1
|
Atefrad S, Yousefnejad A, Faraji N, Keshavarz P. The association between NLGN4 gene variants and the incidence of autism spectrum disorders in Guilan, Iran. IBRO Neurosci Rep 2025; 18:306-310. [PMID: 40034542 PMCID: PMC11874714 DOI: 10.1016/j.ibneur.2025.01.018] [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: 08/27/2024] [Revised: 01/26/2025] [Accepted: 01/31/2025] [Indexed: 03/05/2025] Open
Abstract
Autism Spectrum Disorder (ASD) is a developmental disorder characterized by impaired social interaction, communication skills, and repetitive behaviours. This study aimed to investigate the association between variants of the Neuroligin-4 (NLGN4) gene (rs1882260 and rs3810688) and the incidence of ASD in North of Iran in the ASD group (n = 60) and control group (n = 60). DNA was isolated from whole blood, saliva, or hair samples. The targeted variants were genotyped using the Amplification Refractory Mutation System-Polymerase Chain Reaction (ARMS-PCR) technique. Genetic analyses were conducted using SNPAlyze ver. 8.1. Results revealed a significant difference of rs3810688 polymorphism in the NLGN4 gene in both genotypic and allelic frequency distributions between the ASD and control groups (P < 0.05). The GG genotype of rs3810688 polymorphism exhibited a significant association with an elevated risk of ASD in contrast to the CC genotype, as revealed under the co-dominant model (OR=4.2; 95 %CI, 1.25-14.05; P = 0.019). The study illustrated the possible role of rs3810688 polymorphism of NLGN4 in increasing the incidence of ASD among newborns in Guilan province. Also, the G-C haplotype was found to be a protective variant against ASD.
Collapse
Affiliation(s)
- Sepideh Atefrad
- Department of Genetics, Cellular and Molecular Research Center, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
- Master of Genetics, Razi Clinical Research Development Unit, Guilan University of Medical Sciences, Rasht, Iran
| | - Aidi Yousefnejad
- Department of Genetics, Cellular and Molecular Research Center, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Niloofar Faraji
- Gastrointestinal and Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran
| | - Parvaneh Keshavarz
- Department of Genetics, Cellular and Molecular Research Center, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| |
Collapse
|
2
|
Benner O, Karr CH, Quintero-Gonzalez A, Tamkun MM, Chanda S. The Shab family potassium channels are highly enriched at the presynaptic terminals of human neurons. J Biol Chem 2025; 301:108235. [PMID: 39880095 PMCID: PMC11894309 DOI: 10.1016/j.jbc.2025.108235] [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: 11/12/2024] [Revised: 01/02/2025] [Accepted: 01/21/2025] [Indexed: 01/31/2025] Open
Abstract
The Shab family voltage-gated K+ channels (i.e., Kv2.1, Kv2.2) are widely expressed in mammalian brain and regulate neuronal action-potential firing. In addition to their canonical functions, the Kv2 proteins help establish direct attachments between plasma membrane and endoplasmic reticulum (ER), also known as ER-plasma membrane junctions. However, the biochemical properties and molecular organization of these ion channels have not yet been described in human neurons. Here, we have performed a systematic analysis of endogenous expression, post-translational modification, and subcellular distribution of the major components of Kv2 complex in neurons derived from human stem cells. We found that both Kv2.1, Kv2.2, and their auxiliary subunit AMIGO1 are significantly upregulated during early neurogenesis, localize at the cell surface, and already begin to assemble with each other. Human Kv2.1 and AMIGO1, but not Kv2.2, undergo substantial post-translational modification including phosphorylation and/or N-linked glycosylation. Acute pharmacological inhibition with Kv2 blockers also revealed their functional activation in human neurons. These proteins formed prominent clusters at cell bodies, dendritic branches, and axon initial segments. Interestingly, a large fraction of them also exhibited considerable accumulation at human presynaptic terminals, where they aggregated with the local ER network. This synaptic localization of Kv2 subunits was primarily restricted to presynaptic regions, as they demonstrated limited enrichment at postsynaptic densities. These results were highly reproducible in multiple stem cell lines used and alternative differentiation protocols tested, confirming that human presynaptic compartments can actively recruit the Shab family K+ ion channels.
Collapse
Affiliation(s)
- Orion Benner
- Biochemistry & Molecular Biology, Colorado State University, Fort Collins, Colorado, USA
| | - Charles H Karr
- Biochemistry & Molecular Biology, Colorado State University, Fort Collins, Colorado, USA
| | | | - Michael M Tamkun
- Biomedical Sciences, Colorado State University, Fort Collins, Colorado, USA; Molecular, Cellular & Integrated Neurosciences, Colorado State University, Fort Collins, Colorado, USA
| | - Soham Chanda
- Biochemistry & Molecular Biology, Colorado State University, Fort Collins, Colorado, USA; Molecular, Cellular & Integrated Neurosciences, Colorado State University, Fort Collins, Colorado, USA; Cell & Molecular Biology, Colorado State University, Fort Collins, Colorado, USA.
| |
Collapse
|
3
|
Zhang M, Ma X, Wang Z, Han Y, Jia Z, Chen D, Xu Y, Qiao Z, Jiang X, Wang L, Jiang H, Yu M, Li Y, Shen Y. Genome-wide association analysis study on host resistance against the Aeromonas veronii of largemouth bass Micropterus salmoides. FISH & SHELLFISH IMMUNOLOGY 2025; 157:110093. [PMID: 39736405 DOI: 10.1016/j.fsi.2024.110093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Revised: 11/19/2024] [Accepted: 12/20/2024] [Indexed: 01/01/2025]
Abstract
Largemouth bass (Micropterus salmoides) has become one of the most important freshwater economic fish farmed almost all over China in recent years. At the same time, the increasing outbreaks of diseases in its aquaculture process have caused substantial economic losses to this industry. However, at present, the genetic basis of disease resistance, including resistance against Aeromonas veronii infection, in largemouth bass is very limited. Therefore, a genome-wide association study (GWAS) on host resistance against the A. veronii of largemouth bass was conducted in the present study. A total of 627 largemouth bass were artificially challenged by A. veronii, among which 160 of the earliest deaths and 173 of the final survivals were genotyped. A total of 3076 high-quality SNPs were used for further analysis employing two analysis models, of which six shared SNPs were finally identified as significant molecular markers with the explaining phenotypic variance ranging from 2.28 % to 8.95 %. Furthermore, seven candidate genes were identified, including one gene, T-cell surface antigen CD2, which is directly involved in T cell activation and the cellular immune response. Additionally, the other identified genes play roles in critical processes such as cell survival, inflammatory responses, and signal transduction. This study lays a genetic foundation for research on largemouth bass disease resistance and studies related to A. veronii. It also contributes significantly to the future development of the commercial production of largemouth bass.
Collapse
Affiliation(s)
- Meng Zhang
- College of Fisheries, Henan Normal University, Xinxiang, 453007, China; Observation and Research Station on Water Ecosystem in Danjiangkou Reservoir of Henan Province, Nanyang, 474450, China.
| | - Xiao Ma
- College of Fisheries, Henan Normal University, Xinxiang, 453007, China
| | - Zerui Wang
- College of Fisheries, Henan Normal University, Xinxiang, 453007, China
| | - Yuqing Han
- College of Fisheries, Henan Normal University, Xinxiang, 453007, China
| | - Zhilin Jia
- College of Fisheries, Henan Normal University, Xinxiang, 453007, China
| | - Dongcai Chen
- College of Fisheries, Henan Normal University, Xinxiang, 453007, China
| | - Yue Xu
- College of Fisheries, Henan Normal University, Xinxiang, 453007, China
| | - Zhigang Qiao
- College of Fisheries, Henan Normal University, Xinxiang, 453007, China
| | - Xinyu Jiang
- College of Fisheries, Henan Normal University, Xinxiang, 453007, China
| | - Lei Wang
- College of Fisheries, Henan Normal University, Xinxiang, 453007, China
| | - Hongxia Jiang
- College of Fisheries, Henan Normal University, Xinxiang, 453007, China
| | - Miao Yu
- College of Fisheries, Henan Normal University, Xinxiang, 453007, China
| | - Yongjing Li
- College of Fisheries, Henan Normal University, Xinxiang, 453007, China
| | - Yawei Shen
- College of Fisheries, Henan Normal University, Xinxiang, 453007, China; Observation and Research Station on Water Ecosystem in Danjiangkou Reservoir of Henan Province, Nanyang, 474450, China; Department of Biological Sciences, Clemson University, Clemson, SC, 29634, USA.
| |
Collapse
|
4
|
Mirabella F, Randazzo M, Rinaldi A, Pettinato F, Rizzo R, Sturiale L, Barone R. Glycosylation Pathways Targeted by Deregulated miRNAs in Autism Spectrum Disorder. Int J Mol Sci 2025; 26:783. [PMID: 39859496 PMCID: PMC11766332 DOI: 10.3390/ijms26020783] [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: 11/26/2024] [Revised: 12/22/2024] [Accepted: 01/14/2025] [Indexed: 01/27/2025] Open
Abstract
Autism Spectrum Disorder (ASD) is a complex condition with a multifactorial aetiology including both genetic and epigenetic factors. MicroRNAs (miRNAs) play a role in ASD and may influence metabolic pathways. Glycosylation (the glycoconjugate synthesis pathway) is a necessary process for the optimal development of the central nervous system (CNS). Congenital Disorders of Glycosylation (CDGs) (CDGs) are linked to over 180 genes and are predominantly associated with neurodevelopmental disorders (NDDs) including ASD. From a literature search, we considered 64 miRNAs consistently deregulated in ASD patients (ASD-miRNAs). Computational tools, including DIANA-miRPath v3.0 and TarBase v8, were employed to investigate the potential involvement of ASD-miRNAs in glycosylation pathways. A regulatory network constructed through miRNet 2.0 revealed the involvement of these miRNAs in targeting genes linked to glycosylation. Protein functions were further validated through the Human Protein Atlas. A total of twenty-five ASD-miRNAs were identified, including nine miRNAs that were differentially expressed in cells or brain tissue in ASD patients and associated with glycosylation pathways, specifically protein N- and O-glycosylation and glycosaminoglycan biosynthesis (heparan sulfate). A number of CDG genes and/or ASD-risk genes, including DOLK, GALNT2, and EXT1, were identified as targets, along with validated interactions involving four key miRNAs (hsa-miR-423-5p, hsa-miR-30c-5p, hsa-miR-195-5p, and hsa-miR-132-5p). B4GALT1, an ASD susceptibility gene, emerged as a central regulatory hub, reinforcing the link between glycosylation and ASD. In sum, the evidence presented here supports the hypothesis that ASD-miRNAs mediate the epigenetic regulation of glycosylation, thus unveiling possible novel patho-mechanisms underlying ASD.
Collapse
Affiliation(s)
- Federica Mirabella
- Child Neuropsychiatry Unit, Department of Clinical and Experimental Medicine, University of Catania, 95124 Catania, Italy; (F.M.); (M.R.); (A.R.); (F.P.); (R.R.)
| | - Martina Randazzo
- Child Neuropsychiatry Unit, Department of Clinical and Experimental Medicine, University of Catania, 95124 Catania, Italy; (F.M.); (M.R.); (A.R.); (F.P.); (R.R.)
| | - Alessandro Rinaldi
- Child Neuropsychiatry Unit, Department of Clinical and Experimental Medicine, University of Catania, 95124 Catania, Italy; (F.M.); (M.R.); (A.R.); (F.P.); (R.R.)
| | - Fabio Pettinato
- Child Neuropsychiatry Unit, Department of Clinical and Experimental Medicine, University of Catania, 95124 Catania, Italy; (F.M.); (M.R.); (A.R.); (F.P.); (R.R.)
| | - Renata Rizzo
- Child Neuropsychiatry Unit, Department of Clinical and Experimental Medicine, University of Catania, 95124 Catania, Italy; (F.M.); (M.R.); (A.R.); (F.P.); (R.R.)
| | - Luisa Sturiale
- CNR—Institute for Polymers, Composites and Biomaterials IPCB, 95126 Catania, Italy;
| | - Rita Barone
- Child Neuropsychiatry Unit, Department of Clinical and Experimental Medicine, University of Catania, 95124 Catania, Italy; (F.M.); (M.R.); (A.R.); (F.P.); (R.R.)
- Research Unit of Rare Diseases and Neurodevelopmental Disorders, Oasi Research Institute—IRCCS, 94018 Troina, Italy
| |
Collapse
|
5
|
Lehr AW, McDaniel KF, Roche KW. Analyses of Human Genetic Data to Identify Clinically Relevant Domains of Neuroligins. Genes (Basel) 2024; 15:1601. [PMID: 39766868 PMCID: PMC11675371 DOI: 10.3390/genes15121601] [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: 11/13/2024] [Revised: 12/03/2024] [Accepted: 12/11/2024] [Indexed: 01/30/2025] Open
Abstract
Background/Objectives: Neuroligins (NLGNs) are postsynaptic adhesion molecules critical for neuronal development that are highly associated with autism spectrum disorder (ASD). Here, we provide an overview of the literature on NLGN rare variants. In addition, we introduce a new approach to analyze human variation within NLGN genes to identify sensitive regions that have an increased frequency of ASD-associated variants to better understand NLGN function. Methods: To identify critical protein subdomains within the NLGN gene family, we developed an algorithm that assesses tolerance to missense mutations in human genetic variation by comparing clinical variants from ClinVar to reference variants from gnomAD. This approach provides tolerance values to subdomains within the protein. Results: Our algorithm identified several critical regions that were conserved across multiple NLGN isoforms. Importantly, this approach also identified a previously reported cluster of pathogenic variants in NLGN4X (also conserved in NLGN1 and NLGN3) as well as a region around the highly characterized NLGN3 R451C ASD-associated mutation. Additionally, we highlighted other, as of yet, uncharacterized regions enriched with mutations. Conclusions: The systematic analysis of NLGN ASD-associated variants compared to variants identified in the unaffected population (gnomAD) reveals conserved domains in NLGN isoforms that are tolerant to variation or are enriched in clinically relevant variants. Examination of databases also allows for predictions of the presumed tolerance to loss of an allele. The use of the algorithm we developed effectively allowed the evaluation of subdomains of NLGNs and can be used to examine other ASD-associated genes.
Collapse
Affiliation(s)
- Alexander W. Lehr
- Receptor Biology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA; (A.W.L.); (K.F.M.)
- Department of Neuroscience, Brown University, Providence, RI 02906, USA
| | - Kathryn F. McDaniel
- Receptor Biology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA; (A.W.L.); (K.F.M.)
- Department of Neuroscience, Brown University, Providence, RI 02906, USA
| | - Katherine W. Roche
- Receptor Biology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA; (A.W.L.); (K.F.M.)
| |
Collapse
|
6
|
Wang Y, Lei K, Zhao L, Zhang Y. Clinical glycoproteomics: methods and diseases. MedComm (Beijing) 2024; 5:e760. [PMID: 39372389 PMCID: PMC11450256 DOI: 10.1002/mco2.760] [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: 06/06/2024] [Revised: 09/08/2024] [Accepted: 09/10/2024] [Indexed: 10/08/2024] Open
Abstract
Glycoproteins, representing a significant proportion of posttranslational products, play pivotal roles in various biological processes, such as signal transduction and immune response. Abnormal glycosylation may lead to structural and functional changes of glycoprotein, which is closely related to the occurrence and development of various diseases. Consequently, exploring protein glycosylation can shed light on the mechanisms behind disease manifestation and pave the way for innovative diagnostic and therapeutic strategies. Nonetheless, the study of clinical glycoproteomics is fraught with challenges due to the low abundance and intricate structures of glycosylation. Recent advancements in mass spectrometry-based clinical glycoproteomics have improved our ability to identify abnormal glycoproteins in clinical samples. In this review, we aim to provide a comprehensive overview of the foundational principles and recent advancements in clinical glycoproteomic methodologies and applications. Furthermore, we discussed the typical characteristics, underlying functions, and mechanisms of glycoproteins in various diseases, such as brain diseases, cardiovascular diseases, cancers, kidney diseases, and metabolic diseases. Additionally, we highlighted potential avenues for future development in clinical glycoproteomics. These insights provided in this review will enhance the comprehension of clinical glycoproteomic methods and diseases and promote the elucidation of pathogenesis and the discovery of novel diagnostic biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Yujia Wang
- Department of General Practice Ward/International Medical Center WardGeneral Practice Medical Center and Institutes for Systems GeneticsWest China HospitalSichuan UniversityChengduChina
| | - Kaixin Lei
- Department of General Practice Ward/International Medical Center WardGeneral Practice Medical Center and Institutes for Systems GeneticsWest China HospitalSichuan UniversityChengduChina
| | - Lijun Zhao
- Department of General Practice Ward/International Medical Center WardGeneral Practice Medical Center and Institutes for Systems GeneticsWest China HospitalSichuan UniversityChengduChina
| | - Yong Zhang
- Department of General Practice Ward/International Medical Center WardGeneral Practice Medical Center and Institutes for Systems GeneticsWest China HospitalSichuan UniversityChengduChina
| |
Collapse
|
7
|
Al-Beltagi M, Saeed NK, Bediwy AS, Bediwy EA, Elbeltagi R. Decoding the genetic landscape of autism: A comprehensive review. World J Clin Pediatr 2024; 13:98468. [PMID: 39350903 PMCID: PMC11438927 DOI: 10.5409/wjcp.v13.i3.98468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 07/29/2024] [Accepted: 08/01/2024] [Indexed: 08/30/2024] Open
Abstract
BACKGROUND Autism spectrum disorder (ASD) is a complex neurodevelopmental condition characterized by heterogeneous symptoms and genetic underpinnings. Recent advancements in genetic and epigenetic research have provided insights into the intricate mechanisms contributing to ASD, influencing both diagnosis and therapeutic strategies. AIM To explore the genetic architecture of ASD, elucidate mechanistic insights into genetic mutations, and examine gene-environment interactions. METHODS A comprehensive systematic review was conducted, integrating findings from studies on genetic variations, epigenetic mechanisms (such as DNA methylation and histone modifications), and emerging technologies [including Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR)-Cas9 and single-cell RNA sequencing]. Relevant articles were identified through systematic searches of databases such as PubMed and Google Scholar. RESULTS Genetic studies have identified numerous risk genes and mutations associated with ASD, yet many cases remain unexplained by known factors, suggesting undiscovered genetic components. Mechanistic insights into how these genetic mutations impact neural development and brain connectivity are still evolving. Epigenetic modifications, particularly DNA methylation and non-coding RNAs, also play significant roles in ASD pathogenesis. Emerging technologies like CRISPR-Cas9 and advanced bioinformatics are advancing our understanding by enabling precise genetic editing and analysis of complex genomic data. CONCLUSION Continued research into the genetic and epigenetic underpinnings of ASD is crucial for developing personalized and effective treatments. Collaborative efforts integrating multidisciplinary expertise and international collaborations are essential to address the complexity of ASD and translate genetic discoveries into clinical practice. Addressing unresolved questions and ethical considerations surrounding genetic research will pave the way for improved diagnostic tools and targeted therapies, ultimately enhancing outcomes for individuals affected by ASD.
Collapse
Affiliation(s)
- Mohammed Al-Beltagi
- Department of Pediatric, Faculty of Medicine, Tanta University, Alghrabia, Tanta 31511, Egypt
- Department of Pediatric, University Medical Center, King Abdulla Medical City, Arabian Gulf University, Manama 26671, Bahrain
| | - Nermin Kamal Saeed
- Medical Microbiology Section, Department of Pathology, Salmaniya Medical Complex, Ministry of Health, Kingdom of Bahrain, Manama 12, Bahrain
- Medical Microbiology Section, Department of Pathology, Irish Royal College of Surgeon, Muharraq, Busaiteen 15503, Bahrain
| | - Adel Salah Bediwy
- Department of Pulmonology, Faculty of Medicine, Tanta University, Alghrabia, Tanta 31527, Egypt
- Department of Pulmonology, University Medical Center, King Abdulla Medical City, Arabian Gulf University, Manama 26671, Bahrain
| | - Eman A Bediwy
- Internal Medicine, Faculty of Medicine, Tanta University, Algharbia, Tanta 31527, Egypt
| | - Reem Elbeltagi
- Department of Medicine, The Royal College of Surgeons in Ireland-Bahrain, Muharraq, Busiateen 15503, Bahrain
| |
Collapse
|
8
|
Hatchett CJ, Hall MK, Messer AR, Schwalbe RA. Lowered GnT-I Activity Decreases Complex-Type N-Glycan Amounts and Results in an Aberrant Primary Motor Neuron Structure in the Spinal Cord. J Dev Biol 2024; 12:21. [PMID: 39189261 PMCID: PMC11348029 DOI: 10.3390/jdb12030021] [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: 06/13/2024] [Revised: 07/19/2024] [Accepted: 08/14/2024] [Indexed: 08/28/2024] Open
Abstract
The attachment of sugar to proteins and lipids is a basic modification needed for organismal survival, and perturbations in glycosylation cause severe developmental and neurological difficulties. Here, we investigated the neurological consequences of N-glycan populations in the spinal cord of Wt AB and mgat1b mutant zebrafish. Mutant fish have reduced N-acetylglucosaminyltransferase-I (GnT-I) activity as mgat1a remains intact. GnT-I converts oligomannose N-glycans to hybrid N-glycans, which is needed for complex N-glycan production. MALDI-TOF MS profiles identified N-glycans in the spinal cord for the first time and revealed reduced amounts of complex N-glycans in mutant fish, supporting a lesion in mgat1b. Further lectin blotting showed that oligomannose N-glycans were more prevalent in the spinal cord, skeletal muscle, heart, swim bladder, skin, and testis in mutant fish relative to WT AB, supporting lowered GnT- I activity in a global manner. Developmental delays were noted in hatching and in the swim bladder. Microscopic images of caudal primary (CaP) motor neurons of the spinal cord transiently expressing EGFP in mutant fish were abnormal with significant reductions in collateral branches. Further motor coordination skills were impaired in mutant fish. We conclude that identifying the neurological consequences of aberrant N-glycan processing will enhance our understanding of the role of complex N-glycans in development and nervous system health.
Collapse
Affiliation(s)
| | | | | | - Ruth A. Schwalbe
- Department of Biochemistry and Molecular Biology, Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA; (C.J.H.); (M.K.H.); (A.R.M.)
| |
Collapse
|
9
|
Wang L, Mirabella VR, Dai R, Su X, Xu R, Jadali A, Bernabucci M, Singh I, Chen Y, Tian J, Jiang P, Kwan KY, Pak C, Liu C, Comoletti D, Hart RP, Chen C, Südhof TC, Pang ZP. Analyses of the autism-associated neuroligin-3 R451C mutation in human neurons reveal a gain-of-function synaptic mechanism. Mol Psychiatry 2024; 29:1620-1635. [PMID: 36280753 PMCID: PMC10123180 DOI: 10.1038/s41380-022-01834-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 10/04/2022] [Accepted: 10/10/2022] [Indexed: 12/11/2022]
Abstract
Mutations in many synaptic genes are associated with autism spectrum disorders (ASD), suggesting that synaptic dysfunction is a key driver of ASD pathogenesis. Among these mutations, the R451C substitution in the NLGN3 gene that encodes the postsynaptic adhesion molecule Neuroligin-3 is noteworthy because it was the first specific mutation linked to ASDs. In mice, the corresponding Nlgn3 R451C-knockin mutation recapitulates social interaction deficits of ASD patients and produces synaptic abnormalities, but the impact of the NLGN3 R451C mutation on human neurons has not been investigated. Here, we generated human knockin neurons with the NLGN3 R451C and NLGN3 null mutations. Strikingly, analyses of NLGN3 R451C-mutant neurons revealed that the R451C mutation decreased NLGN3 protein levels but enhanced the strength of excitatory synapses without affecting inhibitory synapses; meanwhile NLGN3 knockout neurons showed reduction in excitatory synaptic strengths. Moreover, overexpression of NLGN3 R451C recapitulated the synaptic enhancement in human neurons. Notably, the augmentation of excitatory transmission was confirmed in vivo with human neurons transplanted into mouse forebrain. Using single-cell RNA-seq experiments with co-cultured excitatory and inhibitory NLGN3 R451C-mutant neurons, we identified differentially expressed genes in relatively mature human neurons corresponding to synaptic gene expression networks. Moreover, gene ontology and enrichment analyses revealed convergent gene networks associated with ASDs and other mental disorders. Our findings suggest that the NLGN3 R451C mutation induces a gain-of-function enhancement in excitatory synaptic transmission that may contribute to the pathophysiology of ASD.
Collapse
Affiliation(s)
- Le Wang
- Child Health Institute of New Jersey and Department of Neuroscience and Cell Biology, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ, 08901, USA
- Center for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, and Department of Psychiatry, The Second Xiangya Hospital, Central South University, 410008, Changsha, China
| | - Vincent R Mirabella
- Child Health Institute of New Jersey and Department of Neuroscience and Cell Biology, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ, 08901, USA
- Department of Molecular & Cellular Physiology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Rujia Dai
- Department of Psychiatry, SUNY Upstate Medical University, Syracuse, NY, 13210, USA
| | - Xiao Su
- Child Health Institute of New Jersey and Department of Neuroscience and Cell Biology, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ, 08901, USA
| | - Ranjie Xu
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, 08854, USA
| | - Azadeh Jadali
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, 08854, USA
| | - Matteo Bernabucci
- Child Health Institute of New Jersey and Department of Neuroscience and Cell Biology, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ, 08901, USA
| | - Ishnoor Singh
- Child Health Institute of New Jersey and Department of Neuroscience and Cell Biology, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ, 08901, USA
| | - Yu Chen
- Center for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, and Department of Psychiatry, The Second Xiangya Hospital, Central South University, 410008, Changsha, China
| | - Jianghua Tian
- Center for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, and Department of Psychiatry, The Second Xiangya Hospital, Central South University, 410008, Changsha, China
| | - Peng Jiang
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, 08854, USA
| | - Kevin Y Kwan
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, 08854, USA
| | - ChangHui Pak
- Department of Biochemistry & Molecular Biology, University of Massachusetts, Amherst, MA, 01003, USA
| | - Chunyu Liu
- Center for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, and Department of Psychiatry, The Second Xiangya Hospital, Central South University, 410008, Changsha, China
- Department of Psychiatry, SUNY Upstate Medical University, Syracuse, NY, 13210, USA
- School of Psychology, Shaanxi Normal University, 710000, Xi'an, Shaanxi, China
| | - Davide Comoletti
- Child Health Institute of New Jersey and Department of Neuroscience and Cell Biology, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ, 08901, USA
- School of Biological Sciences, Victoria University of Wellington, Wellington, 6012, New Zealand
| | - Ronald P Hart
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, 08854, USA
| | - Chao Chen
- Center for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, and Department of Psychiatry, The Second Xiangya Hospital, Central South University, 410008, Changsha, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, 410008, Changsha, Hunan, China.
- Hunan Key Laboratory of Animal Models for Human Diseases, Central South University, 410008, Changsha, Hunan, China.
| | - Thomas C Südhof
- Department of Molecular & Cellular Physiology, Stanford University School of Medicine, Stanford, CA, 94305, USA.
- Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA, 94305, USA.
| | - Zhiping P Pang
- Child Health Institute of New Jersey and Department of Neuroscience and Cell Biology, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ, 08901, USA.
| |
Collapse
|
10
|
Vilela J, Rasga C, Santos JX, Martiniano H, Marques AR, Oliveira G, Vicente AM. Bridging Genetic Insights with Neuroimaging in Autism Spectrum Disorder-A Systematic Review. Int J Mol Sci 2024; 25:4938. [PMID: 38732157 PMCID: PMC11084239 DOI: 10.3390/ijms25094938] [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: 02/29/2024] [Revised: 04/22/2024] [Accepted: 04/25/2024] [Indexed: 05/13/2024] Open
Abstract
Autism Spectrum Disorder (ASD) is an early onset neurodevelopmental disorder characterized by impaired social interaction and communication, and repetitive patterns of behavior. Family studies show that ASD is highly heritable, and hundreds of genes have previously been implicated in the disorder; however, the etiology is still not fully clear. Brain imaging and electroencephalography (EEG) are key techniques that study alterations in brain structure and function. Combined with genetic analysis, these techniques have the potential to help in the clarification of the neurobiological mechanisms contributing to ASD and help in defining novel therapeutic targets. To further understand what is known today regarding the impact of genetic variants in the brain alterations observed in individuals with ASD, a systematic review was carried out using Pubmed and EBSCO databases and following the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines. This review shows that specific genetic variants and altered patterns of gene expression in individuals with ASD may have an effect on brain circuits associated with face processing and social cognition, and contribute to excitation-inhibition imbalances and to anomalies in brain volumes.
Collapse
Affiliation(s)
- Joana Vilela
- Departamento de Promoção da Saúde e Doenças Não Transmissíveis, Instituto Nacional de Saúde Doutor Ricardo Jorge, Avenida Padre Cruz, 1649-016 Lisboa, Portugal; (J.V.); (C.R.); (J.X.S.); (H.M.); (A.R.M.)
- BioISI-Biosystems & Integrative Sciences Institute, Faculty of Sciences, University of Lisboa, Campo Grande, C8, 1749-016 Lisboa, Portugal
| | - Célia Rasga
- Departamento de Promoção da Saúde e Doenças Não Transmissíveis, Instituto Nacional de Saúde Doutor Ricardo Jorge, Avenida Padre Cruz, 1649-016 Lisboa, Portugal; (J.V.); (C.R.); (J.X.S.); (H.M.); (A.R.M.)
- BioISI-Biosystems & Integrative Sciences Institute, Faculty of Sciences, University of Lisboa, Campo Grande, C8, 1749-016 Lisboa, Portugal
| | - João Xavier Santos
- Departamento de Promoção da Saúde e Doenças Não Transmissíveis, Instituto Nacional de Saúde Doutor Ricardo Jorge, Avenida Padre Cruz, 1649-016 Lisboa, Portugal; (J.V.); (C.R.); (J.X.S.); (H.M.); (A.R.M.)
- BioISI-Biosystems & Integrative Sciences Institute, Faculty of Sciences, University of Lisboa, Campo Grande, C8, 1749-016 Lisboa, Portugal
| | - Hugo Martiniano
- Departamento de Promoção da Saúde e Doenças Não Transmissíveis, Instituto Nacional de Saúde Doutor Ricardo Jorge, Avenida Padre Cruz, 1649-016 Lisboa, Portugal; (J.V.); (C.R.); (J.X.S.); (H.M.); (A.R.M.)
- BioISI-Biosystems & Integrative Sciences Institute, Faculty of Sciences, University of Lisboa, Campo Grande, C8, 1749-016 Lisboa, Portugal
| | - Ana Rita Marques
- Departamento de Promoção da Saúde e Doenças Não Transmissíveis, Instituto Nacional de Saúde Doutor Ricardo Jorge, Avenida Padre Cruz, 1649-016 Lisboa, Portugal; (J.V.); (C.R.); (J.X.S.); (H.M.); (A.R.M.)
- BioISI-Biosystems & Integrative Sciences Institute, Faculty of Sciences, University of Lisboa, Campo Grande, C8, 1749-016 Lisboa, Portugal
| | - Guiomar Oliveira
- Unidade de Neurodesenvolvimento e Autismo, Serviço do Centro de Desenvolvimento da Criança, Centro de Investigação e Formação Clínica, Hospital Pediátrico, Centro Hospitalar e Universitário de Coimbra (CHUC), 3000-602 Coimbra, Portugal;
- Coimbra Institute for Biomedical Imaging and Translational Research, University Clinic of Pediatrics, Faculty of Medicine, University of Coimbra, 3000-602 Coimbra, Portugal
| | - Astrid Moura Vicente
- Departamento de Promoção da Saúde e Doenças Não Transmissíveis, Instituto Nacional de Saúde Doutor Ricardo Jorge, Avenida Padre Cruz, 1649-016 Lisboa, Portugal; (J.V.); (C.R.); (J.X.S.); (H.M.); (A.R.M.)
- BioISI-Biosystems & Integrative Sciences Institute, Faculty of Sciences, University of Lisboa, Campo Grande, C8, 1749-016 Lisboa, Portugal
| |
Collapse
|
11
|
Parrella NF, Hill AT, Dipnall LM, Loke YJ, Enticott PG, Ford TC. Inhibitory dysfunction and social processing difficulties in autism: A comprehensive narrative review. J Psychiatr Res 2024; 169:113-125. [PMID: 38016393 DOI: 10.1016/j.jpsychires.2023.11.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 09/04/2023] [Accepted: 11/15/2023] [Indexed: 11/30/2023]
Abstract
The primary inhibitory neurotransmitter γ-aminobutyric acid (GABA) has a prominent role in regulating neural development and function, with disruption to GABAergic signalling linked to behavioural phenotypes associated with neurodevelopmental disorders, particularly autism. Such neurochemical disruption, likely resulting from diverse genetic and molecular mechanisms, particularly during early development, can subsequently affect the cellular balance of excitation and inhibition in neuronal circuits, which may account for the social processing difficulties observed in autism and related conditions. This comprehensive narrative review integrates diverse streams of research from several disciplines, including molecular neurobiology, genetics, epigenetics, and systems neuroscience. In so doing it aims to elucidate the relevance of inhibitory dysfunction to autism, with specific focus on social processing difficulties that represent a core feature of this disorder. Many of the social processing difficulties experienced in autism have been linked to higher levels of the excitatory neurotransmitter glutamate and/or lower levels of inhibitory GABA. While current therapeutic options for social difficulties in autism are largely limited to behavioural interventions, this review highlights the psychopharmacological studies that explore the utility of GABA modulation in alleviating such difficulties.
Collapse
Affiliation(s)
| | - Aron T Hill
- Cognitive Neuroscience Unit, School of Psychology, Deakin University, Geelong, Australia; Department of Psychiatry, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Lillian M Dipnall
- Cognitive Neuroscience Unit, School of Psychology, Deakin University, Geelong, Australia; Early Life Epigenetics Group, Deakin University, Geelong, Australia
| | - Yuk Jing Loke
- Epigenetics Group, Murdoch Children's Research Institute, Melbourne, Victoria, Australia; Department of Paediatrics, The University of Melbourne, Melbourne, Victoria, Australia
| | - Peter G Enticott
- Cognitive Neuroscience Unit, School of Psychology, Deakin University, Geelong, Australia
| | - Talitha C Ford
- Cognitive Neuroscience Unit, School of Psychology, Deakin University, Geelong, Australia; Centre for Human Psychopharmacology, Faculty of Health, Arts and Design, Swinburne University of Technology, Melbourne, Victoria, Australia
| |
Collapse
|
12
|
Tahtamouni LH, Alderfer SA, Kuhn TB, Minamide LS, Chanda S, Ruff MR, Bamburg JR. Characterization of a Human Neuronal Culture System for the Study of Cofilin-Actin Rod Pathology. Biomedicines 2023; 11:2942. [PMID: 38001943 PMCID: PMC10669520 DOI: 10.3390/biomedicines11112942] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 10/17/2023] [Accepted: 10/26/2023] [Indexed: 11/26/2023] Open
Abstract
Cofilactin rod pathology, which can initiate synapse loss, has been extensively studied in rodent neurons, hippocampal slices, and in vivo mouse models of human neurodegenerative diseases such as Alzheimer's disease (AD). In these systems, rod formation induced by disease-associated factors, such as soluble oligomers of Amyloid-β (Aβ) in AD, utilizes a pathway requiring cellular prion protein (PrPC), NADPH oxidase (NOX), and cytokine/chemokine receptors (CCR5 and/or CXCR4). However, rod pathways have not been systematically assessed in a human neuronal model. Here, we characterize glutamatergic neurons differentiated from human-induced pluripotent stem cells (iPSCs) for the formation of rods in response to activators of the PrPC-dependent pathway. Optimization of substratum, cell density, and use of glial-conditioned medium yielded a robust system for studying the development of Aβ-induced rods in the absence of glia, suggesting a cell-autonomous pathway. Rod induction in younger neurons requires ectopic expression of PrPC, but this dependency disappears by Day 55. The quantification of proteins within the rod-inducing pathway suggests that increased PrPC and CXCR4 expression may be factors in the doubling of the rod response to Aβ between Days 35 and 55. FDA-approved antagonists to CXCR4 and CCR5 inhibit the rod response. Rods were predominantly observed in dendrites, although severe cytoskeletal disruptions prevented the assignment of over 40% of the rods to either an axon or dendrite. In the absence of glia, a condition in which rods are more readily observed, neurons mature and fire action potentials but do not form functional synapses. However, PSD95-containing dendritic spines associate with axonal regions of pre-synaptic vesicles containing the glutamate transporter, VGLUT1. Thus, our results identified stem cell-derived neurons as a robust model for studying cofilactin rod formation in a human cellular environment and for developing effective therapeutic strategies for the treatment of dementias arising from multiple proteinopathies with different rod initiators.
Collapse
Affiliation(s)
- Lubna H. Tahtamouni
- Department of Biology and Biotechnology, Faculty of Science, The Hashemite University, Zarqa 13133, Jordan;
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO 80523, USA; (T.B.K.); (L.S.M.); (S.C.)
| | - Sydney A. Alderfer
- Department of Chemical and Biological Engineering and School of Biomedical Engineering, Colorado State University, Fort Collins, CO 80523, USA;
| | - Thomas B. Kuhn
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO 80523, USA; (T.B.K.); (L.S.M.); (S.C.)
| | - Laurie S. Minamide
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO 80523, USA; (T.B.K.); (L.S.M.); (S.C.)
| | - Soham Chanda
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO 80523, USA; (T.B.K.); (L.S.M.); (S.C.)
| | - Michael R. Ruff
- Creative Bio-Peptides, Inc., 10319 Glen Road, Suite 100, Potomac, MD 20854, USA;
| | - James R. Bamburg
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO 80523, USA; (T.B.K.); (L.S.M.); (S.C.)
| |
Collapse
|
13
|
Soeda S, Ito D, Ogushi T, Sano Y, Negoro R, Fujita T, Saito R, Taniura H. Defects in early synaptic formation and neuronal function in Prader-Willi syndrome. Sci Rep 2023; 13:12053. [PMID: 37491450 PMCID: PMC10368700 DOI: 10.1038/s41598-023-39065-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 07/19/2023] [Indexed: 07/27/2023] Open
Abstract
Prader-Willi syndrome (PWS), which is a complex epigenetic disorder caused by the deficiency of paternally expressed genes in chromosome 15q11-q13, is associated with several psychiatric dimensions, including autism spectrum disorder. We have previously reported that iPS cells derived from PWS patients exhibited aberrant differentiation and transcriptomic dysregulation in differentiated neural stem cells (NSCs) and neurons. Here, we identified SLITRK1 as a downregulated gene in NSCs differentiated from PWS patient iPS cells by RNA sequencing analysis. Because SLITRK1 is involved in synaptogenesis, we focused on the synaptic formation and function of neurons differentiated from PWS patient iPS cells and NDN or MAGEL2 single gene defect mutant iPS cells. Although βIII tubulin expression levels in all the neurons were comparable to the level of differentiation in the control, pre- and postsynaptic markers were significantly lower in PWS and mutant neurons than in control neurons. PSD-95 puncta along βIII tubulin neurites were also decreased. Membrane potential responses were measured while exposed to high K+ stimulation. The neuronal excitabilities in PWS and mutant neurons showed significantly lower intensity than that of control neurons. These functional defects in PWS neurons may reflect phenotypes of neurodevelopmental disorders in PWS.
Collapse
Affiliation(s)
- Shuhei Soeda
- Laboratory of Neurochemistry, College of Pharmaceutical Sciences, Ritsumeikan University, 1-1-1 Noji Higashi, Kusatsu, Shiga, 525-8577, Japan.
| | - Daiki Ito
- Laboratory of Neurochemistry, College of Pharmaceutical Sciences, Ritsumeikan University, 1-1-1 Noji Higashi, Kusatsu, Shiga, 525-8577, Japan
| | - Tomoe Ogushi
- Laboratory of Neurochemistry, College of Pharmaceutical Sciences, Ritsumeikan University, 1-1-1 Noji Higashi, Kusatsu, Shiga, 525-8577, Japan
| | - Yui Sano
- Laboratory of Neurochemistry, College of Pharmaceutical Sciences, Ritsumeikan University, 1-1-1 Noji Higashi, Kusatsu, Shiga, 525-8577, Japan
| | - Ryosuke Negoro
- Laboratory of Molecular Pharmacokinetics, College of Pharmaceutical Sciences, Ritsumeikan University, Kusatsu, Shiga, 525-8577, Japan
| | - Takuya Fujita
- Laboratory of Molecular Pharmacokinetics, College of Pharmaceutical Sciences, Ritsumeikan University, Kusatsu, Shiga, 525-8577, Japan
| | - Ryo Saito
- Mitsubishi Tanabe Pharma Corporation, Kamoshida, Aoba, Yokohama, 227-0033, Japan
| | - Hideo Taniura
- Laboratory of Neurochemistry, College of Pharmaceutical Sciences, Ritsumeikan University, 1-1-1 Noji Higashi, Kusatsu, Shiga, 525-8577, Japan
| |
Collapse
|
14
|
Lei W, Cheng Y, Gao J, Liu X, Shao L, Kong Q, Zheng N, Ling Z, Hu W. Akkermansia muciniphila in neuropsychiatric disorders: friend or foe? Front Cell Infect Microbiol 2023; 13:1224155. [PMID: 37492530 PMCID: PMC10363720 DOI: 10.3389/fcimb.2023.1224155] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 06/26/2023] [Indexed: 07/27/2023] Open
Abstract
An accumulating body of evidence suggests that the bacterium Akkermansia muciniphila exhibits positive systemic effects on host health, mainly by improving immunological and metabolic functions, and it is therefore regarded as a promising potential probiotic. Recent clinical and preclinical studies have shown that A. muciniphila plays a vital role in a variety of neuropsychiatric disorders by influencing the host brain through the microbiota-gut-brain axis (MGBA). Numerous studies observed that A. muciniphila and its metabolic substances can effectively improve the symptoms of neuropsychiatric disorders by restoring the gut microbiota, reestablishing the integrity of the gut mucosal barrier, regulating host immunity, and modulating gut and neuroinflammation. However, A. muciniphila was also reported to participate in the development of neuropsychiatric disorders by aggravating inflammation and influencing mucus production. Therefore, the exact mechanism of action of A. muciniphila remains much controversial. This review summarizes the proposed roles and mechanisms of A. muciniphila in various neurological and psychiatric disorders such as depression, anxiety, Parkinson's disease, Alzheimer's disease, multiple sclerosis, strokes, and autism spectrum disorders, and provides insights into the potential therapeutic application of A. muciniphila for the treatment of these conditions.
Collapse
Affiliation(s)
- Wenhui Lei
- Jinan Microecological Biomedicine Shandong Laboratory, Shandong First Medical University, Jinan, Shandong, China
| | - Yiwen Cheng
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, Shandong, China
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Jie Gao
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, Shandong, China
| | - Xia Liu
- Department of Intensive Care Unit, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Li Shao
- School of Clinical Medicine, Institute of Hepatology and Metabolic Diseases, The Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Qingming Kong
- School of Biological Engineering, Hangzhou Medical College, Institute of Parasitic Diseases, Hangzhou, Zhejiang, China
| | - Nengneng Zheng
- Department of Obstetrics, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Zongxin Ling
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, Shandong, China
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Weiming Hu
- Department of Psychiatry, Quzhou Third Hospital, Quzhou, Zhejiang, China
| |
Collapse
|
15
|
Pradeep P, Kang H, Lee B. Glycosylation and behavioral symptoms in neurological disorders. Transl Psychiatry 2023; 13:154. [PMID: 37156804 PMCID: PMC10167254 DOI: 10.1038/s41398-023-02446-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 04/19/2023] [Accepted: 04/24/2023] [Indexed: 05/10/2023] Open
Abstract
Glycosylation, the addition of glycans or carbohydrates to proteins, lipids, or other glycans, is a complex post-translational modification that plays a crucial role in cellular function. It is estimated that at least half of all mammalian proteins undergo glycosylation, underscoring its importance in the functioning of cells. This is reflected in the fact that a significant portion of the human genome, around 2%, is devoted to encoding enzymes involved in glycosylation. Changes in glycosylation have been linked to various neurological disorders, including Alzheimer's disease, Parkinson's disease, autism spectrum disorder, and schizophrenia. Despite its widespread occurrence, the role of glycosylation in the central nervous system remains largely unknown, particularly with regard to its impact on behavioral abnormalities in brain diseases. This review focuses on examining the role of three types of glycosylation: N-glycosylation, O-glycosylation, and O-GlcNAcylation, in the manifestation of behavioral and neurological symptoms in neurodevelopmental, neurodegenerative, and neuropsychiatric disorders.
Collapse
Affiliation(s)
- Prajitha Pradeep
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, 34126, South Korea
- IBS School, University of Science and Technology (UST), Daejeon, 34113, South Korea
| | - Hyeyeon Kang
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, 34126, South Korea
- Department of Biomedical Engineering, College of Information and Biotechnology, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, South Korea
| | - Boyoung Lee
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, 34126, South Korea.
- IBS School, University of Science and Technology (UST), Daejeon, 34113, South Korea.
- Department of Biomedical Engineering, College of Information and Biotechnology, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, South Korea.
| |
Collapse
|
16
|
Muellerleile J, Vnencak M, Sethi MVA, Jungenitz T, Schwarzacher SW, Jedlicka P. Increased Network Inhibition in the Dentate Gyrus of Adult Neuroligin-4 Knock-Out Mice. eNeuro 2023; 10:10/4/ENEURO.0471-22.2023. [PMID: 37080762 PMCID: PMC10121080 DOI: 10.1523/eneuro.0471-22.2023] [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: 11/18/2022] [Revised: 03/01/2023] [Accepted: 03/12/2023] [Indexed: 04/22/2023] Open
Abstract
Loss-of-function mutations in neuroligin-4 (Nlgn4), a member of the neuroligin family of postsynaptic adhesion proteins, cause autism spectrum disorder in humans. Nlgn4 knockout (KO) in mice leads to social behavior deficits and complex alterations of synaptic inhibition or excitation, depending on the brain region. In the present work, we comprehensively analyzed synaptic function and plasticity at the cellular and network levels in hippocampal dentate gyrus of Nlgn4 KO mice. Compared with wild-type littermates, adult Nlgn4 KO mice exhibited increased paired-pulse inhibition of dentate granule cell population spikes, but no impairments in excitatory synaptic transmission or short-term and long-term plasticity in vivo In vitro patch-clamp recordings in neonatal organotypic entorhino-hippocampal slice cultures from Nlgn4 KO and wild-type littermates revealed no significant differences in excitatory or inhibitory synaptic transmission, homeostatic synaptic plasticity, and passive electrotonic properties in dentate granule cells, suggesting that the increased inhibition in vivo is the result of altered network activity in the adult Nlgn4 KO. A comparison with prior studies on Nlgn 1-3 knock-out mice reveals that each of the four neuroligins exerts a characteristic effect on both intrinsic cellular and network activity in the dentate gyrus in vivo.
Collapse
Affiliation(s)
- Julia Muellerleile
- Institute of Clinical Neuroanatomy, Neuroscience Center, Goethe University Frankfurt, 60590 Frankfurt am Main, Germany
- Faculty of Biosciences, Goethe University Frankfurt, 60439 Frankfurt am Main, Germany
| | - Matej Vnencak
- Institute of Clinical Neuroanatomy, Neuroscience Center, Goethe University Frankfurt, 60590 Frankfurt am Main, Germany
| | - Mohammad Valeed Ahmed Sethi
- Institute of Clinical Neuroanatomy, Neuroscience Center, Goethe University Frankfurt, 60590 Frankfurt am Main, Germany
| | - Tassilo Jungenitz
- Institute of Clinical Neuroanatomy, Neuroscience Center, Goethe University Frankfurt, 60590 Frankfurt am Main, Germany
| | - Stephan W Schwarzacher
- Institute of Clinical Neuroanatomy, Neuroscience Center, Goethe University Frankfurt, 60590 Frankfurt am Main, Germany
| | - Peter Jedlicka
- Institute of Clinical Neuroanatomy, Neuroscience Center, Goethe University Frankfurt, 60590 Frankfurt am Main, Germany
- Faculty of Medicine, Justus-Liebig-University Giessen, 35392 Giessen, Germany
| |
Collapse
|
17
|
Impaired OTUD7A-dependent Ankyrin regulation mediates neuronal dysfunction in mouse and human models of the 15q13.3 microdeletion syndrome. Mol Psychiatry 2023; 28:1747-1769. [PMID: 36604605 DOI: 10.1038/s41380-022-01937-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 12/15/2022] [Accepted: 12/19/2022] [Indexed: 01/07/2023]
Abstract
Copy number variations (CNVs) are associated with psychiatric and neurodevelopmental disorders (NDDs), and most, including the recurrent 15q13.3 microdeletion disorder, have unknown disease mechanisms. We used a heterozygous 15q13.3 microdeletion mouse model and patient iPSC-derived neurons to reveal developmental defects in neuronal maturation and network activity. To identify the underlying molecular dysfunction, we developed a neuron-specific proximity-labeling proteomics (BioID2) pipeline, combined with patient mutations, to target the 15q13.3 CNV genetic driver OTUD7A. OTUD7A is an emerging independent NDD risk gene with no known function in the brain, but has putative deubiquitinase function. The OTUD7A protein-protein interaction network included synaptic, axonal, and cytoskeletal proteins and was enriched for ASD and epilepsy risk genes (Ank3, Ank2, SPTAN1, SPTBN1). The interactions between OTUD7A and Ankyrin-G (Ank3) and Ankyrin-B (Ank2) were disrupted by an epilepsy-associated OTUD7A L233F variant. Further investigation of Ankyrin-G in mouse and human 15q13.3 microdeletion and OTUD7AL233F/L233F models revealed protein instability, increased polyubiquitination, and decreased levels in the axon initial segment, while structured illumination microscopy identified reduced Ankyrin-G nanodomains in dendritic spines. Functional analysis of human 15q13.3 microdeletion and OTUD7AL233F/L233F models revealed shared and distinct impairments to axonal growth and intrinsic excitability. Importantly, restoring OTUD7A or Ankyrin-G expression in 15q13.3 microdeletion neurons led to a reversal of abnormalities. These data reveal a critical OTUD7A-Ankyrin pathway in neuronal development, which is impaired in the 15q13.3 microdeletion syndrome, leading to neuronal dysfunction. Furthermore, our study highlights the utility of targeting CNV genes using cell type-specific proteomics to identify shared and unexplored disease mechanisms across NDDs.
Collapse
|
18
|
English J, McSweeney D, Ribbe F, Howell E, Pak C. Generation and Co-culture of Cortical Glutamatergic and GABAergic-Induced Neuronal Cells. Methods Mol Biol 2023; 2683:21-37. [PMID: 37300764 DOI: 10.1007/978-1-0716-3287-1_3] [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] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
The study of neurological disorders requires experimentation on human neurons throughout their development. Primary neurons can be difficult to obtain, and animal models may not fully recapitulate phenotypes observed in human neurons. Human neuronal culturing schemes which contain a balanced mixture of excitatory and inhibitory neurons that resemble physiological ratios seen in vivo will be useful to probe the neurological basis of excitation-inhibition (E-I) balance. Here, we describe a method for directly inducing a homogenous population of cortical excitatory neurons and cortical interneurons from human pluripotent stem cells, as well as the generation of mixed cultures using these induced neurons. The obtained cells display robust neuronal synchronous network activity as well as complex morphologies that are amenable to studies probing the molecular and cellular basis of disease mutations or other aspects of neuronal and synaptic development.
Collapse
Affiliation(s)
- Jay English
- Department of Biochemistry and Molecular Biology, University of Massachusetts Amherst, Amherst, MA, USA
- Molecular and Cellular Biology Graduate Program, University of Massachusetts Amherst, Amherst, MA, USA
| | - Danny McSweeney
- Department of Biochemistry and Molecular Biology, University of Massachusetts Amherst, Amherst, MA, USA
- Molecular and Cellular Biology Graduate Program, University of Massachusetts Amherst, Amherst, MA, USA
| | - Fumiko Ribbe
- Department of Biochemistry and Molecular Biology, University of Massachusetts Amherst, Amherst, MA, USA
| | - Ethan Howell
- Department of Biochemistry and Molecular Biology, University of Massachusetts Amherst, Amherst, MA, USA
| | - ChangHui Pak
- Department of Biochemistry and Molecular Biology, University of Massachusetts Amherst, Amherst, MA, USA.
| |
Collapse
|
19
|
Sperandeo A, Tamburini C, Noakes Z, de la Fuente DC, Keefe F, Petter O, Plumbly W, Clifton N, Li M, Peall K. Cortical neuronal hyperexcitability and synaptic changes in SGCE mutation-positive myoclonus dystonia. Brain 2022; 146:1523-1541. [PMID: 36204995 PMCID: PMC10115238 DOI: 10.1093/brain/awac365] [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: 01/27/2022] [Revised: 07/17/2022] [Accepted: 09/09/2022] [Indexed: 11/13/2022] Open
Abstract
Myoclonus Dystonia is a childhood-onset hyperkinetic movement disorder with a combined motor and psychiatric phenotype. It represents one of the few autosomal dominant inherited dystonic disorders and is caused by mutations in the ε-sarcoglycan (SGCE) gene. Work to date suggests that dystonia is caused by disruption of neuronal networks, principally basal ganglia-cerebello-thalamo-cortical circuits. Investigation of cortical involvement has primarily focused on disruption to interneuron inhibitory activity, rather than the excitatory activity of cortical pyramidal neurons. Here, we have sought to examine excitatory cortical glutamatergic activity using two approaches; the CRISPR/Cas9 editing of a human embryonic cell line, generating an SGCE compound heterozygous mutation, and three patient-derived induced pluripotent stem cell lines (iPSC) each gene edited to generate matched wild-type SGCE control lines. Differentiation towards a cortical neuronal phenotype demonstrated no significant differences in neither early- (PAX6, FOXG1) nor late-stage (CTIP2, TBR1) neurodevelopmental markers. However, functional characterisation using Ca2+ imaging and MEA approaches identified an increase in network activity, while single-cell patch clamp studies found a greater propensity towards action potential generation with larger amplitudes and shorter half-widths associated with SGCE-mutations. Bulk-RNA-seq analysis identified gene ontological enrichment for neuron projection development, synaptic signalling, and synaptic transmission. Examination of dendritic morphology found SGCE-mutations to be associated with a significantly higher number of branches and longer branch lengths, together with longer ion-channel dense axon initial segments, particularly towards the latter stages of differentiation (D80 and D100). Gene expression and protein quantification of key synaptic proteins (synaptophysin, synapsin and PSD95), AMPA and NMDA receptor subunits found no significant differences between the SGCE-mutation and matched wild-type lines. By contrast, significant changes to synaptic adhesion molecule expression were identified, namely higher pre-synaptic neurexin-1 and lower post-synaptic neuroligin-4 levels in the SGCE mutation carrying lines. Our study demonstrates an increased intrinsic excitability of cortical glutamatergic neuronal cells in the context of SGCE mutations, coupled with a more complex neurite morphology and disruption to synaptic adhesion molecules. These changes potentially represent key components to the development of the hyperkinetic clinical phenotype observed in Myoclonus Dystonia, as well a central feature to the wider spectrum of dystonic disorders, potentially providing targets for future therapeutic development.
Collapse
Affiliation(s)
- Alessandra Sperandeo
- Neuroscience and Mental Health Research Institute, Division of Psychological Medicine and Clinical Neuroscience, Cardiff University, Hadyn Ellis Building, Cardiff, CF24 4HQ
| | - Claudia Tamburini
- Neuroscience and Mental Health Research Institute, Division of Psychological Medicine and Clinical Neuroscience, Cardiff University, Hadyn Ellis Building, Cardiff, CF24 4HQ
| | - Zoe Noakes
- Neuroscience and Mental Health Research Institute, Division of Psychological Medicine and Clinical Neuroscience, Cardiff University, Hadyn Ellis Building, Cardiff, CF24 4HQ
| | - Daniel Cabezas de la Fuente
- Neuroscience and Mental Health Research Institute, Division of Psychological Medicine and Clinical Neuroscience, Cardiff University, Hadyn Ellis Building, Cardiff, CF24 4HQ
| | - Francesca Keefe
- Neuroscience and Mental Health Research Institute, Division of Psychological Medicine and Clinical Neuroscience, Cardiff University, Hadyn Ellis Building, Cardiff, CF24 4HQ
| | - Olena Petter
- Neuroscience and Mental Health Research Institute, Division of Psychological Medicine and Clinical Neuroscience, Cardiff University, Hadyn Ellis Building, Cardiff, CF24 4HQ
| | - William Plumbly
- Neuroscience and Mental Health Research Institute, Division of Psychological Medicine and Clinical Neuroscience, Cardiff University, Hadyn Ellis Building, Cardiff, CF24 4HQ
| | - Nicholas Clifton
- Neuroscience and Mental Health Research Institute, Division of Psychological Medicine and Clinical Neuroscience, Cardiff University, Hadyn Ellis Building, Cardiff, CF24 4HQ
| | - Meng Li
- Neuroscience and Mental Health Research Institute, Division of Psychological Medicine and Clinical Neuroscience, Cardiff University, Hadyn Ellis Building, Cardiff, CF24 4HQ
| | - Kathryn Peall
- Neuroscience and Mental Health Research Institute, Division of Psychological Medicine and Clinical Neuroscience, Cardiff University, Hadyn Ellis Building, Cardiff, CF24 4HQ
| |
Collapse
|
20
|
Burlingham SR, Wong NF, Peterkin L, Lubow L, Dos Santos Passos C, Benner O, Ghebrial M, Cast TP, Xu-Friedman MA, Südhof TC, Chanda S. Induction of synapse formation by de novo neurotransmitter synthesis. Nat Commun 2022; 13:3060. [PMID: 35650274 PMCID: PMC9160008 DOI: 10.1038/s41467-022-30756-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 05/17/2022] [Indexed: 11/09/2022] Open
Abstract
A vital question in neuroscience is how neurons align their postsynaptic structures with presynaptic release sites. Although synaptic adhesion proteins are known to contribute in this process, the role of neurotransmitters remains unclear. Here we inquire whether de novo biosynthesis and vesicular release of a noncanonical transmitter can facilitate the assembly of its corresponding postsynapses. We demonstrate that, in both stem cell-derived human neurons as well as in vivo mouse neurons of purely glutamatergic identity, ectopic expression of GABA-synthesis enzymes and vesicular transporters is sufficient to both produce GABA from ambient glutamate and transmit it from presynaptic terminals. This enables efficient accumulation and consistent activation of postsynaptic GABAA receptors, and generates fully functional GABAergic synapses that operate in parallel but independently of their glutamatergic counterparts. These findings suggest that presynaptic release of a neurotransmitter itself can signal the organization of relevant postsynaptic apparatus, which could be directly modified to reprogram the synapse identity of neurons.
Collapse
Affiliation(s)
- Scott R Burlingham
- Biochemistry & Molecular Biology, Colorado State University, Fort Collins, CO, USA
| | - Nicole F Wong
- Biological Sciences, State University of New York at Buffalo, Buffalo, NY, USA
| | - Lindsay Peterkin
- Biochemistry & Molecular Biology, Colorado State University, Fort Collins, CO, USA
| | - Lily Lubow
- Biochemistry & Molecular Biology, Colorado State University, Fort Collins, CO, USA
| | | | - Orion Benner
- Biochemistry & Molecular Biology, Colorado State University, Fort Collins, CO, USA
| | - Michael Ghebrial
- Biological Science, California State University Fullerton, Fullerton, CA, USA
| | - Thomas P Cast
- Biochemistry & Molecular Biology, Colorado State University, Fort Collins, CO, USA
| | | | - Thomas C Südhof
- Molecular & Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA.
| | - Soham Chanda
- Biochemistry & Molecular Biology, Colorado State University, Fort Collins, CO, USA.
- Molecular & Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA.
- Molecular, Cellular & Integrated Neurosciences, Colorado State University, Fort Collins, CO, USA.
| |
Collapse
|
21
|
Maxeiner S, Benseler F, Brose N, Krasteva-Christ G. Of Humans and Gerbils— Independent Diversification of Neuroligin-4 Into X- and Y-Specific Genes in Primates and Rodents. Front Mol Neurosci 2022; 15:838262. [PMID: 35431802 PMCID: PMC9005811 DOI: 10.3389/fnmol.2022.838262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 02/03/2022] [Indexed: 11/13/2022] Open
Abstract
The neural cell adhesion protein neuroligin-4 has puzzled neuroscientists and geneticist alike for almost two decades. Its clinical association with autism spectrum disorders (ASD) is well established, however, its diversification into sex chromosome-specific copies, NLGN4X and NLGN4Y, remains uncharted territory. Just recently, the presence of substantial neuroligin-4 sequence differences between humans and laboratory mice, in which Nlgn4 is a pseudoautosomal gene, could be explained as a consequence of dramatic changes affecting the pseudoautosomal region on both sex chromosomes in a subset of rodents, the clade eumuroida. In this study, we describe the presence of sex chromosome-specific copies of neuroligin-4 genes in the Mongolian gerbil (Meriones unguiculatus) marking the first encounter of its kind in rodents. Gerbils are members of the family Muridae and are closely related to mice and rats. Our results have been incorporated into an extended evolutionary analysis covering primates, rodents, lagomorphs, treeshrews and culogos comprising together the mammalian superorder euarchontoglires. We gathered evidence that substantial changes in neuroligin-4 genes have also occurred outside eumuroida in other rodent species as well as in lagomorphs. These changes feature, e.g., a general reduction of its gene size, an increase in its average GC-content as well as in the third position (GC3) of synonymous codons, and the accumulation of repetitive sequences in line with previous observations. We further show conclusively that the diversification of neuroligin-4 in sex chromosome-specific copies has happened multiple times independently during mammal evolution proving that Y-chromosomal NLGN4Y genes do not originate from a single common NLGN4Y ancestor.
Collapse
Affiliation(s)
- Stephan Maxeiner
- Anatomy and Cell Biology, Saarland University, Homburg, Germany
- *Correspondence: Stephan Maxeiner,
| | - Fritz Benseler
- Department of Molecular Neurobiology, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Nils Brose
- Department of Molecular Neurobiology, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | | |
Collapse
|
22
|
Klarić TS, Lauc G. The dynamic brain N-glycome. Glycoconj J 2022; 39:443-471. [PMID: 35334027 DOI: 10.1007/s10719-022-10055-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 02/27/2022] [Accepted: 03/09/2022] [Indexed: 01/17/2023]
Abstract
The attachment of carbohydrates to other macromolecules, such as proteins or lipids, is an important regulatory mechanism termed glycosylation. One subtype of protein glycosylation is asparagine-linked glycosylation (N-glycosylation) which plays a key role in the development and normal functioning of the vertebrate brain. To better understand the role of N-glycans in neurobiology, it's imperative we analyse not only the functional roles of individual structures, but also the collective impact of large-scale changes in the brain N-glycome. The systematic study of the brain N-glycome is still in its infancy and data are relatively scarce. Nevertheless, the prevailing view has been that the neuroglycome is inherently restricted with limited capacity for variation. The development of improved methods for N-glycomics analysis of brain tissue has facilitated comprehensive characterisation of the complete brain N-glycome under various experimental conditions on a larger scale. Consequently, accumulating data suggest that it's more dynamic than previously recognised and that, within a general framework, it has a given capacity to change in response to both intrinsic and extrinsic stimuli. Here, we provide an overview of the many factors that can alter the brain N-glycome, including neurodevelopment, ageing, diet, stress, neuroinflammation, injury, and disease. Given this emerging evidence, we propose that the neuroglycome has a hitherto underappreciated plasticity and we discuss the therapeutic implications of this regarding the possible reversal of pathological changes via interventions. We also briefly review the merits and limitations of N-glycomics as an analytical method before reflecting on some of the outstanding questions in the field.
Collapse
Affiliation(s)
| | - Gordan Lauc
- Genos Glycoscience Research Laboratory, Zagreb, Croatia.,Faculty of Pharmacy and Biochemistry, University of Zagreb, Zagreb, Croatia
| |
Collapse
|
23
|
Liu J, Gao Z, Liu C, Liu T, Gao J, Cai Y, Fan X. Alteration of Gut Microbiota: New Strategy for Treating Autism Spectrum Disorder. Front Cell Dev Biol 2022; 10:792490. [PMID: 35309933 PMCID: PMC8929512 DOI: 10.3389/fcell.2022.792490] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Accepted: 01/20/2022] [Indexed: 12/12/2022] Open
Abstract
Autism spectrum disorder (ASD) is defined as a complex heterogeneous disorder and characterized by stereotyped behavior and deficits in communication and social interactions. The emerging microbial knowledge has pointed to a potential link between gut microbiota dysbiosis and ASD. Evidence from animal and human studies showed that shifts in composition and activity of the gut microbiota may causally contribute to the etiopathogenesis of core symptoms in the ASD individuals with gastrointestinal tract disturbances and act on microbiota-gut-brain. In this review, we summarized the characterized gut bacterial composition of ASD and the involvement of gut microbiota and their metabolites in the onset and progression of ASD; the possible underlying mechanisms are also highlighted. Given this correlation, we also provide an overview of the microbial-based therapeutic interventions such as probiotics, antibiotics, fecal microbiota transplantation therapy, and dietary interventions and address their potential benefits on behavioral symptoms of ASD. The precise contribution of altering gut microbiome to treating core symptoms in the ASD needs to be further clarified. It seemed to open up promising avenues to develop microbial-based therapies in ASD.
Collapse
Affiliation(s)
- Jiayin Liu
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing, China
- Battalion 5th of Cadet Brigade, Third Military Medical University (Army Medical University), Army Medical University, Chongqing, China
| | - Zhanyuan Gao
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing, China
- Battalion 5th of Cadet Brigade, Third Military Medical University (Army Medical University), Army Medical University, Chongqing, China
| | - Chuanqi Liu
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing, China
- Battalion 5th of Cadet Brigade, Third Military Medical University (Army Medical University), Army Medical University, Chongqing, China
| | - Tianyao Liu
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing, China
| | - Junwei Gao
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yun Cai
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing, China
| | - Xiaotang Fan
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing, China
| |
Collapse
|
24
|
Camporesi E, Nilsson J, Vrillon A, Cognat E, Hourregue C, Zetterberg H, Blennow K, Becker B, Brinkmalm A, Paquet C, Brinkmalm G. Quantification of the trans-synaptic partners neurexin-neuroligin in CSF of neurodegenerative diseases by parallel reaction monitoring mass spectrometry. EBioMedicine 2022; 75:103793. [PMID: 34990894 PMCID: PMC8743209 DOI: 10.1016/j.ebiom.2021.103793] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 12/15/2021] [Accepted: 12/16/2021] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Synaptic proteins are increasingly studied as biomarkers for synaptic dysfunction and loss, which are early and central events in Alzheimer's disease (AD) and strongly correlate with the degree of cognitive decline. In this study, we specifically investigated the synaptic binding partners neurexin (NRXN) and neuroligin (Nlgn) proteins, to assess their biomarker's potential. METHODS we developed a parallel reaction monitoring mass spectrometric method for the simultaneous quantification of NRXNs and Nlgns in cerebrospinal fluid (CSF) of neurodegenerative diseases, focusing on AD. Specifically, NRXN-1α, NRXN-1β, NRXN-2α, NRXN-3α and Nlgn1, Nlgn2, Nlgn3 and Nlgn4 proteins were targeted. FINDINGS The proteins were investigated in a clinical cohort including CSF from controls (n=22), mild cognitive impairment (MCI) due to AD (n=44), MCI due to other conditions (n=46), AD (n=77) and a group of non-AD dementia (n=28). No difference in levels of NRXNs and Nlgns was found between AD (both at dementia and MCI stages) or controls or the non-AD dementia group for any of the targeted proteins. NRXN and Nlgn proteins correlated strongly with each other, but only a weak correlation with the AD core biomarkers and the synaptic biomarkers neurogranin and growth-associated protein 43, was found, possibly reflecting different pathogenic processing at the synapse. INTERPRETATION we conclude that NRXN and Nlgn proteins do not represent suitable biomarkers for synaptic pathology in AD. The panel developed here could aid in future investigations of the potential involvement of NRXNs and Nlgns in synaptic dysfunction in other disorders of the central nervous system. FUNDING a full list of funding can be found under the acknowledgments section.
Collapse
Affiliation(s)
- Elena Camporesi
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at University of Gothenburg, Mölndal, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden.
| | - Johanna Nilsson
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at University of Gothenburg, Mölndal, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Agathe Vrillon
- Université de Paris, Cognitive Neurology Center, GHU Nord APHP Hospital Lariboisière Fernand Widal, Paris, France; Université de Paris, Inserm UMR S11-44 Therapeutic Optimization in Neuropsychopharmacology, Paris, France
| | - Emmanuel Cognat
- Université de Paris, Cognitive Neurology Center, GHU Nord APHP Hospital Lariboisière Fernand Widal, Paris, France; Université de Paris, Inserm UMR S11-44 Therapeutic Optimization in Neuropsychopharmacology, Paris, France
| | - Claire Hourregue
- Université de Paris, Cognitive Neurology Center, GHU Nord APHP Hospital Lariboisière Fernand Widal, Paris, France
| | - Henrik Zetterberg
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at University of Gothenburg, Mölndal, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden; UK Dementia Research Institute at UCL, London, UK; Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK; Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China
| | - Kaj Blennow
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at University of Gothenburg, Mölndal, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Bruno Becker
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at University of Gothenburg, Mölndal, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Ann Brinkmalm
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at University of Gothenburg, Mölndal, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Claire Paquet
- Université de Paris, Cognitive Neurology Center, GHU Nord APHP Hospital Lariboisière Fernand Widal, Paris, France; Université de Paris, Inserm UMR S11-44 Therapeutic Optimization in Neuropsychopharmacology, Paris, France
| | - Gunnar Brinkmalm
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at University of Gothenburg, Mölndal, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| |
Collapse
|
25
|
Doi M, Usui N, Shimada S. Prenatal Environment and Neurodevelopmental Disorders. Front Endocrinol (Lausanne) 2022; 13:860110. [PMID: 35370942 PMCID: PMC8964779 DOI: 10.3389/fendo.2022.860110] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 02/21/2022] [Indexed: 01/23/2023] Open
Abstract
The internal and external environment of the mother during the developmental stages of the fetus affects the offspring's health. According to the developmental origins of health and disease (DOHaD) theory, environmental factors influence the offspring and also affect health in adulthood. Recently, studies based on this theory have gained attracted attention because of their clinical utility in identifying the risk groups for various diseases. Neurodevelopmental disorders (NDDs) such as autism spectrum disorder (ASD) and attention-deficit hyperactivity disorder (ADHD) can be caused by exposure to certain prenatal environments during pregnancy. This review describes the latest findings on the effect of prenatal environment on the onset mechanism of NDDs based on the DOHaD theory. Unravelling the molecular mechanisms underlying the pathogenesis of NDDs is important, because there are no therapeutic drugs for these disorders. Furthermore, elucidating the relationship between the DOHaD theory and NDDs will contribute to the popularization of preventive medicine.
Collapse
Affiliation(s)
- Miyuki Doi
- Department of Neuroscience and Cell Biology, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Noriyoshi Usui
- Department of Neuroscience and Cell Biology, Graduate School of Medicine, Osaka University, Suita, Japan
- United Graduate School of Child Development, Osaka University, Suita, Japan
- Global Center for Medical Engineering and Informatics, Osaka University, Suita, Japan
- Addiction Research Unit, Osaka Psychiatric Research Center, Osaka Psychiatric Medical Center, Osaka, Japan
- *Correspondence: Noriyoshi Usui,
| | - Shoichi Shimada
- Department of Neuroscience and Cell Biology, Graduate School of Medicine, Osaka University, Suita, Japan
- United Graduate School of Child Development, Osaka University, Suita, Japan
- Global Center for Medical Engineering and Informatics, Osaka University, Suita, Japan
- Addiction Research Unit, Osaka Psychiatric Research Center, Osaka Psychiatric Medical Center, Osaka, Japan
| |
Collapse
|
26
|
Niescier RF, Lin YC. The Potential Role of AMPA Receptor Trafficking in Autism and Other Neurodevelopmental Conditions. Neuroscience 2021; 479:180-191. [PMID: 34571086 DOI: 10.1016/j.neuroscience.2021.09.013] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Revised: 09/06/2021] [Accepted: 09/15/2021] [Indexed: 12/21/2022]
Abstract
Autism Spectrum Disorder (ASD) is a multifaceted condition associated with difficulties in social interaction and communication. It also shares several comorbidities with other neurodevelopmental conditions. Intensive research examining the molecular basis and characteristics of ASD has revealed an association with a large number and variety of low-penetrance genes. Many of the variants associated with ASD are in genes underlying pathways involved in long-term potentiation (LTP) or depression (LTD). These mechanisms then control the tuning of neuronal connections in response to experience by modifying and trafficking ionotropic glutamate receptors at the post-synaptic areas. Despite the high genetic heterogeneity in ASD, surface trafficking of the α-amino-3-hydroxy-5-Methyl-4-isoxazolepropionate (AMPA) receptor is a vulnerable pathway in ASD. In this review, we discuss autism-related alterations in the trafficking of AMPA receptors, whose surface density and composition at the post-synapse determine the strength of the excitatory connection between neurons. We highlight genes associated with neurodevelopmental conditions that share the autism comorbidity, including Fragile X syndrome, Rett Syndrome, and Tuberous Sclerosis, as well as the autism-risk genes NLGNs, IQSEC2, DOCK4, and STXBP5, all of which are involved in regulating AMPAR trafficking to the post-synaptic surface.
Collapse
Affiliation(s)
- Robert F Niescier
- Program in Neuroscience, Hussman Institute for Autism, Baltimore, MD 21201, USA.
| | - Yu-Chih Lin
- Program in Neuroscience, Hussman Institute for Autism, Baltimore, MD 21201, USA.
| |
Collapse
|
27
|
Postsynaptic autism spectrum disorder genes and synaptic dysfunction. Neurobiol Dis 2021; 162:105564. [PMID: 34838666 DOI: 10.1016/j.nbd.2021.105564] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 11/17/2021] [Accepted: 11/23/2021] [Indexed: 12/20/2022] Open
Abstract
This review provides an overview of the synaptic dysfunction of neuronal circuits and the ensuing behavioral alterations caused by mutations in autism spectrum disorder (ASD)-linked genes directly or indirectly affecting the postsynaptic neuronal compartment. There are plenty of ASD risk genes, that may be broadly grouped into those involved in gene expression regulation (epigenetic regulation and transcription) and genes regulating synaptic activity (neural communication and neurotransmission). Notably, the effects mediated by ASD-associated genes can vary extensively depending on the developmental time and/or subcellular site of expression. Therefore, in order to gain a better understanding of the mechanisms of disruptions in postsynaptic function, an effort to better model ASD in experimental animals is required to improve standardization and increase reproducibility within and among studies. Such an effort holds promise to provide deeper insight into the development of these disorders and to improve the translational value of preclinical studies.
Collapse
|
28
|
Simone M, Margari L, Pompamea F, De Giacomo A, Gabellone A, Marzulli L, Palumbi R. Autism Spectrum Disorder and Duchenne Muscular Dystrophy: A Clinical Case on the Potential Role of the Dystrophin in Autism Neurobiology. J Clin Med 2021; 10:4370. [PMID: 34640386 PMCID: PMC8509154 DOI: 10.3390/jcm10194370] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 09/16/2021] [Accepted: 09/22/2021] [Indexed: 02/03/2023] Open
Abstract
A diagnosis of autism spectrum disorder is reported in up to 19% of dystrophinopathies. However, over the last ten years, only a few papers have been published on this topic. Therefore, further studies are required to analyze this association in depth and ultimately to understand the role of the brain dystrophin isoform in the pathogenesis of ASD and other neurodevelopmental disorders. In this paper, we report a clinical case of a patient affected by ASD and Duchenne muscular dystrophy, who carries a large deletion of the dystrophin gene. Then we present a brief overview of the literature about similar cases and about the potential role of the dystrophin protein in the neurobiology of autism spectrum disorder.
Collapse
Affiliation(s)
- Marta Simone
- Biomedical Sciences and Human Oncology Department, University of Bari “Aldo Moro”, 70124 Bari, Italy; (M.S.); (F.P.); (A.G.); (L.M.)
| | - Lucia Margari
- Biomedical Sciences and Human Oncology Department, University of Bari “Aldo Moro”, 70124 Bari, Italy; (M.S.); (F.P.); (A.G.); (L.M.)
| | - Francesco Pompamea
- Biomedical Sciences and Human Oncology Department, University of Bari “Aldo Moro”, 70124 Bari, Italy; (M.S.); (F.P.); (A.G.); (L.M.)
| | - Andrea De Giacomo
- Basic Medical Sciences, Neurosciences, and Sensory Organs Department, University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.D.G.); (R.P.)
| | - Alessandra Gabellone
- Biomedical Sciences and Human Oncology Department, University of Bari “Aldo Moro”, 70124 Bari, Italy; (M.S.); (F.P.); (A.G.); (L.M.)
| | - Lucia Marzulli
- Biomedical Sciences and Human Oncology Department, University of Bari “Aldo Moro”, 70124 Bari, Italy; (M.S.); (F.P.); (A.G.); (L.M.)
| | - Roberto Palumbi
- Basic Medical Sciences, Neurosciences, and Sensory Organs Department, University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.D.G.); (R.P.)
| |
Collapse
|
29
|
Neuroinflammation in autism spectrum disorders: Exercise as a "pharmacological" tool. Neurosci Biobehav Rev 2021; 129:63-74. [PMID: 34310976 DOI: 10.1016/j.neubiorev.2021.07.023] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Revised: 03/26/2021] [Accepted: 07/21/2021] [Indexed: 02/06/2023]
Abstract
The worldwide prevalence of ASD is around 1%. Although the pathogenesis of ASD is not entirely understood, it is recognized that a combination of genetic, epigenetics, environmental factors and immune system dysfunction can play an essential role in its development. It has been suggested that autism results from the central nervous system derangements due to low-grade chronic inflammatory reactions associated with the immune system activation. ASD individuals have increased microglial activation, density, and increased proinflammatory cytokines in the several brain regions. Autism has no available pharmacological treatments, however there are pedagogical and psychotherapeutic therapies, and pharmacological treatment, that help to control behavioral symptoms. Recent data indicate that exercise intervention programs may improve cognitive and behavioral symptoms in children with ASD. Exercise can also modify inflammatory profiles that will ameliorate associated metabolic disorders. This review highlights the involvement of neuroinflammation in ASD and the beneficial effects of physical exercise on managing ASD symptoms and associated comorbidities.
Collapse
|