the expression of TrkA in the striatum, but not particularly in the cortex. These results suggest that striatal dysfunction might play a role in the pathology of psychiatric disorders that are associated with a disruption of the Dlg2 gene.
Collapse
Affiliation(s)
- Taesun Yoo
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science, Daejeon, South Korea
| | - Shambhu Joshi
- College of Pharmacy, Yeungnam University, Gyeongsan, South Korea
| | | | - Yi Sul Cho
- Department of Anatomy and Neurobiology, School of Dentistry, Kyungpook National University, Daegu, South Korea
| | - Jinkyeong Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, South Korea
| | - Pil-Hoon Park
- College of Pharmacy, Yeungnam University, Gyeongsan, South Korea
| | - Yong Chul Bae
- Department of Anatomy and Neurobiology, School of Dentistry, Kyungpook National University, Daegu, South Korea
| | - Eunjoon Kim
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science, Daejeon, South Korea
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, South Korea
| | - Soo Young Kim
- College of Pharmacy, Yeungnam University, Gyeongsan, South Korea
- *Correspondence: Soo Young Kim,
| |
Collapse
|
30
|
Doi M, Li M, Usui N, Shimada S. Genomic Strategies for Understanding the Pathophysiology of Autism Spectrum Disorder. Front Mol Neurosci 2022; 15:930941. [PMID: 35813066 PMCID: PMC9263364 DOI: 10.3389/fnmol.2022.930941] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 06/06/2022] [Indexed: 11/25/2022] Open
Abstract
Recent breakthroughs in sequencing technology and technological developments have made it easier to analyze the entire human genome than ever before. In addition to disease-specific genetic mutations and chromosomal aberrations, epigenetic alterations in individuals can also be analyzed using genomics. Autism spectrum disorder (ASD) is a neurodevelopmental disorder (NDD) caused by genetic and/or environmental factors. More than a thousand genes associated with ASD have been identified which are known to be involved in brain development. However, it is difficult to decode the roles of ASD-associated genes without in vitro and in vivo validations, particularly in the process of brain development. In this review, we discuss genomic strategies for understanding the pathological mechanisms underlying ASD. For this purpose, we discuss ASD-associated genes and their functions, as well as analytical strategies and their strengths and weaknesses in cellular and animal models from a basic research perspective.
Collapse
Affiliation(s)
- Miyuki Doi
- Department of Neuroscience and Cell Biology, Graduate School of Medicine, Osaka University, Suita, Japan
- Addiction Research Unit, Osaka Psychiatric Research Center, Osaka Psychiatric Medical Center, Osaka, Japan
| | - Mengwei Li
- Department of Neuroscience and Cell Biology, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Noriyoshi Usui
- Department of Neuroscience and Cell Biology, Graduate School of Medicine, Osaka University, Suita, Japan
- Addiction Research Unit, Osaka Psychiatric Research Center, Osaka Psychiatric Medical Center, Osaka, Japan
- United Graduate School of Child Development, Osaka University, Suita, Japan
- Global Center for Medical Engineering and Informatics, Osaka University, Suita, Japan
- *Correspondence: Noriyoshi Usui
| | - Shoichi Shimada
- Department of Neuroscience and Cell Biology, Graduate School of Medicine, Osaka University, Suita, Japan
- Addiction Research Unit, Osaka Psychiatric Research Center, Osaka Psychiatric Medical Center, Osaka, Japan
- United Graduate School of Child Development, Osaka University, Suita, Japan
- Global Center for Medical Engineering and Informatics, Osaka University, Suita, Japan
| |
Collapse
|
31
|
Analysis of Faecal Microbiota and Small ncRNAs in Autism: Detection of miRNAs and piRNAs with Possible Implications in Host-Gut Microbiota Cross-Talk. Nutrients 2022; 14:nu14071340. [PMID: 35405953 PMCID: PMC9000903 DOI: 10.3390/nu14071340] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 03/07/2022] [Accepted: 03/21/2022] [Indexed: 02/01/2023] Open
Abstract
Intestinal microorganisms impact health by maintaining gut homeostasis and shaping the host immunity, while gut dysbiosis associates with many conditions, including autism, a complex neurodevelopmental disorder with multifactorial aetiology. In autism, gut dysbiosis correlates with symptom severity and is characterised by a reduced bacterial variability and a diminished beneficial commensal relationship. Microbiota can influence the expression of host microRNAs that, in turn, regulate the growth of intestinal bacteria by means of bidirectional host-gut microbiota cross-talk. We investigated possible interactions among intestinal microbes and between them and host transcriptional modulators in autism. To this purpose, we analysed, by "omics" technologies, faecal microbiome, mycobiome, and small non-coding-RNAs (particularly miRNAs and piRNAs) of children with autism and neurotypical development. Patients displayed gut dysbiosis related to a reduction of healthy gut micro- and mycobiota as well as up-regulated transcriptional modulators. The targets of dysregulated non-coding-RNAs are involved in intestinal permeability, inflammation, and autism. Furthermore, microbial families, underrepresented in patients, participate in the production of human essential metabolites negatively influencing the health condition. Here, we propose a novel approach to analyse faeces as a whole, and for the first time, we detected miRNAs and piRNAs in faecal samples of patients with autism.
Collapse
|
32
|
Caubit X, Gubellini P, Roubertoux PL, Carlier M, Molitor J, Chabbert D, Metwaly M, Salin P, Fatmi A, Belaidouni Y, Brosse L, Kerkerian-Le Goff L, Fasano L. Targeted Tshz3 deletion in corticostriatal circuit components segregates core autistic behaviors. Transl Psychiatry 2022; 12:106. [PMID: 35292625 PMCID: PMC8924251 DOI: 10.1038/s41398-022-01865-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 02/18/2022] [Accepted: 02/24/2022] [Indexed: 01/15/2023] Open
Abstract
We previously linked TSHZ3 haploinsufficiency to autism spectrum disorder (ASD) and showed that embryonic or postnatal Tshz3 deletion in mice results in behavioral traits relevant to the two core domains of ASD, namely social interaction deficits and repetitive behaviors. Here, we provide evidence that cortical projection neurons (CPNs) and striatal cholinergic interneurons (SCINs) are two main and complementary players in the TSHZ3-linked ASD syndrome. In the cerebral cortex, TSHZ3 is expressed in CPNs and in a proportion of GABAergic interneurons, but not in cholinergic interneurons or glial cells. In the striatum, TSHZ3 is expressed in all SCINs, while its expression is absent or partial in the other main brain cholinergic systems. We then characterized two new conditional knockout (cKO) models generated by crossing Tshz3flox/flox with Emx1-Cre (Emx1-cKO) or Chat-Cre (Chat-cKO) mice to decipher the respective role of CPNs and SCINs. Emx1-cKO mice show altered excitatory synaptic transmission onto CPNs and impaired plasticity at corticostriatal synapses, with neither cortical neuron loss nor abnormal layer distribution. These animals present social interaction deficits but no repetitive patterns of behavior. Chat-cKO mice exhibit no loss of SCINs but changes in the electrophysiological properties of these interneurons, associated with repetitive patterns of behavior without social interaction deficits. Therefore, dysfunction in either CPNs or SCINs segregates with a distinct ASD behavioral trait. These findings provide novel insights onto the implication of the corticostriatal circuitry in ASD by revealing an unexpected neuronal dichotomy in the biological background of the two core behavioral domains of this disorder.
Collapse
Affiliation(s)
- Xavier Caubit
- grid.462081.90000 0004 0598 4854Aix-Marseille Univ, CNRS, IBDM, UMR7288 Marseille, France
| | - Paolo Gubellini
- grid.462081.90000 0004 0598 4854Aix-Marseille Univ, CNRS, IBDM, UMR7288 Marseille, France
| | - Pierre L. Roubertoux
- grid.5399.60000 0001 2176 4817Aix-Marseille Univ, INSERM, MMG, UMR1251 Marseille, France
| | - Michèle Carlier
- grid.463724.00000 0004 0385 2989Aix-Marseille Univ, CNRS, LPC, UMR7290 Marseille, France
| | - Jordan Molitor
- grid.462081.90000 0004 0598 4854Aix-Marseille Univ, CNRS, IBDM, UMR7288 Marseille, France
| | - Dorian Chabbert
- grid.462081.90000 0004 0598 4854Aix-Marseille Univ, CNRS, IBDM, UMR7288 Marseille, France
| | - Mehdi Metwaly
- grid.462081.90000 0004 0598 4854Aix-Marseille Univ, CNRS, IBDM, UMR7288 Marseille, France
| | - Pascal Salin
- grid.462081.90000 0004 0598 4854Aix-Marseille Univ, CNRS, IBDM, UMR7288 Marseille, France
| | - Ahmed Fatmi
- grid.462081.90000 0004 0598 4854Aix-Marseille Univ, CNRS, IBDM, UMR7288 Marseille, France
| | - Yasmine Belaidouni
- grid.462081.90000 0004 0598 4854Aix-Marseille Univ, CNRS, IBDM, UMR7288 Marseille, France
| | - Lucie Brosse
- grid.462081.90000 0004 0598 4854Aix-Marseille Univ, CNRS, IBDM, UMR7288 Marseille, France
| | | | - Laurent Fasano
- Aix-Marseille Univ, CNRS, IBDM, UMR7288, Marseille, France.
| |
Collapse
|
33
|
Thabault M, Turpin V, Maisterrena A, Jaber M, Egloff M, Galvan L. Cerebellar and Striatal Implications in Autism Spectrum Disorders: From Clinical Observations to Animal Models. Int J Mol Sci 2022; 23:2294. [PMID: 35216408 PMCID: PMC8874522 DOI: 10.3390/ijms23042294] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 02/07/2022] [Accepted: 02/08/2022] [Indexed: 02/06/2023] Open
Abstract
Autism spectrum disorders (ASD) are complex conditions that stem from a combination of genetic, epigenetic and environmental influences during early pre- and postnatal childhood. The review focuses on the cerebellum and the striatum, two structures involved in motor, sensory, cognitive and social functions altered in ASD. We summarize clinical and fundamental studies highlighting the importance of these two structures in ASD. We further discuss the relation between cellular and molecular alterations with the observed behavior at the social, cognitive, motor and gait levels. Functional correlates regarding neuronal activity are also detailed wherever possible, and sexual dimorphism is explored pointing to the need to apprehend ASD in both sexes, as findings can be dramatically different at both quantitative and qualitative levels. The review focuses also on a set of three recent papers from our laboratory where we explored motor and gait function in various genetic and environmental ASD animal models. We report that motor and gait behaviors can constitute an early and quantitative window to the disease, as they often correlate with the severity of social impairments and loss of cerebellar Purkinje cells. The review ends with suggestions as to the main obstacles that need to be surpassed before an appropriate management of the disease can be proposed.
Collapse
Affiliation(s)
- Mathieu Thabault
- Laboratoire de Neurosciences Expérimentales et Cliniques, Institut National de la Santé et de la Recherche Médicale, Université de Poitiers, 86073 Poitiers, France; (M.T.); (V.T.); (A.M.); (M.J.); (M.E.)
| | - Valentine Turpin
- Laboratoire de Neurosciences Expérimentales et Cliniques, Institut National de la Santé et de la Recherche Médicale, Université de Poitiers, 86073 Poitiers, France; (M.T.); (V.T.); (A.M.); (M.J.); (M.E.)
| | - Alexandre Maisterrena
- Laboratoire de Neurosciences Expérimentales et Cliniques, Institut National de la Santé et de la Recherche Médicale, Université de Poitiers, 86073 Poitiers, France; (M.T.); (V.T.); (A.M.); (M.J.); (M.E.)
| | - Mohamed Jaber
- Laboratoire de Neurosciences Expérimentales et Cliniques, Institut National de la Santé et de la Recherche Médicale, Université de Poitiers, 86073 Poitiers, France; (M.T.); (V.T.); (A.M.); (M.J.); (M.E.)
- Centre Hospitalier Universitaire de Poitiers, 86021 Poitiers, France
| | - Matthieu Egloff
- Laboratoire de Neurosciences Expérimentales et Cliniques, Institut National de la Santé et de la Recherche Médicale, Université de Poitiers, 86073 Poitiers, France; (M.T.); (V.T.); (A.M.); (M.J.); (M.E.)
- Centre Hospitalier Universitaire de Poitiers, 86021 Poitiers, France
| | - Laurie Galvan
- Laboratoire de Neurosciences Expérimentales et Cliniques, Institut National de la Santé et de la Recherche Médicale, Université de Poitiers, 86073 Poitiers, France; (M.T.); (V.T.); (A.M.); (M.J.); (M.E.)
| |
Collapse
|
34
|
Meng Y, Xu D, Zhang W, Meng W, Lan X, Wang X, Li M, Zhang X, Zhao Y, Yang H, Zhang R, Zhen Z. Effect of Early Swimming on the Behavior and Striatal Transcriptome of the Shank3 Knockout Rat Model of Autism. Neuropsychiatr Dis Treat 2022; 18:681-694. [PMID: 35387206 PMCID: PMC8979754 DOI: 10.2147/ndt.s357338] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 03/17/2022] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Autism spectrum disorder (ASD) is a developmental disorder characterized by social behavior deficits and stereotyped behaviors in childhood that lacks satisfactory medical intervention. Early swimming intervention is a noninvasive method combining enriched environment and exercise, which has been proven to improve brain development in young children and to treat neurodevelopmental diseases. METHODS In this study, we tested the autism-like behavior of rats with deletions in exons 11-21 of the Shank3 gene and evaluated the effect of early swimming intervention (from postnatal day 8 to 60) on the behavior of this animal model of autism. In addition, the transcriptomes of the striatal tissues of wild-type, Shank3 knockout and Shank3 knockout swimming groups rats were analyzed. RESULTS Shank3 knockout rats exhibit core symptoms of autism, and early swimming improved the social and stereotyped behaviors in this autism rat model. Transcriptomics results revealed that compared to the wild-type group, 291 differentially expressed genes (DEGs) were identified in the striatum of the Shank3 knockout group. Compared to Shank3 knockout group, 534 DEGs were identified in the striatum of Shank3 knockout swimming group. The DEGs annotated by Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway shows that the impacts of Shank3 deletion were primarily reflected in synaptic structure, development, morphology, receptor function and signaling, and swimming primarily changed the terms related to the synapses in the striatum of Shank3 knockout rats, including the morphology, structure, composition, development and regulation of synapses. CONCLUSION Early swimming intervention can ameliorate behavioral abnormalities caused by Shank3 knockout, by a mechanism that may involve the process of striatal synaptic development and should be further investigated.
Collapse
Affiliation(s)
- Yunchen Meng
- College of P.E and Sports, Beijing Normal University, Beijing, People's Republic of China
| | - Dan Xu
- College of P.E and Sports, Beijing Normal University, Beijing, People's Republic of China
| | - Weinan Zhang
- College of P.E and Sports, Beijing Normal University, Beijing, People's Republic of China
| | - Wenshu Meng
- College of Life Sciences, Beijing Normal University, Beijing, People's Republic of China
| | - Xingyu Lan
- Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing, People's Republic of China
| | - Xiaoxi Wang
- Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing, People's Republic of China
| | - Mingjuan Li
- Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing, People's Republic of China
| | - Xiaoyan Zhang
- College of P.E and Sports, Beijing Normal University, Beijing, People's Republic of China
| | - Yu Zhao
- College of P.E and Sports, Beijing Normal University, Beijing, People's Republic of China
| | - Haodong Yang
- College of P.E and Sports, Beijing Normal University, Beijing, People's Republic of China
| | - Rong Zhang
- Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing, People's Republic of China.,Neuroscience Research Institute, Peking University, Beijing, People's Republic of China.,Key Laboratory for Neuroscience, Ministry of Education/National Health and Family Planning Commission, Peking University, Beijing, People's Republic of China.,Autism Research Center of Peking University Health Science Center, Beijing, People's Republic of China
| | - Zhiping Zhen
- College of P.E and Sports, Beijing Normal University, Beijing, People's Republic of China
| |
Collapse
|
35
|
Fernàndez-Castillo N, Cabana-Domínguez J, Kappel DB, Torrico B, Weber H, Lesch KP, Lao O, Reif A, Cormand B. Exploring the Contribution to ADHD of Genes Involved in Mendelian Disorders Presenting with Hyperactivity and/or Inattention. Genes (Basel) 2021; 13:93. [PMID: 35052433 PMCID: PMC8775234 DOI: 10.3390/genes13010093] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 12/16/2021] [Accepted: 12/18/2021] [Indexed: 12/26/2022] Open
Abstract
Attention-deficit hyperactivity disorder (ADHD) is a complex neurodevelopmental disorder characterized by hyperactivity, impulsivity, and/or inattention, which are symptoms also observed in many rare genetic disorders. We searched for genes involved in Mendelian disorders presenting with ADHD symptoms in the Online Mendelian Inheritance in Man (OMIM) database, to curate a list of new candidate risk genes for ADHD. We explored the enrichment of functions and pathways in this gene list, and tested whether rare or common variants in these genes are associated with ADHD or with its comorbidities. We identified 139 genes, causal for 137 rare disorders, mainly related to neurodevelopmental and brain function. Most of these Mendelian disorders also present with other psychiatric traits that are often comorbid with ADHD. Using whole exome sequencing (WES) data from 668 ADHD cases, we found rare variants associated with the dimension of the severity of inattention symptoms in three genes: KIF11, WAC, and CRBN. Then, we focused on common variants and identified six genes associated with ADHD (in 19,099 cases and 34,194 controls): MANBA, UQCC2, HIVEP2, FOPX1, KANSL1, and AUH. Furthermore, HIVEP2, FOXP1, and KANSL1 were nominally associated with autism spectrum disorder (ASD) (18,382 cases and 27,969 controls), as well as HIVEP2 with anxiety (7016 cases and 14,475 controls), and FOXP1 with aggression (18,988 individuals), which is in line with the symptomatology of the rare disorders they are responsible for. In conclusion, inspecting Mendelian disorders and the genes responsible for them constitutes a valuable approach for identifying new risk genes and the mechanisms of complex disorders.
Collapse
Affiliation(s)
- Noèlia Fernàndez-Castillo
- Departament de Genètica, Microbiologia i Estadística, Facultat de Biologia, Universitat de Barcelona, 08028 Barcelona, Spain; (B.T.); (B.C.)
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Institut de Biomedicina de la Universitat de Barcelona (IBUB), 08028 Barcelona, Spain
- Institut de Recerca Sant Joan de Déu (IR-SJD), 08950 Esplugues de Llobregat, Spain
| | - Judit Cabana-Domínguez
- Departament de Genètica, Microbiologia i Estadística, Facultat de Biologia, Universitat de Barcelona, 08028 Barcelona, Spain; (B.T.); (B.C.)
- Psychiatric Genetics Unit, Group of Psychiatry, Mental Health and Addiction, Vall d’Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona, 08035 Barcelona, Spain
- Department of Psychiatry, Hospital Universitari Vall d’Hebron, 08035 Barcelona, Spain
- Biomedical Network Research Centre on Mental Health (CIBERSAM), 28029 Madrid, Spain
| | - Djenifer B. Kappel
- Division of Psychological Medicine and Clinical Neurosciences, MRC Centre for Neuropsychiatric Genetics and Genomics, School of Medicine, Cardiff University, Cardiff CF10 3AT, UK;
| | - Bàrbara Torrico
- Departament de Genètica, Microbiologia i Estadística, Facultat de Biologia, Universitat de Barcelona, 08028 Barcelona, Spain; (B.T.); (B.C.)
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Institut de Biomedicina de la Universitat de Barcelona (IBUB), 08028 Barcelona, Spain
- Institut de Recerca Sant Joan de Déu (IR-SJD), 08950 Esplugues de Llobregat, Spain
| | - Heike Weber
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital Frankfurt, 60590 Frankfurt, Germany; (H.W.); (A.R.)
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital Würzburg, 97080 Wurzburg, Germany
| | - Klaus-Peter Lesch
- Division of Molecular Psychiatry, Center of Mental Health, University of Würzburg, 97080 Wurzburg, Germany;
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, 6221 LK Maastricht, The Netherlands
- Laboratory of Psychiatric Neurobiology, Institute of Molecular Medicine, I.M Sechenov First Moscow State Medical University, 119435 Moscow, Russia
| | - Oscar Lao
- CNAG-CRG, Centre for Genomic Regulation (CRG), 08028 Barcelona, Spain;
- Barcelona Institute of Science and Technology (BIST), 08036 Barcelona, Spain
- Universitat Pompeu Fabra (UPF), 08002 Barcelona, Spain
| | - Andreas Reif
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital Frankfurt, 60590 Frankfurt, Germany; (H.W.); (A.R.)
| | - Bru Cormand
- Departament de Genètica, Microbiologia i Estadística, Facultat de Biologia, Universitat de Barcelona, 08028 Barcelona, Spain; (B.T.); (B.C.)
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Institut de Biomedicina de la Universitat de Barcelona (IBUB), 08028 Barcelona, Spain
- Institut de Recerca Sant Joan de Déu (IR-SJD), 08950 Esplugues de Llobregat, Spain
| |
Collapse
|
36
|
Riemersma IW, Havekes R, Kas MJH. Spatial and Temporal Gene Function Studies in Rodents: Towards Gene-Based Therapies for Autism Spectrum Disorder. Genes (Basel) 2021; 13:28. [PMID: 35052369 PMCID: PMC8774890 DOI: 10.3390/genes13010028] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 12/06/2021] [Accepted: 12/20/2021] [Indexed: 12/26/2022] Open
Abstract
Autism spectrum disorder (ASD) is a complex neurodevelopmental condition that is characterized by differences in social interaction, repetitive behaviors, restricted interests, and sensory differences beginning early in life. Especially sensory symptoms are highly correlated with the severity of other behavioral differences. ASD is a highly heterogeneous condition on multiple levels, including clinical presentation, genetics, and developmental trajectories. Over a thousand genes have been implicated in ASD. This has facilitated the generation of more than two hundred genetic mouse models that are contributing to understanding the biological underpinnings of ASD. Since the first symptoms already arise during early life, it is especially important to identify both spatial and temporal gene functions in relation to the ASD phenotype. To further decompose the heterogeneity, ASD-related genes can be divided into different subgroups based on common functions, such as genes involved in synaptic function. Furthermore, finding common biological processes that are modulated by this subgroup of genes is essential for possible patient stratification and the development of personalized early treatments. Here, we review the current knowledge on behavioral rodent models of synaptic dysfunction by focusing on behavioral phenotypes, spatial and temporal gene function, and molecular targets that could lead to new targeted gene-based therapy.
Collapse
Affiliation(s)
| | | | - Martien J. H. Kas
- Groningen Institute for Evolutionary Life Sciences, Neurobiology, University of Groningen, Nijenborgh 7, 9747 AG Groningen, The Netherlands; (I.W.R.); (R.H.)
| |
Collapse
|
37
|
Postsynaptic autism spectrum disorder genes and synaptic dysfunction. Neurobiol Dis 2021; 162:105564. [PMID: 34838666 DOI: 10.1016/j.nbd.2021.105564] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 11/17/2021] [Accepted: 11/23/2021] [Indexed: 12/20/2022] Open
Abstract
This review provides an overview of the synaptic dysfunction of neuronal circuits and the ensuing behavioral alterations caused by mutations in autism spectrum disorder (ASD)-linked genes directly or indirectly affecting the postsynaptic neuronal compartment. There are plenty of ASD risk genes, that may be broadly grouped into those involved in gene expression regulation (epigenetic regulation and transcription) and genes regulating synaptic activity (neural communication and neurotransmission). Notably, the effects mediated by ASD-associated genes can vary extensively depending on the developmental time and/or subcellular site of expression. Therefore, in order to gain a better understanding of the mechanisms of disruptions in postsynaptic function, an effort to better model ASD in experimental animals is required to improve standardization and increase reproducibility within and among studies. Such an effort holds promise to provide deeper insight into the development of these disorders and to improve the translational value of preclinical studies.
Collapse
|
38
|
Nadeem MS, Hosawi S, Alshehri S, Ghoneim MM, Imam SS, Murtaza BN, Kazmi I. Symptomatic, Genetic, and Mechanistic Overlaps between Autism and Alzheimer's Disease. Biomolecules 2021; 11:1635. [PMID: 34827633 PMCID: PMC8615882 DOI: 10.3390/biom11111635] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 10/26/2021] [Accepted: 11/01/2021] [Indexed: 02/02/2023] Open
Abstract
Autism spectrum disorder (ASD) and Alzheimer's disease (AD) are neurodevelopmental and neurodegenerative disorders affecting two opposite ends of life span, i.e., childhood and old age. Both disorders pose a cumulative threat to human health, with the rate of incidences increasing considerably worldwide. In the context of recent developments, we aimed to review correlated symptoms and genetics, and overlapping aspects in the mechanisms of the pathogenesis of ASD and AD. Dementia, insomnia, and weak neuromuscular interaction, as well as communicative and cognitive impairments, are shared symptoms. A number of genes and proteins linked with both disorders have been tabulated, including MECP2, ADNP, SCN2A, NLGN, SHANK, PTEN, RELN, and FMR1. Theories about the role of neuron development, processing, connectivity, and levels of neurotransmitters in both disorders have been discussed. Based on the recent literature, the roles of FMRP (Fragile X mental retardation protein), hnRNPC (heterogeneous ribonucleoprotein-C), IRP (Iron regulatory proteins), miRNAs (MicroRNAs), and α-, β0, and γ-secretases in the posttranscriptional regulation of cellular synthesis and processing of APP (amyloid-β precursor protein) have been elaborated to describe the parallel and overlapping routes and mechanisms of ASD and AD pathogenesis. However, the interactive role of genetic and environmental factors, oxidative and metal ion stress, mutations in the associated genes, and alterations in the related cellular pathways in the development of ASD and AD needs further investigation.
Collapse
Affiliation(s)
- Muhammad Shahid Nadeem
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (M.S.N.); (S.H.)
| | - Salman Hosawi
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (M.S.N.); (S.H.)
| | - Sultan Alshehri
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (S.A.); (S.S.I.)
| | - Mohammed M. Ghoneim
- Department of Pharmacy Practice, College of Pharmacy, AlMaarefa University, Ad Diriyah 13713, Saudi Arabia;
| | - Syed Sarim Imam
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (S.A.); (S.S.I.)
| | - Bibi Nazia Murtaza
- Department of Zoology, Abbottabad University of Science and Technology (AUST), Abbottabad 22310, Pakistan;
| | - Imran Kazmi
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (M.S.N.); (S.H.)
| |
Collapse
|
39
|
Benthall KN, Cording KR, Agopyan-Miu AHCW, Wong CD, Chen EY, Bateup HS. Loss of Tsc1 from striatal direct pathway neurons impairs endocannabinoid-LTD and enhances motor routine learning. Cell Rep 2021; 36:109511. [PMID: 34380034 PMCID: PMC8404511 DOI: 10.1016/j.celrep.2021.109511] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 05/28/2021] [Accepted: 07/21/2021] [Indexed: 02/08/2023] Open
Abstract
Tuberous sclerosis complex (TSC) is a neurodevelopmental disorder that often presents with psychiatric conditions, including autism spectrum disorder (ASD). ASD is characterized by restricted, repetitive, and inflexible behaviors, which may result from abnormal activity in striatal circuits that mediate motor learning and action selection. To test whether altered striatal activity contributes to aberrant motor behaviors in the context of TSC, we conditionally deleted Tsc1 from direct or indirect pathway striatal projection neurons (dSPNs or iSPNs, respectively). We find that dSPN-specific loss of Tsc1 impairs endocannabinoid-mediated long-term depression (eCB-LTD) at cortico-dSPN synapses and strongly enhances corticostriatal synaptic drive, which is not observed in iSPNs. dSPN-Tsc1 KO, but not iSPN-Tsc1 KO, mice show enhanced motor learning, a phenotype observed in several mouse models of ASD. These findings demonstrate that dSPNs are particularly sensitive to Tsc1 loss and suggest that enhanced corticostriatal activation may contribute to altered motor behaviors in TSC.
Collapse
Affiliation(s)
- Katelyn N Benthall
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Katherine R Cording
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA 94720, USA
| | | | - Corinna D Wong
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Emily Y Chen
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Helen S Bateup
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA; Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA 94720, USA; Chan Zuckerberg Biohub, San Francisco, CA 94158, USA.
| |
Collapse
|
40
|
Caubit X, Arbeille E, Chabbert D, Desprez F, Messak I, Fatmi A, Habermann B, Gubellini P, Fasano L. Camk2a-Cre and Tshz3 Expression in Mouse Striatal Cholinergic Interneurons: Implications for Autism Spectrum Disorder. Front Genet 2021; 12:683959. [PMID: 34349780 PMCID: PMC8328143 DOI: 10.3389/fgene.2021.683959] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 06/21/2021] [Indexed: 12/24/2022] Open
Abstract
Camk2a-Cre mice have been widely used to study the postnatal function of several genes in forebrain projection neurons, including cortical projection neurons (CPNs) and striatal medium-sized spiny neurons (MSNs). We linked heterozygous deletion of TSHZ3/Tshz3 gene to autism spectrum disorder (ASD) and used Camk2a-Cre mice to investigate the postnatal function of Tshz3, which is expressed by CPNs but not MSNs. Recently, single-cell transcriptomics of the adult mouse striatum revealed the expression of Camk2a in interneurons and showed Tshz3 expression in striatal cholinergic interneurons (SCINs), which are attracting increasing interest in the field of ASD. These data and the phenotypic similarity between the mice with Tshz3 haploinsufficiency and Camk2a-Cre-dependent conditional deletion of Tshz3 (Camk2a-cKO) prompted us to better characterize the expression of Tshz3 and the activity of Camk2a-Cre transgene in the striatum. Here, we show that the great majority of Tshz3-expressing cells are SCINs and that all SCINs express Tshz3. Using lineage tracing, we demonstrate that the Camk2a-Cre transgene is expressed in the SCIN lineage where it can efficiently elicit the deletion of the Tshz3-floxed allele. Moreover, transcriptomic and bioinformatic analysis in Camk2a-cKO mice showed dysregulated striatal expression of a number of genes, including genes whose human orthologues are associated with ASD and synaptic signaling. These findings identifying the expression of the Camk2a-Cre transgene in SCINs lineage lead to a reappraisal of the interpretation of experiments using Camk2a-Cre-dependent gene manipulations. They are also useful to decipher the cellular and molecular substrates of the ASD-related behavioral abnormalities observed in Tshz3 mouse models.
Collapse
Affiliation(s)
- Xavier Caubit
- Aix-Marseille University, CNRS, IBDM, UMR 7288, Marseille, France
| | - Elise Arbeille
- Aix-Marseille University, CNRS, IBDM, UMR 7288, Marseille, France
| | - Dorian Chabbert
- Aix-Marseille University, CNRS, IBDM, UMR 7288, Marseille, France
| | - Florence Desprez
- Aix-Marseille University, CNRS, IBDM, UMR 7288, Marseille, France
| | - Imane Messak
- Aix-Marseille University, CNRS, IBDM, UMR 7288, Marseille, France
| | - Ahmed Fatmi
- Aix-Marseille University, CNRS, IBDM, UMR 7288, Marseille, France
| | - Bianca Habermann
- Aix-Marseille University, CNRS, IBDM, UMR 7288, Marseille, France
| | - Paolo Gubellini
- Aix-Marseille University, CNRS, IBDM, UMR 7288, Marseille, France
| | - Laurent Fasano
- Aix-Marseille University, CNRS, IBDM, UMR 7288, Marseille, France
| |
Collapse
|
41
|
Cytogenetic and Array-CGH Characterization of a Simple Case of Reciprocal t(3;10) Translocation Reveals a Hidden Deletion at 5q12. Genes (Basel) 2021; 12:genes12060877. [PMID: 34200357 PMCID: PMC8226940 DOI: 10.3390/genes12060877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 06/01/2021] [Accepted: 06/03/2021] [Indexed: 11/16/2022] Open
Abstract
Chromosome deletions, including band 5q12, have rarely been reported and have been associated with a wide range of clinical manifestations, such as postnatal growth retardation, intellectual disability, hyperactivity, nonspecific ocular defects, facial dysmorphism, and epilepsy. In this study, we describe for the first time a child with growth retardation in which we identified a balanced t(3;10) translocation by conventional cytogenetic analysis in addition to an 8.6 Mb 5q12 deletion through array-CGH. Our results show that the phenotypic abnormalities of a case that had been interpreted as "balanced" by conventional cytogenetics are mainly due to a cryptic deletion, highlighting the need for molecular investigation in subjects with an abnormal phenotype before assuming the cause is an apparently simple cytogenetic rearrangement. Finally, we identify PDE4D and PIK3R1 genes as the two major candidates responsible for the clinical features expressed in our patient.
Collapse
|
42
|
Garozzo MT, Caruso D, La Mendola FMC, Di Nora A, Romano K, Leonardi R, Falsaperla R, Zanghì A, Praticò AD. SYNGAP1 and Its Related Epileptic Syndromes. JOURNAL OF PEDIATRIC NEUROLOGY 2021. [DOI: 10.1055/s-0041-1727144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
AbstractSynaptic Ras GTPase-activating protein 1 (SYNGAP1) is abundantly expressed in the postsynaptic space in brain tissue and has a crucial role in the regulation of the excitatory/inhibitory balance and in brain development. It is estimated that SYNGAP1 loss of function variants have an incidence of 1 to 4/10,000 individuals, mostly occurring de novo, even if few cases of vertical transmission of mosaic mutations have been reported. Loss-of-function mutations within this gene have been related with an epileptic encephalopathy characterized by eyelid myoclonia with absences (EMA) and myoclonic-atonic seizures (MAE) with early onset, commonly resistant to antiepileptic drugs (AED). Epilepsy is often associated with other clinical features, including truncal and/or facial hypotonia and/or ataxia with a wide-based and unsteady gate. Other clinical signs are intellectual disability, developmental delay, and behavioral and speech impairment, in a context of a normal neuroimaging study. In selected cases, dysmorphic features, skeletal abnormalities, and eye involvement are also described. The diagnosis of the disorder is usually established by multigene panel and, in unsolved cases, by exome sequencing. Management of the affected individuals involves different specialists and is mainly symptomatic. No clinical trials about the efficacy of AED in SYNGAP1 encephalopathy have been performed yet and Lamotrigine and valproate are commonly prescribed. In more than half of cases, however, epilepsy is refractory to AED.
Collapse
Affiliation(s)
- Maria Teresa Garozzo
- Unit of Pediatric and Pediatric Emergency, Hospital “Cannizzaro,” Catania, Italy
| | - Daniela Caruso
- Pediatrics Postgraduate Residency Program, Department of Clinical and Experimental Medicine, Section of Pediatrics and Child Neuropsychiatry, University of Catania, Catania, Italy
| | | | - Alessandra Di Nora
- Pediatrics Postgraduate Residency Program, Department of Clinical and Experimental Medicine, Section of Pediatrics and Child Neuropsychiatry, University of Catania, Catania, Italy
| | | | - Roberta Leonardi
- Unit of Rare Diseases of the Nervous System in Childhood, Department of Clinical and Experimental Medicine, Section of Pediatrics and Child Neuropsychiatry, University of Catania, Catania, Italy
| | - Raffaele Falsaperla
- Unit of Pediatrics and Pediatric Emergency, University Hospital “Policlinico Rodolico-San Marco,” Catania, Italy
- Unit of Neonatal Intensive Care and Neonatology, University Hospital “Policlinico Rodolico-San Marco,” Catania, Italy
| | - Antonio Zanghì
- Department of Medical and Surgical Sciences and Advanced Technology “G.F. Ingrassia,” University of Catania, Catania, Italy
| | - Andrea D. Praticò
- Unit of Rare Diseases of the Nervous System in Childhood, Department of Clinical and Experimental Medicine, Section of Pediatrics and Child Neuropsychiatry, University of Catania, Catania, Italy
| |
Collapse
|
43
|
Levchenko A, Kanapin A, Samsonova A, Fedorenko OY, Kornetova EG, Nurgaliev T, Mazo GE, Semke AV, Kibitov AO, Bokhan NA, Gainetdinov RR, Ivanova SA. A genome-wide association study identifies a gene network associated with paranoid schizophrenia and antipsychotics-induced tardive dyskinesia. Prog Neuropsychopharmacol Biol Psychiatry 2021; 105:110134. [PMID: 33065217 DOI: 10.1016/j.pnpbp.2020.110134] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 09/10/2020] [Accepted: 10/06/2020] [Indexed: 02/06/2023]
Abstract
In the present study we conducted a genome-wide association study (GWAS) in a cohort of 505 patients with paranoid schizophrenia (SCZ), of which 95 had tardive dyskinesia (TD), and 503 healthy controls. Using data generated by the PsychENCODE Consortium (PEC) and other bioinformatic databases, we revealed a gene network, implicated in neurodevelopment and brain function, associated with both these disorders. Almost all these genes are in gene or isoform co-expression PEC network modules important for the functioning of the brain; the activity of these networks is also altered in SCZ, bipolar disorder and autism spectrum disorders. The associated PEC network modules are enriched for gene ontology terms relevant to the brain development and function (CNS development, neuron development, axon ensheathment, synapse, synaptic vesicle cycle, and signaling receptor activity) and to the immune system (inflammatory response). Results of the present study suggest that orofacial and limbtruncal types of TD seem to share the molecular network with SCZ. Paranoid SCZ and abnormal involuntary movements that indicate the orofacial type of TD are associated with the same genomic loci on chromosomes 3p22.2, 8q21.13, and 13q14.2. The limbtruncal type of TD is associated with a locus on chromosome 3p13 where the best functional candidate is FOXP1, a high-confidence SCZ gene. The results of this study shed light on common pathogenic mechanisms for SCZ and TD, and indicate that the pathogenesis of the orofacial and limbtruncal types of TD might be driven by interacting genes implicated in neurodevelopment.
Collapse
Affiliation(s)
- Anastasia Levchenko
- Theodosius Dobzhansky Center for Genome Bioinformatics, Saint Petersburg State University, Saint Petersburg, Russia.
| | - Alexander Kanapin
- Theodosius Dobzhansky Center for Genome Bioinformatics, Saint Petersburg State University, Saint Petersburg, Russia
| | - Anastasia Samsonova
- Theodosius Dobzhansky Center for Genome Bioinformatics, Saint Petersburg State University, Saint Petersburg, Russia
| | - Olga Yu Fedorenko
- Mental Health Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia; National Research Tomsk Polytechnic University, Tomsk, Russia
| | - Elena G Kornetova
- Mental Health Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia; Siberian State Medical University, Tomsk, Russia
| | | | - Galina E Mazo
- Department of Endocrine Psychiatry, V.M. Bekhterev National Medical Research Center for Psychiatry and Neurology, Saint Petersburg, Russia
| | - Arkadiy V Semke
- Mental Health Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia
| | - Alexander O Kibitov
- Department of Endocrine Psychiatry, V.M. Bekhterev National Medical Research Center for Psychiatry and Neurology, Saint Petersburg, Russia; Laboratory of Molecular Genetics, Serbsky National Medical Research Center on Psychiatry and Addictions, Moscow, Russia
| | - Nikolay A Bokhan
- Mental Health Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia; Siberian State Medical University, Tomsk, Russia; National Research Tomsk State University, Tomsk, Russia
| | - Raul R Gainetdinov
- Institute of Translational Biomedicine, Saint Petersburg State University, Saint Petersburg, Russia
| | - Svetlana A Ivanova
- Mental Health Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia; National Research Tomsk Polytechnic University, Tomsk, Russia; Siberian State Medical University, Tomsk, Russia
| |
Collapse
|
44
|
Gandhi T, Lee CC. Neural Mechanisms Underlying Repetitive Behaviors in Rodent Models of Autism Spectrum Disorders. Front Cell Neurosci 2021; 14:592710. [PMID: 33519379 PMCID: PMC7840495 DOI: 10.3389/fncel.2020.592710] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 12/09/2020] [Indexed: 12/15/2022] Open
Abstract
Autism spectrum disorder (ASD) is comprised of several conditions characterized by alterations in social interaction, communication, and repetitive behaviors. Genetic and environmental factors contribute to the heterogeneous development of ASD behaviors. Several rodent models display ASD-like phenotypes, including repetitive behaviors. In this review article, we discuss the potential neural mechanisms involved in repetitive behaviors in rodent models of ASD and related neuropsychiatric disorders. We review signaling pathways, neural circuits, and anatomical alterations in rodent models that display robust stereotypic behaviors. Understanding the mechanisms and circuit alterations underlying repetitive behaviors in rodent models of ASD will inform translational research and provide useful insight into therapeutic strategies for the treatment of repetitive behaviors in ASD and other neuropsychiatric disorders.
Collapse
Affiliation(s)
- Tanya Gandhi
- Department of Comparative Biomedical Sciences, Louisiana State University School of Veterinary Medicine, Baton Rouge, LA, United States
| | | |
Collapse
|
45
|
Wang N, Zhao Y, Gao J. Association Between Peripheral Blood Levels of Vitamin A and Autism Spectrum Disorder in Children: A Meta-Analysis. Front Psychiatry 2021; 12:742937. [PMID: 34658977 PMCID: PMC8515042 DOI: 10.3389/fpsyt.2021.742937] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Accepted: 09/06/2021] [Indexed: 11/25/2022] Open
Abstract
Vitamin A is an essential fat-soluble micronutrient that plays important roles in a surprisingly wide variety of biological processes from early growth and development to brain maintenance. Numerous clinical studies have been conducted to explore the relationship between peripheral vitamin A levels and autism spectrum disorder (ASD), but the results of these studies are controversial. Therefore, we assessed the association between peripheral vitamin A levels and ASD in the present meta-analysis. Relevant records were retrieved through the Embase, Web of Knowledge and PubMed databases up to 13 November 2020. Reference lists were also searched and analyzed. Hedges' g with its corresponding 95% confidence interval (CI) was used to assess the association between peripheral vitamin A levels and ASD. A fixed or random effects model was selected according to a heterogeneity test in overall and subgroup analyses. Five records (six studies) with 935 ASD children and 516 healthy children were included in the present study. Significantly decreased peripheral vitamin A concentrations were observed in ASD children compared with healthy children (Hedges' g = -0.600, 95% CI -1.153 to -0.048, P = 0.033). A similar result was also obtained after removing the studies identified by Galbraith plots. In addition, no obvious publication bias was found in the meta-analysis. The findings of our meta-analysis suggested decreased peripheral vitamin A levels in ASD children compared with healthy children. Further investigations into the effects of vitamin A on the development of ASD are warranted.
Collapse
Affiliation(s)
- Ni Wang
- Nursing Office of Beijing Road Medical District, General Hospital of Xinjiang Military Region, Wulumuqi, China
| | | | - Junwei Gao
- Department of Developmental Neuropsychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing, China
| |
Collapse
|
46
|
Two Players in the Field: Hierarchical Model of Interaction between the Dopamine and Acetylcholine Signaling Systems in the Striatum. Biomedicines 2021; 9:biomedicines9010025. [PMID: 33401461 PMCID: PMC7824505 DOI: 10.3390/biomedicines9010025] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 12/25/2020] [Accepted: 12/29/2020] [Indexed: 12/13/2022] Open
Abstract
Tight interactions exist between dopamine and acetylcholine signaling in the striatum. Dopaminergic neurons express muscarinic and nicotinic receptors, and cholinergic interneurons express dopamine receptors. All neurons in the striatum are pacemakers. An increase in dopamine release is activated by stopping acetylcholine release. The coordinated timing or synchrony of the direct and indirect pathways is critical for refined movements. Changes in neurotransmitter ratios are considered a prominent factor in Parkinson’s disease. In general, drugs increase striatal dopamine release, and others can potentiate both dopamine and acetylcholine release. Both neurotransmitters and their receptors show diurnal variations. Recently, it was observed that reward function is modulated by the circadian system, and behavioral changes (hyperactivity and hypoactivity during the light and dark phases, respectively) are present in an animal model of Parkinson’s disease. The striatum is one of the key structures responsible for increased locomotion in the active (dark) period in mice lacking M4 muscarinic receptors. Thus, we propose here a hierarchical model of the interaction between dopamine and acetylcholine signaling systems in the striatum. The basis of this model is their functional morphology. The next highest mode of interaction between these two neurotransmitter systems is their interaction at the neurotransmitter/receptor/signaling level. Furthermore, these interactions contribute to locomotor activity regulation and reward behavior, and the topmost level of interaction represents their biological rhythmicity.
Collapse
|
47
|
Filice F, Janickova L, Henzi T, Bilella A, Schwaller B. The Parvalbumin Hypothesis of Autism Spectrum Disorder. Front Cell Neurosci 2020; 14:577525. [PMID: 33390904 PMCID: PMC7775315 DOI: 10.3389/fncel.2020.577525] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 11/10/2020] [Indexed: 12/11/2022] Open
Abstract
The prevalence of autism spectrum disorder (ASD)-a type of neurodevelopmental disorder-is increasing and is around 2% in North America, Asia, and Europe. Besides the known genetic link, environmental, epigenetic, and metabolic factors have been implicated in ASD etiology. Although highly heterogeneous at the behavioral level, ASD comprises a set of core symptoms including impaired communication and social interaction skills as well as stereotyped and repetitive behaviors. This has led to the suggestion that a large part of the ASD phenotype is caused by changes in a few and common set of signaling pathways, the identification of which is a fundamental aim of autism research. Using advanced bioinformatics tools and the abundantly available genetic data, it is possible to classify the large number of ASD-associated genes according to cellular function and pathways. Cellular processes known to be impaired in ASD include gene regulation, synaptic transmission affecting the excitation/inhibition balance, neuronal Ca2+ signaling, development of short-/long-range connectivity (circuits and networks), and mitochondrial function. Such alterations often occur during early postnatal neurodevelopment. Among the neurons most affected in ASD as well as in schizophrenia are those expressing the Ca2+-binding protein parvalbumin (PV). These mainly inhibitory interneurons present in many different brain regions in humans and rodents are characterized by rapid, non-adaptive firing and have a high energy requirement. PV expression is often reduced at both messenger RNA (mRNA) and protein levels in human ASD brain samples and mouse ASD (and schizophrenia) models. Although the human PVALB gene is not a high-ranking susceptibility/risk gene for either disorder and is currently only listed in the SFARI Gene Archive, we propose and present supporting evidence for the Parvalbumin Hypothesis, which posits that decreased PV level is causally related to the etiology of ASD (and possibly schizophrenia).
Collapse
Affiliation(s)
| | | | | | | | - Beat Schwaller
- Section of Medicine, Anatomy, University of Fribourg, Fribourg, Switzerland
| |
Collapse
|
48
|
Addabbo F, Baglioni V, Schrag A, Schwarz MJ, Dietrich A, Hoekstra PJ, Martino D, Buttiglione M. Anti-dopamine D2 receptor antibodies in chronic tic disorders. Dev Med Child Neurol 2020; 62:1205-1212. [PMID: 32644201 DOI: 10.1111/dmcn.14613] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/18/2020] [Indexed: 12/11/2022]
Abstract
AIM To investigate the association between circulating anti-dopamine D2 receptor (D2R) autoantibodies and the exacerbation of tics in children with chronic tic disorders (CTDs). METHOD One hundred and thirty-seven children with CTDs (108 males, 29 females; mean age [SD] 10y 0mo [2y 7mo], range 4-16y) were recruited over 18 months. Patients were assessed at baseline, at tic exacerbation, and at 2 months after exacerbation. Serum anti-D2R antibodies were evaluated using a cell-based assay and blinded immunofluorescence microscopy scoring was performed by two raters. The association between visit type and presence of anti-D2R antibodies was measured with McNemar's test and repeated-measure logistic regression models, adjusting for potential demographic and clinical confounders. RESULTS At exacerbation, 11 (8%) participants became anti-D2R-positive ('early peri-exacerbation seroconverters'), and nine (6.6%) became anti-D2R-positive at post-exacerbation ('late peri-exacerbation seroconverters'). The anti-D2R antibodies were significantly associated with exacerbations when compared to baseline (McNemar's odds ratio=11, p=0.003) and conditional logistic regression confirmed this association (Z=3.49, p<0.001) after adjustment for demographic and clinical data and use of psychotropic drugs. INTERPRETATION There is a potential association between immune mechanisms and the severity course of tics in adolescents with CTDs.
Collapse
Affiliation(s)
- Francesco Addabbo
- Department of Biomedical Sciences and Human Oncology, University of Bari "Aldo Moro", Bari, Italy
| | - Valentina Baglioni
- Department of Human Neurosciences, Sapienza University of Rome, Institute of Child and Adolescent Neurology and Psychiatry, Rome, Italy
| | - Anette Schrag
- Department of Clinical Neurosciences, UCL Institute of Neurology, Royal Free Campus, London, UK
| | - Markus J Schwarz
- Institute of Laboratory Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Andrea Dietrich
- Department of Child and Adolescent Psychiatry, University Medical Center Groningen, Groningen, the Netherlands
| | - Pieter J Hoekstra
- Department of Child and Adolescent Psychiatry, University Medical Center Groningen, Groningen, the Netherlands
| | - Davide Martino
- Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Health Sciences Centre, Calgary, AB, Canada
| | - Maura Buttiglione
- Department of Biomedical Sciences and Human Oncology, University of Bari "Aldo Moro", Bari, Italy
| | | |
Collapse
|
49
|
Hacohen-Kleiman G, Moaraf S, Kapitansky O, Gozes I. Sex-and Region-Dependent Expression of the Autism-Linked ADNP Correlates with Social- and Speech-Related Genes in the Canary Brain. J Mol Neurosci 2020; 70:1671-1683. [PMID: 32926339 DOI: 10.1007/s12031-020-01700-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/05/2020] [Indexed: 02/07/2023]
Abstract
The activity-dependent neuroprotective protein (ADNP) syndrome is an autistic-like disorder, instigated by mutations in ADNP. This syndrome is characterized by developmental delays, impairments in speech, motor function, abnormal hearing, and intellectual disabilities. In the Adnp-haploinsufficient mouse model, many of these impediments are evident, appearing in a sex-dependent manner. In zebra finch songbird (ZF; Taeniopygia guttata), an animal model used for song/language studies, ADNP mRNA most robust expression is observed in the cerebrum of young males, potentially corroborating with male ZF exclusive singing behavior and developed cerebral song system. Herein, we report a similar sex-dependent ADNP expression profile, with the highest expression in the cerebrum (qRT-PCR) in the brain of another songbird, the domesticated canary (Serinus canaria domestica). Additional analyses for the mRNA transcripts of the ADNP regulator, vasoactive intestinal peptide (VIP), sister gene ADNP2, and speech-related Forkhead box protein P2 (FoxP2) revealed multiple sex and brain region-dependent positive correlations between the genes (including ADNP). Parallel transcript expression patterns for FoxP2 and VIP were observed alongside specific FoxP2 increase in males compared with females as well as VIP/ADNP2 correlations. In spatial view, a sexually independent extensive form of expression was found for ADNP in the canary cerebrum (RNA in situ hybridization). The songbird cerebral mesopallium area stood out as a potentially high-expressing ADNP tissue, further strengthening the association of ADNP with sense integration and auditory memory formation, previously implicated in mouse and human.
Collapse
Affiliation(s)
- Gal Hacohen-Kleiman
- The Elton Laboratory for Neuroendocrinology, Department of Human Molecular Genetics and Biochemistry, Sackler Faculty of Medicine, Sagol School of Neuroscience and Adams Super Center for Brain Studies, Tel Aviv University, 69978, Tel Aviv, Israel
- Department of Natural and Life Sciences, The Open University of Israel, 43107, Ra'anana, Israel
| | - Stan Moaraf
- Department of Natural and Life Sciences, The Open University of Israel, 43107, Ra'anana, Israel
- School of Zoology, Tel Aviv University, 6997801, Tel Aviv, Israel
| | - Oxana Kapitansky
- The Elton Laboratory for Neuroendocrinology, Department of Human Molecular Genetics and Biochemistry, Sackler Faculty of Medicine, Sagol School of Neuroscience and Adams Super Center for Brain Studies, Tel Aviv University, 69978, Tel Aviv, Israel
| | - Illana Gozes
- The Elton Laboratory for Neuroendocrinology, Department of Human Molecular Genetics and Biochemistry, Sackler Faculty of Medicine, Sagol School of Neuroscience and Adams Super Center for Brain Studies, Tel Aviv University, 69978, Tel Aviv, Israel.
| |
Collapse
|
50
|
Zhang H. Synaptic dysregulation in autism spectrum disorders. J Neurosci Res 2020; 98:2111-2114. [PMID: 32780523 DOI: 10.1002/jnr.24711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 07/21/2020] [Accepted: 07/24/2020] [Indexed: 11/07/2022]
Affiliation(s)
- Huaye Zhang
- Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School, Piscataway, NJ, USA
| |
Collapse
|