1
|
Chang KH, Chen CM. The Role of NRF2 in Trinucleotide Repeat Expansion Disorders. Antioxidants (Basel) 2024; 13:649. [PMID: 38929088 PMCID: PMC11200942 DOI: 10.3390/antiox13060649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 05/20/2024] [Accepted: 05/23/2024] [Indexed: 06/28/2024] Open
Abstract
Trinucleotide repeat expansion disorders, a diverse group of neurodegenerative diseases, are caused by abnormal expansions within specific genes. These expansions trigger a cascade of cellular damage, including protein aggregation and abnormal RNA binding. A key contributor to this damage is oxidative stress, an imbalance of reactive oxygen species that harms cellular components. This review explores the interplay between oxidative stress and the NRF2 pathway in these disorders. NRF2 acts as the master regulator of the cellular antioxidant response, orchestrating the expression of enzymes that combat oxidative stress. Trinucleotide repeat expansion disorders often exhibit impaired NRF2 signaling, resulting in inadequate responses to excessive ROS production. NRF2 activation has been shown to upregulate antioxidative gene expression, effectively alleviating oxidative stress damage. NRF2 activators, such as omaveloxolone, vatiquinone, curcumin, sulforaphane, dimethyl fumarate, and resveratrol, demonstrate neuroprotective effects by reducing oxidative stress in experimental cell and animal models of these diseases. However, translating these findings into successful clinical applications requires further research. In this article, we review the literature supporting the role of NRF2 in the pathogenesis of these diseases and the potential therapeutics of NRF2 activators.
Collapse
Affiliation(s)
- Kuo-Hsuan Chang
- Department of Neurology, Chang Gung Memorial Hospital, Linkou Medical Center, Kueishan, Taoyuan 333, Taiwan;
- College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
| | - Chiung-Mei Chen
- Department of Neurology, Chang Gung Memorial Hospital, Linkou Medical Center, Kueishan, Taoyuan 333, Taiwan;
- College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
| |
Collapse
|
2
|
Fok AHK, Huang Y, So BWL, Zheng Q, Tse CSC, Li X, Wong KKY, Huang J, Lai KO, Lai CSW. KIF5B plays important roles in dendritic spine plasticity and dendritic localization of PSD95 and FMRP in the mouse cortex in vivo. Cell Rep 2024; 43:113906. [PMID: 38451812 DOI: 10.1016/j.celrep.2024.113906] [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: 04/19/2023] [Revised: 12/21/2023] [Accepted: 02/16/2024] [Indexed: 03/09/2024] Open
Abstract
Kinesin 1 (KIF5) is one major type of motor protein in neurons, but its members' function in the intact brain remains less studied. Using in vivo two-photon imaging, we find that conditional knockout of Kif5b (KIF5B cKO) in CaMKIIα-Cre-expressing neurons shows heightened turnover and lower stability of dendritic spines in layer 2/3 pyramidal neurons with reduced spine postsynaptic density protein 95 acquisition in the mouse cortex. Furthermore, the RNA-binding protein fragile X mental retardation protein (FMRP) is translocated to the proximity of newly formed spines several hours before the spine formation events in vivo in control mice, but this preceding transport of FMRP is abolished in KIF5B cKO mice. We further find that FMRP is localized closer to newly formed spines after fear extinction, but this learning-dependent localization is disrupted in KIF5B cKO mice. Our findings provide the crucial in vivo evidence that KIF5B is involved in the dendritic targeting of synaptic proteins that underlies dendritic spine plasticity.
Collapse
Affiliation(s)
- Albert Hiu Ka Fok
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong SAR, China
| | - Yuhua Huang
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong SAR, China; Advanced Biomedical Instrumentation Centre, Hong Kong Science Park, Shatin, New Territories, Hong Kong SAR, China
| | - Beth Wing Lam So
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong SAR, China
| | - Qiyu Zheng
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong SAR, China
| | - Chun Sing Carlos Tse
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong SAR, China
| | - Xiaoyang Li
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong SAR, China
| | - Kenneth Kin-Yip Wong
- Advanced Biomedical Instrumentation Centre, Hong Kong Science Park, Shatin, New Territories, Hong Kong SAR, China; Department of Electrical and Electronic Engineering, The University of Hong Kong, Hong Kong SAR, China
| | - Jiandong Huang
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong SAR, China; Chinese Academy of Sciences (CAS) Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China; Clinical Oncology Center, Shenzhen Key Laboratory for Cancer Metastasis and Personalized Therapy, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China; Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-Sen University, Guangzhou, China; State Key Laboratory of Cognitive and Brain Research, The University of Hong Kong, Hong Kong SAR, China
| | - Kwok-On Lai
- Department of Neuroscience, City University of Hong Kong, Hong Kong SAR, China; Hong Kong Institute for Advanced Study, City University of Hong Kong, Hong Kong SAR, China.
| | - Cora Sau Wan Lai
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong SAR, China; State Key Laboratory of Cognitive and Brain Research, The University of Hong Kong, Hong Kong SAR, China.
| |
Collapse
|
3
|
Gundermann DG, Lymer S, Blau J. A rapid and dynamic role for FMRP in the plasticity of adult neurons. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.01.555985. [PMID: 37693612 PMCID: PMC10491314 DOI: 10.1101/2023.09.01.555985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
Fragile X syndrome (FXS) is a neuro-developmental disorder caused by silencing Fmr1, which encodes the RNA-binding protein FMRP. Although Fmr1 is expressed in adult neurons, it has been challenging to separate acute from chronic effects of loss of Fmr1 in models of FXS. We have used the precision of Drosophila genetics to test if Fmr1 acutely affects adult neuronal plasticity in vivo, focusing on the s-LNv circadian pacemaker neurons that show 24 hour rhythms in structural plasticity. We found that over-expressing Fmr1 for only 4 hours blocks the activity-dependent expansion of s-LNv projections without altering the circadian clock or activity-regulated gene expression. Conversely, acutely reducing Fmr1 expression prevented s-LNv projections from retracting. One FMRP target that we identified in s-LNvs is sif, which encodes a Rac1 GEF. Our data indicate that FMRP normally reduces sif mRNA translation at dusk to reduce Rac1 activity. Overall, our data reveal a previously unappreciated rapid and direct role for FMRP in acutely regulating neuronal plasticity in adult neurons, and underscore the importance of RNA-binding proteins in this process.
Collapse
Affiliation(s)
- Daniel G Gundermann
- Department of Biology, New York University, 100 Washington Square East, New York, NY 10003, USA
| | - Seana Lymer
- Department of Biology, New York University, 100 Washington Square East, New York, NY 10003, USA
- Current address: Proteintech Genomics, 11588 Sorrento Valley Rd, San Diego, CA 92121
| | - Justin Blau
- Department of Biology, New York University, 100 Washington Square East, New York, NY 10003, USA
- Center for Genomics and Systems Biology, New York University Abu Dhabi, Abu Dhabi, UAE
| |
Collapse
|
4
|
Caredda E, Pedini G, D'Amico F, Scioli MG, Pacini L, Orsaria P, Vanni G, Buonomo OC, Orlandi A, Bagni C, Palombi L. FMRP expression in primary breast tumor cells correlates with recurrence and specific site of metastasis. PLoS One 2023; 18:e0287062. [PMID: 37379311 DOI: 10.1371/journal.pone.0287062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 05/26/2023] [Indexed: 06/30/2023] Open
Abstract
Breast cancer is the most common cancer among women worldwide. Molecular and clinical evidence indicated that Fragile X Messenger Ribonucleoprotein 1 (FMRP) plays a role in different types of cancer, including breast cancer. FMRP is an RNA binding protein that regulates the metabolism of a large group of mRNAs coding for proteins involved in both neural processes and in epithelial-mesenchymal transition, a pivotal mechanism that in cancer is associated to tumor progression, aggressiveness and chemoresistance. Here, we carried out a retrospective case-control study of 127 patients, to study the expression of FMRP and its correlation with metastasis formation in breast cancer. Consistent with previous findings, we found that FMRP levels are high in tumor tissue. Two categories have been analyzed, tumor with no metastases (referred as control tumors, 84 patients) and tumor with distant metastatic repetition, (referred as cases, 43 patients), with a follow-up of 7 years (mean). We found that FMRP levels were lower in both the nuclei and the cytoplasm in the cases compared to control tumors. Next, within the category cases (tumor with metastases) we evaluated FMRP expression in the specific sites of metastasis revealing a nuclear staining of FMRP. In addition, FMRP expression in both the nuclear and cytoplasmic compartment was significantly lower in patients who developed brain and bone metastases and higher in hepatic and pulmonary sites. While further studies are required to explore the underlying molecular mechanisms of FMRP expression and direct or inverse correlation with the secondary metastatic site, our findings suggest that FMRP levels might be considered a prognostic factor for site-specific metastasis.
Collapse
Affiliation(s)
- E Caredda
- Department of Biomedicine and Prevention, Faculty of Medicine, University of Rome Tor Vergata, Rome, Italy
- Directorate-General for Health Prevention, Ministry of Health, Rome, Italy
| | - G Pedini
- Department of Biomedicine and Prevention, Faculty of Medicine, University of Rome Tor Vergata, Rome, Italy
| | - F D'Amico
- Anatomic Pathology, Department Biomedicine and Prevention, Faculty of Medicine, Tor Vergata University Hospital, Rome, Italy
- Infectious Diseases Unit, Niguarda Hospital, Milan, Italy
| | - M G Scioli
- Anatomic Pathology, Department Biomedicine and Prevention, Faculty of Medicine, Tor Vergata University Hospital, Rome, Italy
| | - L Pacini
- Department of Biomedicine and Prevention, Faculty of Medicine, University of Rome Tor Vergata, Rome, Italy
- UniCamillus, Saint Camillus International, Faculty of Medicine, University of Health and Medical Sciences, Rome, Italy
| | - P Orsaria
- Department of Breast Surgery, University Campus Bio-Medico, Rome, Italy
| | - G Vanni
- Department of Surgery, Faculty of Medicine, Tor Vergata University Hospital, Rome, Italy
| | - O C Buonomo
- Department of Surgery, Faculty of Medicine, Tor Vergata University Hospital, Rome, Italy
| | - A Orlandi
- Anatomic Pathology, Department Biomedicine and Prevention, Faculty of Medicine, Tor Vergata University Hospital, Rome, Italy
| | - C Bagni
- Department of Biomedicine and Prevention, Faculty of Medicine, University of Rome Tor Vergata, Rome, Italy
- Department of Fundamental Neurosciences (DNF), Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - L Palombi
- Department of Biomedicine and Prevention, Faculty of Medicine, University of Rome Tor Vergata, Rome, Italy
| |
Collapse
|
5
|
Mercaldo V, Vidimova B, Gastaldo D, Fernández E, Lo AC, Cencelli G, Pedini G, De Rubeis S, Longo F, Klann E, Smit AB, Grant SGN, Achsel T, Bagni C. Altered striatal actin dynamics drives behavioral inflexibility in a mouse model of fragile X syndrome. Neuron 2023; 111:1760-1775.e8. [PMID: 36996810 DOI: 10.1016/j.neuron.2023.03.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 12/21/2022] [Accepted: 03/06/2023] [Indexed: 03/31/2023]
Abstract
The proteome of glutamatergic synapses is diverse across the mammalian brain and involved in neurodevelopmental disorders (NDDs). Among those is fragile X syndrome (FXS), an NDD caused by the absence of the functional RNA-binding protein FMRP. Here, we demonstrate how the brain region-specific composition of postsynaptic density (PSD) contributes to FXS. In the striatum, the FXS mouse model shows an altered association of the PSD with the actin cytoskeleton, reflecting immature dendritic spine morphology and reduced synaptic actin dynamics. Enhancing actin turnover with constitutively active RAC1 ameliorates these deficits. At the behavioral level, the FXS model displays striatal-driven inflexibility, a typical feature of FXS individuals, which is rescued by exogenous RAC1. Striatal ablation of Fmr1 is sufficient to recapitulate behavioral impairments observed in the FXS model. These results indicate that dysregulation of synaptic actin dynamics in the striatum, a region largely unexplored in FXS, contributes to the manifestation of FXS behavioral phenotypes.
Collapse
Affiliation(s)
- Valentina Mercaldo
- Department of Fundamental Neurosciences, Université de Lausanne, 1005 Lausanne, Switzerland
| | - Barbora Vidimova
- Department of Fundamental Neurosciences, Université de Lausanne, 1005 Lausanne, Switzerland
| | - Denise Gastaldo
- Department of Fundamental Neurosciences, Université de Lausanne, 1005 Lausanne, Switzerland
| | - Esperanza Fernández
- VIB & UGent Center for Medical Biotechnology, Universiteit Gent, 9052 Ghent, Belgium
| | - Adrian C Lo
- Department of Fundamental Neurosciences, Université de Lausanne, 1005 Lausanne, Switzerland
| | - Giulia Cencelli
- Department of Biomedicine and Prevention, Università degli Studi di Roma "Tor Vergata", 00133 Rome, Italy; Institute of Neurosurgery, Fondazione Policlinico Universitario A. Gemelli IRCCS - Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Giorgia Pedini
- Department of Biomedicine and Prevention, Università degli Studi di Roma "Tor Vergata", 00133 Rome, Italy
| | - Silvia De Rubeis
- Seaver Autism Center for Research and Treatment, Department of Psychiatry, Friedman Brain Institute, Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Francesco Longo
- Center for Neural Science, New York University, New York, NY 10029, USA
| | - Eric Klann
- Center for Neural Science, New York University, New York, NY 10029, USA
| | - August B Smit
- Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Seth G N Grant
- Center for the Clinical Brain Sciences and Simons Initiatives for the Developing Brain, The University of Edinburgh, Edinburgh EH16 4SB, Scotland
| | - Tilmann Achsel
- Department of Fundamental Neurosciences, Université de Lausanne, 1005 Lausanne, Switzerland.
| | - Claudia Bagni
- Department of Fundamental Neurosciences, Université de Lausanne, 1005 Lausanne, Switzerland; Department of Biomedicine and Prevention, Università degli Studi di Roma "Tor Vergata", 00133 Rome, Italy.
| |
Collapse
|
6
|
Mitchell ME, Cook LC, Shiers S, Tavares-Ferreira D, Akopian AN, Dussor G, Price TJ. Characterization of Fragile X Mental Retardation Protein expression in human nociceptors and their axonal projections to the spinal dorsal horn. J Comp Neurol 2023; 531:814-835. [PMID: 36808110 PMCID: PMC10038933 DOI: 10.1002/cne.25463] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 12/20/2022] [Accepted: 01/27/2023] [Indexed: 02/23/2023]
Abstract
Fragile X Mental Retardation Protein (FMRP) regulates activity-dependent RNA localization and local translation to modulate synaptic plasticity throughout the central nervous system. Mutations in the FMR1 gene that hinder or ablate FMRP function cause Fragile X Syndrome (FXS), a disorder associated with sensory processing dysfunction. FXS premutations are associated with increased FMRP expression and neurological impairments including sex dimorphic presentations of chronic pain. In mice, FMRP ablation causes dysregulated dorsal root ganglion (DRG) neuron excitability and synaptic vesicle exocytosis, spinal circuit activity, and decreased translation-dependent nociceptive sensitization. Activity-dependent, local translation is a key mechanism for enhancing primary nociceptor excitability that promotes pain in animals and humans. These works indicate that FMRP likely regulates nociception and pain at the level of the primary nociceptor or spinal cord. Therefore, we sought to better understand FMRP expression in the human DRG and spinal cord using immunostaining in organ donor tissues. We find that FMRP is highly expressed in DRG and spinal neuron subsets with substantia gelatinosa exhibiting the most abundant immunoreactivity in spinal synaptic fields. Here, it is expressed in nociceptor axons. FMRP puncta colocalized with Nav1.7 and TRPV1 receptor signals suggesting a pool of axoplasmic FMRP localizes to plasma membrane-associated loci in these branches. Interestingly, FMRP puncta exhibited notable colocalization with calcitonin gene-related peptide (CGRP) immunoreactivity selectively in female spinal cord. Our results support a regulatory role for FMRP in human nociceptor axons of the dorsal horn and implicate it in the sex dimorphic actions of CGRP signaling in nociceptive sensitization and chronic pain.
Collapse
Affiliation(s)
- Molly E Mitchell
- Center for Advanced Pain Studies, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Texas, USA
| | - Lauren C Cook
- Center for Advanced Pain Studies, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Texas, USA
| | - Stephanie Shiers
- Center for Advanced Pain Studies, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Texas, USA
| | - Diana Tavares-Ferreira
- Center for Advanced Pain Studies, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Texas, USA
| | - Armen N Akopian
- Department of Endodontics, UT Health San Antonio, San Antonio, Texas, USA
| | - Gregory Dussor
- Center for Advanced Pain Studies, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Texas, USA
| | - Theodore J Price
- Center for Advanced Pain Studies, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Texas, USA
| |
Collapse
|
7
|
Gredell M, Lu J, Zuo Y. The effect of single-cell knockout of Fragile X Messenger Ribonucleoprotein on synaptic structural plasticity. Front Synaptic Neurosci 2023; 15:1135479. [PMID: 37035256 PMCID: PMC10076639 DOI: 10.3389/fnsyn.2023.1135479] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Accepted: 03/07/2023] [Indexed: 04/11/2023] Open
Abstract
Fragile X Syndrome (FXS) is the best-known form of inherited intellectual disability caused by the loss-of-function mutation in a single gene. The FMR1 gene mutation abolishes the expression of Fragile X Messenger Ribonucleoprotein (FMRP), which regulates the expression of many synaptic proteins. Cortical pyramidal neurons in postmortem FXS patient brains show abnormally high density and immature morphology of dendritic spines; this phenotype is replicated in the Fmr1 knockout (KO) mouse. While FMRP is well-positioned in the dendrite to regulate synaptic plasticity, intriguing in vitro and in vivo data show that wild type neurons embedded in a network of Fmr1 KO neurons or glia exhibit spine abnormalities just as neurons in Fmr1 global KO mice. This raises the question: does FMRP regulate synaptic morphology and dynamics in a cell-autonomous manner, or do the synaptic phenotypes arise from abnormal pre-synaptic inputs? To address this question, we combined viral and mouse genetic approaches to delete FMRP from a very sparse subset of cortical layer 5 pyramidal neurons (L5 PyrNs) either during early postnatal development or in adulthood. We then followed the structural dynamics of dendritic spines on these Fmr1 KO neurons by in vivo two-photon microscopy. We found that, while L5 PyrNs in adult Fmr1 global KO mice have abnormally high density of thin spines, single-cell Fmr1 KO in adulthood does not affect spine density, morphology, or dynamics. On the contrary, neurons with neonatal FMRP deletion have normal spine density but elevated spine formation at 1 month of age, replicating the phenotype in Fmr1 global KO mice. Interestingly, these neurons exhibit elevated thin spine density, but normal total spine density, by adulthood. Together, our data reveal cell-autonomous FMRP regulation of cortical synaptic dynamics during adolescence, but spine defects in adulthood also implicate non-cell-autonomous factors.
Collapse
Affiliation(s)
| | | | - Yi Zuo
- Department of Molecular, Cell and Developmental Biology, University of California, Santa Cruz, Santa Cruz, CA, United States
| |
Collapse
|
8
|
Ardourel M, Ranchon-Cole I, Pâris A, Felgerolle C, Acar N, Lesne F, Briault S, Perche O. FMR protein: Evidence of an emerging role in retinal aging? Exp Eye Res 2022; 225:109282. [PMID: 36265576 DOI: 10.1016/j.exer.2022.109282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 10/05/2022] [Accepted: 10/11/2022] [Indexed: 12/29/2022]
Abstract
Aging is a multifactorial process that affects the entire organism by cumulative alterations. Visual function impairments that go along with aging are commonly observed, causing lower visual acuity, lower contrast sensitivity, and impaired dark adaptation. Electroretinogram analysis revealed that the amplitudes of rod- and cone-mediated responses are reduced in aged mice and humans. Reports suggested that age-related changes observed in both rod and cone photoreceptor functionality were linked to oxidative stress regulation or free radical production homeostasis. Interestingly, several recent reports linked the fragile X mental retardation protein (FMRP) cellular activity with oxidative stress regulation in several tissue including brain tissue where FMRP participates to the response to stress via protein translation in neurite or is involved in free radical production and abnormal glutathione homeostasis. Based on these recent literatures, we raised the question about the effect of FMRP absence in the aging retina of Fmr1-/y compared to their WT littermates. Indeed, up to now, only young or adult mice (<6 months) were investigated and have shown a specific retinal phenotype. Herein, we demonstrated that Fmr1-/y mice do not present the aging effect on retinal function observed in WT littermates since ERG a- and b-waves amplitudes as well as oscillatory potentials amplitudes were not collapsed with age (12/18 months old). Absence of FMRP and its consequences seem to protect the retina against aging effect, rising a pivotal role of FMRP in retinal aging process.
Collapse
Affiliation(s)
- M Ardourel
- UMR7355, CNRS, Orléans, France; Experimental and Molecular Immunology and Neurogenetics, University of Orléans, 3b rue de la Ferollerie, 45071, Orléans, Cedex 2, France
| | - I Ranchon-Cole
- Université Clermont Auvergne, CHU Clermont-Ferrand, Inserm, Neuro-Dol, F-63000, Clermont-Ferrand, France
| | - A Pâris
- UMR7355, CNRS, Orléans, France; Experimental and Molecular Immunology and Neurogenetics, University of Orléans, 3b rue de la Ferollerie, 45071, Orléans, Cedex 2, France
| | - C Felgerolle
- UMR7355, CNRS, Orléans, France; Experimental and Molecular Immunology and Neurogenetics, University of Orléans, 3b rue de la Ferollerie, 45071, Orléans, Cedex 2, France
| | - N Acar
- Eye and Nutrition Research Group, Centre des Sciences du Goût et de l'Alimentation, CNRS, INRAE, Institut Agro, Université Bourgogne Franche-Comté, 21000, Dijon, France
| | - F Lesne
- Genetic Department, Regional Hospital, 14 Avenue de l'hôpital, 45100, Orléans, France
| | - S Briault
- UMR7355, CNRS, Orléans, France; Experimental and Molecular Immunology and Neurogenetics, University of Orléans, 3b rue de la Ferollerie, 45071, Orléans, Cedex 2, France; Genetic Department, Regional Hospital, 14 Avenue de l'hôpital, 45100, Orléans, France
| | - O Perche
- UMR7355, CNRS, Orléans, France; Experimental and Molecular Immunology and Neurogenetics, University of Orléans, 3b rue de la Ferollerie, 45071, Orléans, Cedex 2, France; Genetic Department, Regional Hospital, 14 Avenue de l'hôpital, 45100, Orléans, France.
| |
Collapse
|
9
|
Oe S, Hayashi S, Tanaka S, Koike T, Hirahara Y, Seki-Omura R, Kakizaki R, Sakamoto S, Nakano Y, Noda Y, Yamada H, Kitada M. Cytoplasmic Polyadenylation Element-Binding Protein 1 Post-transcriptionally Regulates Fragile X Mental Retardation 1 Expression Through 3′ Untranslated Region in Central Nervous System Neurons. Front Cell Neurosci 2022; 16:869398. [PMID: 35496917 PMCID: PMC9051318 DOI: 10.3389/fncel.2022.869398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 03/24/2022] [Indexed: 11/13/2022] Open
Abstract
Fragile X syndrome (FXS) is an inherited intellectual disability caused by a deficiency in Fragile X mental retardation 1 (Fmr1) gene expression. Recent studies have proposed the importance of cytoplasmic polyadenylation element-binding protein 1 (CPEB1) in FXS pathology; however, the molecular interaction between Fmr1 mRNA and CPEB1 has not been fully investigated. Here, we revealed that CPEB1 co-localized and interacted with Fmr1 mRNA in hippocampal and cerebellar neurons and culture cells. Furthermore, CPEB1 knockdown upregulated Fmr1 mRNA and protein levels and caused aberrant localization of Fragile X mental retardation protein in neurons. In an FXS cell model, CPEB1 knockdown upregulated the mRNA levels of several mitochondria-related genes and rescued the intracellular heat shock protein family A member 9 distribution. These findings suggest that CPEB1 post-transcriptionally regulated Fmr1 expression through the 3′ untranslated region, and that CPEB1 knockdown might affect mitochondrial function.
Collapse
Affiliation(s)
- Souichi Oe
- Department of Anatomy, Kansai Medical University, Hirakata, Japan
- *Correspondence: Souichi Oe,
| | - Shinichi Hayashi
- Department of Anatomy, Kansai Medical University, Hirakata, Japan
| | - Susumu Tanaka
- Department of Anatomy, Kansai Medical University, Hirakata, Japan
| | - Taro Koike
- Department of Anatomy, Kansai Medical University, Hirakata, Japan
| | - Yukie Hirahara
- Department of Anatomy, Kansai Medical University, Hirakata, Japan
| | | | - Rio Kakizaki
- Department of Anatomy, Kansai Medical University, Hirakata, Japan
| | - Sumika Sakamoto
- Department of Anatomy, Kansai Medical University, Hirakata, Japan
| | - Yosuke Nakano
- Department of Anatomy, Kansai Medical University, Hirakata, Japan
| | - Yasuko Noda
- Department of Anatomy, Bio-Imaging and Neuro-Cell Science, Jichi Medical University, Shimotsuke, Japan
| | - Hisao Yamada
- Biwako Professional University of Rehabilitation, Higashiomi, Japan
| | - Masaaki Kitada
- Department of Anatomy, Kansai Medical University, Hirakata, Japan
- Masaaki Kitada,
| |
Collapse
|
10
|
Carotti S, Zingariello M, Francesconi M, D'Andrea L, Latasa MU, Colyn L, Fernandez-Barrena MG, Flammia RS, Falchi M, Righi D, Pedini G, Pantano F, Bagni C, Perrone G, Rana RA, Avila MA, Morini S, Zalfa F. Fragile X mental retardation protein in intrahepatic cholangiocarcinoma: regulating the cancer cell behavior plasticity at the leading edge. Oncogene 2021; 40:4033-4049. [PMID: 34017076 PMCID: PMC8195741 DOI: 10.1038/s41388-021-01824-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2019] [Revised: 04/15/2021] [Accepted: 04/27/2021] [Indexed: 01/06/2023]
Abstract
Intrahepatic cholangiocarcinoma (iCCA) is a rare malignancy of the intrahepatic biliary tract with a very poor prognosis. Although some clinicopathological parameters can be prognostic factors for iCCA, the molecular prognostic markers and potential mechanisms of iCCA have not been well investigated. Here, we report that the Fragile X mental retardation protein (FMRP), a RNA binding protein functionally absent in patients with the Fragile X syndrome (FXS) and also involved in several types of cancers, is overexpressed in human iCCA and its expression is significantly increased in iCCA metastatic tissues. The silencing of FMRP in metastatic iCCA cell lines affects cell migration and invasion, suggesting a role of FMRP in iCCA progression. Moreover, we show evidence that FMRP is localized at the invasive front of human iCCA neoplastic nests and in pseudopodia and invadopodia protrusions of migrating and invading iCCA cancer cells. Here FMRP binds several mRNAs encoding key proteins involved in the formation and/or function of these protrusions. In particular, we find that FMRP binds to and regulates the expression of Cortactin, a critical regulator of invadopodia formation. Altogether, our findings suggest that FMRP could promote cell invasiveness modulating membrane plasticity and invadopodia formation at the leading edges of invading iCCA cells.
Collapse
Affiliation(s)
- Simone Carotti
- Research Unit of Microscopic and Ultrastructural Anatomy, Department of Medicine, Campus Bio-Medico University, Rome, Italy
- Predictive Molecular Diagnostic Unit, Department of Pathology, Campus Bio-Medico University Hospital, Rome, Italy
| | - Maria Zingariello
- Research Unit of Microscopic and Ultrastructural Anatomy, Department of Medicine, Campus Bio-Medico University, Rome, Italy
| | - Maria Francesconi
- Research Unit of Microscopic and Ultrastructural Anatomy, Department of Medicine, Campus Bio-Medico University, Rome, Italy
| | - Laura D'Andrea
- Research Unit of Microscopic and Ultrastructural Anatomy, Department of Medicine, Campus Bio-Medico University, Rome, Italy
| | - M Ujue Latasa
- Hepatology Program, Center for Applied Medical Research (CIMA), University of Navarra and IdiSNA, Pamplona, Spain
| | - Leticia Colyn
- Hepatology Program, Center for Applied Medical Research (CIMA), University of Navarra and IdiSNA, Pamplona, Spain
| | - Maite G Fernandez-Barrena
- Hepatology Program, Center for Applied Medical Research (CIMA), University of Navarra and IdiSNA, Pamplona, Spain
- Centro de Investigación Biomédica en Red, Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid, Spain
| | - Rocco Simone Flammia
- Research Unit of Microscopic and Ultrastructural Anatomy, Department of Medicine, Campus Bio-Medico University, Rome, Italy
| | - Mario Falchi
- National AIDS Center, Istituto Superiore di Sanità, Rome, Italy
| | - Daniela Righi
- Predictive Molecular Diagnostic Unit, Department of Pathology, Campus Bio-Medico University Hospital, Rome, Italy
| | - Giorgia Pedini
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | - Francesco Pantano
- Medical Oncology Department, Campus Bio-Medico University, Rome, Italy
| | - Claudia Bagni
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, Switzerland
| | - Giuseppe Perrone
- Predictive Molecular Diagnostic Unit, Department of Pathology, Campus Bio-Medico University Hospital, Rome, Italy
- Research Unit of Pathology, Campus Bio-Medico University, Rome, Italy
| | - Rosa Alba Rana
- Medicine and Aging Science Department, University G. D'Annunzio, Chieti-Pescara, Italy
| | - Matias A Avila
- Hepatology Program, Center for Applied Medical Research (CIMA), University of Navarra and IdiSNA, Pamplona, Spain
- Centro de Investigación Biomédica en Red, Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid, Spain
| | - Sergio Morini
- Research Unit of Microscopic and Ultrastructural Anatomy, Department of Medicine, Campus Bio-Medico University, Rome, Italy.
| | - Francesca Zalfa
- Research Unit of Microscopic and Ultrastructural Anatomy, Department of Medicine, Campus Bio-Medico University, Rome, Italy.
- Predictive Molecular Diagnostic Unit, Department of Pathology, Campus Bio-Medico University Hospital, Rome, Italy.
| |
Collapse
|
11
|
Lo AC, Rajan N, Gastaldo D, Telley L, Hilal ML, Buzzi A, Simonato M, Achsel T, Bagni C. Absence of RNA-binding protein FXR2P prevents prolonged phase of kainate-induced seizures. EMBO Rep 2021; 22:e51404. [PMID: 33779029 PMCID: PMC8024897 DOI: 10.15252/embr.202051404] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 01/26/2021] [Accepted: 01/29/2021] [Indexed: 12/26/2022] Open
Abstract
Status epilepticus (SE) is a condition in which seizures are not self-terminating and thereby pose a serious threat to the patient's life. The molecular mechanisms underlying SE are likely heterogeneous and not well understood. Here, we reveal a role for the RNA-binding protein Fragile X-Related Protein 2 (FXR2P) in SE. Fxr2 KO mice display reduced sensitivity specifically to kainic acid-induced SE. Immunoprecipitation of FXR2P coupled to next-generation sequencing of associated mRNAs shows that FXR2P targets are enriched in genes that encode glutamatergic post-synaptic components. Of note, the FXR2P target transcriptome has a significant overlap with epilepsy and SE risk genes. In addition, Fxr2 KO mice fail to show sustained ERK1/2 phosphorylation induced by KA and present reduced burst activity in the hippocampus. Taken together, our findings show that the absence of FXR2P decreases the expression of glutamatergic proteins, and this decrease might prevent self-sustained seizures.
Collapse
Affiliation(s)
- Adrian C Lo
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, Switzerland
| | - Nicholas Rajan
- Department of Neurosciences and Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - Denise Gastaldo
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, Switzerland
| | - Ludovic Telley
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, Switzerland
| | - Muna L Hilal
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, Switzerland
| | - Andrea Buzzi
- Department of Neuroscience and Rehabilitation, University of Ferrara, Ferrara, Italy
| | - Michele Simonato
- Department of Neuroscience and Rehabilitation, University of Ferrara, Ferrara, Italy.,Division of Neuroscience, IRCCS San Raffaele Hospital, Milan, Italy
| | - Tilmann Achsel
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, Switzerland
| | - Claudia Bagni
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, Switzerland.,Department of Neurosciences and Leuven Brain Institute, KU Leuven, Leuven, Belgium.,Department of Biomedicine and Prevention, University of Rome "Tor Vergata", Rome, Italy
| |
Collapse
|
12
|
Adenosine A 2A receptor inhibition reduces synaptic and cognitive hippocampal alterations in Fmr1 KO mice. Transl Psychiatry 2021; 11:112. [PMID: 33547274 PMCID: PMC7864914 DOI: 10.1038/s41398-021-01238-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 12/28/2020] [Accepted: 01/18/2021] [Indexed: 02/07/2023] Open
Abstract
In fragile X syndrome (FXS) the lack of the fragile X mental retardation protein (FMRP) leads to exacerbated signaling through the metabotropic glutamate receptors 5 (mGlu5Rs). The adenosine A2A receptors (A2ARs), modulators of neuronal damage, could play a role in FXS. A synaptic colocalization and a strong permissive interaction between A2A and mGlu5 receptors in the hippocampus have been previously reported, suggesting that blocking A2ARs might normalize the mGlu5R-mediated effects of FXS. To study the cross-talk between A2A and mGlu5 receptors in the absence of FMRP, we performed extracellular electrophysiology experiments in hippocampal slices of Fmr1 KO mouse. The depression of field excitatory postsynaptic potential (fEPSPs) slope induced by the mGlu5R agonist CHPG was completely blocked by the A2AR antagonist ZM241385 and strongly potentiated by the A2AR agonist CGS21680, suggesting that the functional synergistic coupling between the two receptors could be increased in FXS. To verify if chronic A2AR blockade could reverse the FXS phenotypes, we treated the Fmr1 KO mice with istradefylline, an A2AR antagonist. We found that hippocampal DHPG-induced long-term depression (LTD), which is abnormally increased in FXS mice, was restored to the WT level. Furthermore, istradefylline corrected aberrant dendritic spine density, specific behavioral alterations, and overactive mTOR, TrkB, and STEP signaling in Fmr1 KO mice. Finally, we identified A2AR mRNA as a target of FMRP. Our results show that the pharmacological blockade of A2ARs partially restores some of the phenotypes of Fmr1 KO mice, both by reducing mGlu5R functioning and by acting on other A2AR-related downstream targets.
Collapse
|
13
|
Di Grazia A, Marafini I, Pedini G, Di Fusco D, Laudisi F, Dinallo V, Rosina E, Stolfi C, Franzè E, Sileri P, Sica G, Monteleone G, Bagni C, Monteleone I. The Fragile X Mental Retardation Protein Regulates RIPK1 and Colorectal Cancer Resistance to Necroptosis. Cell Mol Gastroenterol Hepatol 2020; 11:639-658. [PMID: 33091622 PMCID: PMC7806864 DOI: 10.1016/j.jcmgh.2020.10.009] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 10/15/2020] [Accepted: 10/16/2020] [Indexed: 12/23/2022]
Abstract
BACKGROUND & AIMS The fragile X mental retardation protein (FMRP) affects multiple steps of the mRNA metabolism during brain development and in different neoplastic processes. However, the contribution of FMRP in colon carcinogenesis has not been investigated. METHODS FMR1 mRNA transcript and FMRP protein expression were analyzed in human colon samples derived from patients with sporadic colorectal cancer (CRC) and healthy subjects. We used a well-established mouse model of sporadic CRC induced by azoxymethane to determine the possible role of FMRP in CRC. To address whether FMRP controls cancer cell survival, we analyzed cell death pathway in CRC human epithelial cell lines and in patient-derived colon cancer organoids in presence or absence of a specific FMR1 antisense oligonucleotide or siRNA. RESULTS We document a significant increase of FMRP in human CRC relative to non-tumor tissues. Next, using an inducible mouse model of CRC, we observed a reduction of colonic tumor incidence and size in the Fmr1 knockout mice. The abrogation of FMRP induced spontaneous cell death in human CRC cell lines activating the necroptotic pathway. Indeed, specific immunoprecipitation experiments on human cell lines and CRC samples indicated that FMRP binds receptor-interacting protein kinase 1 (RIPK1) mRNA, suggesting that FMRP acts as a regulator of necroptosis pathway through the surveillance of RIPK1 mRNA metabolism. Treatment of human CRC cell lines and patient-derived colon cancer organoids with the FMR1 antisense resulted in up-regulation of RIPK1. CONCLUSIONS Altogether, these data support a role for FMRP in controlling RIPK1 expression and necroptotic activation in CRC.
Collapse
Affiliation(s)
- Antonio Di Grazia
- Department of Systems Medicine, University of Rome 'Tor Vergata', Rome, Italy
| | - Irene Marafini
- Department of Systems Medicine, University of Rome 'Tor Vergata', Rome, Italy
| | - Giorgia Pedini
- Department of Biomedicine and Prevention, University of Rome 'Tor Vergata', Rome, Italy
| | - Davide Di Fusco
- Department of Systems Medicine, University of Rome 'Tor Vergata', Rome, Italy
| | - Federica Laudisi
- Department of Systems Medicine, University of Rome 'Tor Vergata', Rome, Italy
| | - Vincenzo Dinallo
- Department of Systems Medicine, University of Rome 'Tor Vergata', Rome, Italy
| | - Eleonora Rosina
- Department of Biomedicine and Prevention, University of Rome 'Tor Vergata', Rome, Italy
| | - Carmine Stolfi
- Department of Systems Medicine, University of Rome 'Tor Vergata', Rome, Italy
| | - Eleonora Franzè
- Department of Systems Medicine, University of Rome 'Tor Vergata', Rome, Italy
| | - Pierpaolo Sileri
- Department of Surgery, University of Rome 'Tor Vergata', Rome, Italy
| | - Giuseppe Sica
- Department of Surgery, University of Rome 'Tor Vergata', Rome, Italy
| | - Giovanni Monteleone
- Department of Systems Medicine, University of Rome 'Tor Vergata', Rome, Italy
| | - Claudia Bagni
- Department of Biomedicine and Prevention, University of Rome 'Tor Vergata', Rome, Italy; Department of Fundamental Neurosciences, University of Lausanne, Lausanne, Switzerland.
| | - Ivan Monteleone
- Department of Biomedicine and Prevention, University of Rome 'Tor Vergata', Rome, Italy.
| |
Collapse
|
14
|
Maltby CJ, Schofield JPR, Houghton SD, O’Kelly I, Vargas-Caballero M, Deinhardt K, Coldwell MJ. A 5' UTR GGN repeat controls localisation and translation of a potassium leak channel mRNA through G-quadruplex formation. Nucleic Acids Res 2020; 48:9822-9839. [PMID: 32870280 PMCID: PMC7515701 DOI: 10.1093/nar/gkaa699] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 08/28/2020] [Indexed: 12/18/2022] Open
Abstract
RNA G-quadruplexes (G4s) are secondary structures proposed to function as regulators of post-transcriptional mRNA localisation and translation. G4s within some neuronal mRNAs are known to control distal localisation and local translation, contributing to distinct local proteomes that facilitate the synaptic remodelling attributed to normal cellular function. In this study, we characterise the G4 formation of a (GGN)13 repeat found within the 5' UTR of the potassium 2-pore domain leak channel Task3 mRNA. Biophysical analyses show that this (GGN)13 repeat forms a parallel G4 in vitro exhibiting the stereotypical potassium specificity of G4s, remaining thermostable under physiological ionic conditions. Through mouse brain tissue G4-RNA immunoprecipitation, we further confirm that Task3 mRNA forms a G4 structure in vivo. The G4 is inhibitory to translation of Task3 in vitro and is overcome through activity of a G4-specific helicase DHX36, increasing K+ leak currents and membrane hyperpolarisation in HEK293 cells. Further, we observe that this G4 is fundamental to ensuring delivery of Task3 mRNA to distal primary cortical neurites. It has been shown that aberrant Task3 expression correlates with neuronal dysfunction, we therefore posit that this G4 is important in regulated local expression of Task3 leak channels that maintain K+ leak within neurons.
Collapse
Affiliation(s)
- Connor J Maltby
- School of Biological Sciences, University of Southampton, Southampton, Hampshire SO17 1BJ, UK
| | - James P R Schofield
- School of Biological Sciences, University of Southampton, Southampton, Hampshire SO17 1BJ, UK
| | - Steven D Houghton
- School of Biological Sciences, University of Southampton, Southampton, Hampshire SO17 1BJ, UK
| | - Ita O’Kelly
- Centre for Human Development, Stem Cells and Regeneration, University of Southampton, Southampton, Hampshire SO17 1BJ, UK
| | | | - Katrin Deinhardt
- School of Biological Sciences, University of Southampton, Southampton, Hampshire SO17 1BJ, UK
| | - Mark J Coldwell
- School of Biological Sciences, University of Southampton, Southampton, Hampshire SO17 1BJ, UK
| |
Collapse
|
15
|
La Rosa P, Petrillo S, Bertini ES, Piemonte F. Oxidative Stress in DNA Repeat Expansion Disorders: A Focus on NRF2 Signaling Involvement. Biomolecules 2020; 10:biom10050702. [PMID: 32369911 PMCID: PMC7277112 DOI: 10.3390/biom10050702] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 04/24/2020] [Accepted: 04/27/2020] [Indexed: 12/13/2022] Open
Abstract
DNA repeat expansion disorders are a group of neuromuscular and neurodegenerative diseases that arise from the inheritance of long tracts of nucleotide repetitions, located in the regulatory region, introns, or inside the coding sequence of a gene. Although loss of protein expression and/or the gain of function of its transcribed mRNA or translated product represent the major pathogenic effect of these pathologies, mitochondrial dysfunction and imbalance in redox homeostasis are reported as common features in these disorders, deeply affecting their severity and progression. In this review, we examine the role that the redox imbalance plays in the pathological mechanisms of DNA expansion disorders and the recent advances on antioxidant treatments, particularly focusing on the expression and the activity of the transcription factor NRF2, the main cellular regulator of the antioxidant response.
Collapse
|
16
|
Nobile V, Palumbo F, Lanni S, Ghisio V, Vitali A, Castagnola M, Marzano V, Maulucci G, De Angelis C, De Spirito M, Pacini L, D'Andrea L, Ragno R, Stazi G, Valente S, Mai A, Chiurazzi P, Genuardi M, Neri G, Tabolacci E. Altered mitochondrial function in cells carrying a premutation or unmethylated full mutation of the FMR1 gene. Hum Genet 2020; 139:227-245. [PMID: 31919630 DOI: 10.1007/s00439-019-02104-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Accepted: 12/21/2019] [Indexed: 12/22/2022]
Abstract
Fragile X-related disorders are due to a dynamic mutation of the CGG repeat at the 5' UTR of the FMR1 gene, coding for the RNA-binding protein FMRP. As the CGG sequence expands from premutation (PM, 56-200 CGGs) to full mutation (> 200 CGGs), FMRP synthesis decreases until it is practically abolished in fragile X syndrome (FXS) patients, mainly due to FMR1 methylation. Cells from rare individuals with no intellectual disability and carriers of an unmethylated full mutation (UFM) produce slightly elevated levels of FMR1-mRNA and relatively low levels of FMRP, like in PM carriers. With the aim of clarifying how UFM cells differ from CTRL and FXS cells, a comparative proteomic approach was undertaken, from which emerged an overexpression of SOD2 in UFM cells, also confirmed in PM but not in FXS. The SOD2-mRNA bound to FMRP in UFM more than in the other cell types. The high SOD2 levels in UFM and PM cells correlated with lower levels of superoxide and reactive oxygen species (ROS), and with morphological anomalies and depolarization of the mitochondrial membrane detected through confocal microscopy. The same effect was observed in CTRL and FXS after treatment with MC2791, causing SOD2 overexpression. These mitochondrial phenotypes reverted after knock-down with siRNA against SOD2-mRNA and FMR1-mRNA in UFM and PM. Overall, these data suggest that in PM and UFM carriers, which have high levels of FMR1 transcription and may develop FXTAS, SOD2 overexpression helps to maintain low levels of both superoxide and ROS with signs of mitochondrial degradation.
Collapse
Affiliation(s)
- Veronica Nobile
- Istituto di Medicina Genomica, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo F. Vito 1, 00168, Roma, Italy
| | - Federica Palumbo
- Istituto di Medicina Genomica, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo F. Vito 1, 00168, Roma, Italy
| | - Stella Lanni
- Program of Genetics and Genome Biology, The Hospital for Sick Children, The Peter Gilgan Centre for Research and Learning, Toronto, ON, Canada
| | - Valentina Ghisio
- UOC Genetica Medica, Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italy
| | - Alberto Vitali
- Institute of Chemistry of Molecular Recognition, CNR, Roma, Italy
- Istituto di Biochimica e Chimica Clinica, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italy
| | - Massimo Castagnola
- Istituto di Biochimica e Chimica Clinica, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italy
| | - Valeria Marzano
- Istituto di Biochimica e Chimica Clinica, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italy
- Human Microbiome Unit, Bambino Gesù Children's Hospital IRCCS, Rome, Italy
| | - Giuseppe Maulucci
- Istituto di Fisica, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italy
| | - Claudio De Angelis
- Istituto di Fisica, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italy
| | - Marco De Spirito
- Istituto di Fisica, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italy
| | - Laura Pacini
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
- UniCamillus, Saint Camillus International University of Health and Medical Sciences, Rome, Italy
| | - Laura D'Andrea
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | - Rino Ragno
- Department of Chemistry and Technologies of Drugs, Sapienza University, Rome, Italy
| | - Giulia Stazi
- Department of Chemistry and Technologies of Drugs, Sapienza University, Rome, Italy
| | - Sergio Valente
- Department of Chemistry and Technologies of Drugs, Sapienza University, Rome, Italy
| | - Antonello Mai
- Department of Chemistry and Technologies of Drugs, Sapienza University, Rome, Italy
| | - Pietro Chiurazzi
- Istituto di Medicina Genomica, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo F. Vito 1, 00168, Roma, Italy
- UOC Genetica Medica, Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italy
| | - Maurizio Genuardi
- Istituto di Medicina Genomica, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo F. Vito 1, 00168, Roma, Italy
- UOC Genetica Medica, Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italy
| | - Giovanni Neri
- Istituto di Medicina Genomica, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo F. Vito 1, 00168, Roma, Italy
- Self Research Institute, Greenwood Genetic Center, Greenwood, SC, USA
| | - Elisabetta Tabolacci
- Istituto di Medicina Genomica, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo F. Vito 1, 00168, Roma, Italy.
| |
Collapse
|
17
|
Formicola N, Vijayakumar J, Besse F. Neuronal ribonucleoprotein granules: Dynamic sensors of localized signals. Traffic 2019; 20:639-649. [PMID: 31206920 DOI: 10.1111/tra.12672] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 06/03/2019] [Accepted: 06/11/2019] [Indexed: 12/14/2022]
Abstract
Membrane-less organelles, because of their capacity to dynamically, selectively and reversibly concentrate molecules, are very well adapted for local information processing and rapid response to environmental fluctuations. These features are particularly important in the context of neuronal cells, where synapse-specific activation, or localized extracellular cues, induce signaling events restricted to specialized axonal or dendritic subcompartments. Neuronal ribonucleoprotein (RNP) particles, or granules, are nonmembrane bound macromolecular condensates that concentrate specific sets of mRNAs and regulatory proteins, promoting their long-distance transport to axons or dendrites. Neuronal RNP granules also have a dual function in regulating the translation of associated mRNAs: while preventing mRNA translation at rest, they fuel local protein synthesis upon activation. As revealed by recent work, rapid and reversible switches between these two functional modes are triggered by modifications of the networks of interactions underlying RNP granule assembly. Such flexible properties also come with a cost, as neuronal RNP granules are prone to transition into pathological aggregates in response to mutations, aging, or cellular stresses, further emphasizing the need to better understand the mechanistic principles governing their dynamic assembly and regulation in living systems.
Collapse
|
18
|
Kinase pathway inhibition restores PSD95 induction in neurons lacking fragile X mental retardation protein. Proc Natl Acad Sci U S A 2019; 116:12007-12012. [PMID: 31118285 DOI: 10.1073/pnas.1812056116] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Fragile X syndrome (FXS) is the leading monogenic cause of autism and intellectual disability. FXS is caused by loss of expression of fragile X mental retardation protein (FMRP), an RNA-binding protein that regulates translation of numerous mRNA targets, some of which are present at synapses. While protein synthesis deficits have long been postulated as an etiology of FXS, how FMRP loss affects distributions of newly synthesized proteins is unknown. Here we investigated the role of FMRP in regulating expression of new copies of the synaptic protein PSD95 in an in vitro model of synaptic plasticity. We find that local BDNF application promotes persistent accumulation of new PSD95 at stimulated synapses and dendrites of cultured neurons, and that this accumulation is absent in FMRP-deficient mouse neurons. New PSD95 accumulation at sites of BDNF stimulation does not require known mechanisms regulating FMRP-mRNA interactions but instead requires the PI3K-mTORC1-S6K1 pathway. Surprisingly, in FMRP-deficient neurons, BDNF induction of new PSD95 accumulation can be restored by mTORC1-S6K1 blockade, suggesting that constitutively high mTORC1-S6K1 activity occludes PSD95 regulation by BDNF and that alternative pathways exist to mediate induction when mTORC1-S6K1 is inhibited. This study provides direct evidence for deficits in local protein synthesis and accumulation of newly synthesized protein in response to local stimulation in FXS, and supports mTORC1-S6K1 pathway inhibition as a potential therapeutic approach for FXS.
Collapse
|
19
|
Bagni C, Zukin RS. A Synaptic Perspective of Fragile X Syndrome and Autism Spectrum Disorders. Neuron 2019; 101:1070-1088. [PMID: 30897358 PMCID: PMC9628679 DOI: 10.1016/j.neuron.2019.02.041] [Citation(s) in RCA: 227] [Impact Index Per Article: 37.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 02/25/2019] [Accepted: 02/27/2019] [Indexed: 12/28/2022]
Abstract
Altered synaptic structure and function is a major hallmark of fragile X syndrome (FXS), autism spectrum disorders (ASDs), and other intellectual disabilities (IDs), which are therefore classified as synaptopathies. FXS and ASDs, while clinically and genetically distinct, share significant comorbidity, suggesting that there may be a common molecular and/or cellular basis, presumably at the synapse. In this article, we review brain architecture and synaptic pathways that are dysregulated in FXS and ASDs, including spine architecture, signaling in synaptic plasticity, local protein synthesis, (m)RNA modifications, and degradation. mRNA repression is a powerful mechanism for the regulation of synaptic structure and efficacy. We infer that there is no single pathway that explains most of the etiology and discuss new findings and the implications for future work directed at improving our understanding of the pathogenesis of FXS and related ASDs and the design of therapeutic strategies to ameliorate these disorders.
Collapse
Affiliation(s)
- Claudia Bagni
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, Switzerland; Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy.
| | - R Suzanne Zukin
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, New York City, NY, USA.
| |
Collapse
|
20
|
Jacquemont S, Pacini L, Jønch AE, Cencelli G, Rozenberg I, He Y, D'Andrea L, Pedini G, Eldeeb M, Willemsen R, Gasparini F, Tassone F, Hagerman R, Gomez-Mancilla B, Bagni C. Protein synthesis levels are increased in a subset of individuals with fragile X syndrome. Hum Mol Genet 2019; 27:2039-2051. [PMID: 29590342 PMCID: PMC5985734 DOI: 10.1093/hmg/ddy099] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Accepted: 03/15/2018] [Indexed: 12/15/2022] Open
Abstract
Fragile X syndrome (FXS) is a monogenic form of intellectual disability and autism spectrum disorder caused by the absence of the fragile X mental retardation protein (FMRP). In biological models for the disease, this leads to upregulated mRNA translation and as a consequence, deficits in synaptic architecture and plasticity. Preclinical studies revealed that pharmacological interventions restore those deficits, which are thought to mediate the FXS cognitive and behavioral symptoms. Here, we characterized the de novo rate of protein synthesis in patients with FXS and their relationship with clinical severity. We measured the rate of protein synthesis in fibroblasts derived from 32 individuals with FXS and from 17 controls as well as in fibroblasts and primary neurons of 27 Fmr1 KO mice and 20 controls. Here, we show that levels of protein synthesis are increased in fibroblasts of individuals with FXS and Fmr1 KO mice. However, this cellular phenotype displays a broad distribution and a proportion of fragile X individuals and Fmr1 KO mice do not show increased levels of protein synthesis, having measures in the normal range. Because the same Fmr1 KO animal measures in fibroblasts predict those in neurons we suggest the validity of this peripheral biomarker. Our study offers a potential explanation for the comprehensive drug development program undertaken thus far yielding negative results and suggests that a significant proportion, but not all individuals with FXS, may benefit from the reduction of excessive levels of protein synthesis.
Collapse
Affiliation(s)
- Sébastien Jacquemont
- Sainte-Justine University Hospital Research Centre, Montreal, QC H3T 1C5.,University of Montreal, Montreal, QC H3T 1J4, Canada
| | - Laura Pacini
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Aia E Jønch
- Department of Clinical Genetics, Odense University Hospital.,Human Genetics, Department of Clinical Research, University of Southern Denmark, 5000 Odense, Denmark
| | - Giulia Cencelli
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Izabela Rozenberg
- Neuroscience Translational Medicine, Novartis Institutes for Biomedical Research, Novartis Pharma AG, 4056 Basel, Switzerland
| | - Yunsheng He
- Biomarker Development, Novartis Institutes for Biomedical Research, Cambridge, MA 02139, USA
| | - Laura D'Andrea
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Giorgia Pedini
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Marwa Eldeeb
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute, University of California, Davis Medical Center, Sacramento, CA 95817, USA
| | - Rob Willemsen
- Department of Clinical Genetics, Erasmus Medical Center, 1738, 3000DR Rotterdam, The Netherlands
| | - Fabrizio Gasparini
- Neuroscience Discovery, Novartis Institutes for BioMedical Research, 4002 Basel, Switzerland
| | - Flora Tassone
- Department of Biochemistry and Molecular Medicine and Medical Investigation of Neurodevelopmental Disorders (MIND) Institute, Sacramento, CA 95817, USA
| | - Randi Hagerman
- Department of Pediatric and Medical Investigation of Neurodevelopmental Disorders (MIND) Institute, University of California Davis, School of Medicine, Sacramento, CA 95817, USA
| | - Baltazar Gomez-Mancilla
- Neuroscience Translational Medicine, Novartis Institutes for Biomedical Research, Novartis Pharma AG, 4056 Basel, Switzerland.,Department of Neurology and Neurosurgery, McGill University, Montreal, QC H3A 0G4, Canada
| | - Claudia Bagni
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy.,Department of Fundamental Neuroscience, University of Lausanne, 1005 Lausanne, Switzerland
| |
Collapse
|
21
|
Rocchi A, Moretti D, Lignani G, Colombo E, Scholz-Starke J, Baldelli P, Tkatch T, Benfenati F. Neurite-Enriched MicroRNA-218 Stimulates Translation of the GluA2 Subunit and Increases Excitatory Synaptic Strength. Mol Neurobiol 2019; 56:5701-5714. [DOI: 10.1007/s12035-019-1492-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Accepted: 01/10/2019] [Indexed: 12/19/2022]
|
22
|
Fernández E, Gennaro E, Pirozzi F, Baldo C, Forzano F, Turolla L, Faravelli F, Gastaldo D, Coviello D, Grasso M, Bagni C. FXS-Like Phenotype in Two Unrelated Patients Carrying a Methylated Premutation of the FMR1 Gene. Front Genet 2018; 9:442. [PMID: 30450110 PMCID: PMC6224343 DOI: 10.3389/fgene.2018.00442] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Accepted: 09/14/2018] [Indexed: 12/31/2022] Open
Abstract
Fragile X syndrome (FXS) is mostly caused by two distinct events that occur in the FMR1 gene (Xq27.3): an expansion above 200 repeats of a CGG triplet located in the 5′UTR of the gene, and methylation of the cytosines located in the CpG islands upstream of the CGG repeats. Here, we describe two unrelated families with one FXS child and another sibling presenting mild intellectual disability and behavioral features evocative of FXS. Genetic characterization of the undiagnosed sibling revealed mosaicism in both the CGG expansion size and the methylation levels in the different tissues analyzed. This report shows that in the same family, two siblings carrying different CGG repeats, one in the full-mutation range and the other in the premutation range, present methylation mosaicism and consequent decreased FMRP production leading to FXS and FXS-like features, respectively. Decreased FMRP levels, more than the number of repeats seem to correlate with the severity of FXS clinical phenotypes.
Collapse
Affiliation(s)
- Esperanza Fernández
- Center for Human Genetics, KU Leuven, Leuven, Belgium.,VIB & KU Leuven Center for Brain & Disease Research, Leuven, Belgium
| | - Elena Gennaro
- Laboratorio di Genetica Umana, Ospedali Galliera, Genoa, Italy
| | - Filomena Pirozzi
- Center for Human Genetics, KU Leuven, Leuven, Belgium.,VIB & KU Leuven Center for Brain & Disease Research, Leuven, Belgium
| | - Chiara Baldo
- Laboratorio di Genetica Umana, Ospedali Galliera, Genoa, Italy
| | - Francesca Forzano
- Clinical Genetics Department, Borough Wing Guy's Hospital, Guy's and St Thomas' NHS Foundation Trust, London, United Kingdom.,S.S.D. Genetica Medica, Ospedali Galliera, Genoa, Italy
| | - Licia Turolla
- U.O.S. Genetica Medica, Azienda ULSS 2, Treviso, Italy
| | - Francesca Faravelli
- Clinical Genetics Department, Great Ormond Street Hospital, London, United Kingdom
| | - Denise Gastaldo
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, Switzerland
| | | | - Marina Grasso
- Laboratorio di Genetica Umana, Ospedali Galliera, Genoa, Italy
| | - Claudia Bagni
- Center for Human Genetics, KU Leuven, Leuven, Belgium.,VIB & KU Leuven Center for Brain & Disease Research, Leuven, Belgium.,Department of Fundamental Neurosciences, University of Lausanne, Lausanne, Switzerland.,Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| |
Collapse
|
23
|
Handling FMRP and its molecular partners: Structural insights into Fragile X Syndrome. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2018; 141:3-14. [PMID: 30905341 DOI: 10.1016/j.pbiomolbio.2018.07.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Accepted: 07/01/2018] [Indexed: 12/29/2022]
Abstract
Fragile X Mental Retardation Protein (FMRP) is a RNA-binding protein (RBP) known to control different steps of mRNA metabolism, even though its complete function is not fully understood yet. Lack or mutations of FMRP lead to Fragile X Syndrome (FXS), the most common form of inherited intellectual disability and a leading monogenic cause of autism spectrum disorder (ASD). It is well established that FMRP has a multi-domain architecture, a feature that allows this RBP to be engaged in a large interaction network with numerous proteins and mRNAs or non-coding RNAs. Insights into the three-dimensional (3D) structure of parts of its three domains (N-terminus, central domain and C-terminus) were obtained using Nuclear Magnetic Resonance and X-ray diffraction, but the complete 3D arrangement of each domain with respect to the others is still missing. Here, we review the structural features of FMRP and of the network of its protein and RNA interactions. Understanding these aspects is the first necessary step towards the design of novel compounds for new therapeutic interventions in FXS.
Collapse
|
24
|
Zalfa F, Panasiti V, Carotti S, Zingariello M, Perrone G, Sancillo L, Pacini L, Luciani F, Roberti V, D'Amico S, Coppola R, Abate SO, Rana RA, De Luca A, Fiers M, Melocchi V, Bianchi F, Farace MG, Achsel T, Marine JC, Morini S, Bagni C. The fragile X mental retardation protein regulates tumor invasiveness-related pathways in melanoma cells. Cell Death Dis 2017; 8:e3169. [PMID: 29144507 PMCID: PMC5775405 DOI: 10.1038/cddis.2017.521] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Revised: 08/24/2017] [Accepted: 08/25/2017] [Indexed: 02/06/2023]
Abstract
The fragile X mental retardation protein (FMRP) is lacking or mutated in patients with the fragile X syndrome (FXS), the most frequent form of inherited intellectual disability. FMRP affects metastasis formation in a mouse model for breast cancer. Here we show that FMRP is overexpressed in human melanoma with high Breslow thickness and high Clark level. Furthermore, meta-analysis of the TCGA melanoma data revealed that high levels of FMRP expression correlate significantly with metastatic tumor tissues, risk of relapsing and disease-free survival. Reduction of FMRP in metastatic melanoma cell lines impinges on cell migration, invasion and adhesion. Next-generation sequencing in human melanoma cells revealed that FMRP regulates a large number of mRNAs involved in relevant processes of melanoma progression. Our findings suggest an association between FMRP levels and the invasive phenotype in melanoma and might open new avenues towards the discovery of novel therapeutic targets.
Collapse
Affiliation(s)
- Francesca Zalfa
- Department of Medicine, Campus Bio-Medico University, via Alvaro del Portillo 21, 00128 Rome, Italy
| | - Vincenzo Panasiti
- Department of Medicine, Campus Bio-Medico University, via Alvaro del Portillo 21, 00128 Rome, Italy
| | - Simone Carotti
- Department of Medicine, Campus Bio-Medico University, via Alvaro del Portillo 21, 00128 Rome, Italy
| | - Maria Zingariello
- Department of Medicine, Campus Bio-Medico University, via Alvaro del Portillo 21, 00128 Rome, Italy
| | - Giuseppe Perrone
- Department of Medicine, Campus Bio-Medico University, via Alvaro del Portillo 21, 00128 Rome, Italy
| | - Laura Sancillo
- Department of Medicine and Science of Aging, University of Chieti 'G d'Annunzio', via dei Vestini 31, 66100 Chieti-Pescara, Italy
| | - Laura Pacini
- Department of Biomedicine and Prevention, University of Rome 'Tor Vergata', via Montpellier 1, 00133 Rome, Italy
| | - Flavie Luciani
- VIB/Center for the Biology of Disease, KU Leuven, O&N 4, Herestraat 49 Box 602, 3000, Leuven, Belgium.,Center for Human Genetics, Leuven Institute for Neuroscience and Disease, KU Leuven, O&N 4, Herestraat 49 Box 602, Leuven, 3000, Belgium
| | - Vincenzo Roberti
- Department of Dermatology, University of Rome 'La Sapienza', viale dell'Università 1, 00185 Rome, Italy
| | - Silvia D'Amico
- Department of Biomedicine and Prevention, University of Rome 'Tor Vergata', via Montpellier 1, 00133 Rome, Italy
| | - Rosa Coppola
- Department of Medicine, Campus Bio-Medico University, via Alvaro del Portillo 21, 00128 Rome, Italy
| | - Simona Osella Abate
- Department of Medical Science and Human Oncology, Section of Dermato-Oncology, University of Turin, via Verdi 8, 10124 Turin, Italy
| | - Rosa Alba Rana
- Department of Medicine and Science of Aging, University of Chieti 'G d'Annunzio', via dei Vestini 31, 66100 Chieti-Pescara, Italy
| | - Anastasia De Luca
- Department of Biomedicine and Prevention, University of Rome 'Tor Vergata', via Montpellier 1, 00133 Rome, Italy
| | - Mark Fiers
- VIB/Center for the Biology of Disease, KU Leuven, O&N 4, Herestraat 49 Box 602, 3000, Leuven, Belgium.,Center for Human Genetics, Leuven Institute for Neuroscience and Disease, KU Leuven, O&N 4, Herestraat 49 Box 602, Leuven, 3000, Belgium
| | - Valentina Melocchi
- ISBREMIT, Institute for Stem-cell Biology, Regenerative Medicine and Innovative Therapies, IRCCS Casa Sollievo della Sofferenza, viale Padre Pio 7, 71013 San Giovanni Rotondo (FG), Italy
| | - Fabrizio Bianchi
- ISBREMIT, Institute for Stem-cell Biology, Regenerative Medicine and Innovative Therapies, IRCCS Casa Sollievo della Sofferenza, viale Padre Pio 7, 71013 San Giovanni Rotondo (FG), Italy
| | - Maria Giulia Farace
- Department of Biomedicine and Prevention, University of Rome 'Tor Vergata', via Montpellier 1, 00133 Rome, Italy
| | - Tilmann Achsel
- VIB/Center for the Biology of Disease, KU Leuven, O&N 4, Herestraat 49 Box 602, 3000, Leuven, Belgium.,Center for Human Genetics, Leuven Institute for Neuroscience and Disease, KU Leuven, O&N 4, Herestraat 49 Box 602, Leuven, 3000, Belgium
| | - Jean-Christophe Marine
- VIB/Center for the Biology of Disease, KU Leuven, O&N 4, Herestraat 49 Box 602, 3000, Leuven, Belgium.,Center for Human Genetics, Leuven Institute for Neuroscience and Disease, KU Leuven, O&N 4, Herestraat 49 Box 602, Leuven, 3000, Belgium
| | - Sergio Morini
- Department of Medicine, Campus Bio-Medico University, via Alvaro del Portillo 21, 00128 Rome, Italy
| | - Claudia Bagni
- Department of Biomedicine and Prevention, University of Rome 'Tor Vergata', via Montpellier 1, 00133 Rome, Italy.,VIB/Center for the Biology of Disease, KU Leuven, O&N 4, Herestraat 49 Box 602, 3000, Leuven, Belgium.,Center for Human Genetics, Leuven Institute for Neuroscience and Disease, KU Leuven, O&N 4, Herestraat 49 Box 602, Leuven, 3000, Belgium.,Department of Fundamental Neuroscience, University of Lausanne, Rue du Bugnon 9, 1005 Lausanne, Switzerland
| |
Collapse
|
25
|
Filippini A, Bonini D, Lacoux C, Pacini L, Zingariello M, Sancillo L, Bosisio D, Salvi V, Mingardi J, La Via L, Zalfa F, Bagni C, Barbon A. Absence of the Fragile X Mental Retardation Protein results in defects of RNA editing of neuronal mRNAs in mouse. RNA Biol 2017. [PMID: 28640668 PMCID: PMC5785225 DOI: 10.1080/15476286.2017.1338232] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022] Open
Abstract
The fragile X syndrome (FXS), the most common form of inherited intellectual disability, is due to the absence of FMRP, a protein regulating RNA metabolism. Recently, an unexpected function of FMRP in modulating the activity of Adenosine Deaminase Acting on RNA (ADAR) enzymes has been reported both in Drosophila and Zebrafish. ADARs are RNA-binding proteins that increase transcriptional complexity through a post-transcriptional mechanism called RNA editing. To evaluate the ADAR2-FMRP interaction in mammals we analyzed several RNA editing re-coding sites in the fmr1 knockout (KO) mice. Ex vivo and in vitro analysis revealed that absence of FMRP leads to an increase in the editing levels of brain specific mRNAs, indicating that FMRP might act as an inhibitor of editing activity. Proximity Ligation Assay (PLA) in mouse primary cortical neurons and in non-neuronal cells revealed that ADAR2 and FMRP co-localize in the nucleus. The ADAR2-FMRP co-localization was further observed by double-immunogold Electron Microscopy (EM) in the hippocampus. Moreover, ADAR2-FMRP interaction appeared to be RNA independent. Because changes in the editing pattern are associated with neuropsychiatric and neurodevelopmental disorders, we propose that the increased editing observed in the fmr1-KO mice might contribute to the FXS molecular phenotypes.
Collapse
Affiliation(s)
- Alice Filippini
- a Biology and Genetic Division; Department of Molecular and Translational Medicine; University of Brescia ; Brescia , Italy
| | - Daniela Bonini
- a Biology and Genetic Division; Department of Molecular and Translational Medicine; University of Brescia ; Brescia , Italy
| | - Caroline Lacoux
- b Department of Biomedicine and Prevention , University of Rome Tor Vergata , Rome , Italy
| | - Laura Pacini
- b Department of Biomedicine and Prevention , University of Rome Tor Vergata , Rome , Italy
| | - Maria Zingariello
- c Department of Medicine , Campus Bio-Medico University , via Álvaro del Portillo 21, Rome , Italy
| | - Laura Sancillo
- d Department of Medicine and Aging Sciences, Section of Human Morphology , University G. D'Annunzio of Chieti-Pescara , Chieti , Italy
| | - Daniela Bosisio
- e Immunology Unit; Department of Molecular and Translational Medicine; University of Brescia ; Brescia , Italy
| | - Valentina Salvi
- e Immunology Unit; Department of Molecular and Translational Medicine; University of Brescia ; Brescia , Italy
| | - Jessica Mingardi
- a Biology and Genetic Division; Department of Molecular and Translational Medicine; University of Brescia ; Brescia , Italy
| | - Luca La Via
- a Biology and Genetic Division; Department of Molecular and Translational Medicine; University of Brescia ; Brescia , Italy
| | - Francesca Zalfa
- c Department of Medicine , Campus Bio-Medico University , via Álvaro del Portillo 21, Rome , Italy
| | - Claudia Bagni
- b Department of Biomedicine and Prevention , University of Rome Tor Vergata , Rome , Italy.,f VIB Center for the Biology of Disease and Center for Human Genetics , Leuven , Belgium.,g Department of Fundamental Neuroscience , University of Lausanne , Lausanne , Switzerland
| | - Alessandro Barbon
- a Biology and Genetic Division; Department of Molecular and Translational Medicine; University of Brescia ; Brescia , Italy
| |
Collapse
|
26
|
The non-coding RNA BC1 regulates experience-dependent structural plasticity and learning. Nat Commun 2017; 8:293. [PMID: 28819097 PMCID: PMC5561022 DOI: 10.1038/s41467-017-00311-2] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Accepted: 06/19/2017] [Indexed: 11/09/2022] Open
Abstract
The brain cytoplasmic (BC1) RNA is a non-coding RNA (ncRNA) involved in neuronal translational control. Absence of BC1 is associated with altered glutamatergic transmission and maladaptive behavior. Here, we show that pyramidal neurons in the barrel cortex of BC1 knock out (KO) mice display larger excitatory postsynaptic currents and increased spontaneous activity in vivo. Furthermore, BC1 KO mice have enlarged spine heads and postsynaptic densities and increased synaptic levels of glutamate receptors and PSD-95. Of note, BC1 KO mice show aberrant structural plasticity in response to whisker deprivation, impaired texture novel object recognition and altered social behavior. Thus, our study highlights a role for BC1 RNA in experience-dependent plasticity and learning in the mammalian adult neocortex, and provides insight into the function of brain ncRNAs regulating synaptic transmission, plasticity and behavior, with potential relevance in the context of intellectual disabilities and psychiatric disorders. Brain cytoplasmic (BC1) RNA is a non-coding RNA that has been implicated in translational regulation, seizure, and anxiety. Here, the authors show that in the cortex, BC1 RNA is required for sensory deprivation-induced structural plasticity of dendritic spines, as well as for correct sensory learning and social behaviors.
Collapse
|
27
|
Pellerin D, Lortie A, Corbin F. Platelets as a surrogate disease model of neurodevelopmental disorders: Insights from Fragile X Syndrome. Platelets 2017; 29:113-124. [PMID: 28660769 DOI: 10.1080/09537104.2017.1317733] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Fragile X Syndrome (FXS) is the most common inherited form of intellectual disability and the leading monogenic cause of autism spectrum disorders (ASD). Despite a large number of therapeutics developed in past years, there is currently no targeted treatment approved for FXS. In fact, translation of the positive and very promising preclinical findings from animal models to human subjects has so far fallen short owing in part to the low predictive validity of the Fmr1 ko mouse, an overly simplistic model of the complex human disease. This issue stresses the critical need to identify new surrogate human peripheral cell models of FXS, which may in fact allow for the identification of novel and more efficient therapies. Of all described models, blood platelets appear to be one of the most promising and appropriate disease models of FXS, in part owing to their close biochemical similarities with neurons. Noteworthy, they also recapitulate some of FXS neuron's core molecular dysregulations, such as hyperactivity of the MAPK/ERK and PI3K/Akt/mTOR pathways, elevated enzymatic activity of MMP9 and decreased production of cAMP. Platelets might therefore help furthering our understanding of FXS pathophysiology and might also lead to the identification of disease-specific biomarkers, as was shown in several psychiatric disorders such as schizophrenia and Alzheimer's disease. Moreover, there is additional evidence suggesting that platelet signaling may assist with prediction of cognitive phenotype and could represent a potent readout of drug efficacy in clinical trials. Globally, given the neurobiological overlap between different forms of intellectual disability, platelets may be a valuable window to access the molecular underpinnings of ASD and other neurodevelopmental disorders (NDD) sharing similar synaptic plasticity defects with FXS. Platelets are indeed an attractive model for unraveling pathophysiological mechanisms involved in NDD as well as to search for diagnostic and therapeutic biomarkers.
Collapse
Affiliation(s)
- David Pellerin
- a Department of Biochemistry, Faculty of Medicine and Health Sciences , Université de Sherbrooke , Sherbrooke , QC , Canada.,b Department of Neurology and Neurosurgery, Faculty of Medicine , McGill University , Montreal , QC , Canada
| | - Audrey Lortie
- a Department of Biochemistry, Faculty of Medicine and Health Sciences , Université de Sherbrooke , Sherbrooke , QC , Canada
| | - François Corbin
- a Department of Biochemistry, Faculty of Medicine and Health Sciences , Université de Sherbrooke , Sherbrooke , QC , Canada
| |
Collapse
|
28
|
McCoy M, Poliquin-Duchesneau D, Corbin F. Molecular dynamics of FMRP and other RNA-binding proteins in MEG-01 differentiation: the role of mRNP complexes in non-neuronal development. Biochem Cell Biol 2016; 94:597-608. [DOI: 10.1139/bcb-2015-0131] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Asymmetrically differentiating cells are formed with the aid of RNA-binding proteins (RBPs), which can bind, stabilize, regulate, and transport target mRNAs. The loss of RBPs in neurons may lead to severe neurodevelopmental diseases such as the Fragile X Syndrome with the absence of the Fragile X Mental Retardation Protein (FMRP). Because the latter is ubiquitous and shares many similarities with other RBPs involved in the development of peripheral cells, we suggest that FMRP would have a role in the differentiation of all tissues where it is expressed. A MEG-01 differentiation model was, therefore, established to study the global developmental functions of FMRP. PMA induction of MEG-01 cells causes important morphological changes driven by cytoskeletal dynamics. Cytoskeleton change and colocalization analyses were performed by confocal microscopy and sucrose gradient fractionation. Total cellular protein content and de novo synthesis were also analyzed. Microtubular transport mediates the displacement of FMRP and other RBP-containing mRNP complexes towards regions of the cell in development. De novo protein synthesis decreases significantly upon differentiation and total protein content composition is altered. Because those results are comparable with those obtained in neurons, the absence of FMRP would have significant consequences in cells everywhere in the body. The latter should be further investigated to give a better understanding of the systemic implications of imbalances of FMRP and other functionally similar RBPs.
Collapse
Affiliation(s)
- M. McCoy
- Department of Biochemistry, Université de Sherbrooke, Sherbrooke, QC, Canada
- Department of Biochemistry, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - D. Poliquin-Duchesneau
- Department of Biochemistry, Université de Sherbrooke, Sherbrooke, QC, Canada
- Department of Biochemistry, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - F. Corbin
- Department of Biochemistry, Université de Sherbrooke, Sherbrooke, QC, Canada
- Department of Biochemistry, Université de Sherbrooke, Sherbrooke, QC, Canada
| |
Collapse
|
29
|
Doll CA, Broadie K. Activity-dependent FMRP requirements in development of the neural circuitry of learning and memory. Development 2016; 142:1346-56. [PMID: 25804740 DOI: 10.1242/dev.117127] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The activity-dependent refinement of neural circuit connectivity during critical periods of brain development is essential for optimized behavioral performance. We hypothesize that this mechanism is defective in fragile X syndrome (FXS), the leading heritable cause of intellectual disability and autism spectrum disorders. Here, we use optogenetic tools in the Drosophila FXS disease model to test activity-dependent dendritogenesis in two extrinsic neurons of the mushroom body (MB) learning and memory brain center: (1) the input projection neuron (PN) innervating Kenyon cells (KCs) in the MB calyx microglomeruli and (2) the output MVP2 neuron innervated by KCs in the MB peduncle. Both input and output neuron classes exhibit distinctive activity-dependent critical period dendritic remodeling. MVP2 arbors expand in Drosophila mutants null for fragile X mental retardation 1 (dfmr1), as well as following channelrhodopsin-driven depolarization during critical period development, but are reduced by halorhodopsin-driven hyperpolarization. Optogenetic manipulation of PNs causes the opposite outcome--reduced dendritic arbors following channelrhodopsin depolarization and expanded arbors following halorhodopsin hyperpolarization during development. Importantly, activity-dependent dendritogenesis in both neuron classes absolutely requires dfmr1 during one developmental window. These results show that dfmr1 acts in a neuron type-specific activity-dependent manner for sculpting dendritic arbors during early-use, critical period development of learning and memory circuitry in the Drosophila brain.
Collapse
Affiliation(s)
- Caleb A Doll
- Department of Biological Sciences, Department of Cell and Developmental Biology, The Vanderbilt Kennedy Center for Research on Human Development, Vanderbilt University and Medical Center, Nashville, TN 37235, USA
| | - Kendal Broadie
- Department of Biological Sciences, Department of Cell and Developmental Biology, The Vanderbilt Kennedy Center for Research on Human Development, Vanderbilt University and Medical Center, Nashville, TN 37235, USA
| |
Collapse
|
30
|
Burguete AS, Almeida S, Gao FB, Kalb R, Akins MR, Bonini NM. GGGGCC microsatellite RNA is neuritically localized, induces branching defects, and perturbs transport granule function. eLife 2015; 4:e08881. [PMID: 26650351 PMCID: PMC4758954 DOI: 10.7554/elife.08881] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Accepted: 11/30/2015] [Indexed: 12/14/2022] Open
Abstract
Microsatellite expansions are the leading cause of numerous neurodegenerative disorders. Here we demonstrate that GGGGCC and CAG microsatellite repeat RNAs associated with C9orf72 in amyotrophic lateral sclerosis/frontotemporal dementia and with polyglutamine diseases, respectively, localize to neuritic granules that undergo active transport into distal neuritic segments. In cultured mammalian spinal cord neurons, the presence of neuritic GGGGCC repeat RNA correlates with neuronal branching defects, and the repeat RNA localizes to granules that label with fragile X mental retardation protein (FMRP), a transport granule component. Using a Drosophila GGGGCC expansion disease model, we characterize dendritic branching defects that are modulated by FMRP and Orb2. The human orthologs of these modifiers are misregulated in induced pluripotent stem cell-differentiated neurons (iPSNs) from GGGGCC expansion carriers. These data suggest that expanded repeat RNAs interact with the messenger RNA transport and translation machinery, causing transport granule dysfunction. This could be a novel mechanism contributing to the neuronal defects associated with C9orf72 and other microsatellite expansion diseases.
Collapse
Affiliation(s)
| | - Sandra Almeida
- Department of Neurology, University of Massachusetts Medical School, Worcester, United States
| | - Fen-Biao Gao
- Department of Neurology, University of Massachusetts Medical School, Worcester, United States
| | - Robert Kalb
- Division of Neurology, Department of Pediatrics, Children's Hospital of Philadelphia, University of Pennsylvania School of Medicine, Philadelphia, United States
| | - Michael R Akins
- Department of Biology, Drexel University, Philadelphia, United States
| | - Nancy M Bonini
- Department of Biology, University of Pennsylvania, Philadelphia, United States
| |
Collapse
|
31
|
A Special Extract of Bacopa monnieri (CDRI-08)-Restored Memory in CoCl2-Hypoxia Mimetic Mice Is Associated with Upregulation of Fmr-1 Gene Expression in Hippocampus. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2015; 2015:347978. [PMID: 26413121 PMCID: PMC4564622 DOI: 10.1155/2015/347978] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Accepted: 04/01/2015] [Indexed: 11/18/2022]
Abstract
Fragile X mental retardation protein (FMRP) is a neuronal translational repressor and has been implicated in learning, memory, and cognition. However, the role of Bacopa monnieri extract (CDRI-08) in enhancing cognitive abilities in hypoxia-induced memory impairment via Fmr-1 gene expression is not known. Here, we have studied effects of CDRI-08 on the expression of Fmr-1 gene in the hippocampus of well validated cobalt chloride (CoCl2)-induced hypoxia mimetic mice and analyzed the data with alterations in spatial memory. Results obtained from Morris water maze test suggest that CoCl2 treatment causes severe loss of spatial memory and CDRI-08 is capable of reversing it towards that in the normal control mice. Our semiquantitative RT-PCR, Western blot, and immunofluorescence microscopic data reveal that CoCl2-induced hypoxia significantly upregulates the expression of Hif-1α and downregulates the Fmr-1 expression in the hippocampus, respectively. Further, CDRI-08 administration reverses the memory loss and this is correlated with significant downregulation of Hif-1α and upregulation of Fmr-1 expression. Our data are novel and may provide mechanisms of hypoxia-induced impairments in the spatial memory and action of CDRI-08 in the recovery of hypoxia led memory impairment involving Fmr-1 gene encoded protein called FMRP.
Collapse
|
32
|
Callaerts-Vegh Z, Ahmed T, Vermaercke B, Marynen P, Balschun D, Froyen G, D'Hooge R. Nxf7 deficiency impairs social exploration and spatio-cognitive abilities as well as hippocampal synaptic plasticity in mice. Front Behav Neurosci 2015. [PMID: 26217206 PMCID: PMC4498129 DOI: 10.3389/fnbeh.2015.00179] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Nuclear RNA export factors (NXF) are conserved in all metazoans and are deemed essential for shuttling RNA across the nuclear envelope and other post-transcriptional processes (such as mRNA metabolism, storage and stability). Disruption of human NXF5 has been implicated in intellectual and psychosocial disabilities. In the present report, we use recently described Nxf7 knockout (KO) mice as an experimental model to analyze in detail the behavioral consequences of clinical NXF5 deficiency. We examined male Nxf7 KO mice using an extended cognitive and behavioral test battery, and recorded extracellular field potentials in the hippocampal CA1 region. We observed various cognitive and behavioral changes including alterations in social exploration, impaired spatial learning and spatio-cognitive abilities. We also defined a new experimental paradigm to discriminate search strategies in Morris water maze and showed significant differences between Nxf7 KO and control animals. Furthermore, while we observed no difference in a nose poke suppression in an conditioned emotional response (CER) protocol, Nxf7 KO mice were impaired in discriminating between differentially reinforced cues in an auditory fear conditioning protocol. This distinct neurocognitive phenotype was accompanied by impaired hippocampal Long-term potentiation (LTP), while long-term depression (LTD) was not affected by Nxf7 deficiency. Our data demonstrate that disruption of murine Nxf7 leads to behavioral phenotypes that may relate to the intellectual and social deficits in patients with NXF5 deficiency.
Collapse
Affiliation(s)
| | - Tariq Ahmed
- Laboratory of Biological Psychology, University of Leuven, KU Leuven Leuven, Belgium
| | - Ben Vermaercke
- Laboratory of Biological Psychology, University of Leuven, KU Leuven Leuven, Belgium
| | - Peter Marynen
- Human Genome Laboratory, University of Leuven and VIB Center for the Biology of Disease Leuven, Belgium
| | - Detlef Balschun
- Laboratory of Biological Psychology, University of Leuven, KU Leuven Leuven, Belgium
| | - Guy Froyen
- Human Genome Laboratory, University of Leuven and VIB Center for the Biology of Disease Leuven, Belgium
| | - Rudi D'Hooge
- Laboratory of Biological Psychology, University of Leuven, KU Leuven Leuven, Belgium
| |
Collapse
|
33
|
Buxbaum AR, Yoon YJ, Singer RH, Park HY. Single-molecule insights into mRNA dynamics in neurons. Trends Cell Biol 2015; 25:468-75. [PMID: 26052005 DOI: 10.1016/j.tcb.2015.05.005] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Revised: 05/07/2015] [Accepted: 05/08/2015] [Indexed: 10/23/2022]
Abstract
Targeting of mRNAs to neuronal dendrites and axons plays an integral role in intracellular signaling, development, and synaptic plasticity. Single-molecule imaging of mRNAs in neurons and brain tissue has led to enhanced understanding of mRNA dynamics. Here we discuss aspects of mRNA regulation as revealed by single-molecule detection, which has led to quantitative analyses of mRNA diversity, localization, transport, and translation. These exciting new discoveries propel our understanding of the life of an mRNA in a neuron and how its activity is regulated at the single-molecule level.
Collapse
Affiliation(s)
- Adina R Buxbaum
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, USA; Gruss Lipper Biophotonics Center, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, USA; Janelia Farm Research Campus, Howard Hughes Medical Institute, 19700 Helix Drive, Ashburn, VA 20147, USA
| | - Young J Yoon
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, USA
| | - Robert H Singer
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, USA; Gruss Lipper Biophotonics Center, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, USA; Janelia Farm Research Campus, Howard Hughes Medical Institute, 19700 Helix Drive, Ashburn, VA 20147, USA
| | - Hye Yoon Park
- Janelia Farm Research Campus, Howard Hughes Medical Institute, 19700 Helix Drive, Ashburn, VA 20147, USA; Department of Physics and Astronomy, Seoul National University, Seoul 151-747, Korea; Institute of Molecular Biology and Genetics, Seoul National University, Seoul 151-742, Korea.
| |
Collapse
|
34
|
Wang Y, Sakano H, Beebe K, Brown MR, de Laat R, Bothwell M, Kulesza RJ, Rubel EW. Intense and specialized dendritic localization of the fragile X mental retardation protein in binaural brainstem neurons: a comparative study in the alligator, chicken, gerbil, and human. J Comp Neurol 2015; 522:2107-28. [PMID: 24318628 DOI: 10.1002/cne.23520] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2013] [Revised: 12/04/2013] [Accepted: 12/05/2013] [Indexed: 11/09/2022]
Abstract
Neuronal dendrites are structurally and functionally dynamic in response to changes in afferent activity. The fragile X mental retardation protein (FMRP) is an mRNA binding protein that regulates activity-dependent protein synthesis and morphological dynamics of dendrites. Loss and abnormal expression of FMRP occur in fragile X syndrome (FXS) and some forms of autism spectrum disorders. To provide further understanding of how FMRP signaling regulates dendritic dynamics, we examined dendritic expression and localization of FMRP in the reptilian and avian nucleus laminaris (NL) and its mammalian analogue, the medial superior olive (MSO), in rodents and humans. NL/MSO neurons are specialized for temporal processing of low-frequency sounds for binaural hearing, which is impaired in FXS. Protein BLAST analyses first demonstrate that the FMRP amino acid sequences in the alligator and chicken are highly similar to human FMRP with identical mRNA-binding and phosphorylation sites, suggesting that FMRP functions similarly across vertebrates. Immunocytochemistry further reveals that NL/MSO neurons have very high levels of dendritic FMRP in low-frequency hearing vertebrates including alligator, chicken, gerbil, and human. Remarkably, dendritic FMRP in NL/MSO neurons often accumulates at branch points and enlarged distal tips, loci known to be critical for branch-specific dendritic arbor dynamics. These observations support an important role for FMRP in regulating dendritic properties of binaural neurons that are essential for low-frequency sound localization and auditory scene segregation, and support the relevance of studying this regulation in nonhuman vertebrates that use low frequencies in order to further understand human auditory processing disorders.
Collapse
Affiliation(s)
- Yuan Wang
- Virginia Merrill Bloedel Hearing Research Center, Department of Otolaryngology-Head and Neck Surgery, University of Washington School of Medicine, Seattle, WA, 98195-7923
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Peredo J, Villacé P, Ortín J, de Lucas S. Human Staufen1 associates to miRNAs involved in neuronal cell differentiation and is required for correct dendritic formation. PLoS One 2014; 9:e113704. [PMID: 25423178 PMCID: PMC4244161 DOI: 10.1371/journal.pone.0113704] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2014] [Accepted: 10/27/2014] [Indexed: 11/19/2022] Open
Abstract
Double-stranded RNA-binding proteins are key elements in the intracellular localization of mRNA and its local translation. Staufen is a double-stranded RNA binding protein involved in the localised translation of specific mRNAs during Drosophila early development and neuronal cell fate. The human homologue Staufen1 forms RNA-containing complexes that include proteins involved in translation and motor proteins to allow their movement within the cell, but the mechanism underlying translation repression in these complexes is poorly understood. Here we show that human Staufen1-containing complexes contain essential elements of the gene silencing apparatus, like Ago1-3 proteins, and we describe a set of miRNAs specifically associated to complexes containing human Staufen1. Among these, miR-124 stands out as particularly relevant because it appears enriched in human Staufen1 complexes and is over-expressed upon differentiation of human neuroblastoma cells in vitro. In agreement with these findings, we show that expression of human Staufen1 is essential for proper dendritic arborisation during neuroblastoma cell differentiation, yet it is not necessary for maintenance of the differentiated state, and suggest potential human Staufen1 mRNA targets involved in this process.
Collapse
Affiliation(s)
- Joan Peredo
- Departamento de Biología Molecular y Celular, Centro Nacional de Biotecnología (CSIC), Madrid, Spain
- Ciber de Enfermedades Respiratorias (ISCIII), Madrid, Spain
| | - Patricia Villacé
- Departamento de Biología Molecular y Celular, Centro Nacional de Biotecnología (CSIC), Madrid, Spain
| | - Juan Ortín
- Departamento de Biología Molecular y Celular, Centro Nacional de Biotecnología (CSIC), Madrid, Spain
- Ciber de Enfermedades Respiratorias (ISCIII), Madrid, Spain
- * E-mail: (JO); (SdL)
| | - Susana de Lucas
- Departamento de Biología Molecular y Celular, Centro Nacional de Biotecnología (CSIC), Madrid, Spain
- Ciber de Enfermedades Respiratorias (ISCIII), Madrid, Spain
- * E-mail: (JO); (SdL)
| |
Collapse
|
36
|
La Fata G, Gärtner A, Domínguez-Iturza N, Dresselaers T, Dawitz J, Poorthuis RB, Averna M, Himmelreich U, Meredith RM, Achsel T, Dotti CG, Bagni C. FMRP regulates multipolar to bipolar transition affecting neuronal migration and cortical circuitry. Nat Neurosci 2014; 17:1693-700. [PMID: 25402856 DOI: 10.1038/nn.3870] [Citation(s) in RCA: 103] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2014] [Accepted: 10/16/2014] [Indexed: 12/20/2022]
Abstract
Deficiencies in fragile X mental retardation protein (FMRP) are the most common cause of inherited intellectual disability, fragile X syndrome (FXS), with symptoms manifesting during infancy and early childhood. Using a mouse model for FXS, we found that Fmrp regulates the positioning of neurons in the cortical plate during embryonic development, affecting their multipolar-to-bipolar transition (MBT). We identified N-cadherin, which is crucial for MBT, as an Fmrp-regulated target in embryonic brain. Furthermore, spontaneous network activity and high-resolution brain imaging revealed defects in the establishment of neuronal networks at very early developmental stages, further confirmed by an unbalanced excitatory and inhibitory network. Finally, reintroduction of Fmrp or N-cadherin in the embryo normalized early postnatal neuron activity. Our findings highlight the critical role of Fmrp in the developing cerebral cortex and might explain some of the clinical features observed in patients with FXS, such as alterations in synaptic communication and neuronal network connectivity.
Collapse
Affiliation(s)
- Giorgio La Fata
- 1] VIB Center for the Biology of Disease, KU Leuven, Leuven, Belgium. [2] Center for Human Genetics and Leuven Institute for Neurodegenerative Diseases (LIND), KU Leuven, Leuven, Belgium
| | - Annette Gärtner
- 1] VIB Center for the Biology of Disease, KU Leuven, Leuven, Belgium. [2] Center for Human Genetics and Leuven Institute for Neurodegenerative Diseases (LIND), KU Leuven, Leuven, Belgium
| | - Nuria Domínguez-Iturza
- 1] VIB Center for the Biology of Disease, KU Leuven, Leuven, Belgium. [2] Center for Human Genetics and Leuven Institute for Neurodegenerative Diseases (LIND), KU Leuven, Leuven, Belgium
| | | | - Julia Dawitz
- Center for Neurogenomics and Cognitive Research (CNCR), VU University Amsterdam, Amsterdam, the Netherlands
| | - Rogier B Poorthuis
- Center for Neurogenomics and Cognitive Research (CNCR), VU University Amsterdam, Amsterdam, the Netherlands
| | - Michele Averna
- 1] VIB Center for the Biology of Disease, KU Leuven, Leuven, Belgium. [2] Center for Human Genetics and Leuven Institute for Neurodegenerative Diseases (LIND), KU Leuven, Leuven, Belgium
| | | | - Rhiannon M Meredith
- Center for Neurogenomics and Cognitive Research (CNCR), VU University Amsterdam, Amsterdam, the Netherlands
| | - Tilmann Achsel
- 1] VIB Center for the Biology of Disease, KU Leuven, Leuven, Belgium. [2] Center for Human Genetics and Leuven Institute for Neurodegenerative Diseases (LIND), KU Leuven, Leuven, Belgium
| | - Carlos G Dotti
- 1] VIB Center for the Biology of Disease, KU Leuven, Leuven, Belgium. [2] Center for Human Genetics and Leuven Institute for Neurodegenerative Diseases (LIND), KU Leuven, Leuven, Belgium. [3] Centro de Biología Molecular Severo Ochoa, Campus de la Universidad Autónoma de Madrid, Spain
| | - Claudia Bagni
- 1] VIB Center for the Biology of Disease, KU Leuven, Leuven, Belgium. [2] Center for Human Genetics and Leuven Institute for Neurodegenerative Diseases (LIND), KU Leuven, Leuven, Belgium. [3] Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| |
Collapse
|
37
|
Santos AR, Kanellopoulos AK, Bagni C. Learning and behavioral deficits associated with the absence of the fragile X mental retardation protein: what a fly and mouse model can teach us. ACTA ACUST UNITED AC 2014; 21:543-55. [PMID: 25227249 PMCID: PMC4175497 DOI: 10.1101/lm.035956.114] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
The Fragile X syndrome (FXS) is the most frequent form of inherited mental disability and is considered a monogenic cause of autism spectrum disorder. FXS is caused by a triplet expansion that inhibits the expression of the FMR1 gene. The gene product, the Fragile X Mental Retardation Protein (FMRP), regulates mRNA metabolism in brain and nonneuronal cells. During brain development, FMRP controls the expression of key molecules involved in receptor signaling, cytoskeleton remodeling, protein synthesis and, ultimately, spine morphology. Symptoms associated with FXS include neurodevelopmental delay, cognitive impairment, anxiety, hyperactivity, and autistic-like behavior. Twenty years ago the first Fmr1 KO mouse to study FXS was generated, and several years later other key models including the mutant Drosophila melanogaster, dFmr1, have further helped the understanding of the cellular and molecular causes behind this complex syndrome. Here, we review to which extent these biological models are affected by the absence of FMRP, pointing out the similarities with the observed human dysfunction. Additionally, we discuss several potential treatments under study in animal models that are able to partially revert some of the FXS abnormalities.
Collapse
Affiliation(s)
- Ana Rita Santos
- VIB Center for the Biology of Disease, 3000 Leuven, Belgium Center for Human Genetics, KU Leuven, 3000 Leuven, Belgium Leuven Institute for Neurodegenerative Diseases (LIND), KU Leuven, 3000 Leuven, Belgium
| | - Alexandros K Kanellopoulos
- VIB Center for the Biology of Disease, 3000 Leuven, Belgium Center for Human Genetics, KU Leuven, 3000 Leuven, Belgium Leuven Institute for Neurodegenerative Diseases (LIND), KU Leuven, 3000 Leuven, Belgium
| | - Claudia Bagni
- VIB Center for the Biology of Disease, 3000 Leuven, Belgium Center for Human Genetics, KU Leuven, 3000 Leuven, Belgium Leuven Institute for Neurodegenerative Diseases (LIND), KU Leuven, 3000 Leuven, Belgium Department of Biomedicine and Prevention, University of Rome "Tor Vergata" 00133, Rome, Italy
| |
Collapse
|
38
|
Georgieva D, Petrova M, Molle E, Daskalovska I, Genova G. Drosophila DFMR1 Interacts with Genes of the Lgl-Pathway in the Brain Synaptic Architecture. BIOTECHNOL BIOTEC EQ 2014. [DOI: 10.5504/50yrtimb.2011.0010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
|
39
|
Fragile X syndrome: a preclinical review on metabotropic glutamate receptor 5 (mGluR5) antagonists and drug development. Psychopharmacology (Berl) 2014; 231:1217-26. [PMID: 24232444 DOI: 10.1007/s00213-013-3330-3] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2013] [Accepted: 10/14/2013] [Indexed: 10/26/2022]
Abstract
RATIONALE Fragile X syndrome (FXS) is considered the leading inherited cause of intellectual disability and autism. In FXS, the fragile X mental retardation 1 (FMR1) gene is silenced and the fragile X mental retardation protein (FMRP) is not expressed, resulting in the characteristic features of the syndrome. Despite recent advances in understanding the pathophysiology of FXS, there is still no cure for this condition; current treatment is symptomatic. Preclinical research is essential in the development of potential therapeutic agents. OBJECTIVES This review provides an overview of the preclinical evidence supporting metabotropic glutamate receptor 5 (mGluR5) antagonists as therapeutic agents for FXS. RESULTS According to the mGluR theory of FXS, the absence of FMRP leads to enhanced glutamatergic signaling via mGluR5, which leads to increased protein synthesis and defects in synaptic plasticity including enhanced long-term depression. As such, efforts to develop agents that target the underlying pathophysiology of FXS have focused on mGluR5 modulation. Animal models, particularly the Fmr1 knockout mouse model, have become invaluable in exploring therapeutic approaches on an electrophysiological, behavioral, biochemical, and neuroanatomical level. Two direct approaches are currently being investigated for FXS treatment: reactivating the FMR1 gene and compensating for the lack of FMRP. The latter approach has yielded promising results, with mGluR5 antagonists showing efficacy in clinical trials. CONCLUSIONS Targeting mGluR5 is a valid approach for the development of therapeutic agents that target the underlying pathophysiology of FXS. Several compounds are currently in development, with encouraging results.
Collapse
|
40
|
Sala C, Segal M. Dendritic spines: the locus of structural and functional plasticity. Physiol Rev 2014; 94:141-88. [PMID: 24382885 DOI: 10.1152/physrev.00012.2013] [Citation(s) in RCA: 346] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
The introduction of high-resolution time lapse imaging and molecular biological tools has changed dramatically the rate of progress towards the understanding of the complex structure-function relations in synapses of central spiny neurons. Standing issues, including the sequence of molecular and structural processes leading to formation, morphological change, and longevity of dendritic spines, as well as the functions of dendritic spines in neurological/psychiatric diseases are being addressed in a growing number of recent studies. There are still unsettled issues with respect to spine formation and plasticity: Are spines formed first, followed by synapse formation, or are synapses formed first, followed by emergence of a spine? What are the immediate and long-lasting changes in spine properties following exposure to plasticity-producing stimulation? Is spine volume/shape indicative of its function? These and other issues are addressed in this review, which highlights the complexity of molecular pathways involved in regulation of spine structure and function, and which contributes to the understanding of central synaptic interactions in health and disease.
Collapse
|
41
|
Doll CA, Broadie K. Impaired activity-dependent neural circuit assembly and refinement in autism spectrum disorder genetic models. Front Cell Neurosci 2014; 8:30. [PMID: 24570656 PMCID: PMC3916725 DOI: 10.3389/fncel.2014.00030] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Accepted: 01/21/2014] [Indexed: 01/23/2023] Open
Abstract
Early-use activity during circuit-specific critical periods refines brain circuitry by the coupled processes of eliminating inappropriate synapses and strengthening maintained synapses. We theorize these activity-dependent (A-D) developmental processes are specifically impaired in autism spectrum disorders (ASDs). ASD genetic models in both mouse and Drosophila have pioneered our insights into normal A-D neural circuit assembly and consolidation, and how these developmental mechanisms go awry in specific genetic conditions. The monogenic fragile X syndrome (FXS), a common cause of heritable ASD and intellectual disability, has been particularly well linked to defects in A-D critical period processes. The fragile X mental retardation protein (FMRP) is positively activity-regulated in expression and function, in turn regulates excitability and activity in a negative feedback loop, and appears to be required for the A-D remodeling of synaptic connectivity during early-use critical periods. The Drosophila FXS model has been shown to functionally conserve the roles of human FMRP in synaptogenesis, and has been centrally important in generating our current mechanistic understanding of the FXS disease state. Recent advances in Drosophila optogenetics, transgenic calcium reporters, highly-targeted transgenic drivers for individually-identified neurons, and a vastly improved connectome of the brain are now being combined to provide unparalleled opportunities to both manipulate and monitor A-D processes during critical period brain development in defined neural circuits. The field is now poised to exploit this new Drosophila transgenic toolbox for the systematic dissection of A-D mechanisms in normal versus ASD brain development, particularly utilizing the well-established Drosophila FXS disease model.
Collapse
Affiliation(s)
- Caleb A Doll
- Department of Biological Sciences, Vanderbilt University Nashville, TN, USA
| | - Kendal Broadie
- Department of Biological Sciences, Vanderbilt University Nashville, TN, USA ; Kennedy Center for Research on Human Development, Vanderbilt University Nashville, TN, USA
| |
Collapse
|
42
|
The Fragile X mental retardation protein regulates matrix metalloproteinase 9 mRNA at synapses. J Neurosci 2014; 33:18234-41. [PMID: 24227732 DOI: 10.1523/jneurosci.2207-13.2013] [Citation(s) in RCA: 91] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Activity-dependent protein synthesis at synapses is dysregulated in the Fragile X syndrome (FXS). This process contributes to dendritic spine dysmorphogenesis and synaptic dysfunction in FXS. Matrix Metalloproteinase 9 (MMP-9) is an enzyme involved in activity-dependent reorganization of dendritic spine architecture and was shown to regulate spine morphology in a mouse model of FXS, the Fmr1 knock-out mice. Here we show that MMP-9 mRNA is part of the FMRP complex and colocalizes in dendrites. In the absence of FMRP MMP-9 mRNA translation is increased at synapses, suggesting that this mechanism contributes to the increased metalloproteinase level at synapses of Fmr1 knock-out mice. We propose that such a local effect can contribute to the aberrant dendritic spine morphology observed in the Fmr1 knock-out mice and in patients with FXS.
Collapse
|
43
|
Fernández E, Rajan N, Bagni C. The FMRP regulon: from targets to disease convergence. Front Neurosci 2013; 7:191. [PMID: 24167470 PMCID: PMC3807044 DOI: 10.3389/fnins.2013.00191] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2013] [Accepted: 10/04/2013] [Indexed: 01/08/2023] Open
Abstract
The fragile X mental retardation protein (FMRP) is an RNA-binding protein that regulates mRNA metabolism. FMRP has been largely studied in the brain, where the absence of this protein leads to fragile X syndrome, the most frequent form of inherited intellectual disability. Since the identification of the FMRP gene in 1991, many studies have primarily focused on understanding the function/s of this protein. Hundreds of potential FMRP mRNA targets and several interacting proteins have been identified. Here, we report the identification of FMRP mRNA targets in the mammalian brain that support the key role of this protein during brain development and in regulating synaptic plasticity. We compared the genes from databases and genome-wide association studies with the brain FMRP transcriptome, and identified several FMRP mRNA targets associated with autism spectrum disorders, mood disorders and schizophrenia, showing a potential common pathway/s for these apparently different disorders.
Collapse
Affiliation(s)
- Esperanza Fernández
- Center for the Biology of Disease, Vlaams Institut voor Biotechnologie Leuven, Belgium ; Center for Human Genetics, Leuven Institute for Neuroscience and Disease, KU Leuven Leuven, Belgium
| | | | | |
Collapse
|
44
|
Bagni C, Oostra BA. Fragile X syndrome: From protein function to therapy. Am J Med Genet A 2013; 161A:2809-21. [PMID: 24115651 DOI: 10.1002/ajmg.a.36241] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2013] [Accepted: 08/28/2013] [Indexed: 12/23/2022]
Abstract
Fragile X syndrome (FXS) is the leading monogenic cause of intellectual disability and autism. The FMR1 gene contains a CGG repeat present in the 5'-untranslated region which can be unstable upon transmission to the next generation. The repeat is up to 55 CGGs long in the normal population. In patients with fragile X syndrome (FXS), a repeat length exceeding 200 CGGs generally leads to methylation of the repeat and the promoter region, which is accompanied by silencing of the FMR1 gene. The disease is a result of lack of expression of the fragile X mental retardation protein leading to severe symptoms, including intellectual disability, hyperactivity, and autistic-like behavior. The FMR1 protein (FMRP) has a number of functions. The translational dysregulation of a subset of mRNAs targeted by FMRP is probably the major contribution to FXS. FMRP is also involved in mRNA transport to synapses where protein synthesis occurs. For some FMRP-bound mRNAs, FMRP is a direct modulator of mRNA stability either by sustaining or preventing mRNA decay. Increased knowledge about the role of FMRP has led to the identification of potential treatments for fragile X syndrome that were often tested first in the different animal models. This review gives an overview about the present knowledge of the function of FMRP and the therapeutic strategies in mouse and man.
Collapse
Affiliation(s)
- Claudia Bagni
- VIB Center for the Biology of Disease, Catholic University of Leuven, Leuven, Belgium; Department of Biomedicine and Prevention, University of Rome, Tor Vergata, Italy
| | | |
Collapse
|
45
|
De Rubeis S, Pasciuto E, Li K, Fernández E, Di Marino D, Buzzi A, Ostroff L, Klann E, Zwartkruis FJ, Komiyama N, Grant SG, Poujol C, Choquet D, Achsel T, Posthuma D, Smit A, Bagni C. CYFIP1 coordinates mRNA translation and cytoskeleton remodeling to ensure proper dendritic spine formation. Neuron 2013; 79:1169-82. [PMID: 24050404 PMCID: PMC3781321 DOI: 10.1016/j.neuron.2013.06.039] [Citation(s) in RCA: 229] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/18/2013] [Indexed: 11/30/2022]
Abstract
The CYFIP1/SRA1 gene is located in a chromosomal region linked to various neurological disorders, including intellectual disability, autism, and schizophrenia. CYFIP1 plays a dual role in two apparently unrelated processes, inhibiting local protein synthesis and favoring actin remodeling. Here, we show that brain-derived neurotrophic factor (BDNF)-driven synaptic signaling releases CYFIP1 from the translational inhibitory complex, triggering translation of target mRNAs and shifting CYFIP1 into the WAVE regulatory complex. Active Rac1 alters the CYFIP1 conformation, as demonstrated by intramolecular FRET, and is key in changing the equilibrium of the two complexes. CYFIP1 thus orchestrates the two molecular cascades, protein translation and actin polymerization, each of which is necessary for correct spine morphology in neurons. The CYFIP1 interactome reveals many interactors associated with brain disorders, opening new perspectives to define regulatory pathways shared by neurological disabilities characterized by spine dysmorphogenesis.
Collapse
Affiliation(s)
- Silvia De Rubeis
- VIB Center for Biology of Disease, KULeuven, 3000 Leuven, Belgium
- Center for Human Genetics and Leuven Institute for Neuroscience and Disease (LIND), KULeuven, 3000 Leuven, Belgium
| | - Emanuela Pasciuto
- VIB Center for Biology of Disease, KULeuven, 3000 Leuven, Belgium
- Center for Human Genetics and Leuven Institute for Neuroscience and Disease (LIND), KULeuven, 3000 Leuven, Belgium
| | - Ka Wan Li
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, VU University Medical Center Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Esperanza Fernández
- VIB Center for Biology of Disease, KULeuven, 3000 Leuven, Belgium
- Center for Human Genetics and Leuven Institute for Neuroscience and Disease (LIND), KULeuven, 3000 Leuven, Belgium
| | - Daniele Di Marino
- VIB Center for Biology of Disease, KULeuven, 3000 Leuven, Belgium
- Center for Human Genetics and Leuven Institute for Neuroscience and Disease (LIND), KULeuven, 3000 Leuven, Belgium
| | - Andrea Buzzi
- VIB Center for Biology of Disease, KULeuven, 3000 Leuven, Belgium
- Center for Human Genetics and Leuven Institute for Neuroscience and Disease (LIND), KULeuven, 3000 Leuven, Belgium
| | | | - Eric Klann
- Center for Neural Science, New York University, New York, NY 10003, USA
| | - Fried J.T. Zwartkruis
- Molecular Cancer Research, Center for Biomedical Genetics and Cancer Genomics Center, University Medical Center Utrecht, 3584 CG Utrecht
| | - Noboru H. Komiyama
- Centre for Clinical Brain Sciences and Centre for Neuroregeneration, University of Edinburgh, Edinburgh EH16 4SB, UK
| | - Seth G.N. Grant
- Centre for Clinical Brain Sciences and Centre for Neuroregeneration, University of Edinburgh, Edinburgh EH16 4SB, UK
| | - Christel Poujol
- CNRS, Bordeaux Imaging Center, UMS 3420, 33000 Bordeaux, France
- University of Bordeaux, UMS 3420, 33077, Bordeaux, France
| | - Daniel Choquet
- CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, 33000 Bordeaux, France
- University of Bordeaux, UMR 5297, 33077, Bordeaux, France
| | - Tilmann Achsel
- VIB Center for Biology of Disease, KULeuven, 3000 Leuven, Belgium
- Center for Human Genetics and Leuven Institute for Neuroscience and Disease (LIND), KULeuven, 3000 Leuven, Belgium
| | - Danielle Posthuma
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, VU University Medical Center Amsterdam, 1081 HV Amsterdam, The Netherlands
- Department of Clinical Genetics, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, VU University Medical Center Amsterdam, 1081 HV Amsterdam, The Netherlands
- Department of Child and Adolescent Psychiatry, Erasmus University Medical Center/Sophia Child Hospital, 3000 CB Rotterdam, The Netherlands
| | - August B. Smit
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, VU University Medical Center Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Claudia Bagni
- VIB Center for Biology of Disease, KULeuven, 3000 Leuven, Belgium
- Center for Human Genetics and Leuven Institute for Neuroscience and Disease (LIND), KULeuven, 3000 Leuven, Belgium
- Department of Biomedicine and Prevention, University “Tor Vergata,” 00133 Rome, Italy
| |
Collapse
|
46
|
Waselus M, Flagel SB, Jedynak JP, Akil H, Robinson TE, Watson SJ. Long-term effects of cocaine experience on neuroplasticity in the nucleus accumbens core of addiction-prone rats. Neuroscience 2013; 248:571-84. [PMID: 23811073 PMCID: PMC3859827 DOI: 10.1016/j.neuroscience.2013.06.042] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2013] [Revised: 06/17/2013] [Accepted: 06/19/2013] [Indexed: 01/17/2023]
Abstract
Repeated exposure to drugs of abuse is associated with structural plasticity in brain reward pathways. Rats selectively bred for locomotor response to novelty differ on a number of neurobehavioral dimensions relevant to addiction. This unique genetic animal model was used here to examine both pre-existing differences and long-term consequences of repeated cocaine treatment on structural plasticity. Selectively bred high-responder (bHR) and low-responder (bLR) rats received repeated saline or cocaine injections for 9 consecutive days. Escalating doses of cocaine (7.5, 15 and 30 mg/kg) were administered on the first (day 1) and last (day 9) days of treatment and a single injection of the intermediate dose (15 mg/kg) was given on days 2-8. Motor activity in response to escalating doses of cocaine was compared on the first and last days of treatment to assess the acute and sensitized response to the drug. Following prolonged cocaine abstinence (28 days), spine density was examined on terminal dendrites of medium spiny neurons in the nucleus accumbens core. Relative to bLRs, bHRs exhibited increased psychomotor activation in response to both the acute and repeated effects of cocaine. There were no differences in spine density between bHR and bLR rats under basal conditions or following repeated saline treatment. However, spine density differed markedly between these two lines following prolonged cocaine abstinence. All spine types were decreased in cocaine-treated bHRs, while only mushroom spines were decreased in bLRs that received cocaine. Changes in spine density occurred specifically near the branch point of terminal dendrites. These findings indicate that structural plasticity associated with prolonged cocaine abstinence varies markedly in two selected strains of rats that vary on numerous traits relevant to addiction. Thus, genetic factors that contribute to individual variation in the behavioral response to cocaine also influence cocaine-induced structural plasticity.
Collapse
Affiliation(s)
- M Waselus
- Molecular and Behavioral Neuroscience Institute, University of Michigan, Ann Arbor, MI, USA.
| | - S B Flagel
- Molecular and Behavioral Neuroscience Institute, University of Michigan, Ann Arbor, MI, USA; Department of Psychiatry, University of Michigan, Ann Arbor, MI, USA; Neuroscience Program, University of Michigan, Ann Arbor, MI, USA; Department of Psychology, University of Michigan, Ann Arbor, MI, USA
| | - J P Jedynak
- Neuroscience Program, University of Michigan, Ann Arbor, MI, USA
| | - H Akil
- Molecular and Behavioral Neuroscience Institute, University of Michigan, Ann Arbor, MI, USA; Department of Psychiatry, University of Michigan, Ann Arbor, MI, USA; Neuroscience Program, University of Michigan, Ann Arbor, MI, USA
| | - T E Robinson
- Neuroscience Program, University of Michigan, Ann Arbor, MI, USA; Department of Psychology, University of Michigan, Ann Arbor, MI, USA
| | - S J Watson
- Molecular and Behavioral Neuroscience Institute, University of Michigan, Ann Arbor, MI, USA; Department of Psychiatry, University of Michigan, Ann Arbor, MI, USA; Neuroscience Program, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
47
|
Lucá R, Averna M, Zalfa F, Vecchi M, Bianchi F, La Fata G, Del Nonno F, Nardacci R, Bianchi M, Nuciforo P, Munck S, Parrella P, Moura R, Signori E, Alston R, Kuchnio A, Farace MG, Fazio VM, Piacentini M, De Strooper B, Achsel T, Neri G, Neven P, Evans DG, Carmeliet P, Mazzone M, Bagni C. The fragile X protein binds mRNAs involved in cancer progression and modulates metastasis formation. EMBO Mol Med 2013; 5:1523-36. [PMID: 24092663 PMCID: PMC3799577 DOI: 10.1002/emmm.201302847] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2013] [Revised: 08/01/2013] [Accepted: 08/06/2013] [Indexed: 01/30/2023] Open
Abstract
The role of the fragile X mental retardation protein (FMRP) is well established in brain, where its absence leads to the fragile X syndrome (FXS). FMRP is almost ubiquitously expressed, suggesting that, in addition to its effects in brain, it may have fundamental roles in other organs. There is evidence that FMRP expression can be linked to cancer. FMR1 mRNA, encoding FMRP, is overexpressed in hepatocellular carcinoma cells. A decreased risk of cancer has been reported in patients with FXS while a patient-case with FXS showed an unusual decrease of tumour brain invasiveness. However, a role for FMRP in regulating cancer biology, if any, remains unknown. We show here that FMRP and FMR1 mRNA levels correlate with prognostic indicators of aggressive breast cancer, lung metastases probability and triple negative breast cancer (TNBC). We establish that FMRP overexpression in murine breast primary tumours enhances lung metastasis while its reduction has the opposite effect regulating cell spreading and invasion. FMRP binds mRNAs involved in epithelial mesenchymal transition (EMT) and invasion including E-cadherin and Vimentin mRNAs, hallmarks of EMT and cancer progression.
Collapse
Affiliation(s)
- Rossella Lucá
- VIB Center for the Biology of Disease, Leuven, Belgium; Center for Human Genetics, KU Leuven, Belgium
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Cook D, Nuro E, Murai KK. Increasing our understanding of human cognition through the study of Fragile X Syndrome. Dev Neurobiol 2013; 74:147-77. [PMID: 23723176 PMCID: PMC4216185 DOI: 10.1002/dneu.22096] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2013] [Accepted: 05/17/2013] [Indexed: 12/16/2022]
Abstract
Fragile X Syndrome (FXS) is considered the most common form of inherited intellectual disability. It is caused by reductions in the expression level or function of a single protein, the Fragile X Mental Retardation Protein (FMRP), a translational regulator which binds to approximately 4% of brain messenger RNAs. Accumulating evidence suggests that FXS is a complex disorder of cognition, involving interactions between genetic and environmental influences, leading to difficulties in acquiring key life skills including motor skills, language, and proper social behaviors. Since many FXS patients also present with one or more features of autism spectrum disorders (ASDs), insights gained from studying the monogenic basis of FXS could pave the way to a greater understanding of underlying features of multigenic ASDs. Here we present an overview of the FXS and FMRP field with the goal of demonstrating how loss of a single protein involved in translational control affects multiple stages of brain development and leads to debilitating consequences on human cognition. We also focus on studies which have rescued or improved FXS symptoms in mice using genetic or therapeutic approaches to reduce protein expression. We end with a brief description of how deficits in translational control are implicated in FXS and certain cases of ASDs, with many recent studies demonstrating that ASDs are likely caused by increases or decreases in the levels of certain key synaptic proteins. The study of FXS and its underlying single genetic cause offers an invaluable opportunity to study how a single gene influences brain development and behavior.
Collapse
Affiliation(s)
- Denise Cook
- Department of Neurology and Neurosurgery, Centre for Research in Neuroscience, The Research Institute of the McGill University Health Centre, Montreal General Hospital, Montreal, Quebec, Canada
| | | | | |
Collapse
|
49
|
Sidorov MS, Auerbach BD, Bear MF. Fragile X mental retardation protein and synaptic plasticity. Mol Brain 2013; 6:15. [PMID: 23566911 PMCID: PMC3636002 DOI: 10.1186/1756-6606-6-15] [Citation(s) in RCA: 170] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2013] [Accepted: 03/25/2013] [Indexed: 12/20/2022] Open
Abstract
Loss of the translational repressor FMRP causes Fragile X syndrome. In healthy neurons, FMRP modulates the local translation of numerous synaptic proteins. Synthesis of these proteins is required for the maintenance and regulation of long-lasting changes in synaptic strength. In this role as a translational inhibitor, FMRP exerts profound effects on synaptic plasticity.
Collapse
Affiliation(s)
- Michael S Sidorov
- The Picower Institute for Learning and Memory, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139 46-3301, USA
| | | | | |
Collapse
|
50
|
Charalambous DC, Pasciuto E, Mercaldo V, Pilo Boyl P, Munck S, Bagni C, Santama N. KIF1Bβ transports dendritically localized mRNPs in neurons and is recruited to synapses in an activity-dependent manner. Cell Mol Life Sci 2013; 70:335-56. [PMID: 22945799 PMCID: PMC11113723 DOI: 10.1007/s00018-012-1108-0] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2011] [Revised: 07/17/2012] [Accepted: 07/24/2012] [Indexed: 12/15/2022]
Abstract
KIF1Bβ is a kinesin-like, microtubule-based molecular motor protein involved in anterograde axonal vesicular transport in vertebrate and invertebrate neurons. Certain KIF1Bβ isoforms have been implicated in different forms of human neurodegenerative disease, with characterization of their functional integration and regulation in the context of synaptic signaling still ongoing. Here, we characterize human KIF1Bβ (isoform NM015074), whose expression we show to be developmentally regulated and elevated in cortical areas of the CNS (including the motor cortex), in the hippocampus, and in spinal motor neurons. KIF1Bβ localizes to the cell body, axon, and dendrites, overlapping with synaptic-vesicle and postsynaptic-density structures. Correspondingly, in purified cortical synaptoneurosomes, KIF1Bβ is enriched in both pre- and postsynaptic structures, forming detergent-resistant complexes. Interestingly, KIF1Bβ forms RNA-protein complexes, containing the dendritically localized Arc and Calmodulin mRNAs, proteins previously shown to be part of RNA transport granules such as Purα, FMRP and FXR2P, and motor protein KIF3A, as well as Calmodulin. The interaction between KIF1Bβ and Calmodulin is Ca(+2)-dependent and takes place through a domain mapped at the carboxy-terminal tail of the motor. Live imaging of cortical neurons reveals active movement by KIF1Bβ at dendritic processes, suggesting that it mediates the transport of dendritically localized mRNAs. Finally, we show that synaptic recruitment of KIF1Bβ is activity-dependent and increased by stimulation of metabotropic or ionotropic glutamate receptors. The activity-dependent synaptic recruitment of KIF1Bβ, its interaction with Ca(2+) sensor Calmodulin, and its new role as a dendritic motor of ribonucleoprotein complexes provide a novel basis for understanding the concerted co-ordination of motor protein mobilization and synaptic signaling pathways.
Collapse
Affiliation(s)
- Despina C. Charalambous
- Department of Biological Sciences, University of Cyprus, University Avenue 1, 1678 Nicosia, Cyprus
| | - Emanuela Pasciuto
- VIB Center for Biology of Disease, Katholieke Universiteit Leuven, Heresraat 49, 3000 Leuven, Belgium
| | - Valentina Mercaldo
- VIB Center for Biology of Disease, Katholieke Universiteit Leuven, Heresraat 49, 3000 Leuven, Belgium
- Present Address: Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Canada
| | - Pietro Pilo Boyl
- VIB Center for Biology of Disease, Katholieke Universiteit Leuven, Heresraat 49, 3000 Leuven, Belgium
- Present Address: Institute of Genetics, University of Bonn, Karlrobert-Kreiten Str. 13, 53115 Bonn, Germany
| | - Sebastian Munck
- VIB Center for Biology of Disease, Katholieke Universiteit Leuven, Heresraat 49, 3000 Leuven, Belgium
| | - Claudia Bagni
- VIB Center for Biology of Disease, Katholieke Universiteit Leuven, Heresraat 49, 3000 Leuven, Belgium
- Department of Biomedicine and Prevention, Faculty of Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00100 Rome, Italy
| | - Niovi Santama
- Department of Biological Sciences, University of Cyprus, University Avenue 1, 1678 Nicosia, Cyprus
| |
Collapse
|