1
|
Yang J, Ma G, Du X, Xie J, Wang M, Wang W, Guo B, Wu S. Deciphering the Role of Shank3 in Dendritic Morphology and Synaptic Function Across Postnatal Developmental Stages in the Shank3B KO Mouse. Neurosci Bull 2025; 41:583-599. [PMID: 39693031 PMCID: PMC11978597 DOI: 10.1007/s12264-024-01330-y] [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: 05/10/2024] [Accepted: 09/14/2024] [Indexed: 12/19/2024] Open
Abstract
Autism Spectrum Disorder (ASD) is marked by early-onset neurodevelopmental anomalies, yet the temporal dynamics of genetic contributions to these processes remain insufficiently understood. This study aimed to elucidate the role of the Shank3 gene, known to be associated with monogenic causes of autism, in early developmental processes to inform the timing and mechanisms for potential interventions for ASD. Utilizing the Shank3B knockout (KO) mouse model, we examined Shank3 expression and its impact on neuronal maturation through Golgi staining for dendritic morphology and electrophysiological recordings to measure synaptic function in the anterior cingulate cortex (ACC) across different postnatal stages. Our longitudinal analysis revealed that, while Shank3B KO mice displayed normal neuronal morphology at one week postnatal, significant impairments in dendritic growth and synaptic activity emerged by two to three weeks. These findings highlight the critical developmental window during which Shank3 is essential for neuronal and synaptic maturation in the ACC.
Collapse
Affiliation(s)
- Jing Yang
- Department of Basic Medicine, School of Medicine, Hangzhou City University, Hangzhou, 310015, China
| | - Guaiguai Ma
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Xiaohui Du
- Department of Basic Medicine, School of Medicine, Hangzhou City University, Hangzhou, 310015, China
| | - Jinyi Xie
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Mengmeng Wang
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Wenting Wang
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Baolin Guo
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China.
- Innovation Research Institute, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China.
| | - Shengxi Wu
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China.
| |
Collapse
|
2
|
Kshetri R, Beavers JO, Hyde R, Ewa R, Schwertman A, Porcayo S, Richardson BD. Behavioral decline in Shank3 Δex4-22 mice during early adulthood parallels cerebellar granule cell glutamatergic synaptic changes. Mol Autism 2024; 15:52. [PMID: 39633421 PMCID: PMC11616285 DOI: 10.1186/s13229-024-00628-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: 08/09/2024] [Accepted: 10/23/2024] [Indexed: 12/07/2024] Open
Abstract
BACKGROUND SHANK3, a gene encoding a synaptic scaffolding protein, is implicated in autism spectrum disorder (ASD) and is disrupted in Phelan-McDermid syndrome (PMS). Despite evidence of regression or worsening of ASD-like symptoms in individuals with PMS, the underlying mechanisms remain unclear. Although Shank3 is highly expressed in the cerebellar cortical granule cells, its role in cerebellar function and contribution to behavioral deficits in ASD models are unknown. This study investigates behavioral changes and cerebellar synaptic alterations in Shank3Δex4-22 mice at two developmental stages. METHODS Shank3Δex4-22 wildtype, heterozygous, and homozygous knockout mice lacking exons 4-22 (all functional isoforms) were subjected to a behavioral battery in both juvenile (5-7 weeks old) and adult (3-5 months old) mouse cohorts of both sexes. Immunostaining was used to show the expression of Shank3 in the cerebellar cortex. Spontaneous excitatory postsynaptic currents (sEPSCs) from cerebellar granule cells (CGCs) were recorded by whole-cell patch-clamp electrophysiology. RESULTS Deletion of Shank3 caused deficits in motor function, heightened anxiety, and repetitive behaviors. These genotype-dependent behavioral alterations were more prominent in adult mice than in juveniles. Reduced social preference was only identified in adult Shank3Δex4-22 knockout male mice, while self-grooming was uniquely elevated in males across both age groups. Heterozygous mice showed little to no changes in behavioral phenotypes in most behavioral tests. Immunofluorescence staining indicated the presence of Shank3 predominantly in the dendrite-containing rosette-like structures in CGCs, colocalizing with presynaptic markers of glutamatergic mossy fiber. Electrophysiological findings identified a parallel relationship between the age-related exacerbation of behavioral impairments and the enhancement of sEPSC amplitude in CGCs. LIMITATIONS Other behavioral tests of muscle strength (grip strength test), memory (Barnes/water maze), and communication (ultrasonic vocalization), were not performed. Further study is necessary to elucidate how Shank3 modulates synaptic function at the mossy fiber-granule cell synapse in the cerebellum and whether these changes shape the behavioral phenotype. CONCLUSIONS Our findings reveal an age-related exacerbation of behavioral impairments in Shank3Δex4-22 mutant mice. These results suggest that Shank3 may alter the function of glutamatergic receptors at the mossy fiber-cerebellar granule cell synapse as a potential mechanism causing cerebellar disruption in ASD.
Collapse
Affiliation(s)
- Rajaram Kshetri
- Department of Pharmacology, Southern Illinois University - School of Medicine, Springfield, IL, 62702, USA
| | - James O Beavers
- Department of Pharmacology, Southern Illinois University - School of Medicine, Springfield, IL, 62702, USA
| | - Romana Hyde
- Department of Biological Sciences, University of Idaho, Moscow, ID, 83844, USA
| | - Roseline Ewa
- Department of Pharmacology, Southern Illinois University - School of Medicine, Springfield, IL, 62702, USA
| | - Amber Schwertman
- Department of Pharmacology, Southern Illinois University - School of Medicine, Springfield, IL, 62702, USA
| | - Sarahi Porcayo
- Department of Pharmacology, Southern Illinois University - School of Medicine, Springfield, IL, 62702, USA
| | - Ben D Richardson
- Department of Pharmacology, Southern Illinois University - School of Medicine, Springfield, IL, 62702, USA.
| |
Collapse
|
3
|
An S, Zhen Z, Wang S, Sang M, Zhang S. Intestinal Microbiota Is a Key Target for Load Swimming to Improve Anxiety Behavior and Muscle Strength in Shank 3 -/- Rats. Mol Neurobiol 2024; 61:9961-9976. [PMID: 37966684 DOI: 10.1007/s12035-023-03670-8] [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: 05/12/2023] [Accepted: 09/20/2023] [Indexed: 11/16/2023]
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder characterized by social disorder and stereotypical behavior, and its incidence rate is increasing yearly. It is considered that acritical period for the prognosis of young children with ASD exists, thus early treatment is crucial. Swimming, due to its comforting effect, is often used to induce enthusiasm in young children for completing activities and has a good effect in the treatment of ASD, but the effective path of swimming has yet to be reported. The intestinal microbiota of ASD patients and animal models has been reported to be different from that of healthy controls, and these changes may affect the brain environment. Therefore, whether the intestinal microbiota is involved in the treatment of ASD by early swimming is our concern. In this study, we used 8-day old Shank3 gene knockout rats with 8 weeks of early load swimming training and conducted behavioral, small intestine morphology, and intestinal content sequencing after training. The results showed that early load swimming significantly reduced the stereotyped and anxious behaviors of Shank3-/- rats, increased their muscle strength, increased the length of intestinal villi and the width of the muscular layer after Shank3 knockout, and affected the abundance of intestinal microorganisms. The abundances with statistical significance were Lactobacillus, Lachnospiraceae, and Alloprevotella. To further confirm the role of intestinal microorganisms in it, we designed a 14-day intestinal stool transplantation experiment. Fecal microbiota transplantation demonstrated that load swimming can significantly reduce the anxiety behavior of Shank3 rats, increase their muscle strength, change the structure of the small intestine, and affect the abundance of intestinal contents. The abundance of Epsilonbateraeota, Prevotella, and Bacteroides significantly changed after transplantation. Our findings confirm the possibility of early load swimming therapy for individuals with ASD and explain that the intestinal microbiota is a key pathway for early exercise therapy for patients with ASD.
Collapse
Affiliation(s)
- Shasha An
- College of Physical Education and Sports, Beijing Normal University, Beijing, 100875, China
| | - Zhiping Zhen
- College of Physical Education and Sports, Beijing Normal University, Beijing, 100875, China.
| | - Shijiao Wang
- College of Physical Education and Sports, Beijing Normal University, Beijing, 100875, China
| | - Mingze Sang
- College of Physical Education and Sports, Beijing Normal University, Beijing, 100875, China
| | - Shuai Zhang
- College of Physical Education and Sports, Beijing Normal University, Beijing, 100875, China
| |
Collapse
|
4
|
Cao W, Li H, Luo J. Prefrontal cortical circuits in social behaviors: an overview. J Zhejiang Univ Sci B 2024; 25:941-955. [PMID: 39626878 PMCID: PMC11634449 DOI: 10.1631/jzus.b2300743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 01/02/2024] [Indexed: 12/13/2024]
Abstract
Social behaviors are fundamental and intricate functions in both humans and animals, governed by the interplay of social cognition and emotions. A noteworthy feature of several neuropsychiatric disorders, including autism spectrum disorder (ASD) and schizophrenia (SCZ), is a pronounced deficit in social functioning. Despite a burgeoning body of research on social behaviors, the precise neural circuit mechanisms underpinning these phenomena remain to be elucidated. In this paper, we review the pivotal role of the prefrontal cortex (PFC) in modulating social behaviors, as well as its functional alteration in social disorders in ASD or SCZ. We posit that PFC dysfunction may represent a critical hub in the pathogenesis of psychiatric disorders characterized by shared social deficits. Furthermore, we delve into the intricate connectivity of the medial PFC (mPFC) with other cortical areas and subcortical brain regions in rodents, which exerts a profound influence on social behaviors. Notably, a substantial body of evidence underscores the role of N-methyl-D-aspartate receptors (NMDARs) and the proper functioning of parvalbumin-positive interneurons within the mPFC for social regulation. Our overarching goal is to furnish a comprehensive understanding of these intricate circuits and thereby contribute to the enhancement of both research endeavors and clinical practices concerning social behavior deficits.
Collapse
Affiliation(s)
- Wei Cao
- Institute of Brain Science and Department of Physiology, School of Basic Medical Sciences, Hangzhou Normal University, Hangzhou 311121, China
- Zhejiang Key Laboratory of Medical Epigenetics, School of Basic Medical Sciences, Hangzhou Normal University, Hangzhou 311121, China
- Zhejiang Philosophy and Social Science Laboratory for Research in Early Development and Childcare, Hangzhou Normal University, Hangzhou 311121, China
| | - Huiyi Li
- Institute of Brain Science and Department of Physiology, School of Basic Medical Sciences, Hangzhou Normal University, Hangzhou 311121, China
- Zhejiang Key Laboratory of Medical Epigenetics, School of Basic Medical Sciences, Hangzhou Normal University, Hangzhou 311121, China
| | - Jianhong Luo
- Affiliated Mental Health Center & Hangzhou Seventh People's Hospital, School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou 310013, China.
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou 310015, China.
- NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|
5
|
Lu X, Ni P, Suarez-Meade P, Ma Y, Forrest EN, Wang G, Wang Y, Quiñones-Hinojosa A, Gerstein M, Jiang YH. Transcriptional determinism and stochasticity contribute to the complexity of autism-associated SHANK family genes. Cell Rep 2024; 43:114376. [PMID: 38900637 PMCID: PMC11328446 DOI: 10.1016/j.celrep.2024.114376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 05/08/2024] [Accepted: 05/31/2024] [Indexed: 06/22/2024] Open
Abstract
Precision of transcription is critical because transcriptional dysregulation is disease causing. Traditional methods of transcriptional profiling are inadequate to elucidate the full spectrum of the transcriptome, particularly for longer and less abundant mRNAs. SHANK3 is one of the most common autism causative genes. Twenty-four Shank3-mutant animal lines have been developed for autism modeling. However, their preclinical validity has been questioned due to incomplete Shank3 transcript structure. We apply an integrative approach combining cDNA-capture and long-read sequencing to profile the SHANK3 transcriptome in humans and mice. We unexpectedly discover an extremely complex SHANK3 transcriptome. Specific SHANK3 transcripts are altered in Shank3-mutant mice and postmortem brain tissues from individuals with autism spectrum disorder. The enhanced SHANK3 transcriptome significantly improves the detection rate for potential deleterious variants from genomics studies of neuropsychiatric disorders. Our findings suggest that both deterministic and stochastic transcription of the genome is associated with SHANK family genes.
Collapse
Affiliation(s)
- Xiaona Lu
- Department of Genetics, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Pengyu Ni
- Program in Computational Biology and Bioinformatics, Yale University, New Haven, CT 06520, USA; Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06520, USA
| | | | - Yu Ma
- Department of Neurology, Children's Hospital of Fudan University, Shanghai 201102, China
| | - Emily Niemitz Forrest
- Department of Genetics, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Guilin Wang
- Keck Microarray Shared Resource, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Yi Wang
- Department of Neurology, Children's Hospital of Fudan University, Shanghai 201102, China
| | | | - Mark Gerstein
- Program in Computational Biology and Bioinformatics, Yale University, New Haven, CT 06520, USA; Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06520, USA; Department of Computer Science, Yale University, New Haven, CT 06520, USA; Department of Statistics and Data Science, Yale University, New Haven, CT 06520, USA; Department of Biomedical Informatics & Data Science, Yale University, New Haven, CT 06520, USA
| | - Yong-Hui Jiang
- Department of Genetics, Yale University School of Medicine, New Haven, CT 06520, USA; Neuroscience, Yale University School of Medicine, New Haven, CT 06520, USA; Pediatrics, Yale University School of Medicine, New Haven, CT 06520, USA.
| |
Collapse
|
6
|
Chung M, Imanaka K, Huang Z, Watarai A, Wang MY, Tao K, Ejima H, Aida T, Feng G, Okuyama T. Conditional knockout of Shank3 in the ventral CA1 by quantitative in vivo genome-editing impairs social memory in mice. Nat Commun 2024; 15:4531. [PMID: 38866749 PMCID: PMC11169449 DOI: 10.1038/s41467-024-48430-x] [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: 12/30/2022] [Accepted: 04/26/2024] [Indexed: 06/14/2024] Open
Abstract
Individuals with autism spectrum disorder (ASD) have a higher prevalence of social memory impairment. A series of our previous studies revealed that hippocampal ventral CA1 (vCA1) neurons possess social memory engram and that the neurophysiological representation of social memory in the vCA1 neurons is disrupted in ASD-associated Shank3 knockout mice. However, whether the dysfunction of Shank3 in vCA1 causes the social memory impairment observed in ASD remains unclear. In this study, we found that vCA1-specific Shank3 conditional knockout (cKO) by the adeno-associated virus (AAV)- or specialized extracellular vesicle (EV)- mediated in vivo gene editing was sufficient to recapitulate the social memory impairment in male mice. Furthermore, the utilization of EV-mediated Shank3-cKO allowed us to quantitatively examine the role of Shank3 in social memory. Our results suggested that there is a certain threshold for the proportion of Shank3-cKO neurons required for social memory disruption. Thus, our study provides insight into the population coding of social memory in vCA1, as well as the pathological mechanisms underlying social memory impairment in ASD.
Collapse
Affiliation(s)
- Myung Chung
- Laboratory of Behavioral Neuroscience, Institute for Quantitative Biosciences, The University of Tokyo, Tokyo, Japan
- Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Katsutoshi Imanaka
- Laboratory of Behavioral Neuroscience, Institute for Quantitative Biosciences, The University of Tokyo, Tokyo, Japan
- Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Ziyan Huang
- Laboratory of Behavioral Neuroscience, Institute for Quantitative Biosciences, The University of Tokyo, Tokyo, Japan
- Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Akiyuki Watarai
- Laboratory of Behavioral Neuroscience, Institute for Quantitative Biosciences, The University of Tokyo, Tokyo, Japan
| | - Mu-Yun Wang
- Laboratory of Behavioral Neuroscience, Institute for Quantitative Biosciences, The University of Tokyo, Tokyo, Japan
| | - Kentaro Tao
- Laboratory of Behavioral Neuroscience, Institute for Quantitative Biosciences, The University of Tokyo, Tokyo, Japan
| | - Hirotaka Ejima
- Department of Materials Engineering, School of Engineering, The University of Tokyo, Tokyo, Japan
| | - Tomomi Aida
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Guoping Feng
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Teruhiro Okuyama
- Laboratory of Behavioral Neuroscience, Institute for Quantitative Biosciences, The University of Tokyo, Tokyo, Japan.
- Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.
| |
Collapse
|
7
|
Lu X, Ni P, Suarez-Meade P, Ma Y, Forrest EN, Wang G, Wang Y, Quiñones-Hinojosa A, Gerstein M, Jiang YH. Transcriptional Determinism and Stochasticity Contribute to the Complexity of Autism Associated SHANK Family Genes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.18.585480. [PMID: 38562714 PMCID: PMC10983920 DOI: 10.1101/2024.03.18.585480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Precision of transcription is critical because transcriptional dysregulation is disease causing. Traditional methods of transcriptional profiling are inadequate to elucidate the full spectrum of the transcriptome, particularly for longer and less abundant mRNAs. SHANK3 is one of the most common autism causative genes. Twenty-four Shank3 mutant animal lines have been developed for autism modeling. However, their preclinical validity has been questioned due to incomplete Shank3 transcript structure. We applied an integrative approach combining cDNA-capture and long-read sequencing to profile the SHANK3 transcriptome in human and mice. We unexpectedly discovered an extremely complex SHANK3 transcriptome. Specific SHANK3 transcripts were altered in Shank3 mutant mice and postmortem brains tissues from individuals with ASD. The enhanced SHANK3 transcriptome significantly improved the detection rate for potential deleterious variants from genomics studies of neuropsychiatric disorders. Our findings suggest the stochastic transcription of genome associated with SHANK family genes.
Collapse
Affiliation(s)
- Xiaona Lu
- Department of Genetics, Yale University School of Medicine New Haven, CT, 06520 USA
| | - Pengyu Ni
- Biomedical Informatics & Data Science, Yale University School of Medicine New Haven, CT, 06520 USA
| | | | - Yu Ma
- Department of Neurology, Children’s Hospital of Fudan University, Shanghai, 201102 China
| | | | - Guilin Wang
- Yale Center for Genome Analysis, Yale University School of Medicine New Haven, CT, 06520 USA
| | - Yi Wang
- Department of Neurology, Children’s Hospital of Fudan University, Shanghai, 201102 China
| | | | - Mark Gerstein
- Biomedical Informatics & Data Science, Yale University School of Medicine New Haven, CT, 06520 USA
- Yale Center for Genome Analysis, Yale University School of Medicine New Haven, CT, 06520 USA
| | - Yong-hui Jiang
- Department of Genetics, Yale University School of Medicine New Haven, CT, 06520 USA
- Neuroscienc, Yale University School of Medicine New Haven, CT, 06520 USA
- Pediatrics, Yale University School of Medicine New Haven, CT, 06520 USA
| |
Collapse
|
8
|
Jeong JH, Lee H, Kim D, Park E, Woo J, Cho Y, Keum G, Lee A, Kang T, Kim J, Choo H, Lee S, Jeon B. Identification of an Antagonist Targeting G Protein and β-Arrestin Signaling Pathways of 5-HT 7R. ACS Chem Neurosci 2024; 15:1026-1041. [PMID: 38387042 DOI: 10.1021/acschemneuro.3c00738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/24/2024] Open
Abstract
In consideration of the limited number of FDA-approved drugs for autism spectrum disorder (ASD), significant efforts have been devoted to identifying novel drug candidates. Among these, 5-HT7R modulators have garnered considerable attention due to their potential in alleviating autism-like behaviors in ASD animal models. In this study, we designed and synthesized biphenyl-3-ylmethylpyrrolidines 3 and biphenyl-3-yl-dihydroimidazoles 4 as 5-HT7R modulators. Through extensive biological tests of 3 and 4 in G protein and β-arrestin signaling pathways of 5-HT7R, it was determined that 2-(2'-methoxy-[1,1'-biphenyl]-3-yl)-4,5-dihydro-1H-imidazole 4h acted as a 5-HT7R antagonist in both signaling pathways. In in vivo study with Shank3-/- transgenic (TG) mice, the self-grooming behavior test was performed with 4h, resulting in a significant reduction in the duration of self-grooming. In addition, an immunohistochemical experiment with 4h restored reduced neurogenesis in Shank3-/- TG mice, which is confirmed by the quantification of doublecortin (DCX) positive neurons, suggesting the promising therapeutic potential of 4h.
Collapse
Affiliation(s)
- Jeong Hyun Jeong
- Brain Science Institute, Korea Institute of Science and Technology, Seongbuk-gu, Seoul 02792, Republic of Korea
| | - Haeun Lee
- Brain Science Institute, Korea Institute of Science and Technology, Seongbuk-gu, Seoul 02792, Republic of Korea
- Division of Bio-Medical Science and Technology, KIST School, University of Science and Technology, Seongbuk-gu, Seoul 02792, Republic of Korea
| | - Doyoung Kim
- Brain Science Institute, Korea Institute of Science and Technology, Seongbuk-gu, Seoul 02792, Republic of Korea
| | - Eunseo Park
- Brain Science Institute, Korea Institute of Science and Technology, Seongbuk-gu, Seoul 02792, Republic of Korea
- Division of Bio-Medical Science and Technology, KIST School, University of Science and Technology, Seongbuk-gu, Seoul 02792, Republic of Korea
| | - Jiwan Woo
- Research Animal Resource Center, Korea Institute of Science and Technology, Seongbuk-gu, Seoul 02792, Republic of Korea
| | - Yakdol Cho
- Research Animal Resource Center, Korea Institute of Science and Technology, Seongbuk-gu, Seoul 02792, Republic of Korea
| | - Gyochang Keum
- Brain Science Institute, Korea Institute of Science and Technology, Seongbuk-gu, Seoul 02792, Republic of Korea
- Division of Bio-Medical Science and Technology, KIST School, University of Science and Technology, Seongbuk-gu, Seoul 02792, Republic of Korea
| | - Ansoo Lee
- Brain Science Institute, Korea Institute of Science and Technology, Seongbuk-gu, Seoul 02792, Republic of Korea
- Division of Bio-Medical Science and Technology, KIST School, University of Science and Technology, Seongbuk-gu, Seoul 02792, Republic of Korea
| | - Taek Kang
- Brain Science Institute, Korea Institute of Science and Technology, Seongbuk-gu, Seoul 02792, Republic of Korea
| | - Jeongjin Kim
- Brain Science Institute, Korea Institute of Science and Technology, Seongbuk-gu, Seoul 02792, Republic of Korea
- Division of Bio-Medical Science and Technology, KIST School, University of Science and Technology, Seongbuk-gu, Seoul 02792, Republic of Korea
| | - Hyunah Choo
- Brain Science Institute, Korea Institute of Science and Technology, Seongbuk-gu, Seoul 02792, Republic of Korea
- Division of Bio-Medical Science and Technology, KIST School, University of Science and Technology, Seongbuk-gu, Seoul 02792, Republic of Korea
| | - Sanghee Lee
- Brain Science Institute, Korea Institute of Science and Technology, Seongbuk-gu, Seoul 02792, Republic of Korea
| | - Byungsun Jeon
- Brain Science Institute, Korea Institute of Science and Technology, Seongbuk-gu, Seoul 02792, Republic of Korea
- Division of Bio-Medical Science and Technology, KIST School, University of Science and Technology, Seongbuk-gu, Seoul 02792, Republic of Korea
| |
Collapse
|
9
|
Qin Y, Zhang XY, Liu Y, Ma Z, Tao S, Li Y, Peng R, Wang F, Wang J, Feng J, Qiu Z, Jin L, Wang H, Gong X. Downregulation of mGluR1-mediated signaling underlying autistic-like core symptoms in Shank1 P1812L-knock-in mice. Transl Psychiatry 2023; 13:329. [PMID: 37880287 PMCID: PMC10600164 DOI: 10.1038/s41398-023-02626-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 09/16/2023] [Accepted: 10/06/2023] [Indexed: 10/27/2023] Open
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder characterized by core symptoms that consist of social deficits and repetitive behaviors. Unfortunately, no effective medication is available thus far to target the core symptoms of ASD, since the pathogenesis remains largely unknown. To investigate the pathogenesis of the core symptoms in ASD, we constructed Shank1 P1812L-knock-in (KI) mice corresponding to a recurrent ASD-related mutation, SHANK1 P1806L, to achieve construct validity and face validity. Shank1 P1812L-KI heterozygous (HET) mice presented with social deficits and repetitive behaviors without the presence of confounding comorbidities. HET mice also exhibited downregulation of metabotropic glutamate receptor (mGluR1) and associated signals, along with structural abnormalities in the dendritic spines and postsynaptic densities. Combined with findings from Shank1 R882H-KI mice, our study confirms that mGluR1-mediated signaling dysfunction is a pivotal mechanism underlying the core symptoms of ASD. Interestingly, Shank1 P1812L-KI homozygous (HOM) mice manifested behavioral signs of impaired long-term memory rather than autistic-like core traits; thus, their phenotype was markedly different from that of Shank1 P1812L-KI HET mice. Correspondingly, at the molecular level, Shank1 P1812L-KI HOM displayed upregulation of AMPA receptor (GluA2)-related signals. The different patterns of protein changes in HOM and HET mice may explain the differences in behaviors. Our study emphasizes the universality of mGluR1-signaling hypofunction in the pathogenesis of the core symptoms in ASD, providing a potential target for therapeutic drugs. The precise correspondence between genotype and phenotype, as shown in HOM and HET mice, indicates the importance of reproducing disease-related genotypes in mouse models.
Collapse
Affiliation(s)
- Yue Qin
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| | - Xiao-Yong Zhang
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China
- Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Ministry of Education, Fudan University, Shanghai, China
- MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Yanyan Liu
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
- Institute of Integrated Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Hefei, China
| | - Zehan Ma
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| | - Shuo Tao
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| | - Ying Li
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| | - Rui Peng
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| | - Fei Wang
- Early Intervention Unit, Department of Psychiatry, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing, China
- Functional Brain Imaging Institute of Nanjing Medical University, Nanjing, China
| | - Jiucun Wang
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| | - Jianfeng Feng
- MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Zilong Qiu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Chinese Academy of Sciences, Shanghai, China
| | - Li Jin
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| | - Hongyan Wang
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China.
| | - Xiaohong Gong
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
10
|
Molloy CJ, Cooke J, Gatford NJF, Rivera-Olvera A, Avazzadeh S, Homberg JR, Grandjean J, Fernandes C, Shen S, Loth E, Srivastava DP, Gallagher L. Bridging the translational gap: what can synaptopathies tell us about autism? Front Mol Neurosci 2023; 16:1191323. [PMID: 37441676 PMCID: PMC10333541 DOI: 10.3389/fnmol.2023.1191323] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 05/24/2023] [Indexed: 07/15/2023] Open
Abstract
Multiple molecular pathways and cellular processes have been implicated in the neurobiology of autism and other neurodevelopmental conditions. There is a current focus on synaptic gene conditions, or synaptopathies, which refer to clinical conditions associated with rare genetic variants disrupting genes involved in synaptic biology. Synaptopathies are commonly associated with autism and developmental delay and may be associated with a range of other neuropsychiatric outcomes. Altered synaptic biology is suggested by both preclinical and clinical studies in autism based on evidence of differences in early brain structural development and altered glutamatergic and GABAergic neurotransmission potentially perturbing excitatory and inhibitory balance. This review focusses on the NRXN-NLGN-SHANK pathway, which is implicated in the synaptic assembly, trans-synaptic signalling, and synaptic functioning. We provide an overview of the insights from preclinical molecular studies of the pathway. Concentrating on NRXN1 deletion and SHANK3 mutations, we discuss emerging understanding of cellular processes and electrophysiology from induced pluripotent stem cells (iPSC) models derived from individuals with synaptopathies, neuroimaging and behavioural findings in animal models of Nrxn1 and Shank3 synaptic gene conditions, and key findings regarding autism features, brain and behavioural phenotypes from human clinical studies of synaptopathies. The identification of molecular-based biomarkers from preclinical models aims to advance the development of targeted therapeutic treatments. However, it remains challenging to translate preclinical animal models and iPSC studies to interpret human brain development and autism features. We discuss the existing challenges in preclinical and clinical synaptopathy research, and potential solutions to align methodologies across preclinical and clinical research. Bridging the translational gap between preclinical and clinical studies will be necessary to understand biological mechanisms, to identify targeted therapies, and ultimately to progress towards personalised approaches for complex neurodevelopmental conditions such as autism.
Collapse
Affiliation(s)
- Ciara J. Molloy
- Department of Psychiatry, School of Medicine, Trinity College Dublin, Dublin, Ireland
| | - Jennifer Cooke
- Forensic and Neurodevelopmental Sciences, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kingdom
| | - Nicholas J. F. Gatford
- Kavli Institute for Nanoscience Discovery, Nuffield Department of Clinical Neurosciences, University of Oxford, Medical Sciences Division, Oxford, United Kingdom
| | - Alejandro Rivera-Olvera
- Donders Institute for Brain, Cognition, and Behaviour, Radboud University Medical Centre, Nijmegen, Netherlands
| | - Sahar Avazzadeh
- Physiology and Cellular Physiology Research Laboratory, CÚRAM SFI Centre for Research in Medical Devices, School of Medicine, Human Biology Building, University of Galway, Galway, Ireland
| | - Judith R. Homberg
- Donders Institute for Brain, Cognition, and Behaviour, Radboud University Medical Centre, Nijmegen, Netherlands
| | - Joanes Grandjean
- Physiology and Cellular Physiology Research Laboratory, CÚRAM SFI Centre for Research in Medical Devices, School of Medicine, Human Biology Building, University of Galway, Galway, Ireland
- Department of Medical Imaging, Radboud University Medical Centre, Nijmegen, Netherlands
| | - Cathy Fernandes
- Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kingdom
- MRC Centre for Neurodevelopmental Disorders, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kingdom
| | - Sanbing Shen
- Regenerative Medicine Institute, School of Medicine, University of Galway, Galway, Ireland
- FutureNeuro, The SFI Research Centre for Chronic and Rare Neurological Diseases, Royal College of Surgeons, Dublin, Ireland
| | - Eva Loth
- Forensic and Neurodevelopmental Sciences, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kingdom
| | - Deepak P. Srivastava
- MRC Centre for Neurodevelopmental Disorders, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kingdom
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kingdom
| | - Louise Gallagher
- Department of Psychiatry, School of Medicine, Trinity College Dublin, Dublin, Ireland
- The Hospital for SickKids, Toronto, ON, Canada
- The Peter Gilgan Centre for Research and Learning, SickKids Research Institute, Toronto, ON, Canada
- The Centre for Addiction and Mental Health, Toronto, ON, Canada
- Department of Psychiatry, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
11
|
Liu XY, Wang K, Deng XH, Wei YH, Guo R, Liu SF, Zhu YF, Zhong JJ, Zheng JY, Wang MD, Ye QH, He JQ, Guo KH, Zhu JR, Huang SQ, Chen ZX, Lv CS, Wen L. Amelioration of olfactory dysfunction in a mouse model of Parkinson's disease via enhancing GABAergic signaling. Cell Biosci 2023; 13:101. [PMID: 37270503 DOI: 10.1186/s13578-023-01049-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 05/06/2023] [Indexed: 06/05/2023] Open
Abstract
BACKGROUND Olfactory dysfunction is among the earliest non-motor symptoms of Parkinson's disease (PD). As the foremost pathological hallmark, α-synuclein initiates the pathology in the olfactory pathway at the early stage of PD, particularly in the olfactory epithelium (OE) and olfactory bulb (OB). However, the local neural microcircuit mechanisms underlying olfactory dysfunction between OE and OB in early PD remain unknown. RESULTS We observed that odor detection and discrimination were impaired in 6-month-old SNCA-A53T mice, while their motor ability remained unaffected. It was confirmed that α-synuclein increased and accumulated in OB but not in OE. Notably, the hyperactivity of mitral/tufted cells and the excitation/inhibition imbalance in OB were found in 6-month-old SNCA-A53T mice, which was attributed to the impaired GABAergic transmission and aberrant expression of GABA transporter 1 and vesicular GABA transporter in OB. We further showed that tiagabine, a potent and selective GABA reuptake inhibitor, could reverse the impaired olfactory function and GABAergic signaling in OB of SNCA-A53T mice. CONCLUSIONS Taken together, our findings demonstrate potential synaptic mechanisms of local neural microcircuit underlying olfactory dysfunction at the early stage of PD. These results highlight the critical role of aberrant GABAergic signaling of OB in early diagnosis and provide a potential therapeutic strategy for early-stage PD.
Collapse
Affiliation(s)
- Xing-Yang Liu
- State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, Xiang'an Hospital, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
- Xiamen Key Laboratory for TCM Dampness Disease, Neurology & Immunology Research, Department of Traditional Chinese Medicine, Longyan Hospital of Traditional Chinese Medicine, School of Medicine, Xiamen University, Longyan, Fujian, 364000, China
| | - Ke Wang
- State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, Xiang'an Hospital, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
- Xiamen Key Laboratory for TCM Dampness Disease, Neurology & Immunology Research, Department of Traditional Chinese Medicine, Longyan Hospital of Traditional Chinese Medicine, School of Medicine, Xiamen University, Longyan, Fujian, 364000, China
| | - Xian-Hua Deng
- State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, Xiang'an Hospital, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
- Xiamen Key Laboratory for TCM Dampness Disease, Neurology & Immunology Research, Department of Traditional Chinese Medicine, Longyan Hospital of Traditional Chinese Medicine, School of Medicine, Xiamen University, Longyan, Fujian, 364000, China
| | - Yi-Hua Wei
- State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, Xiang'an Hospital, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
- Xiamen Key Laboratory for TCM Dampness Disease, Neurology & Immunology Research, Department of Traditional Chinese Medicine, Longyan Hospital of Traditional Chinese Medicine, School of Medicine, Xiamen University, Longyan, Fujian, 364000, China
| | - Rui Guo
- State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, Xiang'an Hospital, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
- Xiamen Key Laboratory for TCM Dampness Disease, Neurology & Immunology Research, Department of Traditional Chinese Medicine, Longyan Hospital of Traditional Chinese Medicine, School of Medicine, Xiamen University, Longyan, Fujian, 364000, China
| | - Sui-Feng Liu
- Zhongshan Hospital, School of Medicine, Xiamen University, Xiamen, Fujian, 361100, China
| | - Yi-Fan Zhu
- State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, Xiang'an Hospital, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
- Xiamen Key Laboratory for TCM Dampness Disease, Neurology & Immunology Research, Department of Traditional Chinese Medicine, Longyan Hospital of Traditional Chinese Medicine, School of Medicine, Xiamen University, Longyan, Fujian, 364000, China
| | - Jia-Jun Zhong
- State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, Xiang'an Hospital, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
- Xiamen Key Laboratory for TCM Dampness Disease, Neurology & Immunology Research, Department of Traditional Chinese Medicine, Longyan Hospital of Traditional Chinese Medicine, School of Medicine, Xiamen University, Longyan, Fujian, 364000, China
| | - Jing-Yuan Zheng
- State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, Xiang'an Hospital, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
- Xiamen Key Laboratory for TCM Dampness Disease, Neurology & Immunology Research, Department of Traditional Chinese Medicine, Longyan Hospital of Traditional Chinese Medicine, School of Medicine, Xiamen University, Longyan, Fujian, 364000, China
| | - Meng-Dan Wang
- State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, Xiang'an Hospital, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
- Xiamen Key Laboratory for TCM Dampness Disease, Neurology & Immunology Research, Department of Traditional Chinese Medicine, Longyan Hospital of Traditional Chinese Medicine, School of Medicine, Xiamen University, Longyan, Fujian, 364000, China
| | - Qiu-Hong Ye
- State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, Xiang'an Hospital, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
- Xiamen Key Laboratory for TCM Dampness Disease, Neurology & Immunology Research, Department of Traditional Chinese Medicine, Longyan Hospital of Traditional Chinese Medicine, School of Medicine, Xiamen University, Longyan, Fujian, 364000, China
| | - Jian-Quan He
- Zhongshan Hospital, School of Medicine, Xiamen University, Xiamen, Fujian, 361100, China
| | - Kai-Hang Guo
- State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, Xiang'an Hospital, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
- Xiamen Key Laboratory for TCM Dampness Disease, Neurology & Immunology Research, Department of Traditional Chinese Medicine, Longyan Hospital of Traditional Chinese Medicine, School of Medicine, Xiamen University, Longyan, Fujian, 364000, China
| | - Jun-Rong Zhu
- State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, Xiang'an Hospital, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
- Xiamen Key Laboratory for TCM Dampness Disease, Neurology & Immunology Research, Department of Traditional Chinese Medicine, Longyan Hospital of Traditional Chinese Medicine, School of Medicine, Xiamen University, Longyan, Fujian, 364000, China
| | - Shu-Qiong Huang
- State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, Xiang'an Hospital, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
- Xiamen Key Laboratory for TCM Dampness Disease, Neurology & Immunology Research, Department of Traditional Chinese Medicine, Longyan Hospital of Traditional Chinese Medicine, School of Medicine, Xiamen University, Longyan, Fujian, 364000, China
| | - Ze-Xu Chen
- State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, Xiang'an Hospital, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
- Xiamen Key Laboratory for TCM Dampness Disease, Neurology & Immunology Research, Department of Traditional Chinese Medicine, Longyan Hospital of Traditional Chinese Medicine, School of Medicine, Xiamen University, Longyan, Fujian, 364000, China
| | - Chong-Shan Lv
- State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, Xiang'an Hospital, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
- Xiamen Key Laboratory for TCM Dampness Disease, Neurology & Immunology Research, Department of Traditional Chinese Medicine, Longyan Hospital of Traditional Chinese Medicine, School of Medicine, Xiamen University, Longyan, Fujian, 364000, China
| | - Lei Wen
- State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, Xiang'an Hospital, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China.
- Xiamen Key Laboratory for TCM Dampness Disease, Neurology & Immunology Research, Department of Traditional Chinese Medicine, Longyan Hospital of Traditional Chinese Medicine, School of Medicine, Xiamen University, Longyan, Fujian, 364000, China.
| |
Collapse
|
12
|
de Coo IF, Jesse S, Le TL, Sala C, Bourgeron T. Consensus recommendations on Epilepsy in Phelan-McDermid syndrome. Eur J Med Genet 2023; 66:104746. [PMID: 36967043 DOI: 10.1016/j.ejmg.2023.104746] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 03/10/2023] [Accepted: 03/18/2023] [Indexed: 03/28/2023]
Abstract
Phelan-McDermid syndrome (PMS) is a 22q13.3 deletion syndrome that presents with a disturbed development, neurological and psychiatric characteristics, and sometimes other comorbidities like seizures. The epilepsy manifests itself in a variety of seizure semiologies. Further diagnostics using electroencephalogram (EEG) and brain magnetic resonance imaging (MRI) are important in conjunction with the clinical picture of the seizures to decide whether anticonvulsant therapy is necessary. As part of the development of European consensus guidelines we focussed on the prevalence and semiology of epileptic seizures in PMS associated with a pathogenic variant in the SHANK3 gene or the 22q13 deletion involving SHANK3, in order to then be able to make recommendations regarding diagnosis and therapy.
Collapse
|
13
|
Huang M, Qi Q, Xu T. Targeting Shank3 deficiency and paresthesia in autism spectrum disorder: A brief review. Front Mol Neurosci 2023; 16:1128974. [PMID: 36846568 PMCID: PMC9948097 DOI: 10.3389/fnmol.2023.1128974] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 01/18/2023] [Indexed: 02/11/2023] Open
Abstract
Autism spectrum disorder (ASD) includes a group of multifactorial neurodevelopmental disorders characterized by impaired social communication, social interaction, and repetitive behaviors. Several studies have shown an association between cases of ASD and mutations in the genes of SH3 and multiple ankyrin repeat domain protein 3 (SHANK3). These genes encode many cell adhesion molecules, scaffold proteins, and proteins involved in synaptic transcription, protein synthesis, and degradation. They have a profound impact on all aspects of synaptic transmission and plasticity, including synapse formation and degeneration, suggesting that the pathogenesis of ASD may be partially attributable to synaptic dysfunction. In this review, we summarize the mechanism of synapses related to Shank3 in ASD. We also discuss the molecular, cellular, and functional studies of experimental models of ASD and current autism treatment methods targeting related proteins.
Collapse
Affiliation(s)
- Min Huang
- Department of Anesthesiology, Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China,Department of Anesthesiology, Suzhou Hospital of Anhui Medical University, Suzhou, China
| | - Qi Qi
- Department of Anesthesiology, Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China,Department of Anesthesiology, Suzhou Hospital of Anhui Medical University, Suzhou, China
| | - Tao Xu
- Department of Anesthesiology, Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China,Department of Anesthesiology, Suzhou Hospital of Anhui Medical University, Suzhou, China,*Correspondence: Tao Xu,
| |
Collapse
|
14
|
Histone deacetylase 4 inhibition ameliorates the social deficits induced by Ephrin-B2 mutation. Prog Neuropsychopharmacol Biol Psychiatry 2023; 120:110622. [PMID: 36029930 DOI: 10.1016/j.pnpbp.2022.110622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 07/26/2022] [Accepted: 08/22/2022] [Indexed: 11/20/2022]
Abstract
Deterioration of inhibitory synapse may be an essential neurological basis underlying abnormal social behaviours. Manipulations that regulate GABAergic transmission are associated with improved behavioural phenotypes in sociability. The synaptic protein, Ephrin-B2 (EB2), plays an important role in the maintenance and reconfiguration of inhibitory synapses in the medial prefrontal cortex (mPFC). However, the inhibitory cell-type specific role of EB2 in the pathophysiology and treatment of social deficits remains unknown. As expected, we revealed that tdTomato-expressing cells were only found in GABAergic neurons instead of excitatory neurons in transgenic EB2-vGATCre mice. This result indicated that depletion of EB2 would occur in those neurons, which further contribute to social deficits. In addition, specific over-expression of mPFC EB2 restored the defective social behaviour abnormalities. These results suggest that the effect of EB2 on social deficits is anatomically and cell-type specific. More importantly, the global upregulation of HDAC4 expression was found in EB2-vGATCre mice. Significant subcellular nuclear shuttling of HDAC4 in vGAT+ neurons was examined and quantified, suggesting a role of nuclear HDAC4 in mediating the mechanism underlying EB2 impairment in vGAT+ neurons. Treatment with LMK235 led to a remarkable rescue of social deficits, thus our data revealed a new domain for the potential utility of HDAC targeting agents to treat social deficits. In conclusion, these results not only revealed a novel molecular mechanism underlying the pathophysiology of social deficits, but also suggested a potential intervention avenue for the treatment of social deficits.
Collapse
|
15
|
Bauer HF, Delling JP, Bockmann J, Boeckers TM, Schön M. Development of sex- and genotype-specific behavioral phenotypes in a Shank3 mouse model for neurodevelopmental disorders. Front Behav Neurosci 2023; 16:1051175. [PMID: 36699652 PMCID: PMC9868822 DOI: 10.3389/fnbeh.2022.1051175] [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: 09/22/2022] [Accepted: 12/05/2022] [Indexed: 01/11/2023] Open
Abstract
Individuals with a SHANK3-related neurodevelopmental disorder, also termed Phelan-McDermid syndrome or abbreviated as PMS, exhibit significant global developmental delay, language impairment, and muscular hypotonia. Also common are repetitive behaviors and altered social interactions, in line with a diagnosis of autism spectrum disorders. This study investigated the developmental aspect of autism-related behaviors and other phenotypes in a Shank3-transgenic mouse model. The animals underwent two sets of identical behavioral experiments, spanning motor skills, social and repetitive behavior, and cognition: baseline began at 5 weeks of age, corresponding to human adolescence, and the follow-up was initiated when aged 13 weeks, resembling early adulthood in humans. Interestingly, the animals displayed relatively stable phenotypes. Moreover, motor coordination and endurance were impaired, while muscle strength was unchanged. Surprisingly, the animals displayed only minor impairments in social behavior, but pronounced stereotypic and repetitive behaviors. Some behavioral tests indicated increased avoidance and anxiety. While spatial learning and memory were unchanged, knockout animals displayed slightly impaired cognitive flexibility. Female animals had similar abnormalities as males in the paradigms testing avoidance, anxiety, and cognition, but were less pathological in motor function and repetitive behavior. In all test paradigms, heterozygous Shank3 knockout animals had either no abnormal or a milder phenotype. Accurate characterization of animal models for genetic diseases is a prerequisite for understanding the pathophysiology. This is subsequently the basis for finding suitable and, ideally, translational biomarkers for therapeutic approaches and, thereby reducing the number of animals needed for preclinical trials.
Collapse
Affiliation(s)
- Helen Friedericke Bauer
- Institute for Anatomy and Cell Biology, Ulm University, Ulm, Germany,International Graduate School in Molecular Medicine Ulm, Ulm University, Ulm, Germany
| | | | - Jürgen Bockmann
- Institute for Anatomy and Cell Biology, Ulm University, Ulm, Germany
| | - Tobias M. Boeckers
- Institute for Anatomy and Cell Biology, Ulm University, Ulm, Germany,Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Ulm Site, Ulm, Germany,*Correspondence: Tobias M. Boeckers,
| | - Michael Schön
- Institute for Anatomy and Cell Biology, Ulm University, Ulm, Germany,Michael Schön,
| |
Collapse
|
16
|
Billeci L, Callara AL, Guiducci L, Prosperi M, Morales MA, Calderoni S, Muratori F, Santocchi E. A randomized controlled trial into the effects of probiotics on electroencephalography in preschoolers with autism. AUTISM : THE INTERNATIONAL JOURNAL OF RESEARCH AND PRACTICE 2023; 27:117-132. [PMID: 35362336 PMCID: PMC9806478 DOI: 10.1177/13623613221082710] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
LAY ABSTRACT This study investigates the effects of a probiotic on preschoolers' brain electrical activity with autism spectrum disorder. Autism is a disorder with an increasing prevalence characterized by an enormous individual, family, and social cost. Although the etiology of autism spectrum disorder is unknown, an interaction between genetic and environmental factors is implicated, converging in altered brain synaptogenesis and, therefore, connectivity. Besides deepening the knowledge on the resting brain electrical activity that characterizes this disorder, this study allows analyzing the positive central effects of a 6-month therapy with a probiotic through a randomized, double-blind placebo-controlled study and the correlations between electroencephalography activity and biochemical and clinical parameters. In subjects treated with probiotics, we observed a decrease of power in frontopolar regions in beta and gamma bands, and increased coherence in the same bands together with a shift in frontal asymmetry, which suggests a modification toward a typical brain activity. Electroencephalography measures were significantly correlated with clinical and biochemical measures. These findings support the importance of further investigations on probiotics' benefits in autism spectrum disorder to better elucidate mechanistic links between probiotics supplementation and changes in brain activity.
Collapse
Affiliation(s)
- Lucia Billeci
- Institute of Clinical Physiology,
National Research Council, Pisa, Italy
| | | | - Letizia Guiducci
- Institute of Clinical Physiology,
National Research Council, Pisa, Italy
| | - Margherita Prosperi
- Department of Developmental
Neuroscience, IRCCS Stella Maris Foundation, Pisa, Italy
| | | | - Sara Calderoni
- Department of Developmental
Neuroscience, IRCCS Stella Maris Foundation, Pisa, Italy
- Department of Clinical and Experimental
Medicine, University of Pisa, Pisa, Italy
| | - Filippo Muratori
- Department of Developmental
Neuroscience, IRCCS Stella Maris Foundation, Pisa, Italy
- Department of Clinical and Experimental
Medicine, University of Pisa, Pisa, Italy
| | - Elisa Santocchi
- UFSMIA zona Valle del Serchio, Azienda
USL Toscana Nord Ovest, Castelnuovo Garfagnana (LU), Italy
| |
Collapse
|
17
|
Shin S, You IJ, Jeong M, Bae Y, Wang XY, Cawley ML, Han A, Lim BK. Early adversity promotes binge-like eating habits by remodeling a leptin-responsive lateral hypothalamus-brainstem pathway. Nat Neurosci 2023; 26:79-91. [PMID: 36510113 PMCID: PMC9829538 DOI: 10.1038/s41593-022-01208-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 10/14/2022] [Indexed: 12/14/2022]
Abstract
Early-life trauma (ELT) is a risk factor for binge eating and obesity later in life, yet the neural circuits that underlie this association have not been addressed. Here, we show in mice that downregulation of the leptin receptor (Lepr) in the lateral hypothalamus (LH) and its effect on neural activity is crucial in causing ELT-induced binge-like eating and obesity upon high-fat diet exposure. We also found that the increased activity of Lepr-expressing LH (LHLepr) neurons encodes sustained binge-like eating in ELT mice. Inhibition of LHLepr neurons projecting to the ventrolateral periaqueductal gray normalizes these behavioral features of ELT mice. Furthermore, activation of proenkephalin-expressing ventrolateral periaqueductal gray neurons, which receive inhibitory inputs from LHLepr neurons, rescues ELT-induced maladaptive eating habits. Our results identify a circuit pathway that mediates ELT-induced maladaptive eating and may lead to the identification of novel therapeutic targets for binge eating and obesity.
Collapse
Affiliation(s)
- Sora Shin
- Fralin Biomedical Research Institute at VTC, Roanoke, VA, USA.
- FBRI Center for Neurobiology Research, Roanoke, VA, USA.
- Department of Human Nutrition, Foods, and Exercise, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA.
- Neurobiology Section, Division of Biological Sciences, University of California, San Diego, La Jolla, CA, USA.
| | - In-Jee You
- Fralin Biomedical Research Institute at VTC, Roanoke, VA, USA
- FBRI Center for Neurobiology Research, Roanoke, VA, USA
| | - Minju Jeong
- Neurobiology Section, Division of Biological Sciences, University of California, San Diego, La Jolla, CA, USA
| | - Yeeun Bae
- Fralin Biomedical Research Institute at VTC, Roanoke, VA, USA
- FBRI Center for Neurobiology Research, Roanoke, VA, USA
- Department of Human Nutrition, Foods, and Exercise, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA
| | - Xiao-Yun Wang
- Neurobiology Section, Division of Biological Sciences, University of California, San Diego, La Jolla, CA, USA
| | - Mikel Leann Cawley
- Fralin Biomedical Research Institute at VTC, Roanoke, VA, USA
- FBRI Center for Neurobiology Research, Roanoke, VA, USA
| | - Abraham Han
- Fralin Biomedical Research Institute at VTC, Roanoke, VA, USA
- FBRI Center for Neurobiology Research, Roanoke, VA, USA
- Department of Human Nutrition, Foods, and Exercise, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA
| | - Byung Kook Lim
- Neurobiology Section, Division of Biological Sciences, University of California, San Diego, La Jolla, CA, USA.
| |
Collapse
|
18
|
Bouquier N, Sakkaki S, Raynaud F, Hemonnot-Girard AL, Seube V, Compan V, Bertaso F, Perroy J, Moutin E. The Shank3 Venus/Venus knock in mouse enables isoform-specific functional studies of Shank3a. Front Neurosci 2022; 16:1081010. [PMID: 36570823 PMCID: PMC9773256 DOI: 10.3389/fnins.2022.1081010] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 11/23/2022] [Indexed: 12/14/2022] Open
Abstract
Background Shank3 is a scaffolding protein essential for the organization and function of the glutamatergic postsynapse. Monogenic mutations in SHANK3 gene are among the leading genetic causes of Autism Spectrum Disorders (ASD). The multiplicity of Shank3 isoforms seems to generate as much functional diversity and yet, there are no tools to study endogenous Shank3 proteins in an isoform-specific manner. Methods In this study, we created a novel transgenic mouse line, the Shank3Venus/Venus knock in mouse, which allows to monitor the endogenous expression of the major Shank3 isoform in the brain, the full-length Shank3a isoform. Results We show that the endogenous Venus-Shank3a protein is localized in spines and is mainly expressed in the striatum, hippocampus and cortex of the developing and adult brain. We show that Shank3Venus/+ and Shank3Venus/Venus mice have no behavioral deficiency. We further crossed Shank3Venus/Venus mice with Shank3ΔC/ΔC mice, a model of ASD, to track the Venus-tagged wild-type copy of Shank3a in physiological (Shank3Venus/+) and pathological (Shank3Venus/ΔC) conditions. We report a developmental delay in brain expression of the Venus-Shank3a isoform in Shank3Venus/ΔC mice, compared to Shank3Venus/+ control mice. Conclusion Altogether, our results show that the Shank3Venus/Venus mouse line is a powerful tool to study endogenous Shank3a expression, in physiological conditions and in ASD.
Collapse
Affiliation(s)
- Nathalie Bouquier
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Sophie Sakkaki
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Fabrice Raynaud
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, Montpellier, France,PhyMedExp, Univ Montpellier, INSERM, CNRS, CHU de Montpellier, Montpellier, France
| | | | - Vincent Seube
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Vincent Compan
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Federica Bertaso
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Julie Perroy
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, Montpellier, France,*Correspondence: Julie Perroy,
| | - Enora Moutin
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, Montpellier, France,Enora Moutin,
| |
Collapse
|
19
|
SHANK family on stem cell fate and development. Cell Death Dis 2022; 13:880. [PMID: 36257935 PMCID: PMC9579136 DOI: 10.1038/s41419-022-05325-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 09/30/2022] [Accepted: 10/05/2022] [Indexed: 11/24/2022]
Abstract
SH3 and multiple ankyrin repeat domains protein (SHANK) 1, SHANK2, and SHANK3 encode a family of postsynaptic scaffolding proteins present at glutamatergic synapses and play a crucial role in synaptogenesis. In the past years, studies have provided a preliminary appreciation and understanding of the influence of the SHANK family in controlling stem cell fate. Here, we review the modulation of SHANK gene expression and their related signaling pathways, allowing for an in-depth understanding of the role of SHANK in stem cells. Besides, their role in governing stem cell self-renewal, proliferation, differentiation, apoptosis, and metabolism are explored in neural stem cells (NSCs), stem cells from apical papilla (SCAPs), and induced pluripotent stem cells (iPSCs). Moreover, iPSCs and embryonic stem cells (ESCs) have been utilized as model systems for analyzing their functions in terms of neuronal development. SHANK-mediated stem cell fate determination is an intricate and multifactorial process. This study aims to achieve a better understanding of the role of SHANK in these processes and their clinical applications, thereby advancing the field of stem cell therapy. This review unravels the regulatory role of the SHANK family in the fate of stem cells.
Collapse
|
20
|
Xu D, Meng Y, An S, Meng W, Li H, Zhang W, Xue Y, Lan X, Wang X, Li M, Zhang X, Zhihao Z, Zhao Y, Yang H, Zhang C, Zhang R, Zhen Z. Swimming exercise is a promising early intervention for autism-like behavior in Shank3 deletion rats. CNS Neurosci Ther 2022; 29:78-90. [PMID: 36221783 PMCID: PMC9804047 DOI: 10.1111/cns.13920] [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: 03/15/2022] [Revised: 06/14/2022] [Accepted: 06/20/2022] [Indexed: 02/06/2023] Open
Abstract
INTRODUCTION SHANK3 is an important excitatory postsynaptic scaffold protein, and its mutations lead to genetic cause of neurodevelopmental diseases including autism spectrum disorders (ASD), Philan McDermid syndrome (PMS), and intellectual disability (ID). Early prevention and treatment are important for Shank3 gene mutation disease. Swimming has been proven to have a positive effect on neurodegenerative diseases. METHODS Shank3 gene exon 11-21 knockout rats were intervened by a 40 min/day, 5 day/week for 8-week protocol. After the intervention, the rats were tested to behavioral measures such as learning and memory, and the volume and H-spectrum of the brain were measured using MRI; hippocampal dendritic spines were measured using Golgi staining and laser confocal. RESULTS The results showed that Shank3-deficient rats had significant deficits in social memory, object recognition, and water maze learning decreased hippocampal volume and number of neurons, and lower levels of related scaffold proteins and receptor proteins were found in Shank3-deficient rats. CONCLUSION It is suggested that early swimming exercise has a positive effect on Shank3 gene-deficient rats, which provides a new therapeutic strategy for the prevention and recovery of neurodevelopmental disorders.
Collapse
Affiliation(s)
- Dan Xu
- College of P.E and SportsBeijing Normal UniversityBeijingChina,Sports and Health Editorial OfficePeople's education pressBeijingChina
| | - Yunchen Meng
- Department of P.E.China University of Mining and Technology‐BeijingBeijingChina
| | - Shasha An
- College of P.E and SportsBeijing Normal UniversityBeijingChina
| | - Wenshu Meng
- College of Life SciencesBeijing Normal UniversityBeijingChina
| | - Hanran Li
- Centre for Cognitive and Brain Sciences and Department of PsychologyUniversity of MacauTaipaMacau
| | - Weinan Zhang
- College of P.E and SportsBeijing Normal UniversityBeijingChina
| | - Yaqi Xue
- College of P.E and SportsBeijing Normal UniversityBeijingChina
| | - Xinyu Lan
- Department of Neurobiology, School of Basic Medical SciencesPeking UniversityBeijingChina,Neuroscience Research InstitutePeking UniversityBeijingChina,Key Laboratory for Neuroscience, Ministry of Education/National Health and Family Planning CommissionPeking UniversityBeijingChina,Autism Research Center of Peking University Health Science CenterBeijingChina
| | - Xiaoxi Wang
- Institute of Acupuncture and MoxibustionChina Academy of Chinese Medical SciencesBeijingChina
| | - Mingjuan Li
- Department of Neurobiology, School of Basic Medical SciencesPeking UniversityBeijingChina,Neuroscience Research InstitutePeking UniversityBeijingChina,Key Laboratory for Neuroscience, Ministry of Education/National Health and Family Planning CommissionPeking UniversityBeijingChina,Autism Research Center of Peking University Health Science CenterBeijingChina
| | - Xiaoyan Zhang
- College of P.E and SportsBeijing Normal UniversityBeijingChina
| | - Zhang Zhihao
- College of P.E and SportsBeijing Normal UniversityBeijingChina
| | - Yu Zhao
- College of P.E and SportsBeijing Normal UniversityBeijingChina
| | - Haodong Yang
- College of P.E and SportsBeijing Normal UniversityBeijingChina
| | - Chen Zhang
- Department of Neurobiology, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Advanced Innovation Center for Human Brain ProtectionCapital Medical UniversityBeijingChina
| | - Rong Zhang
- Department of Neurobiology, School of Basic Medical SciencesPeking UniversityBeijingChina,Neuroscience Research InstitutePeking UniversityBeijingChina,Key Laboratory for Neuroscience, Ministry of Education/National Health and Family Planning CommissionPeking UniversityBeijingChina,Autism Research Center of Peking University Health Science CenterBeijingChina,Department of Integration of Chinese and Western MedicineSchool of Basic Medical Sciences, Peking UniversityBeijingChina
| | - Zhiping Zhen
- College of P.E and SportsBeijing Normal UniversityBeijingChina
| |
Collapse
|
21
|
Moore S, Amatya DN, Chu MM, Besterman AD. Catatonia in autism and other neurodevelopmental disabilities: a state-of-the-art review. NPJ MENTAL HEALTH RESEARCH 2022; 1:12. [PMID: 38609506 PMCID: PMC10955936 DOI: 10.1038/s44184-022-00012-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 08/26/2022] [Indexed: 04/14/2024]
Abstract
Individuals with neurodevelopmental disabilities (NDDs) may be at increased risk for catatonia, which can be an especially challenging condition to diagnose and treat. There may be symptom overlap between catatonia and NDD-associated behaviors, such as stereotypies. The diagnosis of catatonia should perhaps be adjusted to address symptom overlap and to include extreme behaviors observed in patients with NDDs, such as severe self-injury. Risk factors for catatonia in individuals with NDDs may include trauma and certain genetic variants, such as those that disrupt SHANK3. Common etiologic features between neurodevelopmental disabilities and catatonia, such as excitatory/inhibitory imbalance and neuroimmune dysfunction, may partially account for comorbidity. New approaches leveraging genetic testing and neuroimmunologic evaluation may allow for more precise diagnoses and effective treatments.
Collapse
Affiliation(s)
- Shavon Moore
- University of California San Diego, Department of Psychiatry, San Diego, CA, USA
- Rady Children's Hospital San Diego, Division of Behavioral Health Services, San Diego, CA, USA
| | - Debha N Amatya
- University of California San Diego, Department of Psychiatry, San Diego, CA, USA
- UCLA Semel Institute of Neuroscience and Human Behavior, Los Angeles, CA, USA
| | - Michael M Chu
- University of California San Diego, Department of Psychiatry, San Diego, CA, USA
- Rady Children's Hospital San Diego, Division of Behavioral Health Services, San Diego, CA, USA
- Children's Hospital of Orange County, Division of Child and Adolescent Psychiatry, Orange, CA, USA
- University of California Irvine, Department of Psychiatry, Irvine, CA, USA
| | - Aaron D Besterman
- University of California San Diego, Department of Psychiatry, San Diego, CA, USA.
- Rady Children's Hospital San Diego, Division of Behavioral Health Services, San Diego, CA, USA.
- Rady Children's Institute for Genomic Medicine, San Diego, CA, USA.
| |
Collapse
|
22
|
Al-Absi AR, Thambiappa SK, Khan AR, Glerup S, Sanchez C, Landau AM, Nyengaard JR. Df(h22q11)/+ mouse model exhibits reduced binding levels of GABA A receptors and structural and functional dysregulation in the inhibitory and excitatory networks of hippocampus. Mol Cell Neurosci 2022; 122:103769. [PMID: 35988854 DOI: 10.1016/j.mcn.2022.103769] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Revised: 08/11/2022] [Accepted: 08/14/2022] [Indexed: 11/17/2022] Open
Abstract
The 22q11.2 hemizygous deletion confers high risk for multiple neurodevelopmental disorders. Inhibitory signaling, largely regulated through GABAA receptors, is suggested to serve a multitude of brain functions that are disrupted in the 22q11.2 deletion syndrome. We investigated the putative deficit of GABAA receptors and the potential substrates contributing to the inhibitory and excitatory dysregulations in hippocampal networks of the Df(h22q11)/+ mouse model of the 22q11.2 hemizygous deletion. The Df(h22q11)/+ mice exhibited impairments in several hippocampus-related functional domains, represented by impaired spatial memory and sensory gating functions. Autoradiography using the [3H]muscimol tracer revealed a significant reduction in GABAA receptor binding in the CA1 and CA3 subregions, together with a loss of GAD67+ interneurons in CA1 of Df(h22q11)/+ mice. Furthermore, electrophysiology recordings exhibited significantly higher neuronal activity in CA3, in response to the GABAA receptor antagonist, bicuculline, as compared with wild type mice. Density and volume of dendritic spines in pyramidal neurons were reduced and Sholl analysis also showed a reduction in the complexity of basal dendritic tree in CA1 and CA3 subregions of Df(h22q11)/+ mice. Overall, our findings demonstrate that hemizygous deletion in the 22q11.2 locus leads to dysregulations in the inhibitory circuits, involving reduced binding levels of GABAA receptors, in addition to functional and structural modulations of the excitatory networks of hippocampus.
Collapse
Affiliation(s)
- Abdel-Rahman Al-Absi
- Center for Molecular Morphology, Section for Stereology and Microscopy, Department of Clinical Medicine, Aarhus University, Denmark; Department of Pathology, Aarhus University Hospital, Denmark.
| | - Sakeerthi Kethees Thambiappa
- Center for Molecular Morphology, Section for Stereology and Microscopy, Department of Clinical Medicine, Aarhus University, Denmark; Department of Pathology, Aarhus University Hospital, Denmark.
| | - Ahmad Raza Khan
- Centre of Biomedical Research (CBMR), SGPGIMS Campus, Lucknow, India.
| | - Simon Glerup
- Department of Biomedicine, Aarhus University, Denmark.
| | - Connie Sanchez
- Translational Neuropsychiatry Unit, Aarhus University, Denmark.
| | - Anne M Landau
- Translational Neuropsychiatry Unit, Aarhus University, Denmark; Department of Nuclear Medicine and PET Centre, Aarhus University Hospital, Denmark.
| | - Jens R Nyengaard
- Center for Molecular Morphology, Section for Stereology and Microscopy, Department of Clinical Medicine, Aarhus University, Denmark; Department of Pathology, Aarhus University Hospital, Denmark.
| |
Collapse
|
23
|
Wan L, Liu D, Xiao WB, Zhang BX, Yan XX, Luo ZH, Xiao B. Association of SHANK Family with Neuropsychiatric Disorders: An Update on Genetic and Animal Model Discoveries. Cell Mol Neurobiol 2022; 42:1623-1643. [PMID: 33595806 PMCID: PMC11421742 DOI: 10.1007/s10571-021-01054-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 02/02/2021] [Indexed: 12/14/2022]
Abstract
The Shank family proteins are enriched at the postsynaptic density (PSD) of excitatory glutamatergic synapses. They serve as synaptic scaffolding proteins and appear to play a critical role in the formation, maintenance and functioning of synapse. Increasing evidence from genetic association and animal model studies indicates a connection of SHANK genes defects with the development of neuropsychiatric disorders. In this review, we first update the current understanding of the SHANK family genes and their encoded protein products. We then denote the literature relating their alterations to the risk of neuropsychiatric diseases. We further review evidence from animal models that provided molecular insights into the biological as well as pathogenic roles of Shank proteins in synapses, and the potential relationship to the development of abnormal neurobehavioral phenotypes.
Collapse
Affiliation(s)
- Lily Wan
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Du Liu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- Taikang Tongji Hospital, Wuhan, 430050, Hubei, China
| | - Wen-Biao Xiao
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Bo-Xin Zhang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Xiao-Xin Yan
- Department of Anatomy and Neurobiology, Central South University, Changsha, 410013, Hunan, China
| | - Zhao-Hui Luo
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
| | - Bo Xiao
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
| |
Collapse
|
24
|
Jung S, Park M. Shank postsynaptic scaffolding proteins in autism spectrum disorder: Mouse models and their dysfunctions in behaviors, synapses, and molecules. Pharmacol Res 2022; 182:106340. [DOI: 10.1016/j.phrs.2022.106340] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 06/29/2022] [Accepted: 06/30/2022] [Indexed: 01/03/2023]
|
25
|
Chaudry S, Vasudevan N. mTOR-Dependent Spine Dynamics in Autism. Front Mol Neurosci 2022; 15:877609. [PMID: 35782388 PMCID: PMC9241970 DOI: 10.3389/fnmol.2022.877609] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 04/25/2022] [Indexed: 12/12/2022] Open
Abstract
Autism Spectrum Conditions (ASC) are a group of neurodevelopmental disorders characterized by deficits in social communication and interaction as well as repetitive behaviors and restricted range of interests. ASC are complex genetic disorders with moderate to high heritability, and associated with atypical patterns of neural connectivity. Many of the genes implicated in ASC are involved in dendritic spine pruning and spine development, both of which can be mediated by the mammalian target of rapamycin (mTOR) signaling pathway. Consistent with this idea, human postmortem studies have shown increased spine density in ASC compared to controls suggesting that the balance between autophagy and spinogenesis is altered in ASC. However, murine models of ASC have shown inconsistent results for spine morphology, which may underlie functional connectivity. This review seeks to establish the relevance of changes in dendritic spines in ASC using data gathered from rodent models. Using a literature survey, we identify 20 genes that are linked to dendritic spine pruning or development in rodents that are also strongly implicated in ASC in humans. Furthermore, we show that all 20 genes are linked to the mTOR pathway and propose that the mTOR pathway regulating spine dynamics is a potential mechanism underlying the ASC signaling pathway in ASC. We show here that the direction of change in spine density was mostly correlated to the upstream positive or negative regulation of the mTOR pathway and most rodent models of mutant mTOR regulators show increases in immature spines, based on morphological analyses. We further explore the idea that these mutations in these genes result in aberrant social behavior in rodent models that is due to these altered spine dynamics. This review should therefore pave the way for further research on the specific genes outlined, their effect on spine morphology or density with an emphasis on understanding the functional role of these changes in ASC.
Collapse
|
26
|
Kim S, Kim YE, Song I, Ujihara Y, Kim N, Jiang YH, Yin HH, Lee TH, Kim IH. Neural circuit pathology driven by Shank3 mutation disrupts social behaviors. Cell Rep 2022; 39:110906. [PMID: 35675770 PMCID: PMC9210496 DOI: 10.1016/j.celrep.2022.110906] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 03/21/2022] [Accepted: 05/10/2022] [Indexed: 11/30/2022] Open
Abstract
Dysfunctional sociability is a core symptom in autism spectrum disorder (ASD) that may arise from neural-network dysconnectivity between multiple brain regions. However, pathogenic neural-network mechanisms underlying social dysfunction are largely unknown. Here, we demonstrate that circuit-selective mutation (ctMUT) of ASD-risk Shank3 gene within a unidirectional projection from the prefrontal cortex to the basolateral amygdala alters spine morphology and excitatory-inhibitory balance of the circuit. Shank3 ctMUT mice show reduced sociability as well as elevated neural activity and its amplitude variability, which is consistent with the neuroimaging results from human ASD patients. Moreover, the circuit hyper-activity disrupts the temporal correlation of socially tuned neurons to the events of social interactions. Finally, optogenetic circuit activation in wild-type mice partially recapitulates the reduced sociability of Shank3 ctMUT mice, while circuit inhibition in Shank3 ctMUT mice partially rescues social behavior. Collectively, these results highlight a circuit-level pathogenic mechanism of Shank3 mutation that drives social dysfunction.
Collapse
Affiliation(s)
- Sunwhi Kim
- Department of Anatomy and Neurobiology, University of Tennessee Health Science Center, Memphis, TN 38163, USA; Neuroscience Institute, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Yong-Eun Kim
- Department of Anatomy and Neurobiology, University of Tennessee Health Science Center, Memphis, TN 38163, USA; Neuroscience Institute, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Inuk Song
- Department of Psychology, Virginia Tech, Blacksburg, VA 24061, USA
| | - Yusuke Ujihara
- Department of Anatomy and Neurobiology, University of Tennessee Health Science Center, Memphis, TN 38163, USA; Neuroscience Institute, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Namsoo Kim
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA
| | - Yong-Hui Jiang
- Department of Genetics, Pediatrics and Neuroscience, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Henry H Yin
- Department of Psychology and Neuroscience, Duke University, Durham, NC 27708, USA
| | - Tae-Ho Lee
- Department of Psychology, Virginia Tech, Blacksburg, VA 24061, USA
| | - Il Hwan Kim
- Department of Anatomy and Neurobiology, University of Tennessee Health Science Center, Memphis, TN 38163, USA; Neuroscience Institute, University of Tennessee Health Science Center, Memphis, TN 38163, USA.
| |
Collapse
|
27
|
Sauer AK, Hagmeyer S, Grabrucker AM. Prenatal Zinc Deficient Mice as a Model for Autism Spectrum Disorders. Int J Mol Sci 2022; 23:ijms23116082. [PMID: 35682762 PMCID: PMC9181257 DOI: 10.3390/ijms23116082] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 05/18/2022] [Accepted: 05/19/2022] [Indexed: 02/06/2023] Open
Abstract
Epidemiological studies have shown a clear association between early life zinc deficiency and Autism Spectrum Disorders (ASD). In line with this, mouse models have revealed prenatal zinc deficiency as a profound risk factor for neurobiological and behavioral abnormalities in the offspring reminiscent of ASD behavior. From these studies, a complex pathology emerges, with alterations in the gastrointestinal and immune system and synaptic signaling in the brain, as a major consequence of prenatal zinc deficiency. The features represent a critical link in a causal chain that leads to various neuronal dysfunctions and behavioral phenotypes observed in prenatal zinc deficient (PZD) mice and probably other mouse models for ASD. Given that the complete phenotype of PZD mice may be key to understanding how non-genetic factors can modify the clinical features and severity of autistic patients and explain the observed heterogeneity, here, we summarize published data on PZD mice. We critically review the emerging evidence that prenatal zinc deficiency is at the core of several environmental risk factors associated with ASD, being mechanistically linked to ASD-associated genetic factors. In addition, we highlight future directions and outstanding questions, including potential symptomatic, disease-modifying, and preventive treatment strategies.
Collapse
Affiliation(s)
- Ann Katrin Sauer
- Department of Biological Sciences, University of Limerick, V94 T9PX Limerick, Ireland; (A.K.S.); (S.H.)
- Bernal Institute, University of Limerick, V94 T9PX Limerick, Ireland
- Health Research Institute (HRI), University of Limerick, V94 T9PX Limerick, Ireland
| | - Simone Hagmeyer
- Department of Biological Sciences, University of Limerick, V94 T9PX Limerick, Ireland; (A.K.S.); (S.H.)
| | - Andreas M. Grabrucker
- Department of Biological Sciences, University of Limerick, V94 T9PX Limerick, Ireland; (A.K.S.); (S.H.)
- Bernal Institute, University of Limerick, V94 T9PX Limerick, Ireland
- Health Research Institute (HRI), University of Limerick, V94 T9PX Limerick, Ireland
- Correspondence: ; Tel.: +353-61-237756
| |
Collapse
|
28
|
Urrutia-Ruiz C, Rombach D, Cursano S, Gerlach-Arbeiter S, Schoen M, Bockmann J, Demestre M, Boeckers TM. Deletion of the Autism-Associated Protein SHANK3 Abolishes Structural Synaptic Plasticity after Brain Trauma. Int J Mol Sci 2022; 23:ijms23116081. [PMID: 35682760 PMCID: PMC9181590 DOI: 10.3390/ijms23116081] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 05/26/2022] [Accepted: 05/27/2022] [Indexed: 12/14/2022] Open
Abstract
Autism spectrum disorders (ASDs) are characterized by repetitive behaviors and impairments of sociability and communication. About 1% of ASD cases are caused by mutations of SHANK3, a major scaffolding protein of the postsynaptic density. We studied the role of SHANK3 in plastic changes of excitatory synapses within the central nervous system by employing mild traumatic brain injury (mTBI) in WT and Shank3 knockout mice. In WT mice, mTBI triggered ipsi- and contralateral loss of hippocampal dendritic spines and excitatory synapses with a partial recovery over time. In contrast, no significant synaptic alterations were detected in Shank3∆11−/− mice, which showed fewer dendritic spines and excitatory synapses at baseline. In line, mTBI induced the upregulation of synaptic plasticity-related proteins Arc and p-cofilin only in WT mice. Interestingly, microglia proliferation was observed in WT mice after mTBI but not in Shank3∆11−/− mice. Finally, we detected TBI-induced increased fear memory at the behavioral level, whereas in Shank3∆11−/− animals, the already-enhanced fear memory levels increased only slightly after mTBI. Our data show the lack of structural synaptic plasticity in Shank3 knockout mice that might explain at least in part the rigidity of behaviors, problems in adjusting to new situations and cognitive deficits seen in ASDs.
Collapse
Affiliation(s)
- Carolina Urrutia-Ruiz
- Institute for Anatomy and Cell Biology, Albert Einstein Allee 11, 89081 Ulm, Germany; (C.U.-R.); (D.R.); (S.C.); (S.G.-A.); (M.S.); (J.B.); (M.D.)
| | - Daniel Rombach
- Institute for Anatomy and Cell Biology, Albert Einstein Allee 11, 89081 Ulm, Germany; (C.U.-R.); (D.R.); (S.C.); (S.G.-A.); (M.S.); (J.B.); (M.D.)
| | - Silvia Cursano
- Institute for Anatomy and Cell Biology, Albert Einstein Allee 11, 89081 Ulm, Germany; (C.U.-R.); (D.R.); (S.C.); (S.G.-A.); (M.S.); (J.B.); (M.D.)
| | - Susanne Gerlach-Arbeiter
- Institute for Anatomy and Cell Biology, Albert Einstein Allee 11, 89081 Ulm, Germany; (C.U.-R.); (D.R.); (S.C.); (S.G.-A.); (M.S.); (J.B.); (M.D.)
| | - Michael Schoen
- Institute for Anatomy and Cell Biology, Albert Einstein Allee 11, 89081 Ulm, Germany; (C.U.-R.); (D.R.); (S.C.); (S.G.-A.); (M.S.); (J.B.); (M.D.)
| | - Juergen Bockmann
- Institute for Anatomy and Cell Biology, Albert Einstein Allee 11, 89081 Ulm, Germany; (C.U.-R.); (D.R.); (S.C.); (S.G.-A.); (M.S.); (J.B.); (M.D.)
| | - Maria Demestre
- Institute for Anatomy and Cell Biology, Albert Einstein Allee 11, 89081 Ulm, Germany; (C.U.-R.); (D.R.); (S.C.); (S.G.-A.); (M.S.); (J.B.); (M.D.)
| | - Tobias M. Boeckers
- Institute for Anatomy and Cell Biology, Albert Einstein Allee 11, 89081 Ulm, Germany; (C.U.-R.); (D.R.); (S.C.); (S.G.-A.); (M.S.); (J.B.); (M.D.)
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Translational Biochemistry, 89081 Ulm, Germany
- Correspondence: ; Tel.: +49-731-5002-3220
| |
Collapse
|
29
|
GENDER DIFFERENCES IN EFFECTS OF PRENATAL AND POSTNATAL EXPOSURE TO ELECTROMAGNETIC FIELD AND PRENATAL ZINC ON BEHAVIOUR AND SYNAPTIC PROTEINS IN RATS. J Chem Neuroanat 2022; 122:102092. [DOI: 10.1016/j.jchemneu.2022.102092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 03/25/2022] [Accepted: 03/25/2022] [Indexed: 11/21/2022]
|
30
|
Cooke J, Molloy CJ, Cáceres ASJ, Dinneen T, Bourgeron T, Murphy D, Gallagher L, Loth E. The Synaptic Gene Study: Design and Methodology to Identify Neurocognitive Markers in Phelan-McDermid Syndrome and NRXN1 Deletions. Front Neurosci 2022; 16:806990. [PMID: 35250452 PMCID: PMC8894872 DOI: 10.3389/fnins.2022.806990] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 01/26/2022] [Indexed: 11/26/2022] Open
Abstract
Synaptic gene conditions, i.e., “synaptopathies,” involve disruption to genes expressed at the synapse and account for between 0.5 and 2% of autism cases. They provide a unique entry point to understanding the molecular and biological mechanisms underpinning autism-related phenotypes. Phelan-McDermid Syndrome (PMS, also known as 22q13 deletion syndrome) and NRXN1 deletions (NRXN1ds) are two synaptopathies associated with autism and related neurodevelopmental disorders (NDDs). PMS often incorporates disruption to the SHANK3 gene, implicated in excitatory postsynaptic scaffolding, whereas the NRXN1 gene encodes neurexin-1, a presynaptic cell adhesion protein; both are implicated in trans-synaptic signaling in the brain. Around 70% of individuals with PMS and 43–70% of those with NRXN1ds receive a diagnosis of autism, suggesting that alterations in synaptic development may play a crucial role in explaining the aetiology of autism. However, a substantial amount of heterogeneity exists between conditions. Most individuals with PMS have moderate to profound intellectual disability (ID), while those with NRXN1ds have no ID to severe ID. Speech abnormalities are common to both, although appear more severe in PMS. Very little is currently known about the neurocognitive underpinnings of phenotypic presentations in PMS and NRXN1ds. The Synaptic Gene (SynaG) study adopts a gene-first approach and comprehensively assesses these two syndromic forms of autism. The study compliments preclinical efforts within AIMS-2-TRIALS focused on SHANK3 and NRXN1. The aims of the study are to (1) establish the frequency of autism diagnosis and features in individuals with PMS and NRXN1ds, (2) to compare the clinical profile of PMS, NRXN1ds, and individuals with ‘idiopathic’ autism (iASD), (3) to identify mechanistic biomarkers that may account for autistic features and/or heterogeneity in clinical profiles, and (4) investigate the impact of second or multiple genetic hits on heterogeneity in clinical profiles. In the current paper we describe our methodology for phenotyping the sample and our planned comparisons, with information on the necessary adaptations made during the global COVID-19 pandemic. We also describe the demographics of the data collected thus far, including 25 PMS, 36 NRXN1ds, 33 iASD, and 52 NTD participants, and present an interim analysis of autistic features and adaptive functioning.
Collapse
Affiliation(s)
- Jennifer Cooke
- Forensic and Neurodevelopmental Sciences, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, United Kingdom
- *Correspondence: Jennifer Cooke,
| | - Ciara J. Molloy
- Department of Psychiatry, School of Medicine, Trinity College Dublin, Dublin, Ireland
| | - Antonia San José Cáceres
- Forensic and Neurodevelopmental Sciences, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, United Kingdom
- Fundación para la Investigación Biomédica del Hospital General Universitario Gregorio Marañón, Madrid, Spain
- Biomedical Research Networking Center for Mental Health Network (CIBERSAM), Madrid, Spain
| | - Thomas Dinneen
- Department of Psychiatry, School of Medicine, Trinity College Dublin, Dublin, Ireland
| | | | - Declan Murphy
- Forensic and Neurodevelopmental Sciences, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, United Kingdom
| | - Louise Gallagher
- Department of Psychiatry, School of Medicine, Trinity College Dublin, Dublin, Ireland
| | - Eva Loth
- Forensic and Neurodevelopmental Sciences, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, United Kingdom
- Eva Loth,
| |
Collapse
|
31
|
Oberländer K, Witte V, Mallien AS, Gass P, Bengtson CP, Bading H. Dysregulation of Npas4 and Inhba expression and an altered excitation-inhibition balance are associated with cognitive deficits in DBA/2 mice. Learn Mem 2022; 29:55-70. [PMID: 35042829 PMCID: PMC8774195 DOI: 10.1101/lm.053527.121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 11/28/2021] [Indexed: 02/03/2023]
Abstract
Differences in the learning associated transcriptional profiles between mouse strains with distinct learning abilities could provide insight into the molecular basis of learning and memory. The inbred mouse strain DBA/2 shows deficits in hippocampus-dependent memory, yet the transcriptional responses to learning and the underlying mechanisms of the impairments are unknown. Comparing DBA/2J mice with the reference standard C57BL/6N mouse strain we verify an enhanced susceptibility to kainic acid induced seizures, confirm impairments in hippocampus-dependent spatial memory tasks and uncover additional behavioral abnormalities including deficits in hippocampus-independent learning. Surprisingly, we found no broad dysfunction of the DBA/2J strain in immediate early gene (IEG) activation but instead report brain region-specific and gene-specific alterations. The learning-associated IEGs Arc, c-Fos, and Nr4a1 showed no DBA/2J deficits in basal or synaptic activity induced gene expression in hippocampal or cortical primary neuronal cultures or in the CA1, CA3, or retrosplenial cortex following spatial object recognition (SOR) training in vivo. However, the parietal cortex showed reduced and the dentate gyrus showed enhanced SOR-evoked induction of most IEGs. All DBA/2J hippocampal regions exhibited elevated basal expression of inhibin β A (Inhba) and a learning-associated superinduction of the transcription factor neuronal Per-Arnt-Sim domain protein 4 (Npas4) known to regulate the synaptic excitation-inhibition balance. In line with this, CA1 pyramidal neurons of DBA/2J mice showed fewer inhibitory and more excitatory miniature postsynaptic currents but no alteration in most other electrophysiological properties or gross dendritic morphology. The dysregulation of Npas4 and Inhba expression and synaptic connectivity may underlie the cognitive deficits and increased susceptibility to seizures of DBA/2J mice.
Collapse
Affiliation(s)
- Kristin Oberländer
- Department of Neurobiology, Interdisciplinary Center for Neurosciences (IZN), Heidelberg University, 69120 Heidelberg, Germany
| | - Victoria Witte
- Department of Neurobiology, Interdisciplinary Center for Neurosciences (IZN), Heidelberg University, 69120 Heidelberg, Germany
| | - Anne Stephanie Mallien
- Department of Psychiatry and Psychotherapy, Research Group Animal Models in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim/Heidelberg University, 68159 Mannheim, Germany
| | - Peter Gass
- Department of Psychiatry and Psychotherapy, Research Group Animal Models in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim/Heidelberg University, 68159 Mannheim, Germany
| | - C. Peter Bengtson
- Department of Neurobiology, Interdisciplinary Center for Neurosciences (IZN), Heidelberg University, 69120 Heidelberg, Germany
| | - Hilmar Bading
- Department of Neurobiology, Interdisciplinary Center for Neurosciences (IZN), Heidelberg University, 69120 Heidelberg, Germany
| |
Collapse
|
32
|
Krüttner S, Falasconi A, Valbuena S, Galimberti I, Bouwmeester T, Arber S, Caroni P. Absence of familiarity triggers hallmarks of autism in mouse model through aberrant tail-of-striatum and prelimbic cortex signaling. Neuron 2022; 110:1468-1482.e5. [DOI: 10.1016/j.neuron.2022.02.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 01/10/2022] [Accepted: 01/28/2022] [Indexed: 12/28/2022]
|
33
|
Vyas Y, Cheyne JE, Lee K, Jung Y, Cheung PY, Montgomery JM. Shankopathies in the Developing Brain in Autism Spectrum Disorders. Front Neurosci 2022; 15:775431. [PMID: 35002604 PMCID: PMC8727517 DOI: 10.3389/fnins.2021.775431] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 11/22/2021] [Indexed: 11/13/2022] Open
Abstract
The SHANK family of proteins play critical structural and functional roles in the postsynaptic density (PSD) at excitatory glutamatergic synapses. Through their multidomain structure they form a structural platform across the PSD for protein–protein interactions, as well as recruiting protein complexes to strengthen excitatory synaptic transmission. Mutations in SHANKs reflect their importance to synapse development and plasticity. This is evident in autism spectrum disorder (ASD), a neurodevelopmental disorder resulting in behavioural changes including repetitive behaviours, lack of sociability, sensory issues, learning, and language impairments. Human genetic studies have revealed ASD mutations commonly occur in SHANKs. Rodent models expressing these mutations display ASD behavioural impairments, and a subset of these deficits are rescued by reintroduction of Shank in adult animals, suggesting that lack of SHANK during key developmental periods can lead to permanent changes in the brain’s wiring. Here we explore the differences in synaptic function and plasticity from development onward in rodent Shank ASD models. To date the most explored brain regions, relate to the behavioural changes observed, e.g., the striatum, hippocampus, sensory, and prefrontal cortex. In addition, less-studied regions including the hypothalamus, cerebellum, and peripheral nervous system are also affected. Synaptic phenotypes include weakened but also strengthened synaptic function, with NMDA receptors commonly affected, as well as changes in the balance of excitation and inhibition especially in cortical brain circuits. The effects of shankopathies in activity-dependent brain wiring is an important target for therapeutic intervention. We therefore highlight areas of research consensus and identify remaining questions and challenges.
Collapse
Affiliation(s)
- Yukti Vyas
- Department of Physiology, Faculty of Medical and Health Sciences, Centre for Brain Research, University of Auckland, Auckland, New Zealand
| | - Juliette E Cheyne
- Department of Physiology, Faculty of Medical and Health Sciences, Centre for Brain Research, University of Auckland, Auckland, New Zealand
| | - Kevin Lee
- Department of Physiology, Faculty of Medical and Health Sciences, Centre for Brain Research, University of Auckland, Auckland, New Zealand.,Department of Pharmacology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Yewon Jung
- Department of Physiology, Faculty of Medical and Health Sciences, Centre for Brain Research, University of Auckland, Auckland, New Zealand.,Department of Pharmacology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Pang Ying Cheung
- Department of Physiology, Faculty of Medical and Health Sciences, Centre for Brain Research, University of Auckland, Auckland, New Zealand
| | - Johanna M Montgomery
- Department of Physiology, Faculty of Medical and Health Sciences, Centre for Brain Research, University of Auckland, Auckland, New Zealand
| |
Collapse
|
34
|
Garrido D, Beretta S, Grabrucker S, Bauer HF, Bayer D, Sala C, Verpelli C, Roselli F, Bockmann J, Proepper C, Catanese A, Boeckers TM. Shank2/3 double knockout-based screening of cortical subregions links the retrosplenial area to the loss of social memory in autism spectrum disorders. Mol Psychiatry 2022; 27:4994-5006. [PMID: 36100669 PMCID: PMC9763120 DOI: 10.1038/s41380-022-01756-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 08/15/2022] [Accepted: 08/15/2022] [Indexed: 01/19/2023]
Abstract
Members of the Shank protein family are master scaffolds of the postsynaptic architecture and mutations within the SHANK genes are causally associated with autism spectrum disorders (ASDs). We generated a Shank2-Shank3 double knockout mouse that is showing severe autism related core symptoms, as well as a broad spectrum of comorbidities. We exploited this animal model to identify cortical brain areas linked to specific autistic traits by locally deleting Shank2 and Shank3 simultaneously. Our screening of 10 cortical subregions revealed that a Shank2/3 deletion within the retrosplenial area severely impairs social memory, a core symptom of ASD. Notably, DREADD-mediated neuronal activation could rescue the social impairment triggered by Shank2/3 depletion. Data indicate that the retrosplenial area has to be added to the list of defined brain regions that contribute to the spectrum of behavioural alterations seen in ASDs.
Collapse
Affiliation(s)
- Débora Garrido
- grid.6582.90000 0004 1936 9748Institute of Anatomy and Cell Biology, Ulm University, 89081 Ulm, Germany ,grid.6582.90000 0004 1936 9748International Graduate School, Ulm University, 89081 Ulm, Germany
| | - Stefania Beretta
- grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Ulm site, 89081 Ulm, Germany
| | - Stefanie Grabrucker
- grid.6582.90000 0004 1936 9748Institute of Anatomy and Cell Biology, Ulm University, 89081 Ulm, Germany
| | - Helen Friedericke Bauer
- grid.6582.90000 0004 1936 9748Institute of Anatomy and Cell Biology, Ulm University, 89081 Ulm, Germany ,grid.6582.90000 0004 1936 9748International Graduate School, Ulm University, 89081 Ulm, Germany
| | - David Bayer
- grid.6582.90000 0004 1936 9748International Graduate School, Ulm University, 89081 Ulm, Germany ,grid.6582.90000 0004 1936 9748Department of Neurology, Ulm University, 89081 Ulm, Germany
| | - Carlo Sala
- grid.418879.b0000 0004 1758 9800CNR, Institute for Neuroscience, Milano, Italy
| | - Chiara Verpelli
- grid.418879.b0000 0004 1758 9800CNR, Institute for Neuroscience, Milano, Italy
| | - Francesco Roselli
- grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Ulm site, 89081 Ulm, Germany ,grid.6582.90000 0004 1936 9748Department of Neurology, Ulm University, 89081 Ulm, Germany
| | - Juergen Bockmann
- grid.6582.90000 0004 1936 9748Institute of Anatomy and Cell Biology, Ulm University, 89081 Ulm, Germany
| | - Christian Proepper
- grid.6582.90000 0004 1936 9748Institute of Anatomy and Cell Biology, Ulm University, 89081 Ulm, Germany
| | - Alberto Catanese
- grid.6582.90000 0004 1936 9748Institute of Anatomy and Cell Biology, Ulm University, 89081 Ulm, Germany ,grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Ulm site, 89081 Ulm, Germany
| | - Tobias M. Boeckers
- grid.6582.90000 0004 1936 9748Institute of Anatomy and Cell Biology, Ulm University, 89081 Ulm, Germany ,grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), Ulm site, 89081 Ulm, Germany
| |
Collapse
|
35
|
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: 2.0] [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
|
36
|
Lautz JD, Tsegay KB, Zhu Z, Gniffke EP, Welsh JP, Smith SEP. Synaptic protein interaction networks encode experience by assuming stimulus-specific and brain-region-specific states. Cell Rep 2021; 37:110076. [PMID: 34852231 PMCID: PMC8722361 DOI: 10.1016/j.celrep.2021.110076] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 10/01/2021] [Accepted: 11/09/2021] [Indexed: 11/02/2022] Open
Abstract
A core network of widely expressed proteins within the glutamatergic post-synapse mediates activity-dependent synaptic plasticity throughout the brain, but the specific proteomic composition of synapses differs between brain regions. Here, we address the question, how does proteomic composition affect activity-dependent protein-protein interaction networks (PINs) downstream of synaptic activity? Using quantitative multiplex co-immunoprecipitation, we compare the PIN response of in vivo or ex vivo neurons derived from different brain regions to activation by different agonists or different forms of eyeblink conditioning. We report that PINs discriminate between incoming stimuli using differential kinetics of overlapping and non-overlapping PIN parameters. Further, these "molecular logic rules" differ by brain region. We conclude that although the PIN of the glutamatergic post-synapse is expressed widely throughout the brain, its activity-dependent dynamics show remarkable stimulus-specific and brain-region-specific diversity. This diversity may help explain the challenges in developing molecule-specific drug therapies for neurological disorders.
Collapse
Affiliation(s)
- Jonathan D Lautz
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Kaleb B Tsegay
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Zhiyi Zhu
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Edward P Gniffke
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - John P Welsh
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, USA; Department of Pediatrics, University of Washington, Seattle, WA, USA
| | - Stephen E P Smith
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, USA; Department of Pediatrics, University of Washington, Seattle, WA, USA; Graduate Program in Neuroscience, University of Washington, Seattle, WA, USA.
| |
Collapse
|
37
|
Delling JP, Boeckers TM. Comparison of SHANK3 deficiency in animal models: phenotypes, treatment strategies, and translational implications. J Neurodev Disord 2021; 13:55. [PMID: 34784886 PMCID: PMC8594088 DOI: 10.1186/s11689-021-09397-8] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 09/27/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Autism spectrum disorder (ASD) is a neurodevelopmental condition, which is characterized by clinical heterogeneity and high heritability. Core symptoms of ASD include deficits in social communication and interaction, as well as restricted, repetitive patterns of behavior, interests, or activities. Many genes have been identified that are associated with an increased risk for ASD. Proteins encoded by these ASD risk genes are often involved in processes related to fetal brain development, chromatin modification and regulation of gene expression in general, as well as the structural and functional integrity of synapses. Genes of the SH3 and multiple ankyrin repeat domains (SHANK) family encode crucial scaffolding proteins (SHANK1-3) of excitatory synapses and other macromolecular complexes. SHANK gene mutations are highly associated with ASD and more specifically the Phelan-McDermid syndrome (PMDS), which is caused by heterozygous 22q13.3-deletion resulting in SHANK3-haploinsufficiency, or by SHANK3 missense variants. SHANK3 deficiency and potential treatment options have been extensively studied in animal models, especially in mice, but also in rats and non-human primates. However, few of the proposed therapeutic strategies have translated into clinical practice yet. MAIN TEXT This review summarizes the literature concerning SHANK3-deficient animal models. In particular, the structural, behavioral, and neurological abnormalities are described and compared, providing a broad and comprehensive overview. Additionally, the underlying pathophysiologies and possible treatments that have been investigated in these models are discussed and evaluated with respect to their effect on ASD- or PMDS-associated phenotypes. CONCLUSIONS Animal models of SHANK3 deficiency generated by various genetic strategies, which determine the composition of the residual SHANK3-isoforms and affected cell types, show phenotypes resembling ASD and PMDS. The phenotypic heterogeneity across multiple models and studies resembles the variation of clinical severity in human ASD and PMDS patients. Multiple therapeutic strategies have been proposed and tested in animal models, which might lead to translational implications for human patients with ASD and/or PMDS. Future studies should explore the effects of new therapeutic approaches that target genetic haploinsufficiency, like CRISPR-mediated activation of promotors.
Collapse
Affiliation(s)
- Jan Philipp Delling
- Institute for Anatomy and Cell Biology, Ulm University, Albert-Einstein-Allee 11, Ulm, 89081, Germany.
| | - Tobias M Boeckers
- Institute for Anatomy and Cell Biology, Ulm University, Albert-Einstein-Allee 11, Ulm, 89081, Germany. .,Ulm Site, DZNE, Ulm, Germany.
| |
Collapse
|
38
|
Mirabella F, Desiato G, Mancinelli S, Fossati G, Rasile M, Morini R, Markicevic M, Grimm C, Amegandjin C, Termanini A, Peano C, Kunderfranco P, di Cristo G, Zerbi V, Menna E, Lodato S, Matteoli M, Pozzi D. Prenatal interleukin 6 elevation increases glutamatergic synapse density and disrupts hippocampal connectivity in offspring. Immunity 2021; 54:2611-2631.e8. [PMID: 34758338 PMCID: PMC8585508 DOI: 10.1016/j.immuni.2021.10.006] [Citation(s) in RCA: 76] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 04/24/2021] [Accepted: 10/07/2021] [Indexed: 02/07/2023]
Abstract
Early prenatal inflammatory conditions are thought to be a risk factor for different neurodevelopmental disorders. Maternal interleukin-6 (IL-6) elevation during pregnancy causes abnormal behavior in offspring, but whether these defects result from altered synaptic developmental trajectories remains unclear. Here we showed that transient IL-6 elevation via injection into pregnant mice or developing embryos enhanced glutamatergic synapses and led to overall brain hyperconnectivity in offspring into adulthood. IL-6 activated synaptogenesis gene programs in glutamatergic neurons and required the transcription factor STAT3 and expression of the RGS4 gene. The STAT3-RGS4 pathway was also activated in neonatal brains during poly(I:C)-induced maternal immune activation, which mimics viral infection during pregnancy. These findings indicate that IL-6 elevation at early developmental stages is sufficient to exert a long-lasting effect on glutamatergic synaptogenesis and brain connectivity, providing a mechanistic framework for the association between prenatal inflammatory events and brain neurodevelopmental disorders.
Collapse
Affiliation(s)
- Filippo Mirabella
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20090 Pieve Emanuele, Milan, Italy; IRCCS Humanitas Research Hospital, via Manzoni 56, 20089 Rozzano, Milan, Italy
| | - Genni Desiato
- IRCCS Humanitas Research Hospital, via Manzoni 56, 20089 Rozzano, Milan, Italy; Institute of Neuroscience - National Research Council, 20139 Milan, Italy
| | - Sara Mancinelli
- IRCCS Humanitas Research Hospital, via Manzoni 56, 20089 Rozzano, Milan, Italy
| | - Giuliana Fossati
- IRCCS Humanitas Research Hospital, via Manzoni 56, 20089 Rozzano, Milan, Italy
| | - Marco Rasile
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20090 Pieve Emanuele, Milan, Italy
| | - Raffaella Morini
- IRCCS Humanitas Research Hospital, via Manzoni 56, 20089 Rozzano, Milan, Italy
| | - Marija Markicevic
- Neuroscience Center Zürich, ETH Zürich and University of Zürich, Zürich 8057, Switzerland
| | - Christina Grimm
- Neuroscience Center Zürich, ETH Zürich and University of Zürich, Zürich 8057, Switzerland
| | - Clara Amegandjin
- Department of Neurosciences, Université de Montréal, Montréal, QC, Canada; CHU Sainte-Justine Research Center, Montréal, QC, Canada
| | - Alberto Termanini
- Bioinformatic Unit, Humanitas Clinical and Research Center, 20089 Rozzano, Milan, Italy
| | - Clelia Peano
- Institute of Genetic and Biomedical Research, UoS Milan, National Research Council, 20089 Rozzano, Milan, Italy; Genomic Unit, Humanitas Clinical and Research Center, 20089 Rozzano, Milan, Italy
| | - Paolo Kunderfranco
- Bioinformatic Unit, Humanitas Clinical and Research Center, 20089 Rozzano, Milan, Italy
| | - Graziella di Cristo
- Department of Neurosciences, Université de Montréal, Montréal, QC, Canada; CHU Sainte-Justine Research Center, Montréal, QC, Canada
| | - Valerio Zerbi
- Neuroscience Center Zürich, ETH Zürich and University of Zürich, Zürich 8057, Switzerland; Neural Control of Movement Lab, Department of Health Sciences and Technology, ETH Zürich, Zürich 8057, Switzerland
| | - Elisabetta Menna
- IRCCS Humanitas Research Hospital, via Manzoni 56, 20089 Rozzano, Milan, Italy; Institute of Neuroscience - National Research Council, 20139 Milan, Italy
| | - Simona Lodato
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20090 Pieve Emanuele, Milan, Italy; IRCCS Humanitas Research Hospital, via Manzoni 56, 20089 Rozzano, Milan, Italy
| | - Michela Matteoli
- IRCCS Humanitas Research Hospital, via Manzoni 56, 20089 Rozzano, Milan, Italy; Institute of Neuroscience - National Research Council, 20139 Milan, Italy.
| | - Davide Pozzi
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20090 Pieve Emanuele, Milan, Italy; IRCCS Humanitas Research Hospital, via Manzoni 56, 20089 Rozzano, Milan, Italy.
| |
Collapse
|
39
|
Srivastava S, Condy E, Carmody E, Filip-Dhima R, Kapur K, Bernstein JA, Berry-Kravis E, Powell CM, Soorya L, Thurm A, Buxbaum JD, Sahin M, Kolevzon AL. Parent-reported measure of repetitive behavior in Phelan-McDermid syndrome. J Neurodev Disord 2021; 13:53. [PMID: 34740315 PMCID: PMC8570010 DOI: 10.1186/s11689-021-09398-7] [Citation(s) in RCA: 5] [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: 04/06/2021] [Accepted: 10/14/2021] [Indexed: 12/03/2022] Open
Abstract
Background Phelan McDermid syndrome (PMS) is a neurogenetic condition associated with a high prevalence of intellectual disability (ID) and autism spectrum disorder (ASD). This study provides a more comprehensive and quantitative profile of repetitive behaviors within the context of ID seen with the condition. Methods Individuals age 3–21 years with a confirmed PMS diagnosis participated in a multicenter observational study evaluating the phenotype and natural history of the disorder. We evaluated data collected from this study pertaining to repetitive behaviors from the Repetitive Behavior Scales-Revised (RBS-R). Results There were n = 90 participants who were part of this analysis. Forty-seven percent (n = 42/90) were female, and the average age at baseline evaluation was 8.88 ± 4.72 years. The mean best estimate IQ of the cohort was 26.08 ± 17.67 (range = 3.4–88), with n = 8 with mild ID (or no ID), n = 20 with moderate ID, and n = 62 with severe-profound ID. The RBS-R total overall score was 16.46 ± 13.9 (compared to 33.14 ± 20.60 reported in previous studies of ASD) (Lam and Aman, 2007), and the total number of items endorsed was 10.40 ± 6.81 (range = 0–29). After statistical correction for multiple comparisons, IQ correlated with the RBS-R stereotypic behavior subscale score (rs = − 0.33, unadjusted p = 0.0014, adjusted p = 0.01) and RBS-R stereotypic behavior total number of endorsed items (rs = − 0.32, unadjusted p = 0.0019, adjusted p = 0.01). IQ did not correlate with any other RBS-R subscale scores. Conclusions The RBS-R total overall score in a PMS cohort appears milder compared to individuals with ASD characterized in previous studies. Stereotypic behavior in PMS may reflect cognitive functioning.
Collapse
Affiliation(s)
- Siddharth Srivastava
- Department of Neurology, Rosamund Stone Zander Translational Neuroscience Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA.
| | - Emma Condy
- Neurodevelopmental and Behavioral Phenotyping Service, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Erin Carmody
- Department of Neurology, Rosamund Stone Zander Translational Neuroscience Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Rajna Filip-Dhima
- Department of Neurology, Rosamund Stone Zander Translational Neuroscience Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Kush Kapur
- Department of Neurology, Rosamund Stone Zander Translational Neuroscience Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Jonathan A Bernstein
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Elizabeth Berry-Kravis
- Department of Pediatrics, Rush University Medical Center, Chicago, IL, USA.,Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA.,Department of Biochemistry, Rush University Medical Center, Chicago, IL, USA
| | - Craig M Powell
- Department of Neurobiology, University of Alabama at Birmingham School of Medicine, Birmingham, AL, USA.,Civitan International Research Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Latha Soorya
- Department of Psychiatry, Rush University Medical Center, Chicago, IL, USA
| | - Audrey Thurm
- Neurodevelopmental and Behavioral Phenotyping Service, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Joseph D Buxbaum
- Seaver Autism Center for Research and Treatment, Mount Sinai School of Medicine, New York, NY, USA.,Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Genetics and Genomic Sciences, Mount Sinai School of Medicine, New York, NY, USA.,Department of Neuroscience, Mount Sinai School of Medicine, New York, NY, USA
| | - Mustafa Sahin
- Department of Neurology, Rosamund Stone Zander Translational Neuroscience Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA.,F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - A Lexander Kolevzon
- Seaver Autism Center for Research and Treatment, Mount Sinai School of Medicine, New York, NY, USA.,Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | |
Collapse
|
40
|
de Chaumont F, Lemière N, Coqueran S, Bourgeron T, Ey E. LMT USV Toolbox, a Novel Methodological Approach to Place Mouse Ultrasonic Vocalizations in Their Behavioral Contexts-A Study in Female and Male C57BL/6J Mice and in Shank3 Mutant Females. Front Behav Neurosci 2021; 15:735920. [PMID: 34720899 PMCID: PMC8548730 DOI: 10.3389/fnbeh.2021.735920] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Accepted: 09/20/2021] [Indexed: 11/13/2022] Open
Abstract
Ultrasonic vocalizations (USVs) are used as a phenotypic marker in mouse models of neuropsychiatric disorders. Nevertheless, current methodologies still require time-consuming manual input or sound recordings clean of any background noise. We developed a method to overcome these two restraints to boost knowledge on mouse USVs. The methods are freely available and the USV analysis runs online at https://usv.pasteur.cloud. As little is currently known about usage and structure of ultrasonic vocalizations during social interactions over the long-term and in unconstrained context, we investigated mouse spontaneous communication by coupling the analysis of USVs with automatic labeling of behaviors. We continuously recorded during 3 days undisturbed interactions of same-sex pairs of C57BL/6J sexually naive males and females at 5 weeks and 3 and 7 months of age. In same-sex interactions, we observed robust differences between males and females in the amount of USVs produced, in the acoustic structure and in the contexts of emission. The context-specific acoustic variations emerged with increasing age. The emission of USVs also reflected a high level of excitement during social interactions. We finally highlighted the importance of studying long-term spontaneous communication by investigating female mice lacking Shank3, a synaptic protein associated with autism. While the previous short-time constrained investigations could not detect USV emission abnormalities, our analysis revealed robust differences in the usage and structure of the USVs emitted by mutant mice compared to wild-type female pairs.
Collapse
Affiliation(s)
- Fabrice de Chaumont
- Human Genetics and Cognitive Functions, Institut Pasteur, UMR 3571 CNRS, Université de Paris, Paris, France
| | - Nathalie Lemière
- Human Genetics and Cognitive Functions, Institut Pasteur, UMR 3571 CNRS, Université de Paris, Paris, France
| | - Sabrina Coqueran
- Human Genetics and Cognitive Functions, Institut Pasteur, UMR 3571 CNRS, Université de Paris, Paris, France
| | - Thomas Bourgeron
- Human Genetics and Cognitive Functions, Institut Pasteur, UMR 3571 CNRS, Université de Paris, Paris, France
| | - Elodie Ey
- Human Genetics and Cognitive Functions, Institut Pasteur, UMR 3571 CNRS, Université de Paris, Paris, France
| |
Collapse
|
41
|
Wan L, Ai JQ, Yang C, Jiang J, Zhang QL, Luo ZH, Huang RJ, Tu T, Pan A, Tu E, Manavis J, Xiao B, Yan XX. Expression of the Excitatory Postsynaptic Scaffolding Protein, Shank3, in Human Brain: Effect of Age and Alzheimer's Disease. Front Aging Neurosci 2021; 13:717263. [PMID: 34504419 PMCID: PMC8421777 DOI: 10.3389/fnagi.2021.717263] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Accepted: 07/26/2021] [Indexed: 12/14/2022] Open
Abstract
Shank3 is a postsynaptic scaffolding protein of excitatory synapses. Mutations or variations of SHANK3 are associated with various psychiatric and neurological disorders. We set to determine its normal expression pattern in the human brain, and its change, if any, with age and Alzheimer’s disease (AD)-type β-amyloid (Aβ) and Tau pathogenesis. In general, Shank3 immunoreactivity (IR) exhibited largely a neuropil pattern with differential laminar/regional distribution across brain regions. In youth and adults, subsets of pyramidal/multipolar neurons in the cerebrum, striatum, and thalamus showed moderate IR, while some large-sized neurons in the brainstem and the granule cells in the cerebellar cortex exhibited light IR. In double immunofluorescence, Shank3 IR occurred at the sublemmal regions in neuronal somata and large dendrites, apposing to synaptophysin-labeled presynaptic terminals. In aged cases, immunolabeled neuronal somata were reduced, with disrupted neuropil labeling seen in the molecular layer of the dentate gyrus in AD cases. In immunoblot, levels of Shank3 protein were positively correlated with that of the postsynaptic density protein 95 (PSD95) among different brain regions. Levels of Shank3, PSD95, and synaptophysin immunoblotted in the prefrontal, precentral, and cerebellar cortical lysates were reduced in the aged and AD relative to youth and adult groups. Taken together, the differential Shank3 expression among brain structures/regions indicates the varied local density of the excitatory synapses. The enriched Shank3 expression in the forebrain subregions appears inconsistent with a role of this protein in the modulation of high cognitive functions. The decline of its expression in aged and AD brains may relate to the degeneration of excitatory synapses.
Collapse
Affiliation(s)
- Lily Wan
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Jia-Qi Ai
- Department of Anatomy and Neurobiology, Central South University Xiangya School of Medicine, Changsha, China
| | - Chen Yang
- Department of Anatomy and Neurobiology, Central South University Xiangya School of Medicine, Changsha, China
| | - Juan Jiang
- Department of Anatomy and Neurobiology, Central South University Xiangya School of Medicine, Changsha, China
| | - Qi-Lei Zhang
- Department of Anatomy and Neurobiology, Central South University Xiangya School of Medicine, Changsha, China
| | - Zhao-Hui Luo
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Rou-Jie Huang
- Medical Doctor Program, Xiangya School of Medicine, Central South University, Changsha, China
| | - Tian Tu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Aihua Pan
- Department of Anatomy and Neurobiology, Central South University Xiangya School of Medicine, Changsha, China
| | - Ewen Tu
- Department of Neurology, Brain Hospital of Hunan Province, Changsha, China
| | - Jim Manavis
- Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, SA, Australia
| | - Bo Xiao
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Xiao-Xin Yan
- Department of Anatomy and Neurobiology, Central South University Xiangya School of Medicine, Changsha, China
| |
Collapse
|
42
|
Lutz AK, Pfaender S, Incearap B, Ioannidis V, Ottonelli I, Föhr KJ, Cammerer J, Zoller M, Higelin J, Giona F, Stetter M, Stoecker N, Alami NO, Schön M, Orth M, Liebau S, Barbi G, Grabrucker AM, Delorme R, Fauler M, Mayer B, Jesse S, Roselli F, Ludolph AC, Bourgeron T, Verpelli C, Demestre M, Boeckers TM. Autism-associated SHANK3 mutations impair maturation of neuromuscular junctions and striated muscles. Sci Transl Med 2021; 12:12/547/eaaz3267. [PMID: 32522805 DOI: 10.1126/scitranslmed.aaz3267] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 12/09/2019] [Accepted: 04/07/2020] [Indexed: 12/12/2022]
Abstract
Heterozygous mutations of the gene encoding the postsynaptic protein SHANK3 are associated with syndromic forms of autism spectrum disorders (ASDs). One of the earliest clinical symptoms in SHANK3-associated ASD is neonatal skeletal muscle hypotonia. This symptom can be critical for the early diagnosis of affected children; however, the mechanism mediating hypotonia in ASD is not completely understood. Here, we used a combination of patient-derived human induced pluripotent stem cells (hiPSCs), Shank3Δ11(-/-) mice, and Phelan-McDermid syndrome (PMDS) muscle biopsies from patients of different ages to analyze the role of SHANK3 on motor unit development. Our results suggest that the hypotonia in SHANK3 deficiency might be caused by dysfunctions in all elements of the voluntary motor system: motoneurons, neuromuscular junctions (NMJs), and striated muscles. We found that SHANK3 localizes in Z-discs in the skeletal muscle sarcomere and co-immunoprecipitates with α-ACTININ. SHANK3 deficiency lead to shortened Z-discs and severe impairment of acetylcholine receptor clustering in hiPSC-derived myotubes and in muscle from Shank3Δ11(-/-) mice and patients with PMDS, indicating a crucial role for SHANK3 in the maturation of NMJs and striated muscle. Functional motor defects in Shank3Δ11(-/-) mice could be rescued with the troponin activator Tirasemtiv that sensitizes muscle fibers to calcium. Our observations give insight into the function of SHANK3 besides the central nervous system and imply potential treatment strategies for SHANK3-associated ASD.
Collapse
Affiliation(s)
- Anne-Kathrin Lutz
- Institute for Anatomy and Cell Biology, Ulm University, 89081 Ulm, Germany
| | - Stefanie Pfaender
- Institute for Anatomy and Cell Biology, Ulm University, 89081 Ulm, Germany
| | - Berra Incearap
- Institute for Anatomy and Cell Biology, Ulm University, 89081 Ulm, Germany
| | - Valentin Ioannidis
- Institute for Anatomy and Cell Biology, Ulm University, 89081 Ulm, Germany
| | - Ilaria Ottonelli
- Institute for Anatomy and Cell Biology, Ulm University, 89081 Ulm, Germany
| | - Karl J Föhr
- Department of Anesthesiology, Ulm University Hospital, 89081 Ulm, Germany
| | - Judith Cammerer
- Institute for Anatomy and Cell Biology, Ulm University, 89081 Ulm, Germany
| | - Marvin Zoller
- Institute for Anatomy and Cell Biology, Ulm University, 89081 Ulm, Germany
| | - Julia Higelin
- Institute for Anatomy and Cell Biology, Ulm University, 89081 Ulm, Germany
| | - Federica Giona
- CNR Neuroscience Institute, University of Milan, 20129 Milan, Italy.,BIOMETRA University of Milan, 20129 Milan, Italy
| | - Maximilian Stetter
- Institute for Anatomy and Cell Biology, Ulm University, 89081 Ulm, Germany
| | - Nicole Stoecker
- Institute for Anatomy and Cell Biology, Ulm University, 89081 Ulm, Germany
| | | | - Michael Schön
- Institute for Anatomy and Cell Biology, Ulm University, 89081 Ulm, Germany
| | | | - Stefan Liebau
- Institute of Neuroanatomy and Developmental Biology, Eberhard Karls University Tübingen, 72074 Tübingen, Germany
| | - Gotthold Barbi
- Institute for Human Genetics, Ulm University Hospital, 89081 Ulm, Germany
| | - Andreas M Grabrucker
- Cellular Neurobiology and Neuro-Nanotechnology Lab, Department of Biological Sciences, University of Limerick, V94PH61 Limerick, Ireland.,Bernal Institute, University of Limerick, V94T9PX Limerick, Ireland.,Health Research Institute (HRI), University of Limerick, V94T9PX Limerick, Ireland
| | - Richard Delorme
- Child and Adolescent Psychiatry Department, APHP, Robert-Debré Hospital, 750197 Paris, France
| | - Michael Fauler
- Institute of General Physiology, Ulm University, 89081 Ulm, Germany
| | - Benjamin Mayer
- Institute of Epidemiology and Medical Biometry, Ulm University, 89075 Ulm, Germany
| | | | | | | | - Thomas Bourgeron
- Génétique Humaine et Fonctions Cognitives, Université Paris Diderot, Institut Pasteur, 75015 Paris, France
| | - Chiara Verpelli
- CNR Neuroscience Institute, University of Milan, 20129 Milan, Italy.,BIOMETRA University of Milan, 20129 Milan, Italy
| | - Maria Demestre
- Institute for Anatomy and Cell Biology, Ulm University, 89081 Ulm, Germany.
| | - Tobias M Boeckers
- Institute for Anatomy and Cell Biology, Ulm University, 89081 Ulm, Germany. .,DZNE, Ulm Site, 89081 Ulm, Germany
| |
Collapse
|
43
|
Inhibitory regulation of calcium transients in prefrontal dendritic spines is compromised by a nonsense Shank3 mutation. Mol Psychiatry 2021; 26:1945-1966. [PMID: 32161363 PMCID: PMC7483244 DOI: 10.1038/s41380-020-0708-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 02/23/2020] [Accepted: 02/28/2020] [Indexed: 01/11/2023]
Abstract
The SHANK3 gene encodes a postsynaptic scaffold protein in excitatory synapses, and its disruption is implicated in neurodevelopmental disorders such as Phelan-McDermid syndrome, autism spectrum disorder, and schizophrenia. Most studies of SHANK3 in the neocortex and hippocampus have focused on disturbances in pyramidal neurons. However, GABAergic interneurons likewise receive excitatory inputs and presumably would also be a target of constitutive SHANK3 perturbations. In this study, we characterize the prefrontal cortical microcircuit in awake mice using subcellular-resolution two-photon microscopy. We focused on a nonsense R1117X mutation, which leads to truncated SHANK3 and has been linked previously to cortical dysfunction. We find that R1117X mutants have abnormally elevated calcium transients in apical dendritic spines. The synaptic calcium dysregulation is due to a loss of dendritic inhibition via decreased NMDAR currents and reduced firing of dendrite-targeting somatostatin-expressing (SST) GABAergic interneurons. Notably, upregulation of the NMDAR subunit GluN2B in SST interneurons corrects the excessive synaptic calcium signals and ameliorates learning deficits in R1117X mutants. These findings reveal dendrite-targeting interneurons, and more broadly the inhibitory control of dendritic spines, as a key microcircuit mechanism compromised by the SHANK3 dysfunction.
Collapse
|
44
|
Bucher M, Niebling S, Han Y, Molodenskiy D, Hassani Nia F, Kreienkamp HJ, Svergun D, Kim E, Kostyukova AS, Kreutz MR, Mikhaylova M. Autism-associated SHANK3 missense point mutations impact conformational fluctuations and protein turnover at synapses. eLife 2021; 10:66165. [PMID: 33945465 PMCID: PMC8169116 DOI: 10.7554/elife.66165] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Accepted: 05/01/2021] [Indexed: 12/18/2022] Open
Abstract
Members of the SH3- and ankyrin repeat (SHANK) protein family are considered as master scaffolds of the postsynaptic density of glutamatergic synapses. Several missense mutations within the canonical SHANK3 isoform have been proposed as causative for the development of autism spectrum disorders (ASDs). However, there is a surprising paucity of data linking missense mutation-induced changes in protein structure and dynamics to the occurrence of ASD-related synaptic phenotypes. In this proof-of-principle study, we focus on two ASD-associated point mutations, both located within the same domain of SHANK3 and demonstrate that both mutant proteins indeed show distinct changes in secondary and tertiary structure as well as higher conformational fluctuations. Local and distal structural disturbances result in altered synaptic targeting and changes of protein turnover at synaptic sites in rat primary hippocampal neurons.
Collapse
Affiliation(s)
- Michael Bucher
- AG Optobiology, Institute of Biology, Humboldt-University, Berlin, Germany.,DFG Emmy Noether Guest Group 'Neuronal Protein Transport', Institute for Molecular Neurogenetics, Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany.,RG Neuroplasticity, Leibniz-Institute for Neurobiology (LIN), Magdeburg, Germany
| | - Stephan Niebling
- Molecular Biophysics and High-Throughput Crystallization, European Molecular Biology Laboratory (EMBL), Hamburg, Germany
| | - Yuhao Han
- DFG Emmy Noether Guest Group 'Neuronal Protein Transport', Institute for Molecular Neurogenetics, Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany.,Structural Cell Biology of Viruses, Centre for Structural Systems Biology (CSSB) and Leibniz Institute for Experimental Virology, Hamburg, Germany
| | - Dmitry Molodenskiy
- European Molecular Biology Laboratory (EMBL) Hamburg Unit, DESY, Hamburg, Germany
| | - Fatemeh Hassani Nia
- Institute of Human Genetics, Center for Obstetrics and Pediatrics, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Hans-Jürgen Kreienkamp
- Institute of Human Genetics, Center for Obstetrics and Pediatrics, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Dmitri Svergun
- European Molecular Biology Laboratory (EMBL) Hamburg Unit, DESY, Hamburg, Germany
| | - Eunjoon Kim
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science (IBS) and Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Alla S Kostyukova
- DFG Emmy Noether Guest Group 'Neuronal Protein Transport', Institute for Molecular Neurogenetics, Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany.,The Gene and Linda Voiland School of Chemical Engineering and Bioengineering, Washington State University (WSU), Pullman, United States
| | - Michael R Kreutz
- RG Neuroplasticity, Leibniz-Institute for Neurobiology (LIN), Magdeburg, Germany.,Leibniz Group 'Dendritic Organelles and Synaptic Function', Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany.,German Center for Neurodegenerative Diseases, Magdeburg, Germany.,Center for Behavioral Brain Sciences, Magdeburg, Germany
| | - Marina Mikhaylova
- AG Optobiology, Institute of Biology, Humboldt-University, Berlin, Germany.,DFG Emmy Noether Guest Group 'Neuronal Protein Transport', Institute for Molecular Neurogenetics, Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| |
Collapse
|
45
|
Qian KY, Zeng WX, Hao Y, Zeng XT, Liu H, Li L, Chen L, Tian FM, Chang C, Hall Q, Song CX, Gao S, Hu Z, Kaplan JM, Li Q, Tong XJ. Male pheromones modulate synaptic transmission at the C. elegans neuromuscular junction in a sexually dimorphic manner. eLife 2021; 10:e67170. [PMID: 33787493 PMCID: PMC8051947 DOI: 10.7554/elife.67170] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 03/30/2021] [Indexed: 12/24/2022] Open
Abstract
The development of functional synapses in the nervous system is important for animal physiology and behaviors, and its disturbance has been linked with many neurodevelopmental disorders. The synaptic transmission efficacy can be modulated by the environment to accommodate external changes, which is crucial for animal reproduction and survival. However, the underlying plasticity of synaptic transmission remains poorly understood. Here we show that in Caenorhabditis elegans, the male environment increases the hermaphrodite cholinergic transmission at the neuromuscular junction (NMJ), which alters hermaphrodites' locomotion velocity and mating efficiency. We identify that the male-specific pheromones mediate this synaptic transmission modulation effect in a developmental stage-dependent manner. Dissection of the sensory circuits reveals that the AWB chemosensory neurons sense those male pheromones and further transduce the information to NMJ using cGMP signaling. Exposure of hermaphrodites to the male pheromones specifically increases the accumulation of presynaptic CaV2 calcium channels and clustering of postsynaptic acetylcholine receptors at cholinergic synapses of NMJ, which potentiates cholinergic synaptic transmission. Thus, our study demonstrates a circuit mechanism for synaptic modulation and behavioral flexibility by sexual dimorphic pheromones.
Collapse
Affiliation(s)
- Kang-Ying Qian
- School of Life Science and Technology, ShanghaiTech UniversityShanghaiChina
- University of Chinese Academy of SciencesBeijingChina
- Institute of Neuroscience, Shanghai Institutes for Biological Sciences, Chinese Academy of SciencesShanghaiChina
| | - Wan-Xin Zeng
- School of Life Science and Technology, ShanghaiTech UniversityShanghaiChina
- University of Chinese Academy of SciencesBeijingChina
- Institute of Neuroscience, Shanghai Institutes for Biological Sciences, Chinese Academy of SciencesShanghaiChina
| | - Yue Hao
- School of Life Science and Technology, ShanghaiTech UniversityShanghaiChina
- University of Chinese Academy of SciencesBeijingChina
- Institute of Neuroscience, Shanghai Institutes for Biological Sciences, Chinese Academy of SciencesShanghaiChina
| | - Xian-Ting Zeng
- School of Life Science and Technology, ShanghaiTech UniversityShanghaiChina
| | - Haowen Liu
- Queensland Brain Institute, Clem Jones Centre for Ageing Dementia Research (CJCADR), The University of QueenslandBrisbaneAustralia
| | - Lei Li
- Queensland Brain Institute, Clem Jones Centre for Ageing Dementia Research (CJCADR), The University of QueenslandBrisbaneAustralia
| | - Lili Chen
- College of Life Science and Technology, Huazhong University of Science and TechnologyWuhanChina
| | - Fu-min Tian
- School of Life Science and Technology, ShanghaiTech UniversityShanghaiChina
- University of Chinese Academy of SciencesBeijingChina
| | - Cindy Chang
- Department of Molecular Biology, Massachusetts General HospitalBostonUnited States
- Department of Neurobiology, Harvard Medical SchoolBostonUnited States
| | - Qi Hall
- Department of Molecular Biology, Massachusetts General HospitalBostonUnited States
- Department of Neurobiology, Harvard Medical SchoolBostonUnited States
| | - Chun-Xue Song
- Center for Brain Science, Shanghai Children's Medical CenterShanghaiChina
- Department of Anatomy and Physiology, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Shangbang Gao
- College of Life Science and Technology, Huazhong University of Science and TechnologyWuhanChina
| | - Zhitao Hu
- Queensland Brain Institute, Clem Jones Centre for Ageing Dementia Research (CJCADR), The University of QueenslandBrisbaneAustralia
| | - Joshua M Kaplan
- Department of Molecular Biology, Massachusetts General HospitalBostonUnited States
- Department of Neurobiology, Harvard Medical SchoolBostonUnited States
| | - Qian Li
- Center for Brain Science, Shanghai Children's Medical CenterShanghaiChina
- Department of Anatomy and Physiology, Shanghai Jiao Tong University School of MedicineShanghaiChina
- Shanghai Research Center for Brain Science and Brain-Inspired IntelligenceShanghaiChina
| | - Xia-Jing Tong
- School of Life Science and Technology, ShanghaiTech UniversityShanghaiChina
| |
Collapse
|
46
|
Abstract
Social cooperation in rodents was recently validated in rats, and we recently successfully applied a modified automated analysis to mice. Here, we describe a detailed procedure for using this paradigm in mice that relies on reward-based mutual communication that is automatically detected by a software algorithm embedded in the custom-made equipment. We also describe exemplary results of analyses in mice as a guide to broader neuroscience research applications employing transgenic knockout mice modeling neuropsychiatric disorders and mice of various ages. For complete details on the use and execution of this protocol, please refer to Han et al. (2020). Detailed protocol for performing customized social cooperation tests in mice Detailed guidelines for analyzing social cooperation in mice Troubleshooting of the most common pitfalls associated with the procedure
Collapse
Affiliation(s)
- Jungsu Shin
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Korea
| | - Jaewon Ko
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Korea
| |
Collapse
|
47
|
Neklyudova AK, Portnova GV, Rebreikina AB, Voinova VY, Vorsanova SG, Iourov IY, Sysoeva OV. 40-Hz Auditory Steady-State Response (ASSR) as a Biomarker of Genetic Defects in the SHANK3 Gene: A Case Report of 15-Year-Old Girl with a Rare Partial SHANK3 Duplication. Int J Mol Sci 2021; 22:1898. [PMID: 33673024 PMCID: PMC7917917 DOI: 10.3390/ijms22041898] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 01/26/2021] [Accepted: 02/09/2021] [Indexed: 12/02/2022] Open
Abstract
SHANK3 encodes a scaffold protein involved in postsynaptic receptor density in glutamatergic synapses, including those in the parvalbumin (PV)+ inhibitory neurons-the key players in the generation of sensory gamma oscillations, such as 40-Hz auditory steady-state response (ASSR). However, 40-Hz ASSR was not studied in relation to SHANK3 functioning. Here, we present a 15-year-old girl (SH01) with previously unreported duplication of the first seven exons of the SHANK3 gene (22q13.33). SH01's electroencephalogram (EEG) during 40-Hz click trains of 500 ms duration binaurally presented with inter-trial intervals of 500-800 ms were compared with those from typically developing children (n = 32). SH01 was diagnosed with mild mental retardation and learning disabilities (F70.88), dysgraphia, dyslexia, and smaller vocabulary than typically developing (TD) peers. Her clinical phenotype resembled the phenotype of previously described patients with 22q13.33 microduplications (≈30 reported so far). SH01 had mild autistic symptoms but below the threshold for ASD diagnosis and microcephaly. No seizures or MRI abnormalities were reported. While SH01 had relatively preserved auditory event-related potential (ERP) with slightly attenuated P1, her 40-Hz ASSR was totally absent significantly deviating from TD's ASSR. The absence of 40-Hz ASSR in patients with microduplication, which affected the SHANK3 gene, indicates deficient temporal resolution of the auditory system, which might underlie language problems and represent a neurophysiological biomarker of SHANK3 abnormalities.
Collapse
Affiliation(s)
- Anastasia K. Neklyudova
- Laboratory of Human Higher Nervous Activity, Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Science, 117485 Moscow, Russia; (A.K.N.); (G.V.P.); (A.B.R.)
| | - Galina V. Portnova
- Laboratory of Human Higher Nervous Activity, Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Science, 117485 Moscow, Russia; (A.K.N.); (G.V.P.); (A.B.R.)
| | - Anna B. Rebreikina
- Laboratory of Human Higher Nervous Activity, Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Science, 117485 Moscow, Russia; (A.K.N.); (G.V.P.); (A.B.R.)
| | - Victoria Yu Voinova
- Veltischev Research and Clinical Institute for Pediatrics of the Pirogov, Russian National Research Medical University, Ministry of Health of Russian Federation, 125412 Moscow, Russia; (V.Y.V.); (S.G.V.); (I.Y.I.)
- Mental Health Research Center, 117152 Moscow, Russia
| | - Svetlana G. Vorsanova
- Veltischev Research and Clinical Institute for Pediatrics of the Pirogov, Russian National Research Medical University, Ministry of Health of Russian Federation, 125412 Moscow, Russia; (V.Y.V.); (S.G.V.); (I.Y.I.)
- Mental Health Research Center, 117152 Moscow, Russia
| | - Ivan Y. Iourov
- Veltischev Research and Clinical Institute for Pediatrics of the Pirogov, Russian National Research Medical University, Ministry of Health of Russian Federation, 125412 Moscow, Russia; (V.Y.V.); (S.G.V.); (I.Y.I.)
- Mental Health Research Center, 117152 Moscow, Russia
| | - Olga V. Sysoeva
- Laboratory of Human Higher Nervous Activity, Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Science, 117485 Moscow, Russia; (A.K.N.); (G.V.P.); (A.B.R.)
| |
Collapse
|
48
|
Jin C, Kang H, Yoo T, Ryu JR, Yoo YE, Ma R, Zhang Y, Kang HR, Kim Y, Seong H, Bang G, Park S, Kwon SK, Sun W, Kim H, Kim JY, Kim E, Han K. The Neomycin Resistance Cassette in the Targeted Allele of Shank3B Knock-Out Mice Has Potential Off-Target Effects to Produce an Unusual Shank3 Isoform. Front Mol Neurosci 2021; 13:614435. [PMID: 33505245 PMCID: PMC7831789 DOI: 10.3389/fnmol.2020.614435] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 12/02/2020] [Indexed: 01/20/2023] Open
Abstract
Variants of the SH3 and multiple ankyrin repeat domains 3 (SHANK3), which encodes postsynaptic scaffolds, are associated with brain disorders. The targeted alleles in a few Shank3 knock-out (KO) lines contain a neomycin resistance (Neo) cassette, which may perturb the normal expression of neighboring genes; however, this has not been investigated in detail. We previously reported an unexpected increase in the mRNA expression of Shank3 exons 1–12 in the brains of Shank3B KO mice generated by replacing Shank3 exons 13–16 with the Neo cassette. In this study, we confirmed that the increased Shank3 mRNA in Shank3B KO brains produced an unusual ∼60 kDa Shank3 isoform (Shank3-N), which did not properly localize to the synaptic compartment. Functionally, Shank3-N overexpression altered the dendritic spine morphology in cultured neurons. Importantly, Shank3-N expression in Shank3B KO mice was not a compensatory response to a reduction of full-length Shank3 because expression was still detected in the brain after normalizing the level of full-length Shank3. Moreover, in another Shank3 KO line (Shank3 gKO) with a similar Shank3 exonal deletion as that in Shank3B KO mice but without a Neo cassette, the mRNA expression levels of Shank3 exons 1–12 were lower than those of wild-type mice and Shank3-N was not detected in the brain. In addition, the expression levels of genes neighboring Shank3 on chromosome 15 were altered in the striatum of Shank3B KO but not Shank3 gKO mice. These results suggest that the Neo cassette has potential off-target effects in Shank3B KO mice.
Collapse
Affiliation(s)
- Chunmei Jin
- Department of Neuroscience, College of Medicine, Korea University, Seoul, South Korea.,Department of Biomedical Sciences, College of Medicine, Korea University, Seoul, South Korea
| | - Hyojin Kang
- Division of National Supercomputing, Korea Institute of Science and Technology Information, Daejeon, South Korea
| | - Taesun Yoo
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science, Daejeon, South Korea
| | - Jae Ryun Ryu
- Department of Anatomy, College of Medicine, Korea University, Seoul, South Korea
| | - Ye-Eun Yoo
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, South Korea
| | - Ruiying Ma
- Department of Neuroscience, College of Medicine, Korea University, Seoul, South Korea.,Department of Biomedical Sciences, College of Medicine, Korea University, Seoul, South Korea
| | - Yinhua Zhang
- Department of Neuroscience, College of Medicine, Korea University, Seoul, South Korea.,Department of Biomedical Sciences, College of Medicine, Korea University, Seoul, South Korea
| | - Hyae Rim Kang
- Department of Neuroscience, College of Medicine, Korea University, Seoul, South Korea.,Department of Biomedical Sciences, College of Medicine, Korea University, Seoul, South Korea
| | - Yoonhee Kim
- Department of Neuroscience, College of Medicine, Korea University, Seoul, South Korea
| | - Hyunyoung Seong
- Department of Neuroscience, College of Medicine, Korea University, Seoul, South Korea
| | - Geul Bang
- Research Center for Bioconvergence Analysis, Korea Basic Science Institute, Ochang, South Korea.,College of Pharmacy, Korea University, Sejong, South Korea
| | - Sangwoo Park
- Research Center for Bioconvergence Analysis, Korea Basic Science Institute, Ochang, South Korea
| | - Seok-Kyu Kwon
- Center for Functional Connectomics, Korea Institute of Science and Technology, Brain Science Institute, Seoul, South Korea
| | - Woong Sun
- Department of Biomedical Sciences, College of Medicine, Korea University, Seoul, South Korea.,Department of Anatomy, College of Medicine, Korea University, Seoul, South Korea
| | - Hyunkyung Kim
- Department of Biomedical Sciences, College of Medicine, Korea University, Seoul, South Korea.,Department of Biochemistry and Molecular Biology, College of Medicine, Korea University, Seoul, South Korea
| | - Jin Young Kim
- Research Center for Bioconvergence Analysis, Korea Basic Science Institute, Ochang, South Korea
| | - Eunjoon Kim
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science, Daejeon, South Korea.,Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, South Korea
| | - Kihoon Han
- Department of Neuroscience, College of Medicine, Korea University, Seoul, South Korea.,Department of Biomedical Sciences, College of Medicine, Korea University, Seoul, South Korea
| |
Collapse
|
49
|
Chiola S, Napan KL, Wang Y, Lazarenko RM, Armstrong CJ, Cui J, Shcheglovitov A. Defective AMPA-mediated synaptic transmission and morphology in human neurons with hemizygous SHANK3 deletion engrafted in mouse prefrontal cortex. Mol Psychiatry 2021; 26:4670-4686. [PMID: 33558651 PMCID: PMC8349370 DOI: 10.1038/s41380-021-01023-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 12/18/2020] [Accepted: 01/08/2021] [Indexed: 01/12/2023]
Abstract
Genetic abnormalities in synaptic proteins are common in individuals with autism; however, our understanding of the cellular and molecular mechanisms disrupted by these abnormalities is limited. SHANK3 is a postsynaptic scaffolding protein of excitatory synapses that has been found mutated or deleted in most patients with 22q13 deletion syndrome and about 2% of individuals with idiopathic autism and intellectual disability. Here, we generated CRISPR/Cas9-engineered human pluripotent stem cells (PSCs) with complete hemizygous SHANK3 deletion (SHANK3+/-), which is the most common genetic abnormality in patients, and investigated the synaptic and morphological properties of SHANK3-deficient PSC-derived cortical neurons engrafted in the mouse prefrontal cortex. We show that human PSC-derived neurons integrate into the mouse cortex by acquiring appropriate cortical layer identities and by receiving and sending anatomical projections from/to multiple different brain regions. We also demonstrate that SHANK3-deficient human neurons have reduced AMPA-, but not NMDA- or GABA-mediated synaptic transmission and exhibit impaired dendritic arbors and spines, as compared to isogenic control neurons co-engrafted in the same brain region. Together, this study reveals specific synaptic and morphological deficits caused by SHANK3 hemizygosity in human cortical neurons at different developmental stages under physiological conditions and validates the use of co-engrafted control and mutant human neurons as a new platform for studying connectivity deficits in genetic neurodevelopmental disorders associated with autism.
Collapse
Affiliation(s)
- Simone Chiola
- grid.223827.e0000 0001 2193 0096Departments of Neurobiology, University of Utah, Salt Lake City, UT USA
| | - Kandy L. Napan
- grid.223827.e0000 0001 2193 0096Departments of Neurobiology, University of Utah, Salt Lake City, UT USA
| | - Yueqi Wang
- grid.223827.e0000 0001 2193 0096Departments of Neurobiology, University of Utah, Salt Lake City, UT USA ,grid.223827.e0000 0001 2193 0096Neuroscience Graduate Program, University of Utah, Salt Lake City, UT USA
| | - Roman M. Lazarenko
- grid.152326.10000 0001 2264 7217Departments of Pharmacology, Vanderbilt University, Nashville, TN USA
| | - Celeste J. Armstrong
- grid.223827.e0000 0001 2193 0096Departments of Neurobiology, University of Utah, Salt Lake City, UT USA
| | - Jun Cui
- grid.41891.350000 0001 2156 6108Department of Cell Biology and Neurosciences, Montana State University, Bozeman, MT USA
| | - Aleksandr Shcheglovitov
- Departments of Neurobiology, University of Utah, Salt Lake City, UT, USA. .,Neuroscience Graduate Program, University of Utah, Salt Lake City, UT, USA. .,Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, USA. .,Department of Adult Psychiatry, University of Utah, Salt Lake City, UT, USA.
| |
Collapse
|
50
|
Developmental impaired Akt signaling in the Shank1 and Shank3 double knock-out mice. Mol Psychiatry 2021; 26:1928-1944. [PMID: 33402706 PMCID: PMC8440179 DOI: 10.1038/s41380-020-00979-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 11/23/2020] [Accepted: 12/01/2020] [Indexed: 01/30/2023]
Abstract
Human mutations and haploinsufficiency of the SHANK family genes are associated with autism spectrum disorders (ASD) and intellectual disability (ID). Complex phenotypes have been also described in all mouse models of Shank mutations and deletions, consistent with the heterogeneity of the human phenotypes. However, the specific role of Shank proteins in synapse and neuronal functions remain to be elucidated. Here, we generated a new mouse model to investigate how simultaneously deletion of Shank1 and Shank3 affects brain development and behavior in mice. Shank1-Shank3 DKO mice showed a low survival rate, a developmental strong reduction in the activation of intracellular signaling pathways involving Akt, S6, ERK1/2, and eEF2 during development and a severe behavioral impairments. Our study suggests that Shank1 and Shank3 proteins are essential to developmentally regulate the activation of Akt and correlated intracellular pathways crucial for mammalian postnatal brain development and synaptic plasticity. Therefore, Akt function might represent a new therapeutic target for enhancing cognitive abilities of syndromic ASD patients.
Collapse
|