51
|
Bülow P, Wenner PA, Faundez V, Bassell GJ. Mitochondrial Structure and Polarity in Dendrites and the Axon Initial Segment Are Regulated by Homeostatic Plasticity and Dysregulated in Fragile X Syndrome. Front Cell Dev Biol 2021; 9:702020. [PMID: 34350185 PMCID: PMC8327182 DOI: 10.3389/fcell.2021.702020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 06/14/2021] [Indexed: 12/21/2022] Open
Abstract
Mitochondrial dysfunction has long been overlooked in neurodevelopmental disorders, but recent studies have provided new links to genetic forms of autism, including Rett syndrome and fragile X syndrome (FXS). Mitochondria show plasticity in morphology and function in response to neuronal activity, and previous research has reported impairments in mitochondrial morphology and function in disease. We and others have previously reported abnormalities in distinct types of homeostatic plasticity in FXS. It remains unknown if or how activity deprivation triggering homeostatic plasticity affects mitochondria in axons and/or dendrites and whether impairments occur in neurodevelopmental disorders. Here, we test the hypothesis that mitochondria are structurally and functionally modified in a compartment-specific manner during homeostatic plasticity using a model of activity deprivation in cortical neurons from wild-type mice and that this plasticity-induced regulation is altered in Fmr1-knockout (KO) neurons. We uncovered dendrite-specific regulation of the mitochondrial surface area, whereas axon initial segment (AIS) mitochondria show changes in polarity; both responses are lost in the Fmr1 KO. Taken together, our results demonstrate impairments in mitochondrial plasticity in FXS, which has not previously been reported. These results suggest that mitochondrial dysregulation in FXS could contribute to abnormal neuronal plasticity, with broader implications to other neurodevelopmental disorders and therapeutic strategies.
Collapse
Affiliation(s)
- Pernille Bülow
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA, United States
| | - Peter A Wenner
- Department of Physiology, Emory University School of Medicine, Atlanta, GA, United States
| | - Victor Faundez
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA, United States
| | - Gary J Bassell
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA, United States
| |
Collapse
|
52
|
Booker SA, Kind PC. Mechanisms regulating input-output function and plasticity of neurons in the absence of FMRP. Brain Res Bull 2021; 175:69-80. [PMID: 34245842 DOI: 10.1016/j.brainresbull.2021.06.025] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 06/13/2021] [Accepted: 06/29/2021] [Indexed: 12/12/2022]
Abstract
The function of brain circuits relies on high-fidelity information transfer within neurons. Synaptic inputs arrive primarily at dendrites, where they undergo integration and summation throughout the somatodendritic domain, ultimately leading to the generation of precise patterns of action potentials. Emerging evidence suggests that the ability of neurons to transfer synaptic information and modulate their output is impaired in a number of neurodevelopmental disorders including Fragile X Syndrome. In this review we summarise recent findings that have revealed the pathophysiological and plasticity mechanisms that alter the ability of neurons in sensory and limbic circuits to reliably code information in the absence of FMRP. We examine which aspects of this transform may result directly from the loss of FMRP and those that a result from compensatory or homeostatic alterations to neuronal function. Dissection of the mechanisms leading to altered input-output function of neurons in the absence of FMRP and their effects on regulating neuronal plasticity throughout development could have important implications for potential therapies for Fragile X Syndrome, including directing the timing and duration of different treatment options.
Collapse
Affiliation(s)
- Sam A Booker
- Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh, UK; Patrick Wild Centre for Autism Research, University of Edinburgh, Edinburgh, UK; Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK.
| | - Peter C Kind
- Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh, UK; Patrick Wild Centre for Autism Research, University of Edinburgh, Edinburgh, UK; Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK; National Centre for Biological Sciences (NCBS), Bangalore, India.
| |
Collapse
|
53
|
Huebschman JL, Davis MC, Tovar Pensa C, Guo Y, Smith LN. The fragile X mental retardation protein promotes adjustments in cocaine self-administration that preserve reinforcement level. Eur J Neurosci 2021; 54:4920-4933. [PMID: 34133054 DOI: 10.1111/ejn.15356] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 05/08/2021] [Accepted: 06/08/2021] [Indexed: 01/29/2023]
Abstract
The fragile X mental retardation protein (FMRP), an RNA-binding protein, regulates cocaine-induced neuronal plasticity and is critical for the normal development of drug-induced locomotor sensitization, as well as reward-related learning in the conditioned place preference assay. However, it is unknown whether FMRP impacts behaviors that are used to more closely model substance use disorders. Utilizing a cocaine intravenous self-administration (IVSA) assay in Fmr1 knockout (KO) and wild-type (WT) littermate mice, we find that, despite normal acquisition and extinction learning, Fmr1 KO mice fail to make a normal upward shift in responding during dose-response testing. Later, when given access to the original acquisition dose under increasing fixed ratio (FR) schedules of reinforcement (FR1, FR3, and FR5), Fmr1 KO mice earn significantly fewer cocaine infusions than WT mice. Importantly, similar deficits are not present in operant conditioning using a palatable food reinforcer, indicating that our results do not represent broad learning or reward-related deficits in Fmr1 KO mice. Additionally, we find an FMRP target, the activity-regulated cytoskeleton-associated protein (Arc), to be significantly reduced in synaptic cellular fractions prepared from the nucleus accumbens of Fmr1 KO, compared with WT, mice following operant tasks reinforced with cocaine but not food. Overall, our findings suggest that FMRP facilitates adjustments in drug self-administration behavior that generally serve to preserve reinforcement level, and combined with our similar IVSA findings in Arc KO mice may implicate Arc, along with FMRP, in behavioral shifts that occur in drug taking when drug availability is altered.
Collapse
Affiliation(s)
- Jessica L Huebschman
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University Health Science Center, Bryan, Texas, USA.,Texas A&M Institute for Neuroscience, Texas A&M University, College Station, Texas, USA
| | - Megan C Davis
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University Health Science Center, Bryan, Texas, USA
| | - Catherina Tovar Pensa
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University Health Science Center, Bryan, Texas, USA
| | - Yuhong Guo
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University Health Science Center, Bryan, Texas, USA
| | - Laura N Smith
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University Health Science Center, Bryan, Texas, USA.,Texas A&M Institute for Neuroscience, Texas A&M University, College Station, Texas, USA
| |
Collapse
|
54
|
Tiwari A, Rahi S, Mehan S. Elucidation of Abnormal Extracellular Regulated Kinase (ERK) Signaling and Associations with Syndromic and Non-syndromic Autism. Curr Drug Targets 2021; 22:1071-1086. [PMID: 33081671 DOI: 10.2174/1389450121666201020155010] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 09/21/2020] [Accepted: 09/26/2020] [Indexed: 11/22/2022]
Abstract
Autism is a highly inherited and extremely complex disorder in which results from various cases indicate chromosome anomalies, unusual single-gene mutations, and multiplicative effects of particular gene variants, characterized primarily by impaired speech and social interaction and restricted behavior. The precise etiology of Autism Spectrum Disorder (ASD) is currently unclear. The extracellular signal-regulated kinase (ERK) signaling mechanism affects neurogenesis and neuronal plasticity during the development of the central nervous mechanism. In this regard, the pathway of ERK has recently gained significant interest in the pathogenesis of ASD. The mutation occurs in a few ERK components. Besides, the ERK pathway dysfunction lies in the upstream of modified translation and contributes to synapse pathology in syndromic types of autism. In this review, we highlight the ERK pathway as a target for neurodevelopmental disorder autism. In addition, we summarize the regulation of the ERK pathway with ERK inhibitors in neurological disorders. In conclusion, a better understanding of the ERK signaling pathway provides a range of therapeutic options for autism spectrum disorder.
Collapse
Affiliation(s)
- Aarti Tiwari
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India
| | - Saloni Rahi
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India
| | - Sidharth Mehan
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India
| |
Collapse
|
55
|
Gonzalez-Lozano MA, Wortel J, van der Loo RJ, van Weering JRT, Smit AB, Li KW. Reduced mGluR5 Activity Modulates Mitochondrial Function. Cells 2021; 10:cells10061375. [PMID: 34199502 PMCID: PMC8228325 DOI: 10.3390/cells10061375] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 05/23/2021] [Accepted: 05/31/2021] [Indexed: 11/30/2022] Open
Abstract
The metabotropic glutamate receptor 5 (mGluR5) is an essential modulator of synaptic plasticity, learning and memory; whereas in pathological conditions, it is an acknowledged therapeutic target that has been implicated in multiple brain disorders. Despite robust pre-clinical data, mGluR5 antagonists failed in several clinical trials, highlighting the need for a better understanding of the mechanisms underlying mGluR5 function. In this study, we dissected the molecular synaptic modulation mediated by mGluR5 using genetic and pharmacological mouse models to chronically and acutely reduce mGluR5 activity. We found that next to dysregulation of synaptic proteins, the major regulation in protein expression in both models concerned specific processes in mitochondria, such as oxidative phosphorylation. Second, we observed morphological alterations in shape and area of specifically postsynaptic mitochondria in mGluR5 KO synapses using electron microscopy. Third, computational and biochemical assays suggested an increase of mitochondrial function in neurons, with increased level of NADP/H and oxidative damage in mGluR5 KO. Altogether, our observations provide diverse lines of evidence of the modulation of synaptic mitochondrial function by mGluR5. This connection suggests a role for mGluR5 as a mediator between synaptic activity and mitochondrial function, a finding which might be relevant for the improvement of the clinical potential of mGluR5.
Collapse
Affiliation(s)
- Miguel A. Gonzalez-Lozano
- Center for Neurogenomics and Cognitive Research, Department of Molecular and Cellular Neurobiology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, 1081 Amsterdam, The Netherlands; (R.J.v.d.L.); (A.B.S.)
- Correspondence: (M.A.G.-L.); (K.W.L.)
| | - Joke Wortel
- Center for Neurogenomics and Cognitive Research, Department of Functional Genomics, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, 1081 Amsterdam, The Netherlands; (J.W.); (J.R.T.v.W.)
| | - Rolinka J. van der Loo
- Center for Neurogenomics and Cognitive Research, Department of Molecular and Cellular Neurobiology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, 1081 Amsterdam, The Netherlands; (R.J.v.d.L.); (A.B.S.)
| | - Jan R. T. van Weering
- Center for Neurogenomics and Cognitive Research, Department of Functional Genomics, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, 1081 Amsterdam, The Netherlands; (J.W.); (J.R.T.v.W.)
- Center for Neurogenomics and Cognitive Research, Department of Clinical Genetics, Amsterdam Neuroscience, Amsterdam UMC location VUmc, 1081 Amsterdam, The Netherlands
| | - August B. Smit
- Center for Neurogenomics and Cognitive Research, Department of Molecular and Cellular Neurobiology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, 1081 Amsterdam, The Netherlands; (R.J.v.d.L.); (A.B.S.)
| | - Ka Wan Li
- Center for Neurogenomics and Cognitive Research, Department of Molecular and Cellular Neurobiology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, 1081 Amsterdam, The Netherlands; (R.J.v.d.L.); (A.B.S.)
- Correspondence: (M.A.G.-L.); (K.W.L.)
| |
Collapse
|
56
|
Lee J, Avramets D, Jeon B, Choo H. Modulation of Serotonin Receptors in Neurodevelopmental Disorders: Focus on 5-HT7 Receptor. Molecules 2021; 26:molecules26113348. [PMID: 34199418 PMCID: PMC8199608 DOI: 10.3390/molecules26113348] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 05/31/2021] [Accepted: 05/31/2021] [Indexed: 12/17/2022] Open
Abstract
Since neurodevelopmental disorders (NDDs) influence more than 3% of children worldwide, there has been intense investigation to understand the etiology of disorders and develop treatments. Although there are drugs such as aripiprazole, risperidone, and lurasidone, these medications are not cures for the disorders and can only help people feel better or alleviate their symptoms. Thus, it is required to discover therapeutic targets in order to find the ultimate treatments of neurodevelopmental disorders. It is suggested that abnormal neuronal morphology in the neurodevelopment process is a main cause of NDDs, in which the serotonergic system is emerging as playing a crucial role. From this point of view, we noticed the correlation between serotonin receptor subtype 7 (5-HT7R) and NDDs including autism spectrum disorder (ASD), fragile X syndrome (FXS), and Rett syndrome (RTT). 5-HT7R modulators improved altered behaviors in animal models and also affected neuronal morphology via the 5-HT7R/G12 signaling pathway. Through the investigation of recent studies, it is suggested that 5-HT7R could be a potential therapeutic target for the treatment of NDDs.
Collapse
Affiliation(s)
- Jieon Lee
- Brain Science Institute, Korea Institute of Science and Technology, Seongbuk-gu, Seoul 02792, Korea; (J.L.); (D.A.)
- Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology, Seoul 02792, Korea
| | - Diana Avramets
- Brain Science Institute, Korea Institute of Science and Technology, Seongbuk-gu, Seoul 02792, Korea; (J.L.); (D.A.)
- Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology, Seoul 02792, Korea
| | - Byungsun Jeon
- Brain Science Institute, Korea Institute of Science and Technology, Seongbuk-gu, Seoul 02792, Korea; (J.L.); (D.A.)
- Correspondence: (B.J.); (H.C.); Tel.: +82-2-958-5191 (B.J.); +82-2-958-5157 (H.C.)
| | - Hyunah Choo
- Brain Science Institute, Korea Institute of Science and Technology, Seongbuk-gu, Seoul 02792, Korea; (J.L.); (D.A.)
- Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology, Seoul 02792, Korea
- Correspondence: (B.J.); (H.C.); Tel.: +82-2-958-5191 (B.J.); +82-2-958-5157 (H.C.)
| |
Collapse
|
57
|
Dionne O, Corbin F. An "Omic" Overview of Fragile X Syndrome. BIOLOGY 2021; 10:433. [PMID: 34068266 PMCID: PMC8153138 DOI: 10.3390/biology10050433] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 05/01/2021] [Accepted: 05/08/2021] [Indexed: 01/16/2023]
Abstract
Fragile X syndrome (FXS) is a neurodevelopmental disorder associated with a wide range of cognitive, behavioral and medical problems. It arises from the silencing of the fragile X mental retardation 1 (FMR1) gene and, consequently, in the absence of its encoded protein, FMRP (fragile X mental retardation protein). FMRP is a ubiquitously expressed and multifunctional RNA-binding protein, primarily considered as a translational regulator. Pre-clinical studies of the past two decades have therefore focused on this function to relate FMRP's absence to the molecular mechanisms underlying FXS physiopathology. Based on these data, successful pharmacological strategies were developed to rescue fragile X phenotype in animal models. Unfortunately, these results did not translate into humans as clinical trials using same therapeutic approaches did not reach the expected outcomes. These failures highlight the need to put into perspective the different functions of FMRP in order to get a more comprehensive understanding of FXS pathophysiology. This work presents a review of FMRP's involvement on noteworthy molecular mechanisms that may ultimately contribute to various biochemical alterations composing the fragile X phenotype.
Collapse
Affiliation(s)
- Olivier Dionne
- Department of Biochemistry and Functional Genomics, Faculty of Medicine and Health Sciences, Université de Sherbrooke and Centre de Recherche du CHUS, CIUSSS de l’Estrie-CHUS, Sherbrooke, QC J1H 5H4, Canada;
| | | |
Collapse
|
58
|
Dionne O, Lortie A, Gagnon F, Corbin F. Rates of protein synthesis are reduced in peripheral blood mononuclear cells (PBMCs) from fragile X individuals. PLoS One 2021; 16:e0251367. [PMID: 33974659 PMCID: PMC8112704 DOI: 10.1371/journal.pone.0251367] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Accepted: 04/23/2021] [Indexed: 12/30/2022] Open
Abstract
Background Fragile X syndrome (FXS) is the leading inherited cause of intellectual disability and is caused by the loss of expression of the Fragile X mental retardation protein (FMRP). In animal model of FXS, the absence of FMRP leads to an aberrant rate of neuronal protein synthesis, which in turn is believed to be at the origin of defects regarding spine morphology and synaptic plasticity. Normalisation of protein synthesis in these models has been associated with a rescue of FXS behavioral and biochemicals phenotype, thus establishing the rate of protein synthesis as one of the most promising monitoring biomarker for FXS. However, rate of protein synthesis alteration in fragile X individuals is not well characterized. Method We applied a robust radiolabeled assay to measure rate of protein synthesis in freshly extracted peripheral blood mononuclear cells (PBMCs) and blood platelets. We ultimately settle on PBMCs to measure and compare rate of protein synthesis in 13 males with fragile X and 14 matched controls individuals. Results Using this method, we measured a 26.9% decrease (p = 0,0193) in the rate of protein synthesis in fragile X individuals PBMCs. Furthermore, the rate of protein synthesis measurements obtained were highly reproducible, highlighting the robustness of the method. Conclusion Our work presents the first evidence of a diminution of the rate of protein synthesis in a human peripheral model of fragile X. Our results also support the finding of previous studies using brain PET imaging in Fragile X individuals. Since our assay only requires a simple venous puncture, it could be used in other cases of intellectual disability in order to determine if an aberrant rate of protein synthesis is a common general mechanism leading to impairment in synaptic plasticity and to intellectual disability.
Collapse
Affiliation(s)
- Olivier Dionne
- Department of Biochemistry and Functional Genomic, Faculty of Medicine and Health Sciences, Université de Sherbrooke and Centre de Recherche du CHUS, CIUSSS de l’Estrie-CHUS, Sherbrooke, Quebec, Canada
- * E-mail: (OD); (FC)
| | - Audrey Lortie
- Department of Biochemistry and Functional Genomic, Faculty of Medicine and Health Sciences, Université de Sherbrooke and Centre de Recherche du CHUS, CIUSSS de l’Estrie-CHUS, Sherbrooke, Quebec, Canada
| | - Florence Gagnon
- Department of Biochemistry and Functional Genomic, Faculty of Medicine and Health Sciences, Université de Sherbrooke and Centre de Recherche du CHUS, CIUSSS de l’Estrie-CHUS, Sherbrooke, Quebec, Canada
| | - François Corbin
- Department of Biochemistry and Functional Genomic, Faculty of Medicine and Health Sciences, Université de Sherbrooke and Centre de Recherche du CHUS, CIUSSS de l’Estrie-CHUS, Sherbrooke, Quebec, Canada
- * E-mail: (OD); (FC)
| |
Collapse
|
59
|
Khayachi A, Schorova L, Alda M, Rouleau GA, Milnerwood AJ. Posttranslational modifications & lithium's therapeutic effect-Potential biomarkers for clinical responses in psychiatric & neurodegenerative disorders. Neurosci Biobehav Rev 2021; 127:424-445. [PMID: 33971223 DOI: 10.1016/j.neubiorev.2021.05.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 03/14/2021] [Accepted: 05/03/2021] [Indexed: 01/03/2023]
Abstract
Several neurodegenerative diseases and neuropsychiatric disorders display aberrant posttranslational modifications (PTMs) of one, or many, proteins. Lithium treatment has been used for mood stabilization for many decades, and is highly effective for large subsets of patients with diverse neurological conditions. However, the differential effectiveness and mode of action are not fully understood. In recent years, studies have shown that lithium alters several protein PTMs, altering their function, and consequently neuronal physiology. The impetus for this review is to outline the links between lithium's therapeutic mode of action and PTM homeostasis. We first provide an overview of the principal PTMs affected by lithium. We then describe several neuropsychiatric disorders in which PTMs have been implicated as pathogenic. For each of these conditions, we discuss lithium's clinical use and explore the putative mechanism of how it restores PTM homeostasis, and thereby cellular physiology. Evidence suggests that determining specific PTM patterns could be a promising strategy to develop biomarkers for disease and lithium responsiveness.
Collapse
Affiliation(s)
- A Khayachi
- Montreal Neurological Institute, Department of Neurology & Neurosurgery, McGill University, Montréal, Quebec, Canada.
| | - L Schorova
- McGill University Health Center Research Institute, Montréal, Quebec, Canada
| | - M Alda
- Department of Psychiatry, Dalhousie University, Halifax, Nova Scotia, Canada
| | - G A Rouleau
- Montreal Neurological Institute, Department of Neurology & Neurosurgery, McGill University, Montréal, Quebec, Canada; Department of Human Genetics, McGill University, Montréal, Quebec, Canada.
| | - A J Milnerwood
- Montreal Neurological Institute, Department of Neurology & Neurosurgery, McGill University, Montréal, Quebec, Canada.
| |
Collapse
|
60
|
Bülow P, Zlatic SA, Wenner PA, Bassell GJ, Faundez V. FMRP attenuates activity dependent modifications in the mitochondrial proteome. Mol Brain 2021; 14:75. [PMID: 33931071 PMCID: PMC8086361 DOI: 10.1186/s13041-021-00783-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Accepted: 04/21/2021] [Indexed: 12/21/2022] Open
Abstract
Homeostatic plasticity is necessary for the construction and maintenance of functional neuronal networks, but principal molecular mechanisms required for or modified by homeostatic plasticity are not well understood. We recently reported that homeostatic plasticity induced by activity deprivation is dysregulated in cortical neurons from Fragile X Mental Retardation protein (FMRP) knockout mice (Bulow et al. in Cell Rep 26: 1378-1388 e1373, 2019). These findings led us to hypothesize that identifying proteins sensitive to activity deprivation and/or FMRP expression could reveal pathways required for or modified by homeostatic plasticity. Here, we report an unbiased quantitative mass spectrometry used to quantify steady-state proteome changes following chronic activity deprivation in wild type and Fmr1-/y cortical neurons. Proteome hits responsive to both activity deprivation and the Fmr1-/y genotype were significantly annotated to mitochondria. We found an increased number of mitochondria annotated proteins whose expression was sensitive to activity deprivation in Fmr1-/y cortical neurons as compared to wild type neurons. These findings support a novel role of FMRP in attenuating mitochondrial proteome modifications induced by activity deprivation.
Collapse
Affiliation(s)
- Pernille Bülow
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Stephanie A Zlatic
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Peter A Wenner
- Department of Physiology, Emory University School of Medicine, Atlanta, GA, 30322, USA.
| | - Gary J Bassell
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA, 30322, USA.
| | - Victor Faundez
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA, 30322, USA.
| |
Collapse
|
61
|
Perrone-Capano C, Volpicelli F, Penna E, Chun JT, Crispino M. Presynaptic protein synthesis and brain plasticity: From physiology to neuropathology. Prog Neurobiol 2021; 202:102051. [PMID: 33845165 DOI: 10.1016/j.pneurobio.2021.102051] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 03/14/2021] [Accepted: 04/07/2021] [Indexed: 12/12/2022]
Abstract
To form and maintain extremely intricate and functional neural circuitry, mammalian neurons are typically endowed with highly arborized dendrites and a long axon. The synapses that link neurons to neurons or to other cells are numerous and often too remote for the cell body to make and deliver new proteins to the right place in time. Moreover, synapses undergo continuous activity-dependent changes in their number and strength, establishing the basis of neural plasticity. The innate dilemma is then how a highly complex neuron provides new proteins for its cytoplasmic periphery and individual synapses to support synaptic plasticity. Here, we review a growing body of evidence that local protein synthesis in discrete sites of the axon and presynaptic terminals plays crucial roles in synaptic plasticity, and that deregulation of this local translation system is implicated in various pathologies of the nervous system.
Collapse
Affiliation(s)
- Carla Perrone-Capano
- Department of Pharmacy, University of Naples Federico II, Naples, Italy; Institute of Genetics and Biophysics "Adriano Buzzati Traverso", CNR, Naples, Italy.
| | | | - Eduardo Penna
- Department of Biology, University of Naples Federico II, Naples, Italy.
| | - Jong Tai Chun
- Department of Biology and Evolution of Marine Organisms, Stazione Zoologica Anton Dohrn, Naples, Italy.
| | - Marianna Crispino
- Department of Biology, University of Naples Federico II, Naples, Italy.
| |
Collapse
|
62
|
Cell-type-specific profiling of human cellular models of fragile X syndrome reveal PI3K-dependent defects in translation and neurogenesis. Cell Rep 2021; 35:108991. [PMID: 33852833 PMCID: PMC8133829 DOI: 10.1016/j.celrep.2021.108991] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 03/08/2021] [Accepted: 03/23/2021] [Indexed: 02/06/2023] Open
Abstract
Transcriptional silencing of the FMR1 gene in fragile X syndrome (FXS) leads to the loss of the RNA-binding protein FMRP. In addition to regulating mRNA translation and protein synthesis, emerging evidence suggests that FMRP acts to coordinate proliferation and differentiation during early neural development. However, whether loss of FMRP-mediated translational control is related to impaired cell fate specification in the developing human brain remains unknown. Here, we use human patient induced pluripotent stem cell (iPSC)-derived neural progenitor cells and organoids to model neurogenesis in FXS. We developed a high-throughput, in vitro assay that allows for the simultaneous quantification of protein synthesis and proliferation within defined neural subpopulations. We demonstrate that abnormal protein synthesis in FXS is coupled to altered cellular decisions to favor proliferative over neurogenic cell fates during early development. Furthermore, pharmacologic inhibition of elevated phosphoinositide 3-kinase (PI3K) signaling corrects both excess protein synthesis and cell proliferation in a subset of patient neural cells. Raj et al. developed a multiparametric assay to measure cellular and molecular phenotypes during neurogenesis in fragile X syndrome iPSC-derived neural cells. Relative to controls, FXS patient cultures have more proliferative cells with increased protein synthesis. Defects in cell fate acquisition can be normalized by inhibiting overactive PI3K signaling.
Collapse
|
63
|
Holter MC, Hewitt LT, Nishimura KJ, Knowles SJ, Bjorklund GR, Shah S, Fry NR, Rees KP, Gupta TA, Daniels CW, Li G, Marsh S, Treiman DM, Olive MF, Anderson TR, Sanabria F, Snider WD, Newbern JM. Hyperactive MEK1 Signaling in Cortical GABAergic Neurons Promotes Embryonic Parvalbumin Neuron Loss and Defects in Behavioral Inhibition. Cereb Cortex 2021; 31:3064-3081. [PMID: 33570093 DOI: 10.1093/cercor/bhaa413] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 12/22/2020] [Accepted: 12/23/2020] [Indexed: 12/20/2022] Open
Abstract
Many developmental syndromes have been linked to genetic mutations that cause abnormal ERK/MAPK activity; however, the neuropathological effects of hyperactive signaling are not fully understood. Here, we examined whether hyperactivation of MEK1 modifies the development of GABAergic cortical interneurons (CINs), a heterogeneous population of inhibitory neurons necessary for cortical function. We show that GABAergic-neuron specific MEK1 hyperactivation in vivo leads to increased cleaved caspase-3 labeling in a subpopulation of immature neurons in the embryonic subpallial mantle zone. Adult mutants displayed a significant loss of parvalbumin (PV), but not somatostatin, expressing CINs and a reduction in perisomatic inhibitory synapses on excitatory neurons. Surviving mutant PV-CINs maintained a typical fast-spiking phenotype but showed signs of decreased intrinsic excitability that coincided with an increased risk of seizure-like phenotypes. In contrast to other mouse models of PV-CIN loss, we discovered a robust increase in the accumulation of perineuronal nets, an extracellular structure thought to restrict plasticity. Indeed, we found that mutants exhibited a significant impairment in the acquisition of behavioral response inhibition capacity. Overall, our data suggest PV-CIN development is particularly sensitive to hyperactive MEK1 signaling, which may underlie certain neurological deficits frequently observed in ERK/MAPK-linked syndromes.
Collapse
Affiliation(s)
- Michael C Holter
- School of Life Sciences, Arizona State University, Tempe, AZ 85287, USA
| | - Lauren T Hewitt
- School of Life Sciences, Arizona State University, Tempe, AZ 85287, USA.,Interdepartmental Neuroscience Graduate Program, University of Texas, Austin, TX 78712, USA
| | - Kenji J Nishimura
- School of Life Sciences, Arizona State University, Tempe, AZ 85287, USA.,Interdepartmental Neuroscience Graduate Program, University of Texas, Austin, TX 78712, USA
| | - Sara J Knowles
- School of Life Sciences, Arizona State University, Tempe, AZ 85287, USA
| | | | - Shiv Shah
- School of Life Sciences, Arizona State University, Tempe, AZ 85287, USA
| | - Noah R Fry
- School of Life Sciences, Arizona State University, Tempe, AZ 85287, USA
| | - Katherina P Rees
- School of Life Sciences, Arizona State University, Tempe, AZ 85287, USA
| | - Tanya A Gupta
- Department of Psychology, Arizona State University, Tempe, AZ 85287, USA
| | - Carter W Daniels
- Department of Psychology, Arizona State University, Tempe, AZ 85287, USA.,Department of Psychiatry, Columbia University, New York, NY 10032, USA
| | - Guohui Li
- College of Medicine, University of Arizona, Phoenix, AZ 85004, USA
| | - Steven Marsh
- Barrow Neurological Institute, Phoenix, AZ 85013, USA
| | | | | | - Trent R Anderson
- College of Medicine, University of Arizona, Phoenix, AZ 85004, USA
| | - Federico Sanabria
- Department of Psychology, Arizona State University, Tempe, AZ 85287, USA
| | - William D Snider
- University of North Carolina Neuroscience Center, The University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Jason M Newbern
- School of Life Sciences, Arizona State University, Tempe, AZ 85287, USA
| |
Collapse
|
64
|
Sahasrabudhe A, Begum F, Guevara CA, Morrison C, Hsiao K, Kezunovic N, Bozdagi-Gunal O, Benson DL. Cyfip1 Regulates SynGAP1 at Hippocampal Synapses. Front Synaptic Neurosci 2021; 12:581714. [PMID: 33613257 PMCID: PMC7892963 DOI: 10.3389/fnsyn.2020.581714] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Accepted: 12/23/2020] [Indexed: 11/13/2022] Open
Abstract
In humans, copy number variations in CYFIP1 appear to have sweeping physiological and structural consequences in the brain, either producing or altering the severity of intellectual disability, autism, and schizophrenia. Independently, SynGAP1 haploinsufficiency produces intellectual disability and, frequently, autism. Cyfip1 inhibits protein translation and promotes actin polymerization, and SynGAP1 is a synaptically localized Ras/Rap GAP. While these proteins are clearly distinct, studies investigating their functions in mice have shown that each regulates the maturation of synapses in the hippocampus and haploinsufficiency for either produces an exaggerated form of mGluR-dependent long-term depression, suggesting that some signaling pathways converge. In this study, we examined how Cyfip1 haploinsufficiency impacts SynGAP1 levels and localization, as well as potential sites for mechanistic interaction in mouse hippocampus. The data show that synaptic, but not total, levels of SynGAP1 in Cyfip1 +/- mice were abnormally low during early postnatal development and in adults. This may be in response to a shift in the balance of kinases that activate SynGAP1 as levels of Cdk5 were reduced and those of activated CaMKII were maintained in Cyfip1 +/- mice compared to wild-type mice. Alternatively, this could reflect altered actin dynamics as Rac1 activity in Cyfip1 +/- hippocampus was boosted significantly compared to wild-type mice, and levels of synaptic F-actin were generally enhanced due in part to an increase in the activity of the WAVE regulatory complex. Decreased synaptic SynGAP1 coupled with a CaMKII-mediated bias toward Rap1 inactivation at synapses is also consistent with increased levels of synaptic GluA2, increased AMPA receptor-mediated responses to stimulation, and increased levels of synaptic mGluR1/5 compared to wild-type mice. Collectively, our data suggest that Cyfip1 regulates SynGAP1 and the two proteins work coordinately at synapses to appropriately direct actin polymerization and GAP activity.
Collapse
Affiliation(s)
- Abhishek Sahasrabudhe
- Icahn School of Medicine at Mount Sinai, Nash Family Department of Neuroscience, Friedman Brain Institute, New York, NY, United States
| | - Fatema Begum
- Icahn School of Medicine at Mount Sinai, Nash Family Department of Neuroscience, Friedman Brain Institute, New York, NY, United States
| | - Christopher A Guevara
- Icahn School of Medicine at Mount Sinai, Nash Family Department of Neuroscience, Friedman Brain Institute, New York, NY, United States.,Graduate School of Biomedical Sciences, New York, NY, United States
| | - Chenel Morrison
- Icahn School of Medicine at Mount Sinai, Nash Family Department of Neuroscience, Friedman Brain Institute, New York, NY, United States
| | - Kuangfu Hsiao
- Icahn School of Medicine at Mount Sinai, Nash Family Department of Neuroscience, Friedman Brain Institute, New York, NY, United States
| | - Nebojsa Kezunovic
- Icahn School of Medicine at Mount Sinai, Nash Family Department of Neuroscience, Friedman Brain Institute, New York, NY, United States
| | - Ozlem Bozdagi-Gunal
- Department of Psychiatry, Rutgers New Jersey Medical School, Newark, NJ, United States
| | - Deanna L Benson
- Icahn School of Medicine at Mount Sinai, Nash Family Department of Neuroscience, Friedman Brain Institute, New York, NY, United States
| |
Collapse
|
65
|
Adenosine A 2A receptor inhibition reduces synaptic and cognitive hippocampal alterations in Fmr1 KO mice. Transl Psychiatry 2021; 11:112. [PMID: 33547274 PMCID: PMC7864914 DOI: 10.1038/s41398-021-01238-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 12/28/2020] [Accepted: 01/18/2021] [Indexed: 02/07/2023] Open
Abstract
In fragile X syndrome (FXS) the lack of the fragile X mental retardation protein (FMRP) leads to exacerbated signaling through the metabotropic glutamate receptors 5 (mGlu5Rs). The adenosine A2A receptors (A2ARs), modulators of neuronal damage, could play a role in FXS. A synaptic colocalization and a strong permissive interaction between A2A and mGlu5 receptors in the hippocampus have been previously reported, suggesting that blocking A2ARs might normalize the mGlu5R-mediated effects of FXS. To study the cross-talk between A2A and mGlu5 receptors in the absence of FMRP, we performed extracellular electrophysiology experiments in hippocampal slices of Fmr1 KO mouse. The depression of field excitatory postsynaptic potential (fEPSPs) slope induced by the mGlu5R agonist CHPG was completely blocked by the A2AR antagonist ZM241385 and strongly potentiated by the A2AR agonist CGS21680, suggesting that the functional synergistic coupling between the two receptors could be increased in FXS. To verify if chronic A2AR blockade could reverse the FXS phenotypes, we treated the Fmr1 KO mice with istradefylline, an A2AR antagonist. We found that hippocampal DHPG-induced long-term depression (LTD), which is abnormally increased in FXS mice, was restored to the WT level. Furthermore, istradefylline corrected aberrant dendritic spine density, specific behavioral alterations, and overactive mTOR, TrkB, and STEP signaling in Fmr1 KO mice. Finally, we identified A2AR mRNA as a target of FMRP. Our results show that the pharmacological blockade of A2ARs partially restores some of the phenotypes of Fmr1 KO mice, both by reducing mGlu5R functioning and by acting on other A2AR-related downstream targets.
Collapse
|
66
|
Dissociation of functional and structural plasticity of dendritic spines during NMDAR and mGluR-dependent long-term synaptic depression in wild-type and fragile X model mice. Mol Psychiatry 2021; 26:4652-4669. [PMID: 32606374 PMCID: PMC8095717 DOI: 10.1038/s41380-020-0821-6] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2019] [Revised: 06/03/2020] [Accepted: 06/12/2020] [Indexed: 12/14/2022]
Abstract
Many neurodevelopmental disorders are characterized by impaired functional synaptic plasticity and abnormal dendritic spine morphology, but little is known about how these are related. Previous work in the Fmr1-/y mouse model of fragile X (FX) suggests that increased constitutive dendritic protein synthesis yields exaggerated mGluR5-dependent long-term synaptic depression (LTD) in area CA1 of the hippocampus, but an effect on spine structural plasticity remains to be determined. In the current study, we used simultaneous electrophysiology and time-lapse two photon imaging to examine how spines change their structure during LTD induced by activation of mGluRs or NMDA receptors (NMDARs), and how this plasticity is altered in Fmr1-/y mice. We were surprised to find that mGluR activation causes LTD and AMPA receptor internalization, but no spine shrinkage in either wildtype or Fmr1-/y mice. In contrast, NMDAR activation caused spine shrinkage as well as LTD in both genotypes. Spine shrinkage was initiated by non-ionotropic (metabotropic) signaling through NMDARs, and in wild-type mice this structural plasticity required activation of mTORC1 and new protein synthesis. In striking contrast, NMDA-induced spine plasticity in Fmr1-/y mice was no longer dependent on acute activation of mTORC1 or de novo protein synthesis. These findings reveal that the structural consequences of mGluR and metabotropic NMDAR activation differ, and that a brake on spine structural plasticity, normally provided by mTORC1 regulation of protein synthesis, is absent in FX. Increased constitutive protein synthesis in FX appears to modify functional and structural plasticity induced through different glutamate receptors.
Collapse
|
67
|
Shukla T, de la Peña JB, Perish JM, Ploski JE, Stumpf CR, Webster KR, Thorn CA, Campbell ZT. A Highly Selective MNK Inhibitor Rescues Deficits Associated with Fragile X Syndrome in Mice. Neurotherapeutics 2021; 18:624-639. [PMID: 33006091 PMCID: PMC8116363 DOI: 10.1007/s13311-020-00932-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/12/2020] [Indexed: 12/22/2022] Open
Abstract
Fragile X syndrome (FXS) is the most common inherited source of intellectual disability in humans. FXS is caused by mutations that trigger epigenetic silencing of the Fmr1 gene. Loss of Fmr1 results in increased activity of the mitogen-activated protein kinase (MAPK) pathway. An important downstream consequence is activation of the mitogen-activated protein kinase interacting protein kinase (MNK). MNK phosphorylates the mRNA cap-binding protein, eukaryotic initiation factor 4E (eIF4E). Excessive phosphorylation of eIF4E has been directly implicated in the cognitive and behavioral deficits associated with FXS. Pharmacological reduction of eIF4E phosphorylation is one potential strategy for FXS treatment. We demonstrate that systemic dosing of a highly specific, orally available MNK inhibitor, eFT508, attenuates numerous deficits associated with loss of Fmr1 in mice. eFT508 resolves a range of phenotypic abnormalities associated with FXS including macroorchidism, aberrant spinogenesis, and alterations in synaptic plasticity. Key behavioral deficits related to anxiety, social interaction, obsessive and repetitive activities, and object recognition are ameliorated by eFT508. Collectively, this work establishes eFT508 as a potential means to reverse deficits associated with FXS.
Collapse
Affiliation(s)
- Tarjani Shukla
- Department of Biological Sciences, University of Texas at Dallas, 800 W. Campbell Road, Richardson, TX, 75080, USA
- Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX, 75080, USA
| | - June Bryan de la Peña
- Department of Biological Sciences, University of Texas at Dallas, 800 W. Campbell Road, Richardson, TX, 75080, USA
- Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX, 75080, USA
| | - John M Perish
- School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX, 75080, USA
| | - Jonathan E Ploski
- School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX, 75080, USA
| | | | | | - Catherine A Thorn
- School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX, 75080, USA
| | - Zachary T Campbell
- Department of Biological Sciences, University of Texas at Dallas, 800 W. Campbell Road, Richardson, TX, 75080, USA.
- Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX, 75080, USA.
| |
Collapse
|
68
|
Stoppel DC, McCamphill PK, Senter RK, Heynen AJ, Bear MF. mGluR5 Negative Modulators for Fragile X: Treatment Resistance and Persistence. Front Psychiatry 2021; 12:718953. [PMID: 34658956 PMCID: PMC8511445 DOI: 10.3389/fpsyt.2021.718953] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 09/08/2021] [Indexed: 11/13/2022] Open
Abstract
Fragile X syndrome (FXS) is caused by silencing of the human FMR1 gene and is the leading monogenic cause of intellectual disability and autism. Abundant preclinical data indicated that negative allosteric modulators (NAMs) of metabotropic glutamate receptor 5 (mGluR5) might be efficacious in treating FXS in humans. Initial attempts to translate these findings in clinical trials have failed, but these failures provide the opportunity for new discoveries that will improve future trials. The emergence of acquired treatment resistance ("tolerance") after chronic administration of mGluR5 NAMs is a potential factor in the lack of success. Here we confirm that FXS model mice display acquired treatment resistance after chronic treatment with the mGluR5 NAM CTEP in three assays commonly examined in the mouse model of FXS: (1) audiogenic seizure susceptibility, (2) sensory cortex hyperexcitability, and (3) hippocampal protein synthesis. Cross-tolerance experiments suggest that the mechanism of treatment resistance likely occurs at signaling nodes downstream of glycogen synthase kinase 3α (GSK3α), but upstream of protein synthesis. The rapid emergence of tolerance to CTEP begs the question of how previous studies showed an improvement in inhibitory avoidance (IA) cognitive performance after chronic treatment. We show here that this observation was likely explained by timely inhibition of mGluR5 during a critical period, as brief CTEP treatment in juvenile mice is sufficient to provide a persistent improvement of IA behavior measured many weeks later. These data will be important to consider when designing future fragile X clinical trials using compounds that target the mGluR5-to-protein synthesis signaling cascade.
Collapse
Affiliation(s)
- David C Stoppel
- Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Patrick K McCamphill
- Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Rebecca K Senter
- Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Arnold J Heynen
- Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Mark F Bear
- Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, United States
| |
Collapse
|
69
|
Clifton NE, Thomas KL, Wilkinson LS, Hall J, Trent S. FMRP and CYFIP1 at the Synapse and Their Role in Psychiatric Vulnerability. Complex Psychiatry 2020; 6:5-19. [PMID: 34883502 PMCID: PMC7673588 DOI: 10.1159/000506858] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Accepted: 02/27/2020] [Indexed: 12/23/2022] Open
Abstract
There is increasing awareness of the role genetic risk variants have in mediating vulnerability to psychiatric disorders such as schizophrenia and autism. Many of these risk variants encode synaptic proteins, influencing biological pathways of the postsynaptic density and, ultimately, synaptic plasticity. Fragile-X mental retardation 1 (FMR1) and cytoplasmic fragile-X mental retardation protein (FMRP)-interacting protein 1 (CYFIP1) contain 2 such examples of highly penetrant risk variants and encode synaptic proteins with shared functional significance. In this review, we discuss the biological actions of FMRP and CYFIP1, including their regulation of (i) protein synthesis and specifically FMRP targets, (ii) dendritic and spine morphology, and (iii) forms of synaptic plasticity such as long-term depression. We draw upon a range of preclinical studies that have used genetic dosage models of FMR1 and CYFIP1 to determine their biological function. In parallel, we discuss how clinical studies of fragile X syndrome or 15q11.2 deletion patients have informed our understanding of FMRP and CYFIP1, and highlight the latest psychiatric genomic findings that continue to implicate FMRP and CYFIP1. Lastly, we assess the current limitations in our understanding of FMRP and CYFIP1 biology and how they must be addressed before mechanism-led therapeutic strategies can be developed for psychiatric disorders.
Collapse
Affiliation(s)
- Nicholas E. Clifton
- Neuroscience & Mental Health Research Institute, Cardiff University, Cardiff, United Kingdom
- MRC Centre for Neuropsychiatric Genetics and Genomics, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Kerrie L. Thomas
- Neuroscience & Mental Health Research Institute, Cardiff University, Cardiff, United Kingdom
- School of Biosciences, Cardiff University, Cardiff, United Kingdom
| | - Lawrence S. Wilkinson
- Neuroscience & Mental Health Research Institute, Cardiff University, Cardiff, United Kingdom
- School of Psychology, Cardiff University, Cardiff, United Kingdom
| | - Jeremy Hall
- Neuroscience & Mental Health Research Institute, Cardiff University, Cardiff, United Kingdom
- MRC Centre for Neuropsychiatric Genetics and Genomics, School of Medicine, Cardiff University, Cardiff, United Kingdom
- Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Simon Trent
- Neuroscience & Mental Health Research Institute, Cardiff University, Cardiff, United Kingdom
- School of Life Sciences, Faculty of Natural Sciences, Keele University, Keele, United Kingdom
| |
Collapse
|
70
|
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
|
71
|
Booker SA, Simões de Oliveira L, Anstey NJ, Kozic Z, Dando OR, Jackson AD, Baxter PS, Isom LL, Sherman DL, Hardingham GE, Brophy PJ, Wyllie DJ, Kind PC. Input-Output Relationship of CA1 Pyramidal Neurons Reveals Intact Homeostatic Mechanisms in a Mouse Model of Fragile X Syndrome. Cell Rep 2020; 32:107988. [PMID: 32783927 PMCID: PMC7435362 DOI: 10.1016/j.celrep.2020.107988] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 04/01/2020] [Accepted: 07/14/2020] [Indexed: 12/13/2022] Open
Abstract
Cellular hyperexcitability is a salient feature of fragile X syndrome animal models. The cellular basis of hyperexcitability and how it responds to changing activity states is not fully understood. Here, we show increased axon initial segment length in CA1 of the Fmr1-/y mouse hippocampus, with increased cellular excitability. This change in length does not result from reduced AIS plasticity, as prolonged depolarization induces changes in AIS length independent of genotype. However, depolarization does reduce cellular excitability, the magnitude of which is greater in Fmr1-/y neurons. Finally, we observe reduced functional inputs from the entorhinal cortex, with no genotypic difference in the firing rates of CA1 pyramidal neurons. This suggests that AIS-dependent hyperexcitability in Fmr1-/y mice may result from adaptive or homeostatic regulation induced by reduced functional synaptic connectivity. Thus, while AIS length and intrinsic excitability contribute to cellular hyperexcitability, they may reflect a homeostatic mechanism for reduced synaptic input onto CA1 neurons.
Collapse
Affiliation(s)
- Sam A. Booker
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK,Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh, UK,Patrick Wild Centre for Autism Research, University of Edinburgh, Edinburgh, UK,Corresponding author
| | - Laura Simões de Oliveira
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK,Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh, UK,Patrick Wild Centre for Autism Research, University of Edinburgh, Edinburgh, UK
| | - Natasha J. Anstey
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK,Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh, UK,Patrick Wild Centre for Autism Research, University of Edinburgh, Edinburgh, UK,Centre for Brain Development and Repair, InStem, GKVK Campus, Bangalore 560065, India
| | - Zrinko Kozic
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK,Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh, UK
| | - Owen R. Dando
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK,Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh, UK,Patrick Wild Centre for Autism Research, University of Edinburgh, Edinburgh, UK,Dementia Research Institute, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh EH8 9XD, UK,Centre for Brain Development and Repair, InStem, GKVK Campus, Bangalore 560065, India
| | - Adam D. Jackson
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK,Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh, UK,Patrick Wild Centre for Autism Research, University of Edinburgh, Edinburgh, UK,Centre for Brain Development and Repair, InStem, GKVK Campus, Bangalore 560065, India
| | - Paul S. Baxter
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK,Dementia Research Institute, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh EH8 9XD, UK
| | - Lori L. Isom
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, MI 48109-5632, USA
| | - Diane L. Sherman
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Giles E. Hardingham
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK,Dementia Research Institute, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh EH8 9XD, UK
| | - Peter J. Brophy
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - David J.A. Wyllie
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK,Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh, UK,Patrick Wild Centre for Autism Research, University of Edinburgh, Edinburgh, UK,Centre for Brain Development and Repair, InStem, GKVK Campus, Bangalore 560065, India
| | - Peter C. Kind
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK,Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh, UK,Patrick Wild Centre for Autism Research, University of Edinburgh, Edinburgh, UK,Centre for Brain Development and Repair, InStem, GKVK Campus, Bangalore 560065, India,Corresponding author
| |
Collapse
|
72
|
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
|
73
|
A Differential Effect of Lovastatin versus Simvastatin in Neurodevelopmental Disorders. eNeuro 2020; 7:ENEURO.0162-20.2020. [PMID: 32651266 PMCID: PMC7433894 DOI: 10.1523/eneuro.0162-20.2020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 06/08/2020] [Accepted: 06/16/2020] [Indexed: 12/30/2022] Open
|
74
|
Bonnycastle K, Davenport EC, Cousin MA. Presynaptic dysfunction in neurodevelopmental disorders: Insights from the synaptic vesicle life cycle. J Neurochem 2020; 157:179-207. [PMID: 32378740 DOI: 10.1111/jnc.15035] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 04/14/2020] [Accepted: 04/22/2020] [Indexed: 12/11/2022]
Abstract
The activity-dependent fusion, retrieval and recycling of synaptic vesicles is essential for the maintenance of neurotransmission. Until relatively recently it was believed that most mutations in genes that were essential for this process would be incompatible with life, because of this fundamental role. However, an ever-expanding number of mutations in this very cohort of genes are being identified in individuals with neurodevelopmental disorders, including autism, intellectual disability and epilepsy. This article will summarize the current state of knowledge linking mutations in presynaptic genes to neurodevelopmental disorders by sequentially covering the various stages of the synaptic vesicle life cycle. It will also discuss how perturbations of specific stages within this recycling process could translate into human disease. Finally, it will also provide perspectives on the potential for future therapy that are targeted to presynaptic function.
Collapse
Affiliation(s)
- Katherine Bonnycastle
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK.,Muir Maxwell Epilepsy Centre, University of Edinburgh, Edinburgh, UK.,Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh, UK
| | - Elizabeth C Davenport
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK.,Muir Maxwell Epilepsy Centre, University of Edinburgh, Edinburgh, UK.,Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh, UK
| | - Michael A Cousin
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK.,Muir Maxwell Epilepsy Centre, University of Edinburgh, Edinburgh, UK.,Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
75
|
Asiminas A, Jackson AD, Louros SR, Till SM, Spano T, Dando O, Bear MF, Chattarji S, Hardingham GE, Osterweil EK, Wyllie DJA, Wood ER, Kind PC. Sustained correction of associative learning deficits after brief, early treatment in a rat model of Fragile X Syndrome. Sci Transl Med 2020; 11:11/494/eaao0498. [PMID: 31142675 DOI: 10.1126/scitranslmed.aao0498] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Revised: 10/19/2018] [Accepted: 05/09/2019] [Indexed: 12/15/2022]
Abstract
Fragile X Syndrome (FXS) is one of the most common monogenic forms of autism and intellectual disability. Preclinical studies in animal models have highlighted the potential of pharmaceutical intervention strategies for alleviating the symptoms of FXS. However, whether treatment strategies can be tailored to developmental time windows that define the emergence of particular phenotypes is unknown. Similarly, whether a brief, early intervention can have long-lasting beneficial effects, even after treatment cessation, is also unknown. To address these questions, we first examined the developmental profile for the acquisition of associative learning in a rat model of FXS. Associative memory was tested using a range of behavioral paradigms that rely on an animal's innate tendency to explore novelty. Fmr1 knockout (KO) rats showed a developmental delay in their acquisition of object-place recognition and did not demonstrate object-place-context recognition paradigm at any age tested (up to 23 weeks of age). Treatment of Fmr1 KO rats with lovastatin between 5 and 9 weeks of age, during the normal developmental period that this associative memory capability is established, prevents the emergence of deficits but has no effect in wild-type animals. Moreover, we observe no regression of cognitive performance in the FXS rats over several months after treatment. This restoration of the normal developmental trajectory of cognitive function is associated with the sustained rescue of both synaptic plasticity and altered protein synthesis. The findings provide proof of concept that the impaired emergence of the cognitive repertoire in neurodevelopmental disorders may be prevented by brief, early pharmacological intervention.
Collapse
Affiliation(s)
- Antonis Asiminas
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh EH8 9XD, UK.,Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh EH8 9XD, UK.,Patrick Wild Centre, University of Edinburgh, Edinburgh EH8 9XD, UK
| | - Adam D Jackson
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh EH8 9XD, UK.,Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh EH8 9XD, UK.,Patrick Wild Centre, University of Edinburgh, Edinburgh EH8 9XD, UK.,Centre for Brain Development and Repair, InStem, Bangalore 560065, India
| | - Susana R Louros
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh EH8 9XD, UK.,Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh EH8 9XD, UK.,Patrick Wild Centre, University of Edinburgh, Edinburgh EH8 9XD, UK
| | - Sally M Till
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh EH8 9XD, UK.,Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh EH8 9XD, UK.,Patrick Wild Centre, University of Edinburgh, Edinburgh EH8 9XD, UK
| | - Teresa Spano
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh EH8 9XD, UK.,Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh EH8 9XD, UK.,Patrick Wild Centre, University of Edinburgh, Edinburgh EH8 9XD, UK.,Centre for Brain Development and Repair, InStem, Bangalore 560065, India
| | - Owen Dando
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh EH8 9XD, UK.,Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh EH8 9XD, UK.,Patrick Wild Centre, University of Edinburgh, Edinburgh EH8 9XD, UK.,UK Dementia Research Institute at the Edinburgh Medical School, University of Edinburgh, Edinburgh EH8 9XD, UK
| | - Mark F Bear
- Department of Brain and Cognitive Sciences, Howard Hughes Medical Institute, Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Sumantra Chattarji
- Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh EH8 9XD, UK.,Patrick Wild Centre, University of Edinburgh, Edinburgh EH8 9XD, UK.,Centre for Brain Development and Repair, InStem, Bangalore 560065, India
| | - Giles E Hardingham
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh EH8 9XD, UK.,Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh EH8 9XD, UK.,Patrick Wild Centre, University of Edinburgh, Edinburgh EH8 9XD, UK.,UK Dementia Research Institute at the Edinburgh Medical School, University of Edinburgh, Edinburgh EH8 9XD, UK
| | - Emily K Osterweil
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh EH8 9XD, UK.,Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh EH8 9XD, UK.,Patrick Wild Centre, University of Edinburgh, Edinburgh EH8 9XD, UK
| | - David J A Wyllie
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh EH8 9XD, UK.,Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh EH8 9XD, UK.,Patrick Wild Centre, University of Edinburgh, Edinburgh EH8 9XD, UK.,Centre for Brain Development and Repair, InStem, Bangalore 560065, India
| | - Emma R Wood
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh EH8 9XD, UK. .,Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh EH8 9XD, UK.,Patrick Wild Centre, University of Edinburgh, Edinburgh EH8 9XD, UK.,Centre for Brain Development and Repair, InStem, Bangalore 560065, India
| | - Peter C Kind
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh EH8 9XD, UK. .,Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh EH8 9XD, UK.,Patrick Wild Centre, University of Edinburgh, Edinburgh EH8 9XD, UK.,Centre for Brain Development and Repair, InStem, Bangalore 560065, India
| |
Collapse
|
76
|
Schmidt KC, Loutaev I, Quezado Z, Sheeler C, Smith CB. Regional rates of brain protein synthesis are unaltered in dexmedetomidine sedated young men with fragile X syndrome: A L-[1- 11C]leucine PET study. Neurobiol Dis 2020; 143:104978. [PMID: 32569795 PMCID: PMC7425798 DOI: 10.1016/j.nbd.2020.104978] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 06/01/2020] [Accepted: 06/15/2020] [Indexed: 02/06/2023] Open
Abstract
Fragile X syndrome (FXS) is the most common inherited cause of intellectual disability. Fragile X mental retardation protein (FMRP), a putative translation suppressor, is absent or significantly reduced in FXS. One prevailing hypothesis is that rates of protein synthesis are increased by the absence of this regulatory protein. In accord with this hypothesis, we have previously reported increased rates of cerebral protein synthesis (rCPS) in the Fmr1 knockout mouse model of FXS and others have reported similar effects in hippocampal slices. To address the hypothesis in human subjects, we applied the L[1-11C]leucine PET method to measure rCPS in adults with FXS and healthy controls. All subjects were males between the ages of 18 and 24 years and free of psychotropic medication. As most fragile X participants were not able to undergo the PET study awake, we used dexmedetomidine for sedation during the imaging studies. We found no differences between rCPS measured during dexmedetomidine-sedation and the awake state in ten healthy controls. In the comparison of rCPS in dexmedetomidine-sedated fragile X participants (n = 9) and healthy controls (n = 14) we found no statistically significant differences. Our results from in vivo measurements in human brain do not support the hypothesis that rCPS are elevated due to the absence of FMRP. This hypothesis is based on findings in animal models and in vitro measurements in human peripheral cells. The absence of a translation suppressor may produce a more complex response in pathways regulating translation than previously thought. We may need to revise our working hypotheses regarding FXS and our thinking about potential therapeutics.
Collapse
Affiliation(s)
- Kathleen C Schmidt
- Section on Neuroadaptation and Protein Metabolism, National Institute of Mental Health, 10 Center Drive, Room 2D54, Bethesda, MD 20892-1298, United States of America
| | - Inna Loutaev
- Section on Neuroadaptation and Protein Metabolism, National Institute of Mental Health, 10 Center Drive, Room 2D54, Bethesda, MD 20892-1298, United States of America
| | - Zenaide Quezado
- Department of Perioperative Medicine, Clinical Center, National Institutes of Health, 10 Center Drive, Bethesda, MD 20892-1512, United States of America
| | - Carrie Sheeler
- Section on Neuroadaptation and Protein Metabolism, National Institute of Mental Health, 10 Center Drive, Room 2D54, Bethesda, MD 20892-1298, United States of America
| | - Carolyn Beebe Smith
- Section on Neuroadaptation and Protein Metabolism, National Institute of Mental Health, 10 Center Drive, Room 2D54, Bethesda, MD 20892-1298, United States of America.
| |
Collapse
|
77
|
Golden CEM, Breen MS, Koro L, Sonar S, Niblo K, Browne A, Burlant N, Di Marino D, De Rubeis S, Baxter MG, Buxbaum JD, Harony-Nicolas H. Deletion of the KH1 Domain of Fmr1 Leads to Transcriptional Alterations and Attentional Deficits in Rats. Cereb Cortex 2020; 29:2228-2244. [PMID: 30877790 PMCID: PMC6458915 DOI: 10.1093/cercor/bhz029] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Revised: 12/11/2018] [Accepted: 02/06/2019] [Indexed: 12/27/2022] Open
Abstract
Fragile X syndrome (FXS) is a neurodevelopmental disorder caused by mutations in the FMR1 gene. It is a leading monogenic cause of autism spectrum disorder and inherited intellectual disability and is often comorbid with attention deficits. Most FXS cases are due to an expansion of CGG repeats leading to suppressed expression of fragile X mental retardation protein (FMRP), an RNA-binding protein involved in mRNA metabolism. We found that the previously published Fmr1 knockout rat model of FXS expresses an Fmr1 transcript with an in-frame deletion of exon 8, which encodes for the K-homology (KH) RNA-binding domain, KH1. Notably, 3 pathogenic missense mutations associated with FXS lie in the KH domains. We observed that the deletion of exon 8 in rats leads to attention deficits and to alterations in transcriptional profiles within the medial prefrontal cortex (mPFC), which map to 2 weighted gene coexpression network modules. These modules are conserved in human frontal cortex and enriched for known FMRP targets. Hub genes in these modules represent potential therapeutic targets for FXS. Taken together, these findings indicate that attentional testing might be a reliable cross-species tool for investigating FXS and identify dysregulated conserved gene networks in a relevant brain region.
Collapse
Affiliation(s)
- Carla E M Golden
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Michael S Breen
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Lacin Koro
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sankalp Sonar
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kristi Niblo
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Andrew Browne
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Natalie Burlant
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Daniele Di Marino
- Faculty of Biomedical Sciences, Institute of Computational Science, Center for Computational Medicine in Cardiology, Università della Svizzera Italiana (USI), Lugano, Switzerland.,Department of Life and Environmental Sciences, Polytechnic University of Marche, Ancona, Italy
| | - Silvia De Rubeis
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Mark G Baxter
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Joseph D Buxbaum
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Hala Harony-Nicolas
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
78
|
Creus-Muncunill J, Badillos-Rodríguez R, Garcia-Forn M, Masana M, Garcia-Díaz Barriga G, Guisado-Corcoll A, Alberch J, Malagelada C, Delgado-García JM, Gruart A, Pérez-Navarro E. Increased translation as a novel pathogenic mechanism in Huntington's disease. Brain 2020; 142:3158-3175. [PMID: 31365052 DOI: 10.1093/brain/awz230] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Revised: 05/29/2019] [Accepted: 06/02/2019] [Indexed: 11/15/2022] Open
Abstract
Huntington's disease is a neurodegenerative disorder caused by a CAG repeat expansion in exon 1 of the huntingtin gene. Striatal projection neurons are mainly affected, leading to motor symptoms, but molecular mechanisms involved in their vulnerability are not fully characterized. Here, we show that eIF4E binding protein (4E-BP), a protein that inhibits translation, is inactivated in Huntington's disease striatum by increased phosphorylation. Accordingly, we detected aberrant de novo protein synthesis. Proteomic characterization indicates that translation specifically affects sets of proteins as we observed upregulation of ribosomal and oxidative phosphorylation proteins and downregulation of proteins related to neuronal structure and function. Interestingly, treatment with the translation inhibitor 4EGI-1 prevented R6/1 mice motor deficits, although corticostriatal long-term depression was not markedly changed in behaving animals. At the molecular level, injection of 4EGI-1 normalized protein synthesis and ribosomal content in R6/1 mouse striatum. In conclusion, our results indicate that dysregulation of protein synthesis is involved in mutant huntingtin-induced striatal neuron dysfunction.
Collapse
Affiliation(s)
- Jordi Creus-Muncunill
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, Barcelona, Catalonia.,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Catalonia.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Spain
| | - Raquel Badillos-Rodríguez
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, Barcelona, Catalonia.,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Catalonia.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Spain
| | - Marta Garcia-Forn
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, Barcelona, Catalonia.,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Catalonia.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Spain
| | - Mercè Masana
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, Barcelona, Catalonia.,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Catalonia.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Spain
| | - Gerardo Garcia-Díaz Barriga
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, Barcelona, Catalonia.,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Catalonia.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Spain
| | - Anna Guisado-Corcoll
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, Barcelona, Catalonia.,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Catalonia.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Spain
| | - Jordi Alberch
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, Barcelona, Catalonia.,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Catalonia.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Spain
| | - Cristina Malagelada
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, Barcelona, Catalonia
| | | | - Agnès Gruart
- Division of Neurosciences, Pablo de Olavide University, Seville, Spain
| | - Esther Pérez-Navarro
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, Barcelona, Catalonia.,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Catalonia.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Spain
| |
Collapse
|
79
|
Utami KH, Yusof NABM, Kwa JE, Peteri UK, Castrén ML, Pouladi MA. Elevated de novo protein synthesis in FMRP-deficient human neurons and its correction by metformin treatment. Mol Autism 2020; 11:41. [PMID: 32460900 PMCID: PMC7251671 DOI: 10.1186/s13229-020-00350-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 05/11/2020] [Indexed: 12/18/2022] Open
Abstract
FXS is the most common genetic cause of intellectual (ID) and autism spectrum disorders (ASD). FXS is caused by loss of FMRP, an RNA-binding protein involved in the translational regulation of a large number of neuronal mRNAs. Absence of FMRP has been shown to lead to elevated protein synthesis and is thought to be a major cause of the synaptic plasticity and behavioural deficits in FXS. The increase in protein synthesis results in part from abnormal activation of key protein translation pathways downstream of ERK1/2 and mTOR signalling. Pharmacological and genetic interventions that attenuate hyperactivation of these pathways can normalize levels of protein synthesis and improve phenotypic outcomes in animal models of FXS. Several efforts are currently underway to trial this strategy in patients with FXS. To date, elevated global protein synthesis as a result of FMRP loss has not been validated in the context of human neurons. Here, using an isogenic human stem cell-based model, we show that de novo protein synthesis is elevated in FMRP-deficient neural cells. We further show that this increase is associated with elevated ERK1/2 and Akt signalling and can be rescued by metformin treatment. Finally, we examined the effect of normalizing protein synthesis on phenotypic abnormalities in FMRP-deficient neural cells. We find that treatment with metformin attenuates the increase in proliferation of FMRP-deficient neural progenitor cells but not the neuronal deficits in neurite outgrowth. The elevated level of protein synthesis and the normalization of neural progenitor proliferation by metformin treatment were validated in additional control and FXS patient-derived hiPSC lines. Overall, our results validate that loss of FMRP results in elevated de novo protein synthesis in human neurons and suggest that approaches targeting this abnormality are likely to be of partial therapeutic benefit in FXS.
Collapse
Affiliation(s)
- Kagistia Hana Utami
- Translational Laboratory in Genetic Medicine, Agency for Science, Technology and Research, Singapore (A*STAR), 8A Biomedical Grove, Immunos, Level 5, Singapore, 138648, Singapore.
| | - Nur Amirah Binte Mohammad Yusof
- Translational Laboratory in Genetic Medicine, Agency for Science, Technology and Research, Singapore (A*STAR), 8A Biomedical Grove, Immunos, Level 5, Singapore, 138648, Singapore
| | - Jing Eugene Kwa
- Translational Laboratory in Genetic Medicine, Agency for Science, Technology and Research, Singapore (A*STAR), 8A Biomedical Grove, Immunos, Level 5, Singapore, 138648, Singapore
| | - Ulla-Kaisa Peteri
- Department of Physiology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Maija L Castrén
- Department of Physiology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Mahmoud A Pouladi
- Translational Laboratory in Genetic Medicine, Agency for Science, Technology and Research, Singapore (A*STAR), 8A Biomedical Grove, Immunos, Level 5, Singapore, 138648, Singapore.
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
| |
Collapse
|
80
|
McCamphill PK, Stoppel LJ, Senter RK, Lewis MC, Heynen AJ, Stoppel DC, Sridhar V, Collins KA, Shi X, Pan JQ, Madison J, Cottrell JR, Huber KM, Scolnick EM, Holson EB, Wagner FF, Bear MF. Selective inhibition of glycogen synthase kinase 3α corrects pathophysiology in a mouse model of fragile X syndrome. Sci Transl Med 2020; 12:eaam8572. [PMID: 32434848 PMCID: PMC8095719 DOI: 10.1126/scitranslmed.aam8572] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Revised: 07/15/2019] [Accepted: 01/11/2020] [Indexed: 01/06/2023]
Abstract
Fragile X syndrome is caused by FMR1 gene silencing and loss of the encoded fragile X mental retardation protein (FMRP), which binds to mRNA and regulates translation. Studies in the Fmr1-/y mouse model of fragile X syndrome indicate that aberrant cerebral protein synthesis downstream of metabotropic glutamate receptor 5 (mGluR5) signaling contributes to disease pathogenesis, but clinical trials using mGluR5 inhibitors were not successful. Animal studies suggested that treatment with lithium might be an alternative approach. Targets of lithium include paralogs of glycogen synthase kinase 3 (GSK3), and nonselective small-molecule inhibitors of these enzymes improved disease phenotypes in a fragile X syndrome mouse model. However, the potential therapeutic use of GSK3 inhibitors has been hampered by toxicity arising from inhibition of both α and β paralogs. Recently, we developed GSK3 inhibitors with sufficient paralog selectivity to avoid a known toxic consequence of dual inhibition, that is, increased β-catenin stabilization. We show here that inhibition of GSK3α, but not GSK3β, corrected aberrant protein synthesis, audiogenic seizures, and sensory cortex hyperexcitability in Fmr1-/y mice. Although inhibiting either paralog prevented induction of NMDA receptor-dependent long-term depression (LTD) in the hippocampus, only inhibition of GSK3α impaired mGluR5-dependent and protein synthesis-dependent LTD. Inhibition of GSK3α additionally corrected deficits in learning and memory in Fmr1-/y mice; unlike mGluR5 inhibitors, there was no evidence of tachyphylaxis or enhanced psychotomimetic-induced hyperlocomotion. GSK3α selective inhibitors may have potential as a therapeutic approach for treating fragile X syndrome.
Collapse
Affiliation(s)
- Patrick K McCamphill
- Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Laura J Stoppel
- Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Rebecca K Senter
- Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Michael C Lewis
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Arnold J Heynen
- Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - David C Stoppel
- Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Vinay Sridhar
- University of Texas Southwestern Medical Center, Department of Neuroscience, Dallas, TX 75390, USA
| | - Katie A Collins
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Xi Shi
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Jen Q Pan
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Jon Madison
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Jeffrey R Cottrell
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Kimberly M Huber
- University of Texas Southwestern Medical Center, Department of Neuroscience, Dallas, TX 75390, USA
| | - Edward M Scolnick
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Edward B Holson
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Florence F Wagner
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.
| | - Mark F Bear
- Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| |
Collapse
|
81
|
Razak KA, Dominick KC, Erickson CA. Developmental studies in fragile X syndrome. J Neurodev Disord 2020; 12:13. [PMID: 32359368 PMCID: PMC7196229 DOI: 10.1186/s11689-020-09310-9] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Accepted: 02/13/2020] [Indexed: 01/27/2023] Open
Abstract
Fragile X syndrome (FXS) is the most common single gene cause of autism and intellectual disabilities. Humans with FXS exhibit increased anxiety, sensory hypersensitivity, seizures, repetitive behaviors, cognitive inflexibility, and social behavioral impairments. The main purpose of this review is to summarize developmental studies of FXS in humans and in the mouse model, the Fmr1 knockout mouse. The literature presents considerable evidence that a number of early developmental deficits can be identified and that these early deficits chart a course of altered developmental experience leading to symptoms well characterized in adolescents and adults. Nevertheless, a number of critical issues remain unclear or untested regarding the development of symptomology and underlying mechanisms. First, what is the role of FMRP, the protein product of Fmr1 gene, during different developmental ages? Does the absence of FMRP during early development lead to irreversible changes, or could reintroduction of FMRP or therapeutics aimed at FMRP-interacting proteins/pathways hold promise when provided in adults? These questions have implications for clinical trial designs in terms of optimal treatment windows, but few studies have systematically addressed these issues in preclinical and clinical work. Published studies also point to complex trajectories of symptom development, leading to the conclusion that single developmental time point studies are unlikely to disambiguate effects of genetic mutation from effects of altered developmental experience and compensatory plasticity. We conclude by suggesting a number of experiments needed to address these major gaps in the field.
Collapse
Affiliation(s)
- Khaleel A Razak
- Department of Psychology and Graduate Neuroscience Program, University of California, Riverside, USA
| | - Kelli C Dominick
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, Cincinnati, OH, USA.,Division of Child and Adolescent Psychiatry, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue MLC 4002, Cincinnati, OH, 45229, USA
| | - Craig A Erickson
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, Cincinnati, OH, USA. .,Division of Child and Adolescent Psychiatry, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue MLC 4002, Cincinnati, OH, 45229, USA.
| |
Collapse
|
82
|
Thurman AJ, Potter LA, Kim K, Tassone F, Banasik A, Potter SN, Bullard L, Nguyen V, McDuffie A, Hagerman R, Abbeduto L. Controlled trial of lovastatin combined with an open-label treatment of a parent-implemented language intervention in youth with fragile X syndrome. J Neurodev Disord 2020; 12:12. [PMID: 32316911 PMCID: PMC7175541 DOI: 10.1186/s11689-020-09315-4] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Accepted: 03/27/2020] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND The purpose of this study was to conduct a 20-week controlled trial of lovastatin (10 to 40 mg/day) in youth with fragile X syndrome (FXS) ages 10 to 17 years, combined with an open-label treatment of a parent-implemented language intervention (PILI), delivered via distance video teleconferencing to both treatment groups, lovastatin and placebo. METHOD A randomized, double-blind trial was conducted at one site in the Sacramento, California, metropolitan area. Fourteen participants were assigned to the lovastatin group; two participants terminated early from the study. Sixteen participants were assigned to the placebo group. Lovastatin or placebo was administered orally in a capsule form, starting at 10 mg and increasing weekly or as tolerated by 10 mg increments, up to a maximum dose of 40 mg daily. A PILI was delivered to both groups for 12 weeks, with 4 activities per week, through video teleconferencing by an American Speech-Language Association-certified Speech-Language Pathologist, in collaboration with a Board-Certified Behavior Analyst. Parents were taught to use a set of language facilitation strategies while interacting with their children during a shared storytelling activity. The main outcome measures included absolute change from baseline to final visit in the means for youth total number of story-related utterances, youth number of different word roots, and parent total number of story-related utterances. RESULTS Significant increases in all primary outcome measures were observed in both treatment groups. Significant improvements were also observed in parent reports of the severity of spoken language and social impairments in both treatment groups. In all cases, the amount of change observed did not differ across the two treatment groups. Although gains in parental use of the PILI-targeted intervention strategies were observed in both treatment groups, parental use of the PILI strategies was correlated with youth gains in the placebo group and not in the lovastatin group. CONCLUSION Participants in both groups demonstrated significant changes in the primary outcome measures. The magnitude of change observed across the two groups was comparable, providing additional support for the efficacy of the use of PILI in youth with FXS. TRIAL REGISTRATION US National Institutes of Health (ClinicalTrials.gov), NCT02642653. Registered 12/30/2015.
Collapse
Affiliation(s)
- Angela John Thurman
- MIND Institute, University of California Davis Health, 2825 50th Street, Room 2335, Sacramento, CA, 95817, USA.
- Department of Psychiatry and Behavioral Sciences, University of California Davis Health, Sacramento, USA.
| | - Laura A Potter
- MIND Institute, University of California Davis Health, 2825 50th Street, Room 2335, Sacramento, CA, 95817, USA
- Department of Pediatrics, University of California Davis Health, Sacramento, USA
| | - Kyoungmi Kim
- MIND Institute, University of California Davis Health, 2825 50th Street, Room 2335, Sacramento, CA, 95817, USA
- Department of Public Health Sciences, University of California Davis Health, Sacramento, USA
| | - Flora Tassone
- MIND Institute, University of California Davis Health, 2825 50th Street, Room 2335, Sacramento, CA, 95817, USA
- Department of Biochemistry and Molecular Medicine, University of California Davis Health, Sacramento, USA
| | - Amy Banasik
- MIND Institute, University of California Davis Health, 2825 50th Street, Room 2335, Sacramento, CA, 95817, USA
- Department of Psychiatry and Behavioral Sciences, University of California Davis Health, Sacramento, USA
| | - Sarah Nelson Potter
- MIND Institute, University of California Davis Health, 2825 50th Street, Room 2335, Sacramento, CA, 95817, USA
- Department of Human Ecology, University of California Davis, Davis, USA
| | - Lauren Bullard
- MIND Institute, University of California Davis Health, 2825 50th Street, Room 2335, Sacramento, CA, 95817, USA
- Department of Human Ecology, University of California Davis, Davis, USA
| | - Vivian Nguyen
- MIND Institute, University of California Davis Health, 2825 50th Street, Room 2335, Sacramento, CA, 95817, USA
- Department of Psychiatry and Behavioral Sciences, University of California Davis Health, Sacramento, USA
| | - Andrea McDuffie
- MIND Institute, University of California Davis Health, 2825 50th Street, Room 2335, Sacramento, CA, 95817, USA
- Department of Psychiatry and Behavioral Sciences, University of California Davis Health, Sacramento, USA
| | - Randi Hagerman
- MIND Institute, University of California Davis Health, 2825 50th Street, Room 2335, Sacramento, CA, 95817, USA
- Department of Pediatrics, University of California Davis Health, Sacramento, USA
| | - Leonard Abbeduto
- MIND Institute, University of California Davis Health, 2825 50th Street, Room 2335, Sacramento, CA, 95817, USA
- Department of Psychiatry and Behavioral Sciences, University of California Davis Health, Sacramento, USA
| |
Collapse
|
83
|
Griffiths KK, Wang A, Wang L, Tracey M, Kleiner G, Quinzii CM, Sun L, Yang G, Perez-Zoghbi JF, Licznerski P, Yang M, Jonas EA, Levy RJ. Inefficient thermogenic mitochondrial respiration due to futile proton leak in a mouse model of fragile X syndrome. FASEB J 2020; 34:7404-7426. [PMID: 32307754 DOI: 10.1096/fj.202000283rr] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 03/13/2020] [Accepted: 03/16/2020] [Indexed: 12/20/2022]
Abstract
Fragile X syndrome (FXS) is the leading known inherited intellectual disability and the most common genetic cause of autism. The full mutation results in transcriptional silencing of the Fmr1 gene and loss of fragile X mental retardation protein (FMRP) expression. Defects in neuroenergetic capacity are known to cause a variety of neurodevelopmental disorders. Thus, we explored the integrity of forebrain mitochondria in Fmr1 knockout mice during the peak of synaptogenesis. We found inefficient thermogenic respiration due to futile proton leak in Fmr1 KO mitochondria caused by coenzyme Q (CoQ) deficiency and an open cyclosporine-sensitive channel. Repletion of mitochondrial CoQ within the Fmr1 KO forebrain closed the channel, blocked the pathological proton leak, restored rates of protein synthesis during synaptogenesis, and normalized the key phenotypic features later in life. The findings demonstrate that FMRP deficiency results in inefficient oxidative phosphorylation during the neurodevelopment and suggest that dysfunctional mitochondria may contribute to the FXS phenotype.
Collapse
Affiliation(s)
- Keren K Griffiths
- Department of Anesthesiology, Columbia University Medical Center, New York, NY, USA
| | - Aili Wang
- Department of Anesthesiology, Columbia University Medical Center, New York, NY, USA
| | - Lifei Wang
- Department of Anesthesiology, Columbia University Medical Center, New York, NY, USA
| | - Matthew Tracey
- Department of Anesthesiology, Columbia University Medical Center, New York, NY, USA
| | - Giulio Kleiner
- Department of Neurology, Columbia University Medical Center, New York, NY, USA
| | - Catarina M Quinzii
- Department of Neurology, Columbia University Medical Center, New York, NY, USA
| | - Linlin Sun
- Department of Anesthesiology, Columbia University Medical Center, New York, NY, USA
| | - Guang Yang
- Department of Anesthesiology, Columbia University Medical Center, New York, NY, USA
| | - Jose F Perez-Zoghbi
- Department of Anesthesiology, Columbia University Medical Center, New York, NY, USA
| | - Pawel Licznerski
- Section of Endocrinology, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Mu Yang
- Institute of Genomic Medicine and Psychiatry, Columbia University Medical Center, New York, NY, USA
| | - Elizabeth A Jonas
- Section of Endocrinology, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Richard J Levy
- Department of Anesthesiology, Columbia University Medical Center, New York, NY, USA
| |
Collapse
|
84
|
Bülow P, Murphy TJ, Bassell GJ, Wenner P. Homeostatic Intrinsic Plasticity Is Functionally Altered in Fmr1 KO Cortical Neurons. Cell Rep 2020; 26:1378-1388.e3. [PMID: 30726724 PMCID: PMC6443253 DOI: 10.1016/j.celrep.2019.01.035] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Revised: 11/20/2018] [Accepted: 01/09/2019] [Indexed: 12/12/2022] Open
Abstract
Cortical hyperexcitability is a hallmark of fragile X syndrome (FXS). In the Fmr1 knockout (KO) mouse model of FXS,
cortical hyperexcitability is linked to sensory hypersensitivity and seizure susceptibility. It remains unclear why homeostatic
mechanisms fail to prevent such activity. Homeostatic intrinsic plasticity (HIP) adjusts membrane excitability through regulation
of ion channels to maintain activity levels following activity perturbation. Despite the critical role of HIP in the maturation of
excitability, it has not been examined in FXS. Here, we demonstrate that HIP does not operate normally in a disease model, FXS.
HIP was either lost or exaggerated in two distinct neuronal populations from Fmr1 KO cortical cultures. In addition, we have
identified a mechanism for homeostatic intrinsic plasticity. Compromising HIP function during development could leave cortical
neurons in the FXS nervous system vulnerable to hyperexcitability. Fragile X syndrome (FXS) is characterized by cortical hyperexcitability, but the mechanisms driving hyperexcitability are
poorly understood. Homeostatic intrinsic plasticity (HIP) regulates ion channel function to maintain appropriate activity levels.
Bülow et al. show that HIP is functionally altered in FXS neurons, which may leave cortical neurons vulnerable to
hyperexcitability.
Collapse
Affiliation(s)
- Pernille Bülow
- Department of Physiology, Emory University School of Medicine, Atlanta, GA, USA; Department of Cell Biology, Emory University School of Medicine, Atlanta, GA, USA
| | - T J Murphy
- Department of Pharmacology, Emory University School of Medicine, Atlanta, GA, USA
| | - Gary J Bassell
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA, USA.
| | - Peter Wenner
- Department of Physiology, Emory University School of Medicine, Atlanta, GA, USA.
| |
Collapse
|
85
|
Ding Q, Sethna F, Wu XT, Miao Z, Chen P, Zhang Y, Xiao H, Feng W, Feng Y, Li X, Wang H. Transcriptome signature analysis repurposes trifluoperazine for the treatment of fragile X syndrome in mouse model. Commun Biol 2020; 3:127. [PMID: 32179850 PMCID: PMC7075969 DOI: 10.1038/s42003-020-0833-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 02/07/2020] [Indexed: 02/06/2023] Open
Abstract
Fragile X syndrome (FXS) is a prevailing genetic disorder of intellectual disability and autism. There is no efficacious medication for FXS. Through in silico screening with a public database, computational analysis of transcriptome profile in FXS mouse neurons predicts therapeutic value of an FDA-approved drug trifluoperazine. Systemic administration of low-dose trifluoperazine at 0.05 mg/kg attenuates multiple FXS- and autism-related behavioral symptoms. Moreover, computational analysis of transcriptome alteration caused by trifluoperazine suggests a new mechanism of action against PI3K (Phosphatidylinositol-4,5-bisphosphate 3-kinase) activity. Consistently, trifluoperazine suppresses PI3K activity and its down-stream targets Akt (protein kinase B) and S6K1 (S6 kinase 1) in neurons. Further, trifluoperazine normalizes the aberrantly elevated activity of Akt and S6K1 and enhanced protein synthesis in FXS mouse. Together, our data demonstrate a promising value of transcriptome-based computation in identification of therapeutic strategy and repurposing drugs for neurological disorders, and suggest trifluoperazine as a potential treatment for FXS. Qi Ding, Ferzin Sethna et al. perform a computational analysis of the transcriptome profile of Fmr1−/− neurons and identify trifluoperazine as potential therapeutic agent against Fragile X Syndrome. Next, they show that low doses of trifluoperazine ameliorate some of the behavioral and molecular phenotypes present in Fmr1−/− mice.
Collapse
Affiliation(s)
- Qi Ding
- Department of Physiology, Michigan State University, East Lansing, USA
| | - Ferzin Sethna
- Genetics Program, Michigan State University, East Lansing, USA
| | - Xue-Ting Wu
- Key Laboratory of Synthetic Biology, Institute of Plant Physiology and Ecology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 200032, Shanghai, China
| | - Zhuang Miao
- Genetics Program, Michigan State University, East Lansing, USA
| | - Ping Chen
- Key Laboratory of Synthetic Biology, Institute of Plant Physiology and Ecology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 200032, Shanghai, China
| | - Yueqi Zhang
- Department of Physiology, Michigan State University, East Lansing, USA
| | - Hua Xiao
- Department of Physiology, Michigan State University, East Lansing, USA
| | - Wei Feng
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, 30322, Georgia
| | - Yue Feng
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, 30322, Georgia
| | - Xuan Li
- Key Laboratory of Synthetic Biology, Institute of Plant Physiology and Ecology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 200032, Shanghai, China
| | - Hongbing Wang
- Department of Physiology, Michigan State University, East Lansing, USA. .,Neuroscience Program, Michigan State University, East Lansing, MI, 48824, USA.
| |
Collapse
|
86
|
Considerations for Clinical Therapeutic Development of Statins for Neurodevelopmental Disorders. eNeuro 2020; 7:ENEURO.0392-19.2020. [PMID: 32071072 PMCID: PMC7070444 DOI: 10.1523/eneuro.0392-19.2020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2019] [Revised: 01/10/2020] [Accepted: 01/15/2020] [Indexed: 12/18/2022] Open
|
87
|
McCullagh EA, Rotschafer SE, Auerbach BD, Klug A, Kaczmarek LK, Cramer KS, Kulesza RJ, Razak KA, Lovelace JW, Lu Y, Koch U, Wang Y. Mechanisms underlying auditory processing deficits in Fragile X syndrome. FASEB J 2020; 34:3501-3518. [PMID: 32039504 DOI: 10.1096/fj.201902435r] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 12/31/2019] [Accepted: 01/18/2020] [Indexed: 01/14/2023]
Abstract
Autism spectrum disorders (ASD) are strongly associated with auditory hypersensitivity or hyperacusis (difficulty tolerating sounds). Fragile X syndrome (FXS), the most common monogenetic cause of ASD, has emerged as a powerful gateway for exploring underlying mechanisms of hyperacusis and auditory dysfunction in ASD. This review discusses examples of disruption of the auditory pathways in FXS at molecular, synaptic, and circuit levels in animal models as well as in FXS individuals. These examples highlight the involvement of multiple mechanisms, from aberrant synaptic development and ion channel deregulation of auditory brainstem circuits, to impaired neuronal plasticity and network hyperexcitability in the auditory cortex. Though a relatively new area of research, recent discoveries have increased interest in auditory dysfunction and mechanisms underlying hyperacusis in this disorder. This rapidly growing body of data has yielded novel research directions addressing critical questions regarding the timing and possible outcomes of human therapies for auditory dysfunction in ASD.
Collapse
Affiliation(s)
- Elizabeth A McCullagh
- Department of Physiology and Biophysics, University of Colorado Anschutz, Aurora, CO, USA.,Department of Integrative Biology, Oklahoma State University, Stillwater, OK, USA
| | - Sarah E Rotschafer
- Department of Neurobiology and Behavior, University of California, Irvine, CA, USA.,Department of Biomedical Sciences, Mercer University School of Medicine, Savannah, GA, USA
| | - Benjamin D Auerbach
- Center for Hearing and Deafness, Department of Communicative Disorders & Sciences, SUNY at Buffalo, Buffalo, NY, USA
| | - Achim Klug
- Department of Physiology and Biophysics, University of Colorado Anschutz, Aurora, CO, USA
| | - Leonard K Kaczmarek
- Departments of Pharmacology and Cellular and Molecular Physiology, Yale University, New Haven, CT, USA
| | - Karina S Cramer
- Department of Neurobiology and Behavior, University of California, Irvine, CA, USA
| | - Randy J Kulesza
- Department of Anatomy, Lake Erie College of Osteopathic Medicine, Erie, PA, USA
| | - Khaleel A Razak
- Department of Psychology, University of California, Riverside, CA, USA
| | | | - Yong Lu
- Department of Anatomy and Neurobiology, College of Medicine, Northeast Ohio Medical University, Rootstown, OH, USA
| | - Ursula Koch
- Institute of Biology, Neurophysiology, Freie Universität Berlin, Berlin, Germany
| | - Yuan Wang
- Department of Biomedical Sciences, Program in Neuroscience, Florida State University, Tallahassee, FL, USA
| |
Collapse
|
88
|
Loss of fragile X mental retardation protein precedes Lewy pathology in Parkinson's disease. Acta Neuropathol 2020; 139:319-345. [PMID: 31768670 DOI: 10.1007/s00401-019-02099-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 11/10/2019] [Accepted: 11/11/2019] [Indexed: 12/11/2022]
Abstract
Parkinson's disease (PD) is the most common neurodegenerative movement disorder and is characterized by the progressive loss of dopaminergic (DA) neurons in the substantia nigra pars compacta (SNc) and the gradual appearance of α-synuclein (α-syn)-containing neuronal protein aggregates. Although the exact mechanism of α-syn-mediated cell death remains elusive, recent research suggests that α-syn-induced alterations in neuronal excitability contribute to cell death in PD. Because the fragile X mental retardation protein (FMRP) controls the expression and function of numerous neuronal genes related to neuronal excitability and synaptic function, we here investigated the role of FMRP in α-syn-associated pathological changes in cell culture and mouse models of PD as well as in post-mortem human brain tissue from PD patients. We found FMRP to be decreased in cultured DA neurons and in the mouse brain in response to α-syn overexpression. FMRP was, furthermore, lost in the SNc of PD patients and in patients with early stages of incidental Lewy body disease (iLBD). Unlike fragile X syndrome (FXS), FMR1 expression in response to α-syn was regulated by a mechanism involving Protein Kinase C (PKC) and cAMP response element-binding protein (CREB). Reminiscent of FXS neurons, α-syn-overexpressing cells exhibited an increase in membrane N-type calcium channels, increased phosphorylation of ERK1/2, eIF4E and S6, increased overall protein synthesis, and increased expression of Matrix Metalloproteinase 9 (MMP9). FMRP affected neuronal function in a PD animal model, because FMRP-KO mice were resistant to the effect of α-syn on striatal dopamine release. In summary, our results thus reveal a new role of FMRP in PD and support the examination of FMRP-regulated genes in PD disease progression.
Collapse
|
89
|
Seese RR, Le AA, Wang K, Cox CD, Lynch G, Gall CM. A TrkB agonist and ampakine rescue synaptic plasticity and multiple forms of memory in a mouse model of intellectual disability. Neurobiol Dis 2020; 134:104604. [PMID: 31494285 PMCID: PMC7258745 DOI: 10.1016/j.nbd.2019.104604] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 07/26/2019] [Accepted: 09/04/2019] [Indexed: 12/20/2022] Open
Abstract
Fragile X syndrome (FXS) is associated with deficits in various types of learning, including those that require the hippocampus. Relatedly, hippocampal long-term potentiation (LTP) is impaired in the Fmr1 knockout (KO) mouse model of FXS. Prior research found that infusion of brain-derived neurotrophic factor (BDNF) rescues LTP in the KOs. Here, we tested if, in Fmr1 KO mice, up-regulating BDNF production or treatment with an agonist for BDNF's TrkB receptor restores synaptic plasticity and improves learning. In hippocampal slices, bath infusion of the TrkB agonist 7,8-dihydroxyflavone (7,8-DHF) completely restored otherwise impaired hippocampal field CA1 LTP of Fmr1 KOs without effect in wild types (WTs). Similarly, acute, semi-chronic, or chronic treatments with 7,8-DHF rescued a simple hippocampus-dependent form of spatial learning (object location memory: OLM) in Fmr1 KOs without effect in WTs. The agonist also restored object recognition memory, which depends on cortical regions. Semi-chronic, but not acute, treatment with the ampakine CX929, which up-regulates BDNF expression, lowered the training threshold for OLM in WT mice and rescued learning in the KOs. Positive results were also obtained in a test for social recognition. An mGluR5 antagonist did not improve learning. Quantification of synaptic immunolabeling demonstrated that 7,8-DHF and CX929 increase levels of activated TrkB at excitatory synapses. Moreover, CX929 induced a robust synaptic activation of the TrkB effector ERK1/2. These results suggest that enhanced synaptic BDNF signaling constitutes a plausible strategy for treating certain aspects of the cognitive disabilities associated with FXS.
Collapse
Affiliation(s)
- Ronald R Seese
- Department of Anatomy and Neurobiology, University of California, Irvine, CA, United States of America
| | - Aliza A Le
- Department of Anatomy and Neurobiology, University of California, Irvine, CA, United States of America
| | - Kathleen Wang
- Department of Anatomy and Neurobiology, University of California, Irvine, CA, United States of America
| | - Conor D Cox
- Department of Anatomy and Neurobiology, University of California, Irvine, CA, United States of America
| | - Gary Lynch
- Department of Anatomy and Neurobiology, University of California, Irvine, CA, United States of America; Department of Psychiatry and Human Behavior, University of California, Irvine, CA, United States of America.
| | - Christine M Gall
- Department of Anatomy and Neurobiology, University of California, Irvine, CA, United States of America; Department of Neurobiology and Behavior, University of California, Irvine, CA, United States of America.
| |
Collapse
|
90
|
Bhat SA, Henry RJ, Blanchard AC, Stoica BA, Loane DJ, Faden AI. Enhanced Akt/GSK-3β/CREB signaling mediates the anti-inflammatory actions of mGluR5 positive allosteric modulators in microglia and following traumatic brain injury in male mice. J Neurochem 2020; 156:225-248. [PMID: 31926033 DOI: 10.1111/jnc.14954] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Revised: 12/16/2019] [Accepted: 01/02/2020] [Indexed: 12/20/2022]
Abstract
We have previously shown that treatment with a mGluR5 positive allosteric modulator (PAM) is neuroprotective after experimental traumatic brain injury (TBI), limiting post-traumatic neuroinflammation by reducing pro-inflammatory microglial activation and promoting anti-inflammatory and neuroprotective responses. However, the specific molecular mechanisms governing this anti-inflammatory shift in microglia remain unknown. Here we show that the mGluR5 PAM, VU0360172 (VuPAM), regulates microglial inflammatory responses through activation of Akt, resulting in the inhibition of GSK-3β. GSK-3β regulates the phosphorylation of CREB, thereby controlling the expression of inflammation-related genes and microglial plasticity. The anti-inflammatory action of VuPAM in microglia is reversed by inhibiting Akt/GSK-3β/CREB signaling. Using a well-characterized TBI model and CX3CR1gfp/+ mice to visualize microglia in vivo, we demonstrate that VuPAM enhances Akt/GSK-3β/CREB signaling in the injured cortex, as well as anti-inflammatory microglial markers. Furthermore, in situ analysis revealed that GFP + microglia in the cortex of VuPAM-treated TBI mice co-express pCREB and the anti-inflammatory microglial phenotype marker YM1. Taken together, our data show that VuPAM decreases pro-inflammatory microglial activation by modulating Akt/GSK-3β/CREB signaling. These findings serve to clarify the potential neuroprotective mechanisms of mGluR5 PAM treatment after TBI, and suggest novel therapeutic targets for post-traumatic neuroinflammation. Cover Image for this issue: https://doi.org/10.1111/jnc.15048.
Collapse
Affiliation(s)
- Shahnawaz A Bhat
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Rebecca J Henry
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Alexa C Blanchard
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Bogdan A Stoica
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, Baltimore, MD, USA
| | - David J Loane
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, Baltimore, MD, USA.,School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College, Dublin, Ireland
| | - Alan I Faden
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
91
|
Schmitt LM, Wang J, Pedapati EV, Thurman AJ, Abbeduto L, Erickson CA, Sweeney JA. A neurophysiological model of speech production deficits in fragile X syndrome. Brain Commun 2019; 2. [PMID: 32924010 PMCID: PMC7425415 DOI: 10.1093/braincomms/fcz042] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Fragile X syndrome is the most common inherited intellectual disability and monogenic cause of autism spectrum disorder. Expressive language deficits, especially in speech production, are nearly ubiquitous among individuals with fragile X, but understanding of the neurological bases for these deficits remains limited. Speech production depends on feedforward control and the synchronization of neural oscillations between speech-related areas of frontal cortex and auditory areas of temporal cortex. Interaction in this circuitry allows the corollary discharge of intended speech generated from an efference copy of speech commands to be compared against actual speech sounds, which is critical for making adaptive adjustments to optimize future speech. We aimed to determine whether alterations in coherence between frontal and temporal cortices prior to speech production are present in individuals with fragile X and whether they relate to expressive language dysfunction. Twenty-one participants with full-mutation fragile X syndrome (aged 7-55 years, eight females) and 20 healthy controls (matched on age and sex) completed a talk/listen paradigm during high-density EEG recordings. During the talk task, participants repeated pronounced short vocalizations of 'Ah' every 1-2 s for a total of 180 s. During the listen task, participants passively listened to their recordings from the talk task. We compared pre-speech event-related potential activity, N1 suppression to speech sounds, single trial gamma power and fronto-temporal coherence between groups during these tasks and examined their relation to performance during a naturalistic language task. Prior to speech production, fragile X participants showed reduced pre-speech negativity, reduced fronto-temporal connectivity and greater frontal gamma power compared to controls. N1 suppression during self-generated speech did not differ between groups. Reduced pre-speech activity and increased frontal gamma power prior to speech production were related to less intelligible speech as well as broader social communication deficits in fragile X syndrome. Our findings indicate that coordinated pre-speech activity between frontal and temporal cortices is disrupted in individuals with fragile X in a clinically relevant way and represents a mechanism contributing to prominent speech production problems in the disorder.
Collapse
Affiliation(s)
- Lauren M Schmitt
- Division of Developmental and Behavioral Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.,Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Jun Wang
- Department of Psychology, Zhejiang Normal University, Jinhua, Zhejiang 321004, China
| | - Ernest V Pedapati
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati College of Medicine, Cincinnati, OH, USA.,Department of Psychiatry, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Angela John Thurman
- Psychiatry and Behavioral Sciences, University of California, Davis, MIND Institute, Sacramento, CA, USA
| | - Leonard Abbeduto
- Psychiatry and Behavioral Sciences, University of California, Davis, MIND Institute, Sacramento, CA, USA
| | - Craig A Erickson
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati College of Medicine, Cincinnati, OH, USA.,Department of Psychiatry, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - John A Sweeney
- Department of Psychiatry, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| |
Collapse
|
92
|
Gonzalez D, Tomasek M, Hays S, Sridhar V, Ammanuel S, Chang CW, Pawlowski K, Huber KM, Gibson JR. Audiogenic Seizures in the Fmr1 Knock-Out Mouse Are Induced by Fmr1 Deletion in Subcortical, VGlut2-Expressing Excitatory Neurons and Require Deletion in the Inferior Colliculus. J Neurosci 2019; 39:9852-9863. [PMID: 31666356 PMCID: PMC6891051 DOI: 10.1523/jneurosci.0886-19.2019] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 09/06/2019] [Accepted: 10/16/2019] [Indexed: 02/07/2023] Open
Abstract
Fragile X syndrome (FXS) is the most common form of inherited intellectual disability and the leading monogenetic cause of autism. One symptom of FXS and autism is sensory hypersensitivity (also called sensory over-responsivity). Perhaps related to this, the audiogenic seizure (AGS) is arguably the most robust behavioral phenotype in the FXS mouse model-the Fmr1 knock-out (KO) mouse. Therefore, the AGS may be considered a mouse model of sensory hypersensitivity. Hyperactive circuits are hypothesized to underlie dysfunction in a number of brain regions in patients with FXS and Fmr1 KO mice, and the AGS may be a result of this. But the specific cell types and brain regions underlying AGSs in the Fmr1 KO are unknown. We used conditional deletion or expression of Fmr1 in different cell populations to determine whether Fmr1 deletion in those cells was sufficient or necessary, respectively, for the AGS phenotype in males. Our data indicate that Fmr1 deletion in glutamatergic neurons that express vesicular glutamate transporter 2 (VGlut2) and are located in subcortical brain regions is sufficient and necessary to cause AGSs. Furthermore, the deletion of Fmr1 in glutamatergic neurons of the inferior colliculus is necessary for AGSs. When we demonstrate necessity, we show that Fmr1 expression in either the larger population of VGlut2-expressing glutamatergic neurons or the smaller population of inferior collicular glutamatergic neurons-in an otherwise Fmr1 KO mouse-eliminates AGSs. Therefore, targeting these neuronal populations in FXS and autism may be part of a therapeutic strategy to alleviate sensory hypersensitivity.SIGNIFICANCE STATEMENT Sensory hypersensitivity in fragile X syndrome (FXS) and autism patients significantly interferes with quality of life. Audiogenic seizures (AGSs) are arguably the most robust behavioral phenotype in the FXS mouse model-the Fmr1 knockout-and may be considered a model of sensory hypersensitivity in FXS. We provide the clearest and most precise genetic evidence to date for the cell types and brain regions involved in causing AGSs in the Fmr1 knockout and, more broadly, for any mouse mutant. The expression of Fmr1 in these same cell types in an otherwise Fmr1 knockout eliminates AGSs indicating possible cellular targets for alleviating sensory hypersensitivity in FXS and other forms of autism.
Collapse
Affiliation(s)
| | | | - Seth Hays
- Department of Neuroscience, Dallas, and
| | | | | | | | - Karen Pawlowski
- Department of Otolaryngology and Biomedical Engineering Program, University of Texas Southwestern Medical Center, Dallas, Texas 75390-9035
| | | | | |
Collapse
|
93
|
Ciranna L, Costa L. Pituitary Adenylate Cyclase-Activating Polypeptide Modulates Hippocampal Synaptic Transmission and Plasticity: New Therapeutic Suggestions for Fragile X Syndrome. Front Cell Neurosci 2019; 13:524. [PMID: 31827422 PMCID: PMC6890831 DOI: 10.3389/fncel.2019.00524] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Accepted: 11/08/2019] [Indexed: 12/13/2022] Open
Abstract
Pituitary adenylate cyclase-activating polypeptide (PACAP) modulates glutamatergic synaptic transmission and plasticity in the hippocampus, a brain area with a key role in learning and memory. In agreement, several studies have demonstrated that PACAP modulates learning in physiological conditions. Recent publications show reduced PACAP levels and/or alterations in PACAP receptor expression in different conditions associated with cognitive disability. It is noteworthy that PACAP administration rescued impaired synaptic plasticity and learning in animal models of aging, Alzheimer's disease, Parkinson's disease, and Huntington's chorea. In this context, results from our laboratory demonstrate that PACAP rescued metabotropic glutamate receptor-mediated synaptic plasticity in the hippocampus of a mouse model of fragile X syndrome (FXS), a genetic form of intellectual disability. PACAP is actively transported through the blood-brain barrier and reaches the brain following intranasal or intravenous administration. Besides, new studies have identified synthetic PACAP analog peptides with improved selectivity and pharmacokinetic properties with respect to the native peptide. Our review supports the shared idea that pharmacological activation of PACAP receptors might be beneficial for brain pathologies with cognitive disability. In addition, we suggest that the effects of PACAP treatment might be further studied as a possible therapy in FXS.
Collapse
Affiliation(s)
- Lucia Ciranna
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Lara Costa
- Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| |
Collapse
|
94
|
Cooke SK, Russin J, Moulton K, Nadel J, Loutaev I, Gu Q, Li Z, Smith CB. Effects of the presence and absence of amino acids on translation, signaling, and long-term depression in hippocampal slices from Fmr1 knockout mice. J Neurochem 2019; 151:764-776. [PMID: 31539452 DOI: 10.1111/jnc.14874] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 08/02/2019] [Accepted: 08/18/2019] [Indexed: 01/28/2023]
Abstract
Fragile X syndrome (FXS) is caused by silencing of the FMR1 gene and consequent absence of its protein product, fragile X mental retardation protein (FMRP). FMRP is an RNA-binding protein that can suppress translation. The absence of FMRP leads to symptoms of FXS including intellectual disability and has been proposed to lead to abnormalities in synaptic plasticity. Synaptic plasticity, protein synthesis, and cellular growth pathways have been studied extensively in hippocampal slices from a mouse model of FXS (Fmr1 KO). Enhanced metabotropic glutamate receptor 5 (mGluR5)-dependent long-term depression (LTD), increased rates of protein synthesis, and effects on signaling molecules have been reported. These phenotypes were found under amino acid starvation, a condition that has widespread, powerful effects on activation and translation of proteins involved in regulating protein synthesis. We asked if this non-physiological condition could have effects on Fmr1 KO phenotypes reported in hippocampal slices. We performed hippocampal slice experiments in the presence and absence of amino acids. We measured rates of incorporation of a radiolabeled amino acid into protein to determine protein synthesis rates. By means of western blots, we assessed relative levels of total and phosphorylated forms of proteins involved in signaling pathways regulating translation. We measured evoked field potentials in area CA1 to assess the strength of the long-term depression response to mGluR activation. In the absence of amino acids, we replicate many of the reported findings in Fmr1 KO hippocampal slices, but in the more physiological condition of inclusion of amino acids in the medium, we did not find evidence of enhanced mGluR5-dependent LTD. Activation of mGluR5 increased protein synthesis in both wild type and Fmr1 KO. Moreover, mGluR5 activation increased eIF2α phosphorylation and decreased phosphorylation of p70S6k in slices from Fmr1 KO. We propose that the eIF2α response is a cellular attempt to compensate for the lack of regulation of translation by FMRP. Our findings call for a re-examination of the mGluR theory of FXS.
Collapse
Affiliation(s)
- Spencer K Cooke
- Section on Neuroadaptation and Protein Metabolism, Department of Health and Human Services, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland, USA
| | - Jacob Russin
- Section on Neuroadaptation and Protein Metabolism, Department of Health and Human Services, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland, USA
| | - Kristen Moulton
- Section on Neuroadaptation and Protein Metabolism, Department of Health and Human Services, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland, USA
| | - Jeffrey Nadel
- Section on Neuroadaptation and Protein Metabolism, Department of Health and Human Services, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland, USA
| | - Inna Loutaev
- Section on Neuroadaptation and Protein Metabolism, Department of Health and Human Services, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland, USA
| | - Qinhua Gu
- Section on Synapse Development and Plasticity, Department of Health and Human Services, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland, USA
| | - Zheng Li
- Section on Synapse Development and Plasticity, Department of Health and Human Services, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland, USA
| | - Carolyn Beebe Smith
- Section on Neuroadaptation and Protein Metabolism, Department of Health and Human Services, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
95
|
Hui KK, Chen YK, Endo R, Tanaka M. Translation from the Ribosome to the Clinic: Implication in Neurological Disorders and New Perspectives from Recent Advances. Biomolecules 2019; 9:E680. [PMID: 31683805 PMCID: PMC6920867 DOI: 10.3390/biom9110680] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 10/27/2019] [Accepted: 10/29/2019] [Indexed: 12/12/2022] Open
Abstract
De novo protein synthesis by the ribosome and its multitude of co-factors must occur in a tightly regulated manner to ensure that the correct proteins are produced accurately at the right time and, in some cases, also in the proper location. With novel techniques such as ribosome profiling and cryogenic electron microscopy, our understanding of this basic biological process is better than ever and continues to grow. Concurrently, increasing attention is focused on how translational regulation in the brain may be disrupted during the progression of various neurological disorders. In fact, translational dysregulation is now recognized as the de facto pathogenic cause for some disorders. Novel mechanisms including ribosome stalling, ribosome-associated quality control, and liquid-liquid phase separation are closely linked to translational regulation, and may thus be involved in the pathogenic process. The relationships between translational dysregulation and neurological disorders, as well as the ways through which we may be able to reverse those detrimental effects, will be examined in this review.
Collapse
Affiliation(s)
- Kelvin K Hui
- Laboratory for Protein Conformation Diseases, RIKEN Center for Brain Science, Wako, Saitama 351-0198, Japan.
| | - Yi-Kai Chen
- Laboratory for Protein Conformation Diseases, RIKEN Center for Brain Science, Wako, Saitama 351-0198, Japan.
| | - Ryo Endo
- Laboratory for Protein Conformation Diseases, RIKEN Center for Brain Science, Wako, Saitama 351-0198, Japan.
| | - Motomasa Tanaka
- Laboratory for Protein Conformation Diseases, RIKEN Center for Brain Science, Wako, Saitama 351-0198, Japan.
| |
Collapse
|
96
|
Nishiyama J. Plasticity of dendritic spines: Molecular function and dysfunction in neurodevelopmental disorders. Psychiatry Clin Neurosci 2019; 73:541-550. [PMID: 31215705 DOI: 10.1111/pcn.12899] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2018] [Revised: 06/04/2019] [Accepted: 06/06/2019] [Indexed: 02/06/2023]
Abstract
Dendritic spines are tiny postsynaptic protrusions from a dendrite that receive most of the excitatory synaptic input in the brain. Functional and structural changes in dendritic spines are critical for synaptic plasticity, a cellular model of learning and memory. Conversely, altered spine morphology and plasticity are common hallmarks of human neurodevelopmental disorders, such as intellectual disability and autism. The advances in molecular and optical techniques have allowed for exploration of dynamic changes in structure and signal transduction at single-spine resolution, providing significant insights into the molecular regulation underlying spine structural plasticity. Here, I review recent findings on: how synaptic stimulation leads to diverse forms of spine structural plasticity; how the associated biochemical signals are initiated and transmitted into neuronal compartments; and how disruption of single genes associated with neurodevelopmental disorders can lead to abnormal spine structure in human and mouse brains. In particular, I discuss the functions of the Ras superfamily of small GTPases in spatiotemporal regulation of the actin cytoskeleton and protein synthesis in dendritic spines. Multiple lines of evidence implicate disrupted Ras signaling pathways in the spine structural abnormalities observed in neurodevelopmental disorders. Both deficient and excessive Ras activities lead to disrupted spine structure and deficits in learning and memory. Dysregulation of spine Ras signaling, therefore, may play a key role in the pathogenesis of multiple neurodevelopmental disorders with distinct etiologies.
Collapse
Affiliation(s)
- Jun Nishiyama
- Program in Neuroscience and Behavioral Disorders, Duke-National University of Singapore Medical School, Singapore
| |
Collapse
|
97
|
Adenosine A2A receptor as potential therapeutic target in neuropsychiatric disorders. Pharmacol Res 2019; 147:104338. [DOI: 10.1016/j.phrs.2019.104338] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 06/26/2019] [Accepted: 07/01/2019] [Indexed: 01/20/2023]
|
98
|
Cogram P, Deacon RMJ, Warner-Schmidt JL, von Schimmelmann MJ, Abrahams BS, During MJ. Gaboxadol Normalizes Behavioral Abnormalities in a Mouse Model of Fragile X Syndrome. Front Behav Neurosci 2019; 13:141. [PMID: 31293404 PMCID: PMC6603241 DOI: 10.3389/fnbeh.2019.00141] [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: 01/31/2019] [Accepted: 06/07/2019] [Indexed: 12/11/2022] Open
Abstract
Fragile X syndrome (FXS) is the most common inherited form of intellectual disability and autism. FXS is also accompanied by attention problems, hyperactivity, anxiety, aggression, poor sleep, repetitive behaviors, and self-injury. Recent work supports the role of γ-aminobutyric-acid (GABA), the primary inhibitory neurotransmitter in the brain, in mediating symptoms of FXS. Deficits in GABA machinery have been observed in a mouse model of FXS, including a loss of tonic inhibition in the amygdala, which is mediated by extrasynaptic GABAA receptors. Humans with FXS also show reduced GABAA receptor availability. Here, we sought to evaluate the potential of gaboxadol (also called OV101 and THIP), a selective and potent agonist for delta-subunit-containing extrasynaptic GABAA receptors (dSEGA), as a therapeutic agent for FXS by assessing its ability to normalize aberrant behaviors in a relatively uncharacterized mouse model of FXS (Fmr1 KO2 mice). Four behavioral domains (hyperactivity, anxiety, aggression, and repetitive behaviors) were probed using a battery of behavioral assays. The results showed that Fmr1 KO2 mice were hyperactive, had abnormal anxiety-like behavior, were more irritable and aggressive, and had an increased frequency of repetitive behaviors compared to wild-type (WT) littermates, which are all behavioral deficits reminiscent of individuals with FXS. Treatment with gaboxadol normalized all of the aberrant behaviors observed in Fmr1 KO2 mice back to WT levels, providing evidence of its potential benefit for treating FXS. We show that the potentiation of extrasynaptic GABA receptors alone, by gaboxadol, is sufficient to normalize numerous behavioral deficits in the FXS model using endpoints that are directly translatable to the clinical presentation of FXS. Taken together, these data support the future evaluation of gaboxadol in individuals with FXS, particularly with regard to symptoms of hyperactivity, anxiety, irritability, aggression, and repetitive behaviors.
Collapse
Affiliation(s)
- Patricia Cogram
- FRAXA-DVI, FRAXA Research Foundation, Boston, MA, United States.,Centre for Systems Biotechnology, Biomedicine Division, Fraunhofer-Gesellschaft, Santiago, Chile.,GEN.DDI Limited, London, United Kingdom.,Institute of Ecology and Biodiversity (IEB), University of Chile, Santiago, Chile
| | - Robert M J Deacon
- FRAXA-DVI, FRAXA Research Foundation, Boston, MA, United States.,Centre for Systems Biotechnology, Biomedicine Division, Fraunhofer-Gesellschaft, Santiago, Chile.,GEN.DDI Limited, London, United Kingdom.,Institute of Ecology and Biodiversity (IEB), University of Chile, Santiago, Chile
| | | | | | - Brett S Abrahams
- Ovid Therapeutics, New York, NY, United States.,Department of Genetics and Neuroscience, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Matthew J During
- Ovid Therapeutics, New York, NY, United States.,Department of Neurological Surgery and Molecular Virology, Immunology and Medical Genetics, Ohio State University College of Medicine, Columbus, OH, United States
| |
Collapse
|
99
|
Lovastatin, not Simvastatin, Corrects Core Phenotypes in the Fragile X Mouse Model. eNeuro 2019; 6:ENEURO.0097-19.2019. [PMID: 31147392 PMCID: PMC6565377 DOI: 10.1523/eneuro.0097-19.2019] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 05/11/2019] [Accepted: 05/13/2019] [Indexed: 02/07/2023] Open
Abstract
The cholesterol-lowering drug lovastatin corrects neurological phenotypes in animal models of fragile X syndrome (FX), a commonly identified genetic cause of autism and intellectual disability (ID). The therapeutic efficacy of lovastatin is being tested in clinical trials for FX; however, the structurally similar drug simvastatin has been proposed as an alternative due to an increased potency and brain penetrance. Here, we perform a side-by-side comparison of the effects of lovastatin and simvastatin treatment on two core phenotypes in Fmr1-/y mice versus WT littermates: excessive hippocampal protein synthesis and susceptibility to audiogenic seizures (AGSs). We find that simvastatin does not correct excessive hippocampal protein synthesis in the Fmr1-/y hippocampus at any dose tested. In fact, simvastatin significantly increases protein synthesis in both Fmr1-/y and WT. Moreover, injection of simvastatin does not reduce AGS in the Fmr1-/y mouse, while lovastatin significantly reduces AGS incidence and severity versus vehicle-treated animals. These results show that unlike lovastatin, simvastatin does not correct core phenotypes in the Fmr1-/y mouse model.
Collapse
|
100
|
Kinase pathway inhibition restores PSD95 induction in neurons lacking fragile X mental retardation protein. Proc Natl Acad Sci U S A 2019; 116:12007-12012. [PMID: 31118285 DOI: 10.1073/pnas.1812056116] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Fragile X syndrome (FXS) is the leading monogenic cause of autism and intellectual disability. FXS is caused by loss of expression of fragile X mental retardation protein (FMRP), an RNA-binding protein that regulates translation of numerous mRNA targets, some of which are present at synapses. While protein synthesis deficits have long been postulated as an etiology of FXS, how FMRP loss affects distributions of newly synthesized proteins is unknown. Here we investigated the role of FMRP in regulating expression of new copies of the synaptic protein PSD95 in an in vitro model of synaptic plasticity. We find that local BDNF application promotes persistent accumulation of new PSD95 at stimulated synapses and dendrites of cultured neurons, and that this accumulation is absent in FMRP-deficient mouse neurons. New PSD95 accumulation at sites of BDNF stimulation does not require known mechanisms regulating FMRP-mRNA interactions but instead requires the PI3K-mTORC1-S6K1 pathway. Surprisingly, in FMRP-deficient neurons, BDNF induction of new PSD95 accumulation can be restored by mTORC1-S6K1 blockade, suggesting that constitutively high mTORC1-S6K1 activity occludes PSD95 regulation by BDNF and that alternative pathways exist to mediate induction when mTORC1-S6K1 is inhibited. This study provides direct evidence for deficits in local protein synthesis and accumulation of newly synthesized protein in response to local stimulation in FXS, and supports mTORC1-S6K1 pathway inhibition as a potential therapeutic approach for FXS.
Collapse
|