51
|
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
|
52
|
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
|
53
|
Berry-Kravis EM, Harnett MD, Reines SA, Reese MA, Ethridge LE, Outterson AH, Michalak C, Furman J, Gurney ME. Inhibition of phosphodiesterase-4D in adults with fragile X syndrome: a randomized, placebo-controlled, phase 2 clinical trial. Nat Med 2021; 27:862-870. [PMID: 33927413 DOI: 10.1038/s41591-021-01321-w] [Citation(s) in RCA: 84] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 03/15/2021] [Indexed: 12/18/2022]
Abstract
The goal of this study was to determine whether a phosphodiesterase-4D (PDE4D) allosteric inhibitor (BPN14770) would improve cognitive function and behavioral outcomes in patients with fragile X syndrome (FXS). This phase 2 trial was a 24-week randomized, placebo-controlled, two-way crossover study in 30 adult male patients (age 18-41 years) with FXS. Participants received oral doses of BPN14770 25 mg twice daily or placebo. Primary outcomes were prespecified as safety and tolerability with secondary efficacy outcomes of cognitive performance, caregiver rating scales and physician rating scales (ClinicalTrials.gov identifier: NCT03569631 ). The study met the primary outcome measure since BPN14770 was well tolerated with no meaningful differences between the active and placebo treatment arms. The study also met key secondary efficacy measures of cognition and daily function. Cognitive benefit was demonstrated using the National Institutes of Health Toolbox Cognition Battery assessments of Oral Reading Recognition (least squares mean difference +2.81, P = 0.0157), Picture Vocabulary (+5.81, P = 0.0342) and Cognition Crystallized Composite score (+5.31, P = 0.0018). Benefit as assessed by visual analog caregiver rating scales was judged to be clinically meaningful for language (+14.04, P = 0.0051) and daily functioning (+14.53, P = 0.0017). Results from this study using direct, computer-based assessment of cognitive performance by adult males with FXS indicate significant cognitive improvement in domains related to language with corresponding improvement in caregiver scales rating language and daily functioning.
Collapse
Affiliation(s)
- Elizabeth M Berry-Kravis
- Department of Pediatrics, Neurological Sciences, and Biochemistry, Rush University Medical Center, Chicago, IL, USA.
| | | | | | - Melody A Reese
- Department of Psychology, University of Oklahoma, Norman, OK, USA
| | - Lauren E Ethridge
- Department of Psychology, University of Oklahoma, Norman, OK, USA.,Department of Pediatrics, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Abigail H Outterson
- Department of Pediatrics, Neurological Sciences, and Biochemistry, Rush University Medical Center, Chicago, IL, USA
| | - Claire Michalak
- Department of Pediatrics, Neurological Sciences, and Biochemistry, Rush University Medical Center, Chicago, IL, USA
| | - Jeremiah Furman
- Department of Pediatrics, Neurological Sciences, and Biochemistry, Rush University Medical Center, Chicago, IL, USA
| | | |
Collapse
|
54
|
Moskalyuk A, Van De Vijver S, Verstraelen P, De Vos WH, Kooy RF, Giugliano M. Single-Cell and Neuronal Network Alterations in an In Vitro Model of Fragile X Syndrome. Cereb Cortex 2021; 30:31-46. [PMID: 30958540 DOI: 10.1093/cercor/bhz068] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The Fragile X mental retardation protein (FMRP) is involved in many cellular processes and it regulates synaptic and network development in neurons. Its absence is known to lead to intellectual disability, with a wide range of comorbidities including autism. Over the past decades, FMRP research focused on abnormalities both in glutamatergic and GABAergic signaling, and an altered balance between excitation and inhibition has been hypothesized to underlie the clinical consequences of absence of the protein. Using Fmrp knockout mice, we studied an in vitro model of cortical microcircuitry and observed that the loss of FMRP largely affected the electrophysiological correlates of network development and maturation but caused less alterations in single-cell phenotypes. The loss of FMRP also caused a structural increase in the number of excitatory synaptic terminals. Using a mathematical model, we demonstrated that the combination of an increased excitation and reduced inhibition describes best our experimental observations during the ex vivo formation of the network connections.
Collapse
Affiliation(s)
- Anastasiya Moskalyuk
- Molecular, Cellular, and Network Excitability Lab, University of Antwerp, Wilrijk, Flanders, Belgium
| | - Sebastiaan Van De Vijver
- Molecular, Cellular, and Network Excitability Lab, University of Antwerp, Wilrijk, Flanders, Belgium
| | - Peter Verstraelen
- Laboratory of Cell Biology and Histology, University of Antwerp, Wilrijk, Flanders, Belgium
| | - Winnok H De Vos
- Laboratory of Cell Biology and Histology, University of Antwerp, Wilrijk, Flanders, Belgium
| | - R Frank Kooy
- Department of Medical Genetics, University of Antwerp, Edegem, Flanders, Belgium
| | - Michele Giugliano
- Molecular, Cellular, and Network Excitability Lab, University of Antwerp, Wilrijk, Flanders, Belgium.,International School for Advanced Studies (SISSA), Trieste, Italy
| |
Collapse
|
55
|
Bland KM, Aharon A, Widener EL, Song MI, Casey ZO, Zuo Y, Vidal GS. FMRP regulates the subcellular distribution of cortical dendritic spine density in a non-cell-autonomous manner. Neurobiol Dis 2021; 150:105253. [PMID: 33421563 PMCID: PMC7878418 DOI: 10.1016/j.nbd.2021.105253] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 12/15/2020] [Accepted: 01/04/2021] [Indexed: 01/18/2023] Open
Abstract
Fragile X syndrome (FXS) is the most common form of intellectual disability that arises from the dysfunction of a single gene-Fmr1. The main neuroanatomical correlate of FXS is elevated dendritic spine density on cortical pyramidal neurons, which has been modeled in Fmr1-/Y mice. However, the cell-autonomous contribution of Fmr1 on cortical dendritic spine density has not been assessed. Even less is known about the role of Fmr1 in heterozygous female mosaic mice, which are a putative model for human Fmr1 full mutation carriers (i.e., are heterozygous for the full Fmr1-silencing mutation). In this neuroanatomical study, spine density in cortical pyramidal neurons of Fmr1+/- and Fmr1-/Y mice was studied at multiple subcellular compartments, layers, and brain regions. Spine density in Fmr1+/- mice is higher than WT but lower than Fmr1-/Y. Not all subcellular compartments in layer V Fmr1+/- and Fmr1-/Y cortical pyramidal neurons are equally affected: the apical dendrite, a key subcellular compartment, is principally affected over basal dendrites. Within apical dendrites, spine density is differentially affected across branch orders. Finally, identification of FMRP-positive and FMRP-negative neurons within Fmr1+/- permitted the study of the cell-autonomous effect of Fmr1 on spine density. Surprisingly, layer V cortical pyramidal spine density between FMRP-positive and FMRP-negative neurons does not differ, suggesting that the regulation of the primary neuroanatomical defect of FXS-elevated spine density-is non-cell-autonomous.
Collapse
Affiliation(s)
- Katherine M Bland
- Department of Biology, James Madison University, Harrisonburg, VA 22801, United States
| | - Adam Aharon
- Department of Molecular, Cell and Developmental Biology, University of California Santa Cruz, Santa Cruz, CA 95064, United States
| | - Eden L Widener
- Department of Biology, James Madison University, Harrisonburg, VA 22801, United States
| | - M Irene Song
- Department of Biology, James Madison University, Harrisonburg, VA 22801, United States
| | - Zachary O Casey
- Department of Biology, James Madison University, Harrisonburg, VA 22801, United States
| | - Yi Zuo
- Department of Molecular, Cell and Developmental Biology, University of California Santa Cruz, Santa Cruz, CA 95064, United States.
| | - George S Vidal
- Department of Biology, James Madison University, Harrisonburg, VA 22801, United States.
| |
Collapse
|
56
|
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: 12] [Impact Index Per Article: 3.0] [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
|
57
|
Berry-Kravis E, Filipink RA, Frye RE, Golla S, Morris SM, Andrews H, Choo TH, Kaufmann WE. Seizures in Fragile X Syndrome: Associations and Longitudinal Analysis of a Large Clinic-Based Cohort. Front Pediatr 2021; 9:736255. [PMID: 35036394 PMCID: PMC8756611 DOI: 10.3389/fped.2021.736255] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2021] [Accepted: 10/07/2021] [Indexed: 11/15/2022] Open
Abstract
Fragile X syndrome (FXS), the most common inherited cause of intellectual disability, learning disability, and autism spectrum disorder, is associated with an increased prevalence of certain medical conditions including seizures. The goal of this study was to better understand seizures in individuals with FXS using the Fragile X Online Registry with Accessible Research Database, a multisite observational study initiated in 2012 involving FXS clinics in the Fragile X Clinic and Research Consortium. Seizure data were available for 1,607 participants, mostly male (77%) and white (74.5%). The overall prevalence of at least one seizure was 12%, with this rate being significantly higher in males than females (13.7 vs. 6.2%, p < 0.001). As compared to individuals with FXS without seizures, those with seizures were more likely to have autism spectrum disorder, current sleep apnea, later acquisition of expressive language, more severe intellectual disability, hyperactivity, irritability, and stereotyped movements. The mean age of seizure onset was 6.4 (SD 6.1) years of age with the great majority (>80%) having onset of seizures which was before 10. For those with epilepsy, about half (52%) had seizures for more than 3 years. This group was found to have greater cognitive and language impairment, but not behavioral disruptions, compared with those with seizures for <3 years. Antiepileptic drugs were more often used in males (60.6%) than females (34.8%), and females more often required more than one medication. The most commonly used anticonvulsants were oxcarbazepine, valproic acid, lamotrigine, and levetiracetam. The current study is the largest and first longitudinal study ever conducted to describe seizures in FXS. Overall, this study confirms previous reports of seizures in FXS and extends previous findings by further defining the cognitive and behavioral phenotype of those with epilepsy in FXS. Future studies should further investigate the natural history of seizures in FXS and the characteristics of seizures in FXS in adulthood.
Collapse
Affiliation(s)
- Elizabeth Berry-Kravis
- Department of Pediatrics, Rush University Medical Center, Chicago, IL, United States.,Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, United States
| | - Robyn A Filipink
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Richard E Frye
- Barrow Neurological Institute at Phoenix Children's Hospital, Phoenix, AZ, United States.,Department of Child Health, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, United States
| | - Sailaja Golla
- Division of Neurodevelopmental Medicine, Department of Neurology, Thompson Autism Center, Children's Hospital of California, University of Irvine, Orange, CA, United States
| | - Stephanie M Morris
- Division of Pediatric and Developmental Neurology, Department of Neurology, Washington University in St. Louis, St. Louis, MO, United States
| | - Howard Andrews
- Department of Biostatistics, Mailman School of Public Health, Columbia University Medical Center, New York, NY, United States
| | - Tse-Hwei Choo
- Department of Psychiatry, New York State Psychiatric Institute, Columbia University Medical Center, New York, NY, United States
| | | | | |
Collapse
|
58
|
Glutamatergic Dysfunction and Synaptic Ultrastructural Alterations in Schizophrenia and Autism Spectrum Disorder: Evidence from Human and Rodent Studies. Int J Mol Sci 2020; 22:ijms22010059. [PMID: 33374598 PMCID: PMC7793137 DOI: 10.3390/ijms22010059] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 12/15/2020] [Accepted: 12/22/2020] [Indexed: 12/12/2022] Open
Abstract
The correlation between dysfunction in the glutamatergic system and neuropsychiatric disorders, including schizophrenia and autism spectrum disorder, is undisputed. Both disorders are associated with molecular and ultrastructural alterations that affect synaptic plasticity and thus the molecular and physiological basis of learning and memory. Altered synaptic plasticity, accompanied by changes in protein synthesis and trafficking of postsynaptic proteins, as well as structural modifications of excitatory synapses, are critically involved in the postnatal development of the mammalian nervous system. In this review, we summarize glutamatergic alterations and ultrastructural changes in synapses in schizophrenia and autism spectrum disorder of genetic or drug-related origin, and briefly comment on the possible reversibility of these neuropsychiatric disorders in the light of findings in regular synaptic physiology.
Collapse
|
59
|
Banke TG, Barria A. Transient Enhanced GluA2 Expression in Young Hippocampal Neurons of a Fragile X Mouse Model. Front Synaptic Neurosci 2020; 12:588295. [PMID: 33343326 PMCID: PMC7745073 DOI: 10.3389/fnsyn.2020.588295] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 11/10/2020] [Indexed: 12/29/2022] Open
Abstract
AMPA-type glutamate receptors (AMPARs) are tetrameric ligand-gated channels made up of combinations of GluA1-4 subunits and play important roles in synaptic transmission and plasticity. Here, we have investigated the development of AMPAR-mediated synaptic transmission in the hippocampus of the Fmr1 knock-out (KO) mouse, a widely used model of Fragile X syndrome (FXS). FXS is the leading monogenic cause of intellectual disability and autism spectrum disorders (ASD) and it is considered a neurodevelopmental disorder. For that reason, we investigated synaptic properties and dendritic development in animals from an early stage when synapses are starting to form up to adulthood. We found that hippocampal CA1 pyramidal neurons in the Fmr1-KO mouse exhibit a higher AMPAR-NMDAR ratio early in development but reverses to normal values after P13. This increase was accompanied by a larger presence of the GluA2-subunit in synaptic AMPARs that will lead to altered Ca2+ permeability of AMPARs that could have a profound impact upon neural circuits, learning, and diseases. Following this, we found that young KO animals lack Long-term potentiation (LTP), a well-understood model of synaptic plasticity necessary for proper development of circuits, and exhibit an increased frequency of spontaneous miniature excitatory postsynaptic currents, a measure of synaptic density. Furthermore, post hoc morphological analysis of recorded neurons revealed altered dendritic branching in the KO group. Interestingly, all these anomalies are transitory and revert to normal values in older animals. Our data suggest that loss of FMRP during early development leads to temporary upregulation of the GluA2 subunit and this impacts synaptic plasticity and altering morphological dendritic branching.
Collapse
Affiliation(s)
- Tue G Banke
- Department of Physiology and Biophysics, University of Washington, Seattle, WA, United States
| | - Andres Barria
- Department of Physiology and Biophysics, University of Washington, Seattle, WA, United States
| |
Collapse
|
60
|
Feuge J, Scharkowski F, Michaelsen-Preusse K, Korte M. FMRP Modulates Activity-Dependent Spine Plasticity by Binding Cofilin1 mRNA and Regulating Localization and Local Translation. Cereb Cortex 2020; 29:5204-5216. [PMID: 30953439 DOI: 10.1093/cercor/bhz059] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Revised: 02/14/2019] [Accepted: 02/27/2019] [Indexed: 11/13/2022] Open
Abstract
Multiple variants of intellectual disability, e.g., the Fragile X Syndrome are associated with alterations in dendritic spine morphology, thereby pointing to dysregulated actin dynamics during development and processes of synaptic plasticity. Surprisingly, although the necessity of spine actin remodeling was demonstrated repeatedly, the importance and precise role of actin regulators is often undervalued. Here, we provide evidence that structural and functional plasticity are severely impaired after NMDAR-dependent LTP in the hippocampus of Fmr1 KO mice. We can link these defects to an aberrant activity-dependent regulation of Cofilin 1 (cof1) as activity-dependent modulations of local cof1 mRNA availability, local cof1 translation as well as total cof1 expression are impaired in the absence of FMRP. Finally, we can rescue activity-dependent structural plasticity in KO neurons by mimicking the regulation of cof1 observed in WT cells, thereby illustrating the potential of actin modulators to provide novel treatment strategies for the Fragile X Syndrome.
Collapse
Affiliation(s)
- Jonas Feuge
- Division of Cellular Neurobiology, Zoological Institute, TU Braunschweig, Germany
| | | | | | - Martin Korte
- Division of Cellular Neurobiology, Zoological Institute, TU Braunschweig, Germany.,Helmholtz Center for Infection Research, Research group Neuroinflammation and Neurodegeneration, Braunschweig, Germany
| |
Collapse
|
61
|
Rajaratnam A, Potter LA, Biag HMB, Schneider A, Petrasic IC, Hagerman RJ. Review of Autism Profiles and Response to Sertraline in Fragile X Syndrome-Associated Autism vs. Non-syndromic Autism; Next Steps for Targeted Treatment. Front Neurol 2020; 11:581429. [PMID: 33193037 PMCID: PMC7661746 DOI: 10.3389/fneur.2020.581429] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 08/31/2020] [Indexed: 01/17/2023] Open
Abstract
Given significant genetic, molecular, and phenotypic overlaps, researchers have begun to investigate whether targeted treatments for Fragile X Syndrome (FXS) could also be beneficial for patients with Autism Spectrum Disorder (ASD). For example, low-dose sertraline, an SSRI, was used in two recent controlled trials in children with FXS and ASD. The first trial recruited 52 children with FXS, 32 of which were also diagnosed with ASD; the second trial recruited 58 children with non-syndromic ASD. One focus of the present study is to compare the response to sertraline between the FXS-associated ASD and non-syndromic ASD groups. Another focus is to compare baseline ASD-related characteristics between the groups and review these differences within the context of recent literature comparing these populations. Our comparison showed more severe ASD profiles in children with non-syndromic ASD vs. FXS-associated ASD. Regarding response to sertraline, the FXS-ASD group displayed significant improvements in language development, while the non-syndromic group did not show any significant improvements. One possible explanation for this differential response is the distinct anxiety profiles that are seen in these two groups. The heightened anxiety phenotype seen in those with FXS-ASD may have led to a greater relief of anxiety symptoms with sertraline compared to those with non-syndromic ASD; this, in turn, could have led to measurably greater developmental gains. Further research is required to solidify this connection between anxiety relief and developmental gains in these populations.
Collapse
Affiliation(s)
- Akash Rajaratnam
- MIND Institute, University of California, Davis, Sacramento, CA, United States.,School of Medicine, Case Western Reserve University, Cleveland, OH, United States
| | | | | | - Andrea Schneider
- MIND Institute, University of California, Davis, Sacramento, CA, United States
| | - Ignacio Cortina Petrasic
- MIND Institute, University of California, Davis, Sacramento, CA, United States.,School of Medicine, University of California, Davis, Sacramento, CA, United States
| | - Randi Jenssen Hagerman
- MIND Institute, University of California, Davis, Sacramento, CA, United States.,Department of Pediatrics, University of California Davis Health, Sacramento, CA, United States
| |
Collapse
|
62
|
Cogram P, Alkon DL, Crockford D, Deacon RMJ, Hurley MJ, Altimiras F, Sun MK, Tranfaglia M. Chronic bryostatin-1 rescues autistic and cognitive phenotypes in the fragile X mice. Sci Rep 2020; 10:18058. [PMID: 33093534 PMCID: PMC7581799 DOI: 10.1038/s41598-020-74848-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Accepted: 10/05/2020] [Indexed: 12/20/2022] Open
Abstract
Fragile X syndrome (FXS), an X-chromosome linked intellectual disability, is the leading monogenetic cause of autism spectrum disorder (ASD), a neurodevelopmental condition that currently has no specific drug treatment. Building upon the demonstrated therapeutic effects on spatial memory of bryostatin-1, a relatively specific activator of protein kinase C (PKC)ε, (also of PKCα) on impaired synaptic plasticity/maturation and spatial learning and memory in FXS mice, we investigated whether bryostatin-1 might affect the autistic phenotypes and other behaviors, including open field activity, activities of daily living (nesting and marble burying), at the effective therapeutic dose for spatial memory deficits. Further evaluation included other non-spatial learning and memory tasks. Interestingly, a short period of treatment (5 weeks) only produced very limited or no therapeutic effects on the autistic and cognitive phenotypes in the Fmr1 KO2 mice, while a longer treatment (13 weeks) with the same dose of bryostatin-1 effectively rescued the autistic and non-spatial learning deficit cognitive phenotypes. It is possible that longer-term treatment would result in further improvement in these fragile X phenotypes. This effect is clearly different from other treatment strategies tested to date, in that the drug shows little acute effect, but strong long-term effects. It also shows no evidence of tolerance, which has been a problem with other drug classes (mGluR5 antagonists, GABA-A and -B agonists). The results strongly suggest that, at appropriate dosing and therapeutic period, chronic bryostatin-1 may have great therapeutic value for both ASD and FXS.
Collapse
Affiliation(s)
- Patricia Cogram
- FRAXA-DVI, FRAXA, Santiago, Chile. .,IEB, Faculty of Science, University of Chile, Santiago, Chile.
| | | | | | - Robert M J Deacon
- FRAXA-DVI, FRAXA, Santiago, Chile.,IEB, Faculty of Science, University of Chile, Santiago, Chile
| | - Michael J Hurley
- Neuroimmunology, Biological Sciences, Faculty of Environmental and Life Sciences, University of Southampton, Southampton, UK
| | - Francisco Altimiras
- Faculty of Engineering, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile.,Faculty of Engineering and Business, Universidad de las Américas, Santiago, Chile
| | | | | |
Collapse
|
63
|
Dong E, Pandey SC. Prenatal stress induced chromatin remodeling and risk of psychopathology in adulthood. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2020; 156:185-215. [PMID: 33461663 PMCID: PMC7864549 DOI: 10.1016/bs.irn.2020.08.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
New insights into the pathophysiology of psychiatric disorders suggest the existence of a complex interplay between genetics and environment. This notion is supported by evidence suggesting that exposure to stress during pregnancy exerts profound effects on the neurodevelopment and behavior of the offspring and predisposes them to psychiatric disorders later in life. Accumulated evidence suggests that vulnerability to psychiatric disorders may result from permanent negative effects of long-term changes in synaptic plasticity due to altered epigenetic mechanisms (histone modifications and DNA methylation) that lead to condensed chromatin architecture, thereby decreasing the expression of candidate genes during early brain development. In this chapter, we have summarized the literature of clinical studies on psychiatric disorders induced by maternal stress during pregnancy. We also discussed the epigenetic alterations of gene regulations induced by prenatal stress. Because the clinical manifestations of psychiatric disorders are complex, it is obvious that the biological progression of these diseases cannot be studied only in postmortem brains of patients and the use of animal models is required. Therefore, in this chapter, we have introduced a well-established mouse model of prenatal stress (PRS) generated in restrained pregnant dams. The behavioral phenotypes of the offspring (PRS mice) born to the stressed dam and underlying epigenetic changes in key molecules related to synaptic activity were described and highlighted. PRS mice may serve as a useful model for investigating the pathogenesis of psychiatric disorders and may be a useful tool for screening for the potential compounds that may normalize aberrant epigenetic mechanisms induced by prenatal stress.
Collapse
Affiliation(s)
- Erbo Dong
- Center for Alcohol Research in Epigenetics, Department of Psychiatry, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States.
| | - Subhash C Pandey
- Center for Alcohol Research in Epigenetics, Department of Psychiatry, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States; Jesse Brown VA Medical Center, Chicago, IL, United States
| |
Collapse
|
64
|
Huebschman JL, Corona KS, Guo Y, Smith LN. The Fragile X Mental Retardation Protein Regulates Striatal Medium Spiny Neuron Synapse Density and Dendritic Spine Morphology. Front Mol Neurosci 2020; 13:161. [PMID: 33013316 PMCID: PMC7511717 DOI: 10.3389/fnmol.2020.00161] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 08/05/2020] [Indexed: 01/23/2023] Open
Abstract
The fragile X mental retardation protein (FMRP), an RNA-binding protein that mediates the transport, stability, and translation of hundreds of brain RNAs, is critically involved in regulating synaptic function. Loss of FMRP, as in fragile X syndrome (FXS), is a leading monogenic cause of autism and results in altered structural and functional synaptic plasticity, widely described in the hippocampus and cortex. Though FXS is associated with hyperactivity, impaired social interaction, and the development of repetitive or stereotyped behaviors, all of which are influenced by striatal activity, few studies have investigated the function of FMRP here. Utilizing a cortical-striatal co-culture model, we find that striatal medium spiny neurons (MSNs) lacking FMRP fail to make normal increases in PSD95 expression over a short time period and have significant deficits in dendritic spine density and colocalized synaptic puncta at the later measured time point compared to wildtype (WT) MSNs. Acute expression of wtFMRP plasmid in Fmr1 KO co-cultures results in contrasting outcomes for these measures on MSNs at the more mature time point, reducing spine density across multiple spine types but making no significant changes in colocalized puncta. FMRP’s KH2 and RGG RNA-binding domains are required for normal elimination of PSD95, and interruption of these domains slightly favors elimination of immature spine types. Further, KH2 is required for normal levels of colocalized puncta. Our data are largely consistent with a basal role for FMRP and its RNA-binding domains in striatal synapse stabilization on developing MSNs, and in light of previous findings, suggest distinct regional and/or cell type-specific roles for FMRP in regulating synapse structure.
Collapse
Affiliation(s)
- Jessica L Huebschman
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University Health Science Center, Bryan, TX, United States.,Texas A&M Institute for Neuroscience, Texas A&M University, College Station, TX, United States
| | - Kitzia S Corona
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University Health Science Center, Bryan, TX, United States
| | - Yuhong Guo
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University Health Science Center, Bryan, TX, United States
| | - Laura N Smith
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University Health Science Center, Bryan, TX, United States.,Texas A&M Institute for Neuroscience, Texas A&M University, College Station, TX, United States
| |
Collapse
|
65
|
Ostrovskaya OI, Cao G, Eroglu C, Harris KM. Developmental onset of enduring long-term potentiation in mouse hippocampus. Hippocampus 2020; 30:1298-1312. [PMID: 32894631 DOI: 10.1002/hipo.23257] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 07/18/2020] [Accepted: 08/13/2020] [Indexed: 11/10/2022]
Abstract
Analysis of long-term potentiation (LTP) provides a powerful window into cellular mechanisms of learning and memory. Prior work shows late LTP (L-LTP), lasting >3 hr, occurs abruptly at postnatal day 12 (P12) in the stratum radiatum of rat hippocampal area CA1. The goal here was to determine the developmental profile of synaptic plasticity leading to L-LTP in the mouse hippocampus. Two mouse strains and two mutations known to affect synaptic plasticity were chosen: C57BL/6J and Fmr1-/y on the C57BL/6J background, and 129SVE and Hevin-/- (Sparcl1-/- ) on the 129SVE background. Like rats, hippocampal slices from all of the mice showed test pulse-induced depression early during development that was gradually resolved with maturation by 5 weeks. All the mouse strains showed a gradual progression between P10-P35 in the expression of short-term potentiation (STP), lasting ≤1 hr. In the 129SVE mice, L-LTP onset (>25% of slices) occurred by 3 weeks, reliable L-LTP (>50% slices) was achieved by 4 weeks, and Hevin-/- advanced this profile by 1 week. In the C57BL/6J mice, L-LTP onset occurred significantly later, over 3-4 weeks, and reliability was not achieved until 5 weeks. Although some of the Fmr1-/y mice showed L-LTP before 3 weeks, reliable L-LTP also was not achieved until 5 weeks. L-LTP onset was not advanced in any of the mouse genotypes by multiple bouts of theta-burst stimulation at 90 or 180 min intervals. These findings show important species differences in the onset of STP and L-LTP, which occur at the same age in rats but are sequentially acquired in mice.
Collapse
Affiliation(s)
- Olga I Ostrovskaya
- Department of Neuroscience, Center for Learning and Memory, The University of Texas at Austin, Austin, Texas, USA
| | - Guan Cao
- Department of Neuroscience, Center for Learning and Memory, The University of Texas at Austin, Austin, Texas, USA
| | - Cagla Eroglu
- Department of Cell Biology, Duke University Medical Center, Durham, North Carolina, USA.,Department of Neurobiology Regeneration Next Initiative, Duke University Medical Center, Durham, North Carolina, USA
| | - Kristen M Harris
- Department of Neuroscience, Center for Learning and Memory, The University of Texas at Austin, Austin, Texas, USA
| |
Collapse
|
66
|
Twenty Years of SynGAP Research: From Synapses to Cognition. J Neurosci 2020; 40:1596-1605. [PMID: 32075947 DOI: 10.1523/jneurosci.0420-19.2020] [Citation(s) in RCA: 95] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Revised: 01/03/2020] [Accepted: 01/07/2020] [Indexed: 12/11/2022] Open
Abstract
SynGAP is a potent regulator of biochemical signaling in neurons and plays critical roles in neuronal function. It was first identified in 1998, and has since been extensively characterized as a mediator of synaptic plasticity. Because of its involvement in synaptic plasticity, SynGAP has emerged as a critical protein for normal cognitive function. In recent years, mutations in the SYNGAP1 gene have been shown to cause intellectual disability in humans and have been linked to other neurodevelopmental disorders, such as autism spectrum disorders and schizophrenia. While the structure and biochemical function of SynGAP have been well characterized, a unified understanding of the various roles of SynGAP at the synapse and its contributions to neuronal function remains to be achieved. In this review, we summarize and discuss the current understanding of the multifactorial role of SynGAP in regulating neuronal function gathered over the last two decades.
Collapse
|
67
|
Licznerski P, Park HA, Rolyan H, Chen R, Mnatsakanyan N, Miranda P, Graham M, Wu J, Cruz-Reyes N, Mehta N, Sohail S, Salcedo J, Song E, Effman C, Effman S, Brandao L, Xu GN, Braker A, Gribkoff VK, Levy RJ, Jonas EA. ATP Synthase c-Subunit Leak Causes Aberrant Cellular Metabolism in Fragile X Syndrome. Cell 2020; 182:1170-1185.e9. [PMID: 32795412 DOI: 10.1016/j.cell.2020.07.008] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 03/04/2020] [Accepted: 07/10/2020] [Indexed: 12/26/2022]
Abstract
Loss of the gene (Fmr1) encoding Fragile X mental retardation protein (FMRP) causes increased mRNA translation and aberrant synaptic development. We find neurons of the Fmr1-/y mouse have a mitochondrial inner membrane leak contributing to a "leak metabolism." In human Fragile X syndrome (FXS) fibroblasts and in Fmr1-/y mouse neurons, closure of the ATP synthase leak channel by mild depletion of its c-subunit or pharmacological inhibition normalizes stimulus-induced and constitutive mRNA translation rate, decreases lactate and key glycolytic and tricarboxylic acid (TCA) cycle enzyme levels, and triggers synapse maturation. FMRP regulates leak closure in wild-type (WT), but not FX synapses, by stimulus-dependent ATP synthase β subunit translation; this increases the ratio of ATP synthase enzyme to its c-subunit, enhancing ATP production efficiency and synaptic growth. In contrast, in FXS, inability to close developmental c-subunit leak prevents stimulus-dependent synaptic maturation. Therefore, ATP synthase c-subunit leak closure encourages development and attenuates autistic behaviors.
Collapse
Affiliation(s)
- Pawel Licznerski
- Department of Internal Medicine, Section of Endocrinology, Yale University School of Medicine, New Haven, CT 06511, USA.
| | - Han-A Park
- Department of Internal Medicine, Section of Endocrinology, Yale University School of Medicine, New Haven, CT 06511, USA; Department of Human Nutrition and Hospitality Management, College of Human Environmental Sciences, The University of Alabama, Tuscaloosa, AL 35487, USA
| | - Harshvardhan Rolyan
- Department of Internal Medicine, Section of Endocrinology, Yale University School of Medicine, New Haven, CT 06511, USA
| | - Rongmin Chen
- Department of Internal Medicine, Section of Endocrinology, Yale University School of Medicine, New Haven, CT 06511, USA
| | - Nelli Mnatsakanyan
- Department of Internal Medicine, Section of Endocrinology, Yale University School of Medicine, New Haven, CT 06511, USA
| | - Paige Miranda
- Department of Internal Medicine, Section of Endocrinology, Yale University School of Medicine, New Haven, CT 06511, USA
| | - Morven Graham
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Jing Wu
- Department of Internal Medicine, Section of Endocrinology, Yale University School of Medicine, New Haven, CT 06511, USA
| | | | - Nikita Mehta
- Marine Biological Laboratory, Woods Hole, MA 02543, USA
| | - Sana Sohail
- Marine Biological Laboratory, Woods Hole, MA 02543, USA
| | - Jorge Salcedo
- Marine Biological Laboratory, Woods Hole, MA 02543, USA
| | - Erin Song
- Marine Biological Laboratory, Woods Hole, MA 02543, USA
| | | | - Samuel Effman
- Marine Biological Laboratory, Woods Hole, MA 02543, USA
| | - Lucas Brandao
- Department of Biology, Clark University, Worcester, MA 01610, USA
| | - Gulan N Xu
- Department of Internal Medicine, Section of Endocrinology, Yale University School of Medicine, New Haven, CT 06511, USA
| | - Amber Braker
- Department of Internal Medicine, Section of Endocrinology, Yale University School of Medicine, New Haven, CT 06511, USA
| | - Valentin K Gribkoff
- Department of Internal Medicine, Section of Endocrinology, Yale University School of Medicine, New Haven, CT 06511, USA; Marine Biological Laboratory, Woods Hole, MA 02543, USA
| | - Richard J Levy
- Department of Anesthesiology, Columbia University Medical Center, New York, NY 10032, USA
| | - Elizabeth A Jonas
- Department of Internal Medicine, Section of Endocrinology, Yale University School of Medicine, New Haven, CT 06511, USA; Marine Biological Laboratory, Woods Hole, MA 02543, USA.
| |
Collapse
|
68
|
Kennedy T, Rinker D, Broadie K. Genetic background mutations drive neural circuit hyperconnectivity in a fragile X syndrome model. BMC Biol 2020; 18:94. [PMID: 32731855 PMCID: PMC7392683 DOI: 10.1186/s12915-020-00817-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Accepted: 06/19/2020] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND Neural circuits are initially assembled during development when neurons synapse with potential partners and later refined as appropriate connections stabilize into mature synapses while inappropriate contacts are eliminated. Disruptions to this synaptogenic process impair connectivity optimization and can cause neurodevelopmental disorders. Intellectual disability (ID) and autism spectrum disorder (ASD) are often characterized by synaptic overgrowth, with the maintenance of immature or inappropriate synapses. Such synaptogenic defects can occur through mutation of a single gene, such as fragile X mental retardation protein (FMRP) loss causing the neurodevelopmental disorder fragile X syndrome (FXS). FXS represents the leading heritable cause of ID and ASD, but many other genes that play roles in ID and ASD have yet to be identified. RESULTS In a Drosophila FXS disease model, one dfmr150M null mutant stock exhibits previously unreported axonal overgrowths at developmental and mature stages in the giant fiber (GF) escape circuit. These excess axon projections contain both chemical and electrical synapse markers, indicating mixed synaptic connections. Extensive analyses show these supernumerary synapses connect known GF circuit neurons, rather than new, inappropriate partners, indicating hyperconnectivity within the circuit. Despite the striking similarities to well-characterized FXS synaptic defects, this new GF circuit hyperconnectivity phenotype is driven by genetic background mutations in this dfmr150M stock. Similar GF circuit synaptic overgrowth is not observed in independent dfmr1 null alleles. Bulked segregant analysis (BSA) was combined with whole genome sequencing (WGS) to identify the quantitative trait loci (QTL) linked to neural circuit hyperconnectivity. The results reveal 8 QTL associated with inappropriate synapse formation and maintenance in the dfmr150M mutant background. CONCLUSIONS Synaptogenesis is a complex, precisely orchestrated neurodevelopmental process with a large cohort of gene products coordinating the connectivity, synaptic strength, and excitatory/inhibitory balance between neuronal partners. This work identifies a number of genetic regions that contain mutations disrupting proper synaptogenesis within a particularly well-mapped neural circuit. These QTL regions contain potential new genes involved in synapse formation and refinement. Given the similarity of the synaptic overgrowth phenotype to known ID and ASD inherited conditions, identifying these genes should increase our understanding of these devastating neurodevelopmental disease states.
Collapse
Affiliation(s)
- Tyler Kennedy
- Department of Biological Sciences, Vanderbilt University and Medical Center, Nashville, TN, 37235, USA
| | - David Rinker
- Department of Biological Sciences, Vanderbilt University and Medical Center, Nashville, TN, 37235, USA
| | - Kendal Broadie
- Department of Biological Sciences, Vanderbilt University and Medical Center, Nashville, TN, 37235, USA.
- Department of Cell and Developmental Biology, Vanderbilt University and Medical Center, Nashville, TN, 37235, USA.
- Vanderbilt Brain Institute, Vanderbilt University and Medical Center, Nashville, TN, 37235, USA.
| |
Collapse
|
69
|
Seese RR. Working Memory Impairments in Cerebellar Disorders of Childhood. Pediatr Neurol 2020; 107:16-23. [PMID: 32276741 DOI: 10.1016/j.pediatrneurol.2020.02.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2019] [Revised: 01/17/2020] [Accepted: 02/19/2020] [Indexed: 12/20/2022]
Abstract
The cerebellum is a crucial center for motor control and integration. Increasing evidence supports the notion that the cerebellum is also involved in nonmotor functions. Along these lines, multiple cerebellar disorders of childhood and adulthood are associated with behavioral and cognitive symptoms, including impairments in memory. One form of memory commonly affected in cerebellar disorders is working memory, which uses attention to manipulate information that is immediately available to execute cognitive tasks. This article reviews the literature illustrating that working memory impairments are frequently observed in acquired, congenital, and genetic/developmental cerebellar disorders of childhood. Functional neuroimaging studies demonstrate that working memory tasks engage many posterior regions of the cerebellar hemispheres and vermis. Thus, the cerebellum acts as one important node in the working memory circuit, and when the cerebellum is involved in childhood disorders, deficits in working memory commonly occur.
Collapse
Affiliation(s)
- Ronald R Seese
- Division of Child Neurology, Department of Pediatrics, UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania.
| |
Collapse
|
70
|
Das Sharma S, Metz JB, Li H, Hobson BD, Hornstein N, Sulzer D, Tang G, Sims PA. Widespread Alterations in Translation Elongation in the Brain of Juvenile Fmr1 Knockout Mice. Cell Rep 2020; 26:3313-3322.e5. [PMID: 30893603 DOI: 10.1016/j.celrep.2019.02.086] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Revised: 08/27/2018] [Accepted: 02/21/2019] [Indexed: 12/18/2022] Open
Abstract
FMRP (fragile X mental retardation protein) is a polysome-associated RNA-binding protein encoded by Fmr1 that is lost in fragile X syndrome. Increasing evidence suggests that FMRP regulates both translation initiation and elongation, but the gene specificity of these effects is unclear. To elucidate the impact of Fmr1 loss on translation, we utilize ribosome profiling for genome-wide measurements of ribosomal occupancy and positioning in the cortex of 24-day-old Fmr1 knockout mice. We find a remarkably coherent reduction in ribosome footprint abundance per mRNA for previously identified, high-affinity mRNA binding partners of FMRP and an increase for terminal oligopyrimidine (TOP) motif-containing genes canonically controlled by mammalian target of rapamycin-eIF4E-binding protein-eIF4E binding protein-eukaryotic initiation factor 4E (mTOR-4E-BP-eIF4E) signaling. Amino acid motif- and gene-level analyses both show a widespread reduction of translational pausing in Fmr1 knockout mice. Our findings are consistent with a model of FMRP-mediated regulation of both translation initiation through eIF4E and elongation that is disrupted in fragile X syndrome.
Collapse
Affiliation(s)
- Sohani Das Sharma
- Department of Systems Biology, Columbia University Medical Center, New York, NY 10032, USA
| | - Jordan B Metz
- Department of Systems Biology, Columbia University Medical Center, New York, NY 10032, USA; Medical Scientist Training Program, Columbia University Medical Center, New York, NY 10032, USA
| | - Hongyu Li
- Department of Neurology, Columbia University Medical Center, New York, NY 10032, USA
| | - Benjamin D Hobson
- Department of Systems Biology, Columbia University Medical Center, New York, NY 10032, USA; Medical Scientist Training Program, Columbia University Medical Center, New York, NY 10032, USA
| | - Nicholas Hornstein
- Department of Systems Biology, Columbia University Medical Center, New York, NY 10032, USA; Medical Scientist Training Program, Columbia University Medical Center, New York, NY 10032, USA
| | - David Sulzer
- Department of Neurology, Columbia University Medical Center, New York, NY 10032, USA; Department of Psychiatry, Columbia University Medical Center, New York, NY 10032, USA; Department of Pharmacology, Columbia University Medical Center, New York, NY 10032, USA; Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY 10032, USA
| | - Guomei Tang
- Department of Neurology, Columbia University Medical Center, New York, NY 10032, USA
| | - Peter A Sims
- Department of Systems Biology, Columbia University Medical Center, New York, NY 10032, USA; Department of Biochemistry & Molecular Biophysics, Columbia University Medical Center, New York, NY 10032, USA; Sulzberger Columbia Genome Center, Columbia University Medical Center, New York, NY 10032, USA.
| |
Collapse
|
71
|
Cheyne JE, Montgomery JM. The cellular and molecular basis of in vivo synaptic plasticity in rodents. Am J Physiol Cell Physiol 2020; 318:C1264-C1283. [PMID: 32320288 DOI: 10.1152/ajpcell.00416.2019] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Plasticity within the neuronal networks of the brain underlies the ability to learn and retain new information. The initial discovery of synaptic plasticity occurred by measuring synaptic strength in vivo, applying external stimulation and observing an increase in synaptic strength termed long-term potentiation (LTP). Many of the molecular pathways involved in LTP and other forms of synaptic plasticity were subsequently uncovered in vitro. Over the last few decades, technological advances in recording and imaging in live animals have seen many of these molecular mechanisms confirmed in vivo, including structural changes both pre- and postsynaptically, changes in synaptic strength, and changes in neuronal excitability. A well-studied aspect of neuronal plasticity is the capacity of the brain to adapt to its environment, gained by comparing the brains of deprived and experienced animals in vivo, and in direct response to sensory stimuli. Multiple in vivo studies have also strongly linked plastic changes to memory by interfering with the expression of plasticity and by manipulating memory engrams. Plasticity in vivo also occurs in the absence of any form of external stimulation, i.e., during spontaneous network activity occurring with brain development. However, there is still much to learn about how plasticity is induced during natural learning and how this is altered in neurological disorders.
Collapse
Affiliation(s)
- Juliette E Cheyne
- Department of Physiology and Centre for Brain Research, University of Auckland, Auckland, New Zealand
| | - Johanna M Montgomery
- Department of Physiology and Centre for Brain Research, University of Auckland, Auckland, New Zealand
| |
Collapse
|
72
|
Costa JF, Dines M, Lamprecht R. The Role of Rac GTPase in Dendritic Spine Morphogenesis and Memory. Front Synaptic Neurosci 2020; 12:12. [PMID: 32362820 PMCID: PMC7182350 DOI: 10.3389/fnsyn.2020.00012] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Accepted: 03/04/2020] [Indexed: 11/21/2022] Open
Abstract
The ability to form memories in the brain is needed for daily functions, and its impairment is associated with human mental disorders. Evidence indicates that long-term memory (LTM)-related processes such as its consolidation, extinction and forgetting involve changes of synaptic efficacy produced by alterations in neural transmission and morphology. Modulation of the morphology and number of dendritic spines has been proposed to contribute to changes in neuronal transmission mediating such LTM-related processes. Rac GTPase activity is regulated by synaptic activation and it can affect spine morphology by controlling actin-regulatory proteins. Recent evidence shows that changes in Rac GTPase activity affect memory consolidation, extinction, erasure and forgetting and can affect spine morphology in brain areas that mediate these behaviors. Altered Rac GTPase activity is associated with abnormal spine morphology and brain disorders. By affecting Rac GTPase activity we can further understand the roles of spine morphogenesis in memory. Moreover, manipulation of Rac GTPase activity may serve as a therapeutic tool for the treatment of memory-related brain diseases.
Collapse
Affiliation(s)
| | | | - Raphael Lamprecht
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel
| |
Collapse
|
73
|
Hu J, Chen L, Yin J, Yin H, Huang Y, Tian J. Hyperactivity, Memory Defects, and Craniofacial Abnormalities in Zebrafish fmr1 Mutant Larvae. Behav Genet 2020; 50:152-160. [PMID: 32048109 DOI: 10.1007/s10519-020-09995-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2019] [Accepted: 02/04/2020] [Indexed: 01/05/2023]
Abstract
Fragile X syndrome (FXS) is a heritable mental retardation disease caused by unstable trinucleotide repeat sequences in FMR1. FXS is characterized by delayed development, hyperactivity, and autism behavior. Zebrafish is an excellent model to study FXS and the underlying function of fmr1. However, at present, fmr1 function is mainly studied via morpholinos or generated mutants using targeting induced local lesions in genomes. However, both of these methods generate off-target effects, making them suboptimal techniques for studying FXS. In this study, CRISPR/Cas9 technology was used to generate two zebrafish fmr1 mutant lines. High-throughput behavior analysis, qRT-PCR, and alcian blue staining experiments were employed to investigate fmr1 function. The fmr1 mutant line showed abnormal behavior, learning memory defects, and impaired craniofacial cartilage development. These features are similar to the human FXS phenotype, indicating that the fmr1 mutant generated in this study can be used as a new model for studying the molecular pathology of FXS. It also provides a suitable model for high-throughput screening of small molecule drugs for FXS therapeutics.
Collapse
Affiliation(s)
- Jia Hu
- School of Biology & Basic Medical Sciences, Medical College, Soochow University, Suzhou, 215123, Jiangsu, People's Republic of China
| | - Lei Chen
- Department of Medical Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, 215000, Jiangsu, People's Republic of China
| | - Jian Yin
- CAS Key Lab of Bio-medical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, 215163, Jiangsu, People's Republic of China
| | - Huancai Yin
- CAS Key Lab of Bio-medical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, 215163, Jiangsu, People's Republic of China
| | - Yinong Huang
- Shaanxi Institute of Pediatric Diseases, Xi'an Children's Hospital, Xi'an, 710003, Shaanxi, People's Republic of China.
| | - Jingjing Tian
- CAS Key Lab of Bio-medical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, 215163, Jiangsu, People's Republic of China.
- Academy for Engineering & Technology, Fudan University, Shanghai, 200433, People's Republic of China.
| |
Collapse
|
74
|
Kulinich AO, Reinhard SM, Rais M, Lovelace JW, Scott V, Binder DK, Razak KA, Ethell IM. Beneficial effects of sound exposure on auditory cortex development in a mouse model of Fragile X Syndrome. Neurobiol Dis 2020; 134:104622. [PMID: 31698054 DOI: 10.1016/j.nbd.2019.104622] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 08/22/2019] [Accepted: 09/23/2019] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Fragile X syndrome (FXS) is the most common genetic cause of autism and intellectual disability. Fragile X mental retardation gene (Fmr1) knock-out (KO) mice display core deficits of FXS, including abnormally increased sound-evoked responses, and show a delayed development of parvalbumin (PV) cells. Here, we present the surprising result that sound exposure during early development reduces correlates of auditory hypersensitivity in Fmr1 KO mice. METHODS Fmr1 KO and wild-type (WT) mice were raised in a sound-attenuated environment (AE) or sound-exposed (SE) to 14 kHz tones (5 Hz repetition rate) from P9 until P21. At P21-P23, event-related potentials (ERPs), dendritic spine density, PV expression and phosphorylation of tropomyosin receptor kinase B (TrkB) were analyzed in the auditory cortex of AE and SE mice. RESULTS Enhanced N1 amplitude of ERPs, impaired PV cell development, and increased spine density in layers (L) 2/3 and L5/6 excitatory neurons were observed in AE Fmr1 KO compared to WT mice. In contrast, developmental sound exposure normalized ERP N1 amplitude, density of PV cells and dendritic spines in SE Fmr1 KO mice. Finally, TrkB phosphorylation was reduced in AE Fmr1 KO, but was enhanced in SE Fmr1 KO mice, suggesting that BDNF-TrkB signaling may be regulated by sound exposure to influence PV cell development. CONCLUSIONS Our results demonstrate that sound exposure, but not attenuation, during early developmental window restores molecular, cellular and functional properties in the auditory cortex of Fmr1 KO mice, and suggest this approach as a potential treatment for sensory phenotypes in FXS.
Collapse
Affiliation(s)
- Anna O Kulinich
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA, USA
| | - Sarah M Reinhard
- Psychology Department, University of California, Riverside, CA, USA
| | - Maham Rais
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA, USA; Biomedical Sciences Graduate Program, University of California, Riverside, CA, USA
| | | | - Veronica Scott
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA, USA
| | - Devin K Binder
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA, USA; Neuroscience Graduate Program, University of California, Riverside, CA, USA; Biomedical Sciences Graduate Program, University of California, Riverside, CA, USA
| | - Khaleel A Razak
- Psychology Department, University of California, Riverside, CA, USA; Neuroscience Graduate Program, University of California, Riverside, CA, USA; Biomedical Sciences Graduate Program, University of California, Riverside, CA, USA.
| | - Iryna M Ethell
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA, USA; Neuroscience Graduate Program, University of California, Riverside, CA, USA; Biomedical Sciences Graduate Program, University of California, Riverside, CA, USA.
| |
Collapse
|
75
|
Sawicka K, Hale CR, Park CY, Fak JJ, Gresack JE, Van Driesche SJ, Kang JJ, Darnell JC, Darnell RB. FMRP has a cell-type-specific role in CA1 pyramidal neurons to regulate autism-related transcripts and circadian memory. eLife 2019; 8:e46919. [PMID: 31860442 PMCID: PMC6924960 DOI: 10.7554/elife.46919] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2019] [Accepted: 12/02/2019] [Indexed: 12/14/2022] Open
Abstract
Loss of the RNA binding protein FMRP causes Fragile X Syndrome (FXS), the most common cause of inherited intellectual disability, yet it is unknown how FMRP function varies across brain regions and cell types and how this contributes to disease pathophysiology. Here we use conditional tagging of FMRP and CLIP (FMRP cTag CLIP) to examine FMRP mRNA targets in hippocampal CA1 pyramidal neurons, a critical cell type for learning and memory relevant to FXS phenotypes. Integrating these data with analysis of ribosome-bound transcripts in these neurons revealed CA1-enriched binding of autism-relevant mRNAs, and CA1-specific regulation of transcripts encoding circadian proteins. This contrasted with different targets in cerebellar granule neurons, and was consistent with circadian defects in hippocampus-dependent memory in Fmr1 knockout mice. These findings demonstrate differential FMRP-dependent regulation of mRNAs across neuronal cell types that may contribute to phenotypes such as memory defects and sleep disturbance associated with FXS.
Collapse
Affiliation(s)
- Kirsty Sawicka
- Laboratory of Molecular Neuro-OncologyThe Rockefeller UniversityNew YorkUnited States
| | - Caryn R Hale
- Laboratory of Molecular Neuro-OncologyThe Rockefeller UniversityNew YorkUnited States
| | - Christopher Y Park
- Laboratory of Molecular Neuro-OncologyThe Rockefeller UniversityNew YorkUnited States
| | - John J Fak
- Laboratory of Molecular Neuro-OncologyThe Rockefeller UniversityNew YorkUnited States
| | - Jodi E Gresack
- Laboratory of Molecular and Cellular NeuroscienceThe Rockefeller UniversityNew YorkUnited States
| | - Sarah J Van Driesche
- Laboratory of Molecular Neuro-OncologyThe Rockefeller UniversityNew YorkUnited States
| | - Jin Joo Kang
- Laboratory of Molecular Neuro-OncologyThe Rockefeller UniversityNew YorkUnited States
| | - Jennifer C Darnell
- Laboratory of Molecular Neuro-OncologyThe Rockefeller UniversityNew YorkUnited States
| | - Robert B Darnell
- Laboratory of Molecular Neuro-OncologyThe Rockefeller UniversityNew YorkUnited States
- Howard Hughes Medical InstituteChevy ChaseUnited States
| |
Collapse
|
76
|
Hodges SL, Nolan SO, Tomac LA, Muhammad IDA, Binder MS, Taube JH, Lugo JN. Lipopolysaccharide-induced inflammation leads to acute elevations in pro-inflammatory cytokine expression in a mouse model of Fragile X syndrome. Physiol Behav 2019; 215:112776. [PMID: 31838149 DOI: 10.1016/j.physbeh.2019.112776] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 12/11/2019] [Accepted: 12/11/2019] [Indexed: 02/07/2023]
Abstract
Fragile X syndrome (FXS) is a neurodevelopmental disorder caused by a single genetic mutation in the Fmr1 gene, serving as the largest genetic cause of intellectual disability. Trinucleotide expansion mutations in Fmr1 result in silencing and hypermethylation of the gene, preventing synthesis of the RNA binding protein Fragile X mental retardation protein which functions as a translational repressor. Abnormal immune responses have been demonstrated to play a role in FXS pathophysiology, however, whether these alterations impact how those with FXS respond to an immune insult behaviorally is not entirely known. In the current study, we examine how Fmr1 knockout (KO) and wild type (WT) mice respond to the innate immune stimulus lipopolysaccharide (LPS), both on a molecular and behavioral level, to determine if Fmr1 mutations impact the normal physiological response to an immune insult. In response to LPS, Fmr1 KO mice had elevated hippocampal IL-1β and IL-6 mRNA levels 4 h post-treatment compared to WT mice, with no differences detected in any cytokines at baseline or between genotypes 24 h post-LPS administration. Fmr1 KO mice also had upregulated hippocampal BDNF gene expression 4 h post-treatment compared to WT mice, which was not dependent on LPS administration. There were no differences in hippocampal protein expression between genotypes in microglia (Iba1) or astrocyte (GFAP) reactivity. Further, both genotypes displayed the typical sickness response following LPS stimulation, demonstrated by a significant reduction in food burrowed by LPS-treated mice in a burrowing task. Additional investigation is critical to determine if the transient increases in cytokine expression could lead to long-term changes in downstream molecular signaling in FXS.
Collapse
Affiliation(s)
- Samantha L Hodges
- Institute of Biomedical Studies, Baylor University, Waco, TX 76798, USA
| | - Suzanne O Nolan
- Department of Psychology and Neuroscience, Baylor University, One Bear Place # 97334, Waco, TX 76798, USA
| | - Lindsay A Tomac
- Department of Psychology and Neuroscience, Baylor University, One Bear Place # 97334, Waco, TX 76798, USA
| | - Ilyasah D A Muhammad
- Department of Psychology and Neuroscience, Baylor University, One Bear Place # 97334, Waco, TX 76798, USA
| | - Matthew S Binder
- Department of Psychology and Neuroscience, Baylor University, One Bear Place # 97334, Waco, TX 76798, USA
| | - Joseph H Taube
- Institute of Biomedical Studies, Baylor University, Waco, TX 76798, USA; Department of Biology, Baylor University, Waco, TX 76798, USA
| | - Joaquin N Lugo
- Institute of Biomedical Studies, Baylor University, Waco, TX 76798, USA; Department of Psychology and Neuroscience, Baylor University, One Bear Place # 97334, Waco, TX 76798, USA; Department of Biology, Baylor University, Waco, TX 76798, USA.
| |
Collapse
|
77
|
Nussbacher JK, Tabet R, Yeo GW, Lagier-Tourenne C. Disruption of RNA Metabolism in Neurological Diseases and Emerging Therapeutic Interventions. Neuron 2019; 102:294-320. [PMID: 30998900 DOI: 10.1016/j.neuron.2019.03.014] [Citation(s) in RCA: 184] [Impact Index Per Article: 30.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 01/24/2019] [Accepted: 03/12/2019] [Indexed: 02/06/2023]
Abstract
RNA binding proteins are critical to the maintenance of the transcriptome via controlled regulation of RNA processing and transport. Alterations of these proteins impact multiple steps of the RNA life cycle resulting in various molecular phenotypes such as aberrant RNA splicing, transport, and stability. Disruption of RNA binding proteins and widespread RNA processing defects are increasingly recognized as critical determinants of neurological diseases. Here, we describe distinct mechanisms by which the homeostasis of RNA binding proteins is compromised in neurological disorders through their reduced expression level, increased propensity to aggregate or sequestration by abnormal RNAs. These mechanisms all converge toward altered neuronal function highlighting the susceptibility of neurons to deleterious changes in RNA expression and the central role of RNA binding proteins in preserving neuronal integrity. Emerging therapeutic approaches to mitigate or reverse alterations of RNA binding proteins in neurological diseases are discussed.
Collapse
Affiliation(s)
- Julia K Nussbacher
- Department of Cellular and Molecular Medicine, Institute for Genomic Medicine, UCSD Stem Cell Program, University of California, San Diego, La Jolla, CA, USA
| | - Ricardos Tabet
- Department of Neurology, The Sean M. Healey and AMG Center for ALS at Mass General, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA; Broad Institute of Harvard University and MIT, Cambridge, MA 02142, USA
| | - Gene W Yeo
- Department of Cellular and Molecular Medicine, Institute for Genomic Medicine, UCSD Stem Cell Program, University of California, San Diego, La Jolla, CA, USA.
| | - Clotilde Lagier-Tourenne
- Department of Neurology, The Sean M. Healey and AMG Center for ALS at Mass General, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA; Broad Institute of Harvard University and MIT, Cambridge, MA 02142, USA.
| |
Collapse
|
78
|
Evidence for a Contribution of the Nlgn3/Cyfip1/Fmr1 Pathway in the Pathophysiology of Autism Spectrum Disorders. Neuroscience 2019; 445:31-41. [PMID: 31705895 DOI: 10.1016/j.neuroscience.2019.10.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Accepted: 10/06/2019] [Indexed: 12/15/2022]
Abstract
Autism Spectrum Disorders (ASD) are characterized by heterogeneity both in their presentation and their genetic aetiology. In order to discover points of convergence common to different cases of ASD, attempts were made to identify the biological pathways genes associated with ASD contribute to. Many of these genes were found to play a role in neuronal and synaptic development and function. Among these genes are FMR1, CYFIP1 and NLGN3, all present at the synapse and reliably linked to ASD. In this review, we evaluate the evidence for the contribution of these genes to the same biological pathway responsible for the regulation of structural and physiological plasticity. Alterations in dendritic spine density and turnover, as well as long-term depression (LTD), were found in mouse models of mutations of all three genes. This overlap in the phenotypes associated with these mouse models likely arises from the molecular interaction between the protein products of FMR1, CYFIP1, and NLG3. A number of other proteins linked to ASD are also likely to participate in these pathways, resulting in further downstream effects. Overall, a synaptic pathway centered around FMR1, CYFIP1, and NLG3 is likely to contribute to the phenotypes associated with structural and physiological plasticity characteristic of ASD.
Collapse
|
79
|
Lee FHF, Lai TKY, Su P, Liu F. Altered cortical Cytoarchitecture in the Fmr1 knockout mouse. Mol Brain 2019; 12:56. [PMID: 31200759 PMCID: PMC6570929 DOI: 10.1186/s13041-019-0478-8] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Accepted: 06/03/2019] [Indexed: 12/15/2022] Open
Abstract
Fragile X syndrome (FXS) is a neurodevelopmental disorder caused by silencing of the FMR1 gene and subsequent loss of its protein product, fragile X retardation protein (FMRP). One of the most robust neuropathological findings in post-mortem human FXS and Fmr1 KO mice is the abnormal increase in dendritic spine densities, with the majority of spines showing an elongated immature morphology. However, the exact mechanisms of how FMRP can regulate dendritic spine development are still unclear. Abnormal dendritic spines can result from disturbances of multiple factors during neurodevelopment, such as alterations in neuron numbers, position and glial cells. In this study, we undertook a comprehensive histological analysis of the cerebral cortex in Fmr1 KO mice. They displayed significantly fewer neuron and PV-interneuron numbers, along with altered cortical lamination patterns. In terms of glial cells, Fmr1 KO mice exhibited an increase in Olig2-oligodendrocytes, which corresponded to the abnormally higher myelin expression in the corpus callosum. Iba1-microglia were significantly reduced but GFAP-astrocyte numbers and intensity were elevated. Using primary astrocytes derived from KO mice, we further demonstrated the presence of astrogliosis characterized by an increase in GFAP expression and astrocyte hypertrophy. Our findings provide important information on the cortical architecture of Fmr1 KO mice, and insights towards possible mechanisms associated with FXS.
Collapse
Affiliation(s)
- Frankie H F Lee
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, M5T 1R8, Canada
| | - Terence K Y Lai
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, M5T 1R8, Canada.,Department of Physiology, University of Toronto, Toronto, Ontario, M5T 1R8, Canada
| | - Ping Su
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, M5T 1R8, Canada
| | - Fang Liu
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, M5T 1R8, Canada. .,Department of Physiology, University of Toronto, Toronto, Ontario, M5T 1R8, Canada. .,Department of Psychiatry, University of Toronto, Toronto, Ontario, M5T 1R8, Canada.
| |
Collapse
|
80
|
Humble J, Hiratsuka K, Kasai H, Toyoizumi T. Intrinsic Spine Dynamics Are Critical for Recurrent Network Learning in Models With and Without Autism Spectrum Disorder. Front Comput Neurosci 2019; 13:38. [PMID: 31263407 PMCID: PMC6585147 DOI: 10.3389/fncom.2019.00038] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2019] [Accepted: 05/28/2019] [Indexed: 11/13/2022] Open
Abstract
It is often assumed that Hebbian synaptic plasticity forms a cell assembly, a mutually interacting group of neurons that encodes memory. However, in recurrently connected networks with pure Hebbian plasticity, cell assemblies typically diverge or fade under ongoing changes of synaptic strength. Previously assumed mechanisms that stabilize cell assemblies do not robustly reproduce the experimentally reported unimodal and long-tailed distribution of synaptic strengths. Here, we show that augmenting Hebbian plasticity with experimentally observed intrinsic spine dynamics can stabilize cell assemblies and reproduce the distribution of synaptic strengths. Moreover, we posit that strong intrinsic spine dynamics impair learning performance. Our theory explains how excessively strong spine dynamics, experimentally observed in several animal models of autism spectrum disorder, impair learning associations in the brain.
Collapse
Affiliation(s)
- James Humble
- Laboratory for Neural Computation and Adaptation, RIKEN Center for Brain Science, Saitama, Japan
| | - Kazuhiro Hiratsuka
- Laboratory for Neural Computation and Adaptation, RIKEN Center for Brain Science, Saitama, Japan
| | - Haruo Kasai
- Laboratory of Structural Physiology, Faculty of Medicine, Center for Disease Biology and Integrative Medicine, University of Tokyo, Tokyo, Japan
| | - Taro Toyoizumi
- Laboratory for Neural Computation and Adaptation, RIKEN Center for Brain Science, Saitama, Japan
| |
Collapse
|
81
|
Whyte AJ, Kietzman HW, Swanson AM, Butkovich LM, Barbee BR, Bassell GJ, Gross C, Gourley SL. Reward-Related Expectations Trigger Dendritic Spine Plasticity in the Mouse Ventrolateral Orbitofrontal Cortex. J Neurosci 2019; 39:4595-4605. [PMID: 30940719 PMCID: PMC6554633 DOI: 10.1523/jneurosci.2031-18.2019] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 03/07/2019] [Accepted: 03/26/2019] [Indexed: 02/06/2023] Open
Abstract
An essential aspect of goal-directed decision-making is selecting actions based on anticipated consequences, a process that involves the orbitofrontal cortex (OFC) and potentially, the plasticity of dendritic spines in this region. To investigate this possibility, we trained male and female mice to nose poke for food reinforcers, or we delivered the same number of food reinforcers non-contingently to separate mice. We then decreased the likelihood of reinforcement for trained mice, requiring them to modify action-outcome expectations. In a separate experiment, we blocked action-outcome updating via chemogenetic inactivation of the OFC. In both cases, successfully selecting actions based on their likely consequences was associated with fewer immature, thin-shaped dendritic spines and a greater proportion of mature, mushroom-shaped spines in the ventrolateral OFC. This pattern was distinct from spine loss associated with aging, and we identified no effects on hippocampal CA1 neurons. Given that the OFC is involved in prospective calculations of likely outcomes, even when they are not observable, constraining spinogenesis while preserving mature spines may be important for solidifying durable expectations. To investigate causal relationships, we inhibited the RNA-binding protein fragile X mental retardation protein (encoded by Fmr1), which constrains dendritic spine turnover. Ventrolateral OFC-selective Fmr1 knockdown recapitulated the behavioral effects of inducible OFC inactivation (and lesions; also shown here), impairing action-outcome conditioning, and caused dendritic spine excess. Our findings suggest that a proper balance of dendritic spine plasticity within the OFC is necessary for one's ability to select actions based on anticipated consequences.SIGNIFICANCE STATEMENT Navigating a changing environment requires associating actions with their likely outcomes and updating these associations when they change. Dendritic spine plasticity is likely involved, yet relationships are unconfirmed. Using behavioral, chemogenetic, and viral-mediated gene silencing strategies and high-resolution microscopy, we find that modifying action-outcome expectations is associated with fewer immature spines and a greater proportion of mature spines in the ventrolateral orbitofrontal cortex (OFC). Given that the OFC is involved in prospectively calculating the likely outcomes of one's behavior, even when they are not observable, constraining spinogenesis while preserving mature spines may be important for maintaining durable expectations.
Collapse
Affiliation(s)
- Alonzo J Whyte
- Departments of Cell Biology
- Pediatrics, Emory School of Medicine
- Yerkes National Primate Research Center
| | - Henry W Kietzman
- Pediatrics, Emory School of Medicine
- Yerkes National Primate Research Center
- Graduate Program in Neuroscience
| | - Andrew M Swanson
- Pediatrics, Emory School of Medicine
- Yerkes National Primate Research Center
- Graduate Program in Neuroscience
| | - Laura M Butkovich
- Pediatrics, Emory School of Medicine
- Yerkes National Primate Research Center
- Graduate Program in Neuroscience
| | - Britton R Barbee
- Pediatrics, Emory School of Medicine
- Yerkes National Primate Research Center
- Graduate Program in Molecular and Systems Pharmacology, Emory University, Atlanta, Georgia 30329
| | - Gary J Bassell
- Departments of Cell Biology
- Graduate Program in Neuroscience
| | - Christina Gross
- Division of Neurology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio 45229, and
- Department of Pediatrics, University of Cincinnati, College of Medicine, Cincinnati, Ohio 45267
| | - Shannon L Gourley
- Pediatrics, Emory School of Medicine,
- Yerkes National Primate Research Center
- Graduate Program in Neuroscience
- Graduate Program in Molecular and Systems Pharmacology, Emory University, Atlanta, Georgia 30329
| |
Collapse
|
82
|
Bodaleo F, Tapia-Monsalves C, Cea-Del Rio C, Gonzalez-Billault C, Nunez-Parra A. Structural and Functional Abnormalities in the Olfactory System of Fragile X Syndrome Models. Front Mol Neurosci 2019; 12:135. [PMID: 31191246 PMCID: PMC6548058 DOI: 10.3389/fnmol.2019.00135] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Accepted: 05/09/2019] [Indexed: 12/18/2022] Open
Abstract
Fragile X Syndrome (FXS) is the most common inherited form of intellectual disability. It is produced by mutation of the Fmr1 gene that encodes for the Fragile Mental Retardation Protein (FMRP), an important RNA-binding protein that regulates the expression of multiple proteins located in neuronal synapses. Individuals with FXS exhibit abnormal sensory information processing frequently leading to hypersensitivity across sensory modalities and consequently a wide array of behavioral symptoms. Insects and mammals engage primarily their sense of smell to create proper representations of the external world and guide adequate decision-making processes. This feature in combination with the exquisitely organized neuronal circuits found throughout the olfactory system (OS) and the wide expression of FMRP in brain regions that process olfactory information makes it an ideal model to study sensory alterations in FXS models. In the last decade several groups have taken advantage of these features and have used the OS of fruit fly and rodents to understand neuronal alteration giving rise to sensory perception issues. In this review article, we will discuss molecular, morphological and physiological aspects of the olfactory information processing in FXS models. We will highlight the decreased inhibitory/excitatory synaptic balance and the diminished synaptic plasticity found in this system resulting in behavioral alteration of individuals in the presence of odorant stimuli.
Collapse
Affiliation(s)
- Felipe Bodaleo
- Laboratory of Cellular and Neuronal Dynamics, Department of Biology, Faculty of Science, Universidad de Chile, Santiago, Chile.,Geroscience Center for Brain Health and Metabolism (GERO), Santiago, Chile
| | | | - Christian Cea-Del Rio
- Laboratory of Neurophysiopathology, Centro de Investigacion Biomedica y Aplicada (CIBAP), School of Medicine, Universidad de Santiago de Chile, Santiago, Chile
| | - Christian Gonzalez-Billault
- Laboratory of Cellular and Neuronal Dynamics, Department of Biology, Faculty of Science, Universidad de Chile, Santiago, Chile.,Geroscience Center for Brain Health and Metabolism (GERO), Santiago, Chile.,The Buck Institute for Research on Aging, Novato, CA, United States
| | - Alexia Nunez-Parra
- Department of Biology, Faculty of Science, Universidad de Chile, Santiago, Chile.,Cell Physiology Center, Universidad de Chile, Santiago, Chile
| |
Collapse
|
83
|
Bagni C, Zukin RS. A Synaptic Perspective of Fragile X Syndrome and Autism Spectrum Disorders. Neuron 2019; 101:1070-1088. [PMID: 30897358 PMCID: PMC9628679 DOI: 10.1016/j.neuron.2019.02.041] [Citation(s) in RCA: 233] [Impact Index Per Article: 38.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 02/25/2019] [Accepted: 02/27/2019] [Indexed: 12/28/2022]
Abstract
Altered synaptic structure and function is a major hallmark of fragile X syndrome (FXS), autism spectrum disorders (ASDs), and other intellectual disabilities (IDs), which are therefore classified as synaptopathies. FXS and ASDs, while clinically and genetically distinct, share significant comorbidity, suggesting that there may be a common molecular and/or cellular basis, presumably at the synapse. In this article, we review brain architecture and synaptic pathways that are dysregulated in FXS and ASDs, including spine architecture, signaling in synaptic plasticity, local protein synthesis, (m)RNA modifications, and degradation. mRNA repression is a powerful mechanism for the regulation of synaptic structure and efficacy. We infer that there is no single pathway that explains most of the etiology and discuss new findings and the implications for future work directed at improving our understanding of the pathogenesis of FXS and related ASDs and the design of therapeutic strategies to ameliorate these disorders.
Collapse
Affiliation(s)
- Claudia Bagni
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, Switzerland; Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy.
| | - R Suzanne Zukin
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, New York City, NY, USA.
| |
Collapse
|
84
|
Urban P, Rezaei V, Bokota G, Denkiewicz M, Basu S, Plewczyński D. Dendritic Spines Taxonomy: The Functional and Structural Classification • Time-Dependent Probabilistic Model of Neuronal Activation. J Comput Biol 2019; 26:322-335. [PMID: 30810368 DOI: 10.1089/cmb.2018.0155] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Categorizing spines into four subpopulations, stubby, mushroom, thin, or filopodia, is one of the common approaches in morphological analysis. Most cellular models describing synaptic plasticity, long-term potentiation (LTP), and long-term depression associate synaptic strength with either spine enlargement or spine shrinkage. Unfortunately, although we have a lot of available software with automatic spine segmentation and feature extraction methods, at present none of them allows for automatic and unbiased distinction between dendritic spine subpopulations, or for the detailed computational models of spine behavior. Therefore, we propose structural classification based on two different mathematical approaches: unsupervised construction of spine shape taxonomy based on arbitrary features (SpineTool) and supervised classification exploiting convolution kernels theory (2dSpAn). We compared two populations of spines in a form of static and dynamic data sets gathered at three time points. The dynamic data contain two sets of spines: the active set and the control set. The first population was stimulated with LTP, and the other population in its resting state was used as a control population. We propose one equation describing the distribution of variables that best fits all dendritic spine parameters.
Collapse
Affiliation(s)
- Paulina Urban
- 1 Center of New Technologies, University of Warsaw, Warsaw, Poland.,2 College of Inter-Faculty Individual Studies in Mathematics and Natural Sciences, University of Warsaw, Warsaw, Poland
| | - Vahid Rezaei
- 3 Department of Statistics, Faculty of Mathematics and Computer Sciences, Allameh Tabataba'i University, Tehran, Iran.,4 School of Biological Sciences, Institute for Research in Fundamental Sciences (IPM), Tehran, Iran
| | - Grzegorz Bokota
- 1 Center of New Technologies, University of Warsaw, Warsaw, Poland.,5 Faculty of Mathematics, Informatics and Mechanics, University of Warsaw, Warsaw, Poland
| | - Michał Denkiewicz
- 1 Center of New Technologies, University of Warsaw, Warsaw, Poland.,2 College of Inter-Faculty Individual Studies in Mathematics and Natural Sciences, University of Warsaw, Warsaw, Poland
| | - Subhadip Basu
- 6 Department of Computer Science and Engineering, Jadavpur University, Kolkata, India
| | - Dariusz Plewczyński
- 1 Center of New Technologies, University of Warsaw, Warsaw, Poland.,7 Faculty of Mathematics and Information Science, Warsaw University of Technology, Warsaw, Poland
| |
Collapse
|
85
|
Arroyo ED, Fiole D, Mantri SS, Huang C, Portera-Cailliau C. Dendritic Spines in Early Postnatal Fragile X Mice Are Insensitive to Novel Sensory Experience. J Neurosci 2019; 39:412-419. [PMID: 30523064 PMCID: PMC6335755 DOI: 10.1523/jneurosci.1734-18.2018] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2018] [Revised: 10/02/2018] [Accepted: 10/20/2018] [Indexed: 11/21/2022] Open
Abstract
Autism spectrum disorders are often associated with atypical sensory processing and sensory hypersensitivity, which can lead to maladaptive behaviors, such as tactile defensiveness. Such altered sensory perception in autism spectrum disorders could arise from disruptions in experience-dependent maturation of circuits during early brain development. Here, we tested the hypothesis that synaptic structures of primary somatosensory cortex (S1) neurons in Fragile X syndrome (FXS), which is a common inherited cause of autism, are not modulated by novel sensory information during development. We used chronic in vivo two-photon microscopy to image dendritic spines and axon "en passant" boutons of layer 2/3 pyramidal neurons in S1 of male and female WT and Fmr1 KO mice, a model of FXS. We found that a brief (overnight) exposure to dramatically enhance sensory inputs in the second postnatal week led to a significant increase in spine density in WT mice, but not in Fmr1 KO mice. In contrast, axon "en passant" boutons dynamics were impervious to this novel sensory experience in mice of both genotypes. We surmise that the inability of Fmr1 KO mice to modulate postsynaptic dynamics in response to increased sensory input, at a time when sensory information processing first comes online in S1 cortex, could play a role in altered sensory processing in FXS.SIGNIFICANCE STATEMENT Very few longitudinal in vivo imaging studies have investigated synaptic structure and dynamics in early postnatal mice. Moreover, those studies tend to focus on the effects of sensory input deprivation, a process that rarely occurs during normal brain development. Early postnatal imaging experiments are critical because a variety of neurodevelopmental disorders, including those characterized by autism, could result from alterations in how circuits are shaped by incoming sensory inputs during critical periods of development. In this study, we focused on a mouse model of Fragile X syndrome and demonstrate how dendritic spines are insensitive to a brief period of novel sensory experience.
Collapse
Affiliation(s)
- Erica D Arroyo
- Department of Neurology
- Neuroscience Interdepartmental Program, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California 90095
| | | | | | | | | |
Collapse
|
86
|
Lee AW, Ventola P, Budimirovic D, Berry-Kravis E, Visootsak J. Clinical Development of Targeted Fragile X Syndrome Treatments: An Industry Perspective. Brain Sci 2018; 8:E214. [PMID: 30563047 PMCID: PMC6315847 DOI: 10.3390/brainsci8120214] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 11/28/2018] [Accepted: 11/30/2018] [Indexed: 01/03/2023] Open
Abstract
Fragile X syndrome (FXS) is the leading known cause of inherited intellectual disability and autism spectrum disorder. It is caused by a mutation of the fragile X mental retardation 1 (FMR1) gene, resulting in a deficit of fragile X mental retardation protein (FMRP). The clinical presentation of FXS is variable, and is typically associated with developmental delays, intellectual disability, a wide range of behavioral issues, and certain identifying physical features. Over the past 25 years, researchers have worked to understand the complex relationship between FMRP deficiency and the symptoms of FXS and, in the process, have identified several potential targeted therapeutics, some of which have been tested in clinical trials. Whereas most of the basic research to date has been led by experts at academic institutions, the pharmaceutical industry is becoming increasingly involved with not only the scientific community, but also with patient advocacy organizations, as more promising pharmacological agents are moving into the clinical stages of development. The objective of this review is to provide an industry perspective on the ongoing development of mechanism-based treatments for FXS, including identification of challenges and recommendations for future clinical trials.
Collapse
Affiliation(s)
- Anna W Lee
- Ovid Therapeutics Inc., New York, NY 10036, USA.
| | - Pamela Ventola
- Child Study Center, Yale University, New Haven, CT 06520, USA.
| | - Dejan Budimirovic
- Departments of Psychiatry and Behavioral Sciences, Kennedy Krieger Institute and Child Psychiatry, Johns Hopkins University, Baltimore, MD 21205, USA.
| | - Elizabeth Berry-Kravis
- Departments of Pediatrics, Neurological Sciences, Biochemistry, Rush University Medical Center, Chicago, IL 60612, USA.
| | | |
Collapse
|
87
|
Development of GABAergic Inputs Is Not Altered in Early Maturation of Adult Born Dentate Granule Neurons in Fragile X Mice. eNeuro 2018; 5:eN-NRS-0137-18. [PMID: 30627633 PMCID: PMC6325535 DOI: 10.1523/eneuro.0137-18.2018] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Revised: 09/27/2018] [Accepted: 10/17/2018] [Indexed: 02/08/2023] Open
Abstract
Fragile X syndrome (FXS) is the most common form of inherited mental retardation and the most common known cause of autism. Loss of fragile X mental retardation protein (FMRP) in mice (Fmr1 KO) leads to altered synaptic and circuit maturation in the hippocampus that is correlated with alterations in hippocampal-dependent behaviors. Previous studies have demonstrated that loss of FMRP increased the rate of proliferation of progenitor cells and altered their fate specification in adult Fmr1 KO mice. While these studies clearly demonstrate a role for FMRP in adult neurogenesis in the hippocampus, it is not known whether the functional synaptic maturation and integration of adult-born dentate granule cells (abDGCs) into hippocampal circuits is affected in Fmr1 KO mice. Here, we used retroviral labeling to birthdate abDGCs in Fmr1 KO mice which allowed us to perform targeted patch clamp recording to measure the development of synaptic inputs to these neurons at precise time points after differentiation. The frequency and amplitude of spontaneous GABAergic events increased over the first three weeks after differentiation; however, this normal development of GABAergic synapses was not altered in Fmr1 KO mice. Furthermore, the relatively depolarized GABA reversal potential (EGABA) in immature abDGCs was unaffected by loss of FMRP as was the development of dendritic arbor of the adult generated neurons. These studies systematically characterized the functional development of abDGCs during the first four weeks after differentiation and demonstrate that the maturation of GABAergic synaptic inputs to these neurons is not grossly affected by the loss of FMRP.
Collapse
|
88
|
In Vivo Volume Dynamics of Dendritic Spines in the Neocortex of Wild-Type and Fmr1 KO Mice. eNeuro 2018; 5:eN-NWR-0282-18. [PMID: 30417082 PMCID: PMC6223115 DOI: 10.1523/eneuro.0282-18.2018] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Revised: 09/30/2018] [Accepted: 10/03/2018] [Indexed: 02/06/2023] Open
Abstract
Excitatory synapses are often formed at small protrusions of dendrite, called dendritic spines, in most projection neurons, and the spine-head volumes show strong correlations with synaptic connectivity. We examined the dynamics of spine volume in the adult mouse visual cortex using time-lapse in vivo two-photon imaging with a resonant Galvano scanner. Contrary to expectations, we found that the spines in the adult neocortex showed fluctuations to a similar degree as that observed in young hippocampal preparations, but there were systematic differences in how the dynamics were dependent on spine volumes, thus allowing for fewer fluctuations in small spines, which could account for the relatively low turnover rates of neocortical spines in vivo. We found that spine volumes fluctuated to a greater extent in a mouse model (Fmr1 knockout) of fragile X mental retardation than in wild-type mice, and the spine turnover rates were also higher in Fmr1 knock-out mice. Such features of spine dynamics in Fmr1 knock-out mice could be represented by a single slope factor in our model. Our data and model indicate a small but significant change in the average spine volume and more eminent differences in the statistical distribution in Fmr1 knock-out mice even in adulthood, which reflects the abnormal in vivo dynamics of spine volumes.
Collapse
|
89
|
Patel J, Lukkes JL, Shekhar A. Overview of genetic models of autism spectrum disorders. PROGRESS IN BRAIN RESEARCH 2018; 241:1-36. [PMID: 30447752 DOI: 10.1016/bs.pbr.2018.10.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Autism spectrum disorders (ASDs) are a group of neurodevelopment disorders that are characterized by heterogenous cognitive deficits and genetic factors. As more ASD risk genes are identified, genetic animal models have been developed to parse out the underlying neurobiological mechanisms of ASD. In this review, we discuss a subset of genetic models of ASD, focusing on those that have been widely studied and strongly linked to ASD. We focus our discussion of these models in the context of the theories and potential mechanisms of ASD, including disruptions in cell growth and proliferation, spine dynamics, synaptic transmission, excitation/inhibition balance, intracellular signaling, neuroinflammation, and behavior. In addition to ASD pathophysiology, we examine the limitations and challenges that genetic models pose for the study of ASD biology. We end with a review of innovative techniques and concepts of ASD pathology that can be further applied to and studied using genetic ASD models.
Collapse
Affiliation(s)
- Jheel Patel
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, United States; Program in Medical Neuroscience, Paul and Carole Stark Neurosciences Research Institute, Indianapolis, IN, United States
| | - Jodi L Lukkes
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Anantha Shekhar
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, United States; Program in Medical Neuroscience, Paul and Carole Stark Neurosciences Research Institute, Indianapolis, IN, United States; Indiana Clinical and Translation Sciences Institute, Indiana University School of Medicine, Indianapolis, IN, United States.
| |
Collapse
|
90
|
Jose R, Santen L, Shaebani MR. Trapping in and Escape from Branched Structures of Neuronal Dendrites. Biophys J 2018; 115:2014-2025. [PMID: 30366628 DOI: 10.1016/j.bpj.2018.09.029] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Revised: 09/20/2018] [Accepted: 09/26/2018] [Indexed: 10/28/2022] Open
Abstract
We present a coarse-grained model for stochastic transport of noninteracting chemical signals inside neuronal dendrites and show how first-passage properties depend on the key structural factors affected by neurodegenerative disorders or aging: the extent of the tree, the topological bias induced by segmental decrease of dendrite diameter, and the trapping probabilities in biochemical cages and growth cones. We derive an exact expression for the distribution of first-passage times, which follows a universal exponential decay in the long-time limit. The asymptotic mean first-passage time exhibits a crossover from power-law to exponential scaling upon reducing the topological bias. We calibrate the coarse-grained model parameters and obtain the variation range of the mean first-passage time when the geometrical characteristics of the dendritic structure evolve during the course of aging or neurodegenerative disease progression (a few disorders for which clear trends for the pathological changes of dendritic structure have been reported in the literature are chosen and studied). We prove the validity of our analytical approach under realistic fluctuations of structural parameters by comparison to the results of Monte Carlo simulations. Moreover, by constructing local structural irregularities, we analyze the resulting influence on transport of chemical signals and formation of heterogeneous density patterns. Because neural functions rely on chemical signal transmission to a large extent, our results open the possibility of establishing a direct link between the disease progression and neural functions.
Collapse
Affiliation(s)
- Robin Jose
- Department of Theoretical Physics and Center for Biophysics, Saarland University, Saarbrücken, Germany
| | - Ludger Santen
- Department of Theoretical Physics and Center for Biophysics, Saarland University, Saarbrücken, Germany
| | - M Reza Shaebani
- Department of Theoretical Physics and Center for Biophysics, Saarland University, Saarbrücken, Germany.
| |
Collapse
|
91
|
Goel A, Cantu DA, Guilfoyle J, Chaudhari GR, Newadkar A, Todisco B, de Alba D, Kourdougli N, Schmitt LM, Pedapati E, Erickson CA, Portera-Cailliau C. Impaired perceptual learning in a mouse model of Fragile X syndrome is mediated by parvalbumin neuron dysfunction and is reversible. Nat Neurosci 2018; 21:1404-1411. [PMID: 30250263 PMCID: PMC6161491 DOI: 10.1038/s41593-018-0231-0] [Citation(s) in RCA: 116] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2017] [Accepted: 08/06/2018] [Indexed: 12/26/2022]
Abstract
To uncover the circuit-level alterations that underlie atypical sensory processing associated with autism, we adopted a symptom-to-circuit approach in the Fmr1-knockout (Fmr1-/-) mouse model of Fragile X syndrome. Using a go/no-go task and in vivo two-photon calcium imaging, we find that impaired visual discrimination in Fmr1-/- mice correlates with marked deficits in orientation tuning of principal neurons and with a decrease in the activity of parvalbumin interneurons in primary visual cortex. Restoring visually evoked activity in parvalbumin cells in Fmr1-/- mice with a chemogenetic strategy using designer receptors exclusively activated by designer drugs was sufficient to rescue their behavioral performance. Strikingly, human subjects with Fragile X syndrome exhibit impairments in visual discrimination similar to those in Fmr1-/- mice. These results suggest that manipulating inhibition may help sensory processing in Fragile X syndrome.
Collapse
Affiliation(s)
- Anubhuti Goel
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Daniel A Cantu
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Janna Guilfoyle
- Department of Psychiatry, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Gunvant R Chaudhari
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Aditi Newadkar
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Barbara Todisco
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Diego de Alba
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Nazim Kourdougli
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Lauren M Schmitt
- Department of Psychiatry, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Ernest Pedapati
- Department of Psychiatry, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Department of Neurology, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Craig A Erickson
- Department of Psychiatry, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Carlos Portera-Cailliau
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA.
- Department of Neurobiology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA.
| |
Collapse
|
92
|
Melancia F, Trezza V. Modelling fragile X syndrome in the laboratory setting: A behavioral perspective. Behav Brain Res 2018; 350:149-163. [DOI: 10.1016/j.bbr.2018.04.042] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Revised: 04/24/2018] [Accepted: 04/24/2018] [Indexed: 12/13/2022]
|
93
|
Michaelsen‐Preusse K, Feuge J, Korte M. Imbalance of synaptic actin dynamics as a key to fragile X syndrome? J Physiol 2018; 596:2773-2782. [PMID: 29380377 PMCID: PMC6046079 DOI: 10.1113/jp275571] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Accepted: 01/09/2018] [Indexed: 11/08/2022] Open
Abstract
Our experiences and memories define who we are, and evidence has accumulated that memory formation is dependent on functional and structural adaptations of synaptic structures in our brain. Especially dendritic spines, the postsynaptic compartments of synapses show a strong structure-to-function relationship and a high degree of structural plasticity. Although the molecular mechanisms are not completely understood, it is known that these modifications are highly dependent on the actin cytoskeleton, the major cytoskeletal component of the spine. Given the crucial involvement of actin in these mechanisms, dysregulations of spine actin dynamics (reflected by alterations in dendritic spine morphology) can be found in a variety of neurological disorders ranging from schizophrenia to several forms of autism spectrum disorders such as fragile X syndrome (FXS). FXS is caused by a single mutation leading to an inactivation of the X-linked fragile X mental retardation 1 gene and loss of its gene product, the RNA-binding protein fragile X mental retardation protein 1 (FMRP), which normally can be found both pre- and postsynaptically. FMRP is involved in mRNA transport as well as regulation of local translation at the synapse, and although hundreds of FMRP-target mRNAs could be identified only a very few interactions between FMRP and actin-regulating proteins have been reported and validated. In this review we give an overview of recent work by our lab and others providing evidence that dysregulated actin dynamics might indeed be at the very base of a deeper understanding of neurological disorders ranging from cognitive impairment to the autism spectrum.
Collapse
Affiliation(s)
- Kristin Michaelsen‐Preusse
- Zoological Institute, Division of Cellular NeurobiologyTU BraunschweigSpielmannstr. 7Braunschweig38106Germany
| | - Jonas Feuge
- Zoological Institute, Division of Cellular NeurobiologyTU BraunschweigSpielmannstr. 7Braunschweig38106Germany
| | - Martin Korte
- Zoological Institute, Division of Cellular NeurobiologyTU BraunschweigSpielmannstr. 7Braunschweig38106Germany
- Helmholtz Centre for Infection ResearchAG NINDInhoffenstr. 7Braunschweig38124Germany
| |
Collapse
|
94
|
Sengpiel F. Overview: neuroplasticity and synaptic function in neuropsychiatric disorders. J Physiol 2018; 596:2745-2746. [PMID: 30008191 PMCID: PMC6046064 DOI: 10.1113/jp275940] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Affiliation(s)
- Frank Sengpiel
- School of Biosciences, Cardiff UniversityMuseum AvenueCardiffCF10 3AXUK
| |
Collapse
|
95
|
Linda K, Fiuza C, Nadif Kasri N. The promise of induced pluripotent stem cells for neurodevelopmental disorders. Prog Neuropsychopharmacol Biol Psychiatry 2018; 84:382-391. [PMID: 29128445 DOI: 10.1016/j.pnpbp.2017.11.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Revised: 10/30/2017] [Accepted: 11/07/2017] [Indexed: 12/19/2022]
Abstract
A major challenge in clinical genetics and medicine is represented by genetically and phenotypically highly diverse neurodevelopmental disorders, like for example intellectual disability and autism. Intellectual disability is characterized by substantial limitations in cognitive function and adaptive behaviour. At the cellular level, this is reflected by deficits in synaptic structure and plasticity and therefore has been coined as a synaptic disorder or "synaptopathy". In this review, we summarize the findings from recent studies in which iPSCs have been used to model specific neurodevelopmental syndromes, including Fragile X syndrome, Rett syndrome, Williams-Beuren syndrome and Phelan-McDermid syndrome. We discuss what we have learned from these studies and what key issues need to be addressed to move the field forward.
Collapse
Affiliation(s)
- Katrin Linda
- Department of Human Genetics, Department of Cognitive Neuroscience, Radboudumc, 6500 HB, Nijmegen, The Netherlands; Donders Institute for Brain, Cognition, and Behaviour, Centre for Neuroscience, 6525 AJ, Nijmegen, The Netherlands
| | - Carol Fiuza
- Department of Human Genetics, Department of Cognitive Neuroscience, Radboudumc, 6500 HB, Nijmegen, The Netherlands; Donders Institute for Brain, Cognition, and Behaviour, Centre for Neuroscience, 6525 AJ, Nijmegen, The Netherlands
| | - Nael Nadif Kasri
- Department of Human Genetics, Department of Cognitive Neuroscience, Radboudumc, 6500 HB, Nijmegen, The Netherlands; Donders Institute for Brain, Cognition, and Behaviour, Centre for Neuroscience, 6525 AJ, Nijmegen, The Netherlands.
| |
Collapse
|
96
|
Medendorp WE, Petersen ED, Pal A, Wagner LM, Myers AR, Hochgeschwender U, Jenrow KA. Altered Behavior in Mice Socially Isolated During Adolescence Corresponds With Immature Dendritic Spine Morphology and Impaired Plasticity in the Prefrontal Cortex. Front Behav Neurosci 2018; 12:87. [PMID: 29867388 PMCID: PMC5954042 DOI: 10.3389/fnbeh.2018.00087] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2017] [Accepted: 04/20/2018] [Indexed: 11/29/2022] Open
Abstract
Mice socially isolated during adolescence exhibit behaviors of anxiety, depression and impaired social interaction. Although these behaviors are well documented, very little is known about the associated neurobiological changes that accompany these behaviors. It has been hypothesized that social isolation during adolescence alters the development of the prefrontal cortex, based on similar behavioral abnormalities observed in isolated mice and those with disruption of this structure. To establish relationships between behavior and underlying neurobiological changes in the prefrontal cortex, Thy-1-GFP mice were isolated from weaning until adulthood and compared to group-housed littermates regarding behavior, electrophysiological activity and dendritic morphology. Results indicate an immaturity of dendritic spines in single housed animals, with dendritic spines appearing smaller and thinner. Single housed mice additionally show impaired plasticity through measures of long-term potentiation. Together these findings suggest an altered development and impairment of the prefrontal cortex of these animals underlying their behavioral characteristics.
Collapse
Affiliation(s)
- William E Medendorp
- Neuroscience Program, Central Michigan University, Mount Pleasant, MI, United States.,College of Medicine, Central Michigan University, Mount Pleasant, MI, United States
| | - Eric D Petersen
- Neuroscience Program, Central Michigan University, Mount Pleasant, MI, United States.,College of Medicine, Central Michigan University, Mount Pleasant, MI, United States
| | - Akash Pal
- Neuroscience Program, Central Michigan University, Mount Pleasant, MI, United States.,College of Medicine, Central Michigan University, Mount Pleasant, MI, United States
| | - Lina-Marie Wagner
- Neuroscience Program, Central Michigan University, Mount Pleasant, MI, United States
| | - Alexzander R Myers
- Neuroscience Program, Central Michigan University, Mount Pleasant, MI, United States
| | - Ute Hochgeschwender
- Neuroscience Program, Central Michigan University, Mount Pleasant, MI, United States.,College of Medicine, Central Michigan University, Mount Pleasant, MI, United States
| | - Kenneth A Jenrow
- Neuroscience Program, Central Michigan University, Mount Pleasant, MI, United States.,Department of Psychology, Central Michigan University, Mount Pleasant, MI, United States
| |
Collapse
|
97
|
Suresh A, Dunaevsky A. Relationship Between Synaptic AMPAR and Spine Dynamics: Impairments in the FXS Mouse. Cereb Cortex 2018; 27:4244-4256. [PMID: 28541473 PMCID: PMC6057510 DOI: 10.1093/cercor/bhx128] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Accepted: 05/04/2017] [Indexed: 12/26/2022] Open
Abstract
Structural dynamics of dendritic spines are important for memory and learning and are impaired in neurodevelopmental disorders such as fragile X syndrome. Spine dynamics are regulated by activity-dependent mechanisms that involve modulation of AMPA receptors (AMPAR); however, the relationship between AMPAR and spine dynamics in vivo and how these are altered in FXS mouse model is not known. Here, we tracked AMPAR and spines over multiple days in vivo in the cortex and found that dendritic spines in the fmr1 KO mouse were denser, smaller, had higher turnover rates and contained less sGluA2 compared to littermate controls. Although, KO spines maintained the relationship between AMPAR and spine stability, AMPAR levels in the KO were more dynamic with larger proportion of spines showing multiple dynamic events of AMPAR. Directional changes in sGluA2 were also observed in newly formed and eliminated spines, with KO spines displaying greater loss of AMPAR before elimination. Thus, we demonstrate that AMPAR levels within spines not are only continuously dynamic, but are also predictive of spine behavior, with impairments observed in the fmr1 KO mice.
Collapse
Affiliation(s)
| | - Anna Dunaevsky
- Department of Developmental Neuroscience, Munroe-Meyer Institute, University of Nebraska Medical Center, 985960 Nebraska Medical Center, Omaha, NE 68198-5960, USA
| |
Collapse
|
98
|
Banerjee A, Ifrim MF, Valdez AN, Raj N, Bassell GJ. Aberrant RNA translation in fragile X syndrome: From FMRP mechanisms to emerging therapeutic strategies. Brain Res 2018; 1693:24-36. [PMID: 29653083 PMCID: PMC7377270 DOI: 10.1016/j.brainres.2018.04.008] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Revised: 03/30/2018] [Accepted: 04/06/2018] [Indexed: 02/07/2023]
Abstract
Research in the past decades has unfolded the multifaceted role of Fragile X mental retardation protein (FMRP) and how its absence contributes to the pathophysiology of Fragile X syndrome (FXS). Excess signaling through group 1 metabotropic glutamate receptors is commonly observed in mouse models of FXS, which in part is attributed to dysregulated translation and downstream signaling. Considering the wide spectrum of cellular and physiologic functions that loss of FMRP can affect in general, it may be advantageous to pursue disease mechanism based treatments that directly target translational components or signaling factors that regulate protein synthesis. Various FMRP targets upstream and downstream of the translational machinery are therefore being investigated to further our understanding of the molecular mechanism of RNA and protein synthesis dysregulation in FXS as well as test their potential role as therapeutic interventions to alleviate FXS associated symptoms. In this review, we will broadly discuss recent advancements made towards understanding the role of FMRP in translation regulation, new pre-clinical animal models with FMRP targets located at different levels of the translational and signal transduction pathways for therapeutic intervention as well as future use of stem cells to model FXS associated phenotypes.
Collapse
Affiliation(s)
- Anwesha Banerjee
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA 30322, USA.
| | - Marius F Ifrim
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Arielle N Valdez
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Nisha Raj
- Department of Cell Biology, 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; Department of Neurology, Emory University School of Medicine, Atlanta, GA 30322, USA.
| |
Collapse
|
99
|
Oaks AW, Zamarbide M, Tambunan DE, Santini E, Di Costanzo S, Pond HL, Johnson MW, Lin J, Gonzalez DM, Boehler JF, Wu GK, Klann E, Walsh CA, Manzini MC. Cc2d1a Loss of Function Disrupts Functional and Morphological Development in Forebrain Neurons Leading to Cognitive and Social Deficits. Cereb Cortex 2018; 27:1670-1685. [PMID: 26826102 DOI: 10.1093/cercor/bhw009] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Loss-of-function (LOF) mutations in CC2D1A cause a spectrum of neurodevelopmental disorders, including intellectual disability, autism spectrum disorder, and seizures, identifying a critical role for this gene in cognitive and social development. CC2D1A regulates intracellular signaling processes that are critical for neuronal function, but previous attempts to model the human LOF phenotypes have been prevented by perinatal lethality in Cc2d1a-deficient mice. To overcome this challenge, we generated a floxed Cc2d1a allele for conditional removal of Cc2d1a in the brain using Cre recombinase. While removal of Cc2d1a in neuronal progenitors using Cre expressed from the Nestin promoter still causes death at birth, conditional postnatal removal of Cc2d1a in the forebrain via calcium/calmodulin-dependent protein kinase II-alpha (CamKIIa) promoter-driven Cre generates animals that are viable and fertile with grossly normal anatomy. Analysis of neuronal morphology identified abnormal cortical dendrite organization and a reduction in dendritic spine density. These animals display deficits in neuronal plasticity and in spatial learning and memory that are accompanied by reduced sociability, hyperactivity, anxiety, and excessive grooming. Cc2d1a conditional knockout mice therefore recapitulate features of both cognitive and social impairment caused by human CC2D1A mutation, and represent a model that could provide much needed insights into the developmental mechanisms underlying nonsyndromic neurodevelopmental disorders.
Collapse
Affiliation(s)
- Adam W Oaks
- Department of Pharmacology and Physiology and Integrative Systems Biology, The George Washington University School of Medicine and Health Sciences, Washington, DC 20037, USA
| | - Marta Zamarbide
- Department of Pharmacology and Physiology and Integrative Systems Biology, The George Washington University School of Medicine and Health Sciences, Washington, DC 20037, USA
| | - Dimira E Tambunan
- Division of Genetics and Genomics and the Manton Center for Orphan Disease Research, Howard Hughes Medical Institute, Boston Children's Hospital, Boston, MA, USA.,Departments of Pediatrics and Neurology, Harvard Medical School, Boston, MA 02115, USA
| | - Emanuela Santini
- Center for Neural Science, New York University, New York, NY 10003, USA
| | - Stefania Di Costanzo
- Department of Pharmacology and Physiology and Integrative Systems Biology, The George Washington University School of Medicine and Health Sciences, Washington, DC 20037, USA
| | - Heather L Pond
- Department of Pharmacology and Physiology and Integrative Systems Biology, The George Washington University School of Medicine and Health Sciences, Washington, DC 20037, USA
| | - Mark W Johnson
- Department of Pharmacology and Physiology and Integrative Systems Biology, The George Washington University School of Medicine and Health Sciences, Washington, DC 20037, USA
| | - Jeff Lin
- Department of Psychology, The George Washington University, Washington, DC 20052, USA
| | - Dilenny M Gonzalez
- Division of Genetics and Genomics and the Manton Center for Orphan Disease Research, Howard Hughes Medical Institute, Boston Children's Hospital, Boston, MA, USA.,Departments of Pediatrics and Neurology, Harvard Medical School, Boston, MA 02115, USA
| | - Jessica F Boehler
- Department of Pharmacology and Physiology and Integrative Systems Biology, The George Washington University School of Medicine and Health Sciences, Washington, DC 20037, USA
| | - Guangying K Wu
- Department of Psychology, The George Washington University, Washington, DC 20052, USA
| | - Eric Klann
- Center for Neural Science, New York University, New York, NY 10003, USA
| | - Christopher A Walsh
- Division of Genetics and Genomics and the Manton Center for Orphan Disease Research, Howard Hughes Medical Institute, Boston Children's Hospital, Boston, MA, USA.,Departments of Pediatrics and Neurology, Harvard Medical School, Boston, MA 02115, USA
| | - M Chiara Manzini
- Department of Pharmacology and Physiology and Integrative Systems Biology, The George Washington University School of Medicine and Health Sciences, Washington, DC 20037, USA
| |
Collapse
|
100
|
Liu DC, Seimetz J, Lee KY, Kalsotra A, Chung HJ, Lu H, Tsai NP. Mdm2 mediates FMRP- and Gp1 mGluR-dependent protein translation and neural network activity. Hum Mol Genet 2018; 26:3895-3908. [PMID: 29016848 DOI: 10.1093/hmg/ddx276] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Accepted: 07/11/2017] [Indexed: 01/08/2023] Open
Abstract
Activating Group 1 (Gp1) metabotropic glutamate receptors (mGluRs), including mGluR1 and mGluR5, elicits translation-dependent neural plasticity mechanisms that are crucial to animal behavior and circuit development. Dysregulated Gp1 mGluR signaling has been observed in numerous neurological and psychiatric disorders. However, the molecular pathways underlying Gp1 mGluR-dependent plasticity mechanisms are complex and have been elusive. In this study, we identified a novel mechanism through which Gp1 mGluR mediates protein translation and neural plasticity. Using a multi-electrode array (MEA) recording system, we showed that activating Gp1 mGluR elevates neural network activity, as demonstrated by increased spontaneous spike frequency and burst activity. Importantly, we validated that elevating neural network activity requires protein translation and is dependent on fragile X mental retardation protein (FMRP), the protein that is deficient in the most common inherited form of mental retardation and autism, fragile X syndrome (FXS). In an effort to determine the mechanism by which FMRP mediates protein translation and neural network activity, we demonstrated that a ubiquitin E3 ligase, murine double minute-2 (Mdm2), is required for Gp1 mGluR-induced translation and neural network activity. Our data showed that Mdm2 acts as a translation suppressor, and FMRP is required for its ubiquitination and down-regulation upon Gp1 mGluR activation. These data revealed a novel mechanism by which Gp1 mGluR and FMRP mediate protein translation and neural network activity, potentially through de-repressing Mdm2. Our results also introduce an alternative way for understanding altered protein translation and brain circuit excitability associated with Gp1 mGluR in neurological diseases such as FXS.
Collapse
Affiliation(s)
- Dai-Chi Liu
- Department of Molecular and Integrative Physiology, School of Molecular and Cellular Biology.,Neuroscience Program
| | - Joseph Seimetz
- Department of Biochemistry, School of Molecular and Cellular Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Kwan Young Lee
- Department of Molecular and Integrative Physiology, School of Molecular and Cellular Biology
| | - Auinash Kalsotra
- Department of Biochemistry, School of Molecular and Cellular Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA.,Carl R.Woese Institute of Genomic Biology, University of Illinois, Champaign, IL 61801, USA
| | - Hee Jung Chung
- Department of Molecular and Integrative Physiology, School of Molecular and Cellular Biology.,Neuroscience Program.,Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Hua Lu
- Department of Biochemistry and Molecular Biology.,Tulane Cancer Center, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Nien-Pei Tsai
- Department of Molecular and Integrative Physiology, School of Molecular and Cellular Biology.,Neuroscience Program.,Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| |
Collapse
|