101
|
Anreiter I, Tian YW, Soller M. The cap epitranscriptome: Early directions to a complex life as mRNA. Bioessays 2023; 45:e2200198. [PMID: 36529693 DOI: 10.1002/bies.202200198] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 12/07/2022] [Accepted: 12/08/2022] [Indexed: 12/23/2022]
Abstract
Animal, protist and viral messenger RNAs (mRNAs) are most prominently modified at the beginning by methylation of cap-adjacent nucleotides at the 2'-O-position of the ribose (cOMe) by dedicated cap methyltransferases (CMTrs). If the first nucleotide of an mRNA is an adenosine, PCIF1 can methylate at the N6 -position (m6 A), while internally the Mettl3/14 writer complex can methylate. These modifications are introduced co-transcriptionally to affect many aspects of gene expression including localisation to synapses and local translation. Of particular interest, transcription start sites of many genes are heterogeneous leading to sequence diversity at the beginning of mRNAs, which together with cOMe and m6 Am could constitute an extensive novel layer of gene expression control. Given the role of cOMe and m6 A in local gene expression at synapses and higher brain functions including learning and memory, such code could be implemented at the transcriptional level for lasting memories through local gene expression at synapses.
Collapse
Affiliation(s)
- Ina Anreiter
- Department of Biological Sciences, University of Toronto Scarborough, Toronto, Canada
| | - Yuan W Tian
- Birmingham Centre for Genome Biology, University of Birmingham, Birmingham, UK.,School of Biosciences, College of Life and Environmental Sciences, University of Birmingham, Birmingham, UK
| | - Matthias Soller
- Birmingham Centre for Genome Biology, University of Birmingham, Birmingham, UK.,School of Biosciences, College of Life and Environmental Sciences, University of Birmingham, Birmingham, UK
| |
Collapse
|
102
|
Cencelli G, Pacini L, De Luca A, Messia I, Gentile A, Kang Y, Nobile V, Tabolacci E, Jin P, Farace MG, Bagni C. Age-Dependent Dysregulation of APP in Neuronal and Skin Cells from Fragile X Individuals. Cells 2023; 12:758. [PMID: 36899894 PMCID: PMC10000963 DOI: 10.3390/cells12050758] [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: 11/06/2022] [Revised: 01/02/2023] [Accepted: 01/06/2023] [Indexed: 03/04/2023] Open
Abstract
Fragile X syndrome (FXS) is the most common form of monogenic intellectual disability and autism, caused by the absence of the functional fragile X messenger ribonucleoprotein 1 (FMRP). FXS features include increased and dysregulated protein synthesis, observed in both murine and human cells. Altered processing of the amyloid precursor protein (APP), consisting of an excess of soluble APPα (sAPPα), may contribute to this molecular phenotype in mice and human fibroblasts. Here we show an age-dependent dysregulation of APP processing in fibroblasts from FXS individuals, human neural precursor cells derived from induced pluripotent stem cells (iPSCs), and forebrain organoids. Moreover, FXS fibroblasts treated with a cell-permeable peptide that decreases the generation of sAPPα show restored levels of protein synthesis. Our findings suggest the possibility of using cell-based permeable peptides as a future therapeutic approach for FXS during a defined developmental window.
Collapse
Affiliation(s)
- Giulia Cencelli
- Department of Biomedicine and Prevention, Faculty of Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
- Institute of Neurosurgery, Fondazione Policlinico Universitario A. Gemelli Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Catholic University, 00168 Rome, Italy
| | - Laura Pacini
- Department of Biomedicine and Prevention, Faculty of Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
- Faculty of Medicine, UniCamillus, Saint Camillus International University of Health and Medical Sciences, 00131 Rome, Italy
| | - Anastasia De Luca
- Department of Biomedicine and Prevention, Faculty of Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Ilenia Messia
- Department of Biomedicine and Prevention, Faculty of Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Antonietta Gentile
- Department of Biomedicine and Prevention, Faculty of Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Roma, 00166 Rome, Italy
| | - Yunhee Kang
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Veronica Nobile
- Institute of Genomic Medicine, Fondazione Policlinico Universitario A. Gemelli Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Catholic University, 00168 Rome, Italy
| | - Elisabetta Tabolacci
- Institute of Genomic Medicine, Fondazione Policlinico Universitario A. Gemelli Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Catholic University, 00168 Rome, Italy
| | - Peng Jin
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Maria Giulia Farace
- Department of Biomedicine and Prevention, Faculty of Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Claudia Bagni
- Department of Biomedicine and Prevention, Faculty of Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
- Department of Fundamental Neurosciences, Faculty of Biology and Medicine, University of Lausanne, 1005 Lausanne, Switzerland
| |
Collapse
|
103
|
Abstract
The fragile X-related disorders are an important group of hereditary disorders that are caused by expanded CGG repeats in the 5' untranslated region of the FMR1 gene or by mutations in the coding sequence of this gene. Two categories of pathological CGG repeats are associated with these disorders, full mutation alleles and shorter premutation alleles. Individuals with full mutation alleles develop fragile X syndrome, which causes autism and intellectual disability, whereas those with premutation alleles, which have shorter CGG expansions, can develop fragile X-associated tremor/ataxia syndrome, a progressive neurodegenerative disease. Thus, fragile X-related disorders can manifest as neurodegenerative or neurodevelopmental disorders, depending on the size of the repeat expansion. Here, we review mouse models of fragile X-related disorders and discuss how they have informed our understanding of neurodegenerative and neurodevelopmental disorders. We also assess the translational value of these models for developing rational targeted therapies for intellectual disability and autism disorders.
Collapse
Affiliation(s)
- Rob Willemsen
- Department of Clinical Genetics, Erasmus University Medical Center, 3015 CN Rotterdam, the Netherlands. Department of Medical Genetics, University of Antwerp, 2000 Antwerp, Belgium
| | - R. Frank Kooy
- Department of Clinical Genetics, Erasmus University Medical Center, 3015 CN Rotterdam, the Netherlands. Department of Medical Genetics, University of Antwerp, 2000 Antwerp, Belgium
| |
Collapse
|
104
|
Brown RE. Sex Differences in Neurodevelopment and Its Disorders. NEURODEVELOPMENTAL PEDIATRICS 2023:179-212. [DOI: 10.1007/978-3-031-20792-1_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
105
|
Guo M, Xie P, Liu S, Luan G, Li T. Epilepsy and Autism Spectrum Disorder (ASD): The Underlying Mechanisms and Therapy Targets Related to Adenosine. Curr Neuropharmacol 2023; 21:54-66. [PMID: 35794774 PMCID: PMC10193761 DOI: 10.2174/1570159x20666220706100136] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 03/23/2022] [Accepted: 04/26/2022] [Indexed: 02/04/2023] Open
Abstract
Epilepsy and autism spectrum disorder (ASD) are highly mutually comorbid, suggesting potential overlaps in genetic etiology, pathophysiology, and neurodevelopmental abnormalities. Adenosine, an endogenous anticonvulsant and neuroprotective neuromodulator of the brain, has been proved to affect the process of epilepsy and ASD. On the one hand, adenosine plays a crucial role in preventing the progression and development of epilepsy through adenosine receptordependent and -independent ways. On the other hand, adenosine signaling can not only regulate core symptoms but also improve comorbid disorders in ASD. Given the important role of adenosine in epilepsy and ASD, therapeutic strategies related to adenosine, including the ketogenic diet, neuromodulation therapy, and adenosine augmentation therapy, have been suggested for the arrangement of epilepsy and ASD. There are several proposals in this review. Firstly, it is necessary to further discuss the relationship between both diseases based on the comorbid symptoms and mechanisms of epilepsy and ASD. Secondly, it is important to explore the role of adenosine involved in epilepsy and ASD. Lastly, potential therapeutic value and clinical approaches of adenosine-related therapies in treating epilepsy and ASD need to be emphasized.
Collapse
Affiliation(s)
- Mengyi Guo
- Department of Brain Institute, Center of Epilepsy, Beijing Institute for Brain Disorders, Beijing Key Laboratory of Epilepsy Research, Sanbo Brain Hospital, Capital Medical University, Beijing 100093, China
- Department of Neurology, Center of Epilepsy, Beijing Institute for Brain Disorders, Sanbo Brain Hospital, Capital Medical University, Beijing 100093, China
| | - Pandeng Xie
- Department of Brain Institute, Center of Epilepsy, Beijing Institute for Brain Disorders, Beijing Key Laboratory of Epilepsy Research, Sanbo Brain Hospital, Capital Medical University, Beijing 100093, China
- Department of Neurology, Center of Epilepsy, Beijing Institute for Brain Disorders, Sanbo Brain Hospital, Capital Medical University, Beijing 100093, China
| | - Siqi Liu
- Department of Brain Institute, Center of Epilepsy, Beijing Institute for Brain Disorders, Beijing Key Laboratory of Epilepsy Research, Sanbo Brain Hospital, Capital Medical University, Beijing 100093, China
- Department of Neurology, Center of Epilepsy, Beijing Institute for Brain Disorders, Sanbo Brain Hospital, Capital Medical University, Beijing 100093, China
| | - Guoming Luan
- Department of Brain Institute, Center of Epilepsy, Beijing Institute for Brain Disorders, Beijing Key Laboratory of Epilepsy Research, Sanbo Brain Hospital, Capital Medical University, Beijing 100093, China
- Department of Neurology, Center of Epilepsy, Beijing Institute for Brain Disorders, Sanbo Brain Hospital, Capital Medical University, Beijing 100093, China
| | - Tianfu Li
- Department of Brain Institute, Center of Epilepsy, Beijing Institute for Brain Disorders, Beijing Key Laboratory of Epilepsy Research, Sanbo Brain Hospital, Capital Medical University, Beijing 100093, China
- Department of Neurology, Center of Epilepsy, Beijing Institute for Brain Disorders, Sanbo Brain Hospital, Capital Medical University, Beijing 100093, China
| |
Collapse
|
106
|
Buzzelli V, Carbone E, Manduca A, Schiavi S, Feo A, Perederiy JV, Ambert KH, Hausman M, Trezza V. Psilocybin mitigates the cognitive deficits observed in a rat model of Fragile X syndrome. Psychopharmacology (Berl) 2023; 240:137-147. [PMID: 36469097 DOI: 10.1007/s00213-022-06286-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 11/20/2022] [Indexed: 12/12/2022]
Abstract
RATIONALE Fragile X syndrome (FXS) is the most common form of inherited intellectual disability (ID) and the leading monogenic cause of autism spectrum disorder (ASD). Serotonergic neurotransmission has a key role in the modulation of neuronal activity during development, and therefore, it has been hypothesized to be involved in ASD and co-occurring conditions including FXS. As serotonin is involved in synaptic remodeling and maturation, serotonergic insufficiency during childhood may have a compounding effect on brain patterning in neurodevelopmental disorders, manifesting as behavioral and emotional symptoms. Thus, compounds that stimulate serotonergic signaling such as psilocybin may offer promise as effective early interventions for developmental disorders such as ASD and FXS. OBJECTIVES The aim of the present study was to test whether different protocols of psilocybin administration mitigate cognitive deficits displayed by the recently validated Fmr1-Δexon 8 rat model of ASD, which is also a model of FXS. RESULTS Our results revealed that systemic and oral administration of psilocybin microdoses normalizes the aberrant cognitive performance displayed by adolescent Fmr1-Δexon 8 rats in the short-term version of the novel object recognition test-a measure of exploratory behavior, perception, and recognition. CONCLUSIONS These data support the hypothesis that serotonin-modulating drugs such as psilocybin may be useful to ameliorate ASD-related cognitive deficits. Overall, this study provides evidence of the beneficial effects of different schedules of psilocybin treatment in mitigating the short-term cognitive deficit observed in a rat model of FXS.
Collapse
Affiliation(s)
- Valeria Buzzelli
- Department of Science, Section of Biomedical Sciences and Technologies, University "Roma Tre", Viale G. Marconi 446, 00146, Rome, Italy
| | - Emilia Carbone
- Department of Science, Section of Biomedical Sciences and Technologies, University "Roma Tre", Viale G. Marconi 446, 00146, Rome, Italy
| | - Antonia Manduca
- Department of Science, Section of Biomedical Sciences and Technologies, University "Roma Tre", Viale G. Marconi 446, 00146, Rome, Italy.,Neuroendocrinology, Metabolism and Neuropharmacology Unit, IRCSS Fondazione Santa Lucia, Rome, Italy
| | - Sara Schiavi
- Department of Science, Section of Biomedical Sciences and Technologies, University "Roma Tre", Viale G. Marconi 446, 00146, Rome, Italy
| | - Alessandro Feo
- Department of Science, Section of Biomedical Sciences and Technologies, University "Roma Tre", Viale G. Marconi 446, 00146, Rome, Italy
| | | | - Kyle H Ambert
- Nova Mentis Life Science Corp., Vancouver, BC, Canada
| | | | - Viviana Trezza
- Department of Science, Section of Biomedical Sciences and Technologies, University "Roma Tre", Viale G. Marconi 446, 00146, Rome, Italy.
| |
Collapse
|
107
|
Gribkoff VK, Kaczmarek LK. The Difficult Path to the Discovery of Novel Treatments in Psychiatric Disorders. ADVANCES IN NEUROBIOLOGY 2023; 30:255-285. [PMID: 36928854 PMCID: PMC10599454 DOI: 10.1007/978-3-031-21054-9_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/18/2023]
Abstract
CNS diseases, including psychiatric disorders, represent a significant opportunity for the discovery and development of new drugs and therapeutic treatments with the potential to have a significant impact on human health. CNS diseases, however, present particular challenges to therapeutic discovery efforts, and psychiatric diseases/disorders may be among the most difficult. With specific exceptions such as psychostimulants for ADHD, a large number of psychiatric patients are resistant to existing treatments. In addition, clinicians have no way of knowing which psychiatric patients will respond to which drugs. By definition, psychiatric diagnoses are syndromal in nature; determinations of efficacy are often self-reported, and drug discovery is largely model-based. While such models of psychiatric disease are amenable to screening for new drugs, whether cellular or whole-animal based, they have only modest face validity and, more importantly, predictive validity. Multiple academic, pharmaceutical industry, and government agencies are dedicated to the translation of new findings about the neurobiology of major psychiatric disorders into the discovery and advancement of novel therapies. The collaboration of these agencies provide a pathway for developing new therapeutics. These efforts will be greatly helped by recent advances in understanding the genetic bases of psychiatric disorders, the ongoing search for diagnostic and therapy-responsive biomarkers, and the validation of new animal models.
Collapse
Affiliation(s)
- Valentin K Gribkoff
- Department of Internal Medicine, Section on Endocrinology, Yale University School of Medicine, New Haven, CT, USA.
| | - Leonard K Kaczmarek
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT, USA.
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
108
|
Leahy SN, Song C, Vita DJ, Broadie K. FMRP activity and control of Csw/SHP2 translation regulate MAPK-dependent synaptic transmission. PLoS Biol 2023; 21:e3001969. [PMID: 36701299 PMCID: PMC9879533 DOI: 10.1371/journal.pbio.3001969] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 12/16/2022] [Indexed: 01/27/2023] Open
Abstract
Noonan syndrome (NS) and NS with multiple lentigines (NSML) cognitive dysfunction are linked to SH2 domain-containing protein tyrosine phosphatase-2 (SHP2) gain-of-function (GoF) and loss-of-function (LoF), respectively. In Drosophila disease models, we find both SHP2 mutations from human patients and corkscrew (csw) homolog LoF/GoF elevate glutamatergic transmission. Cell-targeted RNAi and neurotransmitter release analyses reveal a presynaptic requirement. Consistently, all mutants exhibit reduced synaptic depression during high-frequency stimulation. Both LoF and GoF mutants also show impaired synaptic plasticity, including reduced facilitation, augmentation, and post-tetanic potentiation. NS/NSML diseases are characterized by elevated MAPK/ERK signaling, and drugs suppressing this signaling restore normal neurotransmission in mutants. Fragile X syndrome (FXS) is likewise characterized by elevated MAPK/ERK signaling. Fragile X Mental Retardation Protein (FMRP) binds csw mRNA and neuronal Csw protein is elevated in Drosophila fragile X mental retardation 1 (dfmr1) nulls. Moreover, phosphorylated ERK (pERK) is increased in dfmr1 and csw null presynaptic boutons. We find presynaptic pERK activation in response to stimulation is reduced in dfmr1 and csw nulls. Trans-heterozygous csw/+; dfmr1/+ recapitulate elevated presynaptic pERK activation and function, showing FMRP and Csw/SHP2 act within the same signaling pathway. Thus, a FMRP and SHP2 MAPK/ERK regulative mechanism controls basal and activity-dependent neurotransmission strength.
Collapse
Affiliation(s)
- Shannon N. Leahy
- Department of Biological Sciences, Vanderbilt University and Medical Center, Nashville, Tennessee, United States of America
| | - Chunzhu Song
- Department of Biological Sciences, Vanderbilt University and Medical Center, Nashville, Tennessee, United States of America
| | - Dominic J. Vita
- Department of Biological Sciences, Vanderbilt University and Medical Center, Nashville, Tennessee, United States of America
| | - Kendal Broadie
- Department of Biological Sciences, Vanderbilt University and Medical Center, Nashville, Tennessee, United States of America
- Department of Cell and Developmental Biology, Vanderbilt University and Medical Center, Nashville, Tennessee, United States of America
- Department of Pharmacology, Vanderbilt University and Medical Center, Nashville, Tennessee, United States of America
- Vanderbilt Brain Institute, Vanderbilt University and Medical Center, Nashville, Tennessee, United States of America
| |
Collapse
|
109
|
Scott DN, Frank MJ. Adaptive control of synaptic plasticity integrates micro- and macroscopic network function. Neuropsychopharmacology 2023; 48:121-144. [PMID: 36038780 PMCID: PMC9700774 DOI: 10.1038/s41386-022-01374-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 06/23/2022] [Accepted: 06/24/2022] [Indexed: 11/09/2022]
Abstract
Synaptic plasticity configures interactions between neurons and is therefore likely to be a primary driver of behavioral learning and development. How this microscopic-macroscopic interaction occurs is poorly understood, as researchers frequently examine models within particular ranges of abstraction and scale. Computational neuroscience and machine learning models offer theoretically powerful analyses of plasticity in neural networks, but results are often siloed and only coarsely linked to biology. In this review, we examine connections between these areas, asking how network computations change as a function of diverse features of plasticity and vice versa. We review how plasticity can be controlled at synapses by calcium dynamics and neuromodulatory signals, the manifestation of these changes in networks, and their impacts in specialized circuits. We conclude that metaplasticity-defined broadly as the adaptive control of plasticity-forges connections across scales by governing what groups of synapses can and can't learn about, when, and to what ends. The metaplasticity we discuss acts by co-opting Hebbian mechanisms, shifting network properties, and routing activity within and across brain systems. Asking how these operations can go awry should also be useful for understanding pathology, which we address in the context of autism, schizophrenia and Parkinson's disease.
Collapse
Affiliation(s)
- Daniel N Scott
- Cognitive Linguistic, and Psychological Sciences, Brown University, Providence, RI, USA.
- Carney Institute for Brain Science, Brown University, Providence, RI, USA.
| | - Michael J Frank
- Cognitive Linguistic, and Psychological Sciences, Brown University, Providence, RI, USA.
- Carney Institute for Brain Science, Brown University, Providence, RI, USA.
| |
Collapse
|
110
|
Ma B, Shan X, Yu J, Zhu T, Li R, Lv H, Cheng H, Zhang T, Wang L, Wei F, Meng B, Yuan X, Mei B, Zhang XY, Li WG, Li F. Social deficits via dysregulated Rac1-dependent excitability control of prefrontal cortical neurons and increased GABA/glutamate ratios. Cell Rep 2022; 41:111722. [PMID: 36450249 DOI: 10.1016/j.celrep.2022.111722] [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: 03/04/2022] [Revised: 04/26/2022] [Accepted: 11/02/2022] [Indexed: 12/03/2022] Open
Abstract
Identifying symptom-specific convergent mechanisms for neurodevelopmental disorders is a promising strategy in advancing therapies. Here, we show that bidirectional dysregulation of Rac1 activity in the medial prefrontal cortex (mPFC) dictates shared social deficits in mice. Selective upregulation or downregulation of Rac1 activity in glutamatergic or fast-spiking GABAergic neurons results in excessive or inadequate control of excitability combined with a decrease in glutamate or an increase in GABA concentrations and an increase in the GABA/glutamate ratio, which is responsible for social deficits. Notably, the autism model of Shank3B knockout mice exhibits aberrantly enhanced Rac1 activity, reduced glutamate concentrations, and pyramidal neuron excitability in mPFC accompanied with social deficits, which were corrected by either excitatory-neuron-specific downregulation of Rac1 activity or upregulation of neuronal excitability. Thus, this work shows a convergence between genetic autism risk factors, dysregulation of Rac1 signaling, and excitation-inhibition imbalance, enabling mechanism-based stratification of patients with social deficits.
Collapse
Affiliation(s)
- Bingke Ma
- Shanghai Key Laboratory of Brain Functional Genomics (Ministry of Education), School of Life Sciences, East China Normal University, Shanghai 200062, China; Developmental and Behavioral Pediatric Department, Brain and Behavioral Research Unit of Shanghai Institute for Pediatric Research and Ministry of Education - Shanghai Key Laboratory for Children's Environmental Health, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China; Developmental and Behavioral Pediatric Department, Shanghai Xinhua Children's Hospital, Shanghai 200092, China
| | - Xingyue Shan
- Shanghai Key Laboratory of Brain Functional Genomics (Ministry of Education), School of Life Sciences, East China Normal University, Shanghai 200062, China; Developmental and Behavioral Pediatric Department, Brain and Behavioral Research Unit of Shanghai Institute for Pediatric Research and Ministry of Education - Shanghai Key Laboratory for Children's Environmental Health, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China; Developmental and Behavioral Pediatric Department, Shanghai Xinhua Children's Hospital, Shanghai 200092, China
| | - Juehua Yu
- Developmental and Behavioral Pediatric Department, Brain and Behavioral Research Unit of Shanghai Institute for Pediatric Research and Ministry of Education - Shanghai Key Laboratory for Children's Environmental Health, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China; Developmental and Behavioral Pediatric Department, Shanghai Xinhua Children's Hospital, Shanghai 200092, China; Center for Experimental Studies and Research, The First Affiliated Hospital of Kunming Medical University, Kunming 650032, China
| | - Tailin Zhu
- Developmental and Behavioral Pediatric Department, Brain and Behavioral Research Unit of Shanghai Institute for Pediatric Research and Ministry of Education - Shanghai Key Laboratory for Children's Environmental Health, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China; Developmental and Behavioral Pediatric Department, Shanghai Xinhua Children's Hospital, Shanghai 200092, China
| | - Ren Li
- Institute of Science and Technology for Brain-Inspired Intelligence, Ministry of Education - Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Ministry of Education Frontiers Center for Brain Science, Fudan University, Shanghai 200433, China
| | - Hui Lv
- Developmental and Behavioral Pediatric Department, Brain and Behavioral Research Unit of Shanghai Institute for Pediatric Research and Ministry of Education - Shanghai Key Laboratory for Children's Environmental Health, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China; Developmental and Behavioral Pediatric Department, Shanghai Xinhua Children's Hospital, Shanghai 200092, China
| | - Haidi Cheng
- Shanghai Key Laboratory of Brain Functional Genomics (Ministry of Education), School of Life Sciences, East China Normal University, Shanghai 200062, China; Developmental and Behavioral Pediatric Department, Brain and Behavioral Research Unit of Shanghai Institute for Pediatric Research and Ministry of Education - Shanghai Key Laboratory for Children's Environmental Health, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China; Developmental and Behavioral Pediatric Department, Shanghai Xinhua Children's Hospital, Shanghai 200092, China
| | - Tiantian Zhang
- Shanghai Key Laboratory of Brain Functional Genomics (Ministry of Education), School of Life Sciences, East China Normal University, Shanghai 200062, China; Developmental and Behavioral Pediatric Department, Brain and Behavioral Research Unit of Shanghai Institute for Pediatric Research and Ministry of Education - Shanghai Key Laboratory for Children's Environmental Health, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China; Developmental and Behavioral Pediatric Department, Shanghai Xinhua Children's Hospital, Shanghai 200092, China
| | - Lihua Wang
- Developmental and Behavioral Pediatric Department, Brain and Behavioral Research Unit of Shanghai Institute for Pediatric Research and Ministry of Education - Shanghai Key Laboratory for Children's Environmental Health, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China; Developmental and Behavioral Pediatric Department, Shanghai Xinhua Children's Hospital, Shanghai 200092, China
| | - Feiyang Wei
- Developmental and Behavioral Pediatric Department, Brain and Behavioral Research Unit of Shanghai Institute for Pediatric Research and Ministry of Education - Shanghai Key Laboratory for Children's Environmental Health, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China; Developmental and Behavioral Pediatric Department, Shanghai Xinhua Children's Hospital, Shanghai 200092, China
| | - Bo Meng
- Shanghai Key Laboratory of Brain Functional Genomics (Ministry of Education), School of Life Sciences, East China Normal University, Shanghai 200062, China
| | - Xiaobing Yuan
- Shanghai Key Laboratory of Brain Functional Genomics (Ministry of Education), School of Life Sciences, East China Normal University, Shanghai 200062, China
| | - Bing Mei
- Shanghai Key Laboratory of Brain Functional Genomics (Ministry of Education), School of Life Sciences, East China Normal University, Shanghai 200062, China.
| | - Xiao-Yong Zhang
- Institute of Science and Technology for Brain-Inspired Intelligence, Ministry of Education - Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Ministry of Education Frontiers Center for Brain Science, Fudan University, Shanghai 200433, China.
| | - Wei-Guang Li
- Department of Rehabilitation Medicine, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, Fudan University, Shanghai 200032, China.
| | - Fei Li
- Developmental and Behavioral Pediatric Department, Brain and Behavioral Research Unit of Shanghai Institute for Pediatric Research and Ministry of Education - Shanghai Key Laboratory for Children's Environmental Health, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China; Developmental and Behavioral Pediatric Department, Shanghai Xinhua Children's Hospital, Shanghai 200092, China.
| |
Collapse
|
111
|
Bernard C, Exposito-Alonso D, Selten M, Sanalidou S, Hanusz-Godoy A, Aguilera A, Hamid F, Oozeer F, Maeso P, Allison L, Russell M, Fleck RA, Rico B, Marín O. Cortical wiring by synapse type-specific control of local protein synthesis. Science 2022; 378:eabm7466. [PMID: 36423280 DOI: 10.1126/science.abm7466] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/14/2024]
Abstract
Neurons use local protein synthesis to support their morphological complexity, which requires independent control across multiple subcellular compartments up to the level of individual synapses. We identify a signaling pathway that regulates the local synthesis of proteins required to form excitatory synapses on parvalbumin-expressing (PV+) interneurons in the mouse cerebral cortex. This process involves regulation of the TSC subunit 2 (Tsc2) by the Erb-B2 receptor tyrosine kinase 4 (ErbB4), which enables local control of messenger RNA {mRNA} translation in a cell type-specific and synapse type-specific manner. Ribosome-associated mRNA profiling reveals a molecular program of synaptic proteins downstream of ErbB4 signaling required to form excitatory inputs on PV+ interneurons. Thus, specific connections use local protein synthesis to control synapse formation in the nervous system.
Collapse
Affiliation(s)
- Clémence Bernard
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, UK
- MRC Centre for Neurodevelopmental Disorders, King's College London, London SE1 1UL, UK
| | - David Exposito-Alonso
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, UK
- MRC Centre for Neurodevelopmental Disorders, King's College London, London SE1 1UL, UK
| | - Martijn Selten
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, UK
- MRC Centre for Neurodevelopmental Disorders, King's College London, London SE1 1UL, UK
| | - Stella Sanalidou
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, UK
- MRC Centre for Neurodevelopmental Disorders, King's College London, London SE1 1UL, UK
| | - Alicia Hanusz-Godoy
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, UK
- MRC Centre for Neurodevelopmental Disorders, King's College London, London SE1 1UL, UK
| | - Alfonso Aguilera
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, UK
- MRC Centre for Neurodevelopmental Disorders, King's College London, London SE1 1UL, UK
| | - Fursham Hamid
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, UK
- MRC Centre for Neurodevelopmental Disorders, King's College London, London SE1 1UL, UK
| | - Fazal Oozeer
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, UK
- MRC Centre for Neurodevelopmental Disorders, King's College London, London SE1 1UL, UK
| | - Patricia Maeso
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, UK
- MRC Centre for Neurodevelopmental Disorders, King's College London, London SE1 1UL, UK
| | - Leanne Allison
- Centre for Ultrastructural Imaging, King's College London, London SE1 1UL, UK
| | - Matthew Russell
- Centre for Ultrastructural Imaging, King's College London, London SE1 1UL, UK
| | - Roland A Fleck
- Centre for Ultrastructural Imaging, King's College London, London SE1 1UL, UK
| | - Beatriz Rico
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, UK
- MRC Centre for Neurodevelopmental Disorders, King's College London, London SE1 1UL, UK
| | - Oscar Marín
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, UK
- MRC Centre for Neurodevelopmental Disorders, King's College London, London SE1 1UL, UK
| |
Collapse
|
112
|
Chatterjee D, Beaulieu JM. Inhibition of glycogen synthase kinase 3 by lithium, a mechanism in search of specificity. Front Mol Neurosci 2022; 15:1028963. [PMID: 36504683 PMCID: PMC9731798 DOI: 10.3389/fnmol.2022.1028963] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 10/24/2022] [Indexed: 11/25/2022] Open
Abstract
Inhibition of Glycogen synthase kinase 3 (GSK3) is a popular explanation for the effects of lithium ions on mood regulation in bipolar disorder and other mental illnesses, including major depression, cyclothymia, and schizophrenia. Contribution of GSK3 is supported by evidence obtained from animal and patient derived model systems. However, the two GSK3 enzymes, GSK3α and GSK3β, have more than 100 validated substrates. They are thus central hubs for major biological functions, such as dopamine-glutamate neurotransmission, synaptic plasticity (Hebbian and homeostatic), inflammation, circadian regulation, protein synthesis, metabolism, inflammation, and mitochondrial functions. The intricate contributions of GSK3 to several biological processes make it difficult to identify specific mechanisms of mood stabilization for therapeutic development. Identification of GSK3 substrates involved in lithium therapeutic action is thus critical. We provide an overview of GSK3 biological functions and substrates for which there is evidence for a contribution to lithium effects. A particular focus is given to four of these: the transcription factor cAMP response element-binding protein (CREB), the RNA-binding protein FXR1, kinesin subunits, and the cytoskeletal regulator CRMP2. An overview of how co-regulation of these substrates may result in shared outcomes is also presented. Better understanding of how inhibition of GSK3 contributes to the therapeutic effects of lithium should allow for identification of more specific targets for future drug development. It may also provide a framework for the understanding of how lithium effects overlap with those of other drugs such as ketamine and antipsychotics, which also inhibit brain GSK3.
Collapse
Affiliation(s)
| | - Jean Martin Beaulieu
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
113
|
Cogram P, Fernández-Beltrán LC, Casarejos MJ, Sánchez-Yepes S, Rodríguez-Martín E, García-Rubia A, Sánchez-Barrena MJ, Gil C, Martínez A, Mansilla A. The inhibition of NCS-1 binding to Ric8a rescues fragile X syndrome mice model phenotypes. Front Neurosci 2022; 16:1007531. [PMID: 36466176 PMCID: PMC9709425 DOI: 10.3389/fnins.2022.1007531] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Accepted: 09/26/2022] [Indexed: 01/01/2024] Open
Abstract
Fragile X syndrome (FXS) is caused by the loss of function of Fragile X mental retardation protein (FMRP). FXS is one of the leading monogenic causes of intellectual disability (ID) and autism. Although it is caused by the failure of a single gene, FMRP that functions as an RNA binding protein affects a large number of genes secondarily. All these genes represent hundreds of potential targets and different mechanisms that account for multiple pathological features, thereby hampering the search for effective treatments. In this scenario, it seems desirable to reorient therapies toward more general approaches. Neuronal calcium sensor 1 (NCS-1), through its interaction with the guanine-exchange factor Ric8a, regulates the number of synapses and the probability of the release of a neurotransmitter, the two neuronal features that are altered in FXS and other neurodevelopmental disorders. Inhibitors of the NCS-1/Ric8a complex have been shown to be effective in restoring abnormally high synapse numbers as well as improving associative learning in FMRP mutant flies. Here, we demonstrate that phenothiazine FD44, an NCS-1/Ric8a inhibitor, has strong inhibition ability in situ and sufficient bioavailability in the mouse brain. More importantly, administration of FD44 to two different FXS mouse models restores well-known FXS phenotypes, such as hyperactivity, associative learning, aggressive behavior, stereotype, or impaired social approach. It has been suggested that dopamine (DA) may play a relevant role in the behavior and in neurodevelopmental disorders in general. We have measured DA and its metabolites in different brain regions, finding a higher metabolic rate in the limbic area, which is also restored with FD44 treatment. Therefore, in addition to confirming that the NCS-1/Ric8a complex is an excellent therapeutic target, we demonstrate the rescue effect of its inhibitor on the behavior of cognitive and autistic FXS mice and show DA metabolism as a FXS biochemical disease marker.
Collapse
Affiliation(s)
- Patricia Cogram
- Department of Genetics, Institute of Ecology and Biodiversity (IEB), Faculty of Sciences, Universidad de Chile, Santiago, Chile
- FRAXA-DVI, FRAXA Research Foundation, Santiago, Chile
| | - Luis C. Fernández-Beltrán
- Department of Neurobiology, Instituto Ramón y Cajal de Investigación Sanitaria, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - María José Casarejos
- Department of Neurobiology, Instituto Ramón y Cajal de Investigación Sanitaria, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Sonia Sánchez-Yepes
- Department of Neurobiology, Instituto Ramón y Cajal de Investigación Sanitaria, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Eulalia Rodríguez-Martín
- Department of Immunology, Instituto Ramón y Cajal de Investigación Sanitaria, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Alfonso García-Rubia
- Centro de Investigaciones Biológicas Margarita Salas-CSIC, Madrid, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | | | - Carmen Gil
- Centro de Investigaciones Biológicas Margarita Salas-CSIC, Madrid, Spain
| | - Ana Martínez
- Centro de Investigaciones Biológicas Margarita Salas-CSIC, Madrid, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Alicia Mansilla
- Department of Neurobiology, Instituto Ramón y Cajal de Investigación Sanitaria, Hospital Universitario Ramón y Cajal, Madrid, Spain
- Department of Biology Systems, Universidad de Alcala, Madrid, Spain
| |
Collapse
|
114
|
Sun H, Wu M, Wang M, Zhang X, Zhu J. The regulatory role of endoplasmic reticulum chaperone proteins in neurodevelopment. Front Neurosci 2022; 16:1032607. [DOI: 10.3389/fnins.2022.1032607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 10/25/2022] [Indexed: 11/16/2022] Open
Abstract
The endoplasmic reticulum (ER) is the largest tubular reticular organelle spanning the cell. As the main site of protein synthesis, Ca2+ homeostasis maintenance and lipid metabolism, the ER plays a variety of essential roles in eukaryotic cells, with ER molecular chaperones participate in all these processes. In recent years, it has been reported that the abnormal expression of ER chaperones often leads to a variety of neurodevelopmental disorders (NDDs), including abnormal neuronal migration, neuronal morphogenesis, and synaptic function. Neuronal development is a complex and precisely regulated process. Currently, the mechanism by which neural development is regulated at the ER level remains under investigation. Therefore, in this work, we reviewed the recent advances in the roles of ER chaperones in neural development and developmental disorders caused by the deficiency of these molecular chaperones.
Collapse
|
115
|
Speranza L, Filiz KD, Goebel S, Perrone-Capano C, Pulcrano S, Volpicelli F, Francesconi A. Combined DiI and Antibody Labeling Reveals Complex Dysgenesis of Hippocampal Dendritic Spines in a Mouse Model of Fragile X Syndrome. Biomedicines 2022; 10:2692. [PMID: 36359212 PMCID: PMC9687937 DOI: 10.3390/biomedicines10112692] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 10/18/2022] [Accepted: 10/19/2022] [Indexed: 11/30/2022] Open
Abstract
Structural, functional, and molecular alterations in excitatory spines are a common hallmark of many neurodevelopmental disorders including intellectual disability and autism. Here, we describe an optimized methodology, based on combined use of DiI and immunofluorescence, for rapid and sensitive characterization of the structure and composition of spines in native brain tissue. We successfully demonstrate the applicability of this approach by examining the properties of hippocampal spines in juvenile Fmr1 KO mice, a mouse model of Fragile X Syndrome. We find that mutant mice display pervasive dysgenesis of spines evidenced by an overabundance of both abnormally elongated thin spines and cup-shaped spines, in combination with reduced density of mushroom spines. We further find that mushroom spines expressing the actin-binding protein Synaptopodin-a marker for spine apparatus-are more prevalent in mutant mice. Previous work identified spines with Synaptopodin/spine apparatus as the locus of mGluR-LTD, which is abnormally elevated in Fmr1 KO mice. Altogether, our data suggest this enhancement may be linked to the preponderance of this subset of spines in the mutant. Overall, these findings demonstrate the sensitivity and versatility of the optimized methodology by uncovering a novel facet of spine dysgenesis in Fmr1 KO mice.
Collapse
Affiliation(s)
- Luisa Speranza
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, New York, NY 10461, USA
| | - Kardelen Dalım Filiz
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, 80131 Naples, Italy
| | - Sarah Goebel
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, New York, NY 10461, USA
| | - Carla Perrone-Capano
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, 80131 Naples, Italy
| | - Salvatore Pulcrano
- Institute of Genetics and Biophysics “A. Buzzati-Traverso”, C.N.R., 80131 Naples, Italy
| | - Floriana Volpicelli
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, 80131 Naples, Italy
| | - Anna Francesconi
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, New York, NY 10461, USA
| |
Collapse
|
116
|
Chakraborty A, Grageda A, Kuznetsov VA, Feng W. A Double Jeopardy: Loss of FMRP Results in DSB and Down-regulated DNA Repair. 21ST CENTURY PATHOLOGY 2022; 2:125. [PMID: 36688938 PMCID: PMC9850805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Our understanding of the molecular functions of the nucleocytoplasmic FMRP protein, which, if absent or dysfunctional, causes the fragile X syndrome (FXS), largely revolves around its involvement in protein translation regulation in the cytoplasm. Recent studies have begun honing in on the nuclear and genomic functions of FMRP. We have shown that during DNA replication stress, cells derived from FXS patients sustain increased level of R-loop formation and DNA double strand breaks. Here, we describe a transcriptomic analysis of these cells in order to identify those genes most impacted by the loss of FMRP with and without replication stress. We show that FMRP loss causes transcriptomic changes previously reported in untreated conditions. Importantly, we also show that replication stress, in addition to causing excess of DSB, results in down-regulation of transcription in virtually all DNA repair pathways. This finding suggests that despite normal DNA damage response, FXS patient-derived cells experience R-loop-induced DNA breakage as well as impaired DNA repair functions, effectively a double jeopardy. We suggest that it is imperative to deepen the understanding of the nuclear functions, particularly a genome protective function, of FMRP, which will lead to discoveries of novel therapeutic interventions for the FXS.
Collapse
Affiliation(s)
- Arijita Chakraborty
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, New York, USA
- Tessera Therapeutics, Somerville, Massachusetts, USA
| | - Andre Grageda
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, New York, USA
- Department of Urology, SUNY Upstate Medical University, Syracuse, New York, USA
| | - Vladimir A. Kuznetsov
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, New York, USA
- Department of Urology, SUNY Upstate Medical University, Syracuse, New York, USA
| | - Wenyi Feng
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, New York, USA
| |
Collapse
|
117
|
Yoo YE, Yoo T, Kang H, Kim E. Brain region and gene dosage-differential transcriptomic changes in Shank2-mutant mice. Front Mol Neurosci 2022; 15:977305. [PMID: 36311025 PMCID: PMC9612946 DOI: 10.3389/fnmol.2022.977305] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 09/16/2022] [Indexed: 12/20/2022] Open
Abstract
Shank2 is an abundant excitatory postsynaptic scaffolding protein that has been implicated in various neurodevelopmental and psychiatric disorders, including autism spectrum disorder (ASD), intellectual disability, attention-deficit/hyperactivity disorder, and schizophrenia. Shank2-mutant mice show ASD-like behavioral deficits and altered synaptic and neuronal functions, but little is known about how different brain regions and gene dosages affect the transcriptomic phenotypes of these mice. Here, we performed RNA-Seq-based transcriptomic analyses of the prefrontal cortex, hippocampus, and striatum in adult Shank2 heterozygous (HT)- and homozygous (HM)-mutant mice lacking exons 6–7. The prefrontal cortical, hippocampal, and striatal regions showed distinct transcriptomic patterns associated with synapse, ribosome, mitochondria, spliceosome, and extracellular matrix (ECM). The three brain regions were also distinct in the expression of ASD-related and ASD-risk genes. These differential patterns were stronger in the prefrontal cortex where the HT transcriptome displayed increased synaptic gene expression and reverse-ASD patterns whereas the HM transcriptome showed decreased synaptic gene expression and ASD-like patterns. These results suggest brain region- and gene dosage-differential transcriptomic changes in Shank2-mutant mice.
Collapse
Affiliation(s)
- Ye-Eun Yoo
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science (IBS), Daejeon, South Korea
| | - Taesun Yoo
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science (IBS), Daejeon, South Korea
| | - Hyojin Kang
- Division of National Supercomputing, Korea Institute of Science and Technology Information (KISTI), Daejeon, South Korea
| | - Eunjoon Kim
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science (IBS), Daejeon, South Korea
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, South Korea
- *Correspondence: Eunjoon Kim,
| |
Collapse
|
118
|
Kim S, Oh H, Choi SH, Yoo YE, Noh YW, Cho Y, Im GH, Lee C, Oh Y, Yang E, Kim G, Chung WS, Kim H, Kang H, Bae Y, Kim SG, Kim E. Postnatal age-differential ASD-like transcriptomic, synaptic, and behavioral deficits in Myt1l-mutant mice. Cell Rep 2022; 40:111398. [PMID: 36130507 DOI: 10.1016/j.celrep.2022.111398] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 06/28/2022] [Accepted: 08/31/2022] [Indexed: 12/29/2022] Open
Abstract
Myelin transcription factor 1 like (Myt1l), a zinc-finger transcription factor, promotes neuronal differentiation and is implicated in autism spectrum disorder (ASD) and intellectual disability. However, it remains unclear whether Myt1l promotes neuronal differentiation in vivo and its deficiency in mice leads to disease-related phenotypes. Here, we report that Myt1l-heterozygous mutant (Myt1l-HT) mice display postnatal age-differential ASD-related phenotypes: newborn Myt1l-HT mice, with strong Myt1l expression, show ASD-like transcriptomic changes involving decreased synaptic gene expression and prefrontal excitatory synaptic transmission and altered righting reflex. Juvenile Myt1l-HT mice, with markedly decreased Myt1l expression, display reverse ASD-like transcriptomes, increased prefrontal excitatory transmission, and largely normal behaviors. Adult Myt1l-HT mice show ASD-like transcriptomes involving astrocytic and microglial gene upregulation, increased prefrontal inhibitory transmission, and behavioral deficits. Therefore, Myt1l haploinsufficiency leads to ASD-related phenotypes in newborn mice, which are temporarily normalized in juveniles but re-appear in adults, pointing to continuing phenotypic changes long after a marked decrease of Myt1l expression in juveniles.
Collapse
Affiliation(s)
- Seongbin Kim
- Department of Biological Sciences, Korea Advanced Institute for Science and Technology (KAIST), Daejeon 34141, Korea
| | - Hyoseon Oh
- Department of Biological Sciences, Korea Advanced Institute for Science and Technology (KAIST), Daejeon 34141, Korea
| | - Sang Han Choi
- Center for Neuroscience Imaging Research, Institute for Basic Science (IBS), Suwon 16419, Korea; Department of Biomedical Engineering, Sungkyunkwan University, Suwon 16419, Korea
| | - Ye-Eun Yoo
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science (IBS), Daejeon 34141, Korea
| | - Young Woo Noh
- Department of Biological Sciences, Korea Advanced Institute for Science and Technology (KAIST), Daejeon 34141, Korea
| | - Yisul Cho
- Department of Anatomy and Neurobiology, School of Dentistry, Kyungpook National University, Daegu 41940, Korea
| | - Geun Ho Im
- Center for Neuroscience Imaging Research, Institute for Basic Science (IBS), Suwon 16419, Korea
| | - Chanhee Lee
- Center for Neuroscience Imaging Research, Institute for Basic Science (IBS), Suwon 16419, Korea
| | - Yusang Oh
- Department of Bio and Brain Engineering, Korea Advanced Institute for Science and Technology (KAIST), Daejeon 34141, Korea
| | - Esther Yang
- Department of Anatomy and BK21 Graduate Program, Biomedical Sciences, College of Medicine, Korea University, Seoul 02841, Korea
| | - Gyuri Kim
- Department of Biological Sciences, Korea Advanced Institute for Science and Technology (KAIST), Daejeon 34141, Korea
| | - Won-Suk Chung
- Department of Biological Sciences, Korea Advanced Institute for Science and Technology (KAIST), Daejeon 34141, Korea
| | - Hyun Kim
- Department of Anatomy and BK21 Graduate Program, Biomedical Sciences, College of Medicine, Korea University, Seoul 02841, Korea
| | - Hyojin Kang
- Division of National Supercomputing, Korea Institute of Science and Technology Information (KISTI), Daejeon 34141, Korea
| | - Yongchul Bae
- Department of Anatomy and Neurobiology, School of Dentistry, Kyungpook National University, Daegu 41940, Korea
| | - Seong-Gi Kim
- Center for Neuroscience Imaging Research, Institute for Basic Science (IBS), Suwon 16419, Korea; Department of Biomedical Engineering, Sungkyunkwan University, Suwon 16419, Korea
| | - Eunjoon Kim
- Department of Biological Sciences, Korea Advanced Institute for Science and Technology (KAIST), Daejeon 34141, Korea; Center for Synaptic Brain Dysfunctions, Institute for Basic Science (IBS), Daejeon 34141, Korea.
| |
Collapse
|
119
|
Hai Y, Leng G. A more than four-fold sex-specific difference of autism spectrum disorders and the possible contribution of pesticide usage in China 1990-2030. Front Public Health 2022; 10:945172. [PMID: 36187693 PMCID: PMC9525129 DOI: 10.3389/fpubh.2022.945172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 08/24/2022] [Indexed: 01/21/2023] Open
Abstract
Autism spectrum disorders (ASDs) are prevalent in children and adolescents and disproportionately affect males, and the main contributing factors underlying male vulnerability remain widely unknown. Pesticide use is widely reported to be associated with ASD risk, and the cases of pesticide poisoning incidence in rural areas are remarkably higher than those in the urban areas while the prevalence of ASDs in rural areas was higher than that in urban areas and the rate of male pesticide poisoning was significantly higher than female. Thus, pesticide usage may be an important contributing factor for causing sex-specific differences of ASD incidence. ASD burden was analyzed by using the data of ASD number, ASD rate (ASD cases per 100,000 persons) and disability-adjusted life years (DALYs) from 1990 to 2019. The changes from 1990 to 2030 were predicted using autoregressive integrated moving average (ARIMA) in time series forecasting based on the small values of Akaike information criterion and Bayesian information criterion. Finally, the relationship between ASD rate and pesticide usage risk index (PURI) was analyzed via Pearson's correlation coefficient. ASD number, ASD rate and DALYs will be reduced by 45.5% ± 8.2% (t = 9.100 and p = 0.0119), 56.6% ± 10.2% (t = 9.111 and p = 0.0118), and 44.9% ± 7.0% (t = 20.90 and p = 0.0023) from 1990 to 2030 in China. PURI has a strong relationship with ASD rate (rho = 0.953 to 0.988 and p < 0.0001). Pesticide poisoning incidence in males is up to 2-fold higher than that in females. ASD number and DALYs in males are 4-fold higher than those in females. Furthermore, there is growing evidence supporting that males are more susceptible than females to pesticides with sex differences in neurotoxicogenetics. Therefore, pesticide poisoning may be a contributing factor for causing the sex differences of ASD. Much work still needs to be done to confirm that.
Collapse
Affiliation(s)
- Yang Hai
- International Education College, Harbin Medical University, Harbin, China,*Correspondence: Yang Hai
| | - Guodong Leng
- College of Business Administration, Shenyang Pharmaceutical University, Shenyang, China
| |
Collapse
|
120
|
Recent Developments in Autism Genetic Research: A Scientometric Review from 2018 to 2022. Genes (Basel) 2022; 13:genes13091646. [PMID: 36140813 PMCID: PMC9498399 DOI: 10.3390/genes13091646] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 09/12/2022] [Accepted: 09/12/2022] [Indexed: 12/13/2022] Open
Abstract
Genetic research in Autism Spectrum Disorder (ASD) has progressed tremendously in recent decades. Dozens of genetic loci and hundreds of alterations in the genetic sequence, expression, epigenetic transformation, and interactions with other physiological and environmental systems have been found to increase the likelihood of developing ASD. There is therefore a need to represent this wide-ranging yet voluminous body of literature in a systematic manner so that this information can be synthesised and understood at a macro level. Therefore, this study made use of scientometric methods, particularly document co-citation analysis (DCA), to systematically review literature on ASD genetic research from 2018 to 2022. A total of 14,818 articles were extracted from Scopus and analyzed with CiteSpace. An optimized DCA analysis revealed that recent literature on ASD genetic research can be broadly organised into 12 major clusters representing various sub-topics. These clusters are briefly described in the manuscript and potential applications of this study are discussed.
Collapse
|
121
|
Deliu LP, Turingan M, Jadir D, Lee B, Ghosh A, Grewal SS. Serotonergic neuron ribosomal proteins regulate the neuroendocrine control of Drosophila development. PLoS Genet 2022; 18:e1010371. [PMID: 36048889 PMCID: PMC9473637 DOI: 10.1371/journal.pgen.1010371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 09/14/2022] [Accepted: 07/29/2022] [Indexed: 11/21/2022] Open
Abstract
The regulation of ribosome function is a conserved mechanism of growth control. While studies in single cell systems have defined how ribosomes contribute to cell growth, the mechanisms that link ribosome function to organismal growth are less clear. Here we explore this issue using Drosophila Minutes, a class of heterozygous mutants for ribosomal proteins. These animals exhibit a delay in larval development caused by decreased production of the steroid hormone ecdysone, the main regulator of larval maturation. We found that this developmental delay is not caused by decreases in either global ribosome numbers or translation rates. Instead, we show that they are due in part to loss of Rp function specifically in a subset of serotonin (5-HT) neurons that innervate the prothoracic gland to control ecdysone production. We find that these effects do not occur due to altered protein synthesis or proteostasis, but that Minute animals have reduced expression of synaptotagmin, a synaptic vesicle protein, and that the Minute developmental delay can be partially reversed by overexpression of synaptic vesicle proteins in 5-HTergic cells. These results identify a 5-HT cell-specific role for ribosomal function in the neuroendocrine control of animal growth and development.
Collapse
Affiliation(s)
- Lisa Patricia Deliu
- Clark H Smith Brain Tumour Centre, Arnie Charbonneau Cancer Institute, Alberta Children’s Hospital Research Institute, and Department of Biochemistry and Molecular Biology Calgary, University of Calgary, Alberta, Canada
| | - Michael Turingan
- Clark H Smith Brain Tumour Centre, Arnie Charbonneau Cancer Institute, Alberta Children’s Hospital Research Institute, and Department of Biochemistry and Molecular Biology Calgary, University of Calgary, Alberta, Canada
| | - Deeshpaul Jadir
- Clark H Smith Brain Tumour Centre, Arnie Charbonneau Cancer Institute, Alberta Children’s Hospital Research Institute, and Department of Biochemistry and Molecular Biology Calgary, University of Calgary, Alberta, Canada
| | - Byoungchun Lee
- Clark H Smith Brain Tumour Centre, Arnie Charbonneau Cancer Institute, Alberta Children’s Hospital Research Institute, and Department of Biochemistry and Molecular Biology Calgary, University of Calgary, Alberta, Canada
| | - Abhishek Ghosh
- Clark H Smith Brain Tumour Centre, Arnie Charbonneau Cancer Institute, Alberta Children’s Hospital Research Institute, and Department of Biochemistry and Molecular Biology Calgary, University of Calgary, Alberta, Canada
| | - Savraj Singh Grewal
- Clark H Smith Brain Tumour Centre, Arnie Charbonneau Cancer Institute, Alberta Children’s Hospital Research Institute, and Department of Biochemistry and Molecular Biology Calgary, University of Calgary, Alberta, Canada
| |
Collapse
|
122
|
Jaudon F, Thalhammer A, Zentilin L, Cingolani LA. CRISPR-mediated activation of autism gene Itgb3 restores cortical network excitability via mGluR5 signaling. MOLECULAR THERAPY - NUCLEIC ACIDS 2022; 29:462-480. [PMID: 36035754 PMCID: PMC9382421 DOI: 10.1016/j.omtn.2022.07.013] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 07/15/2022] [Indexed: 01/12/2023]
Abstract
Many mutations in autism spectrum disorder (ASD) affect a single allele, indicating a key role for gene dosage in ASD susceptibility. Recently, haplo-insufficiency of ITGB3, the gene encoding the extracellular matrix receptor β3 integrin, was associated with ASD. Accordingly, Itgb3 knockout (KO) mice exhibit autism-like phenotypes. The pathophysiological mechanisms of Itgb3 remain, however, unknown, and the potential of targeting this gene for developing ASD therapies uninvestigated. By combining molecular, biochemical, imaging, and pharmacological analyses, we establish that Itgb3 haplo-insufficiency impairs cortical network excitability by promoting extra-synaptic over synaptic signaling of the metabotropic glutamate receptor mGluR5, which is similarly dysregulated in fragile X syndrome, the most frequent monogenic form of ASD. To assess the therapeutic potential of regulating Itgb3 gene dosage, we implemented CRISPR activation and compared its efficacy with that of a pharmacological rescue strategy for fragile X syndrome. Correction of neuronal Itgb3 haplo-insufficiency by CRISPR activation rebalanced network excitability as effectively as blockade of mGluR5 with the selective antagonist MPEP. Our findings reveal an unexpected functional interaction between two ASD genes, thereby validating the pathogenicity of ITGB3 haplo-insufficiency. Further, they pave the way for exploiting CRISPR activation as gene therapy for normalizing gene dosage and network excitability in ASD.
Collapse
Affiliation(s)
- Fanny Jaudon
- Center for Synaptic Neuroscience and Technology (NSYN), Fondazione Istituto Italiano di Tecnologia (IIT), 16132 Genoa, Italy
- Department of Life Sciences, University of Trieste, 34127 Trieste, Italy
| | - Agnes Thalhammer
- Department of Life Sciences, University of Trieste, 34127 Trieste, Italy
- IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
| | - Lorena Zentilin
- AAV Vector Unit, International Centre for Genetic Engineering and Biotechnology (ICGEB), 34149 Trieste, Italy
| | - Lorenzo A. Cingolani
- Center for Synaptic Neuroscience and Technology (NSYN), Fondazione Istituto Italiano di Tecnologia (IIT), 16132 Genoa, Italy
- Department of Life Sciences, University of Trieste, 34127 Trieste, Italy
- Corresponding author Lorenzo A. Cingolani, Department of Life Sciences, University of Trieste, 34127 Trieste, Italy.
| |
Collapse
|
123
|
Jeon SJ, Kwon H, Bae HJ, Gonzales EL, Kim J, Chung HJ, Kim DH, Ryu JH, Shin CY. Agmatine relieves behavioral impairments in Fragile X mice model. Neuropharmacology 2022; 219:109234. [PMID: 36057317 DOI: 10.1016/j.neuropharm.2022.109234] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 07/25/2022] [Accepted: 08/21/2022] [Indexed: 10/31/2022]
Abstract
BACKGROUND Fragile X syndrome (FXS) is the most common heritable form of neurodevelopmental disorder, which is caused by the loss of fragile X mental retardation protein (FMRP) expression. Despite the unceasing efforts to develop therapeutic agents against FXS based on the pathophysiological changes observed in animal models of FXS and human patients, therapeutic candidates including mGluR signaling modulators have failed to provide sufficient effects. Based on the recent successful demonstration of an endogenous polyamine, agmatine, to improve the autism-like symptoms in the valproic acid animal model of autism, we investigated the effects of agmatine against FXS symptoms using Fmr1 knockout (KO) mice. METHODS We used male Fmr1 KO mice for behavioral tests such as marble burying, open-field test, memory tasks, social interaction tests and startle response to confirm the symptoms of FXS. We also checked the electrophysiological profile of neural activity in agmatine-treated Fmr1 KO mice. RESULTS Agmatine reversed the compulsion, learning and memory deficits, hyperactivity, aberrant social interaction, and communication deficit in Fmr1 KO mice while it normalized the aberrant LTP and LTD in the hippocampus. CONCLUSIONS The results highlight the potential of agmatine's novel disease-ameliorating effects in FXS, which warrants further studies to ascertain whether these findings translate into clinical effects in FXS patients.
Collapse
Affiliation(s)
- Se Jin Jeon
- Department of Pharmacology and Department of Advanced Translational Medicine, School of Medicine, Konkuk University, Seoul, 05029, Republic of Korea; Department of Integrative Biotechnology, College of Science and Technology, Sahmyook University, Seoul, 01795, Republic of Korea
| | - Huiyoung Kwon
- Department of Pharmacology and Department of Advanced Translational Medicine, School of Medicine, Konkuk University, Seoul, 05029, Republic of Korea
| | - Ho Jung Bae
- Department of Life and Nanopharmaceutical Science, College of Pharmacy, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Edson Luck Gonzales
- Department of Pharmacology and Department of Advanced Translational Medicine, School of Medicine, Konkuk University, Seoul, 05029, Republic of Korea
| | - Junhyeong Kim
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Gyeongsang National University, Jinju, 52828, Republic of Korea
| | - Hye Jin Chung
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Gyeongsang National University, Jinju, 52828, Republic of Korea
| | - Dong Hyun Kim
- Department of Pharmacology and Department of Advanced Translational Medicine, School of Medicine, Konkuk University, Seoul, 05029, Republic of Korea
| | - Jong Hoon Ryu
- Department of Life and Nanopharmaceutical Science, College of Pharmacy, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Chan Young Shin
- Department of Pharmacology and Department of Advanced Translational Medicine, School of Medicine, Konkuk University, Seoul, 05029, Republic of Korea.
| |
Collapse
|
124
|
Kim H, Kim D, Cho Y, Kim K, Roh JD, Kim Y, Yang E, Kim SS, Ahn S, Kim H, Kang H, Bae Y, Kim E. Early postnatal serotonin modulation prevents adult-stage deficits in Arid1b-deficient mice through synaptic transcriptional reprogramming. Nat Commun 2022; 13:5051. [PMID: 36030255 PMCID: PMC9420115 DOI: 10.1038/s41467-022-32748-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 08/12/2022] [Indexed: 11/19/2022] Open
Abstract
Autism spectrum disorder is characterized by early postnatal symptoms, although little is known about the mechanistic deviations that produce them and whether correcting them has long-lasting preventive effects on adult-stage deficits. ARID1B, a chromatin remodeler implicated in neurodevelopmental disorders, including autism spectrum disorder, exhibits strong embryonic- and early postnatal-stage expression. We report here that Arid1b-happloinsufficient (Arid1b+/-) mice display autistic-like behaviors at juvenile and adult stages accompanied by persistent decreases in excitatory synaptic density and transmission. Chronic treatment of Arid1b+/- mice with fluoxetine, a selective serotonin-reuptake inhibitor, during the first three postnatal weeks prevents synaptic and behavioral deficits in adults. Mechanistically, these rescues accompany transcriptomic changes, including upregulation of FMRP targets and normalization of HDAC4/MEF2A-related transcriptional regulation of the synaptic proteins, SynGAP1 and Arc. These results suggest that chronic modulation of serotonergic receptors during critical early postnatal periods prevents synaptic and behavioral deficits in adult Arid1b+/- mice through transcriptional reprogramming.
Collapse
Affiliation(s)
- Hyosang Kim
- Department of Biological Sciences, Korea Advanced Institute for Science and Technology (KAIST), Daejeon, 34141, Korea
| | - Doyoun Kim
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science (IBS), Daejeon, 34141, Korea
| | - Yisul Cho
- Department of Anatomy and Neurobiology, School of Dentistry, Kyungpook National University, Daegu, 41940, Korea
| | - Kyungdeok Kim
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science (IBS), Daejeon, 34141, Korea
| | - Junyeop Daniel Roh
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science (IBS), Daejeon, 34141, Korea
| | - Yangsik Kim
- Graduate School of Biomedical Engineering, Korea Advanced Institute for Science and Technology (KAIST), Daejeon, 34141, Korea
| | - Esther Yang
- Department of Anatomy and Division of Brain Korea 21, Biomedical Science, College of Medicine, Korea University, Seoul, 02841, Korea
| | - Seong Soon Kim
- Therapeutics and Biotechnology Division, Korea Research Institute of Chemical Technology (KRICT), Daejeon, 34114, Korea
| | - Sunjoo Ahn
- Therapeutics and Biotechnology Division, Korea Research Institute of Chemical Technology (KRICT), Daejeon, 34114, Korea
| | - Hyun Kim
- Department of Anatomy and Division of Brain Korea 21, Biomedical Science, College of Medicine, Korea University, Seoul, 02841, Korea
| | - Hyojin Kang
- Division of National Supercomputing, Korea Institute of Science and Technology Information, Daejeon, 34141, Korea
| | - Yongchul Bae
- Department of Anatomy and Neurobiology, School of Dentistry, Kyungpook National University, Daegu, 41940, Korea.
| | - Eunjoon Kim
- Department of Biological Sciences, Korea Advanced Institute for Science and Technology (KAIST), Daejeon, 34141, Korea.
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science (IBS), Daejeon, 34141, Korea.
| |
Collapse
|
125
|
FMRP modulates the Wnt signalling pathway in glioblastoma. Cell Death Dis 2022; 13:719. [PMID: 35982038 PMCID: PMC9388540 DOI: 10.1038/s41419-022-05019-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 06/06/2022] [Accepted: 06/10/2022] [Indexed: 01/21/2023]
Abstract
Converging evidence indicates that the Fragile X Messenger Ribonucleoprotein (FMRP), which absent or mutated in Fragile X Syndrome (FXS), plays a role in many types of cancers. However, while FMRP roles in brain development and function have been extensively studied, its involvement in the biology of brain tumors remains largely unexplored. Here we show, in human glioblastoma (GBM) biopsies, that increased expression of FMRP directly correlates with a worse patient outcome. In contrast, reductions in FMRP correlate with a diminished tumor growth and proliferation of human GBM stem-like cells (GSCs) in vitro in a cell culture model and in vivo in mouse brain GSC xenografts. Consistently, increased FMRP levels promote GSC proliferation. To characterize the mechanism(s) by which FMRP regulates GSC proliferation, we performed GSC transcriptome analyses in GSCs expressing high levels of FMRP, and in these GSCs after knockdown of FMRP. We show that the WNT signalling is the most significantly enriched among the published FMRP target genes and genes involved in ASD. Consistently, we find that reductions in FMRP downregulate both the canonical WNT/β-Catenin and the non-canonical WNT-ERK1/2 signalling pathways, reducing the stability of several key transcription factors (i.e. β-Catenin, CREB and ETS1) previously implicated in the modulation of malignant features of glioma cells. Our findings support a key role for FMRP in GBM cancer progression, acting via regulation of WNT signalling.
Collapse
|
126
|
Monday HR, Kharod SC, Yoon YJ, Singer RH, Castillo PE. Presynaptic FMRP and local protein synthesis support structural and functional plasticity of glutamatergic axon terminals. Neuron 2022; 110:2588-2606.e6. [PMID: 35728596 PMCID: PMC9391299 DOI: 10.1016/j.neuron.2022.05.024] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 03/31/2022] [Accepted: 05/26/2022] [Indexed: 10/18/2022]
Abstract
Learning and memory rely on long-lasting, synapse-specific modifications. Although postsynaptic forms of plasticity typically require local protein synthesis, whether and how local protein synthesis contributes to presynaptic changes remain unclear. Here, we examined the mouse hippocampal mossy fiber (MF)-CA3 synapse, which expresses both structural and functional presynaptic plasticity and contains presynaptic fragile X messenger ribonucleoprotein (FMRP), an RNA-binding protein involved in postsynaptic protein-synthesis-dependent plasticity. We report that MF boutons contain ribosomes and synthesize protein locally. The long-term potentiation of MF-CA3 synaptic transmission (MF-LTP) was associated with the translation-dependent enlargement of MF boutons. Remarkably, increasing in vitro or in vivo MF activity enhanced the protein synthesis in MFs. Moreover, the deletion of presynaptic FMRP blocked structural and functional MF-LTP, suggesting that FMRP is a critical regulator of presynaptic MF plasticity. Thus, presynaptic FMRP and protein synthesis dynamically control presynaptic structure and function in the mature mammalian brain.
Collapse
Affiliation(s)
- Hannah R Monday
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, New York, NY 10461, USA.
| | - Shivani C Kharod
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, New York, NY 10461, USA
| | - Young J Yoon
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, New York, NY 10461, USA; Department of Cell Biology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, New York, NY 10461, USA
| | - Robert H Singer
- Department of Cell Biology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, New York, NY 10461, USA
| | - Pablo E Castillo
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, New York, NY 10461, USA; Department of Psychiatry and Behavioral Sciences, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, New York, NY 10461, USA.
| |
Collapse
|
127
|
Chen YN, Zheng X, Chen HL, Gao JX, Li XX, Xie JF, Xie YP, Spruyt K, Shao YF, Hou YP. Stereotaxic atlas of the infant rat brain at postnatal days 7–13. Front Neuroanat 2022; 16:968320. [PMID: 36032994 PMCID: PMC9412974 DOI: 10.3389/fnana.2022.968320] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 07/25/2022] [Indexed: 11/13/2022] Open
Abstract
Recently, researchers have paid progressively more attention to the study of neural development in infant rats. However, due to the lack of complete intracerebral localization information, such as clear nuclear cluster boundaries, identified main brain structures, and reliable stereotaxic coordinates, it is difficult and restricted to apply technical neuroscience to infant rat’s brain. The present study was undertaken to refine the atlas of infant rats. As such, we established a stereotaxic atlas of the infant rat’s brain at postnatal days 7–13. Furthermore, dye calibration surgery was performed in P7–P13 infant rats by injecting Methylene blue, and sections were incubated in Nissl solutions. From the panoramic images of the brain sections, atlases were made. Our article has provided the appearance and measurements of P7–P13 Sprague–Dawley rat pups. Whereas the atlas contains a series of about 530 coronal brain section images from olfactory bulbs to the brainstem, a list of abbreviations of the main brain structures, and reliable stereotaxic coordinates, which were demonstrated by vertical and oblique injections with fluorescent dye DiI. The present findings demonstrated that our study of P7–P13 atlases has reasonable nucleus boundaries and accurate and good repeatability of stereotaxic coordinates, which can make up for the shortage of postnatal rat brain atlas currently in the field.
Collapse
Affiliation(s)
- Yu-Nong Chen
- Departments of Neuroscience, Anatomy, Histology, and Embryology, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Xin Zheng
- Departments of Neuroscience, Anatomy, Histology, and Embryology, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Hai-Lin Chen
- Departments of Neuroscience, Anatomy, Histology, and Embryology, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Jin-Xian Gao
- Departments of Neuroscience, Anatomy, Histology, and Embryology, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Xin-Xuan Li
- Departments of Neuroscience, Anatomy, Histology, and Embryology, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Jun-Fan Xie
- Departments of Neuroscience, Anatomy, Histology, and Embryology, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Yu-Ping Xie
- Sleep Medicine Center of Gansu Provincial Hospital, Lanzhou, China
| | - Karen Spruyt
- NeuroDiderot – INSERM, Université de Paris, Paris, France
| | - Yu-Feng Shao
- Departments of Neuroscience, Anatomy, Histology, and Embryology, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
- Key Lab of Neurology of Gansu Province, Lanzhou University, Lanzhou, China
- *Correspondence: Yu-Feng Shao,
| | - Yi-Ping Hou
- Departments of Neuroscience, Anatomy, Histology, and Embryology, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
- Key Lab of Neurology of Gansu Province, Lanzhou University, Lanzhou, China
- Yi-Ping Hou,
| |
Collapse
|
128
|
Cogram P, Deacon RMJ, Klamer D, Rebowe N, Sprouse J, Reyes ST, Missling CU, Kaufmann WE. Brain cell signaling abnormalities are detected in blood in a murine model of Fragile X syndrome and corrected by Sigma-1 receptor agonist Blarcamesine. Am J Med Genet A 2022; 188:2497-2500. [PMID: 35661397 DOI: 10.1002/ajmg.a.62853] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Accepted: 04/30/2022] [Indexed: 11/12/2022]
Affiliation(s)
- Patricia Cogram
- FRAXA-DVI, FRAXA, Newburyport, Massachusetts, USA
- GeN.DDI Ltd, London, UK
- Fraunhofer Chile Research, Center for Systems Biotechnology, Santiago, Chile
| | - Robert M J Deacon
- FRAXA-DVI, FRAXA, Newburyport, Massachusetts, USA
- GeN.DDI Ltd, London, UK
- Fraunhofer Chile Research, Center for Systems Biotechnology, Santiago, Chile
| | | | - Nell Rebowe
- Anavex Life Sciences Corp, New York, New York, USA
| | | | - Samantha T Reyes
- Stanford University, Department of Radiology, Stanford, California, USA
| | | | - Walter E Kaufmann
- Anavex Life Sciences Corp, New York, New York, USA
- Department of Human Genetics, Emory University School of Medicine, Atlanta, Georgia, USA
| |
Collapse
|
129
|
Exome sequencing analysis of Japanese autism spectrum disorder case-control sample supports an increased burden of synaptic function-related genes. Transl Psychiatry 2022; 12:265. [PMID: 35811316 PMCID: PMC9271461 DOI: 10.1038/s41398-022-02033-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 06/15/2022] [Accepted: 06/17/2022] [Indexed: 01/16/2023] Open
Abstract
Autism spectrum disorder (ASD) is a highly heritable, complex disorder in which rare variants contribute significantly to disease risk. Although many genes have been associated with ASD, there have been few genetic studies of ASD in the Japanese population. In whole exomes from a Japanese ASD sample of 309 cases and 299 controls, rare variants were associated with ASD within specific neurodevelopmental gene sets, including highly constrained genes, fragile X mental retardation protein target genes, and genes involved in synaptic function, with the strongest enrichment in trans-synaptic signaling (p = 4.4 × 10-4, Q-value = 0.06). In particular, we strengthen the evidence regarding the role of ABCA13, a synaptic function-related gene, in Japanese ASD. The overall results of this case-control exome study showed that rare variants related to synaptic function are associated with ASD susceptibility in the Japanese population.
Collapse
|
130
|
Song C, Broadie K. Dysregulation of BMP, Wnt, and Insulin Signaling in Fragile X Syndrome. Front Cell Dev Biol 2022; 10:934662. [PMID: 35880195 PMCID: PMC9307498 DOI: 10.3389/fcell.2022.934662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Accepted: 06/06/2022] [Indexed: 01/21/2023] Open
Abstract
Drosophila models of neurological disease contribute tremendously to research progress due to the high conservation of human disease genes, the powerful and sophisticated genetic toolkit, and the rapid generation time. Fragile X syndrome (FXS) is the most prevalent heritable cause of intellectual disability and autism spectrum disorders, and the Drosophila FXS disease model has been critical for the genetic screening discovery of new intercellular secretion mechanisms. Here, we focus on the roles of three major signaling pathways: BMP, Wnt, and insulin-like peptides. We present Drosophila FXS model defects compared to mouse models in stem cells/embryos, the glutamatergic neuromuscular junction (NMJ) synapse model, and the developing adult brain. All three of these secreted signaling pathways are strikingly altered in FXS disease models, giving new mechanistic insights into impaired cellular outcomes and neurological phenotypes. Drosophila provides a powerful genetic screening platform to expand understanding of these secretory mechanisms and to test cellular roles in both peripheral and central nervous systems. The studies demonstrate the importance of exploring broad genetic interactions and unexpected regulatory mechanisms. We discuss a number of research avenues to pursue BMP, Wnt, and insulin signaling in future FXS investigations and the development of potential therapeutics.
Collapse
Affiliation(s)
- Chunzhu Song
- Department of Biological Sciences, College of Arts and Science, Vanderbilt University, Nashville, TN, United States
| | - Kendal Broadie
- Department of Biological Sciences, College of Arts and Science, Vanderbilt University, Nashville, TN, United States
- Department of Cell and Developmental Biology, School of Medicine, Vanderbilt University, Nashville, TN, United States
- Kennedy Center for Research on Human Development, Nashville, TN, United States
- Vanderbilt Brain Institute, School of Medicine, Vanderbilt University and Medical Center, Nashville, TN, United States
| |
Collapse
|
131
|
Acetyl-L-carnitine and/or liposomal co-enzyme Q10 prevent propionic acid-induced neurotoxicity by modulating oxidative tissue injury, inflammation, and ALDH1A1-RA-RARα signaling in rats. Biomed Pharmacother 2022; 153:113360. [PMID: 35785703 DOI: 10.1016/j.biopha.2022.113360] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Revised: 06/15/2022] [Accepted: 06/28/2022] [Indexed: 11/23/2022] Open
Abstract
Propionic acid (PPA) is a short-chain fatty acid produced endogenously by gut microbiota and found in foodstuffs and pharmaceutical products as an additive. Exposure to PPA has been associated with the development of autism spectrum disorder (ASD). The purpose of this study was to investigate the protective effect of acetyl-L-carnitine (ALCAR) and liposomal Co-enzyme Q10 (CoQ10) against cerebral and cerebellar oxidative injury, inflammation, and cell death, and alterations in ALDH1A1-RA-RARα signaling in an autism-like rat model induced by PPA. The rats were treated with PPA and concurrently received ALCAR and/or CoQ10 for 5 days. The animals were sacrificed, and the cerebral cortex and cerebellum were collected for analysis. PPA caused histopathological alterations along with increased malondialdehyde (MDA), NF-κB p65, TNF-α, and IL-6 in the cerebrum and cerebellum of rats. Reduced glutathione (GSH) and antioxidant enzymes were declined in the brain of rats that received PPA. Concurrent treatment with ALCAR and/or CoQ10 prevented tissue injury, decreased MDA, NF-κB p65, and pro-inflammatory cytokines, and enhanced cellular antioxidants in PPA-administered rats. ALCAR and/or CoQ10 upregulated Bcl-2 and decreased Bax and caspase-3 in the brain of rats. In addition, ALCAR and/or CoQ10 upregulated cerebral and cerebellar ALDH1A1 and RARα in PPA-treated rats. The combination of ALCAR and CoQ10 showed more potent effects when compared with the individual treatments. In conclusion, ALCAR and/or CoQ10 prevented tissue injury, ameliorated oxidative stress, inflammatory response, and apoptosis, and upregulated ALDH1A1-RA-RARα signaling in the brain of autistic rats.
Collapse
|
132
|
Aishworiya R, Protic D, Hagerman R. Autism spectrum disorder in the fragile X premutation state: possible mechanisms and implications. J Neurol 2022; 269:4676-4683. [PMID: 35723724 DOI: 10.1007/s00415-022-11209-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 05/26/2022] [Accepted: 05/27/2022] [Indexed: 12/23/2022]
Abstract
There is increasing recognition of the heterogeneity of origin of cases of autism spectrum disorder (ASD) with multiple forms of ASD having been identified over the decades. Among these, a genetic etiology can be identified in 20-40% of cases when a full genetic work-up is completed. The Fragile X premutation state (characterized by the presence of 55-200 CGG repeats in the FMR1 gene) is a relatively newly identified disease state that has since been associated with several disorders including fragile X-associated tremor ataxia syndrome (FXTAS), fragile X-associated primary ovarian insufficiency (FXPOI) and most recently, fragile X-associated neurodevelopmental disorders (FXAND) which commonly includes anxiety and depression. In addition to these associated disorders, extant literature and clinical observations have suggested an association between the premutation state and ASD. In this paper, we review the literature pertinent to this and discuss possible molecular mechanisms that may explain this association. This includes lowered levels of the FMR1 Protein (FMRP), GABA deficits, mitochondrial dysfunction and secondary genetic abnormalities that is seen in premutation carriers as well as their increased vulnerability to environmental stressors. Understanding these mechanisms can facilitate development of targeted treatment for specific sub-groups of ASD and premutation disorders in future.
Collapse
Affiliation(s)
- Ramkumar Aishworiya
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute, University of California Davis, 2825 50th Street, Sacramento, CA, 95817, USA. .,Khoo Teck Puat-National University Children's Medical Institute, National University Health System, 5 Lower Kent Ridge Road, Singapore, 119074, Singapore. .,Department of Pediatrics, Yong Loo Lin School of Medicine, National University of Singapore, 10 Medical Drive, Singapore, 117597, Singapore.
| | - Dragana Protic
- Department of Pharmacology, Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Randi Hagerman
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute, University of California Davis, 2825 50th Street, Sacramento, CA, 95817, USA.,Department of Pediatrics, University of California Davis School of Medicine, 4610 X St, Sacramento, CA, 95817, USA
| |
Collapse
|
133
|
O'Neill AC, Uzbas F, Antognolli G, Merino F, Draganova K, Jäck A, Zhang S, Pedini G, Schessner JP, Cramer K, Schepers A, Metzger F, Esgleas M, Smialowski P, Guerrini R, Falk S, Feederle R, Freytag S, Wang Z, Bahlo M, Jungmann R, Bagni C, Borner GHH, Robertson SP, Hauck SM, Götz M. Spatial centrosome proteome of human neural cells uncovers disease-relevant heterogeneity. Science 2022; 376:eabf9088. [PMID: 35709258 DOI: 10.1126/science.abf9088] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The centrosome provides an intracellular anchor for the cytoskeleton, regulating cell division, cell migration, and cilia formation. We used spatial proteomics to elucidate protein interaction networks at the centrosome of human induced pluripotent stem cell-derived neural stem cells (NSCs) and neurons. Centrosome-associated proteins were largely cell type-specific, with protein hubs involved in RNA dynamics. Analysis of neurodevelopmental disease cohorts identified a significant overrepresentation of NSC centrosome proteins with variants in patients with periventricular heterotopia (PH). Expressing the PH-associated mutant pre-mRNA-processing factor 6 (PRPF6) reproduced the periventricular misplacement in the developing mouse brain, highlighting missplicing of transcripts of a microtubule-associated kinase with centrosomal location as essential for the phenotype. Collectively, cell type-specific centrosome interactomes explain how genetic variants in ubiquitous proteins may convey brain-specific phenotypes.
Collapse
Affiliation(s)
- Adam C O'Neill
- Physiological Genomics, Biomedical Center (BMC), Ludwig-Maximilians-Universitaet (LMU), Großhaderner Straße 9, 82152 Planegg-Martinsried, Germany.,Institute of Stem Cell Research, Helmholtz Center Munich, German Research Center for Environmental Health, Großhaderner Straße 9, 82152 Planegg-Martinsried, Germany
| | - Fatma Uzbas
- Physiological Genomics, Biomedical Center (BMC), Ludwig-Maximilians-Universitaet (LMU), Großhaderner Straße 9, 82152 Planegg-Martinsried, Germany.,Institute of Stem Cell Research, Helmholtz Center Munich, German Research Center for Environmental Health, Großhaderner Straße 9, 82152 Planegg-Martinsried, Germany
| | - Giulia Antognolli
- Physiological Genomics, Biomedical Center (BMC), Ludwig-Maximilians-Universitaet (LMU), Großhaderner Straße 9, 82152 Planegg-Martinsried, Germany.,Institute of Stem Cell Research, Helmholtz Center Munich, German Research Center for Environmental Health, Großhaderner Straße 9, 82152 Planegg-Martinsried, Germany
| | - Florencia Merino
- Physiological Genomics, Biomedical Center (BMC), Ludwig-Maximilians-Universitaet (LMU), Großhaderner Straße 9, 82152 Planegg-Martinsried, Germany.,Institute of Stem Cell Research, Helmholtz Center Munich, German Research Center for Environmental Health, Großhaderner Straße 9, 82152 Planegg-Martinsried, Germany
| | - Kalina Draganova
- Physiological Genomics, Biomedical Center (BMC), Ludwig-Maximilians-Universitaet (LMU), Großhaderner Straße 9, 82152 Planegg-Martinsried, Germany.,Institute of Stem Cell Research, Helmholtz Center Munich, German Research Center for Environmental Health, Großhaderner Straße 9, 82152 Planegg-Martinsried, Germany
| | - Alex Jäck
- Physiological Genomics, Biomedical Center (BMC), Ludwig-Maximilians-Universitaet (LMU), Großhaderner Straße 9, 82152 Planegg-Martinsried, Germany.,Institute of Stem Cell Research, Helmholtz Center Munich, German Research Center for Environmental Health, Großhaderner Straße 9, 82152 Planegg-Martinsried, Germany
| | - Sirui Zhang
- CAS Key Laboratory of Computational Biology, Biomedical Big Data Center, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai 200031, China.,University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China.,CAS Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China
| | - Giorgia Pedini
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy
| | | | - Kimberly Cramer
- Max Planck Institute of Biochemistry, Martinsried, Germany.,Faculty of Physics and Center for Nanoscience, LMU, Munich, Germany
| | - Aloys Schepers
- Monoclonal Antibody Core Facility, Institute for Diabetes and Obesity, Helmholtz Center Munich, German Research Center for Environmental Health, 85764 Neuherberg, Germany
| | - Fabian Metzger
- Research Unit Protein Science and Metabolomics and Proteomics Core, Helmholtz Centre Munich, German Research Center for Environmental Health, 85764 Neuherberg, Germany
| | - Miriam Esgleas
- Physiological Genomics, Biomedical Center (BMC), Ludwig-Maximilians-Universitaet (LMU), Großhaderner Straße 9, 82152 Planegg-Martinsried, Germany.,Institute of Stem Cell Research, Helmholtz Center Munich, German Research Center for Environmental Health, Großhaderner Straße 9, 82152 Planegg-Martinsried, Germany
| | - Pawel Smialowski
- Physiological Genomics, Biomedical Center (BMC), Ludwig-Maximilians-Universitaet (LMU), Großhaderner Straße 9, 82152 Planegg-Martinsried, Germany.,Institute of Stem Cell Research, Helmholtz Center Munich, German Research Center for Environmental Health, Großhaderner Straße 9, 82152 Planegg-Martinsried, Germany
| | - Renzo Guerrini
- Neuroscience Department, Children's Hospital Meyer-University of Florence, Florence, Italy
| | - Sven Falk
- Physiological Genomics, Biomedical Center (BMC), Ludwig-Maximilians-Universitaet (LMU), Großhaderner Straße 9, 82152 Planegg-Martinsried, Germany.,Institute of Stem Cell Research, Helmholtz Center Munich, German Research Center for Environmental Health, Großhaderner Straße 9, 82152 Planegg-Martinsried, Germany
| | - Regina Feederle
- Monoclonal Antibody Core Facility, Institute for Diabetes and Obesity, Helmholtz Center Munich, German Research Center for Environmental Health, 85764 Neuherberg, Germany.,SYNERGY, Excellence Cluster of Systems Neurology, Biomedical Center, LMU, Planegg-Martinsried, Germany
| | - Saskia Freytag
- Personalised Oncology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia.,Department of Medical Biology, University of Melbourne, Melbourne, VIC 3010, Australia
| | - Zefeng Wang
- CAS Key Laboratory of Computational Biology, Biomedical Big Data Center, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai 200031, China.,University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China.,CAS Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China
| | - Melanie Bahlo
- Personalised Oncology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia.,Department of Medical Biology, University of Melbourne, Melbourne, VIC 3010, Australia
| | - Ralf Jungmann
- Max Planck Institute of Biochemistry, Martinsried, Germany.,Faculty of Physics and Center for Nanoscience, LMU, Munich, Germany
| | - Claudia Bagni
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy.,Department of Fundamental Neurosciences, University of Lausanne, Rue du Bugnon 9, 1005 Lausanne, Switzerland
| | | | - Stephen P Robertson
- Department of Women's and Children's Health, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand
| | - Stefanie M Hauck
- Research Unit Protein Science and Metabolomics and Proteomics Core, Helmholtz Centre Munich, German Research Center for Environmental Health, 85764 Neuherberg, Germany
| | - Magdalena Götz
- Physiological Genomics, Biomedical Center (BMC), Ludwig-Maximilians-Universitaet (LMU), Großhaderner Straße 9, 82152 Planegg-Martinsried, Germany.,Institute of Stem Cell Research, Helmholtz Center Munich, German Research Center for Environmental Health, Großhaderner Straße 9, 82152 Planegg-Martinsried, Germany.,SYNERGY, Excellence Cluster of Systems Neurology, Biomedical Center, LMU, Planegg-Martinsried, Germany
| |
Collapse
|
134
|
Li R, Zhang C, Rao Y, Yuan TF. Deep brain stimulation of fornix for memory improvement in Alzheimer's disease: A critical review. Ageing Res Rev 2022; 79:101668. [PMID: 35705176 DOI: 10.1016/j.arr.2022.101668] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 05/16/2022] [Accepted: 06/09/2022] [Indexed: 11/30/2022]
Abstract
Memory reflects the brain function in encoding, storage and retrieval of the data or information, which is a fundamental ability for any live organism. The development of approaches to improve memory attracts much attention due to the underlying mechanistic insight and therapeutic potential to treat neurodegenerative diseases with memory loss, such as Alzheimer's disease (AD). Deep brain stimulation (DBS), a reversible, adjustable, and non-ablative therapy, has been shown to be safe and effective in many clinical trials for neurodegenerative and neuropsychiatric disorders. Among all potential regions with access to invasive electrodes, fornix is considered as it is the major afferent and efferent connection of the hippocampus known to be closely associated with learning and memory. Indeed, clinical trials have demonstrated that fornix DBS globally improved cognitive function in a subset of patients with AD, indicating fornix can serve as a potential target for neurosurgical intervention in treating memory impairment in AD. The present review aims to provide a better understanding of recent progresses in the application of fornix DBS for ameliorating memory impairments in AD patients.
Collapse
Affiliation(s)
- Ruofan Li
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chencheng Zhang
- Department of Neurosurgery, Center for Functional Neurosurgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yanxia Rao
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Department of Laboratory Animal Science, Fudan University, China.
| | - Ti-Fei Yuan
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China; Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital Affiliated to Tongji University School of Medicine, Shanghai, China.
| |
Collapse
|
135
|
Hwang JY, Monday HR, Yan J, Gompers A, Buxbaum AR, Sawicka KJ, Singer RH, Castillo PE, Zukin RS. CPEB3-dependent increase in GluA2 subunits impairs excitatory transmission onto inhibitory interneurons in a mouse model of fragile X. Cell Rep 2022; 39:110853. [PMID: 35675768 PMCID: PMC9671216 DOI: 10.1016/j.celrep.2022.110853] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 10/05/2021] [Accepted: 05/01/2022] [Indexed: 01/29/2023] Open
Abstract
Fragile X syndrome (FXS) is a leading cause of inherited intellectual disability and autism. Whereas dysregulated RNA translation in Fmr1 knockout (KO) mice, a model of FXS, is well studied, little is known about aberrant transcription. Using single-molecule mRNA detection, we show that mRNA encoding the AMPAR subunit GluA2 (but not GluA1) is elevated in dendrites and at transcription sites of hippocampal neurons of Fmr1 KO mice, indicating elevated GluA2 transcription. We identify CPEB3, a protein implicated in memory consolidation, as an upstream effector critical to GluA2 mRNA expression in FXS. Increased GluA2 mRNA is translated into an increase in GluA2 subunits, a switch in synaptic AMPAR phenotype from GluA2-lacking, Ca2+-permeable to GluA2-containing, Ca2+-impermeable, reduced inhibitory synaptic transmission, and loss of NMDAR-independent LTP at glutamatergic synapses onto CA1 inhibitory interneurons. These factors could contribute to an excitatory/inhibitory imbalance-a common theme in FXS and other autism spectrum disorders.
Collapse
Affiliation(s)
- Jee-Yeon Hwang
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, New York, NY 10461, USA,Department of Pharmacology and Neuroscience, Creighton University School of Medicine, Omaha, NE 68178, USA,These authors contributed equally,Lead contact,Correspondence: (J.-Y.H.), (R.S.Z.)
| | - Hannah R. Monday
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, New York, NY 10461, USA,Present address: Department of Molecular and Cellular Biology and Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA 94720, USA,These authors contributed equally
| | - Jingqi Yan
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, New York, NY 10461, USA,Center for Gene Regulation in Health and Disease, Department of Biological, Geological, and Environmental Sciences, Cleveland State University, Cleveland, OH 44115, USA,These authors contributed equally
| | - Andrea Gompers
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, New York, NY 10461, USA,Center for Immunology and Infectious Diseases, University of California, Davis, Davis, CA 95616, USA,These authors contributed equally
| | - Adina R. Buxbaum
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, New York, NY 10461, USA,Department of Structural & Cell Biology, Albert Einstein College of Medicine, New York, NY 10461, USA,Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA,Present address: Department of Neurosciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Kirsty J. Sawicka
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, New York, NY 10461, USA,Present address: Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Cambridge, UK
| | - Robert H. Singer
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, New York, NY 10461, USA,Department of Structural & Cell Biology, Albert Einstein College of Medicine, New York, NY 10461, USA,Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA,These authors contributed equally
| | - Pablo E. Castillo
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, New York, NY 10461, USA,Department of Psychiatry and Behavioral Sciences, Albert Einstein College of Medicine, New York, NY 10461, USA,These authors contributed equally
| | - R. Suzanne Zukin
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, New York, NY 10461, USA,These authors contributed equally,Correspondence: (J.-Y.H.), (R.S.Z.)
| |
Collapse
|
136
|
Chung C, Shin W, Kim E. Early and Late Corrections in Mouse Models of Autism Spectrum Disorder. Biol Psychiatry 2022; 91:934-944. [PMID: 34556257 DOI: 10.1016/j.biopsych.2021.07.021] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 06/18/2021] [Accepted: 07/21/2021] [Indexed: 12/18/2022]
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder characterized by social and repetitive symptoms. A key feature of ASD is early-life manifestations of symptoms, indicative of early pathophysiological mechanisms. In mouse models of ASD, increasing evidence indicates that there are early pathophysiological mechanisms that can be corrected early to prevent phenotypic defects in adults, overcoming the disadvantage of the short-lasting effects that characterize adult-initiated treatments. In addition, the results from gene restorations indicate that ASD-related phenotypes can be rescued in some cases even after the brain has fully matured. These results suggest that we need to consider both temporal and mechanistic aspects in studies of ASD models and carefully compare genetic and nongenetic corrections. Here, we summarize the early and late corrections in mouse models of ASD by genetic and pharmacological interventions and discuss how to better integrate these results to ensure efficient and long-lasting corrections for eventual clinical translation.
Collapse
Affiliation(s)
- Changuk Chung
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science, Daejeon, South Korea; Department of Neurosciences, University of California San Diego, La Jolla, California
| | - Wangyong Shin
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science, Daejeon, South Korea
| | - Eunjoon Kim
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science, Daejeon, South Korea; Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, South Korea.
| |
Collapse
|
137
|
Ma R, Pang K, Kang H, Zhang Y, Bang G, Park S, Hwang E, Ryu JR, Kwon Y, Kang HR, Jin C, Kim Y, Kim SY, Kwon SK, Kim D, Sun W, Kim JY, Han K. Protein interactome and cell-type expression analyses reveal that cytoplasmic FMR1-interacting protein 1 (CYFIP1), but not CYFIP2, associates with astrocytic focal adhesion. J Neurochem 2022; 162:190-206. [PMID: 35567753 DOI: 10.1111/jnc.15622] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 02/24/2022] [Accepted: 05/11/2022] [Indexed: 11/28/2022]
Abstract
The two members of the cytoplasmic FMR1-interacting protein family, CYFIP1 and CYFIP2, are evolutionarily conserved multifunctional proteins whose defects are associated with distinct types of brain disorders. Even with high sequence homology between CYFIP1 and CYFIP2, several lines of evidence indicate their different functions in the brain; however, the underlying mechanisms remain largely unknown. Here, we performed reciprocal immunoprecipitation experiments using CYFIP1-2×Myc and CYFIP2-3×Flag knock-in mice and found that CYFIP1 and CYFIP2 are not significantly co-immunoprecipitated with each other in the knock-in brains compared to negative control wild-type brains. Moreover, CYFIP1 and CYFIP2 showed different size distributions by size-exclusion chromatography of wild-type mouse brains. Specifically, mass spectrometry-based analysis of CYFIP1-2×Myc knock-in brains identified 131 proteins in the CYFIP1 interactome. Comparison of the CYFIP1 interactome with the previously identified brain region- and age-matched CYFIP2 interactome, consisting of 140 proteins, revealed only eight common proteins. Investigations using single-cell RNA-sequencing databases suggested non-neuronal cell- and neuron-enriched expression of Cyfip1 and Cyfip2, respectively. At the protein level, CYFIP1 was detected in both neurons and astrocytes, while CYFIP2 was detected only in neurons, suggesting the predominant expression of CYFIP1 in astrocytes. Bioinformatic characterization of the CYFIP1 interactome, and co-expression analysis of Cyfip1 with astrocytic genes, commonly linked CYFIP1 with focal adhesion proteins. Immunocytochemical analysis and proximity ligation assay suggested partial co-localization of CYFIP1 and focal adhesion proteins in cultured astrocytes. Together, these results suggest a CYFIP1-specific association with astrocytic focal adhesion, which may contribute to the different brain functions and dysfunctions of CYFIP1 and CYFIP2.
Collapse
Affiliation(s)
- Ruiying Ma
- Department of Neuroscience, Korea University College of Medicine, Seoul, 02841, Republic of Korea.,BK21 Graduate Program, Department of Biomedical Sciences, Korea University College of Medicine, Seoul, 02841, Republic of Korea
| | - Kaifang Pang
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas, 77030, USA.,Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, Texas, 77030, USA
| | - Hyojin Kang
- Division of National Supercomputing, Korea Institute of Science and Technology Information (KISTI), Daejeon, 34141, Republic of Korea
| | - Yinhua Zhang
- Department of Neuroscience, Korea University College of Medicine, Seoul, 02841, Republic of Korea.,BK21 Graduate Program, Department of Biomedical Sciences, Korea University College of Medicine, Seoul, 02841, Republic of Korea
| | - Geul Bang
- Research Center for Bioconvergence Analysis, Korea Basic Science Institute (KBSI), Ochang 28119, Republic of Korea.,Korea University College of Pharmacy, Sejong, 30019, Republic of Korea
| | - Sangwoo Park
- Research Center for Bioconvergence Analysis, Korea Basic Science Institute (KBSI), Ochang 28119, Republic of Korea
| | - Eunha Hwang
- Center for Research Equipment, Korea Basic Science Institute (KBSI), Ochang 28119, Republic of Korea
| | - Jae Ryun Ryu
- Department of Anatomy, Korea University College of Medicine, Seoul, 02841, Republic of Korea
| | - Yujin Kwon
- Therapeutics & Biotechnology Division, Drug discovery platform research center, Korea Research Institute of Chemical Technology (KRICT), Daejeon, 34114, Republic of Korea
| | - Hyae Rim Kang
- Department of Neuroscience, Korea University College of Medicine, Seoul, 02841, Republic of Korea.,BK21 Graduate Program, Department of Biomedical Sciences, Korea University College of Medicine, Seoul, 02841, Republic of Korea
| | - Chunmei Jin
- Department of Neuroscience, Korea University College of Medicine, Seoul, 02841, Republic of Korea.,BK21 Graduate Program, Department of Biomedical Sciences, Korea University College of Medicine, Seoul, 02841, Republic of Korea
| | - Yoonhee Kim
- Department of Neuroscience, Korea University College of Medicine, Seoul, 02841, Republic of Korea
| | - Su Yeon Kim
- Department of Neuroscience, Korea University College of Medicine, Seoul, 02841, Republic of Korea.,Korea Institute of Science and Technology (KIST), Brain Science Institute, Seoul, 02792, Republic of Korea
| | - Seok-Kyu Kwon
- Korea Institute of Science and Technology (KIST), Brain Science Institute, Seoul, 02792, Republic of Korea
| | - Doyoun Kim
- Therapeutics & Biotechnology Division, Drug discovery platform research center, Korea Research Institute of Chemical Technology (KRICT), Daejeon, 34114, Republic of Korea.,Medicinal Chemistry and Pharmacology, Korea University of Science and Technology (UST), Daejeon, 34113, Republic of Korea
| | - Woong Sun
- BK21 Graduate Program, Department of Biomedical Sciences, Korea University College of Medicine, Seoul, 02841, Republic of Korea.,Department of Anatomy, Korea University College of Medicine, Seoul, 02841, Republic of Korea
| | - Jin Young Kim
- Research Center for Bioconvergence Analysis, Korea Basic Science Institute (KBSI), Ochang 28119, Republic of Korea
| | - Kihoon Han
- Department of Neuroscience, Korea University College of Medicine, Seoul, 02841, Republic of Korea.,BK21 Graduate Program, Department of Biomedical Sciences, Korea University College of Medicine, Seoul, 02841, Republic of Korea
| |
Collapse
|
138
|
Moreau MM, Pietropaolo S, Ezan J, Robert BJA, Miraux S, Maître M, Cho Y, Crusio WE, Montcouquiol M, Sans N. Scribble Controls Social Motivation Behavior through the Regulation of the ERK/Mnk1 Pathway. Cells 2022; 11:cells11101601. [PMID: 35626639 PMCID: PMC9139383 DOI: 10.3390/cells11101601] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 05/06/2022] [Accepted: 05/06/2022] [Indexed: 02/01/2023] Open
Abstract
Social behavior is a basic domain affected by several neurodevelopmental disorders, including ASD and a heterogeneous set of neuropsychiatric disorders. The SCRIB gene that codes for the polarity protein SCRIBBLE has been identified as a risk gene for spina bifida, the most common type of neural tube defect, found at high frequencies in autistic patients, as well as other congenital anomalies. The deletions and mutations of the 8q24.3 region encompassing SCRIB are also associated with multisyndromic and rare disorders. Nonetheless, the potential link between SCRIB and relevant social phenotypes has not been fully investigated. Hence, we show that Scribcrc/+ mice, carrying a mutated version of Scrib, displayed reduced social motivation behavior and social habituation, while other behavioral domains were unaltered. Social deficits were associated with the upregulation of ERK phosphorylation, together with increased c-Fos activity. Importantly, the social alterations were rescued by both direct and indirect pERK inhibition. These results support a link between polarity genes, social behaviors and hippocampal functionality and suggest a role for SCRIB in the etiopathology of neurodevelopmental disorders. Furthermore, our data demonstrate the crucial role of the MAPK/ERK signaling pathway in underlying social motivation behavior, thus supporting its relevance as a therapeutic target.
Collapse
Affiliation(s)
- Maïté M. Moreau
- Univ. Bordeaux, INSERM, Neurocentre Magendie, U1215, 33077 Bordeaux, France; (J.E.); (B.J.A.R.); (M.M.); (M.M.)
- Correspondence: (M.M.M.); (N.S.)
| | - Susanna Pietropaolo
- Univ. Bordeaux, CNRS, Aquitaine Institute for Cognitive and Integrative Neurosciences, UMR5287, 33405 Bordeaux, France; (S.P.); (Y.C.); (W.E.C.)
| | - Jérôme Ezan
- Univ. Bordeaux, INSERM, Neurocentre Magendie, U1215, 33077 Bordeaux, France; (J.E.); (B.J.A.R.); (M.M.); (M.M.)
| | - Benjamin J. A. Robert
- Univ. Bordeaux, INSERM, Neurocentre Magendie, U1215, 33077 Bordeaux, France; (J.E.); (B.J.A.R.); (M.M.); (M.M.)
| | - Sylvain Miraux
- Univ. Bordeaux, CNRS, Centre de Résonance Magnétique des Systèmes Biologiques UMR5536, 33077 Bordeaux, France;
| | - Marlène Maître
- Univ. Bordeaux, INSERM, Neurocentre Magendie, U1215, 33077 Bordeaux, France; (J.E.); (B.J.A.R.); (M.M.); (M.M.)
| | - Yoon Cho
- Univ. Bordeaux, CNRS, Aquitaine Institute for Cognitive and Integrative Neurosciences, UMR5287, 33405 Bordeaux, France; (S.P.); (Y.C.); (W.E.C.)
| | - Wim E. Crusio
- Univ. Bordeaux, CNRS, Aquitaine Institute for Cognitive and Integrative Neurosciences, UMR5287, 33405 Bordeaux, France; (S.P.); (Y.C.); (W.E.C.)
| | - Mireille Montcouquiol
- Univ. Bordeaux, INSERM, Neurocentre Magendie, U1215, 33077 Bordeaux, France; (J.E.); (B.J.A.R.); (M.M.); (M.M.)
| | - Nathalie Sans
- Univ. Bordeaux, INSERM, Neurocentre Magendie, U1215, 33077 Bordeaux, France; (J.E.); (B.J.A.R.); (M.M.); (M.M.)
- Correspondence: (M.M.M.); (N.S.)
| |
Collapse
|
139
|
I SP, I GD, L B, M S, J GR, A M EO, I M AB, C LM, E M PV, J A A, E B, J L V, R M DP, R R. The Absence of Caspase-8 in the Dopaminergic System Leads to Mild Autism-like Behavior. Front Cell Dev Biol 2022; 10:839715. [PMID: 35493109 PMCID: PMC9045412 DOI: 10.3389/fcell.2022.839715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 03/16/2022] [Indexed: 11/23/2022] Open
Abstract
In the last decade, new non-apoptotic roles have been ascribed to apoptotic caspases. This family of proteins plays an important role in the sculpting of the brain in the early stages of development by eliminating excessive and nonfunctional synapses and extra cells. Consequently, impairments in this process can underlie many neurological and mental illnesses. This view is particularly relevant to dopamine because it plays a pleiotropic role in motor control, motivation, and reward processing. In this study, we analyze the effects of the elimination of caspase-8 (CASP8) on the development of catecholaminergic neurons using neurochemical, ultrastructural, and behavioral tests. To do this, we selectively delete the CASP8 gene in cells that express tyrosine hydroxylase with the help of recombination through the Cre-loxP system. Our results show that the number of dopaminergic neurons increases in the substantia nigra. In the striatum, the basal extracellular level of dopamine and potassium-evoked dopamine release decreased significantly in mice lacking CASP8, clearly showing the low dopamine functioning in tissues innervated by this neurotransmitter. This view is supported by electron microscopy analysis of striatal synapses. Interestingly, behavioral analysis demonstrates that mice lacking CASP8 show changes reminiscent of autism spectrum disorders (ASD). Our research reactivates the possible role of dopamine transmission in the pathogenesis of ASD and provides a mild model of autism.
Collapse
Affiliation(s)
- Suárez-Pereira I
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Sevilla, Spain.,Neuropsychopharmacology and Psychobiology Research Group, Instituto de Investigación e Innovación en Ciencias Biomédicas de Cádiz, INiBICA, University of Cádiz, Cádiz, Spain
| | - García-Domínguez I
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Instituto de Biomedicina de Sevilla-Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
| | - Bravo L
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Sevilla, Spain.,Neuropsychopharmacology and Psychobiology Research Group, Instituto de Investigación e Innovación en Ciencias Biomédicas de Cádiz, INiBICA, University of Cádiz, Cádiz, Spain
| | - Santiago M
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Instituto de Biomedicina de Sevilla-Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
| | - García-Revilla J
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Instituto de Biomedicina de Sevilla-Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
| | - Espinosa-Oliva A M
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Instituto de Biomedicina de Sevilla-Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
| | - Alonso-Bellido I M
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Instituto de Biomedicina de Sevilla-Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
| | - López-Martín C
- Neuropsychopharmacology and Psychobiology Research Group, Instituto de Investigación e Innovación en Ciencias Biomédicas de Cádiz, INiBICA, University of Cádiz, Cádiz, Spain
| | - Pérez-Villegas E M
- Departamento de Fisiología, Anatomía y Biología Celular, Universidad Pablo de Olavide, Sevilla, Spain
| | - Armengol J A
- Departamento de Fisiología, Anatomía y Biología Celular, Universidad Pablo de Olavide, Sevilla, Spain
| | - Berrocoso E
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Sevilla, Spain.,Neuropsychopharmacology and Psychobiology Research Group, Instituto de Investigación e Innovación en Ciencias Biomédicas de Cádiz, INiBICA, University of Cádiz, Cádiz, Spain
| | - Venero J L
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Instituto de Biomedicina de Sevilla-Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
| | - de Pablos R M
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Instituto de Biomedicina de Sevilla-Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
| | - Ruiz R
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Instituto de Biomedicina de Sevilla-Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
| |
Collapse
|
140
|
Song W, Li Q, Wang T, Li Y, Fan T, Zhang J, Wang Q, Pan J, Dong Q, Sun ZS, Wang Y. Putative complement control protein CSMD3 dysfunction impairs synaptogenesis and induces neurodevelopmental disorders. Brain Behav Immun 2022; 102:237-250. [PMID: 35245678 DOI: 10.1016/j.bbi.2022.02.027] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 02/10/2022] [Accepted: 02/26/2022] [Indexed: 12/23/2022] Open
Abstract
Recent studies have reported that complement-related proteins modulate brain development through regulating synapse processes in the cortex. CSMD3 belongs to a group of putative complement control proteins. However, its role in the central nervous system and synaptogenesis remains largely unknown. Here we report that CSMD3 deleterious mutations occur frequently in patients with neurodevelopmental disorders (NDDs). Csmd3 is predominantly expressed in cortical neurons of the developing cortex. In mice, Csmd3 disruption induced retarded development and NDD-related behaviors. Csmd3 deficiency impaired synaptogenesis and neurogenesis, allowing fewer neurons reaching the cortical plate. Csmd3 deficiency also induced perturbed functional networks in the developing cortex, involving a number of downregulated synapse-associated genes that influence early synaptic organization and upregulated genes related to immune activity. Our study provides mechanistic insights into the endogenous regulation of complement-related proteins in synaptic development and supports the pathological role of CSMD3 in NDDs.
Collapse
Affiliation(s)
- Wei Song
- Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing 100101, China; CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Quan Li
- School of Life Sciences, Hebei University, Baoding 071002, China
| | - Tao Wang
- Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing 100101, China
| | - Yuanyuan Li
- Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing 100101, China; CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Tianda Fan
- Institute of Genomic Medicine, Wenzhou Medical University, Wenzhou 325000, China
| | - Jianghong Zhang
- Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing 100101, China; CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qingqing Wang
- Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing 100101, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jinrong Pan
- Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing 100101, China; CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qiwen Dong
- Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing 100101, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhong Sheng Sun
- Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing 100101, China; CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing 100049, China; School of Life Sciences, Hebei University, Baoding 071002, China; Institute of Genomic Medicine, Wenzhou Medical University, Wenzhou 325000, China; State Key Laboratory of Integrated Management of Pest Insects and Rodents, Chinese Academy of Sciences, Beijing 100101, China.
| | - Yan Wang
- Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing 100101, China; CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
141
|
Oe S, Hayashi S, Tanaka S, Koike T, Hirahara Y, Seki-Omura R, Kakizaki R, Sakamoto S, Nakano Y, Noda Y, Yamada H, Kitada M. Cytoplasmic Polyadenylation Element-Binding Protein 1 Post-transcriptionally Regulates Fragile X Mental Retardation 1 Expression Through 3′ Untranslated Region in Central Nervous System Neurons. Front Cell Neurosci 2022; 16:869398. [PMID: 35496917 PMCID: PMC9051318 DOI: 10.3389/fncel.2022.869398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 03/24/2022] [Indexed: 11/13/2022] Open
Abstract
Fragile X syndrome (FXS) is an inherited intellectual disability caused by a deficiency in Fragile X mental retardation 1 (Fmr1) gene expression. Recent studies have proposed the importance of cytoplasmic polyadenylation element-binding protein 1 (CPEB1) in FXS pathology; however, the molecular interaction between Fmr1 mRNA and CPEB1 has not been fully investigated. Here, we revealed that CPEB1 co-localized and interacted with Fmr1 mRNA in hippocampal and cerebellar neurons and culture cells. Furthermore, CPEB1 knockdown upregulated Fmr1 mRNA and protein levels and caused aberrant localization of Fragile X mental retardation protein in neurons. In an FXS cell model, CPEB1 knockdown upregulated the mRNA levels of several mitochondria-related genes and rescued the intracellular heat shock protein family A member 9 distribution. These findings suggest that CPEB1 post-transcriptionally regulated Fmr1 expression through the 3′ untranslated region, and that CPEB1 knockdown might affect mitochondrial function.
Collapse
Affiliation(s)
- Souichi Oe
- Department of Anatomy, Kansai Medical University, Hirakata, Japan
- *Correspondence: Souichi Oe,
| | - Shinichi Hayashi
- Department of Anatomy, Kansai Medical University, Hirakata, Japan
| | - Susumu Tanaka
- Department of Anatomy, Kansai Medical University, Hirakata, Japan
| | - Taro Koike
- Department of Anatomy, Kansai Medical University, Hirakata, Japan
| | - Yukie Hirahara
- Department of Anatomy, Kansai Medical University, Hirakata, Japan
| | | | - Rio Kakizaki
- Department of Anatomy, Kansai Medical University, Hirakata, Japan
| | - Sumika Sakamoto
- Department of Anatomy, Kansai Medical University, Hirakata, Japan
| | - Yosuke Nakano
- Department of Anatomy, Kansai Medical University, Hirakata, Japan
| | - Yasuko Noda
- Department of Anatomy, Bio-Imaging and Neuro-Cell Science, Jichi Medical University, Shimotsuke, Japan
| | - Hisao Yamada
- Biwako Professional University of Rehabilitation, Higashiomi, Japan
| | - Masaaki Kitada
- Department of Anatomy, Kansai Medical University, Hirakata, Japan
- Masaaki Kitada,
| |
Collapse
|
142
|
D’Incal C, Broos J, Torfs T, Kooy RF, Vanden Berghe W. Towards Kinase Inhibitor Therapies for Fragile X Syndrome: Tweaking Twists in the Autism Spectrum Kinase Signaling Network. Cells 2022; 11:cells11081325. [PMID: 35456004 PMCID: PMC9029738 DOI: 10.3390/cells11081325] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 04/01/2022] [Accepted: 04/03/2022] [Indexed: 12/12/2022] Open
Abstract
Absence of the Fragile X Mental Retardation Protein (FMRP) causes autism spectrum disorders and intellectual disability, commonly referred to as the Fragile X syndrome. FMRP is a negative regulator of protein translation and is essential for neuronal development and synapse formation. FMRP is a target for several post-translational modifications (PTMs) such as phosphorylation and methylation, which tightly regulate its cellular functions. Studies have indicated the involvement of FMRP in a multitude of cellular pathways, and an absence of FMRP was shown to affect several neurotransmitter receptors, for example, the GABA receptor and intracellular signaling molecules such as Akt, ERK, mTOR, and GSK3. Interestingly, many of these molecules function as protein kinases or phosphatases and thus are potentially amendable by pharmacological treatment. Several treatments acting on these kinase-phosphatase systems have been shown to be successful in preclinical models; however, they have failed to convincingly show any improvements in clinical trials. In this review, we highlight the different protein kinase and phosphatase studies that have been performed in the Fragile X syndrome. In our opinion, some of the paradoxical study conclusions are potentially due to the lack of insight into integrative kinase signaling networks in the disease. Quantitative proteome analyses have been performed in several models for the FXS to determine global molecular processes in FXS. However, only one phosphoproteomics study has been carried out in Fmr1 knock-out mouse embryonic fibroblasts, and it showed dysfunctional protein kinase and phosphatase signaling hubs in the brain. This suggests that the further use of phosphoproteomics approaches in Fragile X syndrome holds promise for identifying novel targets for kinase inhibitor therapies.
Collapse
Affiliation(s)
- Claudio D’Incal
- Protein Chemistry, Proteomics and Epigenetic Signaling (PPES), Department of Biomedical Sciences, University of Antwerp, 2000 Antwerp, Belgium; (C.D.); (J.B.); (T.T.)
- Department of Medical Genetics, University of Antwerp, 2000 Antwerp, Belgium;
| | - Jitse Broos
- Protein Chemistry, Proteomics and Epigenetic Signaling (PPES), Department of Biomedical Sciences, University of Antwerp, 2000 Antwerp, Belgium; (C.D.); (J.B.); (T.T.)
| | - Thierry Torfs
- Protein Chemistry, Proteomics and Epigenetic Signaling (PPES), Department of Biomedical Sciences, University of Antwerp, 2000 Antwerp, Belgium; (C.D.); (J.B.); (T.T.)
| | - R. Frank Kooy
- Department of Medical Genetics, University of Antwerp, 2000 Antwerp, Belgium;
| | - Wim Vanden Berghe
- Protein Chemistry, Proteomics and Epigenetic Signaling (PPES), Department of Biomedical Sciences, University of Antwerp, 2000 Antwerp, Belgium; (C.D.); (J.B.); (T.T.)
- Correspondence: ; Tel.: +0032-(0)-32-652-657
| |
Collapse
|
143
|
Bonefas KM, Iwase S. Soma-to-germline transformation in chromatin-linked neurodevelopmental disorders? FEBS J 2022; 289:2301-2317. [PMID: 34514717 PMCID: PMC8918023 DOI: 10.1111/febs.16196] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 08/16/2021] [Accepted: 09/10/2021] [Indexed: 01/22/2023]
Abstract
Mutations in numerous chromatin regulators cause neurodevelopmental disorders (NDDs) with unknown mechanisms. Understandably, most research has focused on how chromatin regulators control gene expression that is directly relevant to brain development and function, such as synaptic genes. However, some NDD models surprisingly show ectopic expression of germline genes in the brain. These germline genes are usually expressed only in the primordial germ cells, testis, and ovaries for germ cell development and sexual reproduction. Such ectopic germline gene expression has been reported in several NDDs, including immunodeficiency, centromeric instability, facial anomalies syndrome 1; Kleefstra syndrome 1; MeCP2 duplication syndrome; and mental retardation, X-linked syndromic, Claes-Jensen type. The responsible genes, DNMT3B, G9A/GLP, MECP2, and KDM5C, all encode chromatin regulators for gene silencing. These mutations may therefore lead to germline gene derepression and, in turn, a severe identity crisis of brain cells-potentially interfering with normal brain development. Thus, the ectopic expression of germline genes is a unique hallmark defining this NDD subset and further implicates the importance of germline gene silencing during brain development. The functional impact of germline gene expression on brain development, however, remains undetermined. This perspective article explores how this apparent soma-to-germline transformation arises and how it may interfere with neurodevelopment through genomic instability and impaired sensory cilium formation. Furthermore, we also discuss how to test these hypotheses experimentally to ultimately determine the contribution of ectopic germline transcripts to chromatin-linked NDDs.
Collapse
Affiliation(s)
- Katherine M. Bonefas
- Department of Human Genetics, Michigan Medicine, University of Michigan, Ann Arbor, MI 48109,The University of Michigan Neuroscience Graduate Program,Corresponding authors: Please address correspondence to: , and
| | - Shigeki Iwase
- Department of Human Genetics, Michigan Medicine, University of Michigan, Ann Arbor, MI 48109,The University of Michigan Neuroscience Graduate Program,Corresponding authors: Please address correspondence to: , and
| |
Collapse
|
144
|
Chen G, Han L, Tan S, Jia X, Wu H, Quan Y, Zhang Q, Yu B, Hu Z, Xia K, Guo H. Loss-of-function of KMT5B leads to neurodevelopmental disorder and impairs neuronal development and neurogenesis. J Genet Genomics 2022; 49:881-890. [PMID: 35331928 DOI: 10.1016/j.jgg.2022.03.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 03/04/2022] [Accepted: 03/06/2022] [Indexed: 11/16/2022]
Abstract
Autism spectrum disorder (ASD) is a group of neurodevelopmental disorders that cause severe social, communication and behavioral problems. Recent studies show that the variants of a histone methyltransferase gene KMT5B, cause neurodevelopmental disorders (NDDs), including ASD and the knockout of Kmt5b in mice is embryonic lethal. However, the detailed genotype-phenotype correlations and functional effects of KMT5B in neurodevelopment are unclear. By targeted sequencing of a large Chinese ASD cohort, analyzing published genome-wide sequencing data, and mining literature, we curated 39 KMT5B variants identified from NDD individuals. A genotype-phenotype correlation analysis for ten individuals with KMT5B pathogenic variants reveals common symptoms, including ASD, intellectual disability, languages problem and macrocephaly. In vitro knockdown of the expression of Kmt5b in cultured mouse primary cortical neurons leads to a decrease in neuronal dendritic complexity and an increase in dendritic spine density, which can be rescued by expression of human KMT5B but not that of pathogenic de novo missense mutants. In vivo knockdown of the Kmt5b expression in the mouse embryonic cerebral cortex by in utero electroporation results in decreased proliferation and accelerated migration of neural progenitor cells. Our findings reveal essential roles of histone methyltransferase KMT5B in neuronal development, prenatal neurogenesis, and neuronal migration.
Collapse
Affiliation(s)
- Guodong Chen
- Center of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan 421001, China
| | - Lin Han
- Center of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan 421001, China; Chongqing Reproductive and Genetics Institute, Chongqing Health Center for Women and Children, Chongqing 400010, China
| | - Senwei Tan
- Center of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan 421001, China
| | - Xiangbin Jia
- Center of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan 421001, China
| | - Huidan Wu
- Center of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan 421001, China
| | - Yingting Quan
- Center of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan 421001, China
| | - Qiumeng Zhang
- Center of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan 421001, China
| | - Bin Yu
- Center of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan 421001, China
| | - Zhengmao Hu
- Center of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan 421001, China
| | - Kun Xia
- Center of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan 421001, China; CAS Center for Excellence in Brain Science and Intelligences Technology (CEBSIT), Chinese Academy of Sciences, Shanghai 200030, China
| | - Hui Guo
- Center of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan 421001, China; Hunan Key Laboratory of Animal Models for Human Diseases, Changsha, Hunan 410078, China.
| |
Collapse
|
145
|
CMTr cap-adjacent 2'-O-ribose mRNA methyltransferases are required for reward learning and mRNA localization to synapses. Nat Commun 2022; 13:1209. [PMID: 35260552 PMCID: PMC8904806 DOI: 10.1038/s41467-022-28549-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 01/18/2022] [Indexed: 11/25/2022] Open
Abstract
Cap-adjacent nucleotides of animal, protist and viral mRNAs can be O-methylated at the 2‘ position of the ribose (cOMe). The functions of cOMe in animals, however, remain largely unknown. Here we show that the two cap methyltransferases (CMTr1 and CMTr2) of Drosophila can methylate the ribose of the first nucleotide in mRNA. Double-mutant flies lack cOMe but are viable. Consistent with prominent neuronal expression, they have a reward learning defect that can be rescued by conditional expression in mushroom body neurons before training. Among CMTr targets are cell adhesion and signaling molecules. Many are relevant for learning, and are also targets of Fragile X Mental Retardation Protein (FMRP). Like FMRP, cOMe is required for localization of untranslated mRNAs to synapses and enhances binding of the cap binding complex in the nucleus. Hence, our study reveals a mechanism to co-transcriptionally prime mRNAs by cOMe for localized protein synthesis at synapses. The two cap methyltransferases (CMTrs) redundantly methylate riboses of first cap adjacent nucleotides in messenger RNAs in Drosophila. Here, CMTrs are required for reward learning and localization of untranslated messenger RNAs to synapses.
Collapse
|
146
|
Kim J, Roh JD, Kim S, Kang H, Bae M, Kim E. Slc6a20a Heterozygous and Homozygous Mutant Mice Display Differential Behavioral and Transcriptomic Changes. Front Mol Neurosci 2022; 15:857820. [PMID: 35321029 PMCID: PMC8936588 DOI: 10.3389/fnmol.2022.857820] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 01/28/2022] [Indexed: 11/23/2022] Open
Abstract
SLC6A20A is a proline and glycine transporter known to regulate glycine homeostasis and NMDA receptor (NMDAR) function in the brain. A previous study found increases in ambient glycine levels and NMDA receptor-mediated synaptic transmission in the brains of Slc6a20a-haploinsufficient mice, but it remained unknown whether Slc6a20a deficiency leads to disease-related behavioral deficits in mice. Here, we report that Slc6a20a heterozygous and homozygous mutant mice display differential behavioral phenotypes in locomotor, repetitive behavioral, and spatial and fear memory domains. In addition, these mice show differential transcriptomic changes in synapse, ribosome, mitochondria, autism, epilepsy, and neuron-related genes. These results suggest that heterozygous and homozygous Slc6a20a deletions in mice lead to differential changes in behaviors and transcriptomes.
Collapse
Affiliation(s)
- Junhyung Kim
- Department of Biological Sciences, Korea Advanced Institute for Science and Technology (KAIST), Daejeon, South Korea
| | - Junyeop Daniel Roh
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science (IBS), Daejeon, South Korea
| | - Seongbin Kim
- Department of Biological Sciences, Korea Advanced Institute for Science and Technology (KAIST), Daejeon, South Korea
| | - Hyojin Kang
- Division of National Supercomputing, Korea Institute of Science and Technology Information (KISTI), Daejeon, South Korea
| | - Mihyun Bae
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science (IBS), Daejeon, South Korea
| | - Eunjoon Kim
- Department of Biological Sciences, Korea Advanced Institute for Science and Technology (KAIST), Daejeon, South Korea
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science (IBS), Daejeon, South Korea
- *Correspondence: Eunjoon Kim,
| |
Collapse
|
147
|
Meng L, Kaufmann WE, Frye RE, Ong K, Kaminski JW, Velinov M, Berry-Kravis E. The association between mosaicism type and cognitive and behavioral functioning among males with fragile X syndrome. Am J Med Genet A 2022; 188:858-866. [PMID: 35148024 PMCID: PMC10948005 DOI: 10.1002/ajmg.a.62594] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Revised: 11/09/2021] [Accepted: 11/22/2021] [Indexed: 12/26/2022]
Abstract
Mosaicism in fragile X syndrome (FXS) refers to two different FMR1 allele variations: size mosaicism represents different numbers of CGG repeats between the two alleles, such that in addition to a full mutation allele there is an allele in the normal or premutation range of CGG repeats, while methylation mosaicism indicates whether a full-mutation allele is fully or partially methylated. The present study explored the association between mosaicism type and cognitive and behavioral functioning in a large sample of males 3 years and older (n = 487) with FXS, participating in the Fragile X Online Registry with Accessible Research Database. Participants with methylation mosaicism were less severely cognitively affected as indicated by a less severe intellectual disability rating, higher intelligence quotient and adaptive behavior score, and lower social impairment score. In contrast, the presence of size mosaicism was not significantly associated with better cognitive and behavioral outcomes than full mutation. Our findings suggest that methylation mosaicism is associated with better cognitive functioning and adaptive behavior and less social impairment. Further research could assess to what extent these cognitive and behavioral differences depend on molecular diagnostic methods and the impact of mosaicism on prognosis of individuals with FXS.
Collapse
Affiliation(s)
- Lu Meng
- Centers for Disease Control and Prevention, National Center on Birth Defects and Developmental Disabilities, Atlanta, Georgia, USA
| | - Walter E. Kaufmann
- Department of Human Genetics, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Richard E. Frye
- Section on Neurodevelopmental Disorders, Barrow Neurological Institute at Phoenix Children's Hospital, Phoenix, Arizona, USA
- Department of Child Health, University of Arizona College of Medicine – Phoenix, Phoenix, Arizona, USA
| | - Katherine Ong
- Centers for Disease Control and Prevention, National Center on Birth Defects and Developmental Disabilities, Atlanta, Georgia, USA
| | - Jennifer W. Kaminski
- Centers for Disease Control and Prevention, National Center on Birth Defects and Developmental Disabilities, Atlanta, Georgia, USA
| | - Milen Velinov
- Rutgers Robert Wood Johnson Medical School, New Brunswick, New Jersey, USA
| | - Elizabeth Berry-Kravis
- Departments of Pediatrics and Neurological Sciences, Rush University Medical Center, Chicago, Illinois, USA
| |
Collapse
|
148
|
Brašić JR, Goodman JA, Nandi A, Russell DS, Jennings D, Barret O, Martin SD, Slifer K, Sedlak T, Mathur AK, Seibyl JP, Berry-Kravis EM, Wong DF, Budimirovic DB. Fragile X Mental Retardation Protein and Cerebral Expression of Metabotropic Glutamate Receptor Subtype 5 in Men with Fragile X Syndrome: A Pilot Study. Brain Sci 2022; 12:314. [PMID: 35326270 PMCID: PMC8946825 DOI: 10.3390/brainsci12030314] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 01/26/2022] [Accepted: 02/09/2022] [Indexed: 02/04/2023] Open
Abstract
Multiple lines of evidence suggest that a deficiency of Fragile X Mental Retardation Protein (FMRP) mediates dysfunction of the metabotropic glutamate receptor subtype 5 (mGluR5) in the pathogenesis of fragile X syndrome (FXS), the most commonly known single-gene cause of inherited intellectual disability (ID) and autism spectrum disorder (ASD). Nevertheless, animal and human studies regarding the link between FMRP and mGluR5 expression provide inconsistent or conflicting findings about the nature of those relationships. Since multiple clinical trials of glutamatergic agents in humans with FXS did not demonstrate the amelioration of the behavioral phenotype observed in animal models of FXS, we sought measure if mGluR5 expression is increased in men with FXS to form the basis for improved clinical trials. Unexpectedly marked reductions in mGluR5 expression were observed in cortical and subcortical regions in men with FXS. Reduced mGluR5 expression throughout the living brains of men with FXS provides a clue to examine FMRP and mGluR5 expression in FXS. In order to develop the findings of our previous study and to strengthen the objective tools for future clinical trials of glutamatergic agents in FXS, we sought to assess the possible value of measuring both FMRP levels and mGluR5 expression in men with FXS. We aimed to show the value of measurement of FMRP levels and mGluR5 expression for the diagnosis and treatment of individuals with FXS and related conditions. We administered 3-[18F]fluoro-5-(2-pyridinylethynyl)benzonitrile ([18F]FPEB), a specific mGluR5 radioligand for quantitative measurements of the density and the distribution of mGluR5s, to six men with the full mutation (FM) of FXS and to one man with allele size mosaicism for FXS (FXS-M). Utilizing the seven cortical and subcortical regions affected in neurodegenerative disorders as indicator variables, adjusted linear regression of mGluR5 expression and FMRP showed that mGluR5 expression was significantly reduced in the occipital cortex and the thalamus relative to baseline (anterior cingulate cortex) if FMRP levels are held constant (F(7,47) = 6.84, p < 0.001).These findings indicate the usefulness of cerebral mGluR5 expression measured by PET with [18F]FPEB and FMRP values in men with FXS and related conditions for assessments in community facilities within a hundred-mile radius of a production center with a cyclotron. These initial results of this pilot study advance our previous study regarding the measurement of mGluR5 expression by combining both FMRP levels and mGluR5 expression as tools for meaningful clinical trials of glutamatergic agents for men with FXS. We confirm the feasibility of this protocol as a valuable tool to measure FMRP levels and mGluR5 expression in clinical trials of individuals with FXS and related conditions and to provide the foundations to apply precision medicine to tailor treatment plans to the specific needs of individuals with FXS and related conditions.
Collapse
Affiliation(s)
- James Robert Brašić
- Section of High Resolution Brain Positron Emission Tomography Imaging, Division of Nuclear Medicine and Molecular Imaging, The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; (A.N.); (S.D.M.); (T.S.); (A.K.M.); (D.F.W.)
| | - Jack Alexander Goodman
- Frank H. Netter MD School of Medicine, Quinnipiac University, North Haven, CT 06473, USA;
| | - Ayon Nandi
- Section of High Resolution Brain Positron Emission Tomography Imaging, Division of Nuclear Medicine and Molecular Imaging, The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; (A.N.); (S.D.M.); (T.S.); (A.K.M.); (D.F.W.)
| | - David S. Russell
- Institute for Neurodegenerative Disorders, New Haven, CT 06510, USA; (D.S.R.); (D.J.); (O.B.); (J.P.S.)
- Invicro, New Haven, CT 06510, USA
| | - Danna Jennings
- Institute for Neurodegenerative Disorders, New Haven, CT 06510, USA; (D.S.R.); (D.J.); (O.B.); (J.P.S.)
- Invicro, New Haven, CT 06510, USA
- Denali Therapeutics, Inc., South San Francisco, CA 94080, USA
| | - Olivier Barret
- Institute for Neurodegenerative Disorders, New Haven, CT 06510, USA; (D.S.R.); (D.J.); (O.B.); (J.P.S.)
- Invicro, New Haven, CT 06510, USA
- Laboratoire des Maladies Neurodégénératives, Molecular Imaging Research Center (MIRCen), Institut de Biologie François Jacob, Centre National de la Recherche Scientifique (CNRS), Commissariat à l’Énergie Atomique et aux Énergies Alternatives (CEA), Université Paris-Saclay, CEDEX, 92265 Fontenay-aux-Roses, France
| | - Samuel D. Martin
- Section of High Resolution Brain Positron Emission Tomography Imaging, Division of Nuclear Medicine and Molecular Imaging, The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; (A.N.); (S.D.M.); (T.S.); (A.K.M.); (D.F.W.)
- Department of Neuroscience, Zanvyl Krieger School of Arts and Sciences, The Johns Hopkins University, Baltimore, MD 21218, USA
| | - Keith Slifer
- Department of Psychiatry and Behavioral Sciences-Child Psychiatry, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA;
- Department of Behavioral Psychology, Kennedy Krieger Institute, Baltimore, MD 21205, USA
| | - Thomas Sedlak
- Section of High Resolution Brain Positron Emission Tomography Imaging, Division of Nuclear Medicine and Molecular Imaging, The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; (A.N.); (S.D.M.); (T.S.); (A.K.M.); (D.F.W.)
- Department of Psychiatry and Behavioral Sciences-General Psychiatry, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Anil Kumar Mathur
- Section of High Resolution Brain Positron Emission Tomography Imaging, Division of Nuclear Medicine and Molecular Imaging, The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; (A.N.); (S.D.M.); (T.S.); (A.K.M.); (D.F.W.)
| | - John P. Seibyl
- Institute for Neurodegenerative Disorders, New Haven, CT 06510, USA; (D.S.R.); (D.J.); (O.B.); (J.P.S.)
- Invicro, New Haven, CT 06510, USA
| | - Elizabeth M. Berry-Kravis
- Departments of Pediatrics, Neurological Sciences, and Biochemistry, Rush University Medical Center, Chicago, IL 60612, USA;
| | - Dean F. Wong
- Section of High Resolution Brain Positron Emission Tomography Imaging, Division of Nuclear Medicine and Molecular Imaging, The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; (A.N.); (S.D.M.); (T.S.); (A.K.M.); (D.F.W.)
- Laboratory of Central Nervous System (CNS) Neuropsychopharmacology and Multimodal, Imaging (CNAMI), Mallinckrodt Institute of Radiology, Washington University, Saint Louis, MO 63110, USA
| | - Dejan B. Budimirovic
- Department of Psychiatry and Behavioral Sciences-Child Psychiatry, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA;
- Department of Psychiatry, Kennedy Krieger Institute, Baltimore, MD 21205, USA
| |
Collapse
|
149
|
Protic DD, Aishworiya R, Salcedo-Arellano MJ, Tang SJ, Milisavljevic J, Mitrovic F, Hagerman RJ, Budimirovic DB. Fragile X Syndrome: From Molecular Aspect to Clinical Treatment. Int J Mol Sci 2022; 23:1935. [PMID: 35216055 PMCID: PMC8875233 DOI: 10.3390/ijms23041935] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 02/01/2022] [Accepted: 02/02/2022] [Indexed: 02/01/2023] Open
Abstract
Fragile X syndrome (FXS) is a neurodevelopmental disorder caused by the full mutation as well as highly localized methylation of the fragile X mental retardation 1 (FMR1) gene on the long arm of the X chromosome. Children with FXS are commonly co-diagnosed with Autism Spectrum Disorder, attention and learning problems, anxiety, aggressive behavior and sleep disorder, and early interventions have improved many behavior symptoms associated with FXS. In this review, we performed a literature search of original and review articles data of clinical trials and book chapters using MEDLINE (1990-2021) and ClinicalTrials.gov. While we have reviewed the biological importance of the fragile X mental retardation protein (FMRP), the FXS phenotype, and current diagnosis techniques, the emphasis of this review is on clinical interventions. Early non-pharmacological interventions in combination with pharmacotherapy and targeted treatments aiming to reverse dysregulated brain pathways are the mainstream of treatment in FXS. Overall, early diagnosis and interventions are fundamental to achieve optimal clinical outcomes in FXS.
Collapse
Affiliation(s)
- Dragana D. Protic
- Department of Pharmacology, Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Belgrade, 11129 Belgrade, Serbia
| | - Ramkumar Aishworiya
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute UCDH, University of California Davis, 2825 50th Street, Sacramento, CA 95817, USA; (R.A.); (M.J.S.-A.); (S.J.T.); (R.J.H.)
- Khoo Teck Puat-National University Children’s Medical Institute, National University Health System, 5 Lower Kent Ridge Road, Singapore 119074, Singapore
| | - Maria Jimena Salcedo-Arellano
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute UCDH, University of California Davis, 2825 50th Street, Sacramento, CA 95817, USA; (R.A.); (M.J.S.-A.); (S.J.T.); (R.J.H.)
- Department of Pediatrics, University of California Davis School of Medicine, Sacramento, CA 95817, USA
- Department of Pathology and Laboratory Medicine, University of California Davis School of Medicine, Sacramento, CA 95817, USA
| | - Si Jie Tang
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute UCDH, University of California Davis, 2825 50th Street, Sacramento, CA 95817, USA; (R.A.); (M.J.S.-A.); (S.J.T.); (R.J.H.)
| | - Jelena Milisavljevic
- Faculty of Medicine, University of Belgrade, 11129 Belgrade, Serbia; (J.M.); (F.M.)
| | - Filip Mitrovic
- Faculty of Medicine, University of Belgrade, 11129 Belgrade, Serbia; (J.M.); (F.M.)
| | - Randi J. Hagerman
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute UCDH, University of California Davis, 2825 50th Street, Sacramento, CA 95817, USA; (R.A.); (M.J.S.-A.); (S.J.T.); (R.J.H.)
- Department of Pediatrics, University of California Davis School of Medicine, Sacramento, CA 95817, USA
| | - Dejan B. Budimirovic
- Department of Psychiatry, Fragile X Clinic, Kennedy Krieger Institute, Baltimore, MD 21205, USA
- Department of Psychiatry & Behavioral Sciences-Child Psychiatry, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
150
|
Zhao H, Mao X, Zhu C, Zou X, Peng F, Yang W, Li B, Li G, Ge T, Cui R. GABAergic System Dysfunction in Autism Spectrum Disorders. Front Cell Dev Biol 2022; 9:781327. [PMID: 35198562 PMCID: PMC8858939 DOI: 10.3389/fcell.2021.781327] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 12/16/2021] [Indexed: 12/19/2022] Open
Abstract
Autism spectrum disorder (ASD) refers to a series of neurodevelopmental diseases characterized by two hallmark symptoms, social communication deficits and repetitive behaviors. Gamma-aminobutyric acid (GABA) is one of the most important inhibitory neurotransmitters in the central nervous system (CNS). GABAergic inhibitory neurotransmission is critical for the regulation of brain rhythm and spontaneous neuronal activities during neurodevelopment. Genetic evidence has identified some variations of genes associated with the GABA system, indicating an abnormal excitatory/inhibitory (E/I) neurotransmission ratio implicated in the pathogenesis of ASD. However, the specific molecular mechanism by which GABA and GABAergic synaptic transmission affect ASD remains unclear. Transgenic technology enables translating genetic variations into rodent models to further investigate the structural and functional synaptic dysregulation related to ASD. In this review, we summarized evidence from human neuroimaging, postmortem, and genetic and pharmacological studies, and put emphasis on the GABAergic synaptic dysregulation and consequent E/I imbalance. We attempt to illuminate the pathophysiological role of structural and functional synaptic dysregulation in ASD and provide insights for future investigation.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Ranji Cui
- *Correspondence: Tongtong Ge, ; Ranji Cui,
| |
Collapse
|