1
|
van der Lei MB, Kooy RF. From Discovery to Innovative Translational Approaches in 80 Years of Fragile X Syndrome Research. Biomedicines 2025; 13:805. [PMID: 40299377 PMCID: PMC12024745 DOI: 10.3390/biomedicines13040805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Revised: 03/21/2025] [Accepted: 03/25/2025] [Indexed: 04/30/2025] Open
Abstract
Fragile X syndrome (FXS) is the most common inherited cause of intellectual disability and a major genetic contributor to autism spectrum disorder. It is caused by a CGG trinucleotide repeat expansion in the FMR1 gene, resulting in gene silencing and the loss of FMRP, an RNA-binding protein essential for synaptic plasticity. This review covers over 80 years of FXS research, highlighting key milestones, clinical features, genetic and molecular mechanisms, the FXS mouse model, disrupted molecular pathways, and current therapeutic strategies. Additionally, we discuss recent advances including AI-driven combination therapies, CRISPR-based gene editing, and antisense oligonucleotides (ASOs) therapies. Despite these scientific breakthroughs, translating preclinical findings into effective clinical treatments remains challenging. Clinical trials have faced several difficulties, including patient heterogeneity, inconsistent outcome measures, and variable therapeutic responses. Standardized preclinical testing protocols and refined clinical trial designs are required to overcome these challenges. The development of FXS-specific biomarkers could also improve the precision of treatment assessments. Ultimately, future therapies will need to combine pharmacological and behavioral interventions tailored to individual needs. While significant challenges remain, ongoing research continues to offer hope for transformative breakthroughs that could significantly improve the quality of life for individuals with FXS and their families.
Collapse
Affiliation(s)
| | - R. Frank Kooy
- Center of Medical Genetics, University of Antwerp and Antwerp University Hospital, 2650 Edegem, Belgium;
| |
Collapse
|
2
|
Gunapala KM, Gadban A, Noreen F, Schär P, Benvenisty N, Taylor V. Ascorbic Acid Ameliorates Molecular and Developmental Defects in Human-Induced Pluripotent Stem Cell and Cerebral Organoid Models of Fragile X Syndrome. Int J Mol Sci 2024; 25:12718. [PMID: 39684429 DOI: 10.3390/ijms252312718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 11/15/2024] [Accepted: 11/22/2024] [Indexed: 12/18/2024] Open
Abstract
Fragile X Syndrome (FX) is the most common form of inherited cognitive impairment and falls under the broader category of Autism Spectrum Disorders (ASD). FX is caused by a CGG trinucleotide repeat expansion in the non-coding region of the X-linked Fragile X Messenger Ribonucleoprotein 1 (FMR1) gene, leading to its hypermethylation and epigenetic silencing. Animal models of FX rely on the deletion of the Fmr1 gene, which fails to replicate the epigenetic silencing mechanism of the FMR1 gene observed in human patients. Human stem cells carrying FX repeat expansions have provided a better understanding of the basis of epigenetic silencing of FMR1. Previous studies have found that 5-Azacytidine (5Azac) can reverse this methylation; however, 5Azac can be toxic, which may limit its therapeutic potential. Here, we show that the dietary factor Ascorbic Acid (AsA) can reduce DNA methylation in the FMR1 locus and lead to an increase in FMR1 gene expression in FX iPSCs and cerebral organoids. In addition, AsA treatment rescued neuronal gene expression and morphological defects observed in FX iPSC-derived cerebral organoids. Hence, we demonstrate that the dietary co-factor AsA can partially revert the molecular and morphological defects seen in human FX models in vitro. Our findings have implications for the development of novel therapies for FX in the future.
Collapse
Affiliation(s)
- Keith M Gunapala
- Department of Biomedicine, University of Basel, Mattenstrasse 28, 4058 Basel, Switzerland
- The Azrieli Center for Stem Cells and Genetic Research, Department of Genetics, Institute of Life Sciences, The Hebrew University, Jerusalem 91904, Israel
| | - Aseel Gadban
- The Azrieli Center for Stem Cells and Genetic Research, Department of Genetics, Institute of Life Sciences, The Hebrew University, Jerusalem 91904, Israel
| | - Faiza Noreen
- Department of Biomedicine, University of Basel, Mattenstrasse 28, 4058 Basel, Switzerland
- Swiss Institute of Bioinformatics, 4031 Basel, Switzerland
| | - Primo Schär
- Department of Biomedicine, University of Basel, Mattenstrasse 28, 4058 Basel, Switzerland
| | - Nissim Benvenisty
- The Azrieli Center for Stem Cells and Genetic Research, Department of Genetics, Institute of Life Sciences, The Hebrew University, Jerusalem 91904, Israel
| | - Verdon Taylor
- Department of Biomedicine, University of Basel, Mattenstrasse 28, 4058 Basel, Switzerland
| |
Collapse
|
3
|
Dionne O, Sabatié S, Fortin F, Corbin F, Laurent B. Efficient generation of human induced pluripotent stem cells from urine samples of patients with Fragile X syndrome. Front Cell Dev Biol 2024; 12:1489190. [PMID: 39650724 PMCID: PMC11621072 DOI: 10.3389/fcell.2024.1489190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Accepted: 11/12/2024] [Indexed: 12/11/2024] Open
Abstract
Human induced pluripotent stem cells (iPSCs) are a valuable tool for studying human development and diseases. iPSCs can be generated by reprogramming from any somatic cells, however establishing primary cell cultures can involve invasive procedures (e.g., skin biopsy) and be labor-intensive. In this paper, we describe an efficient, reliable, and non-invasive method for cultivating primary urine-derived cells (UDCs) and efficiently reprogram them into iPSCs using a feeder-free and non-integrative system. This approach has several advantages: (i) UDCs collection and culture are non-invasive, straightforward, and do not require medical personnel; (ii) reprogramming UDCs using commercially available Sendai viruses is highly efficient and reliable; and (iii) iPSCs generated from UDCs demonstrate strong differentiation potential. To showcase the effectiveness of this method, we generated iPSC lines from UDCs of three control individuals and three patients with Fragile X syndrome.
Collapse
Affiliation(s)
- Olivier Dionne
- Department of Biochemistry and Functional Genomics, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Salomé Sabatié
- Department of Biochemistry and Functional Genomics, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Fléchère Fortin
- Medical Genetics division, Centre Hospitalier Universitaire de Sherbrooke (CHUS), Sherbrooke, QC, Canada
| | - François Corbin
- Department of Biochemistry and Functional Genomics, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Benoit Laurent
- Department of Biochemistry and Functional Genomics, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada
- Research Center on Aging, Centre Intégré Universitaire de Santé et Services Sociaux de l’Estrie-Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, QC, Canada
| |
Collapse
|
4
|
Ornoy A, Echefu B, Becker M. Animal Models of Autistic-like Behavior in Rodents: A Scoping Review and Call for a Comprehensive Scoring System. Int J Mol Sci 2024; 25:10469. [PMID: 39408797 PMCID: PMC11477392 DOI: 10.3390/ijms251910469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 09/24/2024] [Accepted: 09/25/2024] [Indexed: 10/20/2024] Open
Abstract
Appropriate animal models of human diseases are a cornerstone in the advancement of science and medicine. To create animal models of neuropsychiatric and neurobehavioral diseases such as autism spectrum disorder (ASD) necessitates the development of sufficient neurobehavioral measuring tools to translate human behavior to expected measurable behavioral features in animals. If possible, the severity of the symptoms should also be assessed. Indeed, at least in rodents, adequate neurobehavioral and neurological tests have been developed. Since ASD is characterized by a number of specific behavioral trends with significant severity, animal models of autistic-like behavior have to demonstrate the specific characteristic features, namely impaired social interactions, communication deficits, and restricted, repetitive behavioral patterns, with association to several additional impairments such as somatosensory, motor, and memory impairments. Thus, an appropriate model must show behavioral impairment of a minimal number of neurobehavioral characteristics using an adequate number of behavioral tests. The proper animal models enable the study of ASD-like-behavior from the etiologic, pathogenetic, and therapeutic aspects. From the etiologic aspects, models have been developed by the use of immunogenic substances like polyinosinic-polycytidylic acid (PolyIC), lipopolysaccharide (LPS), and propionic acid, or other well-documented immunogens or pathogens, like Mycobacterium tuberculosis. Another approach is the use of chemicals like valproic acid, polychlorinated biphenyls (PCBs), organophosphate pesticides like chlorpyrifos (CPF), and others. These substances were administered either prenatally, generally after the period of major organogenesis, or, especially in rodents, during early postnatal life. In addition, using modern genetic manipulation methods, genetic models have been created of almost all human genetic diseases that are manifested by autistic-like behavior (i.e., fragile X, Rett syndrome, SHANK gene mutation, neuroligin genes, and others). Ideally, we should not only evaluate the different behavioral modes affected by the ASD-like behavior, but also assess the severity of the behavioral deviations by an appropriate scoring system, as applied to humans. We therefore propose a scoring system for improved assessment of ASD-like behavior in animal models.
Collapse
Affiliation(s)
- Asher Ornoy
- Department of Morphological Sciences and Teratology, Adelson School of Medicine, Ariel University, Ariel 40700, Israel; (B.E.); (M.B.)
- Hadassah Academic College, Jerusalem 9101001, Israel
- Hadassah Medical School, Hebrew University, Jerusalem 9112102, Israel
| | - Boniface Echefu
- Department of Morphological Sciences and Teratology, Adelson School of Medicine, Ariel University, Ariel 40700, Israel; (B.E.); (M.B.)
| | - Maria Becker
- Department of Morphological Sciences and Teratology, Adelson School of Medicine, Ariel University, Ariel 40700, Israel; (B.E.); (M.B.)
| |
Collapse
|
5
|
Milojevic S, Ghosh A, Makevic V, Stojkovic M, Capovilla M, Tosti T, Budimirovic D, Protic D. Circadian Rhythm and Sleep Analyses in a Fruit Fly Model of Fragile X Syndrome Using a Video-Based Automated Behavioral Research System. Int J Mol Sci 2024; 25:7949. [PMID: 39063191 PMCID: PMC11277495 DOI: 10.3390/ijms25147949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 07/09/2024] [Accepted: 07/18/2024] [Indexed: 07/28/2024] Open
Abstract
Fragile X syndrome (FXS) is caused by the full mutation in the FMR1 gene on the Xq27.3 chromosome region. It is the most common monogenic cause of autism spectrum disorder (ASD) and inherited intellectual disability (ID). Besides ASD and ID and other symptoms, individuals with FXS may exhibit sleep problems and impairment of circadian rhythm (CR). The Drosophila melanogaster models of FXS, such as dFMR1B55, represent excellent models for research in the FXS field. During this study, sleep patterns and CR in dFMR1B55 mutants were analyzed, using a new platform based on continuous high-resolution videography integrated with a highly-customized version of an open-source software. This methodology provides more sensitive results, which could be crucial for all further research in this model of fruit flies. The study revealed that dFMR1B55 male mutants sleep more and can be considered weak rhythmic flies rather than totally arrhythmic and present a good alternative animal model of genetic disorder, which includes impairment of CR and sleep behavior. The combination of affordable videography and software used in the current study is a significant improvement over previous methods and will enable broader adaptation of such high-resolution behavior monitoring methods.
Collapse
Affiliation(s)
- Sara Milojevic
- Department of Pharmacology, Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia; (S.M.); (M.S.)
| | - Arijit Ghosh
- Chronobiology and Behavioral Neurogenetics Laboratory, Neuroscience Unit, Jawaharlal Nehru Center for Advanced Scientific Research, Jakkur, Bangalore 560064, India;
| | - Vedrana Makevic
- Department of Pathophysiology, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia;
| | - Maja Stojkovic
- Department of Pharmacology, Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia; (S.M.); (M.S.)
| | - Maria Capovilla
- UMR7275 CNRS-INSERM-UniCA, Institute of Cellular and Molecular Pharmacology Institute, Sophia Antipolis, 06560 Valbonne, France;
| | - Tomislav Tosti
- Institute of Chemistry, Technology and Metallurgy, National Institute of the Republic of Serbia, University of Belgrade, 11000 Belgrade, Serbia;
| | - Dejan Budimirovic
- Department of Psychiatry, Fragile X Clinic, Kennedy Krieger Institute, Baltimore, MD 21205, USA;
- Department of Psychiatry & Behavioral Sciences-Child Psychiatry, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Dragana Protic
- Department of Pharmacology, Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia; (S.M.); (M.S.)
- Fragile X Clinic, Special Hospital for Cerebral Palsy and Developmental Neurology, 11000 Belgrade, Serbia
| |
Collapse
|
6
|
Martinez JD, Wilson LG, Brancaleone WP, Peterson KG, Popke DS, Garzon VC, Perez Tremble RE, Donnelly MJ, Mendez Ortega SL, Torres D, Shaver JJ, Jiang S, Yang Z, Aton SJ. Hypnotic treatment improves sleep architecture and EEG disruptions and rescues memory deficits in a mouse model of fragile X syndrome. Cell Rep 2024; 43:114266. [PMID: 38787724 PMCID: PMC11910971 DOI: 10.1016/j.celrep.2024.114266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 12/20/2023] [Accepted: 05/08/2024] [Indexed: 05/26/2024] Open
Abstract
Fragile X syndrome (FXS) is associated with disrupted cognition and sleep abnormalities. Sleep loss negatively impacts cognitive function, and one untested possibility is that disrupted cognition in FXS is exacerbated by abnormal sleep. We tested whether ML297, a hypnotic acting on G-protein-activated inward-rectifying potassium (GIRK) channels, could reverse sleep phenotypes and disrupted memory in Fmr1-/y mice. Fmr1-/y mice exhibit reduced non-rapid eye movement (NREM) sleep and fragmented NREM architecture, altered sleep electroencephalogram (EEG) oscillations, and reduced EEG coherence between cortical areas; these are partially reversed following ML297 administration. Treatment following contextual fear or spatial learning restores disrupted memory consolidation in Fmr1-/y mice. During memory recall, Fmr1-/y mice show an altered balance of activity among hippocampal principal neurons vs. parvalbumin-expressing interneurons; this is partially reversed by ML297. Because sleep disruption could impact neurophysiological phenotypes in FXS, augmenting sleep may improve disrupted cognition in this disorder.
Collapse
Affiliation(s)
- Jessy D Martinez
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Lydia G Wilson
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - William P Brancaleone
- Undergraduate Program in Neuroscience, University of Michigan, Ann Arbor, MI 48109, USA
| | - Kathryn G Peterson
- Undergraduate Program in Neuroscience, University of Michigan, Ann Arbor, MI 48109, USA
| | - Donald S Popke
- Undergraduate Program in Neuroscience, University of Michigan, Ann Arbor, MI 48109, USA
| | - Valentina Caicedo Garzon
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Roxanne E Perez Tremble
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Marcus J Donnelly
- Undergraduate Program in Neuroscience, University of Michigan, Ann Arbor, MI 48109, USA
| | | | - Daniel Torres
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - James J Shaver
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Sha Jiang
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Zhongying Yang
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Sara J Aton
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
7
|
Sandoval SO, Méndez-Albelo NM, Xu Z, Zhao X. From wings to whiskers to stem cells: why every model matters in fragile X syndrome research. J Neurodev Disord 2024; 16:30. [PMID: 38872088 PMCID: PMC11177515 DOI: 10.1186/s11689-024-09545-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 05/21/2024] [Indexed: 06/15/2024] Open
Abstract
Fragile X syndrome (FXS) is caused by epigenetic silencing of the X-linked fragile X messenger ribonucleoprotein 1 (FMR1) gene located on chromosome Xq27.3, which leads to the loss of its protein product, fragile X messenger ribonucleoprotein (FMRP). It is the most prevalent inherited form of intellectual disability and the highest single genetic cause of autism. Since the discovery of the genetic basis of FXS, extensive studies using animal models and human pluripotent stem cells have unveiled the functions of FMRP and mechanisms underlying FXS. However, clinical trials have not yielded successful treatment. Here we review what we have learned from commonly used models for FXS, potential limitations of these models, and recommendations for future steps.
Collapse
Affiliation(s)
- Soraya O Sandoval
- Waisman Center, University of Wisconsin-Madison, Madison, WI, 53705, USA
- Department of Neuroscience, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, 53705, USA
- Neuroscience Training Program, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Natasha M Méndez-Albelo
- Waisman Center, University of Wisconsin-Madison, Madison, WI, 53705, USA
- Department of Neuroscience, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, 53705, USA
- Molecular Cellular Pharmacology Training Program, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Zhiyan Xu
- Waisman Center, University of Wisconsin-Madison, Madison, WI, 53705, USA
- Department of Neuroscience, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, 53705, USA
- Graduate Program in Cell and Molecular Biology, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Xinyu Zhao
- Waisman Center, University of Wisconsin-Madison, Madison, WI, 53705, USA.
- Department of Neuroscience, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, 53705, USA.
| |
Collapse
|
8
|
Stojkovic M, Petrovic M, Capovilla M, Milojevic S, Makevic V, Budimirovic DB, Corscadden L, He S, Protic D. Using a Combination of Novel Research Tools to Understand Social Interaction in the Drosophila melanogaster Model for Fragile X Syndrome. BIOLOGY 2024; 13:432. [PMID: 38927312 PMCID: PMC11200401 DOI: 10.3390/biology13060432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 05/31/2024] [Accepted: 06/06/2024] [Indexed: 06/28/2024]
Abstract
Fragile X syndrome (FXS), the most common monogenic cause of inherited intellectual disability and autism spectrum disorder, is caused by a full mutation (>200 CGG repeats) in the Fragile X Messenger Ribonucleoprotein 1 (FMR1) gene. Individuals with FXS experience various challenges related to social interaction (SI). Animal models, such as the Drosophila melanogaster model for FXS where the only ortholog of human FMR1 (dFMR1) is mutated, have played a crucial role in the understanding of FXS. The aim of this study was to investigate SI in the dFMR1B55 mutants (the groups of flies of both sexes simultaneously) using the novel Drosophila Shallow Chamber and a Python data processing pipeline based on social network analysis (SNA). In comparison with wild-type flies (w1118), SNA analysis in dFMR1B55 mutants revealed hypoactivity, fewer connections in their networks, longer interaction duration, a lower ability to transmit information efficiently, fewer alternative pathways for information transmission, a higher variability in the number of interactions they achieved, and flies tended to stay near the boundaries of the testing chamber. These observed alterations indicate the presence of characteristic strain-dependent social networks in dFMR1B55 flies, commonly referred to as the group phenotype. Finally, combining novel research tools is a valuable method for SI research in fruit flies.
Collapse
Affiliation(s)
- Maja Stojkovic
- Department of Pharmacology, Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia; (M.S.); (S.M.)
| | - Milan Petrovic
- Department of Informatics, University of Rijeka, 51000 Rijeka, Croatia;
- Center for Artificial Intelligence and Cybersecurity, University of Rijeka, 51000 Rijeka, Croatia
| | - Maria Capovilla
- UMR7275 CNRS-UCA, Institut de Pharmacologie Moléculaire et Cellulaire, 06560 Valbonne Sophia Antipolis, France;
| | - Sara Milojevic
- Department of Pharmacology, Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia; (M.S.); (S.M.)
| | - Vedrana Makevic
- Department of Pathophysiology, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia;
| | - Dejan B. Budimirovic
- Department of Psychiatry, Fragile X Clinic, Kennedy Krieger Institute, Baltimore, MD 21205, USA;
- Department of Psychiatry & Behavioral Sciences-Child Psychiatry, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
| | | | - Shuhan He
- Lab of Computer Science, Department of Internal Medicine, Harvard Medical School, Boston, MA 02115, USA;
| | - Dragana Protic
- Department of Pharmacology, Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia; (M.S.); (S.M.)
- Fragile X Clinic, Special Hospital for Cerebral Palsy and Developmental Neurology, 11000 Belgrade, Serbia
| |
Collapse
|
9
|
Abbasi DA, Berry-Kravis E, Zhao X, Cologna SM. Proteomics insights into fragile X syndrome: Unraveling molecular mechanisms and therapeutic avenues. Neurobiol Dis 2024; 194:106486. [PMID: 38548140 PMCID: PMC11650894 DOI: 10.1016/j.nbd.2024.106486] [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: 11/21/2023] [Revised: 03/08/2024] [Accepted: 03/25/2024] [Indexed: 04/02/2024] Open
Abstract
Fragile X Syndrome (FXS) is a neurodevelopment disorder characterized by cognitive impairment, behavioral challenges, and synaptic abnormalities, with a genetic basis linked to a mutation in the FMR1 (Fragile X Messenger Ribonucleoprotein 1) gene that results in a deficiency or absence of its protein product, Fragile X Messenger Ribonucleoprotein (FMRP). In recent years, mass spectrometry (MS) - based proteomics has emerged as a powerful tool to uncover the complex molecular landscape underlying FXS. This review provides a comprehensive overview of the proteomics studies focused on FXS, summarizing key findings with an emphasis on dysregulated proteins associated with FXS. These proteins span a wide range of cellular functions including, but not limited to, synaptic plasticity, RNA translation, and mitochondrial function. The work conducted in these proteomic studies provides a more holistic understanding to the molecular pathways involved in FXS and considerably enhances our knowledge into the synaptic dysfunction seen in FXS.
Collapse
Affiliation(s)
- Diana A Abbasi
- Departments of Pediatrics and Neurological Sciences, Rush University Medical Center, Chicago, IL 60612, United States of America
| | - Elizabeth Berry-Kravis
- Departments of Pediatrics and Neurological Sciences, Rush University Medical Center, Chicago, IL 60612, United States of America
| | - Xinyu Zhao
- Department of Neuroscience, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, United States of America
| | - Stephanie M Cologna
- Department of Chemistry, University of Illinois Chicago, Chicago, IL 60607, United States of America.
| |
Collapse
|
10
|
Meneghetti N, Vannini E, Mazzoni A. Rodents' visual gamma as a biomarker of pathological neural conditions. J Physiol 2024; 602:1017-1048. [PMID: 38372352 DOI: 10.1113/jp283858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 01/23/2024] [Indexed: 02/20/2024] Open
Abstract
Neural gamma oscillations (indicatively 30-100 Hz) are ubiquitous: they are associated with a broad range of functions in multiple cortical areas and across many animal species. Experimental and computational works established gamma rhythms as a global emergent property of neuronal networks generated by the balanced and coordinated interaction of excitation and inhibition. Coherently, gamma activity is strongly influenced by the alterations of synaptic dynamics which are often associated with pathological neural dysfunctions. We argue therefore that these oscillations are an optimal biomarker for probing the mechanism of cortical dysfunctions. Gamma oscillations are also highly sensitive to external stimuli in sensory cortices, especially the primary visual cortex (V1), where the stimulus dependence of gamma oscillations has been thoroughly investigated. Gamma manipulation by visual stimuli tuning is particularly easy in rodents, which have become a standard animal model for investigating the effects of network alterations on gamma oscillations. Overall, gamma in the rodents' visual cortex offers an accessible probe on dysfunctional information processing in pathological conditions. Beyond vision-related dysfunctions, alterations of gamma oscillations in rodents were indeed also reported in neural deficits such as migraine, epilepsy and neurodegenerative or neuropsychiatric conditions such as Alzheimer's, schizophrenia and autism spectrum disorders. Altogether, the connections between visual cortical gamma activity and physio-pathological conditions in rodent models underscore the potential of gamma oscillations as markers of neuronal (dys)functioning.
Collapse
Affiliation(s)
- Nicolò Meneghetti
- The Biorobotics Institute, Scuola Superiore Sant'Anna, Pisa, Italy
- Department of Excellence for Robotics and AI, Scuola Superiore Sant'Anna, Pisa, Italy
| | - Eleonora Vannini
- Neuroscience Institute, National Research Council (CNR), Pisa, Italy
| | - Alberto Mazzoni
- The Biorobotics Institute, Scuola Superiore Sant'Anna, Pisa, Italy
- Department of Excellence for Robotics and AI, Scuola Superiore Sant'Anna, Pisa, Italy
| |
Collapse
|
11
|
Fang M, Deibler SK, Krishnamurthy PM, Wang F, Rodriguez P, Banday S, Virbasius CM, Sena-Esteves M, Watts JK, Green MR. EZH2 inhibition reactivates epigenetically silenced FMR1 and normalizes molecular and electrophysiological abnormalities in fragile X syndrome neurons. Front Neurosci 2024; 18:1348478. [PMID: 38449737 PMCID: PMC10915284 DOI: 10.3389/fnins.2024.1348478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Accepted: 02/09/2024] [Indexed: 03/08/2024] Open
Abstract
Fragile X Syndrome (FXS) is a neurological disorder caused by epigenetic silencing of the FMR1 gene. Reactivation of FMR1 is a potential therapeutic approach for FXS that would correct the root cause of the disease. Here, using a candidate-based shRNA screen, we identify nine epigenetic repressors that promote silencing of FMR1 in FXS cells (called FMR1 Silencing Factors, or FMR1- SFs). Inhibition of FMR1-SFs with shRNAs or small molecules reactivates FMR1 in cultured undifferentiated induced pluripotent stem cells, neural progenitor cells (NPCs) and post-mitotic neurons derived from FXS patients. One of the FMR1-SFs is the histone methyltransferase EZH2, for which an FDA-approved small molecule inhibitor, EPZ6438 (also known as tazemetostat), is available. We show that EPZ6438 substantially corrects the characteristic molecular and electrophysiological abnormalities of cultured FXS neurons. Unfortunately, EZH2 inhibitors do not efficiently cross the blood-brain barrier, limiting their therapeutic use for FXS. Recently, antisense oligonucleotide (ASO)-based approaches have been developed as effective treatment options for certain central nervous system disorders. We therefore derived efficacious ASOs targeting EZH2 and demonstrate that they reactivate FMR1 expression and correct molecular and electrophysiological abnormalities in cultured FXS neurons, and reactivate FMR1 expression in human FXS NPCs engrafted within the brains of mice. Collectively, our results establish EZH2 inhibition in general, and EZH2 ASOs in particular, as a therapeutic approach for FXS.
Collapse
Affiliation(s)
- Minggang Fang
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA, United States
| | - Sara K. Deibler
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA, United States
| | | | - Feng Wang
- RNA Therapeutics Institute, University of Massachusetts Chan Medical School, Worcester, MA, United States
| | - Paola Rodriguez
- Department of Neurology, University of Massachusetts Chan Medical School, Worcester, MA, United States
| | - Shahid Banday
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA, United States
| | - Ching-Man Virbasius
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA, United States
| | - Miguel Sena-Esteves
- Department of Neurology, University of Massachusetts Chan Medical School, Worcester, MA, United States
| | - Jonathan K. Watts
- RNA Therapeutics Institute, University of Massachusetts Chan Medical School, Worcester, MA, United States
| | - Michael R. Green
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA, United States
| |
Collapse
|
12
|
Malachowski T, Chandradoss KR, Boya R, Zhou L, Cook AL, Su C, Pham K, Haws SA, Kim JH, Ryu HS, Ge C, Luppino JM, Nguyen SC, Titus KR, Gong W, Wallace O, Joyce EF, Wu H, Rojas LA, Phillips-Cremins JE. Spatially coordinated heterochromatinization of long synaptic genes in fragile X syndrome. Cell 2023; 186:5840-5858.e36. [PMID: 38134876 PMCID: PMC10794044 DOI: 10.1016/j.cell.2023.11.019] [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: 12/03/2022] [Revised: 07/31/2023] [Accepted: 11/16/2023] [Indexed: 12/24/2023]
Abstract
Short tandem repeat (STR) instability causes transcriptional silencing in several repeat expansion disorders. In fragile X syndrome (FXS), mutation-length expansion of a CGG STR represses FMR1 via local DNA methylation. Here, we find megabase-scale H3K9me3 domains on autosomes and encompassing FMR1 on the X chromosome in FXS patient-derived iPSCs, iPSC-derived neural progenitors, EBV-transformed lymphoblasts, and brain tissue with mutation-length CGG expansion. H3K9me3 domains connect via inter-chromosomal interactions and demarcate severe misfolding of TADs and loops. They harbor long synaptic genes replicating at the end of S phase, replication-stress-induced double-strand breaks, and STRs prone to stepwise somatic instability. CRISPR engineering of the mutation-length CGG to premutation length reverses H3K9me3 on the X chromosome and multiple autosomes, refolds TADs, and restores gene expression. H3K9me3 domains can also arise in normal-length iPSCs created with perturbations linked to genome instability, suggesting their relevance beyond FXS. Our results reveal Mb-scale heterochromatinization and trans interactions among loci susceptible to instability.
Collapse
Affiliation(s)
- Thomas Malachowski
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA; Epigenetics Institute, University of Pennsylvania, Philadelphia, PA, USA; Department of Genetics, University of Pennsylvania, Philadelphia, PA, USA
| | - Keerthivasan Raanin Chandradoss
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA; Epigenetics Institute, University of Pennsylvania, Philadelphia, PA, USA; Department of Genetics, University of Pennsylvania, Philadelphia, PA, USA
| | - Ravi Boya
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA; Epigenetics Institute, University of Pennsylvania, Philadelphia, PA, USA; Department of Genetics, University of Pennsylvania, Philadelphia, PA, USA
| | - Linda Zhou
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA; Epigenetics Institute, University of Pennsylvania, Philadelphia, PA, USA; Department of Genetics, University of Pennsylvania, Philadelphia, PA, USA
| | - Ashley L Cook
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA; Epigenetics Institute, University of Pennsylvania, Philadelphia, PA, USA; Department of Genetics, University of Pennsylvania, Philadelphia, PA, USA
| | - Chuanbin Su
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA; Epigenetics Institute, University of Pennsylvania, Philadelphia, PA, USA; Department of Genetics, University of Pennsylvania, Philadelphia, PA, USA
| | - Kenneth Pham
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA; Department of Genetics, University of Pennsylvania, Philadelphia, PA, USA
| | - Spencer A Haws
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA; Epigenetics Institute, University of Pennsylvania, Philadelphia, PA, USA; Department of Genetics, University of Pennsylvania, Philadelphia, PA, USA
| | - Ji Hun Kim
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Han-Seul Ryu
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA; Department of Genetics, University of Pennsylvania, Philadelphia, PA, USA
| | - Chunmin Ge
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Jennifer M Luppino
- Epigenetics Institute, University of Pennsylvania, Philadelphia, PA, USA; Department of Genetics, University of Pennsylvania, Philadelphia, PA, USA
| | - Son C Nguyen
- Epigenetics Institute, University of Pennsylvania, Philadelphia, PA, USA; Department of Genetics, University of Pennsylvania, Philadelphia, PA, USA
| | - Katelyn R Titus
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA; Epigenetics Institute, University of Pennsylvania, Philadelphia, PA, USA; Department of Genetics, University of Pennsylvania, Philadelphia, PA, USA
| | - Wanfeng Gong
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA; Epigenetics Institute, University of Pennsylvania, Philadelphia, PA, USA; Department of Genetics, University of Pennsylvania, Philadelphia, PA, USA
| | - Owen Wallace
- Fulcrum Therapeutics Incorporated, Cambridge, MA, USA
| | - Eric F Joyce
- Epigenetics Institute, University of Pennsylvania, Philadelphia, PA, USA; Department of Genetics, University of Pennsylvania, Philadelphia, PA, USA
| | - Hao Wu
- Fulcrum Therapeutics Incorporated, Cambridge, MA, USA
| | | | - Jennifer E Phillips-Cremins
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA; Epigenetics Institute, University of Pennsylvania, Philadelphia, PA, USA; Department of Genetics, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
13
|
Ziemka-Nalecz M, Pawelec P, Ziabska K, Zalewska T. Sex Differences in Brain Disorders. Int J Mol Sci 2023; 24:14571. [PMID: 37834018 PMCID: PMC10572175 DOI: 10.3390/ijms241914571] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 09/20/2023] [Accepted: 09/22/2023] [Indexed: 10/15/2023] Open
Abstract
A remarkable feature of the brain is its sexual dimorphism. Sexual dimorphism in brain structure and function is associated with clinical implications documented previously in healthy individuals but also in those who suffer from various brain disorders. Sex-based differences concerning some features such as the risk, prevalence, age of onset, and symptomatology have been confirmed in a range of neurological and neuropsychiatric diseases. The mechanisms responsible for the establishment of sex-based differences between men and women are not fully understood. The present paper provides up-to-date data on sex-related dissimilarities observed in brain disorders and highlights the most relevant features that differ between males and females. The topic is very important as the recognition of disparities between the sexes might allow for the identification of therapeutic targets and pharmacological approaches for intractable neurological and neuropsychiatric disorders.
Collapse
Affiliation(s)
| | | | | | - Teresa Zalewska
- NeuroRepair Department, Mossakowski Medical Research Institute, Polish Academy of Sciences, 5, A. Pawinskiego Str., 02-106 Warsaw, Poland; (M.Z.-N.); (P.P.); (K.Z.)
| |
Collapse
|
14
|
Valenti D, Vacca RA. Brain Mitochondrial Bioenergetics in Genetic Neurodevelopmental Disorders: Focus on Down, Rett and Fragile X Syndromes. Int J Mol Sci 2023; 24:12488. [PMID: 37569863 PMCID: PMC10419900 DOI: 10.3390/ijms241512488] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 07/31/2023] [Accepted: 08/04/2023] [Indexed: 08/13/2023] Open
Abstract
Mitochondria, far beyond their prominent role as cellular powerhouses, are complex cellular organelles active as central metabolic hubs that are capable of integrating and controlling several signaling pathways essential for neurological processes, including neurogenesis and neuroplasticity. On the other hand, mitochondria are themselves regulated from a series of signaling proteins to achieve the best efficiency in producing energy, in establishing a network and in performing their own de novo synthesis or clearance. Dysfunctions in signaling processes that control mitochondrial biogenesis, dynamics and bioenergetics are increasingly associated with impairment in brain development and involved in a wide variety of neurodevelopmental disorders. Here, we review recent evidence proving the emerging role of mitochondria as master regulators of brain bioenergetics, highlighting their control skills in brain neurodevelopment and cognition. We analyze, from a mechanistic point of view, mitochondrial bioenergetic dysfunction as causally interrelated to the origins of typical genetic intellectual disability-related neurodevelopmental disorders, such as Down, Rett and Fragile X syndromes. Finally, we discuss whether mitochondria can become therapeutic targets to improve brain development and function from a holistic perspective.
Collapse
Affiliation(s)
- Daniela Valenti
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies (IBIOM), National Research Council (CNR), Via G. Amendola 122/O, 70126 Bari, Italy
| | - Rosa Anna Vacca
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies (IBIOM), National Research Council (CNR), Via G. Amendola 122/O, 70126 Bari, Italy
| |
Collapse
|
15
|
Elhawary NA, AlJahdali IA, Abumansour IS, Azher ZA, Falemban AH, Madani WM, Alosaimi W, Alghamdi G, Sindi IA. Phenotypic variability to medication management: an update on fragile X syndrome. Hum Genomics 2023; 17:60. [PMID: 37420260 PMCID: PMC10329374 DOI: 10.1186/s40246-023-00507-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 07/03/2023] [Indexed: 07/09/2023] Open
Abstract
This review discusses the discovery, epidemiology, pathophysiology, genetic etiology, molecular diagnosis, and medication-based management of fragile X syndrome (FXS). It also highlights the syndrome's variable expressivity and common comorbid and overlapping conditions. FXS is an X-linked dominant disorder associated with a wide spectrum of clinical features, including but not limited to intellectual disability, autism spectrum disorder, language deficits, macroorchidism, seizures, and anxiety. Its prevalence in the general population is approximately 1 in 5000-7000 men and 1 in 4000-6000 women worldwide. FXS is associated with the fragile X messenger ribonucleoprotein 1 (FMR1) gene located at locus Xq27.3 and encodes the fragile X messenger ribonucleoprotein (FMRP). Most individuals with FXS have an FMR1 allele with > 200 CGG repeats (full mutation) and hypermethylation of the CpG island proximal to the repeats, which silences the gene's promoter. Some individuals have mosaicism in the size of the CGG repeats or in hypermethylation of the CpG island, both produce some FMRP and give rise to milder cognitive and behavioral deficits than in non-mosaic individuals with FXS. As in several monogenic disorders, modifier genes influence the penetrance of FMR1 mutations and FXS's variable expressivity by regulating the pathophysiological mechanisms related to the syndrome's behavioral features. Although there is no cure for FXS, prenatal molecular diagnostic testing is recommended to facilitate early diagnosis. Pharmacologic agents can reduce some behavioral features of FXS, and researchers are investigating whether gene editing can be used to demethylate the FMR1 promoter region to improve patient outcomes. Moreover, clustered regularly interspaced palindromic repeats (CRISPR)/Cas9 and developed nuclease defective Cas9 (dCas9) strategies have promised options of genome editing in gain-of-function mutations to rewrite new genetic information into a specified DNA site, are also being studied.
Collapse
Affiliation(s)
- Nasser A. Elhawary
- Department of Medical Genetics, College of Medicine, Umm Al-Qura University, Mecca, 21955 Saudi Arabia
| | - Imad A. AlJahdali
- Department of Community Medicine, College of Medicine, Umm Al-Qura University, Mecca, Saudi Arabia
| | - Iman S. Abumansour
- Department of Medical Genetics, College of Medicine, Umm Al-Qura University, Mecca, 21955 Saudi Arabia
| | - Zohor A. Azher
- Department of Medical Genetics, College of Medicine, Umm Al-Qura University, Mecca, 21955 Saudi Arabia
| | - Alaa H. Falemban
- Department of Pharmacology and Toxicology, College of Medicine, Umm Al-Qura University, Mecca, 24382 Saudi Arabia
| | - Wefaq M. Madani
- Department of Hematology and Immunology, Faculty of Medicine, Umm Al-Qura University, Mecca, Saudi Arabia
| | - Wafaa Alosaimi
- Department of Hematology, Maternity and Children Hospital, Mecca, Saudi Arabia
| | - Ghydda Alghamdi
- Department of Medical Genetics, College of Medicine, Umm Al-Qura University, Mecca, 21955 Saudi Arabia
| | - Ikhlas A. Sindi
- Department of Biology, Faculty of Science, King Abdulaziz University, Jeddah, 21589 Saudi Arabia
- Preparatory Year Program, Batterjee Medical College, Jeddah, 21442 Saudi Arabia
| |
Collapse
|
16
|
Pedapati EV, Sweeney JA, Schmitt LM, Ethridge LE, Miyakoshi M, Liu R, Smith E, Shaffer RC, Wu SW, Gilbert DL, Horn PS, Erickson C. Empirical Frequency Bound Derivation Reveals Prominent Mid-Frontal Alpha Associated with Neurosensory Dysfunction in Fragile X Syndrome. RESEARCH SQUARE 2023:rs.3.rs-2855646. [PMID: 37162907 PMCID: PMC10168472 DOI: 10.21203/rs.3.rs-2855646/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
The FMR1 gene is inactive in Fragile X syndrome (FXS), resulting in low levels of FMRP and consequent neurochemical, synaptic, and local circuit neurophysiological alterations in the fmr1 KO mouse. In FXS patients, electrophysiological studies have demonstrated a marked reduction in global alpha activity and regional increases in gamma oscillations associated with intellectual disability and sensory hypersensitivity. Since alpha activity is associated with a thalamocortical function with widely distributed modulatory effects on neocortical excitability, insight into alpha physiology may provide insight into systems-level disease mechanisms. Herein, we took a data-driven approach to clarify the temporal and spatial properties of alpha and theta activity in participants with FXS. High-resolution resting-state EEG data were collected from participants affected by FXS (n = 65) and matched controls (n = 70). We used a multivariate technique to empirically classify neural oscillatory bands based on their coherent spatiotemporal patterns. Participants with FXS demonstrated: 1) redistribution of lower-frequency boundaries indicating a "slower" dominant alpha rhythm, 2) an anteriorization of alpha frequency activity, and 3) a correlation of increased individualized alpha power measurements with auditory neurosensory dysfunction. These findings suggest an important role for alterations in thalamocortical physiology for the well-established neocortical hyper-excitability in FXS and, thus, a role for neural systems level disruption to cortical hyperexcitability that has been studied primarily at the local circuit level in animal models.
Collapse
Affiliation(s)
| | | | | | | | | | - Rui Liu
- Cincinnati Children's Hospital Medical Center
| | | | | | - Steve W Wu
- Cincinnati Children's Hospital Medical Center
| | | | - Paul S Horn
- Cincinnati Children's Hospital Medical Center
| | | |
Collapse
|
17
|
Veenstra-VanderWeele J, O'Reilly KC, Dennis MY, Uribe-Salazar JM, Amaral DG. Translational Neuroscience Approaches to Understanding Autism. Am J Psychiatry 2023; 180:265-276. [PMID: 37002692 DOI: 10.1176/appi.ajp.20230153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/04/2023]
Abstract
While autism spectrum disorder affects nearly 2% of children in the United States, little is known with certainty concerning the etiologies and brain systems involved. This is due, in part, to the substantial heterogeneity in the presentation of the core symptoms of autism as well as the great number of co-occurring conditions that are common in autistic individuals. Understanding the neurobiology of autism is further hampered by the limited availability of postmortem brain tissue to determine the cellular and molecular alterations that take place in the autistic brain. Animal models therefore provide great translational value in helping to define the neural systems that constitute the social brain and mediate repetitive behaviors or interests. If they are based on genetic or environmental factors that contribute to autism, organisms from flies to nonhuman primates may serve as models of the neural structure or function of the autistic brain. Ultimately, successful models can also be employed to test the safety and effectiveness of potential therapeutics. This is an overview of the major animal species that are currently used as models of autism, including an appraisal of the advantages and limitations of each.
Collapse
Affiliation(s)
- Jeremy Veenstra-VanderWeele
- New York State Psychiatric Institute and Department of Psychiatry, Columbia University, New York (Veenstra-VanderWeele, O'Reilly); Department of Biochemistry and Molecular Medicine, Genome Center (Dennis, Uribe-Salazar), MIND Institute (Dennis, Uribe-Salazar, Amaral), and Department of Psychiatry and Behavioral Sciences (Amaral), University of California, Davis
| | - Kally C O'Reilly
- New York State Psychiatric Institute and Department of Psychiatry, Columbia University, New York (Veenstra-VanderWeele, O'Reilly); Department of Biochemistry and Molecular Medicine, Genome Center (Dennis, Uribe-Salazar), MIND Institute (Dennis, Uribe-Salazar, Amaral), and Department of Psychiatry and Behavioral Sciences (Amaral), University of California, Davis
| | - Megan Y Dennis
- New York State Psychiatric Institute and Department of Psychiatry, Columbia University, New York (Veenstra-VanderWeele, O'Reilly); Department of Biochemistry and Molecular Medicine, Genome Center (Dennis, Uribe-Salazar), MIND Institute (Dennis, Uribe-Salazar, Amaral), and Department of Psychiatry and Behavioral Sciences (Amaral), University of California, Davis
| | - José M Uribe-Salazar
- New York State Psychiatric Institute and Department of Psychiatry, Columbia University, New York (Veenstra-VanderWeele, O'Reilly); Department of Biochemistry and Molecular Medicine, Genome Center (Dennis, Uribe-Salazar), MIND Institute (Dennis, Uribe-Salazar, Amaral), and Department of Psychiatry and Behavioral Sciences (Amaral), University of California, Davis
| | - David G Amaral
- New York State Psychiatric Institute and Department of Psychiatry, Columbia University, New York (Veenstra-VanderWeele, O'Reilly); Department of Biochemistry and Molecular Medicine, Genome Center (Dennis, Uribe-Salazar), MIND Institute (Dennis, Uribe-Salazar, Amaral), and Department of Psychiatry and Behavioral Sciences (Amaral), University of California, Davis
| |
Collapse
|
18
|
Luo L, Yang L, Zhang K, Zhou SM, Wang Y, Yang LK, Feng B, Liu SB, Wu YM, Zhao MG, Yang Q. Caveolin-1-Mediated Cholesterol Accumulation Contributes to Exaggerated mGluR-Dependent Long-Term Depression and Impaired Cognition in Fmr1 Knockout Mice. Mol Neurobiol 2023; 60:3379-3395. [PMID: 36854997 PMCID: PMC10122623 DOI: 10.1007/s12035-023-03269-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 02/08/2023] [Indexed: 03/02/2023]
Abstract
Fragile X syndrome (FXS) is one of the most common inherited mental retardation diseases and is caused by the loss of fragile X mental retardation protein (FMRP) expression. The metabotropic glutamate receptor (mGluR) theory of FXS states that enhanced mGluR-dependent long-term depression (LTD) due to FMRP loss is involved in aberrant synaptic plasticity and autistic-like behaviors, but little is known about the underlying molecular mechanism. Here, we found that only hippocampal mGluR-LTD was exaggerated in adolescent Fmr1 KO mice, while N-methyl-D-aspartate receptor (NMDAR)-LTD was intact in mice of all ages. This development-dependent alteration was related to the differential expression of caveolin-1 (Cav1), which is essential for caveolae formation. Knockdown of Cav1 restored the enhanced mGluR-LTD in Fmr1 KO mice. Moreover, hippocampal Cav1 expression in Fmr1 KO mice induced excessive endocytosis of the α-amino-3-hydroxyl-5-methyl-4-isoxazolepropionate (AMPA) receptor subunit GluA2. This process relied on mGluR1/5 activation rather than NMDAR. Interference with Cav1 expression reversed these changes. Furthermore, massive cholesterol accumulation contributed to redundant caveolae formation, which provided the platform for mGluR-triggered Cav1 coupling to GluA2. Importantly, injection of the cholesterol scavenger methyl-β-cyclodextrin (Mβ-CD) recovered AMPA receptor trafficking and markedly alleviated hyperactivity, hippocampus-dependent fear memory, and spatial memory defects in Fmr1 KO mice. Together, our findings elucidate the important role of Cav1 in mediating mGluR-LTD enhancement and further inducing AMPA receptor endocytosis and suggest that cholesterol depletion by Mβ-CD during caveolae formation may be a novel and safe strategy to treat FXS.
Collapse
Affiliation(s)
- Li Luo
- Precision Pharmacy & Drug Development Center, Department of Pharmacy, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, China
| | - Le Yang
- Precision Pharmacy & Drug Development Center, Department of Pharmacy, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, China
| | - Kun Zhang
- Precision Pharmacy & Drug Development Center, Department of Pharmacy, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, China
| | - Shi-Meng Zhou
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, 710032, China
| | - Yan Wang
- Department of Gastroenterology and Endoscopy Center, Fourth Military Medical University, No.986 Hospital, Xi'an, 710054, China
| | - Liu-Kun Yang
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, 710032, China
| | - Bin Feng
- State Key Laboratory of Military Stomatology, Department of Pharmacy, School of Stomatology, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Fourth Military Medical University, Xi'an, 710054, China
| | - Shui-Bing Liu
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, 710032, China
| | - Yu-Mei Wu
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, 710032, China
| | - Ming-Gao Zhao
- Precision Pharmacy & Drug Development Center, Department of Pharmacy, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, China
| | - Qi Yang
- Precision Pharmacy & Drug Development Center, Department of Pharmacy, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, China.
| |
Collapse
|
19
|
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
|
20
|
Krzisch MA, Wu H, Yuan B, Whitfield TW, Liu XS, Fu D, Garrett-Engele CM, Khalil AS, Lungjangwa T, Shih J, Chang AN, Warren S, Cacace A, Andrykovich KR, Rietjens RGJ, Wallace O, Sur M, Jain B, Jaenisch R. Fragile X Syndrome Patient-Derived Neurons Developing in the Mouse Brain Show FMR1-Dependent Phenotypes. Biol Psychiatry 2023; 93:71-81. [PMID: 36372569 DOI: 10.1016/j.biopsych.2022.08.020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 08/11/2022] [Accepted: 08/11/2022] [Indexed: 01/10/2023]
Abstract
BACKGROUND Fragile X syndrome (FXS) is characterized by physical abnormalities, anxiety, intellectual disability, hyperactivity, autistic behaviors, and seizures. Abnormal neuronal development in FXS is poorly understood. Data on patients with FXS remain scarce, and FXS animal models have failed to yield successful therapies. In vitro models do not fully recapitulate the morphology and function of human neurons. METHODS To mimic human neuron development in vivo, we coinjected neural precursor cells derived from FXS patient-derived induced pluripotent stem cells and neural precursor cells derived from corrected isogenic control induced pluripotent stem cells into the brain of neonatal immune-deprived mice. RESULTS The transplanted cells populated the brain and a proportion differentiated into neurons and glial cells. Immunofluorescence and single and bulk RNA sequencing analyses showed accelerated maturation of FXS neurons after an initial delay. Additionally, we found increased percentages of Arc- and Egr-1-positive FXS neurons and wider dendritic protrusions of mature FXS striatal medium spiny neurons. CONCLUSIONS This transplantation approach provides new insights into the alterations of neuronal development in FXS by facilitating physiological development of cells in a 3-dimensional context.
Collapse
Affiliation(s)
- Marine A Krzisch
- Whitehead Institute for Biomedical Research, Cambridge, Massachusetts.
| | - Hao Wu
- Full Circles Therapeutics, Inc., Cambridge, Massachusetts
| | - Bingbing Yuan
- Whitehead Institute for Biomedical Research, Cambridge, Massachusetts
| | - Troy W Whitfield
- Whitehead Institute for Biomedical Research, Cambridge, Massachusetts
| | - X Shawn Liu
- Department of Physiology and Cellular Biophysics, Columbia University Medical Center, New York, New York
| | - Dongdong Fu
- Whitehead Institute for Biomedical Research, Cambridge, Massachusetts
| | | | - Andrew S Khalil
- Whitehead Institute for Biomedical Research, Cambridge, Massachusetts
| | - Tenzin Lungjangwa
- Whitehead Institute for Biomedical Research, Cambridge, Massachusetts
| | - Jennifer Shih
- Picower Institute for Learning and Memory, Cambridge, Massachusetts
| | | | - Stephen Warren
- Departments of Human Genetics, Biochemistry, and Pediatrics, Emory University School of Medicine, Atlanta, Georgia
| | | | | | | | | | - Mriganka Sur
- Picower Institute for Learning and Memory, Cambridge, Massachusetts
| | - Bhav Jain
- Whitehead Institute for Biomedical Research, Cambridge, Massachusetts
| | - Rudolf Jaenisch
- Whitehead Institute for Biomedical Research, Cambridge, Massachusetts; Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts.
| |
Collapse
|
21
|
Trajković J, Makevic V, Pesic M, Pavković-Lučić S, Milojevic S, Cvjetkovic S, Hagerman R, Budimirovic DB, Protic D. Drosophila melanogaster as a Model to Study Fragile X-Associated Disorders. Genes (Basel) 2022; 14:genes14010087. [PMID: 36672829 PMCID: PMC9859539 DOI: 10.3390/genes14010087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 12/22/2022] [Accepted: 12/23/2022] [Indexed: 12/30/2022] Open
Abstract
Fragile X syndrome (FXS) is a global neurodevelopmental disorder caused by the expansion of CGG trinucleotide repeats (≥200) in the Fragile X Messenger Ribonucleoprotein 1 (FMR1) gene. FXS is the hallmark of Fragile X-associated disorders (FXD) and the most common monogenic cause of inherited intellectual disability and autism spectrum disorder. There are several animal models used to study FXS. In the FXS model of Drosophila, the only ortholog of FMR1, dfmr1, is mutated so that its protein is missing. This model has several relevant phenotypes, including defects in the circadian output pathway, sleep problems, memory deficits in the conditioned courtship and olfactory conditioning paradigms, deficits in social interaction, and deficits in neuronal development. In addition to FXS, a model of another FXD, Fragile X-associated tremor/ataxia syndrome (FXTAS), has also been established in Drosophila. This review summarizes many years of research on FXD in Drosophila models.
Collapse
Affiliation(s)
- Jelena Trajković
- Faculty of Biology, University of Belgrade, 11000 Belgrade, Serbia
| | - Vedrana Makevic
- Department of Pathophysiology, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| | - Milica Pesic
- Institute of Human Genetics, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| | | | - Sara Milojevic
- Department of Pharmacology, Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| | - Smiljana Cvjetkovic
- Department of Humanities, Faculty of Medicine, University of Belgrade, 11000 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, 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
| | - Dragana Protic
- Department of Pharmacology, Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
- Correspondence:
| |
Collapse
|
22
|
Asiminas A, Booker SA, Dando OR, Kozic Z, Arkell D, Inkpen FH, Sumera A, Akyel I, Kind PC, Wood ER. Experience-dependent changes in hippocampal spatial activity and hippocampal circuit function are disrupted in a rat model of Fragile X Syndrome. Mol Autism 2022; 13:49. [PMID: 36536454 PMCID: PMC9764562 DOI: 10.1186/s13229-022-00528-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 12/01/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Fragile X syndrome (FXS) is a common single gene cause of intellectual disability and autism spectrum disorder. Cognitive inflexibility is one of the hallmarks of FXS with affected individuals showing extreme difficulty adapting to novel or complex situations. To explore the neural correlates of this cognitive inflexibility, we used a rat model of FXS (Fmr1-/y). METHODS We recorded from the CA1 in Fmr1-/y and WT littermates over six 10-min exploration sessions in a novel environment-three sessions per day (ITI 10 min). Our recordings yielded 288 and 246 putative pyramidal cells from 7 WT and 7 Fmr1-/y rats, respectively. RESULTS On the first day of exploration of a novel environment, the firing rate and spatial tuning of CA1 pyramidal neurons was similar between wild-type (WT) and Fmr1-/y rats. However, while CA1 pyramidal neurons from WT rats showed experience-dependent changes in firing and spatial tuning between the first and second day of exposure to the environment, these changes were decreased or absent in CA1 neurons of Fmr1-/y rats. These findings were consistent with increased excitability of Fmr1-/y CA1 neurons in ex vivo hippocampal slices, which correlated with reduced synaptic inputs from the medial entorhinal cortex. Lastly, activity patterns of CA1 pyramidal neurons were dis-coordinated with respect to hippocampal oscillatory activity in Fmr1-/y rats. LIMITATIONS It is still unclear how the observed circuit function abnormalities give rise to behavioural deficits in Fmr1-/y rats. Future experiments will focus on this connection as well as the contribution of other neuronal cell types in the hippocampal circuit pathophysiology associated with the loss of FMRP. It would also be interesting to see if hippocampal circuit deficits converge with those seen in other rodent models of intellectual disability. CONCLUSIONS In conclusion, we found that hippocampal place cells from Fmr1-/y rats show similar spatial firing properties as those from WT rats but do not show the same experience-dependent increase in spatial specificity or the experience-dependent changes in network coordination. Our findings offer support to a network-level origin of cognitive deficits in FXS.
Collapse
Affiliation(s)
- Antonis Asiminas
- grid.4305.20000 0004 1936 7988Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, EH8 9XD UK ,grid.4305.20000 0004 1936 7988Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh, EH8 9XD UK ,grid.4305.20000 0004 1936 7988Patrick Wild Centre, University of Edinburgh, Edinburgh, EH8 9XD UK ,grid.5254.60000 0001 0674 042XPresent Address: Center for Translational Neuromedicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Sam A. Booker
- grid.4305.20000 0004 1936 7988Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, EH8 9XD UK ,grid.4305.20000 0004 1936 7988Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh, EH8 9XD UK ,grid.4305.20000 0004 1936 7988Patrick Wild Centre, University of Edinburgh, Edinburgh, EH8 9XD UK
| | - Owen R. Dando
- grid.4305.20000 0004 1936 7988Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, EH8 9XD UK ,grid.4305.20000 0004 1936 7988Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh, EH8 9XD UK ,grid.4305.20000 0004 1936 7988Patrick Wild Centre, University of Edinburgh, Edinburgh, EH8 9XD UK ,grid.4305.20000 0004 1936 7988UK Dementia Research Institute at the Edinburgh Medical School, University of Edinburgh, Edinburgh, EH8 9XD UK
| | - Zrinko Kozic
- grid.4305.20000 0004 1936 7988Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, EH8 9XD UK ,grid.4305.20000 0004 1936 7988Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh, EH8 9XD UK
| | - Daisy Arkell
- grid.4305.20000 0004 1936 7988Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, EH8 9XD UK ,grid.4305.20000 0004 1936 7988Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh, EH8 9XD UK ,grid.4305.20000 0004 1936 7988Patrick Wild Centre, University of Edinburgh, Edinburgh, EH8 9XD UK
| | - Felicity H. Inkpen
- grid.4305.20000 0004 1936 7988Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, EH8 9XD UK ,grid.4305.20000 0004 1936 7988Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh, EH8 9XD UK ,grid.4305.20000 0004 1936 7988Patrick Wild Centre, University of Edinburgh, Edinburgh, EH8 9XD UK
| | - Anna Sumera
- grid.4305.20000 0004 1936 7988Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, EH8 9XD UK ,grid.4305.20000 0004 1936 7988Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh, EH8 9XD UK ,grid.4305.20000 0004 1936 7988Patrick Wild Centre, University of Edinburgh, Edinburgh, EH8 9XD UK
| | - Irem Akyel
- grid.4305.20000 0004 1936 7988Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, EH8 9XD UK ,grid.4305.20000 0004 1936 7988Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh, EH8 9XD UK
| | - Peter C. Kind
- grid.4305.20000 0004 1936 7988Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, EH8 9XD UK ,grid.4305.20000 0004 1936 7988Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh, EH8 9XD UK ,grid.4305.20000 0004 1936 7988Patrick Wild Centre, University of Edinburgh, Edinburgh, EH8 9XD UK ,Centre for Brain Development and Repair, Bangalore, 560065 India
| | - Emma R. Wood
- grid.4305.20000 0004 1936 7988Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, EH8 9XD UK ,grid.4305.20000 0004 1936 7988Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh, EH8 9XD UK ,grid.4305.20000 0004 1936 7988Patrick Wild Centre, University of Edinburgh, Edinburgh, EH8 9XD UK ,Centre for Brain Development and Repair, Bangalore, 560065 India
| |
Collapse
|
23
|
Bonini NM. A perspective on Drosophila genetics and its insight into human neurodegenerative disease. Front Mol Biosci 2022; 9:1060796. [PMID: 36518845 PMCID: PMC9743296 DOI: 10.3389/fmolb.2022.1060796] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 10/28/2022] [Indexed: 09/07/2023] Open
Abstract
Drosophila has been long appreciated as a classic genetic system for its ability to define gene function in vivo. Within the last several decades, the fly has also emerged as a premiere system for modeling and defining mechanisms of human disease by expressing dominant human disease genes and analyzing the effects. Here I discuss key aspects of this latter approach that first intrigued me to focus my laboratory research on this idea. Differences between the loss-of-function vs. the gain-of-function approach are raised-and the insight of these approaches for appreciating mechanisms that contribute to human neurodegenerative disease. The application of modifier genetics, which is a prominent goal of models of human disease, has implications for how specific genes or pathways intersect with the dominant disease-associated mechanisms. Models of human disease will continue to reveal unanticipated insight into fundamental cellular processes-insight that might be harder to glean from classical genetic methodologies vs modifier genetics of disease.
Collapse
Affiliation(s)
- Nancy M. Bonini
- Department of Biology, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
24
|
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
|
25
|
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
|
26
|
Kieffer F, Hilal F, Gay AS, Debayle D, Pronot M, Poupon G, Lacagne I, Bardoni B, Martin S, Gwizdek C. Combining affinity purification and mass spectrometry to define the network of the nuclear proteins interacting with the N-terminal region of FMRP. Front Mol Biosci 2022; 9:954087. [PMID: 36237573 PMCID: PMC9553004 DOI: 10.3389/fmolb.2022.954087] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 08/05/2022] [Indexed: 11/13/2022] Open
Abstract
Fragile X-Syndrome (FXS) represents the most common inherited form of intellectual disability and the leading monogenic cause of Autism Spectrum Disorders. In most cases, this disease results from the absence of expression of the protein FMRP encoded by the FMR1 gene (Fragile X messenger ribonucleoprotein 1). FMRP is mainly defined as a cytoplasmic RNA-binding protein regulating the local translation of thousands of target mRNAs. Interestingly, FMRP is also able to shuttle between the nucleus and the cytoplasm. However, to date, its roles in the nucleus of mammalian neurons are just emerging. To broaden our insight into the contribution of nuclear FMRP in mammalian neuronal physiology, we identified here a nuclear interactome of the protein by combining subcellular fractionation of rat forebrains with pull‐ down affinity purification and mass spectrometry analysis. By this approach, we listed 55 candidate nuclear partners. This interactome includes known nuclear FMRP-binding proteins as Adar or Rbm14 as well as several novel candidates, notably Ddx41, Poldip3, or Hnrnpa3 that we further validated by target‐specific approaches. Through our approach, we identified factors involved in different steps of mRNA biogenesis, as transcription, splicing, editing or nuclear export, revealing a potential central regulatory function of FMRP in the biogenesis of its target mRNAs. Therefore, our work considerably enlarges the nuclear proteins interaction network of FMRP in mammalian neurons and lays the basis for exciting future mechanistic studies deepening the roles of nuclear FMRP in neuronal physiology and the etiology of the FXS.
Collapse
Affiliation(s)
- Félicie Kieffer
- Université Côte d'Azur, Centre National de la Recherche Scientifique, Institut de Pharmacologie Moléculaire et Cellulaire, Valbonne, France
| | - Fahd Hilal
- Université Côte d'Azur, Centre National de la Recherche Scientifique, Institut de Pharmacologie Moléculaire et Cellulaire, Valbonne, France
| | - Anne-Sophie Gay
- Université Côte d'Azur, Centre National de la Recherche Scientifique, Institut de Pharmacologie Moléculaire et Cellulaire, Valbonne, France
| | - Delphine Debayle
- Université Côte d'Azur, Centre National de la Recherche Scientifique, Institut de Pharmacologie Moléculaire et Cellulaire, Valbonne, France
| | - Marie Pronot
- Université Côte d'Azur, Centre National de la Recherche Scientifique, Institut de Pharmacologie Moléculaire et Cellulaire, Valbonne, France
| | - Gwénola Poupon
- Université Côte d'Azur, Centre National de la Recherche Scientifique, Institut de Pharmacologie Moléculaire et Cellulaire, Valbonne, France
| | - Iliona Lacagne
- Université Côte d'Azur, Centre National de la Recherche Scientifique, Institut de Pharmacologie Moléculaire et Cellulaire, Valbonne, France
| | - Barbara Bardoni
- Université Côte d'Azur, Institut National de la Santé Et de la Recherche Médicale, Centre National de la Recherche Scientifique, Institut de Pharmacologie Moléculaire et Cellulaire, Valbonne, France
| | - Stéphane Martin
- Université Côte d'Azur, Institut National de la Santé Et de la Recherche Médicale, Centre National de la Recherche Scientifique, Institut de Pharmacologie Moléculaire et Cellulaire, Valbonne, France
| | - Carole Gwizdek
- Université Côte d'Azur, Centre National de la Recherche Scientifique, Institut de Pharmacologie Moléculaire et Cellulaire, Valbonne, France
- *Correspondence: Carole Gwizdek,
| |
Collapse
|
27
|
Kuznitsov-Yanovsky L, Shapira G, Gildin L, Shomron N, Ben-Yosef D. Transcriptomic Analysis of Human Fragile X Syndrome Neurons Reveals Neurite Outgrowth Modulation by the TGFβ/BMP Pathway. Int J Mol Sci 2022; 23:ijms23169278. [PMID: 36012539 PMCID: PMC9409179 DOI: 10.3390/ijms23169278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 08/11/2022] [Accepted: 08/14/2022] [Indexed: 11/25/2022] Open
Abstract
Fragile X Syndrome (FXS) is the main genetic reason for intellectual disability and is caused by the silencing of fragile X mental retardation protein (FMRP), an RNA-binding protein regulating the translation of many neuronal mRNAs. Neural differentiation of FX human embryonic stem cells (hESC) mimics the neurodevelopment of FXS fetuses and thus serves as a good model to explore the mechanisms underlining the development of FXS. Isogenic hESC clones with and without the FX mutation that share the same genetic background were in vitro differentiated into neurons, and their transcriptome was analyzed by RNA sequencing. FX neurons inactivating FMR1 expression presented delayed neuronal development and maturation, concomitant with dysregulation of the TGFβ/BMP signaling pathway, and genes related to the extracellular matrix. Migration assay showed decreased neurite outgrowth in FX neurons that was rescued by inhibition of the TGFβ/BMP signaling pathway. Our results provide new insights into the molecular pathway by which loss of FMRP affects neuronal network development. In FX neurons, the lack of FMRP dysregulates members of the BMP signaling pathway associated with ECM organization which, in a yet unknown mechanism, reduces the guidance of axonal growth cones, probably leading to the aberrant neuronal network function seen in FXS.
Collapse
Affiliation(s)
- Liron Kuznitsov-Yanovsky
- Wolfe PGD Stem Cell Lab, Racine IVF Unit, Lis Maternity Hospital Tel-Aviv Sourasky Medical Center, Tel Aviv 64239, Israel
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv 69978, Israel
| | - Guy Shapira
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv 69978, Israel
| | - Lital Gildin
- Wolfe PGD Stem Cell Lab, Racine IVF Unit, Lis Maternity Hospital Tel-Aviv Sourasky Medical Center, Tel Aviv 64239, Israel
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv 69978, Israel
| | - Noam Shomron
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv 69978, Israel
- Sagol School of Neuroscience, Tel-Aviv University, Tel Aviv 69978, Israel
| | - Dalit Ben-Yosef
- Wolfe PGD Stem Cell Lab, Racine IVF Unit, Lis Maternity Hospital Tel-Aviv Sourasky Medical Center, Tel Aviv 64239, Israel
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv 69978, Israel
- Sagol School of Neuroscience, Tel-Aviv University, Tel Aviv 69978, Israel
- Correspondence:
| |
Collapse
|
28
|
Yamazaki M, Arai T, Yarimizu J, Matsumoto M. 5-HT5A Receptor Antagonist ASP5736 Ameliorates Several Abnormal Behaviors in an Fmr1-Targeted Transgenic Male Rat Model of Fragile X Syndrome. Int J Neuropsychopharmacol 2022; 25:786-793. [PMID: 35882205 PMCID: PMC9515134 DOI: 10.1093/ijnp/pyac041] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 06/24/2022] [Accepted: 07/23/2022] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Fragile X syndrome (FXS) is a genetic condition that causes a range of developmental problems, including intellectual disability, aggressive behavior, anxiety, abnormal sensory processing, and cognitive impairment. Despite intensive preclinical research in Fmr1-targeted transgenic mice, an effective treatment for FXS has yet to be developed. We previously demonstrated that ASP5736, a 5-Hydroxytryptamine (serotonin) receptor 5A receptor antagonist, ameliorated scopolamine-induced working memory deficits in mice, reference memory impairment in aged rats, and methamphetamine-induced positive symptoms and phencyclidine-induced cognitive impairment in animal models of schizophrenia. We hypothesized that ASP5736 may be effective for ameliorating similar behavior deficits in male Fmr1-targeted transgenic rats as a preclinical model of FXS. METHODS We evaluated the effect of acute oral administration of ASP5736 on the abnormal behavior of hyperactivity (0.01, 0.1 mg/kg), prepulse inhibition (0.01, 0.03, 0.1 mg/kg), and the novel object recognition task (0.1 mg/kg) in Frmr1-knockout (KO) rats. RESULTS Fmr1-KO rats showed body weight gain, hyperactivity, abnormal sensory motor gating, and cognitive impairment. ASP5736 (0.1 mg/kg) reversed the hyperactivity and ameliorated the sensory motor gating deficits (0.03-0.1 mg/kg). ASP5736 (0.01 mg/kg) also improved cognitive impairment. CONCLUSIONS ASP5736 is a potential drug candidate for FXS. Further studies are needed to confirm its clinical efficacy.
Collapse
Affiliation(s)
- Mayako Yamazaki
- Correspondence: Mayako Yamazaki, PhD, Research Fellow, Department of Neuroscience, Drug Discovery Research, Astellas Pharma Inc., 21 Miyukigaoka, Tsukuba-shi, Ibaraki, 305-8585, Japan ()
| | - Takatomo Arai
- Department of Neuroscience, Drug Discovery Research, Astellas Pharma Inc., Tsukuba-shi, Ibaraki, Japan
| | - Junko Yarimizu
- Department of Neuroscience, Drug Discovery Research, Astellas Pharma Inc., Tsukuba-shi, Ibaraki, Japan
| | - Mitsuyuki Matsumoto
- Department of Neuroscience, Drug Discovery Research, Astellas Pharma Inc., Tsukuba-shi, Ibaraki, Japan,Neuroscience, La Jolla Laboratory, Astellas Research Institute of America LLC, San Diego, CA, USA
| |
Collapse
|
29
|
Deficiency in FTSJ1 Affects Neuronal Plasticity in the Hippocampal Formation of Mice. BIOLOGY 2022; 11:biology11071011. [PMID: 36101392 PMCID: PMC9312013 DOI: 10.3390/biology11071011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 07/01/2022] [Accepted: 07/03/2022] [Indexed: 11/16/2022]
Abstract
Simple Summary Neuronal plasticity refers to the brain’s ability to adapt in response to activity-dependent changes. This process, among others, allows the brain to acquire memory or to compensate for a neurocognitive deficit. We analyzed adult FTSJ1-deficient mice in order to gain insight into the role of FTSJ1 in neuronal plasticity. These mice displayed alterations in the hippocampus (a brain structure that is involved in memory and learning, among other functions) e.g., in the form of changes in dendritic spines. Changes in dendritic spines are considered to represent a morphological hallmark of altered neuronal plasticity, and thus FTSJ1 deficiency might have a direct effect upon the capacity of the brain to adapt to plastic changes. Long-term potentiation (LTP) is an electrophysiological correlate of neuronal plasticity, and is related to learning and to processes attributed to memory. Here we show that LTP in FTSJ1-deficient mice is reduced, hinting at disturbed neuronal plasticity. These findings suggest that FTSJ1 deficiency has an impact on neuronal plasticity not only morphologically but also on the physiological level. Abstract The role of the tRNA methyltransferase FTSJ1 in the brain is largely unknown. We analyzed whether FTSJ1-deficient mice (KO) displayed altered neuronal plasticity. We explored open field behavior (10 KO mice (aged 22–25 weeks)) and 11 age-matched control littermates (WT) and examined mean layer thickness (7 KO; 6 WT) and dendritic spines (5 KO; 5 WT) in the hippocampal area CA1 and the dentate gyrus. Furthermore, long-term potentiation (LTP) within area CA1 was investigated (5 KO; 5 WT), and mass spectrometry (MS) using CA1 tissue (2 each) was performed. Compared to controls, KO mice showed a significant reduction in the mean thickness of apical CA1 layers. Dendritic spine densities were also altered in KO mice. Stable LTP could be induced in the CA1 area of KO mice and remained stable at for at least 1 h, although at a lower level as compared to WTs, while MS data indicated differential abundance of several proteins, which play a role in neuronal plasticity. FTSJ1 has an impact on neuronal plasticity in the murine hippocampal area CA1 at the morphological and physiological levels, which, in conjunction with comparable changes in other cortical areas, might accumulate in disturbed learning and memory functions.
Collapse
|
30
|
Bülow P, Segal M, Bassell GJ. Mechanisms Driving the Emergence of Neuronal Hyperexcitability in Fragile X Syndrome. Int J Mol Sci 2022; 23:ijms23116315. [PMID: 35682993 PMCID: PMC9181819 DOI: 10.3390/ijms23116315] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 05/24/2022] [Accepted: 05/27/2022] [Indexed: 02/04/2023] Open
Abstract
Hyperexcitability is a shared neurophysiological phenotype across various genetic neurodevelopmental disorders, including Fragile X syndrome (FXS). Several patient symptoms are associated with hyperexcitability, but a puzzling feature is that their onset is often delayed until their second and third year of life. It remains unclear how and why hyperexcitability emerges in neurodevelopmental disorders. FXS is caused by the loss of FMRP, an RNA-binding protein which has many critical roles including protein synthesis-dependent and independent regulation of ion channels and receptors, as well as global regulation of protein synthesis. Here, we discussed recent literature uncovering novel mechanisms that may drive the progressive onset of hyperexcitability in the FXS brain. We discussed in detail how recent publications have highlighted defects in homeostatic plasticity, providing new insight on the FXS brain and suggest pharmacotherapeutic strategies in FXS and other neurodevelopmental disorders.
Collapse
Affiliation(s)
- Pernille Bülow
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA 30322, USA
- Correspondence: (P.B.); (G.J.B.)
| | - Menahem Segal
- Department of Brain Science, Weizmann Institute of Science, Rehovot 76100, Israel;
| | - Gary J. Bassell
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA 30322, USA
- Correspondence: (P.B.); (G.J.B.)
| |
Collapse
|
31
|
Lee A, Xu J, Wen Z, Jin P. Across Dimensions: Developing 2D and 3D Human iPSC-Based Models of Fragile X Syndrome. Cells 2022; 11:1725. [PMID: 35681419 PMCID: PMC9179297 DOI: 10.3390/cells11111725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 05/11/2022] [Accepted: 05/13/2022] [Indexed: 02/01/2023] Open
Abstract
Fragile X syndrome (FXS) is the most common inherited cause of intellectual disability and autism spectrum disorder. FXS is caused by a cytosine-guanine-guanine (CGG) trinucleotide repeat expansion in the untranslated region of the FMR1 gene leading to the functional loss of the gene's protein product FMRP. Various animal models of FXS have provided substantial knowledge about the disorder. However, critical limitations exist in replicating the pathophysiological mechanisms. Human induced pluripotent stem cells (hiPSCs) provide a unique means of studying the features and processes of both normal and abnormal human neurodevelopment in large sample quantities in a controlled setting. Human iPSC-based models of FXS have offered a better understanding of FXS pathophysiology specific to humans. This review summarizes studies that have used hiPSC-based two-dimensional cellular models of FXS to reproduce the pathology, examine altered gene expression and translation, determine the functions and targets of FMRP, characterize the neurodevelopmental phenotypes and electrophysiological features, and, finally, to reactivate FMR1. We also provide an overview of the most recent studies using three-dimensional human brain organoids of FXS and end with a discussion of current limitations and future directions for FXS research using hiPSCs.
Collapse
Affiliation(s)
- Azalea Lee
- Neuroscience Graduate Program, Emory University, Atlanta, GA 30322, USA;
- MD/PhD Program, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Jie Xu
- Genetics and Molecular Biology Graduate Program, Emory University, Atlanta, GA 30322, USA;
| | - Zhexing Wen
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA 30322, USA
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Peng Jin
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
| |
Collapse
|
32
|
Delprato A, Xiao E, Manoj D. Connecting DCX, COMT and FMR1 in social behavior and cognitive impairment. BEHAVIORAL AND BRAIN FUNCTIONS : BBF 2022; 18:7. [PMID: 35590332 PMCID: PMC9121553 DOI: 10.1186/s12993-022-00191-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 03/14/2022] [Indexed: 11/24/2022]
Abstract
Genetic variants of DCX, COMT and FMR1 have been linked to neurodevelopmental disorders related to intellectual disability and social behavior. In this systematic review we examine the roles of the DCX, COMT and FMR1 genes in the context of hippocampal neurogenesis with respect to these disorders with the aim of identifying important hubs and signaling pathways that may bridge these conditions. Taken together our findings indicate that factors connecting DCX, COMT, and FMR1 in intellectual disability and social behavior may converge at Wnt signaling, neuron migration, and axon and dendrite morphogenesis. Data derived from genomic research has identified a multitude of genes that are linked to brain disorders and developmental differences. Information about where and how these genes function and cooperate is lagging behind. The approach used here may help to shed light on the biological underpinnings in which key genes interface and may prove useful for the testing of specific hypotheses.
Collapse
Affiliation(s)
- Anna Delprato
- Department of Research and Education, BioScience Project, Wakefield, MA, 01880, USA.
| | - Emily Xiao
- Department of Research and Education, BioScience Project, Wakefield, MA, 01880, USA.,Alexander Mackenzie High School, Richmond Hill, ON, 14519, Canada
| | - Devika Manoj
- Department of Research and Education, BioScience Project, Wakefield, MA, 01880, USA.,Lambert High School, Suwanee, GA, 30024, USA
| |
Collapse
|
33
|
Curnow E, Wang Y. New Animal Models for Understanding FMRP Functions and FXS Pathology. Cells 2022; 11:1628. [PMID: 35626665 PMCID: PMC9140010 DOI: 10.3390/cells11101628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 05/03/2022] [Accepted: 05/09/2022] [Indexed: 11/16/2022] Open
Abstract
Fragile X encompasses a range of genetic conditions, all of which result as a function of changes within the FMR1 gene and abnormal production and/or expression of the FMR1 gene products. Individuals with Fragile X syndrome (FXS), the most common heritable form of intellectual disability, have a full-mutation sequence (>200 CGG repeats) which brings about transcriptional silencing of FMR1 and loss of FMR protein (FMRP). Despite considerable progress in our understanding of FXS, safe, effective, and reliable treatments that either prevent or reduce the severity of the FXS phenotype have not been approved. While current FXS animal models contribute their own unique understanding to the molecular, cellular, physiological, and behavioral deficits associated with FXS, no single animal model is able to fully recreate the FXS phenotype. This review will describe the status and rationale in the development, validation, and utility of three emerging animal model systems for FXS, namely the nonhuman primate (NHP), Mongolian gerbil, and chicken. These developing animal models will provide a sophisticated resource in which the deficits in complex functions of perception, action, and cognition in the human disorder are accurately reflected and aid in the successful translation of novel therapeutics and interventions to the clinic setting.
Collapse
Affiliation(s)
- Eliza Curnow
- REI Division, Department of ObGyn, University of Washington, Seattle, WA 98195, USA
- Washington National Primate Research Center, University of Washington, Seattle, WA 98195, USA
| | - Yuan Wang
- Program in Neuroscience, Department of Biomedical Sciences, College of Medicine, Florida State University, Tallahassee, FL 32306, USA
| |
Collapse
|
34
|
Afshar S, Lule S, Yuan G, Qu X, Pan C, Whalen M, Brownell AL, Mody M. Longitudinal PET studies of mGluR5 in FXS using an FMR1 knockout mouse model. Transl Neurosci 2022; 13:80-92. [PMID: 35582646 PMCID: PMC9055256 DOI: 10.1515/tnsci-2022-0217] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 03/28/2022] [Accepted: 04/04/2022] [Indexed: 11/15/2022] Open
Abstract
Abstract
Fragile X syndrome (FXS) is a monogenic disorder characterized by intellectual disability and behavioral challenges. It is caused by aberrant methylation of the fragile X mental retardation 1 (FMR1) gene. Given the failure of clinical trials in FXS and growing evidence of a role of metabotropic glutamate subtype 5 receptors (mGluR5) in the pathophysiology of the disorder, we investigated mGluR5 function in FMR1 Knockout (FMR1-KO) mice and age- and sex-matched control mice using longitudinal positron emission tomography (PET) imaging to better understand the disorder. The studies were repeated at four time points to examine age- and disease-induced changes in mGluR5 availability using 3-fluoro-[18F]5-(2-pyridinylethynyl)benzonitrile ([18F]FPEB). We found that the binding potential (BP) of [18F]FPEB was significantly lower in the KO mice in mGluR5-implicated brain areas including striatum, cortex, hippocampus, thalamus, and olfactory bulb. The BP also changed with age, regardless of disorder status, increasing in early adulthood in male but not in female mice before decreasing later in both sexes. The difference in mGluR5 availability between the FMR1-KO and control mice and the change in BP in the KO mice as a function of age and sex illustrate the nature of the disorder and its progression, providing mechanistic insights for treatment design.
Collapse
Affiliation(s)
- Sepideh Afshar
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School , Charlestown , 02129 MA , United States of America
| | - Sevda Lule
- Department of Pediatrics, Massachusetts General Hospital, Harvard Medical School , Charlestown , 02129 MA , United States of America
| | - Gengyang Yuan
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School , Charlestown , 02129 MA , United States of America
| | - Xiying Qu
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School , Charlestown , 02129 MA , United States of America
| | - Chuzhi Pan
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School , Charlestown , 02129 MA , United States of America
| | - Michael Whalen
- Department of Pediatrics, Massachusetts General Hospital, Harvard Medical School , Charlestown , 02129 MA , United States of America
| | - Anna-Liisa Brownell
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School , Charlestown , 02129 MA , United States of America
| | - Maria Mody
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School , Charlestown , 02129 MA , USA
| |
Collapse
|
35
|
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
|
36
|
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
|
37
|
Chawla A, McCullagh EA. Auditory Brain Stem Responses in the C57BL/6J Fragile X Syndrome-Knockout Mouse Model. Front Integr Neurosci 2022; 15:803483. [PMID: 35111002 PMCID: PMC8802689 DOI: 10.3389/fnint.2021.803483] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 12/14/2021] [Indexed: 01/07/2023] Open
Abstract
Sensory hypersensitivity, especially in the auditory system, is a common symptom in Fragile X syndrome (FXS), the most common monogenic form of intellectual disability. However, linking phenotypes across genetic background strains of mouse models has been a challenge and could underly some of the issues with translatability of drug studies to the human condition. This study is the first to characterize the auditory brain stem response (ABR), a minimally invasive physiological readout of early auditory processing that is also used in humans, in a commonly used mouse background strain model of FXS, C57BL/6J. We measured morphological features of pinna and head and used ABR to measure the hearing range, and monaural and binaural auditory responses in hemizygous males, homozygous females, and heterozygous females compared with those in wild-type mice. Consistent with previous study, we showed no difference in morphological parameters across genotypes or sexes. There was no significant difference in hearing range between the sexes or genotypes, however there was a trend towards high frequency hearing loss in male FXS mice. In contrast, female mice with homozygous FXS had a decreased amplitude of wave IV of the monaural ABR, while there was no difference in males for amplitudes and no change in latency of ABR waveforms across sexes and genotypes. Finally, males with FXS had an increased latency of the binaural interaction component (BIC) at 0 interaural timing difference compared with that in wild-type males. These findings further clarify auditory brain stem processing in FXS by adding more information across genetic background strains allowing for a better understanding of shared phenotypes.
Collapse
Affiliation(s)
| | - Elizabeth A. McCullagh
- Department of Integrative Biology, Oklahoma State University, Stillwater, OK, United States
| |
Collapse
|
38
|
Bach S, Shovlin S, Moriarty M, Bardoni B, Tropea D. Rett Syndrome and Fragile X Syndrome: Different Etiology With Common Molecular Dysfunctions. Front Cell Neurosci 2021; 15:764761. [PMID: 34867203 PMCID: PMC8640214 DOI: 10.3389/fncel.2021.764761] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 10/27/2021] [Indexed: 01/04/2023] Open
Abstract
Rett syndrome (RTT) and Fragile X syndrome (FXS) are two monogenetic neurodevelopmental disorders with complex clinical presentations. RTT is caused by mutations in the Methyl-CpG binding protein 2 gene (MECP2) altering the function of its protein product MeCP2. MeCP2 modulates gene expression by binding methylated CpG dinucleotides, and by interacting with transcription factors. FXS is caused by the silencing of the FMR1 gene encoding the Fragile X Mental Retardation Protein (FMRP), a RNA binding protein involved in multiple steps of RNA metabolism, and modulating the translation of thousands of proteins including a large set of synaptic proteins. Despite differences in genetic etiology, there are overlapping features in RTT and FXS, possibly due to interactions between MeCP2 and FMRP, and to the regulation of pathways resulting in dysregulation of common molecular signaling. Furthermore, basic physiological mechanisms are regulated by these proteins and might concur to the pathophysiology of both syndromes. Considering that RTT and FXS are disorders affecting brain development, and that most of the common targets of MeCP2 and FMRP are involved in brain activity, we discuss the mechanisms of synaptic function and plasticity altered in RTT and FXS, and we consider the similarities and the differences between these two disorders.
Collapse
Affiliation(s)
- Snow Bach
- School of Mathematical Sciences, Dublin City University, Dublin, Ireland.,Neuropsychiatric Genetics, Department of Psychiatry, School of Medicine, Trinity College Dublin, Trinity Translational Medicine Institute, St James's Hospital, Dublin, Ireland
| | - Stephen Shovlin
- Neuropsychiatric Genetics, Department of Psychiatry, School of Medicine, Trinity College Dublin, Trinity Translational Medicine Institute, St James's Hospital, Dublin, Ireland
| | | | - Barbara Bardoni
- Inserm, CNRS UMR 7275, Institute of Molecular and Cellular Pharmacology, Université Côte d'Azur, Valbonne, France
| | - Daniela Tropea
- Neuropsychiatric Genetics, Department of Psychiatry, School of Medicine, Trinity College Dublin, Trinity Translational Medicine Institute, St James's Hospital, Dublin, Ireland.,Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland.,FutureNeuro, The SFI Research Centre for Chronic and Rare Neurological Diseases, Dublin, Ireland
| |
Collapse
|
39
|
Lucas A, Poleg S, Klug A, McCullagh EA. Myelination Deficits in the Auditory Brainstem of a Mouse Model of Fragile X Syndrome. Front Neurosci 2021; 15:772943. [PMID: 34858133 PMCID: PMC8632548 DOI: 10.3389/fnins.2021.772943] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 10/21/2021] [Indexed: 11/13/2022] Open
Abstract
Auditory symptoms are one of the most frequent sensory issues described in people with Fragile X Syndrome (FXS), the most common genetic form of intellectual disability. However, the mechanisms that lead to these symptoms are under explored. In this study, we examined whether there are defects in myelination in the auditory brainstem circuitry. Specifically, we studied myelinated fibers that terminate in the Calyx of Held, which encode temporally precise sound arrival time, and are some of the most heavily myelinated axons in the brain. We measured anatomical myelination characteristics using coherent anti-stokes Raman spectroscopy (CARS) and electron microscopy (EM) in a FXS mouse model in the medial nucleus of the trapezoid body (MNTB) where the Calyx of Held synapses. We measured number of mature oligodendrocytes (OL) and oligodendrocyte precursor cells (OPCs) to determine if changes in myelination were due to changes in the number of myelinating or immature glial cells. The two microscopy techniques (EM and CARS) showed a decrease in fiber diameter in FXS mice. Additionally, EM results indicated reductions in myelin thickness and axon diameter, and an increase in g-ratio, a measure of structural and functional myelination. Lastly, we showed an increase in both OL and OPCs in MNTB sections of FXS mice suggesting that the myelination phenotype is not due to an overall decrease in number of myelinating OLs. This is the first study to show that a myelination defects in the auditory brainstem that may underly auditory phenotypes in FXS.
Collapse
Affiliation(s)
- Alexandra Lucas
- Department of Physiology and Biophysics, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Shani Poleg
- Department of Physiology and Biophysics, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Achim Klug
- Department of Physiology and Biophysics, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Elizabeth A McCullagh
- Department of Integrative Biology, Oklahoma State University, Stillwater, OK, United States
| |
Collapse
|
40
|
Kurosaki T, Sakano H, Pröschel C, Wheeler J, Hewko A, Maquat LE. NMD abnormalities during brain development in the Fmr1-knockout mouse model of fragile X syndrome. Genome Biol 2021; 22:317. [PMID: 34784943 PMCID: PMC8597091 DOI: 10.1186/s13059-021-02530-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 10/28/2021] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND Fragile X syndrome (FXS) is an intellectual disability attributable to loss of fragile X protein (FMRP). We previously demonstrated that FMRP binds mRNAs targeted for nonsense-mediated mRNA decay (NMD) and that FMRP loss results in hyperactivated NMD and inhibition of neuronal differentiation in human stem cells. RESULTS We show here that NMD is hyperactivated during the development of the cerebral cortex, hippocampus, and cerebellum in the Fmr1-knockout (KO) mouse during embryonic and early postnatal periods. Our findings demonstrate that NMD regulates many neuronal mRNAs that are important for mouse brain development. CONCLUSIONS We reveal the abnormal regulation of these mRNAs in the Fmr1-KO mouse, a model of FXS, and highlight the importance of early intervention.
Collapse
Affiliation(s)
- Tatsuaki Kurosaki
- Department of Biochemistry and Biophysics, School of Medicine and Dentistry, University of Rochester, Rochester, NY 14642 USA
- Center for RNA Biology, University of Rochester, Rochester, NY 14642 USA
| | - Hitomi Sakano
- Center for RNA Biology, University of Rochester, Rochester, NY 14642 USA
- Department of Otolaryngology, School of Medicine and Dentistry, University of Rochester, Rochester, NY 14642 USA
| | - Christoph Pröschel
- Department of Biomedical Genetics, School of Medicine and Dentistry, University of Rochester, Rochester, NY 14642 USA
- Stem Cell and Regenerative Medicine Institute, School of Medicine and Dentistry, University of Rochester, Rochester, NY 14642 USA
| | - Jason Wheeler
- Center for RNA Biology, University of Rochester, Rochester, NY 14642 USA
- Department of Otolaryngology, School of Medicine and Dentistry, University of Rochester, Rochester, NY 14642 USA
| | - Alexander Hewko
- Department of Biochemistry and Biophysics, School of Medicine and Dentistry, University of Rochester, Rochester, NY 14642 USA
- Center for RNA Biology, University of Rochester, Rochester, NY 14642 USA
| | - Lynne E. Maquat
- Department of Biochemistry and Biophysics, School of Medicine and Dentistry, University of Rochester, Rochester, NY 14642 USA
- Center for RNA Biology, University of Rochester, Rochester, NY 14642 USA
| |
Collapse
|
41
|
Hagerman RJ, Hagerman PJ. Fragile X Syndrome: Lessons Learned and What New Treatment Avenues Are on the Horizon. Annu Rev Pharmacol Toxicol 2021; 62:365-381. [PMID: 34499526 DOI: 10.1146/annurev-pharmtox-052120-090147] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Fragile X syndrome (FXS) is the most common form of inherited intellectual disability and the leading single-gene form of autism spectrum disorder, encompassing cognitive, behavioral, and physical forms of clinical involvement. FXS is caused by large expansions of a noncoding CGG repeat (>200 repeats) in the FMR1 gene, at which point the gene is generally silenced. Absence of FMR1 protein (FMRP), important for synaptic development and maintenance, gives rise to the neurodevelopmental disorder. There is, at present, no therapeutic approach that directly reverses the loss of FMRP; however, there is an increasing number of potential treatments that target the pathways dysregulated in FXS, including those that address the enhanced activity of the mGluR5 pathway and deficits in GABA pathways. Based on studies of targeted therapeutics to date, the prospects are good for one or more effective therapies for FXS in the near future. Expected final online publication date for the Annual Review of Pharmacology and Toxicology, Volume 62 is January 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Randi J Hagerman
- Department of Pediatrics, University of California, Davis, School of Medicine, Sacramento, California 95817, USA; .,MIND Institute, University of California Davis Health, Sacramento, California 95817, USA
| | - Paul J Hagerman
- MIND Institute, University of California Davis Health, Sacramento, California 95817, USA.,Department of Biochemistry and Molecular Medicine, University of California, Davis, School of Medicine, Davis, California 95616, USA;
| |
Collapse
|
42
|
Bicker F, Nardi L, Maier J, Vasic V, Schmeisser MJ. Criss-crossing autism spectrum disorder and adult neurogenesis. J Neurochem 2021; 159:452-478. [PMID: 34478569 DOI: 10.1111/jnc.15501] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 08/05/2021] [Accepted: 08/28/2021] [Indexed: 12/19/2022]
Abstract
Autism spectrum disorder (ASD) comprises a group of multifactorial neurodevelopmental disorders primarily characterized by deficits in social interaction and repetitive behavior. Although the onset is typically in early childhood, ASD poses a lifelong challenge for both patients and caretakers. Adult neurogenesis (AN) is the process by which new functional neurons are created from neural stem cells existing in the post-natal brain. The entire event is based on a sequence of cellular processes, such as proliferation, specification of cell fate, maturation, and ultimately, synaptic integration into the existing neural circuits. Hence, AN is implicated in structural and functional brain plasticity throughout life. Accumulating evidence shows that impaired AN may underlie some of the abnormal behavioral phenotypes seen in ASD. In this review, we approach the interconnections between the molecular pathways related to AN and ASD. We also discuss existing therapeutic approaches targeting such pathways both in preclinical and clinical studies. A deeper understanding of how ASD and AN reciprocally affect one another could reveal important converging pathways leading to the emergence of psychiatric disorders.
Collapse
Affiliation(s)
- Frank Bicker
- Institute for Microscopic Anatomy and Neurobiology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Leonardo Nardi
- Institute for Microscopic Anatomy and Neurobiology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Jannik Maier
- Institute for Microscopic Anatomy and Neurobiology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Verica Vasic
- Institute for Microscopic Anatomy and Neurobiology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Michael J Schmeisser
- Institute for Microscopic Anatomy and Neurobiology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany.,Focus Program Translational Neurosciences (FTN), University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| |
Collapse
|
43
|
Reyes ST, Deacon RMJ, Guo SG, Altimiras FJ, Castillo JB, van der Wildt B, Morales AP, Park JH, Klamer D, Rosenberg J, Oberman LM, Rebowe N, Sprouse J, Missling CU, McCurdy CR, Cogram P, Kaufmann WE, Chin FT. Effects of the sigma-1 receptor agonist blarcamesine in a murine model of fragile X syndrome: neurobehavioral phenotypes and receptor occupancy. Sci Rep 2021; 11:17150. [PMID: 34433831 PMCID: PMC8387417 DOI: 10.1038/s41598-021-94079-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Accepted: 06/21/2021] [Indexed: 11/08/2022] Open
Abstract
Fragile X syndrome (FXS), a disorder of synaptic development and function, is the most prevalent genetic form of intellectual disability and autism spectrum disorder. FXS mouse models display clinically-relevant phenotypes, such as increased anxiety and hyperactivity. Despite their availability, so far advances in drug development have not yielded new treatments. Therefore, testing novel drugs that can ameliorate FXS' cognitive and behavioral impairments is imperative. ANAVEX2-73 (blarcamesine) is a sigma-1 receptor (S1R) agonist with a strong safety record and preliminary efficacy evidence in patients with Alzheimer's disease and Rett syndrome, other synaptic neurodegenerative and neurodevelopmental disorders. S1R's role in calcium homeostasis and mitochondrial function, cellular functions related to synaptic function, makes blarcamesine a potential drug candidate for FXS. Administration of blarcamesine in 2-month-old FXS and wild type mice for 2 weeks led to normalization in two key neurobehavioral phenotypes: open field test (hyperactivity) and contextual fear conditioning (associative learning). Furthermore, there was improvement in marble-burying (anxiety, perseverative behavior). It also restored levels of BDNF, a converging point of many synaptic regulators, in the hippocampus. Positron emission tomography (PET) and ex vivo autoradiographic studies, using the highly selective S1R PET ligand [18F]FTC-146, demonstrated the drug's dose-dependent receptor occupancy. Subsequent analyses also showed a wide but variable brain regional distribution of S1Rs, which was preserved in FXS mice. Altogether, these neurobehavioral, biochemical, and imaging data demonstrates doses that yield measurable receptor occupancy are effective for improving the synaptic and behavioral phenotype in FXS mice. The present findings support the viability of S1R as a therapeutic target in FXS, and the clinical potential of blarcamesine in FXS and other neurodevelopmental disorders.
Collapse
Affiliation(s)
- Samantha T Reyes
- Department of Radiology, Stanford University, Stanford, CA, 94305, USA
| | - Robert M J Deacon
- FRAXA-DVI, FRAXA, Santiago, Chile
- IEB, Faculty of Science, University of Chile, Santiago, Chile
- Fraunhofer Chile Research, Center for Systems Biotechnology, Santiago, Chile
| | - Scarlett G Guo
- Department of Radiology, Stanford University, Stanford, CA, 94305, USA
| | - Francisco J Altimiras
- FRAXA-DVI, FRAXA, Santiago, Chile
- Faculty of Engineering and Business, Universidad de las Américas, Santiago, Chile
| | - Jessa B Castillo
- Department of Radiology, Stanford University, Stanford, CA, 94305, USA
| | | | - Aimara P Morales
- Department of Radiology, Stanford University, Stanford, CA, 94305, USA
| | - Jun Hyung Park
- Department of Radiology, Stanford University, Stanford, CA, 94305, USA
| | - Daniel Klamer
- Anavex Life Sciences Corp., New York, NY, 10019, USA
| | - Jarrett Rosenberg
- Department of Radiology, Stanford University, Stanford, CA, 94305, USA
| | - Lindsay M Oberman
- Center for Neuroscience & Regenerative Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Nell Rebowe
- Anavex Life Sciences Corp., New York, NY, 10019, USA
| | | | | | - Christopher R McCurdy
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, FL, 32610, USA
| | - Patricia Cogram
- FRAXA-DVI, FRAXA, Santiago, Chile
- IEB, Faculty of Science, University of Chile, Santiago, Chile
- Fraunhofer Chile Research, Center for Systems Biotechnology, Santiago, Chile
| | - Walter E Kaufmann
- Anavex Life Sciences Corp., New York, NY, 10019, USA.
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, 30322, USA.
| | - Frederick T Chin
- Department of Radiology, Stanford University, Stanford, CA, 94305, USA.
| |
Collapse
|
44
|
Transcriptome programs involved in the development and structure of the cerebellum. Cell Mol Life Sci 2021; 78:6431-6451. [PMID: 34406416 PMCID: PMC8558292 DOI: 10.1007/s00018-021-03911-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 08/02/2021] [Indexed: 12/23/2022]
Abstract
In the past two decades, mounting evidence has modified the classical view of the cerebellum as a brain region specifically involved in the modulation of motor functions. Indeed, clinical studies and engineered mouse models have highlighted cerebellar circuits implicated in cognitive functions and behavior. Furthermore, it is now clear that insults occurring in specific time windows of cerebellar development can affect cognitive performance later in life and are associated with neurological syndromes, such as Autism Spectrum Disorder. Despite its almost homogenous cytoarchitecture, how cerebellar circuits form and function is not completely elucidated yet. Notably, the apparently simple neuronal organization of the cerebellum, in which Purkinje cells represent the only output, hides an elevated functional diversity even within the same neuronal population. Such complexity is the result of the integration of intrinsic morphogenetic programs and extracellular cues from the surrounding environment, which impact on the regulation of the transcriptome of cerebellar neurons. In this review, we briefly summarize key features of the development and structure of the cerebellum before focusing on the pathways involved in the acquisition of the cerebellar neuron identity. We focus on gene expression and mRNA processing programs, including mRNA methylation, trafficking and splicing, that are set in motion during cerebellar development and participate to its physiology. These programs are likely to add new layers of complexity and versatility that are fundamental for the adaptability of cerebellar neurons.
Collapse
|
45
|
Dionne O, Corbin F. A new strategy to uncover fragile X proteomic biomarkers using the nascent proteome of peripheral blood mononuclear cells (PBMCs). Sci Rep 2021; 11:15148. [PMID: 34312401 PMCID: PMC8313568 DOI: 10.1038/s41598-021-94027-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 07/05/2021] [Indexed: 02/06/2023] Open
Abstract
Fragile X syndrome (FXS) is the most prevalent inherited cause of intellectual disabilities and autism spectrum disorders. FXS result from the loss of expression of the FMRP protein, an RNA-binding protein that regulates the expression of key synaptic effectors. FXS is also characterized by a wide array of behavioural, cognitive and metabolic impairments. The severity and penetrance of those comorbidities are extremely variable, meaning that a considerable phenotypic heterogeneity is found among fragile X individuals. Unfortunately, clinicians currently have no tools at their disposal to assay a patient prognosis upon diagnosis. Since the absence of FMRP was repeatedly associated with an aberrant protein synthesis, we decided to study the nascent proteome in order to screen for potential proteomic biomarkers of FXS. We used a BONCAT (Biorthogonal Non-canonical Amino Acids Tagging) method coupled to label-free mass spectrometry to purify and quantify nascent proteins of peripheral blood mononuclear cells (PBMCs) from 7 fragile X male patients and 7 age-matched controls. The proteomic analysis identified several proteins which were either up or downregulated in PBMCs from FXS individuals. Eleven of those proteins were considered as potential biomarkers, of which 5 were further validated by Western blot. The gene ontology enrichment analysis highlighted molecular pathways that may contribute to FXS physiopathology. Our results suggest that the nascent proteome of PBMCs is well suited for the discovery of FXS biomarkers.
Collapse
Affiliation(s)
- Olivier Dionne
- Department of Biochemistry and Functional Genomic, Faculty of Medicine and Health Sciences, Université de Sherbrooke and Centre de Recherche du CHUS, CIUSSS de l'Estrie-CHUS, Sherbrooke, QC, Canada.
| | - François Corbin
- Department of Biochemistry and Functional Genomic, Faculty of Medicine and Health Sciences, Université de Sherbrooke and Centre de Recherche du CHUS, CIUSSS de l'Estrie-CHUS, Sherbrooke, QC, Canada.
| |
Collapse
|
46
|
Ellingford RA, Panasiuk MJ, de Meritens ER, Shaunak R, Naybour L, Browne L, Basson MA, Andreae LC. Cell-type-specific synaptic imbalance and disrupted homeostatic plasticity in cortical circuits of ASD-associated Chd8 haploinsufficient mice. Mol Psychiatry 2021; 26:3614-3624. [PMID: 33837267 PMCID: PMC8505247 DOI: 10.1038/s41380-021-01070-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 02/28/2021] [Accepted: 03/19/2021] [Indexed: 12/11/2022]
Abstract
Heterozygous mutation of chromodomain helicase DNA binding protein 8 (CHD8) is strongly associated with autism spectrum disorder (ASD) and results in dysregulated expression of neurodevelopmental and synaptic genes during brain development. To reveal how these changes affect ASD-associated cortical circuits, we studied synaptic transmission in the prefrontal cortex of a haploinsufficient Chd8 mouse model. We report profound alterations to both excitatory and inhibitory synaptic transmission onto deep layer projection neurons, resulting in a reduced excitatory:inhibitory balance, which were found to vary dynamically across neurodevelopment and result from distinct effects of reduced Chd8 expression within individual neuronal subtypes. These changes were associated with disrupted regulation of homeostatic plasticity mechanisms operating via spontaneous neurotransmission. These findings therefore directly implicate CHD8 mutation in the disruption of ASD-relevant circuits in the cortex.
Collapse
Affiliation(s)
- Robert A Ellingford
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
- Centre for Craniofacial & Regenerative Biology, King's College London, London, UK
| | - Martyna J Panasiuk
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
- MRC Centre for Neurodevelopmental Disorders, King's College London, London, UK
| | - Emilie Rabesahala de Meritens
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Raghav Shaunak
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Liam Naybour
- Centre for Craniofacial & Regenerative Biology, King's College London, London, UK
| | - Lorcan Browne
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - M Albert Basson
- Centre for Craniofacial & Regenerative Biology, King's College London, London, UK.
- MRC Centre for Neurodevelopmental Disorders, King's College London, London, UK.
| | - Laura C Andreae
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK.
- MRC Centre for Neurodevelopmental Disorders, King's College London, London, UK.
| |
Collapse
|
47
|
Wen J, Xu J, Mathena RP, Choi JH, Mintz CD. Early Isoflurane Exposure Impairs Synaptic Development in Fmr1 KO Mice via the mTOR Pathway. Neurochem Res 2021; 46:1577-1588. [PMID: 33791908 DOI: 10.1007/s11064-021-03301-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 02/23/2021] [Accepted: 03/17/2021] [Indexed: 12/16/2022]
Abstract
General anesthetics (GAs) may cause disruptions in brain development, and the effect of GA exposure in the setting of pre-existing neurodevelopmental disease is unknown. We tested the hypothesis that synaptic development is more vulnerable to GA-induced deficits in a mouse model of fragile X syndrome than in WT mice and asked whether they were related to the mTOR pathway, a signaling system implicated in both anesthesia toxicity and fragile X syndrome. Early postnatal WT and Fmr1-KO mice were exposed to isoflurane and brain slices were collected in adulthood. Primary neuron cultures isolated from WT and Fmr1-KO mice were exposed to isoflurane during development, in some cases treated with rapamycin, and processed for immunohistochemistry at maturity. Quantitative immunofluorescence microscopy was conducted for synaptic markers and markers of mTOR pathway activity. Isoflurane exposure caused reduction in Synpasin-1, PSD-95, and Gephyrin puncta that was significantly lower in Fmr1-KO mice than in WT mice. Similar results were found in cell culture, where synapse loss was ameliorated with rapamycin treatment. Early developmental exposure to isoflurane causes more profound synapse loss in Fmr1- KO than WT mice, and this effect is mediated by a pathologic increase in mTOR pathway activity.
Collapse
Affiliation(s)
- Jieqiong Wen
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China
- Department of Anesthesiology and Critical Care Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Jing Xu
- Department of Anesthesiology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
- Department of Anesthesiology and Critical Care Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - R Paige Mathena
- Department of Anesthesiology and Critical Care Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Jun H Choi
- Department of Anesthesiology and Critical Care Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - C David Mintz
- Department of Anesthesiology and Critical Care Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
| |
Collapse
|
48
|
Barthelson K, Baer L, Dong Y, Hand M, Pujic Z, Newman M, Goodhill GJ, Richards RI, Pederson SM, Lardelli M. Zebrafish Chromosome 14 Gene Differential Expression in the fmr1 h u2787 Model of Fragile X Syndrome. Front Genet 2021; 12:625466. [PMID: 34135935 PMCID: PMC8203322 DOI: 10.3389/fgene.2021.625466] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Accepted: 04/14/2021] [Indexed: 11/13/2022] Open
Abstract
Zebrafish represent a valuable model for investigating the molecular and cellular basis of Fragile X syndrome (FXS). Reduced expression of the zebrafish FMR1 orthologous gene, fmr1, causes developmental and behavioural phenotypes related to FXS. Zebrafish homozygous for the hu2787 non-sense mutation allele of fmr1 are widely used to model FXS, although FXS-relevant phenotypes seen from morpholino antisense oligonucleotide (morpholino) suppression of fmr1 transcript translation were not observed when hu2787 was first described. The subsequent discovery of transcriptional adaptation (a form of genetic compensation), whereby mutations causing non-sense-mediated decay of transcripts can drive compensatory upregulation of homologous transcripts independent of protein feedback loops, suggested an explanation for the differences reported. We examined the whole-embryo transcriptome effects of homozygosity for fmr1 h u2787 at 2 days post fertilisation. We observed statistically significant changes in expression of a number of gene transcripts, but none from genes showing sequence homology to fmr1. Enrichment testing of differentially expressed genes implied effects on lysosome function and glycosphingolipid biosynthesis. The majority of the differentially expressed genes are located, like fmr1, on Chromosome 14. Quantitative PCR tests did not support that this was artefactual due to changes in relative chromosome abundance. Enrichment testing of the "leading edge" differentially expressed genes from Chromosome 14 revealed that their co-location on this chromosome may be associated with roles in brain development and function. The differential expression of functionally related genes due to mutation of fmr1, and located on the same chromosome as fmr1, is consistent with R.A. Fisher's assertion that the selective advantage of co-segregation of particular combinations of alleles of genes will favour, during evolution, chromosomal rearrangements that place them in linkage disequilibrium on the same chromosome. However, we cannot exclude that the apparent differential expression of genes on Chromosome 14 genes was, (if only in part), caused by differences between the expression of alleles of genes unrelated to the effects of the fmr1 h u2787 mutation and made manifest due to the limited, but non-zero, allelic diversity between the genotypes compared.
Collapse
Affiliation(s)
- Karissa Barthelson
- School of Biological Sciences, University of Adelaide, Adelaide, SA, Australia
| | - Lachlan Baer
- School of Biological Sciences, University of Adelaide, Adelaide, SA, Australia
| | - Yang Dong
- School of Biological Sciences, University of Adelaide, Adelaide, SA, Australia
| | - Melanie Hand
- Bioinformatics Hub, University of Adelaide, Adelaide, SA, Australia
| | - Zac Pujic
- Queensland Brain Institute, University of Queensland, Brisbane, QLD, Australia
| | - Morgan Newman
- School of Biological Sciences, University of Adelaide, Adelaide, SA, Australia
| | - Geoffrey J. Goodhill
- Queensland Brain Institute, University of Queensland, Brisbane, QLD, Australia
- School of Mathematics and Physics, University of Queensland, Brisbane, QLD, Australia
| | - Robert I. Richards
- School of Biological Sciences, University of Adelaide, Adelaide, SA, Australia
| | | | - Michael Lardelli
- School of Biological Sciences, University of Adelaide, Adelaide, SA, Australia
| |
Collapse
|
49
|
Dionne O, Corbin F. An "Omic" Overview of Fragile X Syndrome. BIOLOGY 2021; 10:433. [PMID: 34068266 PMCID: PMC8153138 DOI: 10.3390/biology10050433] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 05/01/2021] [Accepted: 05/08/2021] [Indexed: 01/16/2023]
Abstract
Fragile X syndrome (FXS) is a neurodevelopmental disorder associated with a wide range of cognitive, behavioral and medical problems. It arises from the silencing of the fragile X mental retardation 1 (FMR1) gene and, consequently, in the absence of its encoded protein, FMRP (fragile X mental retardation protein). FMRP is a ubiquitously expressed and multifunctional RNA-binding protein, primarily considered as a translational regulator. Pre-clinical studies of the past two decades have therefore focused on this function to relate FMRP's absence to the molecular mechanisms underlying FXS physiopathology. Based on these data, successful pharmacological strategies were developed to rescue fragile X phenotype in animal models. Unfortunately, these results did not translate into humans as clinical trials using same therapeutic approaches did not reach the expected outcomes. These failures highlight the need to put into perspective the different functions of FMRP in order to get a more comprehensive understanding of FXS pathophysiology. This work presents a review of FMRP's involvement on noteworthy molecular mechanisms that may ultimately contribute to various biochemical alterations composing the fragile X phenotype.
Collapse
Affiliation(s)
- Olivier Dionne
- Department of Biochemistry and Functional Genomics, Faculty of Medicine and Health Sciences, Université de Sherbrooke and Centre de Recherche du CHUS, CIUSSS de l’Estrie-CHUS, Sherbrooke, QC J1H 5H4, Canada;
| | | |
Collapse
|
50
|
Dionne O, Lortie A, Gagnon F, Corbin F. Rates of protein synthesis are reduced in peripheral blood mononuclear cells (PBMCs) from fragile X individuals. PLoS One 2021; 16:e0251367. [PMID: 33974659 PMCID: PMC8112704 DOI: 10.1371/journal.pone.0251367] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Accepted: 04/23/2021] [Indexed: 12/30/2022] Open
Abstract
Background Fragile X syndrome (FXS) is the leading inherited cause of intellectual disability and is caused by the loss of expression of the Fragile X mental retardation protein (FMRP). In animal model of FXS, the absence of FMRP leads to an aberrant rate of neuronal protein synthesis, which in turn is believed to be at the origin of defects regarding spine morphology and synaptic plasticity. Normalisation of protein synthesis in these models has been associated with a rescue of FXS behavioral and biochemicals phenotype, thus establishing the rate of protein synthesis as one of the most promising monitoring biomarker for FXS. However, rate of protein synthesis alteration in fragile X individuals is not well characterized. Method We applied a robust radiolabeled assay to measure rate of protein synthesis in freshly extracted peripheral blood mononuclear cells (PBMCs) and blood platelets. We ultimately settle on PBMCs to measure and compare rate of protein synthesis in 13 males with fragile X and 14 matched controls individuals. Results Using this method, we measured a 26.9% decrease (p = 0,0193) in the rate of protein synthesis in fragile X individuals PBMCs. Furthermore, the rate of protein synthesis measurements obtained were highly reproducible, highlighting the robustness of the method. Conclusion Our work presents the first evidence of a diminution of the rate of protein synthesis in a human peripheral model of fragile X. Our results also support the finding of previous studies using brain PET imaging in Fragile X individuals. Since our assay only requires a simple venous puncture, it could be used in other cases of intellectual disability in order to determine if an aberrant rate of protein synthesis is a common general mechanism leading to impairment in synaptic plasticity and to intellectual disability.
Collapse
Affiliation(s)
- Olivier Dionne
- Department of Biochemistry and Functional Genomic, Faculty of Medicine and Health Sciences, Université de Sherbrooke and Centre de Recherche du CHUS, CIUSSS de l’Estrie-CHUS, Sherbrooke, Quebec, Canada
- * E-mail: (OD); (FC)
| | - Audrey Lortie
- Department of Biochemistry and Functional Genomic, Faculty of Medicine and Health Sciences, Université de Sherbrooke and Centre de Recherche du CHUS, CIUSSS de l’Estrie-CHUS, Sherbrooke, Quebec, Canada
| | - Florence Gagnon
- Department of Biochemistry and Functional Genomic, Faculty of Medicine and Health Sciences, Université de Sherbrooke and Centre de Recherche du CHUS, CIUSSS de l’Estrie-CHUS, Sherbrooke, Quebec, Canada
| | - François Corbin
- Department of Biochemistry and Functional Genomic, Faculty of Medicine and Health Sciences, Université de Sherbrooke and Centre de Recherche du CHUS, CIUSSS de l’Estrie-CHUS, Sherbrooke, Quebec, Canada
- * E-mail: (OD); (FC)
| |
Collapse
|