1
|
Brill J, Clarke B, Hong I, Huganir RL. Dissociation of SYNGAP1 enzymatic and structural roles: Intrinsic excitability and seizure susceptibility. Proc Natl Acad Sci U S A 2025; 122:e2427288122. [PMID: 40294267 PMCID: PMC12067237 DOI: 10.1073/pnas.2427288122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Accepted: 03/12/2025] [Indexed: 04/30/2025] Open
Abstract
SYNGAP1 is a key Ras-GAP protein enriched at excitatory synapses, with mutations causing intellectual disability and epilepsy in humans. Recent studies have revealed that in addition to its role as a negative regulator of G-protein signaling through its GAP enzymatic activity, SYNGAP1 plays an important structural role through its interaction with postsynaptic density proteins. Here, we reveal that intrinsic excitability deficits and seizure phenotypes in heterozygous Syngap1 knockout (KO) mice are differentially dependent on Syngap1 GAP activity. Cortical excitatory neurons in heterozygous KO mice displayed reduced intrinsic excitability, including lower input resistance, and increased rheobase, a phenotype recapitulated in GAP-deficient Syngap1 mutants. However, seizure severity and susceptibility to pentylenetetrazol (PTZ)-induced seizures were significantly elevated in heterozygous KO mice but unaffected in GAP-deficient mutants, implicating the structural rather than enzymatic role of Syngap1 in seizure regulation. These findings highlight the complex interplay between SYNGAP1 structural and catalytic functions in neuronal physiology and disease.
Collapse
Affiliation(s)
- Julia Brill
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD21205
- Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, MD21205
| | - Blaise Clarke
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD21205
- Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, MD21205
| | - Ingie Hong
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD21205
- Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, MD21205
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD21205
| | - Richard L. Huganir
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD21205
- Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, MD21205
| |
Collapse
|
2
|
Singh A, Rizzi M, Seo SS, Osterweil EK. Syngap+/- CA1 Pyramidal Neurons Exhibit Upregulated Translation of Long MRNAs Associated with LTP. eNeuro 2025; 12:ENEURO.0086-25.2025. [PMID: 40295099 PMCID: PMC12091090 DOI: 10.1523/eneuro.0086-25.2025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Revised: 04/01/2025] [Accepted: 04/09/2025] [Indexed: 04/30/2025] Open
Abstract
In the Syngap+/- model of SYNGAP1-related intellectual disability (SRID), excessive neuronal protein synthesis is linked to deficits in synaptic plasticity. Here, we use Translating Ribosome Affinity Purification and RNA-seq (TRAP-seq) to identify mistranslating mRNAs in Syngap+/- CA1 pyramidal neurons that exhibit occluded long-term potentiation (LTP). We find the translation environment is significantly altered in a manner that is distinct from the Fmr1-/y model of fragile X syndrome (FXS), another monogenic model of autism and intellectual disability. The Syngap+/- translatome is enriched for regulators of DNA repair and mimics changes induced with chemical LTP (cLTP) in WT. This includes a striking upregulation in the translation of mRNAs with a longer-length (>2 kb) coding sequence (CDS). In contrast, long CDS transcripts are downregulated with induction of Gp1 metabotropic glutamate receptor-induced long-term depression (mGluR-LTD) in WT, and in the Fmr1-/y model that exhibits occluded mGluR-LTD. Together, our results show the Syngap+/- and Fmr1-/y models mimic the translation environments of LTP and LTD, respectively, consistent with the saturation of plasticity states in each model. Moreover, we show that translation of >2 kb mRNAs is a defining feature of LTP that is oppositely regulated during LTD, revealing a novel mRNA signature of plasticity.
Collapse
Affiliation(s)
- Aditi Singh
- Rosamund Stone Zander Translational Neuroscience Center, F. M. Kirby Center, Department of Neurology, Harvard Medical School, Boston Children's Hospital, Boston, Massachusetts 02115
- Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh EH8 9XD, United Kingdom
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh EH8 9XD, United Kingdom
| | - Manuela Rizzi
- Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh EH8 9XD, United Kingdom
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh EH8 9XD, United Kingdom
| | - Sang S Seo
- Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh EH8 9XD, United Kingdom
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh EH8 9XD, United Kingdom
| | - Emily K Osterweil
- Rosamund Stone Zander Translational Neuroscience Center, F. M. Kirby Center, Department of Neurology, Harvard Medical School, Boston Children's Hospital, Boston, Massachusetts 02115
- Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh EH8 9XD, United Kingdom
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh EH8 9XD, United Kingdom
| |
Collapse
|
3
|
Wang YZ, Savas JN. Reply to "Concerns regarding the interpretation of Shank3 protein isoforms expressed in Shank3B -/- mice: potential off-target effects by a neomycin resistance cassette". Mol Psychiatry 2025; 30:1712-1713. [PMID: 39843547 DOI: 10.1038/s41380-025-02903-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 01/08/2025] [Accepted: 01/17/2025] [Indexed: 01/24/2025]
Affiliation(s)
- Yi-Zhi Wang
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Jeffrey N Savas
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA.
| |
Collapse
|
4
|
McKee JL, Magielski JH, Xian J, Cohen S, Toib J, Harrison A, Chen C, Kim D, Rathod A, Brimble E, Fitter N, Graglia JM, Helde KA, McKeown Ruggiero S, Boland MJ, Prosser BL, Sederman R, Helbig I. Clinical signatures of SYNGAP1-related disorders through data integration. Genet Med 2025; 27:101419. [PMID: 40119723 DOI: 10.1016/j.gim.2025.101419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 03/12/2025] [Accepted: 03/14/2025] [Indexed: 03/24/2025] Open
Abstract
PURPOSE SYNGAP1 is a genetic neurodevelopmental disorder characterized by generalized epilepsy, autism, and intellectual disability. Despite a comparatively high prevalence, the longitudinal landscape remains relatively unexplored, and complete characterization is essential for clinical trial readiness. METHODS We combined electronic medical record data (n = 158) with insurance claims data (n = 246) to evaluate longitudinal progression of symptoms. RESULTS Phenotypes associated with SYNGAP1 included behavioral abnormalities (odds ratio [OR]: 12.35, 95% CI: 9.21-16.78), generalized-onset seizures (OR: 1.56, 95% CI: 1.20-2.02), autism (OR: 12.23, 95% CI: 9.29-16.24), and a developmental profile with prominent deficits in verbal skill acquisition. Several clinical features showed distinct age-related patterns, such as a more than 5-fold risk of autistic behavior emerging between 27 and 30 months. Generalized-onset seizures were significantly increased (OR: 4.05, 95% CI: 2.02-7.59) after 3 years of age and persisted over time. Valproic acid and clobazam were commonly used for epilepsy treatment, whereas risperidone, aripiprazole, and guanfacine were commonly used for behavior management. Valproate and lamotrigine were more effective at reducing seizure frequencies or maintaining seizure freedom than other antiseizure medications. CONCLUSION We delineated the seizure, developmental, and behavioral trajectories in SYNGAP1-related disorders, to improve diagnosis, prognosis, and clinical care, and facilitating clinical trial readiness.
Collapse
Affiliation(s)
- Jillian L McKee
- Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, PA; The Epilepsy NeuroGenetics Initiative (ENGIN), Children's Hospital of Philadelphia, Philadelphia, PA; Department of Biomedical and Health Informatics (DBHi), Children's Hospital of Philadelphia, Philadelphia, PA; Epilepsy and Neurodevelopmental Disorders Center (ENDD), Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA; Department of Neurology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Jan H Magielski
- Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, PA; The Epilepsy NeuroGenetics Initiative (ENGIN), Children's Hospital of Philadelphia, Philadelphia, PA; Department of Biomedical and Health Informatics (DBHi), Children's Hospital of Philadelphia, Philadelphia, PA; Epilepsy and Neurodevelopmental Disorders Center (ENDD), Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Julie Xian
- Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, PA; The Epilepsy NeuroGenetics Initiative (ENGIN), Children's Hospital of Philadelphia, Philadelphia, PA; Department of Biomedical and Health Informatics (DBHi), Children's Hospital of Philadelphia, Philadelphia, PA; Epilepsy and Neurodevelopmental Disorders Center (ENDD), Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Stacey Cohen
- Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, PA; The Epilepsy NeuroGenetics Initiative (ENGIN), Children's Hospital of Philadelphia, Philadelphia, PA; Epilepsy and Neurodevelopmental Disorders Center (ENDD), Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Jonathan Toib
- Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, PA; The Epilepsy NeuroGenetics Initiative (ENGIN), Children's Hospital of Philadelphia, Philadelphia, PA
| | - Alicia Harrison
- Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, PA; The Epilepsy NeuroGenetics Initiative (ENGIN), Children's Hospital of Philadelphia, Philadelphia, PA; Epilepsy and Neurodevelopmental Disorders Center (ENDD), Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | | | - Dan Kim
- Ambit RD, Inc, Morristown, NJ
| | | | | | | | | | | | - Sarah McKeown Ruggiero
- Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, PA; The Epilepsy NeuroGenetics Initiative (ENGIN), Children's Hospital of Philadelphia, Philadelphia, PA; Epilepsy and Neurodevelopmental Disorders Center (ENDD), Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Michael J Boland
- Epilepsy and Neurodevelopmental Disorders Center (ENDD), Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA; Department of Physiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Benjamin L Prosser
- Epilepsy and Neurodevelopmental Disorders Center (ENDD), Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA; Department of Physiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Rob Sederman
- Department of Neurology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Ingo Helbig
- Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, PA; The Epilepsy NeuroGenetics Initiative (ENGIN), Children's Hospital of Philadelphia, Philadelphia, PA; Department of Biomedical and Health Informatics (DBHi), Children's Hospital of Philadelphia, Philadelphia, PA; Epilepsy and Neurodevelopmental Disorders Center (ENDD), Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA; Department of Neurology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA.
| |
Collapse
|
5
|
Yang X, Huang YWA, Marshall J. Targeting TrkB-PSD-95 coupling to mitigate neurological disorders. Neural Regen Res 2025; 20:715-724. [PMID: 38886937 PMCID: PMC11433911 DOI: 10.4103/nrr.nrr-d-23-02000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 03/15/2024] [Accepted: 03/30/2024] [Indexed: 06/20/2024] Open
Abstract
Tropomyosin receptor kinase B (TrkB) signaling plays a pivotal role in dendritic growth and dendritic spine formation to promote learning and memory. The activity-dependent release of brain-derived neurotrophic factor at synapses binds to pre- or postsynaptic TrkB resulting in the strengthening of synapses, reflected by long-term potentiation. Postsynaptically, the association of postsynaptic density protein-95 with TrkB enhances phospholipase Cγ-Ca2+/calmodulin-dependent protein kinase II and phosphatidylinositol 3-kinase-mechanistic target of rapamycin signaling required for long-term potentiation. In this review, we discuss TrkB-postsynaptic density protein-95 coupling as a promising strategy to magnify brain-derived neurotrophic factor signaling towards the development of novel therapeutics for specific neurological disorders. A reduction of TrkB signaling has been observed in neurodegenerative disorders, such as Alzheimer's disease and Huntington's disease, and enhancement of postsynaptic density protein-95 association with TrkB signaling could mitigate the observed deficiency of neuronal connectivity in schizophrenia and depression. Treatment with brain-derived neurotrophic factor is problematic, due to poor pharmacokinetics, low brain penetration, and side effects resulting from activation of the p75 neurotrophin receptor or the truncated TrkB.T1 isoform. Although TrkB agonists and antibodies that activate TrkB are being intensively investigated, they cannot distinguish the multiple human TrkB splicing isoforms or cell type-specific functions. Targeting TrkB-postsynaptic density protein-95 coupling provides an alternative approach to specifically boost TrkB signaling at localized synaptic sites versus global stimulation that risks many adverse side effects.
Collapse
Affiliation(s)
- Xin Yang
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI, USA
| | - Yu-Wen Alvin Huang
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI, USA
- Department of Neurology, Warren Alpert Medical School of Brown University, Providence, RI, USA
- Center for Translational Neuroscience, Robert J. and Nancy D. Carney Institute for Brain Science and Brown Institute for Translational Science, Brown University, Providence, RI, USA
| | - John Marshall
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI, USA
| |
Collapse
|
6
|
Yang S, Zhu Y, Xiao J, Yan L, Zhuang X, Li Q, Li Y, Xie J, Du G, Zhou F, Fan G, Feng C. Hippocampal MEF2C phosphorylation mediates synaptic plasticity in lead-induced learning and memory impairments. Toxicology 2025; 512:154082. [PMID: 39947421 DOI: 10.1016/j.tox.2025.154082] [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: 11/23/2024] [Revised: 02/07/2025] [Accepted: 02/07/2025] [Indexed: 02/24/2025]
Abstract
Lead (Pb), an ancient metallic environmental pollutant, has received widespread attention and has been strictly controlled, but current findings have found the hazards of Pb to be much higher than previously estimated. Pb exposure can lead to memory impairment. However, the underlying molecular mechanisms have not been fully elucidated. Dynamic changes in dendritic spines form the structural basis of memory, and MEF2C, a key regulator of dendritic plasticity, plays an important role in hippocampus-dependent learning and memory. This study focused on the role of ERK-induced MEF2C phosphorylation in Pb-induced learning and memory impairments. A rat model of Pb exposure from the embryo to adolescence and the model of Pb exposure in PC12 cells were established. The results showed that Pb exposure reduced the phosphorylation level of MEF2C in the rat hippocampus by affecting the expression of ERK5. In addition, it reduced the expression of MEF2C at the translation stage, which affected its transcriptional ability and led to abnormal expression of the downstream factor ARC, causing alterations in neuronal dendritic plasticity and affecting its memory ability. Further experiments showed that regulating the phosphorylation level of MEF2C could affect neurite growth in PC12 cells exposed to Pb. Our findings demonstrate that Pb exposure may contribute to memory impairment by affecting MEF2C and its phosphorylation levels, resulting in altered dendritic plasticity.
Collapse
Affiliation(s)
- Shuo Yang
- School of Public Health, Nanchang University, Nanchang, Jiangxi 330006, PR China; Jiangxi Provincial Key Laboratory of Preventive Medicine, School of Public Health, Nanchang University, Nanchang, Jiangxi 330006, PR China
| | - Yanhui Zhu
- School of Public Health, Nanchang University, Nanchang, Jiangxi 330006, PR China; Jiangxi Provincial Key Laboratory of Preventive Medicine, School of Public Health, Nanchang University, Nanchang, Jiangxi 330006, PR China
| | - Jinmei Xiao
- School of Public Health, Nanchang University, Nanchang, Jiangxi 330006, PR China; Jiangxi Provincial Key Laboratory of Preventive Medicine, School of Public Health, Nanchang University, Nanchang, Jiangxi 330006, PR China
| | - Lingyu Yan
- School of Public Health, Nanchang University, Nanchang, Jiangxi 330006, PR China; Jiangxi Provincial Key Laboratory of Preventive Medicine, School of Public Health, Nanchang University, Nanchang, Jiangxi 330006, PR China
| | - Xuebing Zhuang
- School of Public Health, Nanchang University, Nanchang, Jiangxi 330006, PR China; Jiangxi Provincial Key Laboratory of Preventive Medicine, School of Public Health, Nanchang University, Nanchang, Jiangxi 330006, PR China
| | - Qi Li
- School of Public Health, Nanchang University, Nanchang, Jiangxi 330006, PR China; Jiangxi Provincial Key Laboratory of Preventive Medicine, School of Public Health, Nanchang University, Nanchang, Jiangxi 330006, PR China
| | - Yue Li
- School of Public Health, Nanchang University, Nanchang, Jiangxi 330006, PR China; Jiangxi Provincial Key Laboratory of Preventive Medicine, School of Public Health, Nanchang University, Nanchang, Jiangxi 330006, PR China
| | - Jie Xie
- School of Public Health, Nanchang University, Nanchang, Jiangxi 330006, PR China; Jiangxi Provincial Key Laboratory of Preventive Medicine, School of Public Health, Nanchang University, Nanchang, Jiangxi 330006, PR China
| | - Guihua Du
- School of Public Health, Nanchang University, Nanchang, Jiangxi 330006, PR China; Jiangxi Provincial Key Laboratory of Preventive Medicine, School of Public Health, Nanchang University, Nanchang, Jiangxi 330006, PR China
| | - Fankun Zhou
- School of Public Health, Nanchang University, Nanchang, Jiangxi 330006, PR China; Jiangxi Provincial Key Laboratory of Preventive Medicine, School of Public Health, Nanchang University, Nanchang, Jiangxi 330006, PR China
| | - Guangqin Fan
- School of Public Health, Nanchang University, Nanchang, Jiangxi 330006, PR China; Jiangxi Provincial Key Laboratory of Preventive Medicine, School of Public Health, Nanchang University, Nanchang, Jiangxi 330006, PR China.
| | - Chang Feng
- School of Public Health, Nanchang University, Nanchang, Jiangxi 330006, PR China; Jiangxi Provincial Key Laboratory of Preventive Medicine, School of Public Health, Nanchang University, Nanchang, Jiangxi 330006, PR China.
| |
Collapse
|
7
|
Ansari A, Thibault PA, Salapa HE, Clarke JPWE, Levin MC. Mutations in hnRNP A1 drive neurodegeneration and alternative RNA splicing of neuronal gene targets. Neurobiol Dis 2025; 206:106814. [PMID: 39874994 DOI: 10.1016/j.nbd.2025.106814] [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: 10/17/2024] [Revised: 01/23/2025] [Accepted: 01/24/2025] [Indexed: 01/30/2025] Open
Abstract
RNA binding protein dysfunction is a pathogenic feature of multiple neurological diseases, including multiple sclerosis (MS). Neurodegeneration (the loss of, or damage to neurons and axons) is the primary driver of disease progression in MS. Herein, we utilized a novel, neuron-specific model of neurodegeneration by transducing primary mouse neurons with mutant forms of the RNA binding protein heterogeneous nuclear ribonucleoprotein A1 (hnRNP A1) identified from MS patients, including one within the M9-nuclear localization sequence of hnRNP A1 (A1(P275S)) and a second in the prion-like domain of hnRNP A1 (A1(F263S)) to test the hypothesis that neuronal hnRNP A1 dysfunction drives neurodegeneration in MS. Examination of hnRNP A1 localization in neurons revealed an increase in nucleocytoplasmic mislocalization in neurons transduced with A1(P275S), but not A1(F263S). Yet, both A1(F263S) and A1(P275S) induced neurodegeneration evidenced by significant reductions in total neurite length and complexity and an increase in FluoroJade-C neuronal cell body staining. RNA sequencing and differential alternative splicing analysis of mutant-expressing neurons revealed dramatic changes in alternative RNA splicing of transcripts critical to neuronal function. Further, amyloid precursor protein (APP), a marker for neurodegeneration in MS, showed differential splicing in mutant-expressing neurons, which was confirmed in MS brains with hnRNP A1 dysfunction. Overall, we have identified that hnRNP A1 plays a complex role in neuronal function and regulation by mediating the alternative splicing of neuron-specific transcripts. When neuronal hnRNP A1 function is impaired, as in disease, resultant dysfunction propagates through multiple pathways that may influence the progression of neurodegeneration in MS.
Collapse
Affiliation(s)
- Ansalna Ansari
- Office of the Saskatchewan Multiple Sclerosis Clinical Research Chair, University of Saskatchewan, Saskatoon, SK S7K 0M7, Canada; Department of Anatomy, Physiology and Pharmacology, College of Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | - Patricia A Thibault
- Office of the Saskatchewan Multiple Sclerosis Clinical Research Chair, University of Saskatchewan, Saskatoon, SK S7K 0M7, Canada; Cameco MS Neuroscience Research Centre, College of Medicine, University of Saskatchewan, Saskatoon, SK S7K 0M7, Canada; Neurology Division, Department of Medicine, University of Saskatchewan, Saskatoon, SK S7N 0X8, Canada
| | - Hannah E Salapa
- Office of the Saskatchewan Multiple Sclerosis Clinical Research Chair, University of Saskatchewan, Saskatoon, SK S7K 0M7, Canada; Cameco MS Neuroscience Research Centre, College of Medicine, University of Saskatchewan, Saskatoon, SK S7K 0M7, Canada; Neurology Division, Department of Medicine, University of Saskatchewan, Saskatoon, SK S7N 0X8, Canada
| | - Joseph-Patrick W E Clarke
- Office of the Saskatchewan Multiple Sclerosis Clinical Research Chair, University of Saskatchewan, Saskatoon, SK S7K 0M7, Canada; Cameco MS Neuroscience Research Centre, College of Medicine, University of Saskatchewan, Saskatoon, SK S7K 0M7, Canada; Neurology Division, Department of Medicine, University of Saskatchewan, Saskatoon, SK S7N 0X8, Canada
| | - Michael C Levin
- Office of the Saskatchewan Multiple Sclerosis Clinical Research Chair, University of Saskatchewan, Saskatoon, SK S7K 0M7, Canada; Neurology Division, Department of Medicine, University of Saskatchewan, Saskatoon, SK S7N 0X8, Canada.
| |
Collapse
|
8
|
Hong L, Yuan Q. Genotype-Phenotype Correlations in SYNGAP1-Related Mental Retardation Type 5. Clin Genet 2025; 107:136-146. [PMID: 39647930 DOI: 10.1111/cge.14661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 11/15/2024] [Accepted: 11/18/2024] [Indexed: 12/10/2024]
Abstract
Variants in the SYNGAP1 gene leading to decreased SynGAP protein expression are critical for the pathogenesis of mental retardation type 5 (MRD5). This study aims to explore the relationship between SYNGAP1 genotype and clinical phenotype through an expanded sample size, thereby enhancing the understanding of the specific mechanisms underlying MRD5. Data from previously published cases of patients with SYNGAP1 mutations were collected, and the relationship between genotype and clinical phenotype was analyzed. A total of 246 MRD5 patients were included in the analysis. Among them, 98.7% (224/227) were diagnosed with intellectual disability (ID), 91.6% (208/227) with epilepsy, and 57.3% (137/239) with autism spectrum disorder (ASD). The clinical phenotypes of MRD5 patients were found to be associated with their genotypes. Variants located in exons 1 to 6 may correlate with milder ID and reduced risk of ASD, yet they are more likely to present as refractory epilepsy.
Collapse
Affiliation(s)
- Liying Hong
- Department of Functional (ECG Room), Nanchang First Hospital, Nanchang, China
| | - Qifeng Yuan
- Department of Pediatrics, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
9
|
Medyanik AD, Anisimova PE, Kustova AO, Tarabykin VS, Kondakova EV. Developmental and Epileptic Encephalopathy: Pathogenesis of Intellectual Disability Beyond Channelopathies. Biomolecules 2025; 15:133. [PMID: 39858526 PMCID: PMC11763800 DOI: 10.3390/biom15010133] [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: 12/06/2024] [Revised: 01/11/2025] [Accepted: 01/13/2025] [Indexed: 01/27/2025] Open
Abstract
Developmental and epileptic encephalopathies (DEEs) are a group of neuropediatric diseases associated with epileptic seizures, severe delay or regression of psychomotor development, and cognitive and behavioral deficits. What sets DEEs apart is their complex interplay of epilepsy and developmental delay, often driven by genetic factors. These two aspects influence one another but can develop independently, creating diagnostic and therapeutic challenges. Intellectual disability is severe and complicates potential treatment. Pathogenic variants are found in 30-50% of patients with DEE. Many genes mutated in DEEs encode ion channels, causing current conduction disruptions known as channelopathies. Although channelopathies indeed make up a significant proportion of DEE cases, many other mechanisms have been identified: impaired neurogenesis, metabolic disorders, disruption of dendrite and axon growth, maintenance and synapse formation abnormalities -synaptopathies. Here, we review recent publications on non-channelopathies in DEE with an emphasis on the mechanisms linking epileptiform activity with intellectual disability. We focus on three major mechanisms of intellectual disability in DEE and describe several recently identified genes involved in the pathogenesis of DEE.
Collapse
Affiliation(s)
- Alexandra D. Medyanik
- Institute of Neuroscience, Lobachevsky State University of Nizhny Novgorod, 23 Gagarin Ave., 603022 Nizhny Novgorod, Russia; (A.D.M.); (P.E.A.); (A.O.K.); (E.V.K.)
| | - Polina E. Anisimova
- Institute of Neuroscience, Lobachevsky State University of Nizhny Novgorod, 23 Gagarin Ave., 603022 Nizhny Novgorod, Russia; (A.D.M.); (P.E.A.); (A.O.K.); (E.V.K.)
| | - Angelina O. Kustova
- Institute of Neuroscience, Lobachevsky State University of Nizhny Novgorod, 23 Gagarin Ave., 603022 Nizhny Novgorod, Russia; (A.D.M.); (P.E.A.); (A.O.K.); (E.V.K.)
| | - Victor S. Tarabykin
- Institute of Neuroscience, Lobachevsky State University of Nizhny Novgorod, 23 Gagarin Ave., 603022 Nizhny Novgorod, Russia; (A.D.M.); (P.E.A.); (A.O.K.); (E.V.K.)
- Institute of Cell Biology and Neurobiology, Charité—Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Elena V. Kondakova
- Institute of Neuroscience, Lobachevsky State University of Nizhny Novgorod, 23 Gagarin Ave., 603022 Nizhny Novgorod, Russia; (A.D.M.); (P.E.A.); (A.O.K.); (E.V.K.)
| |
Collapse
|
10
|
Brill J, Clarke B, Hong I, Huganir RL. Dissociation of SYNGAP1 Enzymatic and Structural Roles: Intrinsic Excitability and Seizure Susceptibility. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.14.633019. [PMID: 39868300 PMCID: PMC11761602 DOI: 10.1101/2025.01.14.633019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
SYNGAP1 is a key Ras-GAP protein enriched at excitatory synapses, with mutations causing intellectual disability and epilepsy in humans. Recent studies have revealed that in addition to its role as a negative regulator of G-protein signaling through its GAP enzymatic activity, SYNGAP1 plays an important structural role through its interaction with post-synaptic density proteins. Here, we reveal that intrinsic excitability deficits and seizure phenotypes in heterozygous Syngap1 knockout (KO) mice are differentially dependent on Syngap1 GAP activity. Cortical excitatory neurons in heterozygous KO mice displayed reduced intrinsic excitability, including lower input resistance, and increased rheobase, a phenotype recapitulated in GAP-deficient Syngap1 mutants. However, seizure severity and susceptibility to pentylenetetrazol (PTZ)-induced seizures were significantly elevated in heterozygous KO mice but unaffected in GAP-deficient mutants, implicating the structural rather than enzymatic role of Syngap1 in seizure regulation. These findings highlight the complex interplay between SYNGAP1 structural and catalytic functions in neuronal physiology and disease.
Collapse
Affiliation(s)
- Julia Brill
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Blaise Clarke
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Ingie Hong
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Richard L. Huganir
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
11
|
Barão S, Hong I, Müller U, Huganir RL. Syngap1 and the development of murine neocortical progenitor cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.18.629233. [PMID: 39763888 PMCID: PMC11702710 DOI: 10.1101/2024.12.18.629233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/18/2025]
Abstract
SYNGAP1 is a major regulator of synaptic plasticity through its interaction with synaptic scaffold proteins and modulation of Ras and Rap GTPase signaling pathways. SYNGAP1 mutations in humans are often associated with intellectual disability, epilepsy, and autism spectrum disorder. Syngap1 heterozygous loss-of-function results in impaired LTP, premature maturation of dendritic spines, learning disabilities and seizures in mice. More recently, SYNGAP1 was shown to influence cortical neurogenesis and the proliferation of progenitors in human organoids. Here, we show that the absence or haploinsufficiency of Syngap1 does not influence the properties of neocortical progenitors and their cellular output in mice. This discrepancy highlights potential species-specific or methodological differences and raises important questions about the broader applicability of SYNGAP1's role in neurogenesis.
Collapse
Affiliation(s)
- Soraia Barão
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Ingie Hong
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Ulrich Müller
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Richard L. Huganir
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
12
|
Jadhav V, Carreno-Munoz MI, Chehrazi P, Michaud JL, Chattopadhyaya B, Di Cristo G. Developmental Syngap1 Haploinsufficiency in Medial Ganglionic Eminence-Derived Interneurons Impairs Auditory Cortex Activity, Social Behavior, and Extinction of Fear Memory. J Neurosci 2024; 44:e0946242024. [PMID: 39406516 PMCID: PMC11622180 DOI: 10.1523/jneurosci.0946-24.2024] [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: 05/17/2024] [Revised: 07/26/2024] [Accepted: 07/31/2024] [Indexed: 12/06/2024] Open
Abstract
Mutations in SYNGAP1, a protein enriched at glutamatergic synapses, cause intellectual disability associated with epilepsy, autism spectrum disorder, and sensory dysfunctions. Several studies showed that Syngap1 regulates the time course of forebrain glutamatergic synapse maturation; however, the developmental role of Syngap1 in inhibitory GABAergic neurons is less clear. GABAergic neurons can be classified into different subtypes based on their morphology, connectivity, and physiological properties. Whether Syngap1 expression specifically in parvalbumin (PV)-expressing and somatostatin (SST)-expressing interneurons, which are derived from the medial ganglionic eminence (MGE), plays a role in the emergence of distinct brain functions remains largely unknown. We used genetic strategies to generate Syngap1 haploinsufficiency in (1) prenatal interneurons derived from the medial ganglionic eminence, (2) in postnatal PV cells, and (3) in prenatal SST interneurons. We further performed in vivo recordings and behavioral assays to test whether and how these different genetic manipulations alter brain function and behavior in mice of either sex. Mice with prenatal-onset Syngap1 haploinsufficiency restricted to Nkx2.1-expressing neurons show abnormal cortical oscillations and increased entrainment induced by 40 Hz auditory stimulation but lack stimulus-specific adaptation. This latter phenotype was reproduced in mice with Syngap1 haploinsufficiency restricted to PV, but not SST, interneurons. Prenatal-onset Syngap1 haploinsufficiency in Nkx2.1-expressing neurons led to impaired social behavior and inability to extinguish fear memories; however, neither postnatal PV- nor prenatal SST-specific mutant mice show these phenotypes. We speculate that Syngap1 haploinsufficiency in prenatal/perinatal PV interneurons may contribute to cortical activity and cognitive alterations associated with Syngap1 mutations.
Collapse
Affiliation(s)
- Vidya Jadhav
- CHU Sainte-Justine Azrieli Research Centre (CHUSJ), Montréal, Quebec H3T 1C5, Canada
- Department of Neurosciences, Université de Montréal, Montréal, Quebec H3T 1J4, Canada
| | - Maria Isabel Carreno-Munoz
- CHU Sainte-Justine Azrieli Research Centre (CHUSJ), Montréal, Quebec H3T 1C5, Canada
- Department of Neurosciences, Université de Montréal, Montréal, Quebec H3T 1J4, Canada
| | - Pegah Chehrazi
- CHU Sainte-Justine Azrieli Research Centre (CHUSJ), Montréal, Quebec H3T 1C5, Canada
- Department of Neurosciences, Université de Montréal, Montréal, Quebec H3T 1J4, Canada
| | - Jacques L Michaud
- CHU Sainte-Justine Azrieli Research Centre (CHUSJ), Montréal, Quebec H3T 1C5, Canada
- Department of Pediatrics, Université de Montréal, Montréal, Quebec H3T 1C5, Canada
| | | | - Graziella Di Cristo
- CHU Sainte-Justine Azrieli Research Centre (CHUSJ), Montréal, Quebec H3T 1C5, Canada
- Department of Neurosciences, Université de Montréal, Montréal, Quebec H3T 1J4, Canada
| |
Collapse
|
13
|
Mosini A, Moysés-Oliveira M, Adami L, Xavier S, Marquezini B, Kloster A, Balbueno B, de Mello C, Moreira G, Andersen M, Tufik S. Subjective sleep assessment in individuals with SYNGAP1-associated syndrome. J Clin Sleep Med 2024; 20:1879-1885. [PMID: 38958060 PMCID: PMC11609837 DOI: 10.5664/jcsm.11246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 06/14/2024] [Accepted: 06/17/2024] [Indexed: 07/04/2024]
Abstract
STUDY OBJECTIVES Sleep disturbances are common in neurodevelopmental disorders, affecting patients and caregivers' quality of life. SYNGAP1-associated syndrome, a rare neurodevelopmental disorder, is marked by intellectual disability, developmental delay, epilepsy, and sleep issues. However, research on sleep quality in these individuals is limited. This study aimed to evaluate genetic variants, epilepsy, and sleep patterns in SYNGAP1-associated syndrome patients and their caregivers. METHODS An online survey was applied to 11 caregivers of individuals diagnosed with SYNGAP1-associated syndrome. Specific clinical inquiries were included, addressing childbirth, previous surgeries, and medication use. Inquiries about epilepsy included type of epilepsy, type and frequency of seizures, antiseizure medications, and complementary nonpharmacological treatments. Children's Sleep Habits Questionnaire was applied to assess the patients' sleep profile. Pittsburgh Sleep Quality Index was used to evaluate the sleep quality of caregivers. RESULTS Genetic analysis showed heterozygous mutations in SYNGAP1, often leading to loss of function. Epilepsy was present in 82% of participants, with 77.8% having drug-resistant seizures. Using the Children's Sleep Habits Questionnaire, 81.8% of patients exhibited poor sleep habits, including bedtime resistance, anxiety, night awakenings, parasomnias, and daytime sleepiness. Caregivers also reported poor sleep quality according to the Pittsburgh Sleep Quality Index. CONCLUSIONS This study highlights the high prevalence of epilepsy and sleep problems in SYNGAP1-associated syndrome, impacting both patients and caregivers. Further research is crucial to understand the syndrome's effects on sleep disturbances, emphasizing the need for targeted interventions to improve sleep quality in individuals with rare genetic syndromes and their caregivers. CITATION Mosini A, Moysés-Oliveira M, Adami L, et al. Subjective sleep assessment in individuals with SYNGAP1-associated syndrome. J Clin Sleep Med. 2024;20(12):1879-1885.
Collapse
Affiliation(s)
- Amanda Mosini
- Sleep Institute, Associação Fundo de Incentivo à Pesquisa, São Paulo, Brazil
- Departamento de Psicobiologia, Universidade Federal de São Paulo, São Paulo, Brazil
| | | | - Luana Adami
- Sleep Institute, Associação Fundo de Incentivo à Pesquisa, São Paulo, Brazil
- Departamento de Psicobiologia, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Sandra Xavier
- Sleep Institute, Associação Fundo de Incentivo à Pesquisa, São Paulo, Brazil
- Departamento de Psicobiologia, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Bruna Marquezini
- Sleep Institute, Associação Fundo de Incentivo à Pesquisa, São Paulo, Brazil
| | - Anna Kloster
- Sleep Institute, Associação Fundo de Incentivo à Pesquisa, São Paulo, Brazil
| | - Bianca Balbueno
- Sleep Institute, Associação Fundo de Incentivo à Pesquisa, São Paulo, Brazil
| | - Claudia de Mello
- Sleep Institute, Associação Fundo de Incentivo à Pesquisa, São Paulo, Brazil
- Departamento de Psicobiologia, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Gustavo Moreira
- Sleep Institute, Associação Fundo de Incentivo à Pesquisa, São Paulo, Brazil
- Departamento de Psicobiologia, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Monica Andersen
- Sleep Institute, Associação Fundo de Incentivo à Pesquisa, São Paulo, Brazil
- Departamento de Psicobiologia, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Sergio Tufik
- Sleep Institute, Associação Fundo de Incentivo à Pesquisa, São Paulo, Brazil
- Departamento de Psicobiologia, Universidade Federal de São Paulo, São Paulo, Brazil
| |
Collapse
|
14
|
Fenton TA, Haouchine OY, Hallam EB, Smith EM, Jackson KC, Rahbarian D, Canales CP, Adhikari A, Nord AS, Ben-Shalom R, Silverman JL. Hyperexcitability and translational phenotypes in a preclinical mouse model of SYNGAP1-related intellectual disability. Transl Psychiatry 2024; 14:405. [PMID: 39358332 PMCID: PMC11447000 DOI: 10.1038/s41398-024-03077-6] [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: 03/10/2024] [Revised: 08/22/2024] [Accepted: 08/27/2024] [Indexed: 10/04/2024] Open
Abstract
Disruption of SYNGAP1 directly causes a genetically identifiable neurodevelopmental disorder (NDD) called SYNGAP1-related intellectual disability (SRID). Without functional SynGAP1 protein, individuals are developmentally delayed and have prominent features of intellectual disability (ID), motor impairments, and epilepsy. Over the past two decades, there have been numerous discoveries indicating the critical role of Syngap1. Several rodent models with a loss of Syngap1 have been engineered, identifying precise roles in neuronal structure and function, as well as key biochemical pathways key for synapse integrity. Homozygous loss of SYNGAP1/Syngap1 is lethal. Heterozygous mutations of Syngap1 result in a broad range of behavioral phenotypes. Our in vivo functional data, using the original mouse model from the Huganir laboratory, corroborated behaviors including robust hyperactivity and deficits in learning and memory in young adults. Furthermore, we described impairments in the domain of sleep, characterized using neurophysiological data that was collected with wireless, telemetric electroencephalography (EEG). Syngap1+/- mice exhibited elevated spiking events and spike trains, in addition to elevated power, most notably in the delta power frequency. For the first time, we illustrated that primary neurons from Syngap1+/- mice displayed: 1) increased network firing activity, 2) greater bursts, 3) and shorter inter-burst intervals between peaks, by utilizing high density microelectrode arrays (HD-MEA). Our work bridges in vitro electrophysiological neuronal activity and function with in vivo neurophysiological brain activity and function. These data elucidate quantitative, translational biomarkers in vivo and in vitro that can be utilized for the development and efficacy assessment of targeted treatments for SRID.
Collapse
Affiliation(s)
- Timothy A Fenton
- MIND Institute, University of California Davis School of Medicine, Sacramento, CA, 95817, USA
- Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, CA, 95817, USA
| | - Olivia Y Haouchine
- MIND Institute, University of California Davis School of Medicine, Sacramento, CA, 95817, USA
- Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, CA, 95817, USA
| | - Elizabeth B Hallam
- MIND Institute, University of California Davis School of Medicine, Sacramento, CA, 95817, USA
- Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, CA, 95817, USA
| | - Emily M Smith
- UC Davis Center for Neuroscience; Department of Psychiatry and Behavioral Sciences & Department of Neurobiology, Physiology and Behavior, University of California Davis, Davis, CA, 95616, USA
| | - Kiya C Jackson
- UC Davis Center for Neuroscience; Department of Psychiatry and Behavioral Sciences & Department of Neurobiology, Physiology and Behavior, University of California Davis, Davis, CA, 95616, USA
| | - Darlene Rahbarian
- UC Davis Center for Neuroscience; Department of Psychiatry and Behavioral Sciences & Department of Neurobiology, Physiology and Behavior, University of California Davis, Davis, CA, 95616, USA
| | - Cesar P Canales
- Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, CA, 95817, USA
- UC Davis Center for Neuroscience; Department of Psychiatry and Behavioral Sciences & Department of Neurobiology, Physiology and Behavior, University of California Davis, Davis, CA, 95616, USA
| | - Anna Adhikari
- MIND Institute, University of California Davis School of Medicine, Sacramento, CA, 95817, USA
- Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, CA, 95817, USA
| | - Alex S Nord
- MIND Institute, University of California Davis School of Medicine, Sacramento, CA, 95817, USA
- Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, CA, 95817, USA
- UC Davis Center for Neuroscience; Department of Psychiatry and Behavioral Sciences & Department of Neurobiology, Physiology and Behavior, University of California Davis, Davis, CA, 95616, USA
| | - Roy Ben-Shalom
- MIND Institute, University of California Davis School of Medicine, Sacramento, CA, 95817, USA
- Department of Neurology, University of California Davis School of Medicine, Sacramento, CA, 95817, USA
| | - Jill L Silverman
- MIND Institute, University of California Davis School of Medicine, Sacramento, CA, 95817, USA.
- Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, CA, 95817, USA.
| |
Collapse
|
15
|
Katsanevaki D, Till SM, Buller-Peralta I, Nawaz MS, Louros SR, Kapgal V, Tiwari S, Walsh D, Anstey NJ, Petrović NG, Cormack A, Salazar-Sanchez V, Harris A, Farnworth-Rowson W, Sutherland A, Watson TC, Dimitrov S, Jackson AD, Arkell D, Biswal S, Dissanayake KN, Mizen LAM, Perentos N, Jones MW, Cousin MA, Booker SA, Osterweil EK, Chattarji S, Wyllie DJA, Gonzalez-Sulser A, Hardt O, Wood ER, Kind PC. Key roles of C2/GAP domains in SYNGAP1-related pathophysiology. Cell Rep 2024; 43:114733. [PMID: 39269903 DOI: 10.1016/j.celrep.2024.114733] [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: 01/18/2023] [Revised: 07/30/2024] [Accepted: 08/23/2024] [Indexed: 09/15/2024] Open
Abstract
Mutations in SYNGAP1 are a common genetic cause of intellectual disability (ID) and a risk factor for autism. SYNGAP1 encodes a synaptic GTPase-activating protein (GAP) that has both signaling and scaffolding roles. Most pathogenic variants of SYNGAP1 are predicted to result in haploinsufficiency. However, some affected individuals carry missense mutations in its calcium/lipid binding (C2) and GAP domains, suggesting that many clinical features result from loss of functions carried out by these domains. To test this hypothesis, we targeted the exons encoding the C2 and GAP domains of SYNGAP. Rats heterozygous for this deletion exhibit reduced exploration and fear extinction, altered social investigation, and spontaneous seizures-key phenotypes shared with Syngap heterozygous null rats. Together, these findings indicate that the reduction of SYNGAP C2/GAP domain function is a main feature of SYNGAP haploinsufficiency. This rat model provides an important system for the study of ID, autism, and epilepsy.
Collapse
Affiliation(s)
- Danai Katsanevaki
- Simons Initiative for the Developing Brain, Centre for Discovery Brain Sciences, University of Edinburgh, EH8 9XD Edinburgh, UK; Patrick Wild Centre, University of Edinburgh, EH8 9XD Edinburgh, UK
| | - Sally M Till
- Simons Initiative for the Developing Brain, Centre for Discovery Brain Sciences, University of Edinburgh, EH8 9XD Edinburgh, UK; Patrick Wild Centre, University of Edinburgh, EH8 9XD Edinburgh, UK; Centre for Brain Development and Repair, Instem, Bangalore 560065, India
| | - Ingrid Buller-Peralta
- Simons Initiative for the Developing Brain, Centre for Discovery Brain Sciences, University of Edinburgh, EH8 9XD Edinburgh, UK; Patrick Wild Centre, University of Edinburgh, EH8 9XD Edinburgh, UK
| | - Mohammad Sarfaraz Nawaz
- Simons Initiative for the Developing Brain, Centre for Discovery Brain Sciences, University of Edinburgh, EH8 9XD Edinburgh, UK; Centre for Brain Development and Repair, Instem, Bangalore 560065, India
| | - Susana R Louros
- Simons Initiative for the Developing Brain, Centre for Discovery Brain Sciences, University of Edinburgh, EH8 9XD Edinburgh, UK; Patrick Wild Centre, University of Edinburgh, EH8 9XD Edinburgh, UK
| | - Vijayakumar Kapgal
- Centre for Brain Development and Repair, Instem, Bangalore 560065, India; The University of Transdisciplinary Health Sciences and Technology, Bangalore 560065, India
| | - Shashank Tiwari
- Centre for Brain Development and Repair, Instem, Bangalore 560065, India
| | - Darren Walsh
- Simons Initiative for the Developing Brain, Centre for Discovery Brain Sciences, University of Edinburgh, EH8 9XD Edinburgh, UK; Patrick Wild Centre, University of Edinburgh, EH8 9XD Edinburgh, UK
| | - Natasha J Anstey
- Simons Initiative for the Developing Brain, Centre for Discovery Brain Sciences, University of Edinburgh, EH8 9XD Edinburgh, UK; Patrick Wild Centre, University of Edinburgh, EH8 9XD Edinburgh, UK; Centre for Brain Development and Repair, Instem, Bangalore 560065, India
| | - Nina G Petrović
- Simons Initiative for the Developing Brain, Centre for Discovery Brain Sciences, University of Edinburgh, EH8 9XD Edinburgh, UK; Patrick Wild Centre, University of Edinburgh, EH8 9XD Edinburgh, UK
| | - Alison Cormack
- Simons Initiative for the Developing Brain, Centre for Discovery Brain Sciences, University of Edinburgh, EH8 9XD Edinburgh, UK; Patrick Wild Centre, University of Edinburgh, EH8 9XD Edinburgh, UK
| | - Vanesa Salazar-Sanchez
- Simons Initiative for the Developing Brain, Centre for Discovery Brain Sciences, University of Edinburgh, EH8 9XD Edinburgh, UK; Patrick Wild Centre, University of Edinburgh, EH8 9XD Edinburgh, UK
| | - Anjanette Harris
- Simons Initiative for the Developing Brain, Centre for Discovery Brain Sciences, University of Edinburgh, EH8 9XD Edinburgh, UK; Patrick Wild Centre, University of Edinburgh, EH8 9XD Edinburgh, UK
| | - William Farnworth-Rowson
- Simons Initiative for the Developing Brain, Centre for Discovery Brain Sciences, University of Edinburgh, EH8 9XD Edinburgh, UK; Patrick Wild Centre, University of Edinburgh, EH8 9XD Edinburgh, UK
| | - Andrew Sutherland
- Simons Initiative for the Developing Brain, Centre for Discovery Brain Sciences, University of Edinburgh, EH8 9XD Edinburgh, UK; Patrick Wild Centre, University of Edinburgh, EH8 9XD Edinburgh, UK
| | - Thomas C Watson
- Simons Initiative for the Developing Brain, Centre for Discovery Brain Sciences, University of Edinburgh, EH8 9XD Edinburgh, UK; Patrick Wild Centre, University of Edinburgh, EH8 9XD Edinburgh, UK
| | - Siyan Dimitrov
- Simons Initiative for the Developing Brain, Centre for Discovery Brain Sciences, University of Edinburgh, EH8 9XD Edinburgh, UK; Patrick Wild Centre, University of Edinburgh, EH8 9XD Edinburgh, UK
| | - Adam D Jackson
- Simons Initiative for the Developing Brain, Centre for Discovery Brain Sciences, University of Edinburgh, EH8 9XD Edinburgh, UK; Patrick Wild Centre, University of Edinburgh, EH8 9XD Edinburgh, UK; Centre for Brain Development and Repair, Instem, Bangalore 560065, India
| | - Daisy Arkell
- Simons Initiative for the Developing Brain, Centre for Discovery Brain Sciences, University of Edinburgh, EH8 9XD Edinburgh, UK; Patrick Wild Centre, University of Edinburgh, EH8 9XD Edinburgh, UK
| | | | - Kosala N Dissanayake
- Simons Initiative for the Developing Brain, Centre for Discovery Brain Sciences, University of Edinburgh, EH8 9XD Edinburgh, UK; Patrick Wild Centre, University of Edinburgh, EH8 9XD Edinburgh, UK
| | - Lindsay A M Mizen
- Simons Initiative for the Developing Brain, Centre for Discovery Brain Sciences, University of Edinburgh, EH8 9XD Edinburgh, UK; Patrick Wild Centre, University of Edinburgh, EH8 9XD Edinburgh, UK
| | - Nikolas Perentos
- Department of Veterinary Medicine, University of Nicosia School of Veterinary Medicine, 2414 Nicosia, Cyprus
| | - Matt W Jones
- School of Physiology, Pharmacology, and Neuroscience, University of Bristol, Biomedical Sciences Building, University Walk, BS8 1TD Bristol, UK
| | - Michael A Cousin
- Simons Initiative for the Developing Brain, Centre for Discovery Brain Sciences, University of Edinburgh, EH8 9XD Edinburgh, UK; Patrick Wild Centre, University of Edinburgh, EH8 9XD Edinburgh, UK; Centre for Brain Development and Repair, Instem, Bangalore 560065, India
| | - Sam A Booker
- Simons Initiative for the Developing Brain, Centre for Discovery Brain Sciences, University of Edinburgh, EH8 9XD Edinburgh, UK; Patrick Wild Centre, University of Edinburgh, EH8 9XD Edinburgh, UK
| | - Emily K Osterweil
- Simons Initiative for the Developing Brain, Centre for Discovery Brain Sciences, University of Edinburgh, EH8 9XD Edinburgh, UK; Patrick Wild Centre, University of Edinburgh, EH8 9XD Edinburgh, UK
| | - Sumantra Chattarji
- Simons Initiative for the Developing Brain, Centre for Discovery Brain Sciences, University of Edinburgh, EH8 9XD Edinburgh, UK; Patrick Wild Centre, University of Edinburgh, EH8 9XD Edinburgh, UK; Centre for Brain Development and Repair, Instem, Bangalore 560065, India
| | - David J A Wyllie
- Simons Initiative for the Developing Brain, Centre for Discovery Brain Sciences, University of Edinburgh, EH8 9XD Edinburgh, UK; Patrick Wild Centre, University of Edinburgh, EH8 9XD Edinburgh, UK; Centre for Brain Development and Repair, Instem, Bangalore 560065, India
| | - Alfredo Gonzalez-Sulser
- Simons Initiative for the Developing Brain, Centre for Discovery Brain Sciences, University of Edinburgh, EH8 9XD Edinburgh, UK; Patrick Wild Centre, University of Edinburgh, EH8 9XD Edinburgh, UK
| | - Oliver Hardt
- Simons Initiative for the Developing Brain, Centre for Discovery Brain Sciences, University of Edinburgh, EH8 9XD Edinburgh, UK; Patrick Wild Centre, University of Edinburgh, EH8 9XD Edinburgh, UK; Centre for Brain Development and Repair, Instem, Bangalore 560065, India; Department of Psychology, McGill University, Montreal, QC H3A 1G1, Canada
| | - Emma R Wood
- Simons Initiative for the Developing Brain, Centre for Discovery Brain Sciences, University of Edinburgh, EH8 9XD Edinburgh, UK; Patrick Wild Centre, University of Edinburgh, EH8 9XD Edinburgh, UK; Centre for Brain Development and Repair, Instem, Bangalore 560065, India
| | - Peter C Kind
- Simons Initiative for the Developing Brain, Centre for Discovery Brain Sciences, University of Edinburgh, EH8 9XD Edinburgh, UK; Patrick Wild Centre, University of Edinburgh, EH8 9XD Edinburgh, UK; Centre for Brain Development and Repair, Instem, Bangalore 560065, India.
| |
Collapse
|
16
|
Hou K, Zheng X. A 10-Year Review on Advancements in Identifying and Treating Intellectual Disability Caused by Genetic Variations. Genes (Basel) 2024; 15:1118. [PMID: 39336708 PMCID: PMC11431063 DOI: 10.3390/genes15091118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 08/19/2024] [Accepted: 08/23/2024] [Indexed: 09/30/2024] Open
Abstract
Intellectual disability (ID) is a prevalent neurodevelopmental disorder characterized by neurodevelopmental defects such as the congenital impairment of intellectual function and restricted adaptive behavior. However, genetic studies have been significantly hindered by the extreme clinical and genetic heterogeneity of the subjects under investigation. With the development of gene sequencing technologies, more genetic variations have been discovered, assisting efforts in ID identification and treatment. In this review, the physiological basis of gene variations in ID is systematically explained, the diagnosis and therapy of ID is comprehensively described, and the potential of genetic therapies and exercise therapy in the rehabilitation of individuals with intellectual disabilities are highlighted, offering new perspectives for treatment approaches.
Collapse
Affiliation(s)
- Kexin Hou
- School of Exercise and Health, Shanghai University of Sport, 200 Hengren Road, Yangpu, Shanghai 200438, China
| | - Xinyan Zheng
- School of Exercise and Health, Shanghai University of Sport, 200 Hengren Road, Yangpu, Shanghai 200438, China
| |
Collapse
|
17
|
Wang YZ, Perez-Rosello T, Smukowski SN, Surmeier DJ, Savas JN. Neuron type-specific proteomics reveals distinct Shank3 proteoforms in iSPNs and dSPNs lead to striatal synaptopathy in Shank3B -/- mice. Mol Psychiatry 2024; 29:2372-2388. [PMID: 38486049 PMCID: PMC11412912 DOI: 10.1038/s41380-024-02493-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 02/16/2024] [Accepted: 02/22/2024] [Indexed: 09/21/2024]
Abstract
Combinatorial expression of postsynaptic proteins underlies synapse diversity within and between neuron types. Thus, characterization of neuron-type-specific postsynaptic proteomes is key to obtaining a deeper understanding of discrete synaptic properties and how selective dysfunction manifests in synaptopathies. To overcome the limitations associated with bulk measures of synaptic protein abundance, we developed a biotin proximity protein tagging probe to characterize neuron-type-specific postsynaptic proteomes in vivo. We found Shank3 protein isoforms are differentially expressed by direct and indirect pathway spiny projection neurons (dSPNs and iSPNs). Investigation of Shank3B-/- mice lacking exons 13-16 within the Shank3 gene, reveal distinct Shank3 protein isoform expression in iSPNs and dSPNs. In Shank3B-/- striatum, Shank3E and Shank3NT are expressed by dSPNs but are undetectable in iSPNs. Proteomic analysis indicates significant and selective alterations in the postsynaptic proteome of Shank3B-/- iSPNs. Correspondingly, the deletion of exons 13-16 diminishes dendritic spine density, reduces spine head diameter, and hampers corticostriatal synaptic transmission in iSPNs. Remarkably, reintroducing Shank3E in adult Shank3B-/- iSPNs significantly rectifies the observed dendritic spine morphological and corticostriatal synaptic transmission deficits. We report unexpected cell-type specific synaptic protein isoform expression which could play a key causal role in specifying synapse diversity and selective synapse dysfunction in synaptopathies.
Collapse
Affiliation(s)
- Yi-Zhi Wang
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Tamara Perez-Rosello
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Samuel N Smukowski
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - D James Surmeier
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Jeffrey N Savas
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA.
| |
Collapse
|
18
|
Sumathipala SH, Khan S, Kozol RA, Araki Y, Syed S, Huganir RL, Dallman JE. Context-dependent hyperactivity in syngap1a and syngap1b zebrafish models of SYNGAP1-related disorder. Front Mol Neurosci 2024; 17:1401746. [PMID: 39050824 PMCID: PMC11266194 DOI: 10.3389/fnmol.2024.1401746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 06/04/2024] [Indexed: 07/27/2024] Open
Abstract
Background and aims SYNGAP1-related disorder (SYNGAP1-RD) is a prevalent genetic form of Autism Spectrum Disorder and Intellectual Disability (ASD/ID) and is caused by de novo or inherited mutations in one copy of the SYNGAP1 gene. In addition to ASD/ID, SYNGAP1 disorder is associated with comorbid symptoms including treatment-resistant-epilepsy, sleep disturbances, and gastrointestinal distress. Mechanistic links between these diverse symptoms and SYNGAP1 variants remain obscure, therefore, our goal was to generate a zebrafish model in which this range of symptoms can be studied. Methods We used CRISPR/Cas9 to introduce frameshift mutations in the syngap1a and syngap1b zebrafish duplicates (syngap1ab) and validated these stable models for Syngap1 loss-of-function. Because SYNGAP1 is extensively spliced, we mapped splice variants to the two zebrafish syngap1a and b genes and identified mammalian-like isoforms. We then quantified locomotory behaviors in zebrafish syngap1ab larvae under three conditions that normally evoke different arousal states in wild-type larvae: aversive, high-arousal acoustic, medium-arousal dark, and low-arousal light stimuli. Results We show that CRISPR/Cas9 indels in zebrafish syngap1a and syngap1b produced loss-of-function alleles at RNA and protein levels. Our analyses of zebrafish Syngap1 isoforms showed that, as in mammals, zebrafish Syngap1 N- and C-termini are extensively spliced. We identified a zebrafish syngap1 α1-like variant that maps exclusively to the syngap1b gene. Quantifying locomotor behaviors showed that syngap1ab mutant larvae are hyperactive compared to wild-type but to differing degrees depending on the stimulus. Hyperactivity was most pronounced in low arousal settings, and hyperactivity was proportional to the number of mutant syngap1 alleles. Limitations Syngap1 loss-of-function mutations produce relatively subtle phenotypes in zebrafish compared to mammals. For example, while mouse Syngap1 homozygotes die at birth, zebrafish syngap1ab-/- survive to adulthood and are fertile, thus some aspects of symptoms in people with SYNGAP1-Related Disorder are not likely to be reflected in zebrafish. Conclusion Our data support mutations in zebrafish syngap1ab as causal for hyperactivity associated with elevated arousal that is especially pronounced in low-arousal environments.
Collapse
Affiliation(s)
- Sureni H. Sumathipala
- Department of Biology, University of Miami, Coral Gables, FL, United States
- Department of Biological Sciences, North Carolina State University, Raleigh, NC, United States
| | - Suha Khan
- Department of Biology, University of Miami, Coral Gables, FL, United States
| | - Robert A. Kozol
- Department of Biology, University of Miami, Coral Gables, FL, United States
- Department of Biological Sciences, St. John’s University, Queens, NY, United States
| | - Yoichi Araki
- Department of Neuroscience and Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Sheyum Syed
- Department of Physics, University of Miami, Coral Gables, FL, United States
| | - Richard L. Huganir
- Department of Neuroscience and Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Julia E. Dallman
- Department of Biology, University of Miami, Coral Gables, FL, United States
| |
Collapse
|
19
|
Cesari E, Farini D, Medici V, Ehrmann I, Guerra M, Testa E, Naro C, Geloso MC, Pagliarini V, La Barbera L, D’Amelio M, Orsini T, Vecchioli SF, Tamagnone L, Fort P, Viscomi MT, Elliott DJ, Sette C. Differential expression of paralog RNA binding proteins establishes a dynamic splicing program required for normal cerebral cortex development. Nucleic Acids Res 2024; 52:4167-4184. [PMID: 38324473 PMCID: PMC11077083 DOI: 10.1093/nar/gkae071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 01/17/2024] [Accepted: 01/25/2024] [Indexed: 02/09/2024] Open
Abstract
Sam68 and SLM2 are paralog RNA binding proteins (RBPs) expressed in the cerebral cortex and display similar splicing activities. However, their relative functions during cortical development are unknown. We found that these RBPs exhibit an opposite expression pattern during development. Sam68 expression declines postnatally while SLM2 increases after birth, and this developmental pattern is reinforced by hierarchical control of Sam68 expression by SLM2. Analysis of Sam68:Slm2 double knockout (Sam68:Slm2dko) mice revealed hundreds of exons that respond to joint depletion of these proteins. Moreover, parallel analysis of single and double knockout cortices indicated that exons regulated mainly by SLM2 are characterized by a dynamic splicing pattern during development, whereas Sam68-dependent exons are spliced at relatively constant rates. Dynamic splicing of SLM2-sensitive exons is completely suppressed in the Sam68:Slm2dko developing cortex. Sam68:Slm2dko mice die perinatally with defects in neurogenesis and in neuronal differentiation, and develop a hydrocephalus, consistent with splicing alterations in genes related to these biological processes. Thus, our study reveals that developmental control of separate Sam68 and Slm2 paralog genes encoding homologous RBPs enables the orchestration of a dynamic splicing program needed for brain development and viability, while ensuring a robust redundant mechanism that supports proper cortical development.
Collapse
Affiliation(s)
- Eleonora Cesari
- Department of Neuroscience, Section of Human Anatomy, Catholic University of the Sacred Heart, Largo Francesco Vito 1, 00168 Rome, Italy
- Fondazione Policlinico Agostino Gemelli IRCCS, Largo Agostino Gemelli, 00168 Rome, Italy
| | - Donatella Farini
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy
- Fondazione Santa Lucia IRCCS, Via del Fosso di Fiorano, 64, 00143 Rome, Italy
| | - Vanessa Medici
- Department of Neuroscience, Section of Human Anatomy, Catholic University of the Sacred Heart, Largo Francesco Vito 1, 00168 Rome, Italy
| | - Ingrid Ehrmann
- Newcastle University Centre for Cancer, Newcastle University Institute of Biosciences, Newcastle NE1 3BZ, UK
| | - Marika Guerra
- Department of Neuroscience, Section of Human Anatomy, Catholic University of the Sacred Heart, Largo Francesco Vito 1, 00168 Rome, Italy
| | - Erika Testa
- Department of Life Science and Public Health, Section of Histology and Embryology, Catholic University of the Sacred Heart, Rome
| | - Chiara Naro
- Department of Neuroscience, Section of Human Anatomy, Catholic University of the Sacred Heart, Largo Francesco Vito 1, 00168 Rome, Italy
- Fondazione Policlinico Agostino Gemelli IRCCS, Largo Agostino Gemelli, 00168 Rome, Italy
| | - Maria Concetta Geloso
- Department of Neuroscience, Section of Human Anatomy, Catholic University of the Sacred Heart, Largo Francesco Vito 1, 00168 Rome, Italy
| | - Vittoria Pagliarini
- Department of Neuroscience, Section of Human Anatomy, Catholic University of the Sacred Heart, Largo Francesco Vito 1, 00168 Rome, Italy
- Fondazione Policlinico Agostino Gemelli IRCCS, Largo Agostino Gemelli, 00168 Rome, Italy
| | - Livia La Barbera
- Fondazione Santa Lucia IRCCS, Via del Fosso di Fiorano, 64, 00143 Rome, Italy
| | - Marcello D’Amelio
- Fondazione Santa Lucia IRCCS, Via del Fosso di Fiorano, 64, 00143 Rome, Italy
- Università Campus Bio-Medico di Roma, Via Álvaro del Portillo, 21, 00128 Rome, Italy
| | - Tiziana Orsini
- Institute of Biochemistry and Cell Biology, National Research Council (IBBC/CNR), Monterotondo, 00015 Rome, Italy
| | - Stefano Farioli Vecchioli
- Institute of Biochemistry and Cell Biology, National Research Council (IBBC/CNR), Monterotondo, 00015 Rome, Italy
| | - Luca Tamagnone
- Fondazione Policlinico Agostino Gemelli IRCCS, Largo Agostino Gemelli, 00168 Rome, Italy
- Department of Life Science and Public Health, Section of Histology and Embryology, Catholic University of the Sacred Heart, Rome
| | - Philippe Fort
- Centre de Recherche en Biologie Cellulaire de Montpellier, University of Montpellier, CNRS, 1919 Route de Mende, 34293 Montpellier Cedex 05, France
| | - Maria Teresa Viscomi
- Fondazione Policlinico Agostino Gemelli IRCCS, Largo Agostino Gemelli, 00168 Rome, Italy
- Department of Life Science and Public Health, Section of Histology and Embryology, Catholic University of the Sacred Heart, Rome
| | - David J Elliott
- Newcastle University Centre for Cancer, Newcastle University Institute of Biosciences, Newcastle NE1 3BZ, UK
| | - Claudio Sette
- Department of Neuroscience, Section of Human Anatomy, Catholic University of the Sacred Heart, Largo Francesco Vito 1, 00168 Rome, Italy
- Fondazione Policlinico Agostino Gemelli IRCCS, Largo Agostino Gemelli, 00168 Rome, Italy
| |
Collapse
|
20
|
Shi X, Zhou XZ, Chen G, Luo WF, Zhou C, He TJ, Naik MT, Jiang Q, Marshall J, Cao C. Targeting the postsynaptic scaffolding protein PSD-95 enhances BDNF signaling to mitigate depression-like behaviors in mice. Sci Signal 2024; 17:eadn4556. [PMID: 38687826 PMCID: PMC11223518 DOI: 10.1126/scisignal.adn4556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 04/10/2024] [Indexed: 05/02/2024]
Abstract
Signaling mediated by brain-derived neurotrophic factor (BDNF), which is supported by the postsynaptic scaffolding protein PSD-95, has antidepressant effects. Conversely, clinical depression is associated with reduced BDNF signaling. We found that peptidomimetic compounds that bind to PSD-95 promoted signaling by the BDNF receptor TrkB in the hippocampus and reduced depression-like behaviors in mice. The compounds CN2097 and Syn3 both bind to the PDZ3 domain of PSD-95, and Syn3 also binds to an α-helical region of the protein. Syn3 reduced depression-like behaviors in two mouse models of stress-induced depression; CN2097 had similar but less potent effects. In hippocampal neurons, application of Syn3 enhanced the formation of TrkB-Gαi1/3-PSD-95 complexes and potentiated downstream PI3K-Akt-mTOR signaling. In mice subjected to chronic mild stress (CMS), systemic administration of Syn3 reversed the CMS-induced, depression-associated changes in PI3K-Akt-mTOR signaling, dendrite complexity, spine density, and autophagy in the hippocampus and reduced depression-like behaviors. Knocking out Gαi1/3 in hippocampal neurons prevented the therapeutic effects of Syn3, indicating dependence of these effects on the TrkB pathway. The findings suggest that compounds that induce the formation of PSD-95-TrkB complexes have therapeutic potential to alleviate depression.
Collapse
Affiliation(s)
- Xin Shi
- Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Institution of Neuroscience, Soochow University, Suzhou 215123, China
| | - Xiao-zhong Zhou
- Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Institution of Neuroscience, Soochow University, Suzhou 215123, China
- Department of Orthopedics, the Second Affiliated Hospital of Soochow University, Suzhou, 215004, Jiangsu, China
| | - Gang Chen
- Department of Neurosurgery, the First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China
| | - Wei-feng Luo
- Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Institution of Neuroscience, Soochow University, Suzhou 215123, China
| | - Chengyu Zhou
- Department of Neuroscience, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Tian-ju He
- Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Institution of Neuroscience, Soochow University, Suzhou 215123, China
| | - Mandar T. Naik
- Department of Molecular Biology, Cell Biology & Biochemistry, Brown University, Providence, RI 02912, USA
| | - Qin Jiang
- The Fourth School of Clinical Medicine, Nanjing Medical University, Nanjing 210029, China
| | - John Marshall
- Department of Molecular Biology, Cell Biology & Biochemistry, Brown University, Providence, RI 02912, USA
| | - Cong Cao
- Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Institution of Neuroscience, Soochow University, Suzhou 215123, China
| |
Collapse
|
21
|
Fenton TA, Haouchine OY, Hallam EL, Smith EM, Jackson KC, Rahbarian D, Canales C, Adhikari A, Nord AS, Ben-Shalom R, Silverman JL. Hyperexcitability and translational phenotypes in a preclinical mouse model of SYNGAP1-Related Intellectual Disability. RESEARCH SQUARE 2024:rs.3.rs-4067746. [PMID: 38562838 PMCID: PMC10984035 DOI: 10.21203/rs.3.rs-4067746/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Disruption of SYNGAP1 directly causes a genetically identifiable neurodevelopmental disorder (NDD) called SYNGAP1-related intellectual disability (SRID). Without functional SynGAP1 protein, individuals are developmentally delayed and have prominent features of intellectual disability, motor impairments, and epilepsy. Over the past two decades, there have been numerous discoveries indicting the critical role of Syngap1. Several rodent models with a loss of Syngap1 have been engineered identifying precise roles in neuronal structure and function, as well as key biochemical pathways key for synapse integrity. Homozygous loss of SYNGAP1/Syngap1 is lethal. Heterozygous mutations of Syngap1 result in a broad range of behavioral phenotypes. Our in vivo functional data, using the original mouse model from the Huganir laboratory, corroborated behaviors including robust hyperactivity and deficits in learning and memory in young adults. Furthermore, we described impairments in the domain of sleep, characterized using neurophysiological data collected with wireless, telemetric electroencephalography (EEG). Syngap1+/- mice exhibited elevated spiking events and spike trains, in addition to elevated power, most notably in the delta power frequency. For the first time, we illustrated primary neurons from Syngap1+/- mice displayed increased network firing activity, greater bursts, and shorter inter-burst intervals between peaks by employing high density microelectrode arrays (HD-MEA). Our work bridges in-vitro electrophysiological neuronal activity and function with in vivo neurophysiological brain activity and function. These data elucidate quantitative, translational biomarkers in vivo and in vitro that can be utilized for the development and efficacy assessment of targeted treatments for SRID.
Collapse
Affiliation(s)
- Timothy A Fenton
- MIND Institute, University of California Davis School of Medicine, Sacramento, CA 95817
- Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, CA 95817
| | - Olivia Y Haouchine
- MIND Institute, University of California Davis School of Medicine, Sacramento, CA 95817
- Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, CA 95817
| | - Elizabeth L Hallam
- MIND Institute, University of California Davis School of Medicine, Sacramento, CA 95817
- Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, CA 95817
| | - Emily M Smith
- UC Davis Center for Neuroscience; Department of Psychiatry and Behavioral Sciences & Department of Neurobiology, Physiology and Behavior, University of California Davis, Davis, CA 95616
| | - Kiya C. Jackson
- UC Davis Center for Neuroscience; Department of Psychiatry and Behavioral Sciences & Department of Neurobiology, Physiology and Behavior, University of California Davis, Davis, CA 95616
| | - Darlene Rahbarian
- UC Davis Center for Neuroscience; Department of Psychiatry and Behavioral Sciences & Department of Neurobiology, Physiology and Behavior, University of California Davis, Davis, CA 95616
| | - Cesar Canales
- Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, CA 95817
- UC Davis Center for Neuroscience; Department of Psychiatry and Behavioral Sciences & Department of Neurobiology, Physiology and Behavior, University of California Davis, Davis, CA 95616
| | - Anna Adhikari
- MIND Institute, University of California Davis School of Medicine, Sacramento, CA 95817
- Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, CA 95817
| | - Alexander S. Nord
- MIND Institute, University of California Davis School of Medicine, Sacramento, CA 95817
- Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, CA 95817
- UC Davis Center for Neuroscience; Department of Psychiatry and Behavioral Sciences & Department of Neurobiology, Physiology and Behavior, University of California Davis, Davis, CA 95616
| | - Roy Ben-Shalom
- MIND Institute, University of California Davis School of Medicine, Sacramento, CA 95817
- Department of Neurology, University of California Davis School of Medicine, Sacramento, CA 95817
| | - Jill L Silverman
- MIND Institute, University of California Davis School of Medicine, Sacramento, CA 95817
- Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, CA 95817
| |
Collapse
|
22
|
Yook Y, Lee KY, Kim E, Lizarazo S, Yu X, Tsai NP. Hyperfunction of post-synaptic density protein 95 promotes seizure response in early-stage aβ pathology. EMBO Rep 2024; 25:1233-1255. [PMID: 38413732 PMCID: PMC10933348 DOI: 10.1038/s44319-024-00090-0] [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: 05/30/2023] [Revised: 01/24/2024] [Accepted: 01/30/2024] [Indexed: 02/29/2024] Open
Abstract
Accumulation of amyloid-beta (Aβ) can lead to the formation of aggregates that contribute to neurodegeneration in Alzheimer's disease (AD). Despite globally reduced neural activity during AD onset, recent studies have suggested that Aβ induces hyperexcitability and seizure-like activity during the early stages of the disease that ultimately exacerbate cognitive decline. However, the underlying mechanism is unknown. Here, we reveal an Aβ-induced elevation of postsynaptic density protein 95 (PSD-95) in cultured neurons in vitro and in an in vivo AD model using APP/PS1 mice at 8 weeks of age. Elevation of PSD-95 occurs as a result of reduced ubiquitination caused by Akt-dependent phosphorylation of E3 ubiquitin ligase murine-double-minute 2 (Mdm2). The elevation of PSD-95 is consistent with the facilitation of excitatory synapses and the surface expression of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors induced by Aβ. Inhibition of PSD-95 corrects these Aβ-induced synaptic defects and reduces seizure activity in APP/PS1 mice. Our results demonstrate a mechanism underlying elevated seizure activity during early-stage Aβ pathology and suggest that PSD-95 could be an early biomarker and novel therapeutic target for AD.
Collapse
Affiliation(s)
- Yeeun Yook
- Department of Molecular and Integrative Physiology, School of Molecular and Cellular Biology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Kwan Young Lee
- Department of Molecular and Integrative Physiology, School of Molecular and Cellular Biology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Eunyoung Kim
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Simon Lizarazo
- Department of Molecular and Integrative Physiology, School of Molecular and Cellular Biology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Xinzhu Yu
- Department of Molecular and Integrative Physiology, School of Molecular and Cellular Biology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Nien-Pei Tsai
- Department of Molecular and Integrative Physiology, School of Molecular and Cellular Biology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA.
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA.
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA.
| |
Collapse
|
23
|
Araki Y, Rajkovich KE, Gerber EE, Gamache TR, Johnson RC, Tran THN, Liu B, Zhu Q, Hong I, Kirkwood A, Huganir R. SynGAP regulates synaptic plasticity and cognition independently of its catalytic activity. Science 2024; 383:eadk1291. [PMID: 38422154 PMCID: PMC11188940 DOI: 10.1126/science.adk1291] [Citation(s) in RCA: 28] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 12/28/2023] [Indexed: 03/02/2024]
Abstract
SynGAP is an abundant synaptic GTPase-activating protein (GAP) critical for synaptic plasticity, learning, memory, and cognition. Mutations in SYNGAP1 in humans result in intellectual disability, autistic-like behaviors, and epilepsy. Heterozygous Syngap1-knockout mice display deficits in synaptic plasticity, learning, and memory and exhibit seizures. It is unclear whether SynGAP imparts structural properties at synapses independently of its GAP activity. Here, we report that inactivating mutations within the GAP domain do not inhibit synaptic plasticity or cause behavioral deficits. Instead, SynGAP modulates synaptic strength by physically competing with the AMPA-receptor-TARP excitatory receptor complex in the formation of molecular condensates with synaptic scaffolding proteins. These results have major implications for developing therapeutic treatments for SYNGAP1-related neurodevelopmental disorders.
Collapse
Affiliation(s)
| | | | | | | | - Richard C. Johnson
- Department of Neuroscience, Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Thanh Hai N. Tran
- Department of Neuroscience, Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Bian Liu
- Department of Neuroscience, Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Qianwen Zhu
- Department of Neuroscience, Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Ingie Hong
- Department of Neuroscience, Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Alfredo Kirkwood
- Department of Neuroscience, Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Richard Huganir
- Department of Neuroscience, Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
24
|
Choquet D. Shifting rules in a brain disorder. Science 2024; 383:950-951. [PMID: 38422158 DOI: 10.1126/science.adn8707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/02/2024]
Abstract
The mode of action of a synaptic protein is challenged.
Collapse
Affiliation(s)
- Daniel Choquet
- Interdisciplinary Institute for Neuroscience, University of Bordeaux, CNRS, UMR 5297, F-33000 Bordeaux, France
- Bordeaux Imaging Center, University of Bordeaux, CNRS, INSERM, US4, UAR 3420, F-33000 Bordeaux, France
| |
Collapse
|
25
|
Birtele M, Del Dosso A, Xu T, Nguyen T, Wilkinson B, Hosseini N, Nguyen S, Urenda JP, Knight G, Rojas C, Flores I, Atamian A, Moore R, Sharma R, Pirrotte P, Ashton RS, Huang EJ, Rumbaugh G, Coba MP, Quadrato G. Non-synaptic function of the autism spectrum disorder-associated gene SYNGAP1 in cortical neurogenesis. Nat Neurosci 2023; 26:2090-2103. [PMID: 37946050 PMCID: PMC11349286 DOI: 10.1038/s41593-023-01477-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 09/29/2023] [Indexed: 11/12/2023]
Abstract
Genes involved in synaptic function are enriched among those with autism spectrum disorder (ASD)-associated rare genetic variants. Dysregulated cortical neurogenesis has been implicated as a convergent mechanism in ASD pathophysiology, yet it remains unknown how 'synaptic' ASD risk genes contribute to these phenotypes, which arise before synaptogenesis. Here, we show that the synaptic Ras GTPase-activating (RASGAP) protein 1 (SYNGAP1, a top ASD risk gene) is expressed within the apical domain of human radial glia cells (hRGCs). In a human cortical organoid model of SYNGAP1 haploinsufficiency, we find dysregulated cytoskeletal dynamics that impair the scaffolding and division plane of hRGCs, resulting in disrupted lamination and accelerated maturation of cortical projection neurons. Additionally, we confirmed an imbalance in the ratio of progenitors to neurons in a mouse model of Syngap1 haploinsufficiency. Thus, SYNGAP1-related brain disorders may arise through non-synaptic mechanisms, highlighting the need to study genes associated with neurodevelopmental disorders (NDDs) in diverse human cell types and developmental stages.
Collapse
Affiliation(s)
- Marcella Birtele
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research at USC, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Ashley Del Dosso
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research at USC, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Tiantian Xu
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research at USC, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Xiangya Hospital, Central South University, Changsha, China
| | - Tuan Nguyen
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research at USC, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Brent Wilkinson
- Department of Psychiatry and Behavioral Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Negar Hosseini
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research at USC, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Sarah Nguyen
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research at USC, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Jean-Paul Urenda
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research at USC, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Gavin Knight
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI, USA
| | - Camilo Rojas
- Departments of Neuroscience and Molecular Medicine, University of Florida Scripps Biomedical Research Institute, Jupiter, FL, USA
- Skaggs Graduate School of Chemical and Biological Sciences, Scripps Research, Jupiter, FL, USA
| | - Ilse Flores
- Department of Psychiatry and Behavioral Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Alexander Atamian
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research at USC, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Roger Moore
- Integrated Mass Spectrometry Shared Resource, City of Hope Comprehensive Cancer Center, Duarte, CA, USA
| | - Ritin Sharma
- Integrated Mass Spectrometry Shared Resource, City of Hope Comprehensive Cancer Center, Duarte, CA, USA
- Cancer and Cell Biology Division, Translational Genomics Research Institute, Phoenix, AZ, USA
| | - Patrick Pirrotte
- Integrated Mass Spectrometry Shared Resource, City of Hope Comprehensive Cancer Center, Duarte, CA, USA
- Cancer and Cell Biology Division, Translational Genomics Research Institute, Phoenix, AZ, USA
| | - Randolph S Ashton
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI, USA
| | - Eric J Huang
- Department of Pathology, University of California, San Francisco, CA, USA
- Eli and Edythe Broad Institute for Stem Cell Research and Regeneration Medicine, University of California, San Francisco, CA, USA
| | - Gavin Rumbaugh
- Departments of Neuroscience and Molecular Medicine, University of Florida Scripps Biomedical Research Institute, Jupiter, FL, USA
- Skaggs Graduate School of Chemical and Biological Sciences, Scripps Research, Jupiter, FL, USA
| | - Marcelo P Coba
- Department of Psychiatry and Behavioral Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Giorgia Quadrato
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
- Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research at USC, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
26
|
Michel JC, Grivette MMB, Harshfield AT, Huynh L, Komons AP, Loomis B, McKinnis K, Miller BT, Nguyen EQ, Huang TW, Lauf S, Michel ES, Michel ME, Kissinger JS, Marsh AJ, Crow WE, Kaye LE, Lasseigne AM, Lukowicz-Bedford RM, Farnsworth DR, Martin EA, Miller AC. Electrical synapse structure requires distinct isoforms of a postsynaptic scaffold. PLoS Genet 2023; 19:e1011045. [PMID: 38011265 PMCID: PMC10703405 DOI: 10.1371/journal.pgen.1011045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 12/07/2023] [Accepted: 11/01/2023] [Indexed: 11/29/2023] Open
Abstract
Electrical synapses are neuronal gap junction (GJ) channels associated with a macromolecular complex called the electrical synapse density (ESD), which regulates development and dynamically modifies electrical transmission. However, the proteomic makeup and molecular mechanisms utilized by the ESD that direct electrical synapse formation are not well understood. Using the Mauthner cell of zebrafish as a model, we previously found that the intracellular scaffolding protein ZO1b is a member of the ESD, localizing postsynaptically, where it is required for GJ channel localization, electrical communication, neural network function, and behavior. Here, we show that the complexity of the ESD is further diversified by the genomic structure of the ZO1b gene locus. The ZO1b gene is alternatively initiated at three transcriptional start sites resulting in isoforms with unique N-termini that we call ZO1b-Alpha, -Beta, and -Gamma. We demonstrate that ZO1b-Beta and ZO1b-Gamma are broadly expressed throughout the nervous system and localize to electrical synapses. By contrast, ZO1b-Alpha is expressed mainly non-neuronally and is not found at synapses. We generate mutants in all individual isoforms, as well as double mutant combinations in cis on individual chromosomes, and find that ZO1b-Beta is necessary and sufficient for robust GJ channel localization. ZO1b-Gamma, despite its localization to the synapse, plays an auxiliary role in channel localization. This study expands the notion of molecular complexity at the ESD, revealing that an individual genomic locus can contribute distinct isoforms to the macromolecular complex at electrical synapses. Further, independent scaffold isoforms have differential contributions to developmental assembly of the interneuronal GJ channels. We propose that ESD molecular complexity arises both from the diversity of unique genes and from distinct isoforms encoded by single genes. Overall, ESD proteomic diversity is expected to have critical impacts on the development, structure, function, and plasticity of electrical transmission.
Collapse
Affiliation(s)
- Jennifer Carlisle Michel
- Institute of Neuroscience, Department of Biology, University of Oregon, Eugene, Oregon, United States of America
| | - Margaret M. B. Grivette
- Institute of Neuroscience, Department of Biology, University of Oregon, Eugene, Oregon, United States of America
| | - Amber T. Harshfield
- Institute of Neuroscience, Department of Biology, University of Oregon, Eugene, Oregon, United States of America
| | - Lisa Huynh
- Institute of Neuroscience, Department of Biology, University of Oregon, Eugene, Oregon, United States of America
| | - Ava P. Komons
- Institute of Neuroscience, Department of Biology, University of Oregon, Eugene, Oregon, United States of America
| | - Bradley Loomis
- Institute of Neuroscience, Department of Biology, University of Oregon, Eugene, Oregon, United States of America
| | - Kaitlan McKinnis
- Institute of Neuroscience, Department of Biology, University of Oregon, Eugene, Oregon, United States of America
| | - Brennen T. Miller
- Institute of Neuroscience, Department of Biology, University of Oregon, Eugene, Oregon, United States of America
| | - Ethan Q. Nguyen
- Institute of Neuroscience, Department of Biology, University of Oregon, Eugene, Oregon, United States of America
| | - Tiffany W. Huang
- Institute of Neuroscience, Department of Biology, University of Oregon, Eugene, Oregon, United States of America
| | - Sophia Lauf
- Institute of Neuroscience, Department of Biology, University of Oregon, Eugene, Oregon, United States of America
| | - Elias S. Michel
- Institute of Neuroscience, Department of Biology, University of Oregon, Eugene, Oregon, United States of America
| | - Mia E. Michel
- Institute of Neuroscience, Department of Biology, University of Oregon, Eugene, Oregon, United States of America
| | - Jane S. Kissinger
- Institute of Neuroscience, Department of Biology, University of Oregon, Eugene, Oregon, United States of America
| | - Audrey J. Marsh
- Institute of Neuroscience, Department of Biology, University of Oregon, Eugene, Oregon, United States of America
| | - William E. Crow
- Institute of Neuroscience, Department of Biology, University of Oregon, Eugene, Oregon, United States of America
| | - Lila E. Kaye
- Institute of Neuroscience, Department of Biology, University of Oregon, Eugene, Oregon, United States of America
| | - Abagael M. Lasseigne
- Institute of Neuroscience, Department of Biology, University of Oregon, Eugene, Oregon, United States of America
| | - Rachel M. Lukowicz-Bedford
- Institute of Neuroscience, Department of Biology, University of Oregon, Eugene, Oregon, United States of America
| | - Dylan R. Farnsworth
- Institute of Neuroscience, Department of Biology, University of Oregon, Eugene, Oregon, United States of America
| | - E. Anne Martin
- Institute of Neuroscience, Department of Biology, University of Oregon, Eugene, Oregon, United States of America
| | - Adam C. Miller
- Institute of Neuroscience, Department of Biology, University of Oregon, Eugene, Oregon, United States of America
| |
Collapse
|
27
|
Sumathipala SH, Khan S, Kozol RA, Araki Y, Syed S, Huganir RL, Dallman JE. Context-dependent hyperactivity in syngap1a and syngap1b zebrafish autism models. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.20.557316. [PMID: 37786701 PMCID: PMC10541574 DOI: 10.1101/2023.09.20.557316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/04/2023]
Abstract
Background and Aims SYNGAP1 disorder is a prevalent genetic form of Autism Spectrum Disorder and Intellectual Disability (ASD/ID) and is caused by de novo or inherited mutations in one copy of the SYNGAP1 gene. In addition to ASD/ID, SYNGAP1 disorder is associated with comorbid symptoms including treatment-resistant-epilepsy, sleep disturbances, and gastrointestinal distress. Mechanistic links between these diverse symptoms and SYNGAP1 variants remain obscure, therefore, our goal was to generate a zebrafish model in which this range of symptoms can be studied. Methods We used CRISPR/Cas9 to introduce frameshift mutations in the syngap1a and syngap1b zebrafish duplicates (syngap1ab) and validated these stable models for Syngap1 loss-of-function. Because SYNGAP1 is extensively spliced, we mapped splice variants to the two zebrafish syngap1a and b genes and identified mammalian-like isoforms. We then quantified locomotory behaviors in zebrafish syngap1ab larvae under three conditions that normally evoke different arousal states in wild type larvae: aversive, high-arousal acoustic, medium-arousal dark, and low-arousal light stimuli. Results We show that CRISPR/Cas9 indels in zebrafish syngap1a and syngap1b produced loss-of-function alleles at RNA and protein levels. Our analyses of zebrafish Syngap1 isoforms showed that, as in mammals, zebrafish Syngap1 N- and C-termini are extensively spliced. We identified a zebrafish syngap1 α1-like variant that maps exclusively to the syngap1b gene. Quantifying locomotor behaviors showed that syngap1ab larvae are hyperactive compared to wild type but to differing degrees depending on the stimulus. Hyperactivity was most pronounced in low arousal settings, with overall movement increasing with the number of mutant syngap1 alleles. Conclusions Our data support mutations in zebrafish syngap1ab as causal for hyperactivity associated with elevated arousal that is especially pronounced in low-arousal environments.
Collapse
Affiliation(s)
- Sureni H. Sumathipala
- Department of Biology, University of Miami, Coral Gables, FL USA
- Department of Biological Sciences, North Carolina State University, Raleigh, NC, USA
| | - Suha Khan
- Department of Biology, University of Miami, Coral Gables, FL USA
| | - Robert A. Kozol
- Department of Biology, University of Miami, Coral Gables, FL USA
- Jupiter Life Science Initiative, Florida Atlantic University, Jupiter FL, USA
| | - Yoichi Araki
- Department of Neuroscience and Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, MD USA
| | - Sheyum Syed
- Department of Physics, University of Miami, Coral Gables, FL USA
| | - Richard L. Huganir
- Department of Neuroscience and Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, MD USA
| | - Julia E. Dallman
- Department of Biology, University of Miami, Coral Gables, FL USA
| |
Collapse
|
28
|
Silverman JL, Fenton T, Haouchine O, Hallam E, Smith E, Jackson K, Rahbarian D, Canales C, Adhikari A, Nord A, Ben-Shalom R. Hyperexcitability and translational phenotypes in a preclinical model of SYNGAP1 mutations. RESEARCH SQUARE 2023:rs.3.rs-3246655. [PMID: 37790402 PMCID: PMC10543290 DOI: 10.21203/rs.3.rs-3246655/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/05/2023]
Abstract
SYNGAP1 is a critical gene for neuronal development, synaptic structure, and function. Although rare, the disruption of SYNGAP1 directly causes a genetically identifiable neurodevelopmental disorder (NDD) called SYNGAP1 -related intellectual disability. Without functional SynGAP1 protein, patients present with intellectual disability, motor impairments, and epilepsy. Previous work using mouse models with a variety of germline and conditional mutations has helped delineate SynGAP1's critical roles in neuronal structure and function, as well as key biochemical signaling pathways essential to synapse integrity. Homozygous loss of SYNGAP1 is embryonically lethal. Heterozygous mutations of SynGAP1 result in a broad range of phenotypes including increased locomotor activity, impaired working spatial memory, impaired cued fear memory, and increased stereotypic behavior. Our in vivo functional data, using the original germline mutation mouse line from the Huganir laboratory, corroborated robust hyperactivity and learning and memory deficits. Here, we describe impairments in the translational biomarker domain of sleep, characterized using neurophysiological data collected with wireless telemetric electroencephalography (EEG). We discovered Syngap1+/- mice exhibited elevated spike trains in both number and duration, in addition to elevated power, most notably in the delta power band. Primary neurons from Syngap1+/- mice displayed increased network firing activity, greater spikes per burst, and shorter inter-burst intervals between peaks using high density micro-electrode arrays (HD-MEA). This work is translational, innovative, and highly significant as it outlines functional impairments in Syngap1 mutant mice. Simultaneously, the work utilized untethered, wireless neurophysiology that can discover potential biomarkers of Syngap1 RI-D, for clinical trials, as it has done with other NDDs. Our work is substantial forward progress toward translational work for SynGAP1R-ID as it bridges in-vitro electrophysiological neuronal activity and function with in vivo neurophysiological brain activity and function. These data elucidate multiple quantitative, translational biomarkers in vivo and in vitro for the development of treatments for SYNGAP1-related intellectual disability.
Collapse
Affiliation(s)
- Jill L Silverman
- MIND Institute and Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Araki Y, Gerber EE, Rajkovich KE, Hong I, Johnson RC, Lee HK, Kirkwood A, Huganir RL. Mouse models of SYNGAP1-related intellectual disability. Proc Natl Acad Sci U S A 2023; 120:e2308891120. [PMID: 37669379 PMCID: PMC10500186 DOI: 10.1073/pnas.2308891120] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Accepted: 07/28/2023] [Indexed: 09/07/2023] Open
Abstract
SYNGAP1 is a Ras-GTPase-activating protein highly enriched at excitatory synapses in the brain. De novo loss-of-function mutations in SYNGAP1 are a major cause of genetically defined neurodevelopmental disorders (NDDs). These mutations are highly penetrant and cause SYNGAP1-related intellectual disability (SRID), an NDD characterized by cognitive impairment, social deficits, early-onset seizures, and sleep disturbances. Studies in rodent neurons have shown that Syngap1 regulates developing excitatory synapse structure and function, and heterozygous Syngap1 knockout mice have deficits in synaptic plasticity, learning, and memory and have seizures. However, how specific SYNGAP1 mutations found in humans lead to disease has not been investigated in vivo. To explore this, we utilized the CRISPR-Cas9 system to generate knock-in mouse models with two distinct known causal variants of SRID: one with a frameshift mutation leading to a premature stop codon, SYNGAP1; L813RfsX22, and a second with a single-nucleotide mutation in an intron that creates a cryptic splice acceptor site leading to premature stop codon, SYNGAP1; c.3583-9G>A. While reduction in Syngap1 mRNA varies from 30 to 50% depending on the specific mutation, both models show ~50% reduction in Syngap1 protein, have deficits in synaptic plasticity, and recapitulate key features of SRID including hyperactivity and impaired working memory. These data suggest that half the amount of SYNGAP1 protein is key to the pathogenesis of SRID. These results provide a resource to study SRID and establish a framework for the development of therapeutic strategies for this disorder.
Collapse
Affiliation(s)
- Yoichi Araki
- Department of Neuroscience, Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, MD21205
| | - Elizabeth E. Gerber
- Department of Neuroscience, Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, MD21205
| | - Kacey E. Rajkovich
- Department of Neuroscience, Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, MD21205
| | - Ingie Hong
- Department of Neuroscience, Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, MD21205
| | - Richard C. Johnson
- Department of Neuroscience, Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, MD21205
| | - Hey-Kyoung Lee
- Department of Neuroscience, Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, MD21205
| | - Alfredo Kirkwood
- Department of Neuroscience, Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, MD21205
| | - Richard L. Huganir
- Department of Neuroscience, Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, MD21205
| |
Collapse
|
30
|
Zhao M, Kwon SE. Interneuron-Targeted Disruption of SYNGAP1 Alters Sensory Representations in the Neocortex and Impairs Sensory Learning. J Neurosci 2023; 43:6212-6226. [PMID: 37558489 PMCID: PMC10476640 DOI: 10.1523/jneurosci.1997-22.2023] [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: 10/25/2022] [Revised: 07/18/2023] [Accepted: 07/20/2023] [Indexed: 08/11/2023] Open
Abstract
SYNGAP1 haploinsufficiency in humans leads to severe neurodevelopmental disorders characterized by intellectual disability, autism, epilepsy, and sensory processing deficits. However, the circuit mechanisms underlying these disorders are not well understood. In mice, a decrease of SynGAP levels results in cognitive deficits by interfering with the development of excitatory glutamatergic connections. Recent evidence suggests that SynGAP also plays a crucial role in the development and function of GABAergic inhibitory interneurons. Nevertheless, it remains uncertain whether and to what extent the expression of SYNGAP1 in inhibitory interneurons contributes to cortical circuit function and related behaviors. The activity of cortical neurons has not been measured simultaneously with behavior. To address these gaps, we recorded from layer 2/3 neurons in the primary whisker somatosensory cortex (wS1) of mice while they learned to perform a whisker tactile detection task. Our results demonstrate that mice with interneuron-specific SYNGAP1 haploinsufficiency exhibit learning deficits characterized by heightened behavioral responses in the absence of relevant sensory input and premature responses to unrelated sensory stimuli not associated with reward acquisition. These behavioral deficits are accompanied by specific circuit abnormalities within wS1. Interneuron-specific SYNGAP1 haploinsufficiency increases detrimental neuronal correlations directly related to task performance and enhances responses to irrelevant sensory stimuli unrelated to the reward acquisition. In summary, our findings indicate that a reduction of SynGAP in inhibitory interneurons impairs sensory representation in the primary sensory cortex by disrupting neuronal correlations, which likely contributes to the observed cognitive deficits in mice with pan-neuronal SYNGAP1 haploinsufficiency.SIGNIFICANCE STATEMENT SYNGAP1 haploinsufficiency leads to severe neurodevelopmental disorders. The exact nature of neural circuit dysfunction caused by SYNGAP1 haploinsufficiency remains poorly understood. SynGAP plays a critical role in the function of GABAergic inhibitory interneurons as well as glutamatergic pyramidal neurons in the neocortex. Whether and how decreasing SYNGAP1 level in inhibitory interneurons disrupts a behaviorally relevant circuit remains unclear. We measure neural activity and behavior in mice learning a perceptual task. Mice with interneuron-targeted disruption of SYNGAP1 display increased detrimental neuronal correlations and elevated responses to irrelevant sensory inputs, which are related to impaired task performance. These results show that cortical interneuron dysfunction contributes to sensory deficits in SYNGAP1 haploinsufficiency with important implications for identifying therapeutic targets.
Collapse
Affiliation(s)
- Meiling Zhao
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, Michigan 48109
| | - Sung Eun Kwon
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, Michigan 48109
| |
Collapse
|
31
|
Cho H, Yoo T, Moon H, Kang H, Yang Y, Kang M, Yang E, Lee D, Hwang D, Kim H, Kim D, Kim JY, Kim E. Adnp-mutant mice with cognitive inflexibility, CaMKIIα hyperactivity, and synaptic plasticity deficits. Mol Psychiatry 2023; 28:3548-3562. [PMID: 37365244 PMCID: PMC10618100 DOI: 10.1038/s41380-023-02129-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 05/14/2023] [Accepted: 06/13/2023] [Indexed: 06/28/2023]
Abstract
ADNP syndrome, involving the ADNP transcription factor of the SWI/SNF chromatin-remodeling complex, is characterized by developmental delay, intellectual disability, and autism spectrum disorders (ASD). Although Adnp-haploinsufficient (Adnp-HT) mice display various phenotypic deficits, whether these mice display abnormal synaptic functions remain poorly understood. Here, we report synaptic plasticity deficits associated with cognitive inflexibility and CaMKIIα hyperactivity in Adnp-HT mice. These mice show impaired and inflexible contextual learning and memory, additional to social deficits, long after the juvenile-stage decrease of ADNP protein levels to ~10% of the newborn level. The adult Adnp-HT hippocampus shows hyperphosphorylated CaMKIIα and its substrates, including SynGAP1, and excessive long-term potentiation that is normalized by CaMKIIα inhibition. Therefore, Adnp haploinsufficiency in mice leads to cognitive inflexibility involving CaMKIIα hyperphosphorylation and excessive LTP in adults long after its marked expressional decrease in juveniles.
Collapse
Affiliation(s)
- Heejin Cho
- Department of Biological Sciences, Korea Advanced Institute for Science and Technology (KAIST), Daejeon, 34141, Korea
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science (IBS), Daejeon, 34141, Korea
| | - Taesun Yoo
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science (IBS), Daejeon, 34141, Korea
| | - Heera Moon
- Department of Biological Sciences, Korea Advanced Institute for Science and Technology (KAIST), Daejeon, 34141, Korea
| | - Hyojin Kang
- Division of National Supercomputing, Korea Institute of Science and Technology Information, Daejeon, 34141, Korea
| | - Yeji Yang
- Department of Biological Sciences, Korea Advanced Institute for Science and Technology (KAIST), Daejeon, 34141, Korea
- Research Center for Bioconvergence Analysis, Korea Basic Science Institute, 162 Yeongudanjiro, Ochang, Cheongju, Chungbuk, 28119, Korea
| | - MinSoung Kang
- Therapeutics & Biotechnology Division, Drug discovery platform research center, Korea Research Institute of Chemical Technology (KRICT), Daejeon, 34114, Korea
| | - Esther Yang
- Department of Anatomy and BK21 Graduate Program, Biomedical Sciences, College of Medicine, Korea University, Seoul, 02841, Korea
| | - Dowoon Lee
- School of Biological Sciences, Seoul National University, Seoul, 08826, Korea
| | - Daehee Hwang
- School of Biological Sciences, Seoul National University, Seoul, 08826, Korea
| | - Hyun Kim
- Department of Anatomy and BK21 Graduate Program, Biomedical Sciences, College of Medicine, Korea University, Seoul, 02841, Korea
| | - Doyoun Kim
- Therapeutics & Biotechnology Division, Drug discovery platform research center, Korea Research Institute of Chemical Technology (KRICT), Daejeon, 34114, Korea
- Medicinal Chemistry and Pharmacology, Korea University of Science and Technology (UST), Daejeon, 34113, Korea
| | - Jin Young Kim
- Research Center for Bioconvergence Analysis, Korea Basic Science Institute, 162 Yeongudanjiro, Ochang, Cheongju, Chungbuk, 28119, Korea
| | - Eunjoon Kim
- Department of Biological Sciences, Korea Advanced Institute for Science and Technology (KAIST), Daejeon, 34141, Korea.
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science (IBS), Daejeon, 34141, Korea.
| |
Collapse
|
32
|
Fenton TA, Haouchine OY, Hallam EL, Smith EM, Jackson KC, Rahbarian D, Canales C, Adhikari A, Nord AS, Ben-Shalom R, Silverman JL. Hyperexcitability and translational phenotypes in a preclinical model of SYNGAP1 mutations. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.24.550093. [PMID: 37546838 PMCID: PMC10402099 DOI: 10.1101/2023.07.24.550093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
Abstract
SYNGAP1 is a critical gene for neuronal development, synaptic structure, and function. Although rare, the disruption of SYNGAP1 directly causes a genetically identifiable neurodevelopmental disorder (NDD) called SYNGAP1-related intellectual disability. Without functional SynGAP1 protein, patients present with intellectual disability, motor impairments, and epilepsy. Previous work using mouse models with a variety of germline and conditional mutations has helped delineate SynGAP1's critical roles in neuronal structure and function, as well as key biochemical signaling pathways essential to synapse integrity. Homozygous loss of SYNGAP1 is embryonically lethal. Heterozygous mutations of SynGAP1 result in a broad range of phenotypes including increased locomotor activity, impaired working spatial memory, impaired cued fear memory, and increased stereotypic behavior. Our in vivo functional data, using the original germline mutation mouse line from the Huganir laboratory, corroborated robust hyperactivity and learning and memory deficits. Here, we describe impairments in the translational biomarker domain of sleep, characterized using neurophysiological data collected with wireless telemetric electroencephalography (EEG). We discovered Syngap1 +/- mice exhibited elevated spike trains in both number and duration, in addition to elevated power, most notably in the delta power band. Primary neurons from Syngap1 +/- mice displayed increased network firing activity, greater spikes per burst, and shorter inter-burst intervals between peaks using high density micro-electrode arrays (HD-MEA). This work is translational, innovative, and highly significant as it outlines functional impairments in Syngap1 mutant mice. Simultaneously, the work utilized untethered, wireless neurophysiology that can discover potential biomarkers of Syngap1R-ID, for clinical trials, as it has done with other NDDs. Our work is substantial forward progress toward translational work for SynGAP1R-ID as it bridges in-vitro electrophysiological neuronal activity and function with in vivo neurophysiological brain activity and function. These data elucidate multiple quantitative, translational biomarkers in vivo and in vitro for the development of treatments for SYNGAP1-related intellectual disability.
Collapse
|
33
|
Daniel JA, Elizarova S, Shaib AH, Chouaib AA, Magnussen HM, Wang J, Brose N, Rhee J, Tirard M. An intellectual-disability-associated mutation of the transcriptional regulator NACC1 impairs glutamatergic neurotransmission. Front Mol Neurosci 2023; 16:1115880. [PMID: 37533751 PMCID: PMC10393139 DOI: 10.3389/fnmol.2023.1115880] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Accepted: 06/14/2023] [Indexed: 08/04/2023] Open
Abstract
Advances in genome sequencing technologies have favored the identification of rare de novo mutations linked to neurological disorders in humans. Recently, a de novo autosomal dominant mutation in NACC1 was identified (NM_052876.3: c.892C > T, NP_443108.1; p.Arg298Trp), associated with severe neurological symptoms including intellectual disability, microcephaly, and epilepsy. As NACC1 had never before been associated with neurological diseases, we investigated how this mutation might lead to altered brain function. We examined neurotransmission in autaptic glutamatergic mouse neurons expressing the murine homolog of the human mutant NACC1, i.e., Nacc1-R284W. We observed that expression of Nacc1-R284W impaired glutamatergic neurotransmission in a cell-autonomous manner, likely through a dominant negative mechanism. Furthermore, by screening for Nacc1 interaction targets in the brain, we identified SynGAP1, GluK2A, and several SUMO E3 ligases as novel Nacc1 interaction partners. At a biochemical level, Nacc1-R284W exhibited reduced binding to SynGAP1 and GluK2A, and also showed greatly increased SUMOylation. Ablating the SUMOylation of Nacc1-R284W partially restored its interaction with SynGAP1 but did not restore binding to GluK2A. Overall, these data indicate a role for Nacc1 in regulating glutamatergic neurotransmission, which is substantially impaired by the expression of a disease-associated Nacc1 mutant. This study provides the first functional insights into potential deficits in neuronal function in patients expressing the de novo mutant NACC1 protein.
Collapse
Affiliation(s)
- James A. Daniel
- Department of Molecular Neurobiology, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Sofia Elizarova
- Department of Molecular Neurobiology, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Ali H. Shaib
- Institute for Neuro- and Sensory Physiology, University Medical Center Göttingen, Göttingen, Germany
| | - Abed A. Chouaib
- Department of Cellular Neurophysiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), Saarland University, Homburg, Germany
| | - Helge M. Magnussen
- MRC Protein Phosphorylation and Ubiquitination Unit, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Jianlong Wang
- Department of Medicine, Columbia Center for Human Development and Stem Cell Therapies, Columbia University Irving Medical Center, New York, NY, United States
| | - Nils Brose
- Department of Molecular Neurobiology, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - JeongSeop Rhee
- Department of Molecular Neurobiology, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Marilyn Tirard
- Department of Molecular Neurobiology, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| |
Collapse
|
34
|
Bygrave AM, Sengupta A, Jackert EP, Ahmed M, Adenuga B, Nelson E, Goldschmidt HL, Johnson RC, Zhong H, Yeh FL, Sheng M, Huganir RL. Btbd11 supports cell-type-specific synaptic function. Cell Rep 2023; 42:112591. [PMID: 37261953 PMCID: PMC10592477 DOI: 10.1016/j.celrep.2023.112591] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 04/21/2023] [Accepted: 05/16/2023] [Indexed: 06/03/2023] Open
Abstract
Synapses in the brain exhibit cell-type-specific differences in basal synaptic transmission and plasticity. Here, we evaluated cell-type-specific specializations in the composition of glutamatergic synapses, identifying Btbd11 as an inhibitory interneuron-specific, synapse-enriched protein. Btbd11 is highly conserved across species and binds to core postsynaptic proteins, including Psd-95. Intriguingly, we show that Btbd11 can undergo liquid-liquid phase separation when expressed with Psd-95, supporting the idea that the glutamatergic postsynaptic density in synapses in inhibitory interneurons exists in a phase-separated state. Knockout of Btbd11 decreased glutamatergic signaling onto parvalbumin-positive interneurons. Further, both in vitro and in vivo, Btbd11 knockout disrupts network activity. At the behavioral level, Btbd11 knockout from interneurons alters exploratory behavior, measures of anxiety, and sensitizes mice to pharmacologically induced hyperactivity following NMDA receptor antagonist challenge. Our findings identify a cell-type-specific mechanism that supports glutamatergic synapse function in inhibitory interneurons-with implications for circuit function and animal behavior.
Collapse
Affiliation(s)
- Alexei M Bygrave
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| | - Ayesha Sengupta
- National Institute on Drug Abuse, Bayview Boulevard, Baltimore, MD 21224, USA
| | - Ella P Jackert
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Mehroz Ahmed
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Beloved Adenuga
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Erik Nelson
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Hana L Goldschmidt
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Richard C Johnson
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Haining Zhong
- Vollum Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Felix L Yeh
- Department of Neuroscience, Genentech, Inc, South San Francisco, CA 94080, USA
| | - Morgan Sheng
- Department of Neuroscience, Genentech, Inc, South San Francisco, CA 94080, USA
| | - Richard L Huganir
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Kavli Neuroscience Discovery Institute, Johns Hopkins University, Baltimore, MD 21205, USA.
| |
Collapse
|
35
|
Araki Y, Gerber EE, Rajkovich KE, Hong I, Johnson RC, Lee HK, Kirkwood A, Huganir RL. Mouse models of SYNGAP1 -related intellectual disability. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.25.542312. [PMID: 37293116 PMCID: PMC10245951 DOI: 10.1101/2023.05.25.542312] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
SYNGAP1 is a Ras-GTPase activating protein highly enriched at excitatory synapses in the brain. De novo loss-of-function mutations in SYNGAP1 are a major cause of genetically defined neurodevelopmental disorders (NDD). These mutations are highly penetrant and cause SYNGAP1 -related intellectual disability (SRID), a NDD characterized by cognitive impairment, social deficits, early-onset seizures, and sleep disturbances (1-5). Studies in rodent neurons have shown that Syngap1 regulates developing excitatory synapse structure and function (6-11), and heterozygous Syngap1 knockout mice have deficits in synaptic plasticity, learning and memory, and have seizures (9, 12-14). However, how specific SYNGAP1 mutations found in humans lead to disease has not been investigated in vivo. To explore this, we utilized the CRISPR-Cas9 system to generate knock-in mouse models with two distinct known causal variants of SRID: one with a frameshift mutation leading to a premature stop codon, SYNGAP1; L813RfsX22, and a second with a single-nucleotide mutation in an intron that creates a cryptic splice acceptor site leading to premature stop codon, SYNGAP1; c.3583-9G>A . While reduction in Syngap1 mRNA varies from 30-50% depending on the specific mutation, both models show ∼50% reduction in Syngap1 protein, have deficits in synaptic plasticity, and recapitulate key features of SRID including hyperactivity and impaired working memory. These data suggest that half the amount of SYNGAP1 protein is key to the pathogenesis of SRID. These results provide a resource to study SRID and establish a framework for the development of therapeutic strategies for this disorder. Significance Statement SYNGAP1 is a protein enriched at excitatory synapses in the brain that is an important regulator of synapse structure and function. SYNGAP1 mutations cause SYNGAP1 -related intellectual disability (SRID), a neurodevelopmental disorder with cognitive impairment, social deficits, seizures, and sleep disturbances. To explore how SYNGAP1 mutations found in humans lead to disease, we generated the first knock-in mouse models with causal SRID variants: one with a frameshift mutation and a second with an intronic mutation that creates a cryptic splice acceptor site. Both models show decreased Syngap1 mRNA and Syngap1 protein and recapitulate key features of SRID including hyperactivity and impaired working memory. These results provide a resource to study SRID and establish a framework for the development of therapeutic strategies. Highlights Two mouse models with SYNGAP1 -related intellectual disability (SRID) mutations found in humans were generated: one with a frameshift mutation that results in a premature stop codon and the other with an intronic mutation resulting in a cryptic splice acceptor site and premature stop codon. Both SRID mouse models show 35∼50% reduction in mRNA and ∼50% reduction in Syngap1 protein.Both SRID mouse models display deficits in synaptic plasticity and behavioral phenotypes found in people. RNA-seq confirmed cryptic splice acceptor activity in one SRID mouse model and revealed broad transcriptional changes also identified in Syngap1 +/- mice. Novel SRID mouse models generated here provide a resource and establish a framework for development of future therapeutic intervention.
Collapse
Affiliation(s)
- Yoichi Araki
- Department of Neuroscience, Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine
| | - Elizabeth E Gerber
- Department of Neuroscience, Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine
| | - Kacey E Rajkovich
- Department of Neuroscience, Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine
| | - Ingie Hong
- Department of Neuroscience, Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine
| | - Richard C Johnson
- Department of Neuroscience, Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine
| | - Hey-Kyoung Lee
- Department of Neuroscience, Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine
| | - Alfredo Kirkwood
- Department of Neuroscience, Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine
| | - Richard L Huganir
- Department of Neuroscience, Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine
| |
Collapse
|
36
|
Li Z. The ALS-Related SynGAP1 Pathogenic Variant Causes Dendritic Spine Loss: Potential Mechanisms of Early-Stage ALS Progression. J Neurosci 2023; 43:2819-2821. [PMID: 37076290 PMCID: PMC10124941 DOI: 10.1523/jneurosci.0099-23.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Revised: 02/27/2023] [Accepted: 02/27/2023] [Indexed: 04/21/2023] Open
Affiliation(s)
- Zhanxu Li
- Xiangya School of Medicine, Central South University, Changsha, Hunan 410078, China
| |
Collapse
|
37
|
Zhang P, Omanska A, Ander B, Gandal M, Stamova B, Schumann C. Neuron-specific transcriptomic signatures indicate neuroinflammation and altered neuronal activity in ASD temporal cortex. Proc Natl Acad Sci U S A 2023; 120:e2206758120. [PMID: 36862688 PMCID: PMC10013873 DOI: 10.1073/pnas.2206758120] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 12/28/2022] [Indexed: 03/03/2023] Open
Abstract
Autism spectrum disorder (ASD) is a highly heterogeneous disorder, yet transcriptomic profiling of bulk brain tissue has identified substantial convergence among dysregulated genes and pathways in ASD. However, this approach lacks cell-specific resolution. We performed comprehensive transcriptomic analyses on bulk tissue and laser-capture microdissected (LCM) neurons from 59 postmortem human brains (27 ASD and 32 controls) in the superior temporal gyrus (STG) of individuals ranging from 2 to 73 years of age. In bulk tissue, synaptic signaling, heat shock protein-related pathways, and RNA splicing were significantly altered in ASD. There was age-dependent dysregulation of genes involved in gamma aminobutyric acid (GABA) (GAD1 and GAD2) and glutamate (SLC38A1) signaling pathways. In LCM neurons, AP-1-mediated neuroinflammation and insulin/IGF-1 signaling pathways were upregulated in ASD, while mitochondrial function, ribosome, and spliceosome components were downregulated. GABA synthesizing enzymes GAD1 and GAD2 were both downregulated in ASD neurons. Mechanistic modeling suggested a direct link between inflammation and ASD in neurons, and prioritized inflammation-associated genes for future study. Alterations in small nucleolar RNAs (snoRNAs) associated with splicing events suggested interplay between snoRNA dysregulation and splicing disruption in neurons of individuals with ASD. Our findings supported the fundamental hypothesis of altered neuronal communication in ASD, demonstrated that inflammation was elevated at least in part in ASD neurons, and may reveal windows of opportunity for biotherapeutics to target the trajectory of gene expression and clinical manifestation of ASD throughout the human lifespan.
Collapse
Affiliation(s)
- Pan Zhang
- Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California, Los Angeles, CA90095
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, CA90095
- Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, CA90095
| | - Alicja Omanska
- Department of Psychiatry and Behavioral Sciences, School of Medicine, University of California, Davis, Sacramento, CA95817
- University of California, Davis, MIND Institute, Sacramento, CA95817
| | - Bradley P. Ander
- University of California, Davis, MIND Institute, Sacramento, CA95817
- Department of Neurology, University of California, Davis, School of Medicine, Sacramento, CA95817
| | - Michael J. Gandal
- Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, CA90095
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104
- Lifespan Brain Institute, Penn Med and the Children’s Hospital of Philadelphia, Philadelphia, PA19104
| | - Boryana Stamova
- University of California, Davis, MIND Institute, Sacramento, CA95817
- Department of Neurology, University of California, Davis, School of Medicine, Sacramento, CA95817
| | - Cynthia M. Schumann
- Department of Psychiatry and Behavioral Sciences, School of Medicine, University of California, Davis, Sacramento, CA95817
- University of California, Davis, MIND Institute, Sacramento, CA95817
| |
Collapse
|
38
|
Zeng X, Chen Y, Yu X, Che Y, Chen H, Yi Z, Qin J, Zhong J. Novel variants of SYNGAP1 associated epileptic encephalopathy: two cases report and literature review. ACTA EPILEPTOLOGICA 2023; 5:6. [PMID: 40217345 PMCID: PMC11960226 DOI: 10.1186/s42494-022-00114-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 10/07/2022] [Indexed: 02/22/2023] Open
Abstract
BACKGROUND SYNGAP1 is a significant genetic risk factor for global developmental delay, autism spectrum disorder, and epileptic encephalopathy. De novo loss-of-function variants in this gene cause a neurodevelopmental disorder, for example, early-onset and drug-refractory seizures. We report two children with global developmental delay and epileptic encephalopathy, which are caused by SYNGAP1 gene novel mutations, and drug treatment is effective. CASE PRESENTATION We report a boy and a girl presented with global developmental delay when they were young babies; as they grew up, cognitive impairment and social-communication disorder became more and more prominent; unfortunately, the patients developed into various seizure types, including eyelid myoclonia, myoclonic and absences when the boy was 1 year 8 mouths old and the girl was 3 years old. The two patients were found two previously unknown mutations by high throughput sequencing [c.3271_ c.3272insT; (p.L1091L fs*62), c.2515A > T (p.K839*)] in exon 15 of the SYNGAP in the proband. Sanger sequencing confirmed the heterozygous nature, and neither of their parents carried the same mutation. The girl treated with valproic acid and prednisone became seizure-free, and valproic acid and levetiracetam combined with clonazepam were influential in the other. CONCLUSIONS The global developmental delay and epileptic encephalopathy of the children were probably due to the pathogenic mutation of the SYNGAP1 gene, and prednisone and clonazepam may be effective in achieving seizure-free.
Collapse
Affiliation(s)
- Xingying Zeng
- Molecular Diagnostic Laboratory, Children's Hospital of Jiangxi Province, Nanchang, 330006, China
| | - Yong Chen
- Department of Neurology, Children's Hospital of Jiangxi Province, Nanchang, 330006, China
| | - Xiongying Yu
- Department of Neurology, Children's Hospital of Jiangxi Province, Nanchang, 330006, China
| | - Yuanyuan Che
- Department of Neurology, Children's Hospital of Jiangxi Province, Nanchang, 330006, China
| | - Hui Chen
- Department of Neurology, Children's Hospital of Jiangxi Province, Nanchang, 330006, China
| | - Zhaoshi Yi
- Molecular Diagnostic Laboratory, Children's Hospital of Jiangxi Province, Nanchang, 330006, China
| | - Jie Qin
- Department of Neurology, Children's Hospital of Jiangxi Province, Nanchang, 330006, China
| | - Jianmin Zhong
- Department of Neurology, Children's Hospital of Jiangxi Province, Nanchang, 330006, China.
| |
Collapse
|
39
|
Hitrec T, Petit C, Cryer E, Muir C, Tal N, Fustin JM, Hughes AT, Piggins HD. Timed exercise stabilizes behavioral rhythms but not molecular programs in the brain's suprachiasmatic clock. iScience 2023; 26:106002. [PMID: 36866044 PMCID: PMC9971895 DOI: 10.1016/j.isci.2023.106002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 11/25/2022] [Accepted: 01/13/2023] [Indexed: 01/20/2023] Open
Abstract
Timed daily access to a running-wheel (scheduled voluntary exercise; SVE) synchronizes rodent circadian rhythms and promotes stable, 24h rhythms in animals with genetically targeted impairment of neuropeptide signaling (Vipr2 -/- mice). Here we used RNA-seq and/or qRT-PCR to assess how this neuropeptide signaling impairment as well as SVE shapes molecular programs in the brain clock (suprachiasmatic nuclei; SCN) and peripheral tissues (liver and lung). Compared to Vipr2 +/+ animals, the SCN transcriptome of Vipr2 -/- mice showed extensive dysregulation which included core clock components, transcription factors, and neurochemicals. Furthermore, although SVE stabilized behavioral rhythms in these animals, the SCN transcriptome remained dysregulated. The molecular programs in the lung and liver of Vipr2 -/- mice were partially intact, although their response to SVE differed to that of these peripheral tissues in the Vipr2 +/+ mice. These findings highlight that SVE can correct behavioral abnormalities in circadian rhythms without causing large scale alterations to the SCN transcriptome.
Collapse
Affiliation(s)
- Timna Hitrec
- School of Physiology, Pharmacology, and Neuroscience, Faculty of Life Sciences, University of Bristol, Bristol BS8 1TD, UK
| | - Cheryl Petit
- School of Medical Sciences, Faculty of Biology, Medicine, and Health, University of Manchester, Manchester M13 9PT, UK
| | - Emily Cryer
- School of Biological Sciences, Faculty of Life Sciences, University of Bristol, Bristol BS8 1TQ, UK
| | - Charlotte Muir
- School of Physiology, Pharmacology, and Neuroscience, Faculty of Life Sciences, University of Bristol, Bristol BS8 1TD, UK
| | - Natalie Tal
- School of Medical Sciences, Faculty of Biology, Medicine, and Health, University of Manchester, Manchester M13 9PT, UK
| | - Jean-Michel Fustin
- School of Medical Sciences, Faculty of Biology, Medicine, and Health, University of Manchester, Manchester M13 9PT, UK
| | - Alun T.L. Hughes
- School of Medical Sciences, Faculty of Biology, Medicine, and Health, University of Manchester, Manchester M13 9PT, UK,School of Biological and Environmental Sciences, Faculty of Science, Liverpool John Moores University, Liverpool L3 3AF, UK,Corresponding author
| | - Hugh D. Piggins
- School of Physiology, Pharmacology, and Neuroscience, Faculty of Life Sciences, University of Bristol, Bristol BS8 1TD, UK,School of Medical Sciences, Faculty of Biology, Medicine, and Health, University of Manchester, Manchester M13 9PT, UK,Corresponding author
| |
Collapse
|
40
|
Synaptoproteomic Analysis of the Prefrontal Cortex Reveals Spatio-Temporal Changes in SYNGAP1 Following Cannabinoid Exposure in Rat Adolescence. Int J Mol Sci 2022; 24:ijms24010698. [PMID: 36614142 PMCID: PMC9820805 DOI: 10.3390/ijms24010698] [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: 11/20/2022] [Revised: 12/19/2022] [Accepted: 12/27/2022] [Indexed: 01/04/2023] Open
Abstract
The regular use of cannabis during adolescence has been associated with a number of negative life outcomes, including psychopathology and cognitive impairments. However, the exact molecular mechanisms that underlie these outcomes are just beginning to be understood. Moreover, very little is known about the spatio-temporal molecular changes that occur following cannabinoid exposure in adolescence. To understand these changes, we exposed mid-adolescent male rats to a synthetic cannabinoid (WIN 55,212-2 mesylate; WIN) and, following drug abstinence through late adolescence, we subjected the synaptosomal fractions of the prefrontal cortex (PFC) to proteomic analyses. A total of N = 487 differentially expressed proteins were found in WIN-exposed animals compared to controls. Gene ontology analyses revealed enrichment of terms related to the gamma-aminobutyric acid (GABA)-ergic neurotransmitter system. Among the top differentially expressed proteins was the synaptic Ras GTPase-activating protein 1 (SYNGAP1). Using Western blotting experiments, we found that the WIN-induced upregulation of SYNGAP1 was spatio-temporal in nature, arising only in the synaptosomal fractions (not in the cytosol) and only following prolonged drug abstinence (not on abstinence day 1). Moreover, the SYNGAP1 changes were found to be specific to WIN-exposure in adolescence and not adulthood. Adolescent animals exposed to a natural cannabinoid (Δ9-tetrahydrocannabinol; THC) were also found to have increased levels of SYNGAP1 in the PFC. THC exposure also led to a pronounced upregulation of SYNGAP1 in the amygdala, but without any changes in the dorsal striatum, hippocampus, or nucleus accumbens. To our knowledge, this is the first study to uncover a link between cannabinoid exposure and changes in SYNGAP1 that are spatio-temporal and developmental in nature. Future studies are needed to investigate the putative role of SYNGAP1 in the negative behavioral consequences of cannabis use in adolescence.
Collapse
|
41
|
Wang Y, Lv Y, Li Z, Gao M, Yang X, Li Y, Shi J, Gao Z, Liu Y, Gai Z. Phenotype and genotype analyses of Chinese patients with autosomal dominant mental retardation type 5 caused by SYNGAP1 gene mutations. Front Genet 2022; 13:957915. [PMID: 36583017 PMCID: PMC9792850 DOI: 10.3389/fgene.2022.957915] [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: 06/17/2022] [Accepted: 11/09/2022] [Indexed: 12/15/2022] Open
Abstract
Background: Autosomal dominant mental retardation type 5 (MRD5), a rare neurodevelopmental disorder (NDD) characterized by intellectual disability (ID), developmental delay (DD), and epilepsy predominantly, is caused by a heterozygous mutation in the SYNGAP1 gene. SYNGAP1 mutations have been rarely reported in the Chinese population. Here, we present an investigation of SYNGAP1 mutations in a clinical cohort with ID and DD in Shandong, a northern province in China, to further explore the genotype and phenotype correlations. Methods: A retrospective study was conducted on 10 children with SYNGAP1 mutations presenting ID, DD, and epilepsy who were diagnosed between January 2014 and May 2022. Clinical data and genetic tests were collected. Treatment and regular follow-ups were carried out to pay close attention to the prognosis of the patients. Results: We described 10 unrelated affected individuals with SYNGAP1 mutations, displaying ID, DD, epilepsy, or seizures. All mutations of SYNGAP1 in the 10 patients were de novo, except patient 3 whose father was unavailable, including five nonsense mutations, two frameshift mutations, two splicing mutations, and one codon deletion. Among these mutations, five were novel and the other five were previously reported. Significantly, all patients with epilepsy were sensitive to anti-seizure drugs, especially sodium valproate. Furthermore, rehabilitation training seemed to exert a more improved effect on motor development than language development for the patients. Conclusion The 10 patients carrying SYNGAP1 mutations were diagnosed as MRD5. Five novel genetic mutations were found, which expanded the mutational spectrum of the SYNGAP1 gene. The identification of these mutations in this study helps explore the relationship between genotypes and phenotypes and contributes to genetic counseling and therapeutic intervention for patients with MRD5.
Collapse
Affiliation(s)
- Yanxin Wang
- Department of Pediatrics, Children’s Hospital Affiliated to Shandong University, Ji’nan, China
| | - Yuqiang Lv
- Pediatric Research Institute, Children’s Hospital Affiliated to Shandong University, Ji’nan, China,Shandong Provincial Clinical Research Center for Children’s Health and Disease, Ji’nan, China
| | - Zilong Li
- Pediatric Research Institute, Children’s Hospital Affiliated to Shandong University, Ji’nan, China,Shandong Provincial Clinical Research Center for Children’s Health and Disease, Ji’nan, China
| | - Min Gao
- Pediatric Research Institute, Children’s Hospital Affiliated to Shandong University, Ji’nan, China,Shandong Provincial Clinical Research Center for Children’s Health and Disease, Ji’nan, China
| | - Xiaomeng Yang
- Pediatric Research Institute, Children’s Hospital Affiliated to Shandong University, Ji’nan, China,Shandong Provincial Clinical Research Center for Children’s Health and Disease, Ji’nan, China
| | - Yue Li
- Pediatric Research Institute, Children’s Hospital Affiliated to Shandong University, Ji’nan, China,Shandong Provincial Clinical Research Center for Children’s Health and Disease, Ji’nan, China
| | - Jianguo Shi
- Shandong Provincial Clinical Research Center for Children’s Health and Disease, Ji’nan, China,Epilepsy Center, Children’s Hospital Affiliated to Shandong University, Ji’nan, China
| | - Zaifen Gao
- Shandong Provincial Clinical Research Center for Children’s Health and Disease, Ji’nan, China,Epilepsy Center, Children’s Hospital Affiliated to Shandong University, Ji’nan, China
| | - Yi Liu
- Pediatric Research Institute, Children’s Hospital Affiliated to Shandong University, Ji’nan, China,Shandong Provincial Clinical Research Center for Children’s Health and Disease, Ji’nan, China,*Correspondence: Yi Liu, ; Zhongtao Gai,
| | - Zhongtao Gai
- Pediatric Research Institute, Children’s Hospital Affiliated to Shandong University, Ji’nan, China,Shandong Provincial Clinical Research Center for Children’s Health and Disease, Ji’nan, China,Epilepsy Center, Children’s Hospital Affiliated to Shandong University, Ji’nan, China,*Correspondence: Yi Liu, ; Zhongtao Gai,
| |
Collapse
|
42
|
Megagiannis P, Suresh R, Rouleau GA, Zhou Y. Reversibility and therapeutic development for neurodevelopmental disorders, insights from genetic animal models. Adv Drug Deliv Rev 2022; 191:114562. [PMID: 36183904 DOI: 10.1016/j.addr.2022.114562] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 08/30/2022] [Accepted: 09/24/2022] [Indexed: 01/24/2023]
Abstract
Neurodevelopmental Disorders (NDDs) encompass a broad spectrum of conditions resulting from atypical brain development. Over the past decades, we have had the fortune to witness enormous progress in diagnosis, etiology discovery, modeling, and mechanistic understanding of NDDs from both fundamental and clinical research. Here, we review recent neurobiological advances from experimental models of NDDs. We introduce several examples and highlight breakthroughs in reversal studies of phenotypes using genetically engineered models of NDDs. The in-depth understanding of brain pathophysiology underlying NDDs and evaluations of reversibility in animal models paves the foundation for discovering novel treatment options. We discuss how the expanding property of cutting-edge technologies, such as gene editing and AAV-mediated gene delivery, are leveraged in animal models for the therapeutic development of NDDs. We envision opportunities and challenges toward faithful modeling and fruitful clinical translation.
Collapse
Affiliation(s)
- Platon Megagiannis
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital; Faculty of Medicine and Health Sciences, McGill University, Montreal, Quebec H3A 2B4, Canada
| | - Rahul Suresh
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital; Faculty of Medicine and Health Sciences, McGill University, Montreal, Quebec H3A 2B4, Canada
| | - Guy A Rouleau
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital; Faculty of Medicine and Health Sciences, McGill University, Montreal, Quebec H3A 2B4, Canada
| | - Yang Zhou
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital; Faculty of Medicine and Health Sciences, McGill University, Montreal, Quebec H3A 2B4, Canada.
| |
Collapse
|
43
|
Yokoi S, Ito T, Sahashi K, Nakatochi M, Nakamura R, Tohnai G, Fujioka Y, Ishigaki S, Udagawa T, Izumi Y, Morita M, Kano O, Oda M, Sone T, Okano H, Atsuta N, Katsuno M, Okada Y, Sobue G. The SYNGAP1 3'UTR Variant in ALS Patients Causes Aberrant SYNGAP1 Splicing and Dendritic Spine Loss by Recruiting HNRNPK. J Neurosci 2022; 42:8881-8896. [PMID: 36261283 PMCID: PMC9698725 DOI: 10.1523/jneurosci.0455-22.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 08/28/2022] [Accepted: 10/06/2022] [Indexed: 12/29/2022] Open
Abstract
Fused in sarcoma (FUS) is a pathogenic RNA-binding protein in amyotrophic lateral sclerosis (ALS). We previously reported that FUS stabilizes Synaptic Ras-GTPase activating protein 1 (Syngap1) mRNA at its 3' untranslated region (UTR) and maintains spine maturation. To elucidate the pathologic roles of this mechanism in ALS patients, we identified the SYNGAP1 3'UTR variant rs149438267 in seven (four males and three females) out of 807 ALS patients at the FUS binding site from a multicenter cohort in Japan. Human-induced pluripotent stem cell (hiPSC)-derived motor neurons with the SYNGAP1 variant showed aberrant splicing, increased isoform α1 levels, and decreased isoform γ levels, which caused dendritic spine loss. Moreover, the SYNGAP1 variant excessively recruited FUS and heterogeneous nuclear ribonucleoprotein K (HNRNPK), and antisense oligonucleotides (ASOs) blocking HNRNPK altered aberrant splicing and ameliorated dendritic spine loss. These data suggest that excessive recruitment of RNA-binding proteins, especially HNRNPK, as well as changes in SYNGAP1 isoforms, are crucial for spine formation in motor neurons.SIGNIFICANCE STATEMENT It is not yet known which RNAs cause the pathogenesis of amyotrophic lateral sclerosis (ALS). We previously reported that Fused in sarcoma (FUS), a pathogenic RNA-binding protein in ALS, stabilizes synaptic Ras-GTPase activating protein 1 (Syngap1) mRNA at its 3' untranslated region (UTR) and maintains dendritic spine maturation. To elucidate whether this mechanism is crucial for ALS, we identified the SYNGAP1 3'UTR variant rs149438267 at the FUS binding site. Human-induced pluripotent stem cell (hiPSC)-derived motor neurons with the SYNGAP1 variant showed aberrant splicing, which caused dendritic spine loss along with excessive recruitment of FUS and heterogeneous nuclear ribonucleoprotein K (HNRNPK). Our findings that dendritic spine loss is because of excess recruitment of RNA-binding proteins provide a basis for the future exploration of ALS-related RNA-binding proteins.
Collapse
Affiliation(s)
- Satoshi Yokoi
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Takuji Ito
- Department of Neurology, Aichi Medical University School of Medicine, Aichi 480-1195, Japan
- Department of Neural iPSC Research, Institute for Medical Science of Aging, Aichi Medical University, Aichi 480-1195, Japan
| | - Kentaro Sahashi
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Masahiro Nakatochi
- Public Health Informatics Unit, Department of Integrated Health Sciences, Nagoya University Graduate School of Medicine, Nagoya 461-8673, Japan
| | - Ryoichi Nakamura
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
- Department of Neurology, Aichi Medical University School of Medicine, Aichi 480-1195, Japan
| | - Genki Tohnai
- Division of ALS Research, Aichi Medical University, Aichi 480-1195, Japan
| | - Yusuke Fujioka
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Shinsuke Ishigaki
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
- Research Division of Dementia and Neurodegenerative Disease, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Tsuyoshi Udagawa
- Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya 467-8603, Japan
| | - Yuishin Izumi
- Department of Neurology, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima 770-8503, Japan
| | - Mitsuya Morita
- Division of Neurology, Department of Internal Medicine, Jichi Medical University, Shimotsuke 329-0498, Japan
| | - Osamu Kano
- Department of Neurology, Toho University Faculty of Medicine, Tokyo 143-8540, Japan
| | - Masaya Oda
- Department of Neurology, Vihara Hananosato Hospital, Miyoshi 728-0001, Japan
| | - Takefumi Sone
- Department of Physiology, Keio University School of Medicine, Tokyo 160-0016, Japan
| | - Hideyuki Okano
- Department of Physiology, Keio University School of Medicine, Tokyo 160-0016, Japan
| | - Naoki Atsuta
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
- Department of Neurology, Aichi Medical University School of Medicine, Aichi 480-1195, Japan
| | - Masahisa Katsuno
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Yohei Okada
- Department of Neurology, Aichi Medical University School of Medicine, Aichi 480-1195, Japan
- Department of Neural iPSC Research, Institute for Medical Science of Aging, Aichi Medical University, Aichi 480-1195, Japan
| | - Gen Sobue
- Research Division of Dementia and Neurodegenerative Disease, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
- Aichi Medical University, Aichi 480-1195, Japan
| |
Collapse
|
44
|
Gross C. Some (Syn)Gaps are Worse than Others: Deciphering The Role of Syngap Isoforms in Excitatory Synaptic Function. Epilepsy Curr 2022; 22:312-314. [PMID: 36285208 PMCID: PMC9549232 DOI: 10.1177/15357597221111686] [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] [Indexed: 11/17/2022] Open
Abstract
Endogenous Syngap Alpha Splice Forms Promote Cognitive Function and Seizure
Protection M Kilinc, V Arora, TK Creson, et al. eLife. 2022;11:e75707.
doi:10.7554/eLife.75707. Loss-of-function variants in SYNGAP cause a developmental encephalopathy defined by
cognitive impairment, autistic features, and epilepsy. SYNGAP splicing leads to
expression of distinct functional protein isoforms. Splicing imparts multiple cellular
functions of SynGAP proteins through coding of distinct C-terminal motifs. However, it
remains unknown how these different splice sequences function in vivo to regulate
neuronal function and behavior. Reduced expression of SynGAP-α1/2 C-terminal splice
variants in mice caused severe phenotypes, including reduced survival, impaired
learning, and reduced seizure latency. In contrast, upregulation of α1/2 expression
improved learning and increased seizure latency. Mice expressing α1-specific
mutations, which disrupted SynGAP cellular functions without altering protein
expression, promoted seizure, disrupted synapse plasticity, and impaired learning.
These findings demonstrate that endogenous SynGAP isoforms with α1/2 spliced sequences
promote cognitive function and impart seizure protection. Regulation of
SynGAP-αexpression or function may be a viable therapeutic strategy to broadly improve
cognitive function and mitigate seizure.
Collapse
|
45
|
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
|
46
|
Screening membraneless organelle participants with machine-learning models that integrate multimodal features. Proc Natl Acad Sci U S A 2022; 119:e2115369119. [PMID: 35687670 PMCID: PMC9214545 DOI: 10.1073/pnas.2115369119] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Protein self-assembly is one of the formation mechanisms of biomolecular condensates. However, most phase-separating systems (PS) demand multiple partners in biological conditions. In this study, we divided PS proteins into two groups according to the mechanism by which they undergo PS: PS-Self proteins can self-assemble spontaneously to form droplets, while PS-Part proteins interact with partners to undergo PS. Analysis of the amino acid composition revealed differences in the sequence pattern between the two protein groups. Existing PS predictors, when evaluated on two test protein sets, preferentially predicted self-assembling proteins. Thus, a comprehensive predictor is required. Herein, we propose that properties other than sequence composition can provide crucial information in screening PS proteins. By incorporating phosphorylation frequencies and immunofluorescence image-based droplet-forming propensity with other PS-related features, we built two independent machine-learning models to separately predict the two protein categories. Results of independent testing suggested the superiority of integrating multimodal features. We performed experimental verification on the top-scored proteins DHX9, Ki-67, and NIFK. Their PS behavior in vitro revealed the effectiveness of our models in PS prediction. Further validation on the proteome of membraneless organelles confirmed the ability of our models to identify PS-Part proteins. We implemented a web server named PhaSePred (http://predict.phasep.pro/) that incorporates our two models together with representative PS predictors. PhaSePred displays proteome-level quantiles of different features, thus profiling PS propensity and providing crucial information for identification of candidate proteins.
Collapse
|
47
|
The Emerging Roles of Long Non-Coding RNAs in Intellectual Disability and Related Neurodevelopmental Disorders. Int J Mol Sci 2022; 23:ijms23116118. [PMID: 35682796 PMCID: PMC9181295 DOI: 10.3390/ijms23116118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 05/23/2022] [Accepted: 05/27/2022] [Indexed: 02/05/2023] Open
Abstract
In the human brain, long non-coding RNAs (lncRNAs) are widely expressed in an exquisitely temporally and spatially regulated manner, thus suggesting their contribution to normal brain development and their probable involvement in the molecular pathology of neurodevelopmental disorders (NDD). Bypassing the classic protein-centric conception of disease mechanisms, some studies have been conducted to identify and characterize the putative roles of non-coding sequences in the genetic pathogenesis and diagnosis of complex diseases. However, their involvement in NDD, and more specifically in intellectual disability (ID), is still poorly documented and only a few genomic alterations affecting the lncRNAs function and/or expression have been causally linked to the disease endophenotype. Considering that a significant fraction of patients still lacks a genetic or molecular explanation, we expect that a deeper investigation of the non-coding genome will unravel novel pathogenic mechanisms, opening new translational opportunities. Here, we present evidence of the possible involvement of many lncRNAs in the etiology of different forms of ID and NDD, grouping the candidate disease-genes in the most frequently affected cellular processes in which ID-risk genes were previously collected. We also illustrate new approaches for the identification and prioritization of NDD-risk lncRNAs, together with the current strategies to exploit them in diagnosis.
Collapse
|
48
|
O-GlcNAcylation modulates liquid–liquid phase separation of SynGAP/PSD-95. Nat Chem 2022; 14:831-840. [DOI: 10.1038/s41557-022-00946-9] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 04/07/2022] [Indexed: 12/13/2022]
|
49
|
Shih PY, Fang YL, Shankar S, Lee SP, Hu HT, Chen H, Wang TF, Hsia KC, Hsueh YP. Phase separation and zinc-induced transition modulate synaptic distribution and association of autism-linked CTTNBP2 and SHANK3. Nat Commun 2022; 13:2664. [PMID: 35562389 PMCID: PMC9106668 DOI: 10.1038/s41467-022-30353-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 04/26/2022] [Indexed: 11/09/2022] Open
Abstract
Many synaptic proteins form biological condensates via liquid-liquid phase separation (LLPS). Synaptopathy, a key feature of autism spectrum disorders (ASD), is likely relevant to the impaired phase separation and/or transition of ASD-linked synaptic proteins. Here, we report that LLPS and zinc-induced liquid-to-gel phase transition regulate the synaptic distribution and protein-protein interaction of cortactin-binding protein 2 (CTTNBP2), an ASD-linked protein. CTTNBP2 forms self-assembled condensates through its C-terminal intrinsically disordered region and facilitates SHANK3 co-condensation at dendritic spines. Zinc binds the N-terminal coiled-coil region of CTTNBP2, promoting higher-order assemblies. Consequently, it leads to reduce CTTNBP2 mobility and enhance the stability and synaptic retention of CTTNBP2 condensates. Moreover, ASD-linked mutations alter condensate formation and synaptic retention of CTTNBP2 and impair mouse social behaviors, which are all ameliorated by zinc supplementation. Our study suggests the relevance of condensate formation and zinc-induced phase transition to the synaptic distribution and function of ASD-linked proteins. Autism impacts synapses. This study reports that autism-linked mutations of CTTNBP2 regulate phase separation to control synaptic enrichment of that protein. A zinc-induced liquid-to-gel transition improves synaptic retention of CTTNBP2 and SHANK3.
Collapse
Affiliation(s)
- Pu-Yun Shih
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan, ROC.,Department of Neurology, University of California San Francisco, San Francisco, USA
| | - Yu-Lun Fang
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan, ROC.,Department and Graduate Institute of Biochemistry, National Defense Medical Center, Taipei, Taiwan, ROC
| | - Sahana Shankar
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan, ROC.,Molecular and Cell Biology, Taiwan International Graduate Program, Institute of Molecular Biology, Academia Sinica and Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan, ROC
| | - Sue-Ping Lee
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan, ROC
| | - Hsiao-Tang Hu
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan, ROC
| | - Hsin Chen
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan, ROC.,Undergraduate Program in Neuroscience, John Hopkins University, Baltimore, USA
| | - Ting-Fang Wang
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan, ROC.,Molecular and Cell Biology, Taiwan International Graduate Program, Institute of Molecular Biology, Academia Sinica and Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan, ROC
| | - Kuo-Chiang Hsia
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan, ROC. .,Molecular and Cell Biology, Taiwan International Graduate Program, Institute of Molecular Biology, Academia Sinica and Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan, ROC.
| | - Yi-Ping Hsueh
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan, ROC. .,Molecular and Cell Biology, Taiwan International Graduate Program, Institute of Molecular Biology, Academia Sinica and Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan, ROC.
| |
Collapse
|
50
|
Kilinc M, Arora V, Creson TK, Rojas C, Le AA, Lauterborn J, Wilkinson B, Hartel N, Graham N, Reich A, Gou G, Araki Y, Bayés À, Coba M, Lynch G, Miller CA, Rumbaugh G. Endogenous Syngap1 alpha splice forms promote cognitive function and seizure protection. eLife 2022; 11:e75707. [PMID: 35394425 PMCID: PMC9064290 DOI: 10.7554/elife.75707] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 04/08/2022] [Indexed: 11/13/2022] Open
Abstract
Loss-of-function variants in SYNGAP1 cause a developmental encephalopathy defined by cognitive impairment, autistic features, and epilepsy. SYNGAP1 splicing leads to expression of distinct functional protein isoforms. Splicing imparts multiple cellular functions of SynGAP proteins through coding of distinct C-terminal motifs. However, it remains unknown how these different splice sequences function in vivo to regulate neuronal function and behavior. Reduced expression of SynGAP-α1/2 C-terminal splice variants in mice caused severe phenotypes, including reduced survival, impaired learning, and reduced seizure latency. In contrast, upregulation of α1/2 expression improved learning and increased seizure latency. Mice expressing α1-specific mutations, which disrupted SynGAP cellular functions without altering protein expression, promoted seizure, disrupted synapse plasticity, and impaired learning. These findings demonstrate that endogenous SynGAP isoforms with α1/2 spliced sequences promote cognitive function and impart seizure protection. Regulation of SynGAP-αexpression or function may be a viable therapeutic strategy to broadly improve cognitive function and mitigate seizure.
Collapse
Affiliation(s)
- Murat Kilinc
- Graduate School of Chemical and Biological Sciences, The Scripps Research InstituteJupiterUnited States
- Departments of Neuroscience and Molecular Medicine, The Scripps Research InstituteJupiterUnited States
| | - Vineet Arora
- Departments of Neuroscience and Molecular Medicine, The Scripps Research InstituteJupiterUnited States
| | - Thomas K Creson
- Departments of Neuroscience and Molecular Medicine, The Scripps Research InstituteJupiterUnited States
| | - Camilo Rojas
- Departments of Neuroscience and Molecular Medicine, The Scripps Research InstituteJupiterUnited States
| | - Aliza A Le
- Department of Anatomy and Neurobiology, The University of CaliforniaIrvineUnited States
| | - Julie Lauterborn
- Department of Anatomy and Neurobiology, The University of CaliforniaIrvineUnited States
| | - Brent Wilkinson
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern CaliforniaLos AngelesUnited States
| | - Nicolas Hartel
- Mork Family Department of Chemical Engineering and Materials Science, University of Southern CaliforniaLos AngelesUnited States
| | - Nicholas Graham
- Mork Family Department of Chemical Engineering and Materials Science, University of Southern CaliforniaLos AngelesUnited States
| | - Adrian Reich
- Bioinformatics and Statistics Core, The Scripps Research InstituteJupiterUnited States
| | - Gemma Gou
- Molecular Physiology of the Synapse Laboratory, Institut d'Investigació Biomèdica Sant PauBarcelonaSpain
- Universitat Autònoma de BarcelonaBellaterraSpain
| | - Yoichi Araki
- Department of Neuroscience, Johns Hopkins School of MedicineBaltimoreUnited States
| | - Àlex Bayés
- Molecular Physiology of the Synapse Laboratory, Institut d'Investigació Biomèdica Sant PauBarcelonaSpain
| | - Marcelo Coba
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern CaliforniaLos AngelesUnited States
| | - Gary Lynch
- Department of Anatomy and Neurobiology, The University of CaliforniaIrvineUnited States
| | - Courtney A Miller
- Graduate School of Chemical and Biological Sciences, The Scripps Research InstituteJupiterUnited States
- Departments of Neuroscience and Molecular Medicine, The Scripps Research InstituteJupiterUnited States
| | - Gavin Rumbaugh
- Graduate School of Chemical and Biological Sciences, The Scripps Research InstituteJupiterUnited States
- Departments of Neuroscience and Molecular Medicine, The Scripps Research InstituteJupiterUnited States
| |
Collapse
|