1
|
Protic D, Breeze E, Mendoza G, Zafarullah M, Abbeduto L, Hagerman R, Coffey C, Cudkowicz M, Durbin-Johnson B, Ashwood P, Berry-Kravis E, Erickson CA, Filipink R, Gropman A, Lehwald L, Maxwell-Horn A, Morris S, Bennett AP, Prock L, Talboy A, Tartaglia N, Veenstra-VanderWeele J, Tassone F. Negative effect of treatment with mGluR5 negative allosteric modulator AFQ056 on blood biomarkers in young individuals with Fragile X syndrome. SAGE Open Med 2024; 12:20503121241282401. [PMID: 39483619 PMCID: PMC11526204 DOI: 10.1177/20503121241282401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 08/22/2024] [Indexed: 11/03/2024] Open
Abstract
Background Fragile X syndrome, with an approximate incidence rate of 1 in 4000 males to 1 in 8000 females, is the most prevalent genetic cause of heritable intellectual disability and the most common monogenic cause of autism spectrum disorder. The full mutation of the Fragile X Messenger Ribonucleoprotein-1 gene, characterized by an expansion of CGG trinucleotide repeats (>200 CGG repeats), leads to fragile X syndrome. Currently, there are no targeted treatments available for fragile X syndrome. In a recent large multi-site trial, FXLEARN, the effects of the mGluR5 negative allosteric modulator, AFQ056 (mavoglurant), were investigated, but did not show a significant impact of AFQ056 on language development in children with fragile X syndrome aged 3-6 years. Objectives The current analyses from biospecimens collected in the FXLEARN study aimed to determine whether AFQ056 affects the level of potential biomarkers associated with Akt/mTOR and matrix metalloproteinase 9 signaling in young individuals with fragile X syndrome. Previous research has indicated that these biomarkers play crucial roles in the pathophysiology of fragile X syndrome. Design A double-blind placebo-controlled parallel-group flexible-dose forced titration design. Methods Blood samples for biomarkers were collected during the FXLEARN at baseline and subsequent visits (1- and 8-month visits). Biomarker analyses included fragile X messenger ribonucleoprotein-1 genotyping by Southern blot and PCR approaches, fragile X messenger ribonucleoprotein-1 mRNA levels determined by PCR, matrix metalloproteinase 9 levels' detection using a magnetic bead panel, and targets of the Akt/mTOR signaling pathway with their phosphorylation levels detected. Results This research revealed that administering AFQ056 does not affect the expression levels of the investigated blood biomarkers in young children with fragile X syndrome. Conclusion Our findings of the lack of association between clinical improvement and biomarkers' levels in the treatment group are in line with the lack of benefit observed in the FXLEARN study. These findings indicate that AFQ056 does not provide benefits as assessed by primary or secondary endpoints. Registration ClincalTrials.gov NCT02920892.
Collapse
Affiliation(s)
- Dragana Protic
- Faculty of Medicine, Department of Pharmacology, Clinical Pharmacology, and Toxicology, University of Belgrade, Belgrade, Serbia
- Fragile X Clinic, Special Hospital for Cerebral Palsy and Developmental Neurology, Belgrade, Serbia
| | - Elizabeth Breeze
- Department of Medical Microbiology and Immunology, School of Medicine, University of California Davis, Davis, CA, USA
- MIND Institute, University of California Davis, Sacramento, CA, USA
| | - Guadalupe Mendoza
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California Davis, Sacramento, CA, USA
| | - Marwa Zafarullah
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California Davis, Sacramento, CA, USA
| | - Leonard Abbeduto
- MIND Institute, University of California Davis, Sacramento, CA, USA
- Department of Psychiatry and Behavioral Sciences, University of California Davis, Sacramento, CA, USA
| | - Randi Hagerman
- MIND Institute, University of California Davis, Sacramento, CA, USA
- Department of Pediatrics, University of California Davis, Sacramento, CA, USA
| | | | - Merit Cudkowicz
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
| | - Blythe Durbin-Johnson
- Division of Biostatistics, Department of Public Health Sciences, University of California, Davis, CA, USA
| | - Paul Ashwood
- Department of Medical Microbiology and Immunology, School of Medicine, University of California Davis, Davis, CA, USA
| | - Elizabeth Berry-Kravis
- Department of Pediatrics, Neurological Sciences, Anatomy, and Cell Biology, Rush University Medical Center, Chicago, IL, USA
| | | | | | | | | | | | - Stephanie Morris
- Washington University Medical Center, Saint Louis Children’s Hospital, St. Louis, MO, USA
| | | | - Lisa Prock
- Boston Children’s Hospital, Boston, MA, USA
| | - Amy Talboy
- Emory University Medical Center, Atlanta, GA, USA
| | | | - Jeremy Veenstra-VanderWeele
- Center for Autism and the Developing Brain, New York-Presbyterian, New York, NY, USA
- Department of Psychiatry, Columbia University, New York, NY, USA
- New York State Psychiatric Institute, New York, NY, USA
| | - Flora Tassone
- MIND Institute, University of California Davis, Sacramento, CA, USA
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California Davis, Sacramento, CA, USA
| |
Collapse
|
2
|
Milojevic S, Ghosh A, Makevic V, Stojkovic M, Capovilla M, Tosti T, Budimirovic D, Protic D. Circadian Rhythm and Sleep Analyses in a Fruit Fly Model of Fragile X Syndrome Using a Video-Based Automated Behavioral Research System. Int J Mol Sci 2024; 25:7949. [PMID: 39063191 PMCID: PMC11277495 DOI: 10.3390/ijms25147949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 07/09/2024] [Accepted: 07/18/2024] [Indexed: 07/28/2024] Open
Abstract
Fragile X syndrome (FXS) is caused by the full mutation in the FMR1 gene on the Xq27.3 chromosome region. It is the most common monogenic cause of autism spectrum disorder (ASD) and inherited intellectual disability (ID). Besides ASD and ID and other symptoms, individuals with FXS may exhibit sleep problems and impairment of circadian rhythm (CR). The Drosophila melanogaster models of FXS, such as dFMR1B55, represent excellent models for research in the FXS field. During this study, sleep patterns and CR in dFMR1B55 mutants were analyzed, using a new platform based on continuous high-resolution videography integrated with a highly-customized version of an open-source software. This methodology provides more sensitive results, which could be crucial for all further research in this model of fruit flies. The study revealed that dFMR1B55 male mutants sleep more and can be considered weak rhythmic flies rather than totally arrhythmic and present a good alternative animal model of genetic disorder, which includes impairment of CR and sleep behavior. The combination of affordable videography and software used in the current study is a significant improvement over previous methods and will enable broader adaptation of such high-resolution behavior monitoring methods.
Collapse
Affiliation(s)
- Sara Milojevic
- Department of Pharmacology, Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia; (S.M.); (M.S.)
| | - Arijit Ghosh
- Chronobiology and Behavioral Neurogenetics Laboratory, Neuroscience Unit, Jawaharlal Nehru Center for Advanced Scientific Research, Jakkur, Bangalore 560064, India;
| | - Vedrana Makevic
- Department of Pathophysiology, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia;
| | - Maja Stojkovic
- Department of Pharmacology, Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia; (S.M.); (M.S.)
| | - Maria Capovilla
- UMR7275 CNRS-INSERM-UniCA, Institute of Cellular and Molecular Pharmacology Institute, Sophia Antipolis, 06560 Valbonne, France;
| | - Tomislav Tosti
- Institute of Chemistry, Technology and Metallurgy, National Institute of the Republic of Serbia, University of Belgrade, 11000 Belgrade, Serbia;
| | - Dejan Budimirovic
- Department of Psychiatry, Fragile X Clinic, Kennedy Krieger Institute, Baltimore, MD 21205, USA;
- Department of Psychiatry & Behavioral Sciences-Child Psychiatry, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Dragana Protic
- Department of Pharmacology, Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia; (S.M.); (M.S.)
- Fragile X Clinic, Special Hospital for Cerebral Palsy and Developmental Neurology, 11000 Belgrade, Serbia
| |
Collapse
|
3
|
Protic D, Hagerman R. State-of-the-art therapies for fragile X syndrome. Dev Med Child Neurol 2024; 66:863-871. [PMID: 38385885 PMCID: PMC11144093 DOI: 10.1111/dmcn.15885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 01/22/2024] [Accepted: 01/24/2024] [Indexed: 02/23/2024]
Abstract
Fragile X syndrome (FXS) is a neurodevelopmental disorder caused by a full mutation (> 200 CGG repeats) in the FMR1 gene. FXS is the leading cause of inherited intellectual disabilities and the most commonly known genetic cause of autism spectrum disorder. Children with FXS experience behavioral and sleep problems, anxiety, inattention, learning difficulties, and speech and language delays. There are no approved medications for FXS; however, there are several interventions and treatments aimed at managing the symptoms and improving the quality of life of individuals with FXS. A combination of non-pharmacological therapies and pharmacotherapy is currently the most effective treatment for FXS. Currently, several targeted treatments, such as metformin, sertraline, and cannabidiol, can be used by clinicians to treat FXS. Gene therapy is rapidly developing and holds potential as a prospective treatment option. Soon its efficacy and safety in patients with FXS will be demonstrated. WHAT THIS PAPER ADDS: Targeted treatment of fragile X syndrome (FXS) is the best current therapeutic approach. Gene therapy holds potential as a prospective treatment for FXS in the future.
Collapse
Affiliation(s)
- Dragana Protic
- Department of Pharmacology, Clinical Pharmacology and Toxicology, Faculty of Medicine University of Belgrade, Belgrade, Serbia
- Fragile X Clinic, Special Hospital for Cerebral Palsy and Developmental Neurology, Belgrade, Serbia
| | - Randi Hagerman
- Medical Investigation of Neurodevelopmental Disorders Institute, University of California, Davis, CA, USA
- Department of Pediatrics, University of California, Davis School of Medicine, Sacramento, CA, USA
| |
Collapse
|
4
|
D'Antoni S, Spatuzza M, Bonaccorso CM, Catania MV. Role of fragile X messenger ribonucleoprotein 1 in the pathophysiology of brain disorders: a glia perspective. Neurosci Biobehav Rev 2024; 162:105731. [PMID: 38763180 DOI: 10.1016/j.neubiorev.2024.105731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 05/14/2024] [Accepted: 05/15/2024] [Indexed: 05/21/2024]
Abstract
Fragile X messenger ribonucleoprotein 1 (FMRP) is a widely expressed RNA binding protein involved in several steps of mRNA metabolism. Mutations in the FMR1 gene encoding FMRP are responsible for fragile X syndrome (FXS), a leading genetic cause of intellectual disability and autism spectrum disorder, and fragile X-associated tremor-ataxia syndrome (FXTAS), a neurodegenerative disorder in aging men. Although FMRP is mainly expressed in neurons, it is also present in glial cells and its deficiency or altered expression can affect functions of glial cells with implications for the pathophysiology of brain disorders. The present review focuses on recent advances on the role of glial subtypes, astrocytes, oligodendrocytes and microglia, in the pathophysiology of FXS and FXTAS, and describes how the absence or reduced expression of FMRP in these cells can impact on glial and neuronal functions. We will also briefly address the role of FMRP in radial glial cells and its effects on neural development, and gliomas and will speculate on the role of glial FMRP in other brain disorders.
Collapse
Affiliation(s)
- S D'Antoni
- Institute for Biomedical Research and Innovation (IRIB), National Research Council (CNR), Via Paolo Gaifami 18, Catania 95126, Italy
| | - M Spatuzza
- Institute for Biomedical Research and Innovation (IRIB), National Research Council (CNR), Via Paolo Gaifami 18, Catania 95126, Italy
| | - C M Bonaccorso
- Oasi Research Institute - IRCCS, via Conte Ruggero 73, Troina 94018, Italy
| | - M V Catania
- Institute for Biomedical Research and Innovation (IRIB), National Research Council (CNR), Via Paolo Gaifami 18, Catania 95126, Italy.
| |
Collapse
|
5
|
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
|
6
|
Geng J, Li S, Li Y, Wu Z, Bhurtel S, Rimal S, Khan D, Ohja R, Brandman O, Lu B. Stalled translation by mitochondrial stress upregulates a CNOT4-ZNF598 ribosomal quality control pathway important for tissue homeostasis. Nat Commun 2024; 15:1637. [PMID: 38388640 PMCID: PMC10883933 DOI: 10.1038/s41467-024-45525-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 01/26/2024] [Indexed: 02/24/2024] Open
Abstract
Translational control exerts immediate effect on the composition, abundance, and integrity of the proteome. Ribosome-associated quality control (RQC) handles ribosomes stalled at the elongation and termination steps of translation, with ZNF598 in mammals and Hel2 in yeast serving as key sensors of translation stalling and coordinators of downstream resolution of collided ribosomes, termination of stalled translation, and removal of faulty translation products. The physiological regulation of RQC in general and ZNF598 in particular in multicellular settings is underexplored. Here we show that ZNF598 undergoes regulatory K63-linked ubiquitination in a CNOT4-dependent manner and is upregulated upon mitochondrial stresses in mammalian cells and Drosophila. ZNF598 promotes resolution of stalled ribosomes and protects against mitochondrial stress in a ubiquitination-dependent fashion. In Drosophila models of neurodegenerative diseases and patient cells, ZNF598 overexpression aborts stalled translation of mitochondrial outer membrane-associated mRNAs, removes faulty translation products causal of disease, and improves mitochondrial and tissue health. These results shed lights on the regulation of ZNF598 and its functional role in mitochondrial and tissue homeostasis.
Collapse
Affiliation(s)
- Ji Geng
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, 94305, USA.
| | - Shuangxi Li
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, 94305, USA
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology, School of Life Sciences, Shandong University, Qingdao, 266237, China
| | - Yu Li
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, 94305, USA
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology, School of Life Sciences, Shandong University, Qingdao, 266237, China
| | - Zhihao Wu
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Sunil Bhurtel
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Suman Rimal
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Danish Khan
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Rani Ohja
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Onn Brandman
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Bingwei Lu
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, 94305, USA.
| |
Collapse
|
7
|
Momoi MY. Overview: Research on the Genetic Architecture of the Developing Cerebral Cortex in Norms and Diseases. Methods Mol Biol 2024; 2794:1-12. [PMID: 38630215 DOI: 10.1007/978-1-0716-3810-1_1] [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] [Indexed: 04/19/2024]
Abstract
The human brain is characterized by high cell numbers, diverse cell types with diverse functions, and intricate connectivity with an exceedingly broad surface of the cortex. Human-specific brain development was accomplished by a long timeline for maturation from the prenatal period to the third decade of life. The long timeline makes complicated architecture and circuits of human cerebral cortex possible, and it makes human brain vulnerable to intrinsic and extrinsic insults resulting in the development of variety of neuropsychiatric disorders. Unraveling the molecular and cellular processes underlying human brain development under the elaborate regulation of gene expression in a spatiotemporally specific manner, especially that of the cortex will provide a biological understanding of human cognition and behavior in health and diseases. Global research consortia and the advancing technologies in brain science including functional genomics equipped with emergent neuroinformatics such as single-cell multiomics, novel human models, and high-volume databases with high-throughput computation facilitate the biological understanding of the development of the human brain cortex. Knowing the process of interplay of the genome and the environment in cortex development will lead us to understand the human-specific cognitive function and its individual diversity. Thus, it is worthwhile to overview the recent progress in neurotechnology to foresee further understanding of the human brain and norms and diseases.
Collapse
Affiliation(s)
- Mariko Y Momoi
- Ryomo Seishi Ryogoen Rehabilitation Hospital for Children with Disabilities, Gunma, Japan
| |
Collapse
|
8
|
Westmark CJ. Toward an understanding of the role of the exposome on fragile X phenotypes. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2023; 173:141-170. [PMID: 37993176 DOI: 10.1016/bs.irn.2023.08.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/24/2023]
Abstract
Fragile X syndrome (FXS) is the leading known monogenetic cause of autism with an estimated 21-50% of FXS individuals meeting autism diagnostic criteria. A critical gap in medical care for persons with autism is an understanding of how environmental exposures and gene-environment interactions affect disease outcomes. Our research indicates more severe neurological and metabolic outcomes (seizures, autism, increased body weight) in mouse and human models of autism spectrum disorders (ASD) as a function of diet. Thus, early-life exposure to chemicals in the diet could cause or exacerbate disease outcomes. Herein, we review the effects of potential dietary toxins, i.e., soy phytoestrogens, glyphosate, and polychlorinated biphenyls (PCB) in FXS and other autism models. The rationale is that potentially toxic chemicals in the diet, particularly infant formula, could contribute to the development and/or severity of ASD and that further study in this area has potential to improve ASD outcomes through dietary modification.
Collapse
Affiliation(s)
- Cara J Westmark
- Department of Neurology, University of Wisconsin-Madison, Medical Sciences Center, Room 3619, 1300 University Avenue, Madison, WI, United States; Molecular Environmental Toxicology Center, University of Wisconsin-Madison, Medical Sciences Center, Room 3619, 1300 University Avenue, Madison, WI, United States.
| |
Collapse
|
9
|
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
|
10
|
Milla LA, Corral L, Rivera J, Zuñiga N, Pino G, Nunez-Parra A, Cea-Del Rio CA. Neurodevelopment and early pharmacological interventions in Fragile X Syndrome. Front Neurosci 2023; 17:1213410. [PMID: 37599992 PMCID: PMC10433175 DOI: 10.3389/fnins.2023.1213410] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 07/17/2023] [Indexed: 08/22/2023] Open
Abstract
Fragile X Syndrome (FXS) is a neurodevelopmental disorder and the leading monogenic cause of autism and intellectual disability. For years, several efforts have been made to develop an effective therapeutic approach to phenotypically rescue patients from the disorder, with some even advancing to late phases of clinical trials. Unfortunately, none of these attempts have completely succeeded, bringing urgency to further expand and refocus research on FXS therapeutics. FXS arises at early stages of postnatal development due to the mutation and transcriptional silencing of the Fragile X Messenger Ribonucleoprotein 1 gene (FMR1) and consequent loss of the Fragile X Messenger Ribonucleoprotein (FMRP) expression. Importantly, FMRP expression is critical for the normal adult nervous system function, particularly during specific windows of embryogenic and early postnatal development. Cellular proliferation, migration, morphology, axonal guidance, synapse formation, and in general, neuronal network establishment and maturation are abnormally regulated in FXS, underlying the cognitive and behavioral phenotypes of the disorder. In this review, we highlight the relevance of therapeutically intervening during critical time points of development, such as early postnatal periods in infants and young children and discuss past and current clinical trials in FXS and their potential to specifically target those periods. We also discuss potential benefits, limitations, and disadvantages of these pharmacological tools based on preclinical and clinical research.
Collapse
Affiliation(s)
- Luis A. Milla
- Centro de Investigacion Biomedica y Aplicada (CIBAP), Escuela de Medicina, Facultad de Ciencias Medicas, Universidad de Santiago de Chile, Santiago, Chile
| | - Lucia Corral
- Laboratorio de Neurofisiopatologia, Centro de Investigacion Biomedica y Aplicada (CIBAP), Escuela de Medicina, Facultad de Ciencias Medicas, Universidad de Santiago de Chile, Santiago, Chile
| | - Jhanpool Rivera
- Laboratorio de Neurofisiopatologia, Centro de Investigacion Biomedica y Aplicada (CIBAP), Escuela de Medicina, Facultad de Ciencias Medicas, Universidad de Santiago de Chile, Santiago, Chile
| | - Nolberto Zuñiga
- Laboratorio de Neurofisiopatologia, Centro de Investigacion Biomedica y Aplicada (CIBAP), Escuela de Medicina, Facultad de Ciencias Medicas, Universidad de Santiago de Chile, Santiago, Chile
| | - Gabriela Pino
- Laboratorio de Neurofisiopatologia, Centro de Investigacion Biomedica y Aplicada (CIBAP), Escuela de Medicina, Facultad de Ciencias Medicas, Universidad de Santiago de Chile, Santiago, Chile
| | - Alexia Nunez-Parra
- Physiology Laboratory, Department of Biology, Faculty of Science, Universidad de Chile, Santiago, Chile
- Cell Physiology Center, Universidad de Chile, Santiago, Chile
| | - Christian A. Cea-Del Rio
- Laboratorio de Neurofisiopatologia, Centro de Investigacion Biomedica y Aplicada (CIBAP), Escuela de Medicina, Facultad de Ciencias Medicas, Universidad de Santiago de Chile, Santiago, Chile
| |
Collapse
|
11
|
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
|
12
|
Wang X, Sela-Donenfeld D, Wang Y. Axonal and presynaptic FMRP: Localization, signal, and functional implications. Hear Res 2023; 430:108720. [PMID: 36809742 PMCID: PMC9998378 DOI: 10.1016/j.heares.2023.108720] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Revised: 01/22/2023] [Accepted: 02/09/2023] [Indexed: 02/12/2023]
Abstract
Fragile X mental retardation protein (FMRP) binds a selected set of mRNAs and proteins to guide neural circuit assembly and regulate synaptic plasticity. Loss of FMRP is responsible for Fragile X syndrome, a neuropsychiatric disorder characterized with auditory processing problems and social difficulty. FMRP actions in synaptic formation, maturation, and plasticity are site-specific among the four compartments of a synapse: presynaptic and postsynaptic neurons, astrocytes, and extracellular matrix. This review summarizes advancements in understanding FMRP localization, signals, and functional roles in axons and presynaptic terminals.
Collapse
Affiliation(s)
- Xiaoyu Wang
- Division of Histology & Embryology, Key Laboratory for Regenerative Medicine of the Ministry of Education, Medical College, Jinan University, Guangzhou 510632, China
| | - Dalit Sela-Donenfeld
- Koret School of Veterinary Medicine, Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot 76100, Israel
| | - Yuan Wang
- Department of Biomedical Sciences, Program in Neuroscience, Florida State University College of Medicine, Tallahassee, FL 32306, USA.
| |
Collapse
|
13
|
Svalina MN, Sullivan R, Restrepo D, Huntsman MM. From circuits to behavior: Amygdala dysfunction in fragile X syndrome. Front Integr Neurosci 2023; 17:1128529. [PMID: 36969493 PMCID: PMC10034113 DOI: 10.3389/fnint.2023.1128529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 02/23/2023] [Indexed: 03/12/2023] Open
Abstract
Fragile X syndrome (FXS) is a neurodevelopmental disorder caused by a repeat expansion mutation in the promotor region of the FMR1 gene resulting in transcriptional silencing and loss of function of fragile X messenger ribonucleoprotein 1 protein (FMRP). FMRP has a well-defined role in the early development of the brain. Thus, loss of the FMRP has well-known consequences for normal cellular and synaptic development leading to a variety of neuropsychiatric disorders including an increased prevalence of amygdala-based disorders. Despite our detailed understanding of the pathophysiology of FXS, the precise cellular and circuit-level underpinnings of amygdala-based disorders is incompletely understood. In this review, we discuss the development of the amygdala, the role of neuromodulation in the critical period plasticity, and recent advances in our understanding of how synaptic and circuit-level changes in the basolateral amygdala contribute to the behavioral manifestations seen in FXS.
Collapse
Affiliation(s)
- Matthew N. Svalina
- Medical Scientist Training Program, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- Neuroscience Graduate Program, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- Department of Cell and Developmental Biology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Regina Sullivan
- Brain Institute, Nathan Kline Institute, Orangeburg, NY, United States
- Child and Adolescent Psychiatry, Child Study Center, New York University School of Medicine, New York, NY, United States
| | - Diego Restrepo
- Neuroscience Graduate Program, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- Department of Cell and Developmental Biology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Molly M. Huntsman
- Neuroscience Graduate Program, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- *Correspondence: Molly M. Huntsman,
| |
Collapse
|
14
|
Cozachenco D, Ribeiro FC, Ferreira ST. Defective proteostasis in Alzheimer's disease. Ageing Res Rev 2023; 85:101862. [PMID: 36693451 DOI: 10.1016/j.arr.2023.101862] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 01/11/2023] [Accepted: 01/20/2023] [Indexed: 01/22/2023]
Abstract
The homeostasis of cellular proteins, or proteostasis, is critical for neuronal function and for brain processes, including learning and memory. Increasing evidence indicates that defective proteostasis contributes to the progression of neurodegenerative disorders, including Alzheimer's disease (AD), the most prevalent form of dementia in the elderly. Proteostasis comprises a set of cellular mechanisms that control protein synthesis, folding, post-translational modification and degradation, all of which are deregulated in AD. Importantly, deregulation of proteostasis plays a key role in synapse dysfunction and in memory impairment, the major clinical manifestation of AD. Here, we discuss molecular pathways involved in protein synthesis and degradation that are altered in AD, and possible pharmacological approaches to correct these defects.
Collapse
Affiliation(s)
- Danielle Cozachenco
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil.
| | - Felipe C Ribeiro
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil.
| | - Sergio T Ferreira
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil; Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil.
| |
Collapse
|
15
|
Abstract
The fragile X-related disorders are an important group of hereditary disorders that are caused by expanded CGG repeats in the 5' untranslated region of the FMR1 gene or by mutations in the coding sequence of this gene. Two categories of pathological CGG repeats are associated with these disorders, full mutation alleles and shorter premutation alleles. Individuals with full mutation alleles develop fragile X syndrome, which causes autism and intellectual disability, whereas those with premutation alleles, which have shorter CGG expansions, can develop fragile X-associated tremor/ataxia syndrome, a progressive neurodegenerative disease. Thus, fragile X-related disorders can manifest as neurodegenerative or neurodevelopmental disorders, depending on the size of the repeat expansion. Here, we review mouse models of fragile X-related disorders and discuss how they have informed our understanding of neurodegenerative and neurodevelopmental disorders. We also assess the translational value of these models for developing rational targeted therapies for intellectual disability and autism disorders.
Collapse
Affiliation(s)
- Rob Willemsen
- Department of Clinical Genetics, Erasmus University Medical Center, 3015 CN Rotterdam, the Netherlands. Department of Medical Genetics, University of Antwerp, 2000 Antwerp, Belgium
| | - R. Frank Kooy
- Department of Clinical Genetics, Erasmus University Medical Center, 3015 CN Rotterdam, the Netherlands. Department of Medical Genetics, University of Antwerp, 2000 Antwerp, Belgium
| |
Collapse
|
16
|
Scott DN, Frank MJ. Adaptive control of synaptic plasticity integrates micro- and macroscopic network function. Neuropsychopharmacology 2023; 48:121-144. [PMID: 36038780 PMCID: PMC9700774 DOI: 10.1038/s41386-022-01374-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 06/23/2022] [Accepted: 06/24/2022] [Indexed: 11/09/2022]
Abstract
Synaptic plasticity configures interactions between neurons and is therefore likely to be a primary driver of behavioral learning and development. How this microscopic-macroscopic interaction occurs is poorly understood, as researchers frequently examine models within particular ranges of abstraction and scale. Computational neuroscience and machine learning models offer theoretically powerful analyses of plasticity in neural networks, but results are often siloed and only coarsely linked to biology. In this review, we examine connections between these areas, asking how network computations change as a function of diverse features of plasticity and vice versa. We review how plasticity can be controlled at synapses by calcium dynamics and neuromodulatory signals, the manifestation of these changes in networks, and their impacts in specialized circuits. We conclude that metaplasticity-defined broadly as the adaptive control of plasticity-forges connections across scales by governing what groups of synapses can and can't learn about, when, and to what ends. The metaplasticity we discuss acts by co-opting Hebbian mechanisms, shifting network properties, and routing activity within and across brain systems. Asking how these operations can go awry should also be useful for understanding pathology, which we address in the context of autism, schizophrenia and Parkinson's disease.
Collapse
Affiliation(s)
- Daniel N Scott
- Cognitive Linguistic, and Psychological Sciences, Brown University, Providence, RI, USA.
- Carney Institute for Brain Science, Brown University, Providence, RI, USA.
| | - Michael J Frank
- Cognitive Linguistic, and Psychological Sciences, Brown University, Providence, RI, USA.
- Carney Institute for Brain Science, Brown University, Providence, RI, USA.
| |
Collapse
|
17
|
Trajković J, Makevic V, Pesic M, Pavković-Lučić S, Milojevic S, Cvjetkovic S, Hagerman R, Budimirovic DB, Protic D. Drosophila melanogaster as a Model to Study Fragile X-Associated Disorders. Genes (Basel) 2022; 14:genes14010087. [PMID: 36672829 PMCID: PMC9859539 DOI: 10.3390/genes14010087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 12/22/2022] [Accepted: 12/23/2022] [Indexed: 12/30/2022] Open
Abstract
Fragile X syndrome (FXS) is a global neurodevelopmental disorder caused by the expansion of CGG trinucleotide repeats (≥200) in the Fragile X Messenger Ribonucleoprotein 1 (FMR1) gene. FXS is the hallmark of Fragile X-associated disorders (FXD) and the most common monogenic cause of inherited intellectual disability and autism spectrum disorder. There are several animal models used to study FXS. In the FXS model of Drosophila, the only ortholog of FMR1, dfmr1, is mutated so that its protein is missing. This model has several relevant phenotypes, including defects in the circadian output pathway, sleep problems, memory deficits in the conditioned courtship and olfactory conditioning paradigms, deficits in social interaction, and deficits in neuronal development. In addition to FXS, a model of another FXD, Fragile X-associated tremor/ataxia syndrome (FXTAS), has also been established in Drosophila. This review summarizes many years of research on FXD in Drosophila models.
Collapse
Affiliation(s)
- Jelena Trajković
- Faculty of Biology, University of Belgrade, 11000 Belgrade, Serbia
| | - Vedrana Makevic
- Department of Pathophysiology, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| | - Milica Pesic
- Institute of Human Genetics, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| | | | - Sara Milojevic
- Department of Pharmacology, Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| | - Smiljana Cvjetkovic
- Department of Humanities, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| | - Randi Hagerman
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute, University of California Davis, 2825 50th Street, Sacramento, CA 95817, USA
- Department of Pediatrics, University of California Davis School of Medicine, Sacramento, CA 95817, USA
| | - Dejan B. Budimirovic
- Department of Psychiatry, Fragile X Clinic, Kennedy Krieger Institute, Baltimore, MD 21205, USA
- Department of Psychiatry & Behavioral Sciences-Child Psychiatry, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Dragana Protic
- Department of Pharmacology, Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
- Correspondence:
| |
Collapse
|
18
|
Yu X, Wang Y. Tonotopic differentiation of presynaptic neurotransmitter-releasing machinery in the auditory brainstem during the prehearing period and its selective deficits in Fmr1 knockout mice. J Comp Neurol 2022; 530:3248-3269. [PMID: 36067267 PMCID: PMC9588645 DOI: 10.1002/cne.25406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 07/29/2022] [Accepted: 08/24/2022] [Indexed: 11/07/2022]
Abstract
Tonotopic organization is a fundamental feature of the auditory system. In the developing auditory brainstem, the ontogeny and maturation of neurotransmission progress from high to low frequencies along the tonotopic axis. To explore the underlying mechanism of this tonotopic development, we aim to determine whether the presynaptic machinery responsible for neurotransmitter release is tonotopically differentiated during development. In the current study, we examined vesicular neurotransmitter transporters and calcium sensors, two central players responsible for loading neurotransmitter into synaptic vesicles and for triggering neurotransmitter release in a calcium-dependent manner, respectively. Using immunocytochemistry, we characterized the distribution patterns of vesicular glutamate transporters (VGLUTs) 1 and 2, vesicular gamma-aminobutyric acid transporter (VGAT), and calcium sensor synaptotagmin (Syt) 1 and 2 in the developing mouse medial nucleus of the trapezoid body (MNTB). We identified tonotopic gradients of VGLUT1, VGAT, Syt1, and Syt2 in the first postnatal week, with higher protein densities in the more medial (high-frequency) portion of the MNTB. These gradients gradually flattened before the onset of hearing. In contrast, VGLUT2 was distributed relatively uniformly along the tonotopic axis during this prehearing period. In mice lacking Fragile X mental retardation protein, an mRNA-binding protein that regulates synaptic development and plasticity, progress to achieve the mature-like organization was altered for VGLUT1, Syt1, and Syt2, but not for VGAT. Together, our results identified novel organization patterns of selective presynaptic proteins in immature auditory synapses, providing a potential mechanism that may contribute to tonotopic differentiation of neurotransmission during normal and abnormal development.
Collapse
Affiliation(s)
- Xiaoyan Yu
- Program in Neuroscience, Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, Florida, USA
| | - Yuan Wang
- Program in Neuroscience, Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, Florida, USA
| |
Collapse
|
19
|
Morrill NK, Joly-Amado A, Li Q, Prabhudeva S, Weeber EJ, Nash KR. Reelin central fragment supplementation improves cognitive deficits in a mouse model of Fragile X Syndrome. Exp Neurol 2022; 357:114170. [PMID: 35863501 DOI: 10.1016/j.expneurol.2022.114170] [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: 05/24/2022] [Revised: 07/10/2022] [Accepted: 07/14/2022] [Indexed: 11/04/2022]
Abstract
Fragile X Syndrome (FXS) is the most common form of inherited intellectual disability and is characterized by autistic behaviors, childhood seizures, and deficits in learning and memory. FXS has a loss of function of the FMR1 gene that leads to a lack of Fragile X Mental Retardation Protein (FMRP) expression. FMRP is critical for synaptic plasticity, spatial learning, and memory. Reelin is a large extracellular glycoprotein essential for synaptic plasticity and numerous neurodevelopmental processes. Reduction in Reelin signaling is implicated as a contributing factor in disease etiology in several neurological disorders, including schizophrenia, and autism. However, the role of Reelin in FXS is poorly understood. We demonstrate a reduction in Reelin in Fmr1 knock-out (KO) mice, suggesting that a loss of Reelin activity may contribute to FXS. We demonstrate here that Reelin signaling enhancement via a single intracerebroventricular injection of the Reelin central fragment into Fmr1 KO mice can profoundly rescue cognitive deficits in hidden platform water maze and fear conditioning, as well as hyperactivity during the open field. Improvements in behavior were associated with rescued levels of post synaptic marker in Fmr1 KO mice when compared to controls. These data suggest that increasing Reelin signaling in FXS could offer a novel therapeutic for improving cognition in FXS.
Collapse
Affiliation(s)
- Nicole K Morrill
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, 12901 Bruce B Downs Blvd, Tampa FL-33612, USA
| | - Aurelie Joly-Amado
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, 12901 Bruce B Downs Blvd, Tampa FL-33612, USA
| | - Qingyou Li
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, 12901 Bruce B Downs Blvd, Tampa FL-33612, USA
| | - Sahana Prabhudeva
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, 12901 Bruce B Downs Blvd, Tampa FL-33612, USA
| | - Edwin J Weeber
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, 12901 Bruce B Downs Blvd, Tampa FL-33612, USA
| | - Kevin R Nash
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, 12901 Bruce B Downs Blvd, Tampa FL-33612, USA.
| |
Collapse
|
20
|
Kalinowska M, van der Lei MB, Kitiashvili M, Mamcarz M, Oliveira MM, Longo F, Klann E. Deletion of Fmr1 in parvalbumin-expressing neurons results in dysregulated translation and selective behavioral deficits associated with fragile X syndrome. Mol Autism 2022; 13:29. [PMID: 35768828 PMCID: PMC9245312 DOI: 10.1186/s13229-022-00509-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 06/10/2022] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Fragile X syndrome (FXS), the most common genetic cause of autism spectrum disorder and intellectual disability, is caused by the lack of fragile X mental retardation protein (FMRP) expression. FMRP is an mRNA binding protein with functions in mRNA transport, localization, and translational control. In Fmr1 knockout mice, dysregulated translation has been linked to pathophysiology, including abnormal synaptic function and dendritic morphology, and autistic-like behavioral phenotypes. The role of FMRP in morphology and function of excitatory neurons has been well studied in mice lacking Fmr1, but the impact of Fmr1 deletion on inhibitory neurons remains less characterized. Moreover, the contribution of FMRP in different cell types to FXS pathophysiology is not well defined. We sought to characterize whether FMRP loss in parvalbumin or somatostatin-expressing neurons results in FXS-like deficits in mice. METHODS We used Cre-lox recombinase technology to generate two lines of conditional knockout mice lacking FMRP in either parvalbumin or somatostatin-expressing cells and carried out a battery of behavioral tests to assess motor function, anxiety, repetitive, stereotypic, social behaviors, and learning and memory. In addition, we used fluorescent non-canonical amino acid tagging along with immunostaining to determine whether de novo protein synthesis is dysregulated in parvalbumin or somatostatin-expressing neurons. RESULTS De novo protein synthesis was elevated in hippocampal parvalbumin and somatostatin-expressing inhibitory neurons in Fmr1 knockout mice. Cell type-specific deletion of Fmr1 in parvalbumin-expressing neurons resulted in anxiety-like behavior, impaired social behavior, and dysregulated de novo protein synthesis. In contrast, deletion of Fmr1 in somatostatin-expressing neurons did not result in behavioral abnormalities and did not significantly impact de novo protein synthesis. This is the first report of how loss of FMRP in two specific subtypes of inhibitory neurons is associated with distinct FXS-like abnormalities. LIMITATIONS The mouse models we generated are limited by whole body knockout of FMRP in parvalbumin or somatostatin-expressing cells and further studies are needed to establish a causal relationship between cellular deficits and FXS-like behaviors. CONCLUSIONS Our findings indicate a cell type-specific role for FMRP in parvalbumin-expressing neurons in regulating distinct behavioral features associated with FXS.
Collapse
Affiliation(s)
- Magdalena Kalinowska
- grid.137628.90000 0004 1936 8753Center for Neural Science, New York University, New York, NY USA
| | - Mathijs B. van der Lei
- grid.5284.b0000 0001 0790 3681Department of Medical Genetics, University of Antwerp, Antwerp, Belgium
| | - Michael Kitiashvili
- grid.137628.90000 0004 1936 8753Center for Neural Science, New York University, New York, NY USA
| | - Maggie Mamcarz
- grid.137628.90000 0004 1936 8753Center for Neural Science, New York University, New York, NY USA
| | - Mauricio M. Oliveira
- grid.137628.90000 0004 1936 8753Center for Neural Science, New York University, New York, NY USA
| | - Francesco Longo
- grid.8761.80000 0000 9919 9582Institute for Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden ,grid.8761.80000 0000 9919 9582Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Eric Klann
- Center for Neural Science, New York University, New York, NY, USA. .,NYU Neuroscience Institute, New York University Langone Medical Center, New York, NY, USA.
| |
Collapse
|
21
|
Protic DD, Aishworiya R, Salcedo-Arellano MJ, Tang SJ, Milisavljevic J, Mitrovic F, Hagerman RJ, Budimirovic DB. Fragile X Syndrome: From Molecular Aspect to Clinical Treatment. Int J Mol Sci 2022; 23:1935. [PMID: 35216055 PMCID: PMC8875233 DOI: 10.3390/ijms23041935] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 02/01/2022] [Accepted: 02/02/2022] [Indexed: 02/01/2023] Open
Abstract
Fragile X syndrome (FXS) is a neurodevelopmental disorder caused by the full mutation as well as highly localized methylation of the fragile X mental retardation 1 (FMR1) gene on the long arm of the X chromosome. Children with FXS are commonly co-diagnosed with Autism Spectrum Disorder, attention and learning problems, anxiety, aggressive behavior and sleep disorder, and early interventions have improved many behavior symptoms associated with FXS. In this review, we performed a literature search of original and review articles data of clinical trials and book chapters using MEDLINE (1990-2021) and ClinicalTrials.gov. While we have reviewed the biological importance of the fragile X mental retardation protein (FMRP), the FXS phenotype, and current diagnosis techniques, the emphasis of this review is on clinical interventions. Early non-pharmacological interventions in combination with pharmacotherapy and targeted treatments aiming to reverse dysregulated brain pathways are the mainstream of treatment in FXS. Overall, early diagnosis and interventions are fundamental to achieve optimal clinical outcomes in FXS.
Collapse
Affiliation(s)
- Dragana D. Protic
- Department of Pharmacology, Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Belgrade, 11129 Belgrade, Serbia
| | - Ramkumar Aishworiya
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute UCDH, University of California Davis, 2825 50th Street, Sacramento, CA 95817, USA; (R.A.); (M.J.S.-A.); (S.J.T.); (R.J.H.)
- Khoo Teck Puat-National University Children’s Medical Institute, National University Health System, 5 Lower Kent Ridge Road, Singapore 119074, Singapore
| | - Maria Jimena Salcedo-Arellano
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute UCDH, University of California Davis, 2825 50th Street, Sacramento, CA 95817, USA; (R.A.); (M.J.S.-A.); (S.J.T.); (R.J.H.)
- Department of Pediatrics, University of California Davis School of Medicine, Sacramento, CA 95817, USA
- Department of Pathology and Laboratory Medicine, University of California Davis School of Medicine, Sacramento, CA 95817, USA
| | - Si Jie Tang
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute UCDH, University of California Davis, 2825 50th Street, Sacramento, CA 95817, USA; (R.A.); (M.J.S.-A.); (S.J.T.); (R.J.H.)
| | - Jelena Milisavljevic
- Faculty of Medicine, University of Belgrade, 11129 Belgrade, Serbia; (J.M.); (F.M.)
| | - Filip Mitrovic
- Faculty of Medicine, University of Belgrade, 11129 Belgrade, Serbia; (J.M.); (F.M.)
| | - Randi J. Hagerman
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute UCDH, University of California Davis, 2825 50th Street, Sacramento, CA 95817, USA; (R.A.); (M.J.S.-A.); (S.J.T.); (R.J.H.)
- Department of Pediatrics, University of California Davis School of Medicine, Sacramento, CA 95817, USA
| | - Dejan B. Budimirovic
- Department of Psychiatry, Fragile X Clinic, Kennedy Krieger Institute, Baltimore, MD 21205, USA
- Department of Psychiatry & Behavioral Sciences-Child Psychiatry, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
22
|
Liu X, Kumar V, Tsai NP, Auerbach BD. Hyperexcitability and Homeostasis in Fragile X Syndrome. Front Mol Neurosci 2022; 14:805929. [PMID: 35069112 PMCID: PMC8770333 DOI: 10.3389/fnmol.2021.805929] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Accepted: 12/14/2021] [Indexed: 01/13/2023] Open
Abstract
Fragile X Syndrome (FXS) is a leading inherited cause of autism and intellectual disability, resulting from a mutation in the FMR1 gene and subsequent loss of its protein product FMRP. Despite this simple genetic origin, FXS is a phenotypically complex disorder with a range of physical and neurocognitive disruptions. While numerous molecular and cellular pathways are affected by FMRP loss, there is growing evidence that circuit hyperexcitability may be a common convergence point that can account for many of the wide-ranging phenotypes seen in FXS. The mechanisms for hyperexcitability in FXS include alterations to excitatory synaptic function and connectivity, reduced inhibitory neuron activity, as well as changes to ion channel expression and conductance. However, understanding the impact of FMR1 mutation on circuit function is complicated by the inherent plasticity in neural circuits, which display an array of homeostatic mechanisms to maintain activity near set levels. FMRP is also an important regulator of activity-dependent plasticity in the brain, meaning that dysregulated plasticity can be both a cause and consequence of hyperexcitable networks in FXS. This makes it difficult to separate the direct effects of FMR1 mutation from the myriad and pleiotropic compensatory changes associated with it, both of which are likely to contribute to FXS pathophysiology. Here we will: (1) review evidence for hyperexcitability and homeostatic plasticity phenotypes in FXS models, focusing on similarities/differences across brain regions, cell-types, and developmental time points; (2) examine how excitability and plasticity disruptions interact with each other to ultimately contribute to circuit dysfunction in FXS; and (3) discuss how these synaptic and circuit deficits contribute to disease-relevant behavioral phenotypes like epilepsy and sensory hypersensitivity. Through this discussion of where the current field stands, we aim to introduce perspectives moving forward in FXS research.
Collapse
Affiliation(s)
- Xiaopeng Liu
- Deparment of Molecular & Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL, United States
- Beckman Institute for Advanced Science & Technology, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Vipendra Kumar
- Deparment of Molecular & Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Nien-Pei Tsai
- Deparment of Molecular & Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Benjamin D. Auerbach
- Deparment of Molecular & Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL, United States
- Beckman Institute for Advanced Science & Technology, University of Illinois at Urbana-Champaign, Urbana, IL, United States
- *Correspondence: Benjamin D. Auerbach
| |
Collapse
|
23
|
FAZELI Z, GHADERIAN SMH, NAJMABADI H, OMRANI MD. Understanding the Molecular Basis of Fragile X Syndrome Using Differentiated Mesenchymal Stem Cells. IRANIAN JOURNAL OF CHILD NEUROLOGY 2022; 16:85-95. [PMID: 35222660 PMCID: PMC8753000 DOI: 10.22037/ijcn.v15i4.22070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/08/2018] [Accepted: 02/21/2021] [Indexed: 11/09/2022]
Abstract
OBJECTIVES Fragile X syndrome (FXS) has been known as the most common cause of inherited intellectual disability and autism. This disease results from the loss of fragile X mental retardation protein expression due to the expansion of CGG repeats located on the 5' untranslated region of the fragile X mental retardation 1 (FMR1) gene. MATERIALS & METHODS In the present study, the peripheral blood-mesenchymal stem cells (PB-MSCs) of two female full mutation carriers were differentiated into neuronal cells by the suppression of bone morphogenesis pathway signaling. Then, the expression of genes adjacent to CGG repeats expansion, including SLIT and NTRK-like protein 2 (SLITRK2), SLIT and NTRK-like protein 4 (SLITRK4), methyl CpG binding protein 2 (MECP2), and gamma-aminobutyric acid receptor subunit alpha-3 (GABRA3), were evaluated in these cells using SYBR Green real-time polymerase chain reaction. RESULTS The obtained results indicated that the expression of SLITRK2 and SLITRK4 were upregulated and downregulated in the neuron-like cells differentiated from the PB-MSCs of females with FMR1 full mutation, compared to that of the normal females, respectively. Furthermore, the expression of MECP2 and GABRA3 genes were observed to be related to the phenotypic differences observed in the female FMR1 full mutation carriers. CONCLUSION The observed association of expression of genes located upstream of the FMR1 gene with phenotypic differences in the female carriers could increase the understanding of novel therapeutic targets for patients with mild symptoms of FXS and the patients affected by other FMR1-related disorders.
Collapse
Affiliation(s)
- Zahra FAZELI
- Department of Medical Genetics, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Hossein NAJMABADI
- Genetics Research Center, University of Social Welfare and Rehabilitation Sciences, Tehran, Iran
| | - Mir Davood OMRANI
- Urogenital Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
24
|
Hale CR, Sawicka K, Mora K, Fak JJ, Kang JJ, Cutrim P, Cialowicz K, Carroll TS, Darnell RB. FMRP regulates mRNAs encoding distinct functions in the cell body and dendrites of CA1 pyramidal neurons. eLife 2021; 10:e71892. [PMID: 34939924 PMCID: PMC8820740 DOI: 10.7554/elife.71892] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 12/13/2021] [Indexed: 12/02/2022] Open
Abstract
Neurons rely on translation of synaptic mRNAs in order to generate activity-dependent changes in plasticity. Here, we develop a strategy combining compartment-specific crosslinking immunoprecipitation (CLIP) and translating ribosome affinity purification (TRAP) in conditionally tagged mice to precisely define the ribosome-bound dendritic transcriptome of CA1 pyramidal neurons. We identify CA1 dendritic transcripts with differentially localized mRNA isoforms generated by alternative polyadenylation and alternative splicing, including many that have altered protein-coding capacity. Among dendritic mRNAs, FMRP targets were found to be overrepresented. Cell-type-specific FMRP-CLIP and TRAP in microdissected CA1 neuropil revealed 383 dendritic FMRP targets and suggests that FMRP differentially regulates functionally distinct modules in CA1 dendrites and cell bodies. FMRP regulates ~15-20% of mRNAs encoding synaptic functions and 10% of chromatin modulators, in the dendrite and cell body, respectively. In the absence of FMRP, dendritic FMRP targets had increased ribosome association, consistent with a function for FMRP in synaptic translational repression. Conversely, downregulation of FMRP targets involved in chromatin regulation in cell bodies suggests a role for FMRP in stabilizing mRNAs containing stalled ribosomes in this compartment. Together, the data support a model in which FMRP regulates the translation and expression of synaptic and nuclear proteins within different compartments of a single neuronal cell type.
Collapse
Affiliation(s)
- Caryn R Hale
- Laboratory of Molecular Neuro-Oncology, Rockefeller UniversityNew YorkUnited States
| | - Kirsty Sawicka
- Laboratory of Molecular Neuro-Oncology, Rockefeller UniversityNew YorkUnited States
| | - Kevin Mora
- Laboratory of Molecular Neuro-Oncology, Rockefeller UniversityNew YorkUnited States
| | - John J Fak
- Laboratory of Molecular Neuro-Oncology, Rockefeller UniversityNew YorkUnited States
| | - Jin Joo Kang
- Laboratory of Molecular Neuro-Oncology, Rockefeller UniversityNew YorkUnited States
| | - Paula Cutrim
- Laboratory of Molecular Neuro-Oncology, Rockefeller UniversityNew YorkUnited States
| | - Katarzyna Cialowicz
- Bio-Imaging Resource Center, The Rockefeller UniversityNew YorkUnited States
| | - Thomas S Carroll
- Bioinformatics Resource Center, The Rockefeller UniversityNew YorkUnited States
| | - Robert B Darnell
- Laboratory of Molecular Neuro-Oncology, Rockefeller UniversityNew YorkUnited States
- Howard Hughes Medical Institute, Rockefeller UniversityNew YorkUnited States
| |
Collapse
|
25
|
Bach S, Shovlin S, Moriarty M, Bardoni B, Tropea D. Rett Syndrome and Fragile X Syndrome: Different Etiology With Common Molecular Dysfunctions. Front Cell Neurosci 2021; 15:764761. [PMID: 34867203 PMCID: PMC8640214 DOI: 10.3389/fncel.2021.764761] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 10/27/2021] [Indexed: 01/04/2023] Open
Abstract
Rett syndrome (RTT) and Fragile X syndrome (FXS) are two monogenetic neurodevelopmental disorders with complex clinical presentations. RTT is caused by mutations in the Methyl-CpG binding protein 2 gene (MECP2) altering the function of its protein product MeCP2. MeCP2 modulates gene expression by binding methylated CpG dinucleotides, and by interacting with transcription factors. FXS is caused by the silencing of the FMR1 gene encoding the Fragile X Mental Retardation Protein (FMRP), a RNA binding protein involved in multiple steps of RNA metabolism, and modulating the translation of thousands of proteins including a large set of synaptic proteins. Despite differences in genetic etiology, there are overlapping features in RTT and FXS, possibly due to interactions between MeCP2 and FMRP, and to the regulation of pathways resulting in dysregulation of common molecular signaling. Furthermore, basic physiological mechanisms are regulated by these proteins and might concur to the pathophysiology of both syndromes. Considering that RTT and FXS are disorders affecting brain development, and that most of the common targets of MeCP2 and FMRP are involved in brain activity, we discuss the mechanisms of synaptic function and plasticity altered in RTT and FXS, and we consider the similarities and the differences between these two disorders.
Collapse
Affiliation(s)
- Snow Bach
- School of Mathematical Sciences, Dublin City University, Dublin, Ireland.,Neuropsychiatric Genetics, Department of Psychiatry, School of Medicine, Trinity College Dublin, Trinity Translational Medicine Institute, St James's Hospital, Dublin, Ireland
| | - Stephen Shovlin
- Neuropsychiatric Genetics, Department of Psychiatry, School of Medicine, Trinity College Dublin, Trinity Translational Medicine Institute, St James's Hospital, Dublin, Ireland
| | | | - Barbara Bardoni
- Inserm, CNRS UMR 7275, Institute of Molecular and Cellular Pharmacology, Université Côte d'Azur, Valbonne, France
| | - Daniela Tropea
- Neuropsychiatric Genetics, Department of Psychiatry, School of Medicine, Trinity College Dublin, Trinity Translational Medicine Institute, St James's Hospital, Dublin, Ireland.,Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland.,FutureNeuro, The SFI Research Centre for Chronic and Rare Neurological Diseases, Dublin, Ireland
| |
Collapse
|
26
|
Faust TE, Gunner G, Schafer DP. Mechanisms governing activity-dependent synaptic pruning in the developing mammalian CNS. Nat Rev Neurosci 2021; 22:657-673. [PMID: 34545240 PMCID: PMC8541743 DOI: 10.1038/s41583-021-00507-y] [Citation(s) in RCA: 171] [Impact Index Per Article: 42.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/27/2021] [Indexed: 02/08/2023]
Abstract
Almost 60 years have passed since the initial discovery by Hubel and Wiesel that changes in neuronal activity can elicit developmental rewiring of the central nervous system (CNS). Over this period, we have gained a more comprehensive picture of how both spontaneous neural activity and sensory experience-induced changes in neuronal activity guide CNS circuit development. Here we review activity-dependent synaptic pruning in the mammalian CNS, which we define as the removal of a subset of synapses, while others are maintained, in response to changes in neural activity in the developing nervous system. We discuss the mounting evidence that immune and cell-death molecules are important mechanistic links by which changes in neural activity guide the pruning of specific synapses, emphasizing the role of glial cells in this process. Finally, we discuss how these developmental pruning programmes may go awry in neurodevelopmental disorders of the human CNS, focusing on autism spectrum disorder and schizophrenia. Together, our aim is to give an overview of how the field of activity-dependent pruning research has evolved, led to exciting new questions and guided the identification of new, therapeutically relevant mechanisms that result in aberrant circuit development in neurodevelopmental disorders.
Collapse
Affiliation(s)
- Travis E Faust
- Department of Neurobiology, Brudnick Neuropsychiatric Research Institute, University of Massachusetts Medical School, Worcester, MA, USA
| | - Georgia Gunner
- Department of Neurobiology, Brudnick Neuropsychiatric Research Institute, University of Massachusetts Medical School, Worcester, MA, USA
| | - Dorothy P Schafer
- Department of Neurobiology, Brudnick Neuropsychiatric Research Institute, University of Massachusetts Medical School, Worcester, MA, USA.
| |
Collapse
|
27
|
Bush L, Scott MN. Neuropsychological and ASD phenotypes in rare genetic syndromes: A critical review of the literature. Clin Neuropsychol 2021; 36:993-1027. [PMID: 34569897 DOI: 10.1080/13854046.2021.1980111] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
OBJECTIVE Autism spectrum disorder (ASD) is a complex neurodevelopmental condition characterized by core deficits in social communication and restricted and repetitive behaviors and interests. Recent advances in clinical genetics have improved our understanding of genetic syndromes associated with ASD, which has helped clarify distinct etiologies of ASD and document syndrome-specific profiles of neurocognitive strengths and weaknesses. Pediatric neuropsychologists have the potential to be impactful members of the care team for children with genetic syndromes and their families. METHOD We provide a critical review of the current literature related to the neuropsychological profiles of children with four genetic syndromes associated with ASD, including Tuberous Sclerosis Complex (TSC), fragile X syndrome (FXS), 22q11.2 deletion syndrome, and Angelman syndrome. Recommendations for assessment, intervention, and future directions are provided. RESULTS There is vast heterogeneity in terms of the cognitive, language, and developmental abilities of these populations. The within- and across-syndrome variability characteristic of genetic syndromes should be carefully considered during clinical evaluations, including possible measurement limitations, presence of intellectual disability, and important qualitative differences in the ASD-phenotypes across groups. CONCLUSIONS Individuals with genetic disorders pose challenging diagnostic and assessment questions. Pediatric neuropsychologists with expertise in neurodevelopmental processes are well suited to address these questions and identify profiles of neurocognitive strengths and weaknesses, tailor individualized recommendations, and provide diagnostic clarification.
Collapse
Affiliation(s)
- Lauren Bush
- Department of Psychiatry and Behavioral Sciences, Rush University Medical Center, Chicago, IL, USA
| | - Megan N Scott
- The Pritzker Department of Psychiatry and Behavioral Health, Ann & Robert H. Lurie Children's Hospital of Chicago, IL, USA
| |
Collapse
|
28
|
Mody M, Petibon Y, Han P, Kuruppu D, Ma C, Yokell D, Neelamegam R, Normandin MD, Fakhri GE, Brownell AL. In vivo imaging of mGlu5 receptor expression in humans with Fragile X Syndrome towards development of a potential biomarker. Sci Rep 2021; 11:15897. [PMID: 34354107 PMCID: PMC8342610 DOI: 10.1038/s41598-021-94967-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 07/16/2021] [Indexed: 11/21/2022] Open
Abstract
Fragile X Syndrome (FXS) is a neurodevelopmental disorder caused by silencing of the Fragile X Mental Retardation (FMR1) gene. The resulting loss of Fragile X Mental Retardation Protein (FMRP) leads to excessive glutamate signaling via metabotropic glutamate subtype 5 receptors (mGluR5) which has been implicated in the pathogenesis of the disorder. In the present study we used the radioligand 3-[18F]fluoro-5-(2-pyridinylethynyl)benzonitrile ([18F]FPEB) in simultaneous PET-MR imaging of males with FXS and age- and gender-matched controls to assess the availability of mGlu5 receptors in relevant brain areas. Patients with FXS showed lower [18F]FPEB binding potential (p < 0.01), reflecting reduced mGluR5 availability, than the healthy controls throughout the brain, with significant group differences in insula, anterior cingulate, parahippocampal, inferior temporal and olfactory cortices, regions associated with deficits in inhibition, memory, and visuospatial processes characteristic of the disorder. The results are among the first to provide in vivo evidence of decreased availability of mGluR5 in the brain in individuals with FXS than in healthy controls. The consistent results across the subjects, despite the tremendous challenges with neuroimaging this population, highlight the robustness of the protocol and support for its use in drug occupancy studies; extending our radiotracer development and application efforts from mice to humans.
Collapse
Affiliation(s)
- Maria Mody
- Athinoula A Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02129, USA.
| | - Yoann Petibon
- Gordon Center for Medical Imaging, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02129, USA
| | - Paul Han
- Gordon Center for Medical Imaging, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02129, USA
| | - Darshini Kuruppu
- Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02129, USA
| | - Chao Ma
- Gordon Center for Medical Imaging, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02129, USA
| | - Daniel Yokell
- Gordon Center for Medical Imaging, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02129, USA
| | - Ramesh Neelamegam
- Department of Radiology, University of Texas Health Science at San Antonio, San Antonio, TX, 78229, USA
| | - Marc D Normandin
- Gordon Center for Medical Imaging, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02129, USA
| | - Georges El Fakhri
- Gordon Center for Medical Imaging, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02129, USA
| | - Anna-Liisa Brownell
- Gordon Center for Medical Imaging, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02129, USA
| |
Collapse
|
29
|
Yang Y, Zhao J, Li Y, Li X, Chen X, Feng Z. Fragile X mental retardation protein-regulated proinflammatory cytokine expression in the spinal cord contributes to the pathogenesis of inflammatory pain induced by complete Freund's adjuvant. J Neurochem 2021; 159:512-524. [PMID: 34338322 DOI: 10.1111/jnc.15485] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 07/18/2021] [Accepted: 07/26/2021] [Indexed: 11/25/2022]
Abstract
Studies have verified that Fragile X mental retardation protein (FMRP), an RNA-binding protein, plays a potential role in the pathogenesis of formalin- and (RS)-3,5-dihydroxyphenylglycine (DHPG)-induced abnormal pain sensations. However, the role of FMRP in inflammatory pain has not been reported. Here, we showed an increase in FMRP expression in the spinal dorsal horn (SDH) in a rat model of inflammatory pain induced by complete Freund's adjuvant (CFA). Double immunofluorescence staining revealed that FMRP was mainly expressed in spinal neurons and colocalized with proinflammatory cytokines [tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6)]. After consecutive intrathecal injection of fragile X mental retardation 1 (Fmr1) small interfering RNA (siRNA) for 3 days post-CFA injection, FMRP expression in the SDH was reduced, and CFA-induced hyperalgesia was decreased. In addition, the CFA-induced increase in spinal TNF-α and IL-6 production was significantly suppressed by intrathecal administration of Fmr1 siRNA. Together, these results suggest that FMRP regulates TNF-α and IL-6 levels in the SDH and plays an important role in inflammatory pain.
Collapse
Affiliation(s)
- Yixin Yang
- Department of Pain Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine.,Department of Anesthesiology, Ningbo City First Hospital, Ningbo, Zhejiang, China
| | - Jinsong Zhao
- Department of Pain Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine.,Department of Anesthesiology, Ningbo NO.6 Hospital, Ningbo, Zhejiang, China
| | - Yunze Li
- Department of Pain Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine
| | - Xiangyao Li
- Institute of Neuroscience, Key Laboratory of Medical Neurobiology, Ministry of Health of China, School of Medicine, Zhejiang University
| | - Xiaowei Chen
- Zhejiang Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, China
| | - Zhiying Feng
- Department of Pain Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine
| |
Collapse
|
30
|
Min R, Chen Z, Wang Y, Deng Z, Zhang Y, Deng Y. Quantitative proteomic analysis of cortex in the depressive-like behavior of rats induced by the simulated complex space environment. J Proteomics 2021; 237:104144. [PMID: 33581354 DOI: 10.1016/j.jprot.2021.104144] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 01/29/2021] [Accepted: 01/29/2021] [Indexed: 12/29/2022]
Abstract
Long-term spaceflight has always been challenging for astronauts due to the extremely complicated space environmental conditions, including microgravity, noise, confinement, and circadian rhythms disorders, which may cause adverse effects on astronauts' mental health, such as anxiety and depression. Unfortunately, so far, the underlying mechanism is not fully understood. Hence, a novel type of box and rat cage was designed and built in order to simulate complex space environment on the ground. After earth-based simulation for 21 days, the rats exhibited the depressive-like behavior according to the sucrose preference and forced swimming test. We applied label-free quantitative proteomics to explore the molecular mechanisms of depressive-like behavior through global changes in cortical protein abundance, given that the cortex is the hub of emotional management. The results revealed up-regulated spliceosome proteins in contrast to down-regulated oxidative phosphorylation (OXPHOS), glutamatergic, and GABAergic synapse related proteins in the simulated complex space environment (SCSE) group. Furthermore, PSD-95 protein was found down-regulated in mass spectrometry, reflecting its role in the psychopathology of depression, which was further validated by Western blotting. These findings provide valuable information to better understand the mechanisms of depressive-like behavior. SIGNIFICANCE: Quantitative proteomic analysis can quantify differentially abundant proteins related to a variety of potential signaling pathways in the rat cortex in the simulated complex space environment. These findings not only provide valuable information to better understand the mechanisms of depressive-like behavior, but also might offer the potential targets and develop countermeasures for the mental disorders to maintain the health of astronauts during the long-term spaceflight.
Collapse
Affiliation(s)
- Rui Min
- School of Life Science, Beijing Institute of Technology, Beijing 100081, China
| | - Zixuan Chen
- School of Life Science, Beijing Institute of Technology, Beijing 100081, China
| | - Yun Wang
- School of Life Science, Beijing Institute of Technology, Beijing 100081, China; Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100007, China
| | - Zixuan Deng
- School of Life Science, Beijing Institute of Technology, Beijing 100081, China
| | - Yongqian Zhang
- School of Life Science, Beijing Institute of Technology, Beijing 100081, China.
| | - Yulin Deng
- School of Life Science, Beijing Institute of Technology, Beijing 100081, China
| |
Collapse
|
31
|
Nie PY, Tong L, Li MD, Fu CH, Peng JB, Ji LL. miR-142 downregulation alleviates rat PTSD-like behaviors, reduces the level of inflammatory cytokine expression and apoptosis in hippocampus, and upregulates the expression of fragile X mental retardation protein. J Neuroinflammation 2021; 18:17. [PMID: 33407653 PMCID: PMC7788709 DOI: 10.1186/s12974-020-02064-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 12/16/2020] [Indexed: 11/30/2022] Open
Abstract
Background FMRP is a selective mRNA-binding protein that regulates protein synthesis at synapses, and its loss may lead to the impairment of trace fear memory. Previously, we found that FMRP levels in the hippocampus of rats with post-traumatic stress disorder (PTSD) were decreased. However, the mechanism underlying these changes remains unclear. Methods Forty-eight male Sprague-Dawley rats were randomly divided into four groups. The experimental groups were treated with the single-prolonged stress (SPS) procedure and injected with a lentivirus-mediated inhibitor of miR-142-5p. Behavior test as well as morphology and molecular biology experiments were performed to detect the effect of miR-142 downregulation on PTSD, which was further verified by in vitro experiments. Results We found that silence of miRNA-142 (miR-142), an upstream regulator of FMRP, could alleviate PTSD-like behaviors of rats exposed to the SPS paradigm. MiR-142 silence not only decreased the levels of proinflammatory mediators, such as interleukin-1β, interleukin-6, and tumor necrosis factor-α, but also increased the expressive levels of synaptic proteins including PSD95 and synapsin I in the hippocampus, which was one of the key brain regions associated with PTSD. We further detected that miR-142 silence also downregulated the transportation of nuclear factor kappa-B (NF-κB) into the nuclei of neurons and might further affect the morphology of neurons. Conclusions The results revealed miR-142 downregulation could alleviate PTSD-like behaviors through attenuating neuroinflammation in the hippocampus of SPS rats by binding to FMRP.
Collapse
Affiliation(s)
- Peng-Yin Nie
- Department of Anatomy, College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Lei Tong
- Department of Anatomy, College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Ming-Da Li
- Department of 1st Clinical Medicine, China Medical University, Shenyang, China
| | - Chang-Hai Fu
- Department of Anatomy, College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Jun-Bo Peng
- Department of Anatomy, College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Li-Li Ji
- Department of Anatomy, College of Basic Medical Sciences, China Medical University, Shenyang, China.
| |
Collapse
|
32
|
Feuge J, Scharkowski F, Michaelsen-Preusse K, Korte M. FMRP Modulates Activity-Dependent Spine Plasticity by Binding Cofilin1 mRNA and Regulating Localization and Local Translation. Cereb Cortex 2020; 29:5204-5216. [PMID: 30953439 DOI: 10.1093/cercor/bhz059] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Revised: 02/14/2019] [Accepted: 02/27/2019] [Indexed: 11/13/2022] Open
Abstract
Multiple variants of intellectual disability, e.g., the Fragile X Syndrome are associated with alterations in dendritic spine morphology, thereby pointing to dysregulated actin dynamics during development and processes of synaptic plasticity. Surprisingly, although the necessity of spine actin remodeling was demonstrated repeatedly, the importance and precise role of actin regulators is often undervalued. Here, we provide evidence that structural and functional plasticity are severely impaired after NMDAR-dependent LTP in the hippocampus of Fmr1 KO mice. We can link these defects to an aberrant activity-dependent regulation of Cofilin 1 (cof1) as activity-dependent modulations of local cof1 mRNA availability, local cof1 translation as well as total cof1 expression are impaired in the absence of FMRP. Finally, we can rescue activity-dependent structural plasticity in KO neurons by mimicking the regulation of cof1 observed in WT cells, thereby illustrating the potential of actin modulators to provide novel treatment strategies for the Fragile X Syndrome.
Collapse
Affiliation(s)
- Jonas Feuge
- Division of Cellular Neurobiology, Zoological Institute, TU Braunschweig, Germany
| | | | | | - Martin Korte
- Division of Cellular Neurobiology, Zoological Institute, TU Braunschweig, Germany.,Helmholtz Center for Infection Research, Research group Neuroinflammation and Neurodegeneration, Braunschweig, Germany
| |
Collapse
|
33
|
Westmark CJ, Kiso M, Halfmann P, Westmark PR, Kawaoka Y. Repurposing Fragile X Drugs to Inhibit SARS-CoV-2 Viral Reproduction. Front Cell Dev Biol 2020; 8:856. [PMID: 32984339 PMCID: PMC7479061 DOI: 10.3389/fcell.2020.00856] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Accepted: 08/10/2020] [Indexed: 12/12/2022] Open
Abstract
The COVID-19 pandemic is a global health crisis that requires the application of interdisciplinary research to address numerous knowledge gaps including molecular strategies to prevent viral reproduction in affected individuals. In response to the Frontiers Research Topic, "Coronavirus disease (COVID-19): Pathophysiology, Epidemiology, Clinical Management, and Public Health Response," this Hypothesis article proposes a novel therapeutic strategy to repurpose metabotropic glutamate 5 receptor (mGluR5) inhibitors to interfere with viral hijacking of the host protein synthesis machinery. We review pertinent background on SARS-CoV-2, fragile X syndrome (FXS) and metabotropic glutamate receptor 5 (mGluR5) and provide a mechanistic-based hypothesis and preliminary data to support testing mGluR5 inhibitors in COVID-19 research.
Collapse
Affiliation(s)
- Cara J Westmark
- Department of Neurology, University of Wisconsin-Madison, Madison, WI, United States
| | - Maki Kiso
- Division of Virology, Department of Microbiology and Immunology, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Peter Halfmann
- Department of Pathobiological Sciences, School of Veterinary Medicine, Influenza Research Institute, University of Wisconsin-Madison, Madison, WI, United States
| | - Pamela R Westmark
- Department of Neurology, University of Wisconsin-Madison, Madison, WI, United States
| | - Yoshihiro Kawaoka
- Division of Virology, Department of Microbiology and Immunology, Institute of Medical Science, The University of Tokyo, Tokyo, Japan.,Department of Pathobiological Sciences, School of Veterinary Medicine, Influenza Research Institute, University of Wisconsin-Madison, Madison, WI, United States.,Department of Special Pathogens, International Research Center for Infectious Diseases, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
34
|
Prieto M, Folci A, Martin S. Post-translational modifications of the Fragile X Mental Retardation Protein in neuronal function and dysfunction. Mol Psychiatry 2020; 25:1688-1703. [PMID: 31822816 DOI: 10.1038/s41380-019-0629-4] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 11/22/2019] [Accepted: 11/27/2019] [Indexed: 12/17/2022]
Abstract
The Fragile X Mental Retardation Protein (FMRP) is an RNA-binding protein essential to the regulation of local translation at synapses. In the mammalian brain, synapses are constantly formed and eliminated throughout development to achieve functional neuronal networks. At the molecular level, thousands of proteins cooperate to accomplish efficient neuronal communication. Therefore, synaptic protein levels and their functional interactions need to be tightly regulated. FMRP generally acts as a translational repressor of its mRNA targets. FMRP is the target of several post-translational modifications (PTMs) that dynamically regulate its function. Here we provide an overview of the PTMs controlling the FMRP function and discuss how their spatiotemporal interplay contributes to the physiological regulation of FMRP. Importantly, FMRP loss-of-function leads to Fragile X syndrome (FXS), a rare genetic developmental condition causing a range of neurological alterations including intellectual disability (ID), learning and memory impairments, autistic-like features and seizures. Here, we also explore the possibility that recently reported missense mutations in the FMR1 gene disrupt the PTM homoeostasis of FMRP, thus participating in the aetiology of FXS. This suggests that the pharmacological targeting of PTMs may be a promising strategy to develop innovative therapies for patients carrying such missense mutations.
Collapse
Affiliation(s)
- Marta Prieto
- Université Côte d'Azur, CNRS, IPMC, Valbonne, France
| | | | - Stéphane Martin
- Université Côte d'Azur, INSERM, CNRS, IPMC, Valbonne, France.
| |
Collapse
|
35
|
Chan WK, Griffiths R, Price DJ, Mason JO. Cerebral organoids as tools to identify the developmental roots of autism. Mol Autism 2020; 11:58. [PMID: 32660622 PMCID: PMC7359249 DOI: 10.1186/s13229-020-00360-3] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 06/23/2020] [Indexed: 12/13/2022] Open
Abstract
Some autism spectrum disorders (ASD) likely arise as a result of abnormalities during early embryonic development of the brain. Studying human embryonic brain development directly is challenging, mainly due to ethical and practical constraints. However, the recent development of cerebral organoids provides a powerful tool for studying both normal human embryonic brain development and, potentially, the origins of neurodevelopmental disorders including ASD. Substantial evidence now indicates that cerebral organoids can mimic normal embryonic brain development and neural cells found in organoids closely resemble their in vivo counterparts. However, with prolonged culture, significant differences begin to arise. We suggest that cerebral organoids, in their current form, are most suitable to model earlier neurodevelopmental events and processes such as neurogenesis and cortical lamination. Processes implicated in ASDs which occur at later stages of development, such as synaptogenesis and neural circuit formation, may also be modeled using organoids. The accuracy of such models will benefit from continuous improvements to protocols for organoid differentiation.
Collapse
Affiliation(s)
- Wai Kit Chan
- Centre for Discovery Brain Sciences and Simons Initiative for the Developing Brain, University of Edinburgh, George Square, Edinburgh, EH8 9XD, UK
| | - Rosie Griffiths
- Centre for Discovery Brain Sciences and Simons Initiative for the Developing Brain, University of Edinburgh, George Square, Edinburgh, EH8 9XD, UK
| | - David J Price
- Centre for Discovery Brain Sciences and Simons Initiative for the Developing Brain, University of Edinburgh, George Square, Edinburgh, EH8 9XD, UK
| | - John O Mason
- Centre for Discovery Brain Sciences and Simons Initiative for the Developing Brain, University of Edinburgh, George Square, Edinburgh, EH8 9XD, UK.
| |
Collapse
|
36
|
Yu X, Wang X, Sakano H, Zorio DAR, Wang Y. Dynamics of the fragile X mental retardation protein correlates with cellular and synaptic properties in primary auditory neurons following afferent deprivation. J Comp Neurol 2020; 529:481-500. [PMID: 32449186 DOI: 10.1002/cne.24959] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 04/26/2020] [Accepted: 05/14/2020] [Indexed: 01/01/2023]
Abstract
Afferent activity dynamically regulates neuronal properties and connectivity in the central nervous system. The Fragile X mental retardation protein (FMRP) is an RNA-binding protein that regulates cellular and synaptic properties in an activity-dependent manner. Whether and how FMRP level and localization are regulated by afferent input remains sparsely examined and how such regulation is associated with neuronal response to changes in sensory input is unknown. We characterized changes in FMRP level and localization in the chicken nucleus magnocellularis (NM), a primary cochlear nucleus, following afferent deprivation by unilateral cochlea removal. We observed rapid (within 2 hr) aggregation of FMRP immunoreactivity into large granular structures in a subset of deafferented NM neurons. Neurons that exhibited persistent FMRP aggregation at 12-24 hr eventually lost cytoplasmic Nissl substance, indicating cell death. A week later, FMRP expression in surviving neurons regained its homeostasis, with a slightly reduced immunostaining intensity and enhanced heterogeneity. Correlation analyses under the homeostatic status (7-14 days) revealed that neurons expressing relatively more FMRP had a higher capability of maintaining cell body size and ribosomal activity, as well as a better ability to detach inactive presynaptic terminals. Additionally, the intensity of an inhibitory postsynaptic protein, gephyrin, was reduced following deafferentation and was positively correlated with FMRP intensity, implicating an involvement of FMRP in synaptic dynamics in response to reduced afferent inputs. Collectively, this study demonstrates that afferent input regulates FMRP expression and localization in ways associated with multiple types of neuronal responses and synaptic rearrangements.
Collapse
Affiliation(s)
- Xiaoyan Yu
- Program in Neuroscience, Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, Florida, USA
| | - Xiaoyu Wang
- Program in Neuroscience, Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, Florida, USA.,Division of Histology & Embryology, Key Laboratory for Regenerative Medicine of the Ministry of Education, Medical College, Jinan University, Guangzhou, China
| | - Hitomi Sakano
- Department of Otolaryngology, Bloedel Hearing Research Center, University of Washington, Seattle, Washington, USA.,Department of Otolaryngology, University of Rochester, Rochester, New York, USA
| | - Diego A R Zorio
- Program in Neuroscience, Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, Florida, USA
| | - Yuan Wang
- Program in Neuroscience, Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, Florida, USA
| |
Collapse
|
37
|
Sehovic E, Spahic L, Smajlovic-Skenderagic L, Pistoljevic N, Dzanko E, Hajdarpasic A. Identification of developmental disorders including autism spectrum disorder using salivary miRNAs in children from Bosnia and Herzegovina. PLoS One 2020; 15:e0232351. [PMID: 32353026 PMCID: PMC7192422 DOI: 10.1371/journal.pone.0232351] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Accepted: 04/14/2020] [Indexed: 02/06/2023] Open
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder characterized by major social, communication and behavioural challenges. The cause of ASD is still unclear and it is assumed that environmental, genetic and epigenetic factors influence the risk of ASD occurrence. MicroRNAs (miRNAs) are short 21-25 nucleotide long RNA molecules which post-transcriptionally regulate gene expression. MiRNAs play an important role in central nervous system development; therefore, dysregulation of miRNAs is connected to changes in behaviour and cognition observed in many disorders including ASD. Based on previously published work, on diagnosing ASD using miRNAs, we hypothesized that miRNAs can be used as biomarkers in children with suspected developmental disorders (DD) including ASD within Bosnian-Herzegovinian (B&H) population. 14 selected miRNAs were tested on saliva of children with suspected developmental disorders including ASD. The method of choice was qRT-PCR as a relatively cheap method available in most diagnostic laboratories in low to mid-income countries (LMIC). Out of 14 analysed miRNAs, 6 were differentially expressed between typically developing children and children with some type of developmental disorder including autism spectrum disorder. Using the most optimal logistic regression, we were able to distinguish between ASD and typically developing (TD) children. We have found 5 miRNAs as potential biomarkers. From those, 3 were differentially expressed within the ASD cohort. All 5 miRNAs had shown good chi-square statistics within the logistic regression performed on all 14 analysed miRNAs. The accuracy of 5-miRNAs model training set was 90.2%, while the validation set had a 90% accuracy. This study has shown that miRNAs may be considered as biomarkers for ASD detection and may be used to identify children with ASD along with standard developmental screening tests. By combining these methods we may be able to reach a reliable and accessible diagnostic model for children with ASD in LMIC such as B&H.
Collapse
Affiliation(s)
- Emir Sehovic
- Genetics and Bioengineering, International Burch University, Sarajevo, Bosnia and Herzegovina
| | - Lemana Spahic
- Genetics and Bioengineering, International Burch University, Sarajevo, Bosnia and Herzegovina
| | | | | | - Eldin Dzanko
- Education for All (EDUS), Sarajevo, Bosnia and Herzegovina
| | - Aida Hajdarpasic
- Department of Medical Biology, Sarajevo Medical School, Sarajevo School of Science and Technology, Sarajevo, Bosnia and Herzegovina
- * E-mail:
| |
Collapse
|
38
|
Rodriguez CM, Wright SE, Kearse MG, Haenfler JM, Flores BN, Liu Y, Ifrim MF, Glineburg MR, Krans A, Jafar-Nejad P, Sutton MA, Bassell GJ, Parent JM, Rigo F, Barmada SJ, Todd PK. A native function for RAN translation and CGG repeats in regulating fragile X protein synthesis. Nat Neurosci 2020; 23:386-397. [PMID: 32066985 PMCID: PMC7668390 DOI: 10.1038/s41593-020-0590-1] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Accepted: 01/10/2020] [Indexed: 02/07/2023]
Abstract
Repeat-associated non-AUG-initiated translation of expanded CGG repeats (CGG RAN) from the FMR1 5'-leader produces toxic proteins that contribute to neurodegeneration in fragile X-associated tremor/ataxia syndrome. Here we describe how unexpanded CGG repeats and their translation play conserved roles in regulating fragile X protein (FMRP) synthesis. In neurons, CGG RAN acts as an inhibitory upstream open reading frame to suppress basal FMRP production. Activation of mGluR5 receptors enhances FMRP synthesis. This enhancement requires both the CGG repeat and CGG RAN initiation sites. Using non-cleaving antisense oligonucleotides (ASOs), we selectively blocked CGG RAN. This ASO blockade enhanced endogenous FMRP expression in human neurons. In human and rodent neurons, CGG RAN-blocking ASOs suppressed repeat toxicity and prolonged survival. These findings delineate a native function for CGG repeats and RAN translation in regulating basal and activity-dependent FMRP synthesis, and they demonstrate the therapeutic potential of modulating CGG RAN translation in fragile X-associated disorders.
Collapse
Affiliation(s)
- Caitlin M Rodriguez
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI, USA
| | - Shannon E Wright
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI, USA
| | - Michael G Kearse
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA
- Department of Biological Chemistry and Pharmacology, Center for RNA Biology, Ohio State University, Columbus, OH, USA
| | - Jill M Haenfler
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA
| | - Brittany N Flores
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA
- Cellular and Molecular Biology Graduate Program, University of Michigan, Ann Arbor, MI, USA
| | - Yu Liu
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA
| | - Marius F Ifrim
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA, USA
| | - Mary R Glineburg
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA
| | - Amy Krans
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA
- VA Ann Arbor Healthcare System, Ann Arbor, MI, USA
| | | | - Michael A Sutton
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Gary J Bassell
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA, USA
| | - Jack M Parent
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA
- VA Ann Arbor Healthcare System, Ann Arbor, MI, USA
| | - Frank Rigo
- Ionis Pharmaceuticals, Carlsbad, CA, USA
| | - Sami J Barmada
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA
| | - Peter K Todd
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA.
- VA Ann Arbor Healthcare System, Ann Arbor, MI, USA.
| |
Collapse
|
39
|
Choe HK, Cho J. Comprehensive Genome-Wide Approaches to Activity-Dependent Translational Control in Neurons. Int J Mol Sci 2020; 21:ijms21051592. [PMID: 32111062 PMCID: PMC7084349 DOI: 10.3390/ijms21051592] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 02/21/2020] [Accepted: 02/25/2020] [Indexed: 02/06/2023] Open
Abstract
Activity-dependent regulation of gene expression is critical in experience-mediated changes in the brain. Although less appreciated than transcriptional control, translational control is a crucial regulatory step of activity-mediated gene expression in physiological and pathological conditions. In the first part of this review, we overview evidence demonstrating the importance of translational controls under the context of synaptic plasticity as well as learning and memory. Then, molecular mechanisms underlying the translational control, including post-translational modifications of translation factors, mTOR signaling pathway, and local translation, are explored. We also summarize how activity-dependent translational regulation is associated with neurodevelopmental and psychiatric disorders, such as autism spectrum disorder and depression. In the second part, we highlight how recent application of high-throughput sequencing techniques has added insight into genome-wide studies on translational regulation of neuronal genes. Sequencing-based strategies to identify molecular signatures of the active neuronal population responding to a specific stimulus are discussed. Overall, this review aims to highlight the implication of translational control for neuronal gene regulation and functions of the brain and to suggest prospects provided by the leading-edge techniques to study yet-unappreciated translational regulation in the nervous system.
Collapse
Affiliation(s)
- Han Kyoung Choe
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Korea
- Correspondence: (H.K.C.); (J.C.)
| | - Jun Cho
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Korea
- Correspondence: (H.K.C.); (J.C.)
| |
Collapse
|
40
|
Suardi GAM, Haddad LA. FMRP ribonucleoprotein complexes and RNA homeostasis. ADVANCES IN GENETICS 2020; 105:95-136. [PMID: 32560791 DOI: 10.1016/bs.adgen.2020.01.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The Fragile Mental Retardation 1 gene (FMR1), at Xq27.3, encodes the fragile mental retardation protein (FMRP), and displays in its 5'-untranslated region a series of polymorphic CGG triplet repeats that may undergo dynamic mutation. Fragile X syndrome (FXS) is the leading cause of inherited intellectual disability among men, and is most frequently due to FMR1 full mutation and consequent transcription repression. FMR1 premutations may associate with at least two other clinical conditions, named fragile X-associated primary ovarian insufficiency (FXPOI) and tremor and ataxia syndrome (FXTAS). While FXPOI and FXTAS appear to be mediated by FMR1 mRNA accumulation, relative reduction of FMRP, and triplet repeat translation, FXS is due to the lack of the RNA-binding protein FMRP. Besides its function as mRNA translation repressor in neuronal and stem/progenitor cells, RNA editing roles have been assigned to FMRP. In this review, we provide a brief description of FMR1 transcribed microsatellite and associated clinical disorders, and discuss FMRP molecular roles in ribonucleoprotein complex assembly and trafficking, as well as aspects of RNA homeostasis affected in FXS cells.
Collapse
Affiliation(s)
- Gabriela Aparecida Marcondes Suardi
- Human Genome and Stem Cell Research Center, Department of Genetics and Evolutionary Biology, Instituto de Biociências, Universidade de São Paulo, São Paulo, Brazil
| | - Luciana Amaral Haddad
- Human Genome and Stem Cell Research Center, Department of Genetics and Evolutionary Biology, Instituto de Biociências, Universidade de São Paulo, São Paulo, Brazil.
| |
Collapse
|
41
|
Graef JD, Wu H, Ng C, Sun C, Villegas V, Qadir D, Jesseman K, Warren ST, Jaenisch R, Cacace A, Wallace O. Partial FMRP expression is sufficient to normalize neuronal hyperactivity in Fragile X neurons. Eur J Neurosci 2020; 51:2143-2157. [PMID: 31880363 PMCID: PMC7318714 DOI: 10.1111/ejn.14660] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 12/04/2019] [Accepted: 12/11/2019] [Indexed: 12/12/2022]
Abstract
Fragile X syndrome (FXS) is the most common genetic form of intellectual disability caused by a CGG repeat expansion in the 5′‐UTR of the Fragile X mental retardation gene FMR1, triggering epigenetic silencing and the subsequent absence of the protein, FMRP. Reactivation of FMR1 represents an attractive therapeutic strategy targeting the genetic root cause of FXS. However, largely missing in the FXS field is an understanding of how much FMR1 reactivation is required to rescue FMRP‐dependent mutant phenotypes. Here, we utilize FXS patient‐derived excitatory neurons to model FXS in vitro and confirm that the absence of FMRP leads to neuronal hyperactivity. We further determined the levels of FMRP and the percentage of FMRP‐positive cells necessary to correct this phenotype utilizing a mixed and mosaic neuronal culture system and a combination of CRISPR, antisense and expression technologies to titrate FMRP in FXS and WT neurons. Our data demonstrate that restoration of greater than 5% of overall FMRP expression levels or greater than 20% FMRP‐expressing neurons in a mosaic pattern is sufficient to normalize a FMRP‐dependent, hyperactive phenotype in FXS iPSC‐derived neurons.
Collapse
Affiliation(s)
| | - Hao Wu
- Fulcrum Therapeutics, Cambridge, MA, USA
| | - Carrie Ng
- Fulcrum Therapeutics, Cambridge, MA, USA
| | | | | | | | | | - Stephen T Warren
- Departments of Human Genetics, Biochemistry, and Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
| | - Rudolf Jaenisch
- Department of Biology, MIT, 9 Cambridge Center, Whitehead Institute, Cambridge, MA, USA
| | | | | |
Collapse
|
42
|
Kainic acid-induced status epilepticus decreases mGlu 5 receptor and phase-specifically downregulates Homer1b/c expression. Brain Res 2019; 1730:146640. [PMID: 31891692 DOI: 10.1016/j.brainres.2019.146640] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 12/05/2019] [Accepted: 12/27/2019] [Indexed: 12/25/2022]
Abstract
Globally, over 50 million people are affected by epilepsy, which is characterized by the occurrence of spontaneous recurrent seizures. Almost one-third of the patients show resistance to current anti-epileptic drugs, making the exploration of new molecular targets necessary. An interesting target may be Homer1, due to its diverse roles in epileptogenesis and synaptic plasticity. Indeed, Homer1 regulates group I metabotropic glutamate (mGlu) receptors (i.e. mGlu1 and mGlu5) scaffolding and signaling in neurons. In the present work, using the systemic kainic acid (KA)-induced status epilepticus (SE) model in adult rats, we investigated the mRNA and protein expression patterns of the mGlu5 receptor, Homer1a and Homer1b/c at 10, 80 and 120 days post-SE (i.e. T10, T80 and T120). Epileptogenesis was validated by electrophysiological recordings of seizures via electroencephalography (EEG) monitoring and through upregulation of glial fibrillary acidic protein. At the protein level, the mGlu5 receptor was downregulated in the late latent phase (T10) and the early- and late exponential growth phase (T80 and T120, respectively), which was best observed in the hippocampal CA1 region. At mRNA level, significant downregulation of the mGlu5 receptor was only detected in the late exponential growth phase. Homer1a expression did not change at any investigated time point. Interestingly, Homer1b/c was only downregulated in the late latent phase, a period where spontaneous seizures are extremely rare. Thus, this phase-specific downregulation may be indicative of an endogenous neuroprotective mechanism. In conclusion, these results suggest that Homer1b/c may be an interesting molecular target to prevent epileptogenesis and/or control seizures.
Collapse
|
43
|
Nussbacher JK, Tabet R, Yeo GW, Lagier-Tourenne C. Disruption of RNA Metabolism in Neurological Diseases and Emerging Therapeutic Interventions. Neuron 2019; 102:294-320. [PMID: 30998900 DOI: 10.1016/j.neuron.2019.03.014] [Citation(s) in RCA: 166] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 01/24/2019] [Accepted: 03/12/2019] [Indexed: 02/06/2023]
Abstract
RNA binding proteins are critical to the maintenance of the transcriptome via controlled regulation of RNA processing and transport. Alterations of these proteins impact multiple steps of the RNA life cycle resulting in various molecular phenotypes such as aberrant RNA splicing, transport, and stability. Disruption of RNA binding proteins and widespread RNA processing defects are increasingly recognized as critical determinants of neurological diseases. Here, we describe distinct mechanisms by which the homeostasis of RNA binding proteins is compromised in neurological disorders through their reduced expression level, increased propensity to aggregate or sequestration by abnormal RNAs. These mechanisms all converge toward altered neuronal function highlighting the susceptibility of neurons to deleterious changes in RNA expression and the central role of RNA binding proteins in preserving neuronal integrity. Emerging therapeutic approaches to mitigate or reverse alterations of RNA binding proteins in neurological diseases are discussed.
Collapse
Affiliation(s)
- Julia K Nussbacher
- Department of Cellular and Molecular Medicine, Institute for Genomic Medicine, UCSD Stem Cell Program, University of California, San Diego, La Jolla, CA, USA
| | - Ricardos Tabet
- Department of Neurology, The Sean M. Healey and AMG Center for ALS at Mass General, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA; Broad Institute of Harvard University and MIT, Cambridge, MA 02142, USA
| | - Gene W Yeo
- Department of Cellular and Molecular Medicine, Institute for Genomic Medicine, UCSD Stem Cell Program, University of California, San Diego, La Jolla, CA, USA.
| | - Clotilde Lagier-Tourenne
- Department of Neurology, The Sean M. Healey and AMG Center for ALS at Mass General, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA; Broad Institute of Harvard University and MIT, Cambridge, MA 02142, USA.
| |
Collapse
|
44
|
Park H, Kaang BK. Balanced actions of protein synthesis and degradation in memory formation. ACTA ACUST UNITED AC 2019; 26:299-306. [PMID: 31416903 PMCID: PMC6699412 DOI: 10.1101/lm.048785.118] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 07/02/2019] [Indexed: 11/24/2022]
Abstract
Storage of long-term memory requires not only protein synthesis but also protein degradation. In this article, we overview recent publications related to this issue, stressing that the balanced actions of protein synthesis and degradation are critical for long-term memory formation. We particularly focused on the brain-derived neurotrophic factor signaling that leads to protein synthesis; proteasome- and autophagy-dependent protein degradation that removes molecular constraints; the role of Fragile X mental retardation protein in translational suppression; and epigenetic modifications that control gene expression at the genomic level. Numerous studies suggest that an imbalance between protein synthesis and degradation leads to intellectual impairment and cognitive disorders.
Collapse
Affiliation(s)
- Hyungju Park
- Department of Structure and Function of Neural Network, Korea Brain Research Institute (KBRI), Daegu 41062, South Korea.,Department of Brain and Cognitive Sciences, DGIST, Daegu 42988, South Korea
| | - Bong-Kiun Kaang
- School of Biological Sciences, Seoul National University, Seoul 08826, South Korea
| |
Collapse
|
45
|
Bellosta P, Soldano A. Dissecting the Genetics of Autism Spectrum Disorders: A Drosophila Perspective. Front Physiol 2019; 10:987. [PMID: 31481894 PMCID: PMC6709880 DOI: 10.3389/fphys.2019.00987] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2019] [Accepted: 07/18/2019] [Indexed: 01/10/2023] Open
Abstract
Autism Spectrum Disorder (ASD) is a complex group of multi-factorial developmental disorders that leads to communication and behavioral defects. Genetic alterations have been identified in around 20% of ASD patients and the use of genetic models, such as Drosophila melanogaster, has been of paramount importance in deciphering the significance of these alterations. In fact, many of the ASD associated genes, such as FMR1, Neurexin, Neuroligins and SHANK encode for proteins that have conserved functions in neurons and during synapse development, both in humans and in the fruit fly. Drosophila is a prominent model in neuroscience due to the conserved genetic networks that control neurodevelopmental processes and to the ease of manipulating its genetics. In the present review we will describe recent advances in the field of ASD with a particular focus on the characterization of genes where the use of Drosophila has been fundamental to better understand their function.
Collapse
Affiliation(s)
- Paola Bellosta
- Laboratory of Metabolism of Cell Growth and Neuronal Survival, Department of Cellular, Computational and Integrative Biology (CIBio), University of Trento, Trento, Italy.,Department of Medicine, New York University Langone Medical Center, New York, NY, United States
| | - Alessia Soldano
- Laboratory of Translational Genomics, Department of Cellular, Computational and Integrative Biology (CIBio), University of Trento, Trento, Italy
| |
Collapse
|
46
|
Kute PM, Ramakrishna S, Neelagandan N, Chattarji S, Muddashetty RS. NMDAR mediated translation at the synapse is regulated by MOV10 and FMRP. Mol Brain 2019; 12:65. [PMID: 31291981 PMCID: PMC6617594 DOI: 10.1186/s13041-019-0473-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Accepted: 05/08/2019] [Indexed: 01/08/2023] Open
Abstract
Protein synthesis is crucial for maintaining synaptic plasticity and synaptic signalling. Here we have attempted to understand the role of RNA binding proteins, Fragile X Mental Retardation Protein (FMRP) and Moloney Leukemia Virus 10 (MOV10) protein in N-Methyl-D-Aspartate Receptor (NMDAR) mediated translation regulation. We show that FMRP is required for translation downstream of NMDAR stimulation and MOV10 is the key specificity factor in this process. In rat cortical synaptoneurosomes, MOV10 in association with FMRP and Argonaute 2 (AGO2) forms the inhibitory complex on a subset of NMDAR responsive mRNAs. On NMDAR stimulation, MOV10 dissociates from AGO2 and promotes the translation of its target mRNAs. FMRP is required to form MOV10-AGO2 inhibitory complex and to promote translation of MOV10 associated mRNAs. Phosphorylation of FMRP appears to be the potential switch for NMDAR mediated translation and in the absence of FMRP, the distinct translation response to NMDAR stimulation is lost. Thus, FMRP and MOV10 have an important regulatory role in NMDAR mediated translation at the synapse.
Collapse
Affiliation(s)
- Preeti Madhav Kute
- Centre for Brain Development and Repair (CBDR), Institute for Stem Cell Science and Regenerative Medicine (inStem), Bangalore, 560065, India.,School of Chemical and Biotechnology, Shanmugha Arts, Science and Technology & Research Academy (SASTRA) University, Thanjavur, 613401, India
| | - Sarayu Ramakrishna
- Centre for Brain Development and Repair (CBDR), Institute for Stem Cell Science and Regenerative Medicine (inStem), Bangalore, 560065, India.,The University of Trans-Disciplinary Health Sciences and Technology, Bangalore, 560064, India
| | - Nagammal Neelagandan
- Centre for Brain Development and Repair (CBDR), Institute for Stem Cell Science and Regenerative Medicine (inStem), Bangalore, 560065, India
| | - Sumantra Chattarji
- Centre for Brain Development and Repair (CBDR), Institute for Stem Cell Science and Regenerative Medicine (inStem), Bangalore, 560065, India.,National Centre for Biological Sciences (NCBS), Bangalore, 560065, India.,Centre for Discovery Brain Sciences, Deanery of Biomedical Sciences, University of Edinburgh, Edinburgh, EH89XD, UK
| | - Ravi S Muddashetty
- Centre for Brain Development and Repair (CBDR), Institute for Stem Cell Science and Regenerative Medicine (inStem), Bangalore, 560065, India.
| |
Collapse
|
47
|
Verma V, Paul A, Amrapali Vishwanath A, Vaidya B, Clement JP. Understanding intellectual disability and autism spectrum disorders from common mouse models: synapses to behaviour. Open Biol 2019; 9:180265. [PMID: 31185809 PMCID: PMC6597757 DOI: 10.1098/rsob.180265] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Normal brain development is highly dependent on the timely coordinated actions of genetic and environmental processes, and an aberration can lead to neurodevelopmental disorders (NDDs). Intellectual disability (ID) and autism spectrum disorders (ASDs) are a group of co-occurring NDDs that affect between 3% and 5% of the world population, thus presenting a great challenge to society. This problem calls for the need to understand the pathobiology of these disorders and to design new therapeutic strategies. One approach towards this has been the development of multiple analogous mouse models. This review discusses studies conducted in the mouse models of five major monogenic causes of ID and ASDs: Fmr1, Syngap1, Mecp2, Shank2/3 and Neuroligins/Neurnexins. These studies reveal that, despite having a diverse molecular origin, the effects of these mutations converge onto similar or related aetiological pathways, consequently giving rise to the typical phenotype of cognitive, social and emotional deficits that are characteristic of ID and ASDs. This convergence, therefore, highlights common pathological nodes that can be targeted for therapy. Other than conventional therapeutic strategies such as non-pharmacological corrective methods and symptomatic alleviation, multiple studies in mouse models have successfully proved the possibility of pharmacological and genetic therapy enabling functional recovery.
Collapse
Affiliation(s)
- Vijaya Verma
- Neuroscience Unit, Jawaharlal Nehru Centre for Advanced Scientific Research , Jakkur, Bengaluru 560 064, Karnataka, India
| | - Abhik Paul
- Neuroscience Unit, Jawaharlal Nehru Centre for Advanced Scientific Research , Jakkur, Bengaluru 560 064, Karnataka, India
| | - Anjali Amrapali Vishwanath
- Neuroscience Unit, Jawaharlal Nehru Centre for Advanced Scientific Research , Jakkur, Bengaluru 560 064, Karnataka, India
| | - Bhupesh Vaidya
- Neuroscience Unit, Jawaharlal Nehru Centre for Advanced Scientific Research , Jakkur, Bengaluru 560 064, Karnataka, India
| | - James P Clement
- Neuroscience Unit, Jawaharlal Nehru Centre for Advanced Scientific Research , Jakkur, Bengaluru 560 064, Karnataka, India
| |
Collapse
|
48
|
Westmark CJ. Fragile X and APP: a Decade in Review, a Vision for the Future. Mol Neurobiol 2019; 56:3904-3921. [PMID: 30225775 PMCID: PMC6421119 DOI: 10.1007/s12035-018-1344-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Accepted: 09/05/2018] [Indexed: 10/28/2022]
Abstract
Fragile X syndrome (FXS) is a devastating developmental disability that has profound effects on cognition, behavior, and seizure susceptibility. There are currently no treatments that target the underlying cause of the disorder, and recent clinical trials have been unsuccessful. In 2007, seminal work demonstrated that amyloid-beta protein precursor (APP) is dysregulated in Fmr1KO mice through a metabotropic glutamate receptor 5 (mGluR5)-dependent pathway. These findings raise the hypotheses that: (1) APP and/or APP metabolites are potential therapeutic targets as well as biomarkers for FXS and (2) mGluR5 inhibitors may be beneficial in the treatment of Alzheimer's disease. Herein, advances in the field over the past decade that have reproduced and greatly expanded upon these original findings are reviewed, and required experimentation to validate APP metabolites as potential disease biomarkers as well as therapeutic targets for FXS are discussed.
Collapse
Affiliation(s)
- Cara J Westmark
- Department of Neurology, University of Wisconsin-Madison, Medical Sciences Center, Room 3619, 1300 University Avenue, Madison, WI, USA.
| |
Collapse
|
49
|
Huang G, Zhu H, Wu S, Cui M, Xu T. Long Noncoding RNA Can Be a Probable Mechanism and a Novel Target for Diagnosis and Therapy in Fragile X Syndrome. Front Genet 2019; 10:446. [PMID: 31191598 PMCID: PMC6541098 DOI: 10.3389/fgene.2019.00446] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Accepted: 04/30/2019] [Indexed: 01/06/2023] Open
Abstract
Fragile X syndrome (FXS) is the most common congenital hereditary disease of low intelligence after Down syndrome. Its main pathogenic gene is fragile X mental retardation 1 (FMR1) gene associated with intellectual disability, autism, and fragile X-related primary ovarian insufficiency (FXPOI) and fragile X-associated tremor/ataxia syndrome (FXTAS). FMR1 gene transcription leads to the absence of fragile X mental retardation protein (FMRP). How to relieve or cure disorders associated with FXS has also become a clinically disturbing problem. Previous studies have recently shown that long noncoding RNAs (lncRNAs) contribute to the pathogenesis. And it has been identified that several lncRNAs including FMR4, FMR5, and FMR6 contribute to developing FXPOI/FXTAS, originating from the FMR1 gene locus. FMR4 is a product of RNA polymerase II and can regulate the expression of relevant genes during differentiation of human neural precursor cells. FMR5 is a sense-oriented transcript while FMR6 is an antisense lncRNA produced by the 3' UTR of FMR1. FMR6 is likely to contribute to developing FXPOI, and it overlaps exons 15-17 of FMR1 as well as two microRNA binding sites. Additionally, BC1 can bind FMRP to form an inhibitory complex and lncRNA TUG1 also can control axonal development by directly interacting with FMRP through modulating SnoN-Ccd1 pathway. Therefore, these lncRNAs provide pharmaceutical targets and novel biomarkers. This review will: (1) describe the clinical manifestations and traditional pathogenesis of FXS and FXTAS/FXPOI; (2) summarize what is known about the role of lncRNAs in the pathogenesis of FXS and FXTAS/FXPOI; and (3) provide an outlook of potential effects and future directions of lncRNAs in FXS and FXTAS/FXPOI researches.
Collapse
Affiliation(s)
- Ge Huang
- The Second Hospital of Jilin University, Changchun, China
| | - He Zhu
- The Second Hospital of Jilin University, Changchun, China
| | - Shuying Wu
- The Second Hospital of Jilin University, Changchun, China
| | - Manhua Cui
- The Second Hospital of Jilin University, Changchun, China
| | - Tianmin Xu
- The Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
50
|
hnRNP Q Regulates Internal Ribosome Entry Site-Mediated fmr1 Translation in Neurons. Mol Cell Biol 2019; 39:MCB.00371-18. [PMID: 30478144 DOI: 10.1128/mcb.00371-18] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Accepted: 11/15/2018] [Indexed: 01/05/2023] Open
Abstract
Fragile X syndrome (FXS) caused by loss of fragile X mental retardation protein (FMRP), is the most common cause of inherited intellectual disability. Numerous studies show that FMRP is an RNA binding protein that regulates translation of its binding targets and plays key roles in neuronal functions. However, the regulatory mechanism for FMRP expression is incompletely understood. Conflicting results regarding internal ribosome entry site (IRES)-mediated fmr1 translation have been reported. Here, we unambiguously demonstrate that the fmr1 gene, which encodes FMRP, exploits both IRES-mediated translation and canonical cap-dependent translation. Furthermore, we find that heterogeneous nuclear ribonucleoprotein Q (hnRNP Q) acts as an IRES-transacting factor (ITAF) for IRES-mediated fmr1 translation in neurons. We also show that semaphorin 3A (Sema3A)-induced axonal growth cone collapse is due to upregulation of hnRNP Q and subsequent IRES-mediated expression of FMRP. These data elucidate the regulatory mechanism of FMRP expression and its role in axonal growth cone collapse.
Collapse
|