1
|
Ouardouz M, Jasinski P, Khalife M, Mahoney JM, Hernan AE, Scott RC. Hippocampal-prefrontal functional neural networks in a rat model of fragile X syndrome are poorly organized with limited resiliency. Sci Rep 2025; 15:16089. [PMID: 40341845 PMCID: PMC12062414 DOI: 10.1038/s41598-025-99408-8] [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: 01/09/2025] [Accepted: 04/21/2025] [Indexed: 05/11/2025] Open
Abstract
Fragile X Syndrome (FXS) is a common cause of autism spectrum symptoms. The genetic mutation results in multiple molecular alterations that are hypothesized to negatively impact neural circuit development although the nature of any functional neural dynamic consequences remain unclear. Therefore, the characteristics of hippocampal-prefrontal (H-PFC) network dysfunction were investigated in a rat model of FXS. FMR-KO and control rats underwent behavioral tests assessing sociability, memory, and anxiety to validate and replicate previously recognized deficits. Single-unit electrophysiology in the H-PFC circuit during exploration was used to measure patterns of action potential firing that were then compared between groups using generalized linear mixed models. FMR-KO rats demonstrated significant behavioral deficits in sociability, spatial learning, and anxiety. These rats also exhibited abnormal firing patterns outside of times when specific behavioral tasks were being performed. The network firing is less precise, more fragmented and with poor H-PFC communication in FXS. These findings suggest that disruptions in 'exploration' neural network dynamics impair the ability of networks to be appropriately engaged during specific behavioral tasks, leading to the observed deficits in social behavior, memory, and anxiety.
Collapse
Affiliation(s)
- Mohamed Ouardouz
- Nemours Children's Hospital, 1600 Rockland Road, Wilmington, DE, 19803, USA
| | - Patrick Jasinski
- Nemours Children's Hospital, 1600 Rockland Road, Wilmington, DE, 19803, USA
| | - Mohamed Khalife
- Nemours Children's Hospital, 1600 Rockland Road, Wilmington, DE, 19803, USA
- University of Delaware, 210 South College Street, Newark, DE, 19716, USA
| | | | - Amanda E Hernan
- Nemours Children's Hospital, 1600 Rockland Road, Wilmington, DE, 19803, USA
- University of Delaware, 210 South College Street, Newark, DE, 19716, USA
| | - Rod C Scott
- Nemours Children's Hospital, 1600 Rockland Road, Wilmington, DE, 19803, USA.
- University of Delaware, 210 South College Street, Newark, DE, 19716, USA.
- Sidney Kimmel College of Medicine, Thomas Jefferson University, 1035 Walnut Street, Philadelphia, PA, 19107, USA.
- Great Ormond Street Hospital NHS Trust, Great Ormond Street, London, WC1N 3JH, UK.
| |
Collapse
|
2
|
Müller L, Hatzfeld M. Emerging functions of Plakophilin 4 in the control of cell contact dynamics. Cell Commun Signal 2025; 23:109. [PMID: 40001215 PMCID: PMC11863852 DOI: 10.1186/s12964-025-02106-1] [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: 10/22/2024] [Accepted: 02/11/2025] [Indexed: 02/27/2025] Open
Abstract
Plakophilin 4 (PKP4, also called p0071) is a unique armadillo family protein localized at adherens junctions that acts as a scaffold protein capable of clustering cadherins. PKP4 also regulates cadherin recycling which is vital to enable junction dynamics. In addition, PKP4 controls the mechanical properties of cells by regulating actin filament organization through small Rho-GTPases. In this setting, PKP4 controls the localization and activity of specific guanine exchange factors (GEFs) and of their opponents, the GTPase activating proteins (GAPs). Through the formation of multiprotein complexes with Rho-GTPases, their regulators and their effectors, PKP4 controls the spatio-temporal activity of Rho signaling to regulate cell adhesion and cell mechanics. In keratinocytes, PKP4 prevents differentiation and at the same time dampens proliferation. This is, in part achieved through an interaction with the Hippo pathway, which controls the activity of the transcriptional co-factors YAP and TAZ. In a feedback loop, YAP/TAZ modulate PKP4 localization and function. Here, we review the various functions of PKP4 in cell signaling, cell mechanics, cell adhesion and growth control. We discuss how these functions converge in the regulation of cell adhesion dynamics to allow cells to adapt to their changing environment and enable proliferation, delamination but, at the same time, guarantee cell barrier function.
Collapse
Affiliation(s)
- Lisa Müller
- Institute of Molecular Medicine, Section for RNA biology and Pathogenesis, Martin Luther University Halle-Wittenberg, Charles Tanford Protein Research Center, Kurt-Mothes-Str. 3A, 06120, Halle, Germany.
| | - Mechthild Hatzfeld
- Institute of Molecular Medicine, Section for Pathobiochemistry, Martin Luther University Halle-Wittenberg, Charles Tanford Protein Research Center, Kurt-Mothes-Str. 3A, 06120, Halle, Germany
| |
Collapse
|
3
|
Rava A, Buzzelli V, Feo A, Ascone F, Di Trapano M, Schiavi S, Carbone E, Pasquadibisceglie A, Polticelli F, Manduca A, Trezza V. Role of peroxisome proliferator-activated receptors α and γ in mediating the beneficial effects of β-caryophyllene in a rat model of fragile X syndrome. Prog Neuropsychopharmacol Biol Psychiatry 2025; 136:111234. [PMID: 39725014 DOI: 10.1016/j.pnpbp.2024.111234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 12/17/2024] [Accepted: 12/21/2024] [Indexed: 12/28/2024]
Abstract
β-Caryophyllene (BCP) is a naturally occurring sesquiterpene found in numerous plant species, including Cannabis sativa. BCP has shown a high safety profile and a wide range of biological functions, including beneficial effects in neurodegenerative and inflammatory diseases. Here, we used behavioral, pharmacological, and in-silico docking analyses to investigate the effects and mechanism of action of BCP in Fragile X Syndrome (FXS), the most common inherited cause of Autism Spectrum Disorder (ASD) and intellectual disability. To this aim, we used the recently validated Fmr1-Δexon 8 rat model of FXS, that is also a genetic rat model of ASD. Acute and repeated oral administration of BCP rescued the cognitive deficits displayed by Fmr1-Δexon 8 rats, without inducing tolerance after repeated administration. These beneficial effects were mediated by activation of hippocampal peroxisome proliferator-activated receptors (PPARs) α and γ, and were mimicked by the PPARα agonist Fenofibrate and the PPARγ agonist Pioglitazone. Conversely, CB2 cannabinoid receptors were not involved. Docking analyses further confirmed the ability of BCP to bind rat PPARs. Together, our findings demonstrate that hippocampal PPARs α and γ play a role in the cognitive deficits observed in a rat model of FXS, and provide first preclinical evidence about the efficacy and mechanism of action of BCP in neurodevelopmental disorders.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Antonia Manduca
- Dept. Science, Roma Tre University, Rome, Italy; Dept. Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy; Neuroendocrinology, Metabolism and Neuropharmacology Unit, IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Viviana Trezza
- Dept. Science, Roma Tre University, Rome, Italy; Neuroendocrinology, Metabolism and Neuropharmacology Unit, IRCCS Fondazione Santa Lucia, Rome, Italy.
| |
Collapse
|
4
|
Jiang HY, Gu WW, Gan J, Yang Q, Shi Y, Lian WB, Xu HR, Yang SH, Yang L, Zhang X, Wang J. MNSFβ promotes LPS-induced TNFα expression by increasing the localization of RC3H1 to stress granules, and the interfering peptide HEPN2 reduces TNFα production by disrupting the MNSFβ-RC3H1 interaction in macrophages. Int Immunopharmacol 2024; 142:113053. [PMID: 39260307 DOI: 10.1016/j.intimp.2024.113053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 08/29/2024] [Accepted: 08/29/2024] [Indexed: 09/13/2024]
Abstract
Abnormally elevated tumor necrosis factor-α (TNFα) levels at the maternal-fetal interface can lead to adverse pregnancy outcomes, including recurrent miscarriage (RM), but the mechanism underlying upregulated TNFα expression is not fully understood. We previously reported that the interaction between monoclonal nonspecific suppressor factor-β (MNSFβ) and RC3H1 upregulates TNFα expression, but the precise mechanisms are unknown. In this study, we found that MNSFβ stimulated the LPS-induced TNFα expression by inactivating the promoting effect of RC3H1 on TNFα mRNA degradation rather than directly inhibiting the expression of RC3H1 in THP1-Mϕs. Mechanistically, the 81-326 aa region of the RC3H1 protein binds to the 101-133 aa region of the MNSFβ protein, and MNSFβ facilitated stress granules (SGs) formation and the translocation of RC3H1 to SGs by interacting with RC3H1 and fragile X mental retardation 1 (FMR1) in response to LPS-induced stress. The SGs-localization of RC3H1 reduced its inhibitory effect on TNFα expression in LPS-treated THP1-Mϕs. The designed HEPN2 peptide effectively reduced the LPS-induced expression of TNFα in THP1-Mϕs by interfering with the MNSFβ-RC3H1 interaction. Treatment with the HEPN2 peptide significantly improved adverse pregnancy outcomes, including early pregnancy loss (EPL) and lower fetal weight (LFW), which are induced by LPS in mice. These data indicated that MNSFβ promoted TNFα expression at least partially by increasing the localization of RC3H1 to SGs under inflammatory stimulation and that the HEPN2 peptide improved the adverse pregnancy outcomes induced by LPS in mice, suggesting that MNSFβ is a potential pharmacological target for adverse pregnancy outcomes caused by abnormally increased inflammation at early pregnancy.
Collapse
Affiliation(s)
- Han-Yu Jiang
- Shanghai Key Lab of Disease and Health Genomics, Shanghai Institute for Biomedical and Pharmaceutical Technologies, School of Pharmacy, Fudan University, Shanghai 20032, China
| | - Wen-Wen Gu
- Shanghai Key Lab of Disease and Health Genomics, Shanghai Institute for Biomedical and Pharmaceutical Technologies, School of Pharmacy, Fudan University, Shanghai 20032, China
| | - Jie Gan
- Shanghai Key Lab of Disease and Health Genomics, Shanghai Institute for Biomedical and Pharmaceutical Technologies, School of Pharmacy, Fudan University, Shanghai 20032, China
| | - Qian Yang
- Shanghai Key Lab of Disease and Health Genomics, Shanghai Institute for Biomedical and Pharmaceutical Technologies, School of Pharmacy, Fudan University, Shanghai 20032, China
| | - Yan Shi
- Shanghai Key Lab of Disease and Health Genomics, Shanghai Institute for Biomedical and Pharmaceutical Technologies, School of Pharmacy, Fudan University, Shanghai 20032, China
| | - Wen-Bo Lian
- Shanghai Key Lab of Disease and Health Genomics, Shanghai Institute for Biomedical and Pharmaceutical Technologies, School of Pharmacy, Fudan University, Shanghai 20032, China
| | - Hao-Ran Xu
- Shanghai Key Lab of Disease and Health Genomics, Shanghai Institute for Biomedical and Pharmaceutical Technologies, School of Pharmacy, Fudan University, Shanghai 20032, China
| | - Shu-Han Yang
- Shanghai Key Lab of Disease and Health Genomics, Shanghai Institute for Biomedical and Pharmaceutical Technologies, School of Pharmacy, Fudan University, Shanghai 20032, China
| | - Long Yang
- Shanghai Key Lab of Disease and Health Genomics, Shanghai Institute for Biomedical and Pharmaceutical Technologies, School of Pharmacy, Fudan University, Shanghai 20032, China.
| | - Xuan Zhang
- Shanghai Key Lab of Disease and Health Genomics, Shanghai Institute for Biomedical and Pharmaceutical Technologies, School of Pharmacy, Fudan University, Shanghai 20032, China
| | - Jian Wang
- Shanghai Key Lab of Disease and Health Genomics, Shanghai Institute for Biomedical and Pharmaceutical Technologies, School of Pharmacy, Fudan University, Shanghai 20032, China.
| |
Collapse
|
5
|
Juárez JCC, Gómez AA, Díaz AES, Arévalo GS. Understanding pathophysiology in fragile X syndrome: a comprehensive review. Neurogenetics 2024; 26:6. [PMID: 39585476 DOI: 10.1007/s10048-024-00794-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Accepted: 11/20/2024] [Indexed: 11/26/2024]
Abstract
Fragile X syndrome (FXS) is the leading hereditary cause of intellectual disability and the most commonly associated genetic cause of autism. Historically, research into its pathophysiology has focused predominantly on neurons; however, emerging evidence suggests involvement of additional cell types and systems. The objective of this study was to review and synthesize current evidence regarding the pathophysiology of Fragile X syndrome. A comprehensive literature review was conducted using databases such as PubMed and Google Scholar, employing MeSH terms including "Fragile X Syndrome," "FMR1 gene," and "FMRP." Studies on both human and animal models, from inception to 2022, published in recognized journals were included. The evidence supports those neurons, glial cells, stem cells, the immune system, and lipid metabolism pathways contribute to the pathophysiology of Fragile X syndrome. Further research is necessary to explore these fields independently and to elucidate their interactions.
Collapse
Affiliation(s)
| | - Alejandro Aguilar Gómez
- Faculty of Medical Sciences, Universidad of San Carlos of Guatemala, Guatemala City, Guatemala
| | | | - Gabriel Silva Arévalo
- Genetics and Metabolic Clinic Coordinator, Hospital Obras Sociales del Santo Hermano Pedro, Antigua Guatemala City, Guatemala
| |
Collapse
|
6
|
Ouardouz M, Jasinski P, Khalife M, Mahoney JM, Hernan AE, Scott RC. Disrupted Hippocampal-Prefrontal Networks in a Rat Model of Fragile X Syndrome: A Study Linking Neural Dynamics to Autism-Like Behavioral Impairments. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.15.617900. [PMID: 39464036 PMCID: PMC11507762 DOI: 10.1101/2024.10.15.617900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
Fragile X Syndrome (FXS) is associated with autism spectrum disorder (ASD) symptoms that are associated with cognitive, learning, and behavioral challenges. We investigated how known molecular disruptions in the Fmr1 knockout (FMR-KO) rat model of FXS negatively impact hippocampal-prefrontal cortex (H-PFC) neural network activity and consequent behavior. Methods FMR-KO and control rats underwent a battery of behavioral tests assessing sociability, memory, and anxiety. Single-unit electrophysiology recordings were then conducted to measure patterns of neural activity in H-PFC circuit. Advanced mathematical models were used to characterize the patterns that were then compared between groups using generalized linear mixed models. Results FMR-KO rats demonstrated significant behavioral deficits in sociability, spatial learning, and anxiety, aligning with symptoms of ASD. At the neural level, these rats exhibited abnormal firing patterns in the H-PFC circuit that is critical for learning, memory, and social behavior. The neural networks in FMR-KO rats were also less densely connected and more fragmented, particularly in hippocampal-PFC correlated firing. These findings suggest that disruptions in neural network dynamics underlie the observed behavioral impairments in FMR-KO rats. Conclusion FMR-KO significantly disrupts several characteristics of action potential firing in the H-PFC network, leading to deficits in social behavior, memory, and anxiety, as seen in FXS. This disruption is characterized by less organized and less resilient hippocampal-PFC networks. These findings suggest that therapeutic strategies aimed at normalizing neural dynamics, such as with brain stimulation, could potentially improve behavior and cognitive functions in autistic individuals. HIGHLIGHTS Fragile X Syndrome is associated with autism, cognitive challenges and anxietyThe loss of Fmr1 protein disrupts processes involved in building neural networksThe consequence is abnormal neural dynamics in hippocampal-prefrontal cortex networksNormalization of dynamics could improve outcomes in FXS and ASD.
Collapse
|
7
|
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
|
8
|
D'Antoni S, Spatuzza M, Bonaccorso CM, Catania MV. Role of fragile X messenger ribonucleoprotein 1 in the pathophysiology of brain disorders: a glia perspective. Neurosci Biobehav Rev 2024; 162:105731. [PMID: 38763180 DOI: 10.1016/j.neubiorev.2024.105731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 05/14/2024] [Accepted: 05/15/2024] [Indexed: 05/21/2024]
Abstract
Fragile X messenger ribonucleoprotein 1 (FMRP) is a widely expressed RNA binding protein involved in several steps of mRNA metabolism. Mutations in the FMR1 gene encoding FMRP are responsible for fragile X syndrome (FXS), a leading genetic cause of intellectual disability and autism spectrum disorder, and fragile X-associated tremor-ataxia syndrome (FXTAS), a neurodegenerative disorder in aging men. Although FMRP is mainly expressed in neurons, it is also present in glial cells and its deficiency or altered expression can affect functions of glial cells with implications for the pathophysiology of brain disorders. The present review focuses on recent advances on the role of glial subtypes, astrocytes, oligodendrocytes and microglia, in the pathophysiology of FXS and FXTAS, and describes how the absence or reduced expression of FMRP in these cells can impact on glial and neuronal functions. We will also briefly address the role of FMRP in radial glial cells and its effects on neural development, and gliomas and will speculate on the role of glial FMRP in other brain disorders.
Collapse
Affiliation(s)
- S D'Antoni
- Institute for Biomedical Research and Innovation (IRIB), National Research Council (CNR), Via Paolo Gaifami 18, Catania 95126, Italy
| | - M Spatuzza
- Institute for Biomedical Research and Innovation (IRIB), National Research Council (CNR), Via Paolo Gaifami 18, Catania 95126, Italy
| | - C M Bonaccorso
- Oasi Research Institute - IRCCS, via Conte Ruggero 73, Troina 94018, Italy
| | - M V Catania
- Institute for Biomedical Research and Innovation (IRIB), National Research Council (CNR), Via Paolo Gaifami 18, Catania 95126, Italy.
| |
Collapse
|
9
|
Manduca A, Buzzelli V, Rava A, Feo A, Carbone E, Schiavi S, Peruzzi B, D'Oria V, Pezzullo M, Pasquadibisceglie A, Polticelli F, Micale V, Kuchar M, Trezza V. Cannabidiol and positive effects on object recognition memory in an in vivo model of Fragile X Syndrome: Obligatory role of hippocampal GPR55 receptors. Pharmacol Res 2024; 203:107176. [PMID: 38583687 DOI: 10.1016/j.phrs.2024.107176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 04/03/2024] [Accepted: 04/04/2024] [Indexed: 04/09/2024]
Abstract
Cannabidiol (CBD), a non-psychotomimetic constituent of Cannabis sativa, has been recently approved for epileptic syndromes often associated with Autism spectrum disorder (ASD). However, the putative efficacy and mechanism of action of CBD in patients suffering from ASD and related comorbidities remain debated, especially because of the complex pharmacology of CBD. We used pharmacological, immunohistochemical and biochemical approaches to investigate the effects and mechanisms of action of CBD in the recently validated Fmr1-Δexon 8 rat model of ASD, that is also a model of Fragile X Syndrome (FXS), the leading monogenic cause of autism. CBD rescued the cognitive deficits displayed by juvenile Fmr1-Δexon 8 animals, without inducing tolerance after repeated administration. Blockade of CA1 hippocampal GPR55 receptors prevented the beneficial effect of both CBD and the fatty acid amide hydrolase (FAAH) inhibitor URB597 in the short-term recognition memory deficits displayed by Fmr1-Δexon 8 rats. Thus, CBD may exert its beneficial effects through CA1 hippocampal GPR55 receptors. Docking analysis further confirmed that the mechanism of action of CBD might involve competition for brain fatty acid binding proteins (FABPs) that deliver anandamide and related bioactive lipids to their catabolic enzyme FAAH. These findings demonstrate that CBD reduced cognitive deficits in a rat model of FXS and provide initial mechanistic insights into its therapeutic potential in neurodevelopmental disorders.
Collapse
Affiliation(s)
- Antonia Manduca
- Dept. Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy; Dept. Science, Roma Tre University, Rome, Italy; Neuroendocrinology, Metabolism and Neuropharmacology Unit, IRCCS Fondazione Santa Lucia, Rome, Italy.
| | | | | | | | | | | | - Barbara Peruzzi
- Bone Physiopathology Research Unit, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Valentina D'Oria
- Confocal Microscopy Core Facility, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Marco Pezzullo
- Histology Core Facility, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | | | | | - Vincenzo Micale
- Dept. Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Martin Kuchar
- Forensic Laboratory of Biologically Active Substances, Dept. Chemistry of Natural Compounds, University of Chemistry and Technologies, Prague, Czech Republic; Psychedelic Research Center, National Institute of Mental Health, Klecany, Czech Republic
| | - Viviana Trezza
- Dept. Science, Roma Tre University, Rome, Italy; Neuroendocrinology, Metabolism and Neuropharmacology Unit, IRCCS Fondazione Santa Lucia, Rome, Italy.
| |
Collapse
|
10
|
Taha MS, Ahmadian MR. Fragile X Messenger Ribonucleoprotein Protein and Its Multifunctionality: From Cytosol to Nucleolus and Back. Biomolecules 2024; 14:399. [PMID: 38672417 PMCID: PMC11047961 DOI: 10.3390/biom14040399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 03/17/2024] [Accepted: 03/19/2024] [Indexed: 04/28/2024] Open
Abstract
Silencing of the fragile X messenger ribonucleoprotein 1 (FMR1) gene and a consequent lack of FMR protein (FMRP) synthesis are associated with fragile X syndrome, one of the most common inherited intellectual disabilities. FMRP is a multifunctional protein that is involved in many cellular functions in almost all subcellular compartments under both normal and cellular stress conditions in neuronal and non-neuronal cell types. This is achieved through its trafficking signals, nuclear localization signal (NLS), nuclear export signal (NES), and nucleolar localization signal (NoLS), as well as its RNA and protein binding domains, and it is modulated by various post-translational modifications such as phosphorylation, ubiquitination, sumoylation, and methylation. This review summarizes the recent advances in understanding the interaction networks of FMRP with a special focus on FMRP stress-related functions, including stress granule formation, mitochondrion and endoplasmic reticulum plasticity, ribosome biogenesis, cell cycle control, and DNA damage response.
Collapse
Affiliation(s)
- Mohamed S. Taha
- Institute of Biochemistry and Molecular Biology II, Medical Faculty, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany;
- Research on Children with Special Needs Department, Institute of Medical Research and Clinical Studies, National Research Centre, Cairo 12622, Egypt
| | - Mohammad Reza Ahmadian
- Institute of Biochemistry and Molecular Biology II, Medical Faculty, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany;
| |
Collapse
|
11
|
Annear DJ, Kooy RF. Unravelling the link between neurodevelopmental disorders and short tandem CGG-repeat expansions. Emerg Top Life Sci 2023; 7:265-275. [PMID: 37768318 PMCID: PMC10754333 DOI: 10.1042/etls20230021] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 08/23/2023] [Accepted: 09/11/2023] [Indexed: 09/29/2023]
Abstract
Neurodevelopmental disorders (NDDs) encompass a diverse group of disorders characterised by impaired cognitive abilities and developmental challenges. Short tandem repeats (STRs), repetitive DNA sequences found throughout the human genome, have emerged as potential contributors to NDDs. Specifically, the CGG trinucleotide repeat has been implicated in a wide range of NDDs, including Fragile X Syndrome (FXS), the most common inherited form of intellectual disability and autism. This review focuses on CGG STR expansions associated with NDDs and their impact on gene expression through repeat expansion-mediated epigenetic silencing. We explore the molecular mechanisms underlying CGG-repeat expansion and the resulting epigenetic modifications, such as DNA hypermethylation and gene silencing. Additionally, we discuss the involvement of other CGG STRs in neurodevelopmental diseases. Several examples, including FMR1, AFF2, AFF3, XYLT1, FRA10AC1, CBL, and DIP2B, highlight the complex relationship between CGG STR expansions and NDDs. Furthermore, recent advancements in this field are highlighted, shedding light on potential future research directions. Understanding the role of STRs, particularly CGG-repeats, in NDDs has the potential to uncover novel diagnostic and therapeutic strategies for these challenging disorders.
Collapse
Affiliation(s)
- Dale J Annear
- Department of Medical Genetics, University of Antwerp, Antwerp, Belgium
| | - R Frank Kooy
- Department of Medical Genetics, University of Antwerp, Antwerp, Belgium
| |
Collapse
|
12
|
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: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [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
|
13
|
Jin C, Zhang X, Lei Q, Chen P, Hu H, Shen S, Liu J, Ye S. Case report: genetic analysis of a novel frameshift mutation in FMR1 gene in a Chinese family. Front Genet 2023; 14:1228682. [PMID: 37745859 PMCID: PMC10512415 DOI: 10.3389/fgene.2023.1228682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Accepted: 08/21/2023] [Indexed: 09/26/2023] Open
Abstract
Fragile X syndrome (FXS) [OMIM 300624] is a common X-linked inherited syndrome with an incidence only second to that of trisomy 21. More than 95% of fragile X syndrome is caused by reduced or absent fragile X intellectual disability protein 1 (FMRP) synthesis due to dynamic mutation expansion of the CGG triplet repeat in the 5'UTR and abnormal methylation of the FMR1 (fragile X messenger ribonucleoprotein 1) gene [OMIM 309550]. Less than 5% of cases are caused by abnormal function of the FMRP due to point mutations or deletions in the FMR1 gene. In a proband with clinical suspicion of FXS and no CGG duplication, we found the presence of c.585_586del (p.Lys195AsnfsTer8) in exon 7 of the FMR1 gene using whole exome sequencing (WES). This variant resulted in frameshift and a premature stop codon after 8 aberrant amino acids. This variant is a novel pathogenic mutation, as determined by pedigree analysis, which has not been reported in any database or literature.
Collapse
Affiliation(s)
- Chunlei Jin
- Center of Medical Prenatal Diagnosis, Lishui Maternity and Child Health Care Hospital, Lishui, China
| | - Xiangdong Zhang
- Center of Medical Prenatal Diagnosis, Lishui Maternity and Child Health Care Hospital, Lishui, China
| | - Qiang Lei
- Center of Medical Prenatal Diagnosis, Lishui Maternity and Child Health Care Hospital, Lishui, China
| | - Penglong Chen
- Center of Medical Prenatal Diagnosis, Lishui Maternity and Child Health Care Hospital, Lishui, China
| | - Hui Hu
- Center of Medical Prenatal Diagnosis, Lishui Maternity and Child Health Care Hospital, Lishui, China
| | - Shuangshuang Shen
- Center of Medical Prenatal Diagnosis, Jinhua Maternity and Child Health Care Hospital, Jinhua, China
| | - Jiao Liu
- Center of Medical Prenatal Diagnosis, Lishui Maternity and Child Health Care Hospital, Lishui, China
| | - Shixuanbao Ye
- Center of Medical Prenatal Diagnosis, Lishui Maternity and Child Health Care Hospital, Lishui, China
| |
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
|
Schiavi S, Manduca A, Carbone E, Buzzelli V, Rava A, Feo A, Ascone F, Morena M, Campolongo P, Hill MN, Trezza V. Anandamide and 2-arachidonoylglycerol differentially modulate autistic-like traits in a genetic model of autism based on FMR1 deletion in rats. Neuropsychopharmacology 2023; 48:897-907. [PMID: 36114286 PMCID: PMC10156791 DOI: 10.1038/s41386-022-01454-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 07/20/2022] [Accepted: 09/01/2022] [Indexed: 11/09/2022]
Abstract
Autism spectrum disorder (ASD) has a multifactorial etiology. Major efforts are underway to understand the neurobiological bases of ASD and to develop efficacious treatment strategies. Recently, the use of cannabinoid compounds in children with neurodevelopmental disorders including ASD has received increasing attention. Beyond anecdotal reports of efficacy, however, there is limited current evidence supporting such an intervention and the clinical studies currently available have intrinsic limitations that make the interpretation of the findings challenging. Furthermore, as the mechanisms underlying the beneficial effects of cannabinoid compounds in neurodevelopmental disorders are still largely unknown, the use of drugs targeting the endocannabinoid system remains controversial. Here, we studied the role of endocannabinoid neurotransmission in the autistic-like traits displayed by the recently validated Fmr1-Δexon 8 rat model of autism. Fmr1-Δexon 8 rats showed reduced anandamide levels in the hippocampus and increased 2-arachidonoylglycerol (2-AG) content in the amygdala. Systemic and intra-hippocampal potentiation of anandamide tone through administration of the anandamide hydrolysis inhibitor URB597 ameliorated the cognitive deficits displayed by Fmr1-Δexon 8 rats along development, as assessed through the novel object and social discrimination tasks. Moreover, blockade of amygdalar 2-AG signaling through intra-amygdala administration of the CB1 receptor antagonist SR141716A prevented the altered sociability displayed by Fmr1-Δexon 8 rats. These findings demonstrate that anandamide and 2-AG differentially modulate specific autistic-like traits in Fmr1-Δexon 8 rats in a brain region-specific manner, suggesting that fine changes in endocannabinoid mechanisms contribute to ASD-related behavioral phenotypes.
Collapse
Affiliation(s)
- Sara Schiavi
- Department of Science, Roma Tre University, Rome, Italy
| | - Antonia Manduca
- Department of Science, Roma Tre University, Rome, Italy
- Neuroendocrinology, Metabolism and Neuropharmacology Unit, IRCSS Fondazione Santa Lucia, Rome, Italy
| | | | | | | | | | | | - Maria Morena
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy
- Neuropsychopharmacology Unit, IRCSS Fondazione Santa Lucia, Rome, Italy
- Departments of Cell Biology and Anatomy & Psychiatry, Hotchkiss Brain Institute and Mathison Center for Mental Health Research and Education, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Patrizia Campolongo
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy
- Neuropsychopharmacology Unit, IRCSS Fondazione Santa Lucia, Rome, Italy
| | - Matthew N Hill
- Departments of Cell Biology and Anatomy & Psychiatry, Hotchkiss Brain Institute and Mathison Center for Mental Health Research and Education, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Viviana Trezza
- Department of Science, Roma Tre University, Rome, Italy.
| |
Collapse
|
16
|
Villalonga E, Mosrin C, Normand T, Girardin C, Serrano A, Žunar B, Doudeau M, Godin F, Bénédetti H, Vallée B. LIM Kinases, LIMK1 and LIMK2, Are Crucial Node Actors of the Cell Fate: Molecular to Pathological Features. Cells 2023; 12:cells12050805. [PMID: 36899941 PMCID: PMC10000741 DOI: 10.3390/cells12050805] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 02/28/2023] [Accepted: 03/02/2023] [Indexed: 03/08/2023] Open
Abstract
LIM kinase 1 (LIMK1) and LIM kinase 2 (LIMK2) are serine/threonine and tyrosine kinases and the only two members of the LIM kinase family. They play a crucial role in the regulation of cytoskeleton dynamics by controlling actin filaments and microtubule turnover, especially through the phosphorylation of cofilin, an actin depolymerising factor. Thus, they are involved in many biological processes, such as cell cycle, cell migration, and neuronal differentiation. Consequently, they are also part of numerous pathological mechanisms, especially in cancer, where their involvement has been reported for a few years and has led to the development of a wide range of inhibitors. LIMK1 and LIMK2 are known to be part of the Rho family GTPase signal transduction pathways, but many more partners have been discovered over the decades, and both LIMKs are suspected to be part of an extended and various range of regulation pathways. In this review, we propose to consider the different molecular mechanisms involving LIM kinases and their associated signalling pathways, and to offer a better understanding of their variety of actions within the physiology and physiopathology of the cell.
Collapse
Affiliation(s)
- Elodie Villalonga
- Centre de Biophysique Moléculaire; UPR4301, CNRS, University of Orleans and INSERM, CEDEX 2, 45071 Orleans, France
| | - Christine Mosrin
- Centre de Biophysique Moléculaire; UPR4301, CNRS, University of Orleans and INSERM, CEDEX 2, 45071 Orleans, France
| | - Thierry Normand
- Centre de Biophysique Moléculaire; UPR4301, CNRS, University of Orleans and INSERM, CEDEX 2, 45071 Orleans, France
| | - Caroline Girardin
- Centre de Biophysique Moléculaire; UPR4301, CNRS, University of Orleans and INSERM, CEDEX 2, 45071 Orleans, France
| | - Amandine Serrano
- Centre de Biophysique Moléculaire; UPR4301, CNRS, University of Orleans and INSERM, CEDEX 2, 45071 Orleans, France
| | - Bojan Žunar
- Laboratory for Biochemistry, Department of Chemistry and Biochemistry, Faculty of Food Technology and Biotechnology, University of Zagreb, 10000 Zagreb, Croatia
| | - Michel Doudeau
- Centre de Biophysique Moléculaire; UPR4301, CNRS, University of Orleans and INSERM, CEDEX 2, 45071 Orleans, France
| | - Fabienne Godin
- Centre de Biophysique Moléculaire; UPR4301, CNRS, University of Orleans and INSERM, CEDEX 2, 45071 Orleans, France
| | - Hélène Bénédetti
- Centre de Biophysique Moléculaire; UPR4301, CNRS, University of Orleans and INSERM, CEDEX 2, 45071 Orleans, France
| | - Béatrice Vallée
- Centre de Biophysique Moléculaire; UPR4301, CNRS, University of Orleans and INSERM, CEDEX 2, 45071 Orleans, France
- Correspondence: ; Tel.: +33-(0)2-38-25-76-11
| |
Collapse
|
17
|
Sex-Related Changes in the Clinical, Genetic, Electrophysiological, Connectivity, and Molecular Presentations of ASD: A Comparison between Human and Animal Models of ASD with Reference to Our Data. Int J Mol Sci 2023; 24:ijms24043287. [PMID: 36834699 PMCID: PMC9965966 DOI: 10.3390/ijms24043287] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 01/28/2023] [Accepted: 02/03/2023] [Indexed: 02/11/2023] Open
Abstract
The etiology of autism spectrum disorder (ASD) is genetic, environmental, and epigenetic. In addition to sex differences in the prevalence of ASD, which is 3-4 times more common in males, there are also distinct clinical, molecular, electrophysiological, and pathophysiological differences between sexes. In human, males with ASD have more externalizing problems (i.e., attention-deficit hyperactivity disorder), more severe communication and social problems, as well as repetitive movements. Females with ASD generally exhibit fewer severe communication problems, less repetitive and stereotyped behavior, but more internalizing problems, such as depression and anxiety. Females need a higher load of genetic changes related to ASD compared to males. There are also sex differences in brain structure, connectivity, and electrophysiology. Genetic or non-genetic experimental animal models of ASD-like behavior, when studied for sex differences, showed some neurobehavioral and electrophysiological differences between male and female animals depending on the specific model. We previously carried out studies on behavioral and molecular differences between male and female mice treated with valproic acid, either prenatally or early postnatally, that exhibited ASD-like behavior and found distinct differences between the sexes, the female mice performing better on tests measuring social interaction and undergoing changes in the expression of more genes in the brain compared to males. Interestingly, co-administration of S-adenosylmethionine alleviated the ASD-like behavioral symptoms and the gene-expression changes to the same extent in both sexes. The mechanisms underlying the sex differences are not yet fully understood.
Collapse
|
18
|
Therapeutic Potential and Limitation of Serotonin Type 7 Receptor Modulation. Int J Mol Sci 2023; 24:ijms24032070. [PMID: 36768393 PMCID: PMC9916679 DOI: 10.3390/ijms24032070] [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: 01/06/2023] [Revised: 01/15/2023] [Accepted: 01/18/2023] [Indexed: 01/21/2023] Open
Abstract
Although a number of mood-stabilising atypical antipsychotics and antidepressants modulate serotonin type 7 receptor (5-HT7), the detailed contributions of 5-HT7 function to clinical efficacy and pathophysiology have not been fully understood. The mood-stabilising antipsychotic agent, lurasidone, and the serotonin partial agonist reuptake inhibitor, vortioxetine, exhibit higher binding affinity to 5-HT7 than other conventional antipsychotics and antidepressants. To date, the initially expected rapid onset of antidepressant effects-in comparison with conventional antidepressants or mood-stabilising antipsychotics-due to 5-HT7 inhibition has not been observed with lurasidone and vortioxetine; however, several clinical studies suggest that 5-HT7 inhibition likely contributes to quality of life of patients with schizophrenia and mood disorders via the improvement of cognition. Furthermore, recent preclinical studies reported that 5-HT7 inhibition might mitigate antipsychotic-induced weight gain and metabolic complication by blocking other monoamine receptors. Further preclinical studies for the development of 5-HT7 modulation against neurodevelopmental disorders and neurodegenerative diseases have been ongoing. To date, various findings from various preclinical studies indicate the possibility that 5-HT7 modifications can provide two independent strategies. The first is that 5-HT7 inhibition ameliorates the dysfunction of inter-neuronal transmission in mature networks. The other is that activation of 5-HT7 can improve transmission dysfunction due to microstructure abnormality in the neurotransmission network-which could be unaffected by conventional therapeutic agents-via modulating intracellular signalling during the neurodevelopmental stage or via loss of neural networks with aging. This review attempts to describe the current and novel clinical applications of 5-HT7 modulation based on preclinical findings.
Collapse
|
19
|
Tempio A, Boulksibat A, Bardoni B, Delhaye S. Fragile X Syndrome as an interneuronopathy: a lesson for future studies and treatments. Front Neurosci 2023; 17:1171895. [PMID: 37188005 PMCID: PMC10176609 DOI: 10.3389/fnins.2023.1171895] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 04/05/2023] [Indexed: 05/17/2023] Open
Abstract
Fragile X Syndrome (FXS) is the most common form of inherited intellectual disability (ID) and a primary genetic cause of autism spectrum disorder (ASD). FXS arises from the silencing of the FMR1 gene causing the lack of translation of its encoded protein, the Fragile X Messenger RibonucleoProtein (FMRP), an RNA-binding protein involved in translational control and in RNA transport along dendrites. Although a large effort during the last 20 years has been made to investigate the cellular roles of FMRP, no effective and specific therapeutic intervention is available to treat FXS. Many studies revealed a role for FMRP in shaping sensory circuits during developmental critical periods to affect proper neurodevelopment. Dendritic spine stability, branching and density abnormalities are part of the developmental delay observed in various FXS brain areas. In particular, cortical neuronal networks in FXS are hyper-responsive and hyperexcitable, making these circuits highly synchronous. Overall, these data suggest that the excitatory/inhibitory (E/I) balance in FXS neuronal circuitry is altered. However, not much is known about how interneuron populations contribute to the unbalanced E/I ratio in FXS even if their abnormal functioning has an impact on the behavioral deficits of patients and animal models affected by neurodevelopmental disorders. We revise here the key literature concerning the role of interneurons in FXS not only with the purpose to better understand the pathophysiology of this disorder, but also to explore new possible therapeutic applications to treat FXS and other forms of ASD or ID. Indeed, for instance, the re-introduction of functional interneurons in the diseased brains has been proposed as a promising therapeutic approach for neurological and psychiatric disorders.
Collapse
Affiliation(s)
- Alessandra Tempio
- Université Côte d’Azur, CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, Valbonne, France
- Alessandra Tempio,
| | - Asma Boulksibat
- Université Côte d’Azur, CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, Valbonne, France
| | - Barbara Bardoni
- Université Côte d’Azur, CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, Valbonne, France
- Inserm, Université Côte d’Azur, CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, Valbonne, France
- *Correspondence: Barbara Bardoni,
| | - Sébastien Delhaye
- Université Côte d’Azur, CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, Valbonne, France
| |
Collapse
|
20
|
Dwivedi Y, Shelton RC. Genomics in Treatment Development. ADVANCES IN NEUROBIOLOGY 2023; 30:363-385. [PMID: 36928858 DOI: 10.1007/978-3-031-21054-9_15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/18/2023]
Abstract
The Human Genome Project mapped the 3 billion base pairs in the human genome, which ushered in a new generation of genomically focused treatment development. While this has been very successful in other areas, neuroscience has been largely devoid of such developments. This is in large part because there are very few neurological or mental health conditions that are related to single-gene variants. While developments in pharmacogenomics have been somewhat successful, the use of genetic information in practice has to do with drug metabolism and adverse reactions. Studies of drug metabolism related to genetic variations are an important part of drug development. However, outside of cancer biology, the actual translation of genomic information into novel therapies has been limited. Epigenetics, which relates in part to the effects of the environment on DNA, is a promising newer area of relevance to CNS disorders. The environment can induce chemical modifications of DNA (e.g., cytosine methylation), which can be induced by the environment and may represent either shorter- or longer-term changes. Given the importance of environmental influences on CNS disorders, epigenetics may identify important treatment targets in the future.
Collapse
Affiliation(s)
- Yogesh Dwivedi
- Department of Psychiatry and Behavioral Neurobiology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Richard C Shelton
- Department of Psychiatry and Behavioral Neurobiology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
21
|
D’Elia A, Schiavi S, Manduca A, Rava A, Buzzelli V, Ascone F, Orsini T, Putti S, Soluri A, Galli F, Soluri A, Mattei M, Cicconi R, Massari R, Trezza V. FMR1 deletion in rats induces hyperactivity with no changes in striatal dopamine transporter availability. Sci Rep 2022; 12:22535. [PMID: 36581671 PMCID: PMC9800572 DOI: 10.1038/s41598-022-26986-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Accepted: 12/22/2022] [Indexed: 12/30/2022] Open
Abstract
Autism Spectrum Disorder (ASD) is a pervasive neurodevelopmental disorder emerging in early life characterized by impairments in social interaction, poor verbal and non-verbal communication, and repetitive patterns of behaviors. Among the best-known genetic risk factors for ASD, there are mutations causing the loss of the Fragile X Messenger Ribonucleoprotein 1 (FMRP) leading to Fragile X syndrome (FXS), a common form of inherited intellectual disability and the leading monogenic cause of ASD. Being a pivotal regulator of motor activity, motivation, attention, and reward processing, dopaminergic neurotransmission has a key role in several neuropsychiatric disorders, including ASD. Fmr1 Δexon 8 rats have been validated as a genetic model of ASD based on FMR1 deletion, and they are also a rat model of FXS. Here, we performed behavioral, biochemical and in vivo SPECT neuroimaging experiments to investigate whether Fmr1 Δexon 8 rats display ASD-like repetitive behaviors associated with changes in striatal dopamine transporter (DAT) availability assessed through in vivo SPECT neuroimaging. At the behavioral level, Fmr1 Δexon 8 rats displayed hyperactivity in the open field test in the absence of repetitive behaviors in the hole board test. However, these behavioral alterations were not associated with changes in striatal DAT availability as assessed by non-invasive in vivo SPECT and Western blot analyses.
Collapse
Affiliation(s)
- Annunziata D’Elia
- grid.5326.20000 0001 1940 4177Institute of Biochemistry and Cell Biology (IBBC), National Research Council of Italy (CNR), c/o International Campus “A. Buzzati-Traverso”, Via E. Ramarini, 32, 00015 Monterotondo Scalo (Rome), Italy ,grid.8509.40000000121622106Department of Science, Section of Biomedical Sciences and Technologies, Roma Tre University, Viale G. Marconi 446, 00146 Rome, Italy
| | - Sara Schiavi
- grid.8509.40000000121622106Department of Science, Section of Biomedical Sciences and Technologies, Roma Tre University, Viale G. Marconi 446, 00146 Rome, Italy
| | - Antonia Manduca
- grid.8509.40000000121622106Department of Science, Section of Biomedical Sciences and Technologies, Roma Tre University, Viale G. Marconi 446, 00146 Rome, Italy ,grid.417778.a0000 0001 0692 3437Neuroendocrinology, Metabolism and Neuropharmacology Unit, IRCSS Fondazione Santa Lucia, Rome, Italy
| | - Alessandro Rava
- grid.8509.40000000121622106Department of Science, Section of Biomedical Sciences and Technologies, Roma Tre University, Viale G. Marconi 446, 00146 Rome, Italy
| | - Valeria Buzzelli
- grid.8509.40000000121622106Department of Science, Section of Biomedical Sciences and Technologies, Roma Tre University, Viale G. Marconi 446, 00146 Rome, Italy
| | - Fabrizio Ascone
- grid.8509.40000000121622106Department of Science, Section of Biomedical Sciences and Technologies, Roma Tre University, Viale G. Marconi 446, 00146 Rome, Italy
| | - Tiziana Orsini
- grid.5326.20000 0001 1940 4177Institute of Biochemistry and Cell Biology (IBBC), National Research Council of Italy (CNR), c/o International Campus “A. Buzzati-Traverso”, Via E. Ramarini, 32, 00015 Monterotondo Scalo (Rome), Italy
| | - Sabrina Putti
- grid.5326.20000 0001 1940 4177Institute of Biochemistry and Cell Biology (IBBC), National Research Council of Italy (CNR), c/o International Campus “A. Buzzati-Traverso”, Via E. Ramarini, 32, 00015 Monterotondo Scalo (Rome), Italy
| | - Andrea Soluri
- grid.5326.20000 0001 1940 4177Institute of Biochemistry and Cell Biology (IBBC), National Research Council of Italy (CNR), c/o International Campus “A. Buzzati-Traverso”, Via E. Ramarini, 32, 00015 Monterotondo Scalo (Rome), Italy ,grid.9657.d0000 0004 1757 5329Unit of Molecular Neurosciences, University Campus Bio-Medico, Rome, Rome, Italy
| | - Filippo Galli
- grid.7841.aNuclear Medicine Unit, Department of Medical-Surgical Sciences and of Translational Medicine, Faculty of Medicine and Psychology, “Sapienza” University of Rome, Rome, Italy
| | - Alessandro Soluri
- grid.5326.20000 0001 1940 4177Institute of Biochemistry and Cell Biology (IBBC), National Research Council of Italy (CNR), c/o International Campus “A. Buzzati-Traverso”, Via E. Ramarini, 32, 00015 Monterotondo Scalo (Rome), Italy
| | - Maurizio Mattei
- grid.6530.00000 0001 2300 0941Department of Biology and Centro di Servizi Interdipartimentale-Stazione per la Tecnologia Animale, “Tor Vergata” University, Rome, Italy
| | - Rosella Cicconi
- grid.6530.00000 0001 2300 0941Department of Biology and Centro di Servizi Interdipartimentale-Stazione per la Tecnologia Animale, “Tor Vergata” University, Rome, Italy
| | - Roberto Massari
- grid.5326.20000 0001 1940 4177Institute of Biochemistry and Cell Biology (IBBC), National Research Council of Italy (CNR), c/o International Campus “A. Buzzati-Traverso”, Via E. Ramarini, 32, 00015 Monterotondo Scalo (Rome), Italy
| | - Viviana Trezza
- grid.8509.40000000121622106Department of Science, Section of Biomedical Sciences and Technologies, Roma Tre University, Viale G. Marconi 446, 00146 Rome, Italy
| |
Collapse
|
22
|
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
|
23
|
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
|
24
|
Boldyreva LV, Andreyeva EN, Pindyurin AV. Position Effect Variegation: Role of the Local Chromatin Context in Gene Expression Regulation. Mol Biol 2022. [DOI: 10.1134/s0026893322030049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
25
|
Napolitano A, Schiavi S, La Rosa P, Rossi-Espagnet MC, Petrillo S, Bottino F, Tagliente E, Longo D, Lupi E, Casula L, Valeri G, Piemonte F, Trezza V, Vicari S. Sex Differences in Autism Spectrum Disorder: Diagnostic, Neurobiological, and Behavioral Features. Front Psychiatry 2022; 13:889636. [PMID: 35633791 PMCID: PMC9136002 DOI: 10.3389/fpsyt.2022.889636] [Citation(s) in RCA: 73] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 04/25/2022] [Indexed: 12/25/2022] Open
Abstract
Autism Spectrum Disorder (ASD) is a complex neurodevelopmental disorder with a worldwide prevalence of about 1%, characterized by impairments in social interaction, communication, repetitive patterns of behaviors, and can be associated with hyper- or hypo-reactivity of sensory stimulation and cognitive disability. ASD comorbid features include internalizing and externalizing symptoms such as anxiety, depression, hyperactivity, and attention problems. The precise etiology of ASD is still unknown and it is undoubted that the disorder is linked to some extent to both genetic and environmental factors. It is also well-documented and known that one of the most striking and consistent finding in ASD is the higher prevalence in males compared to females, with around 70% of ASD cases described being males. The present review looked into the most significant studies that attempted to investigate differences in ASD males and females thus trying to shade some light on the peculiar characteristics of this prevalence in terms of diagnosis, imaging, major autistic-like behavior and sex-dependent uniqueness. The study also discussed sex differences found in animal models of ASD, to provide a possible explanation of the neurological mechanisms underpinning the different presentation of autistic symptoms in males and females.
Collapse
Affiliation(s)
- Antonio Napolitano
- Medical Physics Department, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Sara Schiavi
- Section of Biomedical Sciences and Technologies, Science Department, Roma Tre University, Rome, Italy
| | - Piergiorgio La Rosa
- Division of Neuroscience, Department of Psychology, Sapienza University of Rome, Rome, Italy
| | - Maria Camilla Rossi-Espagnet
- Neuroradiology Unit, Imaging Department, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
- NESMOS, Neuroradiology Department, S. Andrea Hospital Sapienza University, Rome, Italy
| | - Sara Petrillo
- Head Child and Adolescent Psychiatry Unit, Neuroscience Department, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Francesca Bottino
- Medical Physics Department, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Emanuela Tagliente
- Medical Physics Department, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Daniela Longo
- Neuroradiology Unit, Imaging Department, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Elisabetta Lupi
- Head Child and Adolescent Psychiatry Unit, Neuroscience Department, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Laura Casula
- Head Child and Adolescent Psychiatry Unit, Neuroscience Department, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Giovanni Valeri
- Head Child and Adolescent Psychiatry Unit, Neuroscience Department, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Fiorella Piemonte
- Neuromuscular and Neurodegenerative Diseases Unit, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Viviana Trezza
- Section of Biomedical Sciences and Technologies, Science Department, Roma Tre University, Rome, Italy
| | - Stefano Vicari
- Child Neuropsychiatry Unit, Neuroscience Department, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
- Life Sciences and Public Health Department, Catholic University, Rome, Italy
| |
Collapse
|
26
|
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
|
27
|
Sathyanarayana SH, Saunders JA, Slaughter J, Tariq K, Chakrabarti R, Sadanandappa MK, Luikart BW, Bosco G. Pten heterozygosity restores neuronal morphology in fragile X syndrome mice. Proc Natl Acad Sci U S A 2022; 119:e2109448119. [PMID: 35394871 PMCID: PMC9169627 DOI: 10.1073/pnas.2109448119] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 02/11/2022] [Indexed: 11/30/2022] Open
Abstract
Genetic studies of hippocampal granule neuron development have been used to elucidate cellular functions of Pten and Fmr1. While mutations in each gene cause neurodevelopmental disorders such as autism and fragile X syndrome, how Pten and Fmr1 function alone or together during normal development is not known. Moreover, Pten mRNA is bound by the fragile X mental retardation protein (FMRP) RNA binding protein, but how this physical interaction impinges on phosphatase and tensin homolog protein (PTEN) expression is not known. To understand the interaction of PTEN and FMRP, we investigated the dentate gyrus granule neuron development in Pten and Fmr1 knockout (KO) mice. Interestingly, heterozygosity of Pten restored Fmr1 KO cellular phenotypes, including dendritic arborization, and spine density, while PTEN protein expression was significantly increased in Fmr1 KO animals. However, complete deletion of both Pten and Fmr1 resulted in a dramatic increase in dendritic length, spine density, and spine length. In addition, overexpression of PTEN in Fmr1 KO Pten heterozygous background reduced dendritic length, arborization, spine density, and spine length including pS6 levels. Our findings suggest that PTEN levels are negatively regulated by FMRP, and some Fmr1 KO phenotypes are caused by dysregulation of PTEN protein. These observations provide evidence for the genetic interaction of PTEN and FMRP and a possible mechanistic basis for the pathogenesis of Fmr1-related fragile X neurodevelopmental disorders.
Collapse
Affiliation(s)
| | - Jasmine A. Saunders
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, NH 03755
| | - Jacob Slaughter
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, NH 03755
| | - Kamran Tariq
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, NH 03755
| | - Rajarshi Chakrabarti
- Department of Biochemistry and Cellular Biology, Geisel School of Medicine at Dartmouth, Hanover, NH 03755
| | - Madhumala K. Sadanandappa
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, NH 03755
| | - Bryan W. Luikart
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, NH 03755
| | - Giovanni Bosco
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, NH 03755
| |
Collapse
|
28
|
Brain Cholesterol Biosynthetic Pathway Is Altered in a Preclinical Model of Fragile X Syndrome. Int J Mol Sci 2022; 23:ijms23063408. [PMID: 35328827 PMCID: PMC8955806 DOI: 10.3390/ijms23063408] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 03/08/2022] [Accepted: 03/18/2022] [Indexed: 12/02/2022] Open
Abstract
Fragile X Syndrome (FXS) is the most frequent form of inherited X-linked pathology, associated with an intellectual and developmental disability, and currently considered the first monogenic cause of autism spectrum disorder (ASD). Low levels of total cholesterol reported in the serum of FXS patients, and evidence that FMRP targets a subset of mRNAs encoding proteins of lipid synthesis and transport suggests that the cholesterol metabolism impairments could be involved in FXS. Thus, the aim of the presented work was to investigate the modulations of the cholesterol biosynthetic pathway and its end-products in a recently developed Fmr1-Δexon 8 rat model of FXS. Here, we show that this experimental model mimics what is found in FXS patients, exhibiting a lower serum cholesterol content, accompanied by a reduction in food intake and body weight compared to WT animals. Moreover, alterations of proteins committed to cholesterol synthesis and uptake have been observed in the amygdala, prefrontal cortex and nucleus accumbens. Interestingly, the end-products show a brain region-dependent modulation in Fmr1-Δexon 8 rats. Overall, our results demonstrate that the cholesterol biosynthetic pathway is altered in some brain regions of this preclinical model of FXS. This finding has relevance for future studies to delve deeper into the involvement of this metabolic process in FXS, and thus its possible role as a therapeutic target.
Collapse
|
29
|
Apgar TL, Sanders CR. Compendium of causative genes and their encoded proteins for common monogenic disorders. Protein Sci 2022; 31:75-91. [PMID: 34515378 PMCID: PMC8740837 DOI: 10.1002/pro.4183] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 08/29/2021] [Accepted: 08/31/2021] [Indexed: 01/19/2023]
Abstract
A compendium is presented of inherited monogenic disorders that have a prevalence of >1:20,000 in the human population, along with their causative genes and encoded proteins. "Simple" monogenic diseases are those for which the clinical features are caused by mutations impacting a single gene, usually in a manner that alters the sequence of the encoded protein. Of course, for a given "monogenic disorder", there is sometimes more than one potential disease gene, mutations in any one of which is sufficient to cause phenotypes of that disorder. Disease-causing mutations for monogenic disorders are usually passed on from generation to generation in a Mendelian fashion, and originate from spontaneous (de novo) germline founder mutations. In the past monogenic disorders have often been written off as targets for drug discovery because they sometimes are assumed to be rare disorders, for which the meager projected financial payoff of drug discovery and development has discouraged investment. However, not all monogenic diseases are rare. Here, we report that that currently available data identifies 72 disorders with a prevalence of at least 1 in 20,000 humans. For each, we tabulate the gene(s) for which mutations cause the spectrum of phenotypes associated with that disorder. We also identify the gene and protein that most commonly causes each disease. 34 of these disorders are caused exclusively by mutations in only a single gene and encoded protein.
Collapse
Affiliation(s)
- Tucker L. Apgar
- Department of Biochemistry and Center for Structural BiologyVanderbilt University School of Medicine Basic SciencesNashvilleTennesseeUSA
| | - Charles R. Sanders
- Department of Biochemistry and Center for Structural BiologyVanderbilt University School of Medicine Basic SciencesNashvilleTennesseeUSA
| |
Collapse
|
30
|
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
|
31
|
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
|
32
|
D'Antoni S, de Bari L, Valenti D, Borro M, Bonaccorso CM, Simmaco M, Vacca RA, Catania MV. Aberrant mitochondrial bioenergetics in the cerebral cortex of the Fmr1 knockout mouse model of fragile X syndrome. Biol Chem 2021; 401:497-503. [PMID: 31702995 DOI: 10.1515/hsz-2019-0221] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Accepted: 10/14/2019] [Indexed: 12/12/2022]
Abstract
Impaired energy metabolism may play a role in the pathogenesis of neurodevelopmental disorders including fragile X syndrome (FXS). We checked brain energy status and some aspects of cell bioenergetics, namely the activity of key glycolytic enzymes, glycerol-3-phosphate shuttle and mitochondrial respiratory chain (MRC) complexes, in the cerebral cortex of the Fmr1 knockout (KO) mouse model of FXS. We found that, despite a hyperactivation of MRC complexes, adenosine triphosphate (ATP) production via mitochondrial oxidative phosphorylation (OXPHOS) is compromised, resulting in brain energy impairment in juvenile and late-adult Fmr1 KO mice. Thus, an altered mitochondrial energy metabolism may contribute to neurological impairment in FXS.
Collapse
Affiliation(s)
- Simona D'Antoni
- Institute for Biomedical Research and Innovation (IRIB), National Research Council (CNR), Via Paolo Gaifami 18, I-95126 Catania, Italy
| | - Lidia de Bari
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies (IBIOM), CNR, Via Giovanni Amendola 165/A, I-70126 Bari, Italy
| | - Daniela Valenti
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies (IBIOM), CNR, Via Giovanni Amendola 165/A, I-70126 Bari, Italy
| | - Marina Borro
- Department of Neurosciences, Mental Health and Sensory Organs (NESMOS), Sapienza University of Rome, Via di Grottarossa 1035, I-00189 Rome, Italy
| | | | - Maurizio Simmaco
- Department of Neurosciences, Mental Health and Sensory Organs (NESMOS), Sapienza University of Rome, Via di Grottarossa 1035, I-00189 Rome, Italy
| | - Rosa Anna Vacca
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies (IBIOM), CNR, Via Giovanni Amendola 165/A, I-70126 Bari, Italy
| | - Maria Vincenza Catania
- Institute for Biomedical Research and Innovation (IRIB), National Research Council (CNR), Via Paolo Gaifami 18, I-95126 Catania, Italy.,Oasi Research Institute - IRCCS, Via Conte Ruggero 73, I-94018 Troina, Italy
| |
Collapse
|
33
|
Nagano S, Araki T. Axonal Transport and Local Translation of mRNA in Neurodegenerative Diseases. Front Mol Neurosci 2021; 14:697973. [PMID: 34194300 PMCID: PMC8236635 DOI: 10.3389/fnmol.2021.697973] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 05/25/2021] [Indexed: 12/13/2022] Open
Abstract
Since neurons have long neurites including axons, it is crucial for the axons to transport many intracellular substances such as proteins and mitochondria in order to maintain their morphology and function. In addition, mRNAs have also been shown to be transported within axons. RNA-binding proteins form complexes with mRNAs, and regulate transport of the mRNAs to axons, as well as locally translate them into proteins. Local translation of mRNAs actively occurs during the development and damage of neurons, and plays an important role in axon elongation, regeneration, and synapse formation. In recent years, it has been reported that impaired axonal transport and local translation of mRNAs may be involved in the pathogenesis of some neurodegenerative diseases. In this review, we discuss the significance of mRNA axonal transport and their local translation in amyotrophic lateral sclerosis/frontotemporal dementia, spinal muscular atrophy, Alzheimer’s disease, and fragile X syndrome.
Collapse
Affiliation(s)
- Seiichi Nagano
- Department of Neurotherapeutics, Osaka University Graduate School of Medicine, Osaka, Japan.,Department of Peripheral Nervous System Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Toshiyuki Araki
- Department of Peripheral Nervous System Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan
| |
Collapse
|
34
|
Lee J, Avramets D, Jeon B, Choo H. Modulation of Serotonin Receptors in Neurodevelopmental Disorders: Focus on 5-HT7 Receptor. Molecules 2021; 26:molecules26113348. [PMID: 34199418 PMCID: PMC8199608 DOI: 10.3390/molecules26113348] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 05/31/2021] [Accepted: 05/31/2021] [Indexed: 12/17/2022] Open
Abstract
Since neurodevelopmental disorders (NDDs) influence more than 3% of children worldwide, there has been intense investigation to understand the etiology of disorders and develop treatments. Although there are drugs such as aripiprazole, risperidone, and lurasidone, these medications are not cures for the disorders and can only help people feel better or alleviate their symptoms. Thus, it is required to discover therapeutic targets in order to find the ultimate treatments of neurodevelopmental disorders. It is suggested that abnormal neuronal morphology in the neurodevelopment process is a main cause of NDDs, in which the serotonergic system is emerging as playing a crucial role. From this point of view, we noticed the correlation between serotonin receptor subtype 7 (5-HT7R) and NDDs including autism spectrum disorder (ASD), fragile X syndrome (FXS), and Rett syndrome (RTT). 5-HT7R modulators improved altered behaviors in animal models and also affected neuronal morphology via the 5-HT7R/G12 signaling pathway. Through the investigation of recent studies, it is suggested that 5-HT7R could be a potential therapeutic target for the treatment of NDDs.
Collapse
Affiliation(s)
- Jieon Lee
- Brain Science Institute, Korea Institute of Science and Technology, Seongbuk-gu, Seoul 02792, Korea; (J.L.); (D.A.)
- Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology, Seoul 02792, Korea
| | - Diana Avramets
- Brain Science Institute, Korea Institute of Science and Technology, Seongbuk-gu, Seoul 02792, Korea; (J.L.); (D.A.)
- Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology, Seoul 02792, Korea
| | - Byungsun Jeon
- Brain Science Institute, Korea Institute of Science and Technology, Seongbuk-gu, Seoul 02792, Korea; (J.L.); (D.A.)
- Correspondence: (B.J.); (H.C.); Tel.: +82-2-958-5191 (B.J.); +82-2-958-5157 (H.C.)
| | - Hyunah Choo
- Brain Science Institute, Korea Institute of Science and Technology, Seongbuk-gu, Seoul 02792, Korea; (J.L.); (D.A.)
- Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology, Seoul 02792, Korea
- Correspondence: (B.J.); (H.C.); Tel.: +82-2-958-5191 (B.J.); +82-2-958-5157 (H.C.)
| |
Collapse
|
35
|
Shen Z, Liu B, Wu B, Zhou H, Wang X, Cao J, Jiang M, Zhou Y, Guo F, Xue C, Wu ZS. FMRP regulates STAT3 mRNA localization to cellular protrusions and local translation to promote hepatocellular carcinoma metastasis. Commun Biol 2021; 4:540. [PMID: 33972660 PMCID: PMC8110961 DOI: 10.1038/s42003-021-02071-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2019] [Accepted: 03/10/2021] [Indexed: 12/21/2022] Open
Abstract
Most hepatocellular carcinoma (HCC)-associated mortalities are related to the metastasis of cancer cells. The localization of mRNAs and their products to cell protrusions has been reported to play a crucial role in the metastasis. Our previous findings demonstrated that STAT3 mRNA accumulated in the protrusions of metastatic HCC cells. However, the underlying mechanism and functional significance of this localization of STAT3 mRNA has remained unexplored. Here we show that fragile X mental retardation protein (FMRP) modulates the localization and translation of STAT3 mRNA, accelerating HCC metastasis. The results of molecular analyses reveal that the 3′UTR of STAT3 mRNA is responsible for the localization of STAT3 mRNA to cell protrusions. FMRP is able to interact with the 3′UTR of STAT3 mRNA and facilitates its localization to protrusions. Importantly, FMRP could promote the IL-6-mediated translation of STAT3, and serine 114 of FMRP is identified as a potential phosphorylation site required for IL-6-mediated STAT3 translation. Furthermore, FMRP is highly expressed in HCC tissues and FMRP knockdown efficiently suppresses HCC metastasis in vitro and in vivo. Collectively, our findings provide further insights into the mechanism of HCC metastasis associated with the regulation of STAT3 mRNA localization and translation. Shen et al. propose a mechanism for the metastasis of hepatocellular carcinoma (HCC) cells through the localization and translation modulation of the STAT3 oncogene by fragile X mental retardation protein (FMRP). To this end, the authors also find that FMRP knockdown efficiently suppresses HCC metastasis in vitro and in vivo.
Collapse
Affiliation(s)
- Zhifa Shen
- Key Laboratory of Laboratory Medicine, Ministry of Education of China, and Zhejiang Provincial Key Laboratory of Medical Genetics, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China. .,Cancer Metastasis Alert and Prevention Center, Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, National & Local Joint Biomedical Engineering Research Center on Photodynamic Technologies, State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou, China.
| | - Bowen Liu
- Research Center for Molecular Oncology and Functional Nucleic Acids, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, Henan, China
| | - Biting Wu
- Key Laboratory of Laboratory Medicine, Ministry of Education of China, and Zhejiang Provincial Key Laboratory of Medical Genetics, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China
| | - Hongyin Zhou
- Key Laboratory of Laboratory Medicine, Ministry of Education of China, and Zhejiang Provincial Key Laboratory of Medical Genetics, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China
| | - Xiangyun Wang
- Research Center for Molecular Oncology and Functional Nucleic Acids, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, Henan, China
| | - Jinling Cao
- Research Center for Molecular Oncology and Functional Nucleic Acids, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, Henan, China
| | - Min Jiang
- Key Laboratory of Laboratory Medicine, Ministry of Education of China, and Zhejiang Provincial Key Laboratory of Medical Genetics, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China
| | - Yingying Zhou
- Key Laboratory of Laboratory Medicine, Ministry of Education of China, and Zhejiang Provincial Key Laboratory of Medical Genetics, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China
| | - Feixia Guo
- Key Laboratory of Laboratory Medicine, Ministry of Education of China, and Zhejiang Provincial Key Laboratory of Medical Genetics, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China
| | - Chang Xue
- Cancer Metastasis Alert and Prevention Center, Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, National & Local Joint Biomedical Engineering Research Center on Photodynamic Technologies, State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou, China
| | - Zai-Sheng Wu
- Cancer Metastasis Alert and Prevention Center, Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, National & Local Joint Biomedical Engineering Research Center on Photodynamic Technologies, State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou, China.
| |
Collapse
|
36
|
Di Marco B, Dell'Albani P, D'Antoni S, Spatuzza M, Bonaccorso CM, Musumeci SA, Drago F, Bardoni B, Catania MV. Fragile X mental retardation protein (FMRP) and metabotropic glutamate receptor subtype 5 (mGlu5) control stress granule formation in astrocytes. Neurobiol Dis 2021; 154:105338. [PMID: 33775821 DOI: 10.1016/j.nbd.2021.105338] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 03/12/2021] [Accepted: 03/17/2021] [Indexed: 12/18/2022] Open
Abstract
Fragile X syndrome (FXS) is a common form of intellectual disability and autism caused by the lack of Fragile X Mental Retardation Protein (FMRP), an RNA-binding protein involved in RNA transport and protein synthesis. Upon cellular stress, global protein synthesis is blocked and mRNAs are recruited into stress granules (SGs), together with RNA-binding proteins including FMRP. Activation of group-I metabotropic glutamate (mGlu) receptors stimulates FMRP-mediated mRNA transport and protein synthesis, but their role in SGs formation is unexplored. To this aim, we pre-treated wild type (WT) and Fmr1 knockout (KO) cultured astrocytes with the group-I-mGlu receptor agonist (S)-3,5-Dihydroxyphenylglycine (DHPG) and exposed them to sodium arsenite (NaAsO2), a widely used inducer of SGs formation. In WT cultures the activation of group-I mGlu receptors reduced SGs formation and recruitment of FMRP into SGs, and also attenuated phosphorylation of eIF2α, a key event crucially involved in SGs formation and inhibition of protein synthesis. In contrast, Fmr1 KO astrocytes, which exhibited a lower number of SGs than WT astrocytes, did not respond to agonist stimulation. Interestingly, the mGlu5 receptor negative allosteric modulator (NAM) 2-methyl-6-(phenylethynyl)pyridine (MPEP) antagonized DHPG-mediated SGs reduction in WT and reversed SGs formation in Fmr1 KO cultures. Our findings reveal a novel function of mGlu5 receptor as modulator of SGs formation and open new perspectives for understanding cellular response to stress in FXS pathophysiology.
Collapse
Affiliation(s)
- B Di Marco
- Institute for Biomedical Research and Innovation - The National Research Council of Italy (IRIB-CNR), Catania, Italy
| | - P Dell'Albani
- Institute for Biomedical Research and Innovation - The National Research Council of Italy (IRIB-CNR), Catania, Italy
| | - S D'Antoni
- Institute for Biomedical Research and Innovation - The National Research Council of Italy (IRIB-CNR), Catania, Italy
| | - M Spatuzza
- Oasi Research Institute - IRCCS, Troina, Italy
| | | | | | - F Drago
- Department of Biomedical and Biotecnological Sciences, University of Catania, Italy
| | - B Bardoni
- Université Côte d'Azur, Inserm, CNRS UMR7275, Institute of Molecular and Cellular Pharmacology, Valbonne 06560, France
| | - M V Catania
- Institute for Biomedical Research and Innovation - The National Research Council of Italy (IRIB-CNR), Catania, Italy; Oasi Research Institute - IRCCS, Troina, Italy.
| |
Collapse
|
37
|
Prieto M, Folci A, Poupon G, Schiavi S, Buzzelli V, Pronot M, François U, Pousinha P, Lattuada N, Abelanet S, Castagnola S, Chafai M, Khayachi A, Gwizdek C, Brau F, Deval E, Francolini M, Bardoni B, Humeau Y, Trezza V, Martin S. Missense mutation of Fmr1 results in impaired AMPAR-mediated plasticity and socio-cognitive deficits in mice. Nat Commun 2021; 12:1557. [PMID: 33692361 PMCID: PMC7946954 DOI: 10.1038/s41467-021-21820-1] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 02/16/2021] [Indexed: 11/22/2022] Open
Abstract
Fragile X syndrome (FXS) is the most frequent form of inherited intellectual disability and the best-described monogenic cause of autism. CGG-repeat expansion in the FMR1 gene leads to FMR1 silencing, loss-of-expression of the Fragile X Mental Retardation Protein (FMRP), and is a common cause of FXS. Missense mutations in the FMR1 gene were also identified in FXS patients, including the recurrent FMRP-R138Q mutation. To investigate the mechanisms underlying FXS caused by this mutation, we generated a knock-in mouse model (Fmr1R138Q) expressing the FMRP-R138Q protein. We demonstrate that, in the hippocampus of the Fmr1R138Q mice, neurons show an increased spine density associated with synaptic ultrastructural defects and increased AMPA receptor-surface expression. Combining biochemical assays, high-resolution imaging, electrophysiological recordings, and behavioural testing, we also show that the R138Q mutation results in impaired hippocampal long-term potentiation and socio-cognitive deficits in mice. These findings reveal the functional impact of the FMRP-R138Q mutation in a mouse model of FXS.
Collapse
Affiliation(s)
- Marta Prieto
- Université Côte d'Azur, CNRS, IPMC, Valbonne, France
| | | | | | | | | | - Marie Pronot
- Université Côte d'Azur, CNRS, IPMC, Valbonne, France
| | | | | | - Norma Lattuada
- Università degli Studi di Milano, Dept. of Medical Biotechnology and Translational Medicine, Milan, Italy
| | | | | | - Magda Chafai
- Université Côte d'Azur, CNRS, IPMC, Valbonne, France
| | | | | | - Frédéric Brau
- Université Côte d'Azur, CNRS, IPMC, Valbonne, France
| | | | - Maura Francolini
- Università degli Studi di Milano, Dept. of Medical Biotechnology and Translational Medicine, Milan, Italy
| | - Barbara Bardoni
- Université Côte d'Azur, Inserm, CNRS, IPMC, Valbonne, France
| | - Yann Humeau
- University of Bordeaux, CNRS, IINS, Bordeaux, France
| | | | - Stéphane Martin
- Université Côte d'Azur, Inserm, CNRS, IPMC, Valbonne, France.
| |
Collapse
|
38
|
Worpenberg L, Paolantoni C, Longhi S, Mulorz MM, Lence T, Wessels HH, Dassi E, Aiello G, Sutandy FXR, Scheibe M, Edupuganti RR, Busch A, Möckel MM, Vermeulen M, Butter F, König J, Notarangelo M, Ohler U, Dieterich C, Quattrone A, Soldano A, Roignant JY. Ythdf is a N6-methyladenosine reader that modulates Fmr1 target mRNA selection and restricts axonal growth in Drosophila. EMBO J 2021; 40:e104975. [PMID: 33428246 PMCID: PMC7883056 DOI: 10.15252/embj.2020104975] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 11/18/2020] [Accepted: 11/30/2020] [Indexed: 12/19/2022] Open
Abstract
N6‐methyladenosine (m6A) regulates a variety of physiological processes through modulation of RNA metabolism. This modification is particularly enriched in the nervous system of several species, and its dysregulation has been associated with neurodevelopmental defects and neural dysfunctions. In Drosophila, loss of m6A alters fly behavior, albeit the underlying molecular mechanism and the role of m6A during nervous system development have remained elusive. Here we find that impairment of the m6A pathway leads to axonal overgrowth and misguidance at larval neuromuscular junctions as well as in the adult mushroom bodies. We identify Ythdf as the main m6A reader in the nervous system, being required to limit axonal growth. Mechanistically, we show that the m6A reader Ythdf directly interacts with Fmr1, the fly homolog of Fragile X mental retardation RNA binding protein (FMRP), to inhibit the translation of key transcripts involved in axonal growth regulation. Altogether, this study demonstrates that the m6A pathway controls development of the nervous system and modulates Fmr1 target transcript selection.
Collapse
Affiliation(s)
- Lina Worpenberg
- Center for Integrative Genomics, Génopode Building, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Chiara Paolantoni
- Center for Integrative Genomics, Génopode Building, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Sara Longhi
- Laboratory of Translational Genomics, Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Trento, Italy
| | | | - Tina Lence
- Institute of Molecular Biology (IMB), Mainz, Germany
| | - Hans-Hermann Wessels
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin Institute for Medical Systems Biology (BIMSB), Berlin, Germany.,Department of Biology, Humboldt University Berlin, Berlin, Germany
| | - Erik Dassi
- Laboratory of RNA Regulatory Networks, Department CIBIO, University of Trento, Trento, Italy
| | - Giuseppe Aiello
- Armenise-Harvard Laboratory of Brain Disorders and Cancer, Department CIBIO, University of Trento, Trento, Italy
| | | | | | - Raghu R Edupuganti
- Department of Molecular Biology, Faculty of Science, Radboud Institute for Molecular Life Sciences, Oncode Institute, Radboud University Nijmegen, Nijmegen, The Netherlands
| | - Anke Busch
- Bioinformatics Core Facility, IMB, Mainz, Germany
| | | | - Michiel Vermeulen
- Department of Molecular Biology, Faculty of Science, Radboud Institute for Molecular Life Sciences, Oncode Institute, Radboud University Nijmegen, Nijmegen, The Netherlands
| | - Falk Butter
- Institute of Molecular Biology (IMB), Mainz, Germany
| | - Julian König
- Institute of Molecular Biology (IMB), Mainz, Germany
| | - Michela Notarangelo
- Laboratory of Translational Genomics, Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Trento, Italy
| | - Uwe Ohler
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin Institute for Medical Systems Biology (BIMSB), Berlin, Germany.,Department of Biology, Humboldt University Berlin, Berlin, Germany
| | - Christoph Dieterich
- Klaus Tschira Institute for Integrative Computational Cardiology and Department of Internal Medicine III, University Hospital Heidelberg, Heidelberg, Germany.,German Center for Cardiovascular Research (DZHK), Partner site Heidelberg-Mannheim, Heidelberg, Germany
| | - Alessandro Quattrone
- Center for Integrative Genomics, Génopode Building, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland.,Laboratory of Translational Genomics, Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Trento, Italy
| | - Alessia Soldano
- Laboratory of Translational Genomics, Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Trento, Italy
| | - Jean-Yves Roignant
- Center for Integrative Genomics, Génopode Building, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland.,Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg-University Mainz, Mainz, Germany
| |
Collapse
|
39
|
Clifton NE, Thomas KL, Wilkinson LS, Hall J, Trent S. FMRP and CYFIP1 at the Synapse and Their Role in Psychiatric Vulnerability. Complex Psychiatry 2020; 6:5-19. [PMID: 34883502 PMCID: PMC7673588 DOI: 10.1159/000506858] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Accepted: 02/27/2020] [Indexed: 12/23/2022] Open
Abstract
There is increasing awareness of the role genetic risk variants have in mediating vulnerability to psychiatric disorders such as schizophrenia and autism. Many of these risk variants encode synaptic proteins, influencing biological pathways of the postsynaptic density and, ultimately, synaptic plasticity. Fragile-X mental retardation 1 (FMR1) and cytoplasmic fragile-X mental retardation protein (FMRP)-interacting protein 1 (CYFIP1) contain 2 such examples of highly penetrant risk variants and encode synaptic proteins with shared functional significance. In this review, we discuss the biological actions of FMRP and CYFIP1, including their regulation of (i) protein synthesis and specifically FMRP targets, (ii) dendritic and spine morphology, and (iii) forms of synaptic plasticity such as long-term depression. We draw upon a range of preclinical studies that have used genetic dosage models of FMR1 and CYFIP1 to determine their biological function. In parallel, we discuss how clinical studies of fragile X syndrome or 15q11.2 deletion patients have informed our understanding of FMRP and CYFIP1, and highlight the latest psychiatric genomic findings that continue to implicate FMRP and CYFIP1. Lastly, we assess the current limitations in our understanding of FMRP and CYFIP1 biology and how they must be addressed before mechanism-led therapeutic strategies can be developed for psychiatric disorders.
Collapse
Affiliation(s)
- Nicholas E. Clifton
- Neuroscience & Mental Health Research Institute, Cardiff University, Cardiff, United Kingdom
- MRC Centre for Neuropsychiatric Genetics and Genomics, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Kerrie L. Thomas
- Neuroscience & Mental Health Research Institute, Cardiff University, Cardiff, United Kingdom
- School of Biosciences, Cardiff University, Cardiff, United Kingdom
| | - Lawrence S. Wilkinson
- Neuroscience & Mental Health Research Institute, Cardiff University, Cardiff, United Kingdom
- School of Psychology, Cardiff University, Cardiff, United Kingdom
| | - Jeremy Hall
- Neuroscience & Mental Health Research Institute, Cardiff University, Cardiff, United Kingdom
- MRC Centre for Neuropsychiatric Genetics and Genomics, School of Medicine, Cardiff University, Cardiff, United Kingdom
- Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Simon Trent
- Neuroscience & Mental Health Research Institute, Cardiff University, Cardiff, United Kingdom
- School of Life Sciences, Faculty of Natural Sciences, Keele University, Keele, United Kingdom
| |
Collapse
|
40
|
Majumder M, Johnson RH, Palanisamy V. Fragile X-related protein family: a double-edged sword in neurodevelopmental disorders and cancer. Crit Rev Biochem Mol Biol 2020; 55:409-424. [PMID: 32878499 DOI: 10.1080/10409238.2020.1810621] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The fragile X-related (FXR) family proteins FMRP, FXR1, and FXR2 are RNA binding proteins that play a critical role in RNA metabolism, neuronal plasticity, and muscle development. These proteins share significant homology in their protein domains, which are functionally and structurally similar to each other. FXR family members are known to play an essential role in causing fragile X mental retardation syndrome (FXS), the most common genetic form of autism spectrum disorder. Recent advances in our understanding of this family of proteins have occurred in tandem with discoveries of great importance to neurological disorders and cancer biology via the identification of their novel RNA and protein targets. Herein, we review the FXR family of proteins as they pertain to FXS, other mental illnesses, and cancer. We emphasize recent findings and analyses that suggest contrasting functions of this protein family in FXS and tumorigenesis based on their expression patterns in human tissues. Finally, we discuss current gaps in our knowledge regarding the FXR protein family and their role in FXS and cancer and suggest future studies to facilitate bench to bedside translation of the findings.
Collapse
Affiliation(s)
- Mrinmoyee Majumder
- Department of Biochemistry and Molecular Biology, School of Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - Roger H Johnson
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, USA
| | - Viswanathan Palanisamy
- Department of Biochemistry and Molecular Biology, School of Medicine, Medical University of South Carolina, Charleston, SC, USA
| |
Collapse
|
41
|
Maurin T, Melancia F, Jarjat M, Castro L, Costa L, Delhaye S, Khayachi A, Castagnola S, Mota E, Di Giorgio A, Servadio M, Drozd M, Poupon G, Schiavi S, Sardone L, Azoulay S, Ciranna L, Martin S, Vincent P, Trezza V, Bardoni B. Involvement of Phosphodiesterase 2A Activity in the Pathophysiology of Fragile X Syndrome. Cereb Cortex 2020; 29:3241-3252. [PMID: 30137253 DOI: 10.1093/cercor/bhy192] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 07/12/2018] [Accepted: 07/20/2018] [Indexed: 12/26/2022] Open
Abstract
The fragile X mental retardation protein (FMRP) is an RNA-binding protein involved in translational regulation of mRNAs that play key roles in synaptic morphology and plasticity. The functional absence of FMRP causes the fragile X syndrome (FXS), the most common form of inherited intellectual disability and the most common monogenic cause of autism. No effective treatment is available for FXS. We recently identified the Phosphodiesterase 2A (Pde2a) mRNA as a prominent target of FMRP. PDE2A enzymatic activity is increased in the brain of Fmr1-KO mice, a recognized model of FXS, leading to decreased levels of cAMP and cGMP. Here, we pharmacologically inhibited PDE2A in Fmr1-KO mice and observed a rescue both of the maturity of dendritic spines and of the exaggerated hippocampal mGluR-dependent long-term depression. Remarkably, PDE2A blockade rescued the social and communicative deficits of both mouse and rat Fmr1-KO animals. Importantly, chronic inhibition of PDE2A in newborn Fmr1-KO mice followed by a washout interval, resulted in the rescue of the altered social behavior observed in adolescent mice. Altogether, these results reveal the key role of PDE2A in the physiopathology of FXS and suggest that its pharmacological inhibition represents a novel therapeutic approach for FXS.
Collapse
Affiliation(s)
- Thomas Maurin
- Université Côte d'Azur, CNRS, IPMC, Valbonne, France.,CNRS LIA «Neogenex», Valbonne, France
| | | | - Marielle Jarjat
- Université Côte d'Azur, CNRS, IPMC, Valbonne, France.,CNRS LIA «Neogenex», Valbonne, France
| | - Liliana Castro
- Sorbonne Université, CNRS, Biological Adaptation and Ageing, Paris, France.,Labex BioPsy, Paris, France
| | - Lara Costa
- Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| | - Sébastien Delhaye
- Université Côte d'Azur, CNRS, IPMC, Valbonne, France.,CNRS LIA «Neogenex», Valbonne, France
| | | | - Sara Castagnola
- Université Côte d'Azur, CNRS, IPMC, Valbonne, France.,CNRS LIA «Neogenex», Valbonne, France
| | - Elia Mota
- Sorbonne Université, CNRS, Biological Adaptation and Ageing, Paris, France.,Labex BioPsy, Paris, France
| | - Audrey Di Giorgio
- Université Côte d'Azur, CNRS, Institut de Chimie de Nice, Nice, France
| | | | - Malgorzata Drozd
- Université Côte d'Azur, CNRS, IPMC, Valbonne, France.,CNRS LIA «Neogenex», Valbonne, France
| | | | - Sara Schiavi
- Department of Sciences, Università RomaTre, Roma, Italy
| | - Lara Sardone
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Stéphane Azoulay
- Université Côte d'Azur, CNRS, Institut de Chimie de Nice, Nice, France
| | - Lucia Ciranna
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Stéphane Martin
- Université Côte d'Azur, INSERM, CNRS, IPMC, Valbonne, France
| | - Pierre Vincent
- Sorbonne Université, CNRS, Biological Adaptation and Ageing, Paris, France.,Labex BioPsy, Paris, France
| | | | - Barbara Bardoni
- CNRS LIA «Neogenex», Valbonne, France.,Université Côte d'Azur, INSERM, CNRS, IPMC, Valbonne, France
| |
Collapse
|
42
|
Yu X, Wang X, Sakano H, Zorio DAR, Wang Y. Dynamics of the fragile X mental retardation protein correlates with cellular and synaptic properties in primary auditory neurons following afferent deprivation. J Comp Neurol 2020; 529:481-500. [PMID: 32449186 DOI: 10.1002/cne.24959] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 04/26/2020] [Accepted: 05/14/2020] [Indexed: 01/01/2023]
Abstract
Afferent activity dynamically regulates neuronal properties and connectivity in the central nervous system. The Fragile X mental retardation protein (FMRP) is an RNA-binding protein that regulates cellular and synaptic properties in an activity-dependent manner. Whether and how FMRP level and localization are regulated by afferent input remains sparsely examined and how such regulation is associated with neuronal response to changes in sensory input is unknown. We characterized changes in FMRP level and localization in the chicken nucleus magnocellularis (NM), a primary cochlear nucleus, following afferent deprivation by unilateral cochlea removal. We observed rapid (within 2 hr) aggregation of FMRP immunoreactivity into large granular structures in a subset of deafferented NM neurons. Neurons that exhibited persistent FMRP aggregation at 12-24 hr eventually lost cytoplasmic Nissl substance, indicating cell death. A week later, FMRP expression in surviving neurons regained its homeostasis, with a slightly reduced immunostaining intensity and enhanced heterogeneity. Correlation analyses under the homeostatic status (7-14 days) revealed that neurons expressing relatively more FMRP had a higher capability of maintaining cell body size and ribosomal activity, as well as a better ability to detach inactive presynaptic terminals. Additionally, the intensity of an inhibitory postsynaptic protein, gephyrin, was reduced following deafferentation and was positively correlated with FMRP intensity, implicating an involvement of FMRP in synaptic dynamics in response to reduced afferent inputs. Collectively, this study demonstrates that afferent input regulates FMRP expression and localization in ways associated with multiple types of neuronal responses and synaptic rearrangements.
Collapse
Affiliation(s)
- Xiaoyan Yu
- Program in Neuroscience, Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, Florida, USA
| | - Xiaoyu Wang
- Program in Neuroscience, Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, Florida, USA.,Division of Histology & Embryology, Key Laboratory for Regenerative Medicine of the Ministry of Education, Medical College, Jinan University, Guangzhou, China
| | - Hitomi Sakano
- Department of Otolaryngology, Bloedel Hearing Research Center, University of Washington, Seattle, Washington, USA.,Department of Otolaryngology, University of Rochester, Rochester, New York, USA
| | - Diego A R Zorio
- Program in Neuroscience, Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, Florida, USA
| | - Yuan Wang
- Program in Neuroscience, Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, Florida, USA
| |
Collapse
|
43
|
Taha MS, Haghighi F, Stefanski A, Nakhaei-Rad S, Kazemein Jasemi NS, Al Kabbani MA, Görg B, Fujii M, Lang PA, Häussinger D, Piekorz RP, Stühler K, Ahmadian MR. Novel FMRP interaction networks linked to cellular stress. FEBS J 2020; 288:837-860. [PMID: 32525608 DOI: 10.1111/febs.15443] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 04/09/2020] [Accepted: 06/03/2020] [Indexed: 12/12/2022]
Abstract
Silencing of the fragile X mental retardation 1 (FMR1) gene and consequently lack of synthesis of FMR protein (FMRP) are associated with fragile X syndrome, which is one of the most prevalent inherited intellectual disabilities, with additional roles in increased viral infection, liver disease, and reduced cancer risk. FMRP plays critical roles in chromatin dynamics, RNA binding, mRNA transport, and mRNA translation. However, the underlying molecular mechanisms, including the (sub)cellular FMRP protein networks, remain elusive. Here, we employed affinity pull-down and quantitative LC-MS/MS analyses with FMRP. We identified known and novel candidate FMRP-binding proteins as well as protein complexes. FMRP interacted with 180 proteins, 28 of which interacted with its N terminus. Interaction with the C terminus of FMRP was observed for 102 proteins, and 48 proteins interacted with both termini. This FMRP interactome comprises known FMRP-binding proteins, including the ribosomal proteins FXR1P, NUFIP2, Caprin-1, and numerous novel FMRP candidate interacting proteins that localize to different subcellular compartments, including CARF, LARP1, LEO1, NOG2, G3BP1, NONO, NPM1, SKIP, SND1, SQSTM1, and TRIM28. Our data considerably expand the protein and RNA interaction networks of FMRP, which thereby suggest that, in addition to its known functions, FMRP participates in transcription, RNA metabolism, ribonucleoprotein stress granule formation, translation, DNA damage response, chromatin dynamics, cell cycle regulation, ribosome biogenesis, miRNA biogenesis, and mitochondrial organization. Thus, FMRP seems associated with multiple cellular processes both under normal and cell stress conditions in neuronal as well as non-neuronal cell types, as exemplified by its role in the formation of stress granules.
Collapse
Affiliation(s)
- Mohamed S Taha
- Institute of Biochemistry and Molecular Biology II, Medical Faculty of the Heinrich Heine University, Düsseldorf, Germany.,Research on Children with Special Needs Department, Medical Research Branch, National Research Centre, Cairo, Egypt
| | - Fereshteh Haghighi
- Institute of Biochemistry and Molecular Biology II, Medical Faculty of the Heinrich Heine University, Düsseldorf, Germany
| | - Anja Stefanski
- Molecular Proteomics Laboratory, Heinrich Heine-University, Düsseldorf, Germany
| | - Saeideh Nakhaei-Rad
- Institute of Biochemistry and Molecular Biology II, Medical Faculty of the Heinrich Heine University, Düsseldorf, Germany
| | - Neda S Kazemein Jasemi
- Institute of Biochemistry and Molecular Biology II, Medical Faculty of the Heinrich Heine University, Düsseldorf, Germany
| | - Mohamed Aghyad Al Kabbani
- Institute of Biochemistry and Molecular Biology II, Medical Faculty of the Heinrich Heine University, Düsseldorf, Germany
| | - Boris Görg
- Clinic of Gastroenterology, Hepatology and Infectious Diseases, Medical Faculty of the Heinrich Heine-University, Düsseldorf, Germany
| | - Masahiro Fujii
- Division of Virology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Phillip A Lang
- Department of Molecular Medicine II, Medical Faculty, Heinrich Heine-University, Düsseldorf, Germany
| | - Dieter Häussinger
- Clinic of Gastroenterology, Hepatology and Infectious Diseases, Medical Faculty of the Heinrich Heine-University, Düsseldorf, Germany
| | - Roland P Piekorz
- Institute of Biochemistry and Molecular Biology II, Medical Faculty of the Heinrich Heine University, Düsseldorf, Germany
| | - Kai Stühler
- Molecular Proteomics Laboratory, Heinrich Heine-University, Düsseldorf, Germany
| | - Mohammad R Ahmadian
- Institute of Biochemistry and Molecular Biology II, Medical Faculty of the Heinrich Heine University, Düsseldorf, Germany
| |
Collapse
|
44
|
Golden CEM, Breen MS, Koro L, Sonar S, Niblo K, Browne A, Burlant N, Di Marino D, De Rubeis S, Baxter MG, Buxbaum JD, Harony-Nicolas H. Deletion of the KH1 Domain of Fmr1 Leads to Transcriptional Alterations and Attentional Deficits in Rats. Cereb Cortex 2020; 29:2228-2244. [PMID: 30877790 PMCID: PMC6458915 DOI: 10.1093/cercor/bhz029] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Revised: 12/11/2018] [Accepted: 02/06/2019] [Indexed: 12/27/2022] Open
Abstract
Fragile X syndrome (FXS) is a neurodevelopmental disorder caused by mutations in the FMR1 gene. It is a leading monogenic cause of autism spectrum disorder and inherited intellectual disability and is often comorbid with attention deficits. Most FXS cases are due to an expansion of CGG repeats leading to suppressed expression of fragile X mental retardation protein (FMRP), an RNA-binding protein involved in mRNA metabolism. We found that the previously published Fmr1 knockout rat model of FXS expresses an Fmr1 transcript with an in-frame deletion of exon 8, which encodes for the K-homology (KH) RNA-binding domain, KH1. Notably, 3 pathogenic missense mutations associated with FXS lie in the KH domains. We observed that the deletion of exon 8 in rats leads to attention deficits and to alterations in transcriptional profiles within the medial prefrontal cortex (mPFC), which map to 2 weighted gene coexpression network modules. These modules are conserved in human frontal cortex and enriched for known FMRP targets. Hub genes in these modules represent potential therapeutic targets for FXS. Taken together, these findings indicate that attentional testing might be a reliable cross-species tool for investigating FXS and identify dysregulated conserved gene networks in a relevant brain region.
Collapse
Affiliation(s)
- Carla E M Golden
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Michael S Breen
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Lacin Koro
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sankalp Sonar
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kristi Niblo
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Andrew Browne
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Natalie Burlant
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Daniele Di Marino
- Faculty of Biomedical Sciences, Institute of Computational Science, Center for Computational Medicine in Cardiology, Università della Svizzera Italiana (USI), Lugano, Switzerland.,Department of Life and Environmental Sciences, Polytechnic University of Marche, Ancona, Italy
| | - Silvia De Rubeis
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Mark G Baxter
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Joseph D Buxbaum
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Hala Harony-Nicolas
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
45
|
Cascade Testing for Fragile X Syndrome in a Rural Setting in Cameroon (Sub-Saharan Africa). Genes (Basel) 2020; 11:genes11020136. [PMID: 32012997 PMCID: PMC7074341 DOI: 10.3390/genes11020136] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Revised: 01/13/2020] [Accepted: 01/22/2020] [Indexed: 11/17/2022] Open
Abstract
Fragile X Syndrome (FXS), an X-linked dominant monogenic condition, is the main genetic cause of intellectual disability (ID) and autism spectrum disorder (ASD). FXS is associated with an expansion of CGG repeat sequence in the Fragile X Mental Retardation gene 1 (FMR1) on chromosome X. Following a neuropediatric assessment of two male siblings who presented with signs of FXS that was confirmed with molecular testing, we provided cascade counselling and testing to the extended family. A total of 46 individuals were tested for FXS; among them, 58.70% (n = 27) were females. The mean age was 9.4 (±5) years for children and 45.9 (±15.9) years for adults. Pedigree analysis suggested that the founder of these families was likely a normal transmitting male. Four out of 19 males with clinical ID were confirmed to have a full mutation for FXS, while 14/27 females had a pathologic CGG expansion (>56 CGG repeats) on one of their X chromosomes. Two women with premature menopause were confirmed of being carriers of premutation (91 and 101 CGG repeats). We also identified maternal alleles (91 and 126 CGG repeats) which expanded to a full mutation in their offspring (>200 CGG repeats). This study is a rare report on FXS from Africa and illustrates the case scenario of implementing genetic medicine for a neurogenetic condition in a rural setting.
Collapse
|
46
|
Mei X, Yang Y, Zhao J, Wang Y, Chen Q, Qian X, Li X, Feng Z. Role of fragile X mental retardation protein in chronic pain. Mol Pain 2020; 16:1744806920928619. [PMID: 32496847 PMCID: PMC7273537 DOI: 10.1177/1744806920928619] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 04/08/2020] [Accepted: 04/08/2020] [Indexed: 11/15/2022] Open
Abstract
Chronic pain has detrimental effects on one's quality of life. However, its treatment options are very limited, and its underlying pathogenesis remains unclear. Recent research has suggested that fragile X mental retardation protein is involved in the development of chronic pain, making it a potential target for prevention and treatment. The current review of literature will examine the function of fragile X mental retardation protein and its associated pathways, through which we hope to gain insight into how fragile X mental retardation protein may contribute to nociceptive sensitization and chronic pain.
Collapse
Affiliation(s)
- Xiangyang Mei
- Department of Pain Medicine,
The First Affiliated Hospital,
Zhejiang
University School of Medicine,
Hangzhou, Zhejiang, China
| | - Yixin Yang
- Department of Pain Medicine,
The First Affiliated Hospital,
Zhejiang
University School of Medicine,
Hangzhou, Zhejiang, China
| | - Jinsong Zhao
- Department of Pain Medicine,
The First Affiliated Hospital,
Zhejiang
University School of Medicine,
Hangzhou, Zhejiang, China
| | - Yongjie Wang
- Institute of Neuroscience,
Key Laboratory of Medical Neurobiology of the Ministry of Health of
China, School of Medicine,
Zhejiang
University, Hangzhou,
Zhejiang, China
| | - QiLiang Chen
- Department of
Anesthesiology, Perioperative and Pain Medicine, Stanford Health Care,
Stanford
University, Stanford, CA,
USA
| | - Xiang Qian
- Department of
Anesthesiology, Perioperative and Pain Medicine, Stanford Health Care,
Stanford
University, Stanford, CA,
USA
| | - Xiangyao Li
- Institute of Neuroscience,
Key Laboratory of Medical Neurobiology of the Ministry of Health of
China, School of Medicine,
Zhejiang
University, Hangzhou,
Zhejiang, China
| | - Zhiying Feng
- Department of Pain Medicine,
The First Affiliated Hospital,
Zhejiang
University School of Medicine,
Hangzhou, Zhejiang, China
| |
Collapse
|
47
|
Cid-Samper F, Gelabert-Baldrich M, Lang B, Lorenzo-Gotor N, Ponti RD, Severijnen LAWFM, Bolognesi B, Gelpi E, Hukema RK, Botta-Orfila T, Tartaglia GG. An Integrative Study of Protein-RNA Condensates Identifies Scaffolding RNAs and Reveals Players in Fragile X-Associated Tremor/Ataxia Syndrome. Cell Rep 2019; 25:3422-3434.e7. [PMID: 30566867 PMCID: PMC6315285 DOI: 10.1016/j.celrep.2018.11.076] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Revised: 09/26/2018] [Accepted: 11/19/2018] [Indexed: 12/21/2022] Open
Abstract
Recent evidence indicates that specific RNAs promote the formation of ribonucleoprotein condensates by acting as scaffolds for RNA-binding proteins (RBPs). We systematically investigated RNA-RBP interaction networks to understand ribonucleoprotein assembly. We found that highly contacted RNAs are structured, have long UTRs, and contain nucleotide repeat expansions. Among the RNAs with such properties, we identified the FMR1 3' UTR that harbors CGG expansions implicated in fragile X-associated tremor/ataxia syndrome (FXTAS). We studied FMR1 binding partners in silico and in vitro and prioritized the splicing regulator TRA2A for further characterization. In a FXTAS cellular model, we validated the TRA2A-FMR1 interaction and investigated implications of its sequestration at both transcriptomic and post-transcriptomic levels. We found that TRA2A co-aggregates with FMR1 in a FXTAS mouse model and in post-mortem human samples. Our integrative study identifies key components of ribonucleoprotein aggregates, providing links to neurodegenerative disease and allowing the discovery of therapeutic targets.
Collapse
Affiliation(s)
- Fernando Cid-Samper
- Centre for Genomic Regulation (CRG), The Barcelona Institute for Science and Technology, Dr. Aiguader 88, 08003 Barcelona, Spain
| | - Mariona Gelabert-Baldrich
- Centre for Genomic Regulation (CRG), The Barcelona Institute for Science and Technology, Dr. Aiguader 88, 08003 Barcelona, Spain
| | - Benjamin Lang
- Centre for Genomic Regulation (CRG), The Barcelona Institute for Science and Technology, Dr. Aiguader 88, 08003 Barcelona, Spain
| | - Nieves Lorenzo-Gotor
- Centre for Genomic Regulation (CRG), The Barcelona Institute for Science and Technology, Dr. Aiguader 88, 08003 Barcelona, Spain
| | - Riccardo Delli Ponti
- Centre for Genomic Regulation (CRG), The Barcelona Institute for Science and Technology, Dr. Aiguader 88, 08003 Barcelona, Spain
| | | | - Benedetta Bolognesi
- Centre for Genomic Regulation (CRG), The Barcelona Institute for Science and Technology, Dr. Aiguader 88, 08003 Barcelona, Spain
| | - Ellen Gelpi
- Neurological Tissue Biobank of the Hospital Clinic and Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Carrer del Rosselló, 149, 08036, Barcelona, Spain; Institute of Neurology, Medical University of Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria
| | - Renate K Hukema
- Department of Clinical Genetics, Erasmus MC, 3000 CA Rotterdam, the Netherlands
| | - Teresa Botta-Orfila
- Centre for Genomic Regulation (CRG), The Barcelona Institute for Science and Technology, Dr. Aiguader 88, 08003 Barcelona, Spain.
| | - Gian Gaetano Tartaglia
- Centre for Genomic Regulation (CRG), The Barcelona Institute for Science and Technology, Dr. Aiguader 88, 08003 Barcelona, Spain; Universitat Pompeu Fabra (UPF), 08003 Barcelona, Spain; Department of Biology 'Charles Darwin', Sapienza University of Rome, P.le A. Moro 5, Rome 00185, Italy; Institució Catalana de Recerca i Estudis Avançats (ICREA), 23 Passeig Lluís Companys, 08010 Barcelona, Spain.
| |
Collapse
|
48
|
Ciranna L, Costa L. Pituitary Adenylate Cyclase-Activating Polypeptide Modulates Hippocampal Synaptic Transmission and Plasticity: New Therapeutic Suggestions for Fragile X Syndrome. Front Cell Neurosci 2019; 13:524. [PMID: 31827422 PMCID: PMC6890831 DOI: 10.3389/fncel.2019.00524] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Accepted: 11/08/2019] [Indexed: 12/13/2022] Open
Abstract
Pituitary adenylate cyclase-activating polypeptide (PACAP) modulates glutamatergic synaptic transmission and plasticity in the hippocampus, a brain area with a key role in learning and memory. In agreement, several studies have demonstrated that PACAP modulates learning in physiological conditions. Recent publications show reduced PACAP levels and/or alterations in PACAP receptor expression in different conditions associated with cognitive disability. It is noteworthy that PACAP administration rescued impaired synaptic plasticity and learning in animal models of aging, Alzheimer's disease, Parkinson's disease, and Huntington's chorea. In this context, results from our laboratory demonstrate that PACAP rescued metabotropic glutamate receptor-mediated synaptic plasticity in the hippocampus of a mouse model of fragile X syndrome (FXS), a genetic form of intellectual disability. PACAP is actively transported through the blood-brain barrier and reaches the brain following intranasal or intravenous administration. Besides, new studies have identified synthetic PACAP analog peptides with improved selectivity and pharmacokinetic properties with respect to the native peptide. Our review supports the shared idea that pharmacological activation of PACAP receptors might be beneficial for brain pathologies with cognitive disability. In addition, we suggest that the effects of PACAP treatment might be further studied as a possible therapy in FXS.
Collapse
Affiliation(s)
- Lucia Ciranna
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Lara Costa
- Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| |
Collapse
|
49
|
Medishetti R, Rani R, Kavati S, Mahilkar A, Akella V, Saxena U, Kulkarni P, Sevilimedu A. A DNAzyme based knockdown model for Fragile-X syndrome in zebrafish reveals a critical window for therapeutic intervention. J Pharmacol Toxicol Methods 2019; 101:106656. [PMID: 31734279 DOI: 10.1016/j.vascn.2019.106656] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Revised: 11/05/2019] [Accepted: 11/10/2019] [Indexed: 11/16/2022]
Abstract
INTRODUCTION FXS is the leading cause of intellectual disabilities in males and a major monogenic cause of ASD (Autism spectrum disorders). It occurs due to the loss of FMRP, whose role in early development is not well understood. In this study, we have used a novel DNAzyme based approach to create a larval model of FXS in zebrafish with specific focus on the early developmental window. METHODS Fmr1specific DNAzymes were electroporated into embryos to create the knockdown. Changes in RNA and protein levels of FMRP and relevant biomarkers were measured in the 0-7dpf window. Behavioral tests to measure anxiety, cognitive impairments and irritability in the larvae were conducted at the 7dpf stage. Drug treatment was carried out at various time points in the 0-7dpf window to identify the critical window for pharmacological intervention. RESULTS The DNAzyme based knockdown approach led to a significant knockdown of FMRP in the zebrafish embryos, accompanied by increased anxiety, irritability and cognitive impairments at 7dpf, thus creating a robust larval model of FXS. Treatment with the Mavoglurant was able to rescue the behavioral phenotypes in the FXS larvae, and found to be more efficacious in the 0-3dpf window. DISCUSSION The results from this study have revealed that a) a DNAzyme based knockdown approach can be used to create robust larval zebrafish model of disease, in a high-throughput manner and b) optimal window for therapeutic intervention for FXS as well as other pediatric diseases with a monogenic cause can be identified using such a model.
Collapse
Affiliation(s)
- Raghavender Medishetti
- Center for Innovation in Molecular and Pharmaceutical Sciences (CIMPS), Dr. Reddy's Institute of Life Sciences, University of Hyderabad Campus, Gachibowli, Hyderabad, Telangana, 500046, India
| | - Rita Rani
- Center for Innovation in Molecular and Pharmaceutical Sciences (CIMPS), Dr. Reddy's Institute of Life Sciences, University of Hyderabad Campus, Gachibowli, Hyderabad, Telangana, 500046, India
| | - Srinivas Kavati
- Center for Innovation in Molecular and Pharmaceutical Sciences (CIMPS), Dr. Reddy's Institute of Life Sciences, University of Hyderabad Campus, Gachibowli, Hyderabad, Telangana, 500046, India
| | - Anjali Mahilkar
- Center for Innovation in Molecular and Pharmaceutical Sciences (CIMPS), Dr. Reddy's Institute of Life Sciences, University of Hyderabad Campus, Gachibowli, Hyderabad, Telangana, 500046, India
| | - Venkateswarlu Akella
- Center for Innovation in Molecular and Pharmaceutical Sciences (CIMPS), Dr. Reddy's Institute of Life Sciences, University of Hyderabad Campus, Gachibowli, Hyderabad, Telangana, 500046, India
| | - Uday Saxena
- Center for Innovation in Molecular and Pharmaceutical Sciences (CIMPS), Dr. Reddy's Institute of Life Sciences, University of Hyderabad Campus, Gachibowli, Hyderabad, Telangana, 500046, India
| | - Pushkar Kulkarni
- Center for Innovation in Molecular and Pharmaceutical Sciences (CIMPS), Dr. Reddy's Institute of Life Sciences, University of Hyderabad Campus, Gachibowli, Hyderabad, Telangana, 500046, India.
| | - Aarti Sevilimedu
- Center for Innovation in Molecular and Pharmaceutical Sciences (CIMPS), Dr. Reddy's Institute of Life Sciences, University of Hyderabad Campus, Gachibowli, Hyderabad, Telangana, 500046, India.
| |
Collapse
|
50
|
Maurin T, Lebrigand K, Castagnola S, Paquet A, Jarjat M, Popa A, Grossi M, Rage F, Bardoni B. HITS-CLIP in various brain areas reveals new targets and new modalities of RNA binding by fragile X mental retardation protein. Nucleic Acids Res 2019; 46:6344-6355. [PMID: 29668986 PMCID: PMC6158598 DOI: 10.1093/nar/gky267] [Citation(s) in RCA: 115] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Accepted: 03/29/2018] [Indexed: 01/19/2023] Open
Abstract
Fragile X syndrome (FXS), the most common form of inherited intellectual disability, is due to the functional deficiency of the fragile X mental retardation protein (FMRP), an RNA-binding protein involved in translational regulation of many messenger RNAs, playing key roles in synaptic morphology and plasticity. To date, no effective treatment for FXS is available. We searched for FMRP targets by HITS-CLIP during early development of multiple mouse brain regions (hippocampus, cortex and cerebellum) at a time of brain development when FMRP is most highly expressed and synaptogenesis reaches a peak. We identified the largest dataset of mRNA targets of FMRP available in brain and we defined their cellular origin. We confirmed the G-quadruplex containing structure as an enriched motif in FMRP RNA targets. In addition to four less represented motifs, our study points out that, in the brain, CTGKA is the prominent motif bound by FMRP, which recognizes it when not engaged in Watson–Crick pairing. All of these motifs negatively modulated the expression level of a reporter protein. While the repertoire of FMRP RNA targets in cerebellum is quite divergent, the ones of cortex and hippocampus are vastly overlapping. In these two brain regions, the Phosphodiesterase 2a (Pde2a) mRNA is a prominent target of FMRP, which modulates its translation and intracellular transport. This enzyme regulates the homeostasis of cAMP and cGMP and represents a novel and attractive therapeutic target to treat FXS.
Collapse
Affiliation(s)
- Thomas Maurin
- Université Côte d'Azur, CNRS, IPMC, 06560 Valbonne, France.,CNRS LIA « Neogenex », 06560 Valbonne, France
| | | | - Sara Castagnola
- Université Côte d'Azur, CNRS, IPMC, 06560 Valbonne, France.,CNRS LIA « Neogenex », 06560 Valbonne, France
| | - Agnès Paquet
- Université Côte d'Azur, CNRS, IPMC, 06560 Valbonne, France
| | - Marielle Jarjat
- Université Côte d'Azur, CNRS, IPMC, 06560 Valbonne, France.,CNRS LIA « Neogenex », 06560 Valbonne, France
| | - Alexandra Popa
- Research Center for Molecular Medicine of the Austrian Academy of Sciences, A-1090 Vienna, Austria
| | - Mauro Grossi
- Université Côte d'Azur, CNRS, IPMC, 06560 Valbonne, France.,CNRS LIA « Neogenex », 06560 Valbonne, France
| | - Florence Rage
- CNRS, Institut de Génétique Moléculaire, 34293 Montpellier, France
| | - Barbara Bardoni
- CNRS LIA « Neogenex », 06560 Valbonne, France.,Université Côte d'Azur, INSERM, CNRS, IPMC, 06560 Valbonne, France
| |
Collapse
|