1
|
Reynolds KE, Huang E, Sabbineni M, Wiseman E, Murtaza N, Ahuja D, Napier M, Murphy KM, Singh KK, Scott AL. Purinergic Signalling Mediates Aberrant Excitability of Developing Neuronal Circuits in the Fmr1 Knockout Mouse Model. Mol Neurobiol 2024; 61:9507-9528. [PMID: 38652351 DOI: 10.1007/s12035-024-04181-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Accepted: 04/15/2024] [Indexed: 04/25/2024]
Abstract
Neuronal hyperexcitability within developing cortical circuits is a common characteristic of several heritable neurodevelopmental disorders, including Fragile X Syndrome (FXS), intellectual disability and autism spectrum disorders (ASD). While this aberrant circuitry is typically studied from a neuron-centric perspective, glial cells secrete soluble factors that regulate both neurite extension and synaptogenesis during development. The nucleotide-mediated purinergic signalling system is particularly instrumental in facilitating these effects. We recently reported that within a FXS animal model, the Fmr1 KO mouse, the purinergic signalling system is upregulated in cortical astrocytes leading to altered secretion of synaptogenic and plasticity-related proteins. In this study, we examined whether elevated astrocyte purinergic signalling also impacts neuronal morphology and connectivity of Fmr1 KO cortical neurons. Here, we found that conditioned media from primary Fmr1 KO astrocytes was sufficient to enhance neurite extension and complexity of both wildtype and Fmr1 KO neurons to a similar degree as UTP-mediated outgrowth. Significantly enhanced firing was also observed in Fmr1 KO neuron-astrocyte co-cultures grown on microelectrode arrays but was associated with large deficits in firing synchrony. The selective P2Y2 purinergic receptor antagonist AR-C 118925XX effectively normalized much of the aberrant Fmr1 KO activity, designating P2Y2 as a potential therapeutic target in FXS. These results not only demonstrate the importance of astrocyte soluble factors in the development of neural circuitry, but also show that P2Y purinergic receptors play a distinct role in pathological FXS neuronal activity.
Collapse
Affiliation(s)
- Kathryn E Reynolds
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| | - Eileen Huang
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| | - Monica Sabbineni
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| | - Eliza Wiseman
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| | - Nadeem Murtaza
- McMaster Stem Cell and Cancer Research Institute, McMaster University, Hamilton, ON, Canada
| | - Desmond Ahuja
- Department of Psychology, Neuroscience, and Behaviour, McMaster University, Hamilton, ON, Canada
| | - Matt Napier
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
- Department of Molecular and Cellular Biology, University of Guelph, 488 Gordon St, Guelph, ON, Canada
| | - Kathryn M Murphy
- Department of Psychology, Neuroscience, and Behaviour, McMaster University, Hamilton, ON, Canada
| | | | - Angela L Scott
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada.
- Department of Molecular and Cellular Biology, University of Guelph, 488 Gordon St, Guelph, ON, Canada.
| |
Collapse
|
2
|
Napier M, Reynolds K, Scott AL. Glial-mediated dysregulation of neurodevelopment in Fragile X Syndrome. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2023; 173:187-215. [PMID: 37993178 DOI: 10.1016/bs.irn.2023.08.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/24/2023]
Abstract
Astrocytes are highly involved in a multitude of developmental processes that are known to be dysregulated in Fragile X Syndrome. Here, we examine these processes individually and review the roles astrocytes play in contributing to the pathology of this syndrome. As a growing area of interest in the field, new and exciting insight is continually emerging. Understanding these glial-mediated roles is imperative for elucidating the underlying molecular mechanisms at play, not only in Fragile X Syndrome, but also other ASD-related disorders. Understanding these roles will be central to the future development of effective, clinically-relevant treatments of these disorders.
Collapse
Affiliation(s)
- M Napier
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, Canada; Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Canada
| | - K Reynolds
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Canada; Department of Neuroscience, Tufts University School of Medicine, Boston, United States
| | - A L Scott
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, Canada; Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Canada.
| |
Collapse
|
3
|
Scott DN, Frank MJ. Adaptive control of synaptic plasticity integrates micro- and macroscopic network function. Neuropsychopharmacology 2023; 48:121-144. [PMID: 36038780 PMCID: PMC9700774 DOI: 10.1038/s41386-022-01374-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 06/23/2022] [Accepted: 06/24/2022] [Indexed: 11/09/2022]
Abstract
Synaptic plasticity configures interactions between neurons and is therefore likely to be a primary driver of behavioral learning and development. How this microscopic-macroscopic interaction occurs is poorly understood, as researchers frequently examine models within particular ranges of abstraction and scale. Computational neuroscience and machine learning models offer theoretically powerful analyses of plasticity in neural networks, but results are often siloed and only coarsely linked to biology. In this review, we examine connections between these areas, asking how network computations change as a function of diverse features of plasticity and vice versa. We review how plasticity can be controlled at synapses by calcium dynamics and neuromodulatory signals, the manifestation of these changes in networks, and their impacts in specialized circuits. We conclude that metaplasticity-defined broadly as the adaptive control of plasticity-forges connections across scales by governing what groups of synapses can and can't learn about, when, and to what ends. The metaplasticity we discuss acts by co-opting Hebbian mechanisms, shifting network properties, and routing activity within and across brain systems. Asking how these operations can go awry should also be useful for understanding pathology, which we address in the context of autism, schizophrenia and Parkinson's disease.
Collapse
Affiliation(s)
- Daniel N Scott
- Cognitive Linguistic, and Psychological Sciences, Brown University, Providence, RI, USA.
- Carney Institute for Brain Science, Brown University, Providence, RI, USA.
| | - Michael J Frank
- Cognitive Linguistic, and Psychological Sciences, Brown University, Providence, RI, USA.
- Carney Institute for Brain Science, Brown University, Providence, RI, USA.
| |
Collapse
|
4
|
Afshar S, Lule S, Yuan G, Qu X, Pan C, Whalen M, Brownell AL, Mody M. Longitudinal PET studies of mGluR5 in FXS using an FMR1 knockout mouse model. Transl Neurosci 2022; 13:80-92. [PMID: 35582646 PMCID: PMC9055256 DOI: 10.1515/tnsci-2022-0217] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 03/28/2022] [Accepted: 04/04/2022] [Indexed: 11/15/2022] Open
Abstract
Abstract
Fragile X syndrome (FXS) is a monogenic disorder characterized by intellectual disability and behavioral challenges. It is caused by aberrant methylation of the fragile X mental retardation 1 (FMR1) gene. Given the failure of clinical trials in FXS and growing evidence of a role of metabotropic glutamate subtype 5 receptors (mGluR5) in the pathophysiology of the disorder, we investigated mGluR5 function in FMR1 Knockout (FMR1-KO) mice and age- and sex-matched control mice using longitudinal positron emission tomography (PET) imaging to better understand the disorder. The studies were repeated at four time points to examine age- and disease-induced changes in mGluR5 availability using 3-fluoro-[18F]5-(2-pyridinylethynyl)benzonitrile ([18F]FPEB). We found that the binding potential (BP) of [18F]FPEB was significantly lower in the KO mice in mGluR5-implicated brain areas including striatum, cortex, hippocampus, thalamus, and olfactory bulb. The BP also changed with age, regardless of disorder status, increasing in early adulthood in male but not in female mice before decreasing later in both sexes. The difference in mGluR5 availability between the FMR1-KO and control mice and the change in BP in the KO mice as a function of age and sex illustrate the nature of the disorder and its progression, providing mechanistic insights for treatment design.
Collapse
Affiliation(s)
- Sepideh Afshar
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School , Charlestown , 02129 MA , United States of America
| | - Sevda Lule
- Department of Pediatrics, Massachusetts General Hospital, Harvard Medical School , Charlestown , 02129 MA , United States of America
| | - Gengyang Yuan
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School , Charlestown , 02129 MA , United States of America
| | - Xiying Qu
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School , Charlestown , 02129 MA , United States of America
| | - Chuzhi Pan
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School , Charlestown , 02129 MA , United States of America
| | - Michael Whalen
- Department of Pediatrics, Massachusetts General Hospital, Harvard Medical School , Charlestown , 02129 MA , United States of America
| | - Anna-Liisa Brownell
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School , Charlestown , 02129 MA , United States of America
| | - Maria Mody
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School , Charlestown , 02129 MA , USA
| |
Collapse
|
5
|
Han YMY, Yau SY, Chan MMY, Wong CK, Chan AS. Altered Cytokine and BDNF Levels in Individuals with Autism Spectrum Disorders. Brain Sci 2022; 12:brainsci12040460. [PMID: 35447993 PMCID: PMC9026457 DOI: 10.3390/brainsci12040460] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 03/20/2022] [Accepted: 03/25/2022] [Indexed: 12/10/2022] Open
Abstract
Previous studies have shown that immunological factors are involved in the pathogenesis of autism spectrum disorders (ASDs). The present study examined whether immunological abnormalities are associated with cognitive and behavioral deficits in children with ASD and whether children with ASD show different immunological biomarkers and brain-derived neurotrophic factor BDNF levels than typically developing (TD) children. Sixteen children with TD and 18 children with ASD, aged 6–18 years, voluntarily participated in the study. Participants’ executive functions were measured using neuropsychological tests, and behavioral measures were measured using parent ratings. Immunological measures were assessed by measuring the participants’ blood serum levels of chemokine ligand 2 (CCL2) and chemokine ligand 5 (CCL5). Children with ASD showed greater deficits in cognitive functions as well as altered levels of immunological measures when compared to TD children, and their cognitive functions and behavioral deficits were significantly associated with increased CCL5 levels and decreased BDNF levels. These results provide evidence to support the notion that altered immune functions and neurotrophin deficiency are involved in the pathogenesis of ASD.
Collapse
Affiliation(s)
- Yvonne M. Y. Han
- Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, Hong Kong SAR, China; (S.-Y.Y.); (M.M.Y.C.)
- Correspondence: ; Tel.: +852-2766-7578
| | - Suk-Yu Yau
- Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, Hong Kong SAR, China; (S.-Y.Y.); (M.M.Y.C.)
| | - Melody M. Y. Chan
- Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, Hong Kong SAR, China; (S.-Y.Y.); (M.M.Y.C.)
| | - Chun-Kwok Wong
- Department of Chemical Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong SAR, China;
| | - Agnes S. Chan
- Department of Psychology, The Chinese University of Hong Kong, Hong Kong SAR, China;
| |
Collapse
|
6
|
Wang XX, Zhang S, Dong PP, Li YH, Zhang L, Shi SH, Yu ZQ, Chen S. MRCKβ links Dasm1 to actin rearrangements to promote dendrite development. J Biol Chem 2021; 296:100730. [PMID: 33933448 PMCID: PMC8191314 DOI: 10.1016/j.jbc.2021.100730] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Revised: 04/23/2021] [Accepted: 04/28/2021] [Indexed: 01/12/2023] Open
Abstract
Proper dendrite morphogenesis and synapse formation are essential for neuronal development and function. Dasm1, a member of the immunoglobulin superfamily, is known to promote dendrite outgrowth and excitatory synapse maturation in vitro. However, the in vivo function of Dasm1 in neuronal development and the underlying mechanisms are not well understood. To learn more, Dasm1 knockout mice were constructed and employed to confirm that Dasm1 regulates dendrite arborization and spine formation in vivo. We performed a yeast two-hybrid screen using Dasm1, revealing MRCKβ as a putative partner; additional lines of evidence confirmed this interaction and identified cytoplasmic proline-rich region (823–947 aa) of Dasm1 and MRCKβ self-activated kinase domain (CC1, 410–744 aa) as necessary and sufficient for binding. Using co-immunoprecipitation assay, autophosphorylation assay, and BS3 cross-linking assay, we show that Dasm1 binding triggers a change in MRCKβ’s conformation and subsequent dimerization, resulting in autophosphorylation and activation. Activated MRCKβ in turn phosphorylates a class 2 regulatory myosin light chain, which leads to enhanced actin rearrangement, causing the dendrite outgrowth and spine formation observed before. Removal of Dasm1 in mice leads to behavioral abnormalities. Together, these results reveal a crucial molecular pathway mediating cell surface and intracellular signaling communication to regulate actin dynamics and neuronal development in the mammalian brain.
Collapse
Affiliation(s)
- Xiao-Xiao Wang
- NHC Key Laboratory of Glycoconjugate Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China; Department of Gastroenterology and Hepatology, Shanghai Institute of Liver Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Si Zhang
- NHC Key Laboratory of Glycoconjugate Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Ping-Ping Dong
- Department of Gastroenterology and Hepatology, Shanghai Institute of Liver Diseases, Zhongshan Hospital, Fudan University, Shanghai, China; Department of Surgery, Faculty of Medicine, Centre for Cancer Research, The University of Hong Kong, Hong Kong, China
| | - Yao-Hua Li
- NHC Key Laboratory of Glycoconjugate Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Li Zhang
- NHC Key Laboratory of Glycoconjugate Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Song-Hai Shi
- IDG/McGovern Institute for Brain Research, Tsinghua-Peking Center for Life Sciences, Beijing Frontier Research Center of Biological Structure, Beijing Advanced Innovation Center for Structural Biology, School of Life Sciences, Tsinghua University, Beijing, China; Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Zhi-Qiang Yu
- NHC Key Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai, China; Eye Department, Eye & ENT Hospital, Fudan University, Shanghai, China.
| | - She Chen
- NHC Key Laboratory of Glycoconjugate Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China.
| |
Collapse
|
7
|
Reynolds KE, Wong CR, Scott AL. Astrocyte-mediated purinergic signaling is upregulated in a mouse model of Fragile X syndrome. Glia 2021; 69:1816-1832. [PMID: 33754385 DOI: 10.1002/glia.23997] [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: 07/09/2020] [Revised: 03/10/2021] [Accepted: 03/11/2021] [Indexed: 12/12/2022]
Abstract
Fragile X syndrome (FXS) is the leading monogenic cause of intellectual disability and autism spectrum disorders. With increasing investigation into the molecular mechanisms underlying FXS, there is growing evidence that perturbations in glial signaling are widely associated with neurological pathology. Purinergic signaling, which utilizes nucleoside triphosphates as signaling molecules, provides one of the most ubiquitous signaling systems for glial-neuronal and glial-glial crosstalk. Here, we sought to identify whether purinergic signaling is dysregulated within the FXS mouse cortex, and whether this dysregulation contributes to aberrant intercellular communication. In primary astrocyte cultures derived from the Fmr1 knockout (KO) mouse model of FXS, we found that application of exogenous ATP and UTP evoked elevated intracellular calcium responses compared to wildtype levels. Accordingly, purinergic P2Y2 and P2Y6 receptor expression was increased in Fmr1 KO astrocytes both in vitro and in acutely dissociated tissue, while P2Y antagonism via suramin prevented intracellular calcium elevations, suggesting a role for these receptors in aberrant FXS astrocyte activation. To investigate the impact of elevated purinergic signaling on astrocyte-mediated synaptogenesis, we quantified synaptogenic protein TSP-1, known to be regulated by P2Y activation. TSP-1 secretion and expression were both heightened in Fmr1 KO vs wildtype astrocytes following UTP application, while naïve TSP-1 cortical expression was also transiently elevated in vivo, indicating increased potential for excitatory TSP-1-mediated synaptogenesis in the FXS cortex. Together, our results demonstrate novel and significant purinergic signaling elevations in Fmr1 KO astrocytes, which may serve as a potential therapeutic target to mitigate the signaling aberrations observed in FXS.
Collapse
Affiliation(s)
- Kathryn E Reynolds
- Neuroscience Graduate Program, McMaster University, Hamilton, Ontario, Canada
| | - Chloe R Wong
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Angela L Scott
- Neuroscience Graduate Program, McMaster University, Hamilton, Ontario, Canada.,Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
8
|
Motanis H, Buonomano D. Decreased reproducibility and abnormal experience-dependent plasticity of network dynamics in Fragile X circuits. Sci Rep 2020; 10:14535. [PMID: 32884028 PMCID: PMC7471942 DOI: 10.1038/s41598-020-71333-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 08/10/2020] [Indexed: 02/06/2023] Open
Abstract
Fragile X syndrome is a neurodevelopmental disorder associated with a broad range of neural phenotypes. Interpreting these findings has proven challenging because some phenotypes may reflect compensatory mechanisms or normal forms of plasticity differentially engaged by experiential differences. To help minimize compensatory and experiential influences, we used an ex vivo approach to study network dynamics and plasticity of cortical microcircuits. In Fmr1-/y circuits, the spatiotemporal structure of Up-states was less reproducible, suggesting alterations in the plasticity mechanisms governing network activity. Chronic optical stimulation revealed normal homeostatic plasticity of Up-states, however, Fmr1-/y circuits exhibited abnormal experience-dependent plasticity as they did not adapt to chronically presented temporal patterns in an interval-specific manner. These results, suggest that while homeostatic plasticity is normal, Fmr1-/y circuits exhibit deficits in the ability to orchestrate multiple forms of synaptic plasticity and to adapt to sensory patterns in an experience-dependent manner-which is likely to contribute to learning deficits.
Collapse
Affiliation(s)
- Helen Motanis
- Departments of Neurobiology and Psychology, and Integrative Center for Learning and Memory, University of California, 630 Charles E Young Dr S, Center for Health Sciences Building, Los Angeles, CA, 90095, USA
| | - Dean Buonomano
- Departments of Neurobiology and Psychology, and Integrative Center for Learning and Memory, University of California, 630 Charles E Young Dr S, Center for Health Sciences Building, Los Angeles, CA, 90095, USA.
| |
Collapse
|
9
|
McCullagh EA, Rotschafer SE, Auerbach BD, Klug A, Kaczmarek LK, Cramer KS, Kulesza RJ, Razak KA, Lovelace JW, Lu Y, Koch U, Wang Y. Mechanisms underlying auditory processing deficits in Fragile X syndrome. FASEB J 2020; 34:3501-3518. [PMID: 32039504 DOI: 10.1096/fj.201902435r] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 12/31/2019] [Accepted: 01/18/2020] [Indexed: 01/14/2023]
Abstract
Autism spectrum disorders (ASD) are strongly associated with auditory hypersensitivity or hyperacusis (difficulty tolerating sounds). Fragile X syndrome (FXS), the most common monogenetic cause of ASD, has emerged as a powerful gateway for exploring underlying mechanisms of hyperacusis and auditory dysfunction in ASD. This review discusses examples of disruption of the auditory pathways in FXS at molecular, synaptic, and circuit levels in animal models as well as in FXS individuals. These examples highlight the involvement of multiple mechanisms, from aberrant synaptic development and ion channel deregulation of auditory brainstem circuits, to impaired neuronal plasticity and network hyperexcitability in the auditory cortex. Though a relatively new area of research, recent discoveries have increased interest in auditory dysfunction and mechanisms underlying hyperacusis in this disorder. This rapidly growing body of data has yielded novel research directions addressing critical questions regarding the timing and possible outcomes of human therapies for auditory dysfunction in ASD.
Collapse
Affiliation(s)
- Elizabeth A McCullagh
- Department of Physiology and Biophysics, University of Colorado Anschutz, Aurora, CO, USA.,Department of Integrative Biology, Oklahoma State University, Stillwater, OK, USA
| | - Sarah E Rotschafer
- Department of Neurobiology and Behavior, University of California, Irvine, CA, USA.,Department of Biomedical Sciences, Mercer University School of Medicine, Savannah, GA, USA
| | - Benjamin D Auerbach
- Center for Hearing and Deafness, Department of Communicative Disorders & Sciences, SUNY at Buffalo, Buffalo, NY, USA
| | - Achim Klug
- Department of Physiology and Biophysics, University of Colorado Anschutz, Aurora, CO, USA
| | - Leonard K Kaczmarek
- Departments of Pharmacology and Cellular and Molecular Physiology, Yale University, New Haven, CT, USA
| | - Karina S Cramer
- Department of Neurobiology and Behavior, University of California, Irvine, CA, USA
| | - Randy J Kulesza
- Department of Anatomy, Lake Erie College of Osteopathic Medicine, Erie, PA, USA
| | - Khaleel A Razak
- Department of Psychology, University of California, Riverside, CA, USA
| | | | - Yong Lu
- Department of Anatomy and Neurobiology, College of Medicine, Northeast Ohio Medical University, Rootstown, OH, USA
| | - Ursula Koch
- Institute of Biology, Neurophysiology, Freie Universität Berlin, Berlin, Germany
| | - Yuan Wang
- Department of Biomedical Sciences, Program in Neuroscience, Florida State University, Tallahassee, FL, USA
| |
Collapse
|
10
|
Pekarek BT, Hunt PJ, Arenkiel BR. Oxytocin and Sensory Network Plasticity. Front Neurosci 2020; 14:30. [PMID: 32063835 PMCID: PMC7000660 DOI: 10.3389/fnins.2020.00030] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 01/10/2020] [Indexed: 12/22/2022] Open
Abstract
An essential characteristic of nervous systems is their capacity to reshape functional connectivity in response to physiological and environmental cues. Endogenous signals, including neuropeptides, governs nervous system plasticity. Particularly, oxytocin has been recognized for its role in mediating activity-dependent circuit changes. These oxytocin-dependent changes occur at the synaptic level and consequently shape the cellular composition of circuits. Here we discuss recent advances that illustrate how oxytocin functions to reshape neural circuitry in response to environmental changes. Excitingly, recent findings pave the way for promising therapeutic applications of oxytocin to treat neurodevelopmental and neuropsychiatric diseases.
Collapse
Affiliation(s)
- Brandon T. Pekarek
- Genetics and Genomics Program, Baylor College of Medicine, Houston, TX, United States
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, United States
| | - Patrick J. Hunt
- Genetics and Genomics Program, Baylor College of Medicine, Houston, TX, United States
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, United States
- Medical Scientist Training Program, Baylor College of Medicine, Houston, TX, United States
| | - Benjamin R. Arenkiel
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, United States
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, United States
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, Houston, TX, United States
| |
Collapse
|
11
|
Huang JY, Lu HC. mGluR5 Tunes NGF/TrkA Signaling to Orient Spiny Stellate Neuron Dendrites Toward Thalamocortical Axons During Whisker-Barrel Map Formation. Cereb Cortex 2019; 28:1991-2006. [PMID: 28453662 DOI: 10.1093/cercor/bhx105] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2016] [Indexed: 12/12/2022] Open
Abstract
Neurons receive and integrate synaptic inputs at their dendrites, thus dendritic patterning shapes neural connectivity and behavior. Aberrant dendritogenesis is present in neurodevelopmental disorders such as Down's syndrome and autism. Abnormal glutamatergic signaling has been observed in these diseases, as has dysfunction of the metabotropic glutamate receptor 5 (mGluR5). Deleting mGluR5 in cortical glutamatergic neurons disrupted their coordinated dendritic outgrowth toward thalamocortical axons and perturbed somatosensory circuits. Here we show that mGluR5 loss-of-function disrupts dendritogenesis of cortical neurons by increasing mRNA levels of nerve growth factor (NGF) and fibroblast growth factor 10 (FGF10), in part through calcium-permeable AMPA receptors (CP-AMPARs), as the whisker-barrel map is forming. Postnatal NGF and FGF10 expression in cortical layer IV spiny stellate neurons differentially impacted dendritic patterns. Remarkably, NGF-expressing neurons exhibited dendritic patterns resembling mGluR5 knockout neurons: increased total dendritic length/complexity and reduced polarity. Furthermore, suppressing the kinase activity of TrkA, a major NGF receptor, prevents aberrant dendritic patterning in barrel cortex of mGluR5 knockout neurons. These results reveal novel roles for NGF-TrkA signaling and CP-AMPARs for proper dendritic development of cortical neurons. This is the first in vivo demonstration that cortical neuronal NGF expression modulates dendritic patterning during postnatal brain development.
Collapse
Affiliation(s)
- Jui-Yen Huang
- Department of Psychological and Brain Sciences, the Linda and Jack Gill Center for Biomolecular Sciences, Indiana University, Bloomington, IN 47405, USA.,The Cain Foundation Laboratories, Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX 77030, USA.,Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Hui-Chen Lu
- Department of Psychological and Brain Sciences, the Linda and Jack Gill Center for Biomolecular Sciences, Indiana University, Bloomington, IN 47405, USA.,The Cain Foundation Laboratories, Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX 77030, USA.,Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
12
|
Abstract
Fragile X syndrome (FXS) is a neurodevelopmental disorder that causes intellectual disability. It is a leading known genetic cause of autism. In addition to cognitive, social, and communication deficits, humans with FXS demonstrate abnormal sensory processing including sensory hypersensitivity. Sensory hypersensitivity commonly manifests as auditory, tactile, or visual defensiveness or avoidance. Clinical, behavioral, and electrophysiological studies consistently show auditory hypersensitivity, impaired habituation to repeated sounds, and reduced auditory attention in humans with FXS. Children with FXS also exhibit significant visuospatial impairments. Studies in infants and toddlers with FXS have documented impairments in processing texture-defined motion stimuli, temporal flicker, perceiving ordinal numerical sequence, and the ability to maintain the identity of dynamic object information during occlusion. Consistent with the observations in humans with FXS, fragile X mental retardation 1 ( Fmr1) gene knockout (KO) rodent models of FXS also show seizures, abnormal visual-evoked responses, auditory hypersensitivity, and abnormal processing at multiple levels of the auditory system, including altered acoustic startle responses. Among other sensory symptoms, individuals with FXS exhibit tactile defensiveness. Fmr1 KO mice also show impaired encoding of tactile stimulation frequency and larger size of receptive fields in the somatosensory cortex. Since sensory deficits are relatively more tractable from circuit mechanisms and developmental perspectives than more complex social behaviors, the focus of this review is on clinical, functional, and structural studies that outline the auditory, visual, and somatosensory processing deficits in FXS. The similarities in sensory phenotypes between humans with FXS and animal models suggest a likely conservation of basic sensory processing circuits across species and may provide a translational platform to not just develop biomarkers but also to understand underlying mechanisms. We argue that preclinical studies in animal models of FXS can facilitate the ongoing search for new therapeutic approaches in FXS by understanding mechanisms of basic sensory processing circuits and behaviors that are conserved across species.
Collapse
Affiliation(s)
- Maham Rais
- 1 Division of Biomedical Sciences, University of California Riverside School of Medicine, CA, USA.,2 Biomedical Sciences Graduate Program, University of California Riverside, CA, USA
| | - Devin K Binder
- 1 Division of Biomedical Sciences, University of California Riverside School of Medicine, CA, USA.,2 Biomedical Sciences Graduate Program, University of California Riverside, CA, USA.,3 Neuroscience Graduate Program, University of California Riverside, CA, USA
| | - Khaleel A Razak
- 2 Biomedical Sciences Graduate Program, University of California Riverside, CA, USA.,3 Neuroscience Graduate Program, University of California Riverside, CA, USA.,4 Psychology Department, University of California Riverside, CA, USA
| | - Iryna M Ethell
- 1 Division of Biomedical Sciences, University of California Riverside School of Medicine, CA, USA.,2 Biomedical Sciences Graduate Program, University of California Riverside, CA, USA.,3 Neuroscience Graduate Program, University of California Riverside, CA, USA
| |
Collapse
|
13
|
Verma V, Paul A, Amrapali Vishwanath A, Vaidya B, Clement JP. Understanding intellectual disability and autism spectrum disorders from common mouse models: synapses to behaviour. Open Biol 2019; 9:180265. [PMID: 31185809 PMCID: PMC6597757 DOI: 10.1098/rsob.180265] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Normal brain development is highly dependent on the timely coordinated actions of genetic and environmental processes, and an aberration can lead to neurodevelopmental disorders (NDDs). Intellectual disability (ID) and autism spectrum disorders (ASDs) are a group of co-occurring NDDs that affect between 3% and 5% of the world population, thus presenting a great challenge to society. This problem calls for the need to understand the pathobiology of these disorders and to design new therapeutic strategies. One approach towards this has been the development of multiple analogous mouse models. This review discusses studies conducted in the mouse models of five major monogenic causes of ID and ASDs: Fmr1, Syngap1, Mecp2, Shank2/3 and Neuroligins/Neurnexins. These studies reveal that, despite having a diverse molecular origin, the effects of these mutations converge onto similar or related aetiological pathways, consequently giving rise to the typical phenotype of cognitive, social and emotional deficits that are characteristic of ID and ASDs. This convergence, therefore, highlights common pathological nodes that can be targeted for therapy. Other than conventional therapeutic strategies such as non-pharmacological corrective methods and symptomatic alleviation, multiple studies in mouse models have successfully proved the possibility of pharmacological and genetic therapy enabling functional recovery.
Collapse
Affiliation(s)
- Vijaya Verma
- Neuroscience Unit, Jawaharlal Nehru Centre for Advanced Scientific Research , Jakkur, Bengaluru 560 064, Karnataka, India
| | - Abhik Paul
- Neuroscience Unit, Jawaharlal Nehru Centre for Advanced Scientific Research , Jakkur, Bengaluru 560 064, Karnataka, India
| | - Anjali Amrapali Vishwanath
- Neuroscience Unit, Jawaharlal Nehru Centre for Advanced Scientific Research , Jakkur, Bengaluru 560 064, Karnataka, India
| | - Bhupesh Vaidya
- Neuroscience Unit, Jawaharlal Nehru Centre for Advanced Scientific Research , Jakkur, Bengaluru 560 064, Karnataka, India
| | - James P Clement
- Neuroscience Unit, Jawaharlal Nehru Centre for Advanced Scientific Research , Jakkur, Bengaluru 560 064, Karnataka, India
| |
Collapse
|
14
|
Yau SY, Bettio L, Chiu J, Chiu C, Christie BR. Fragile-X Syndrome Is Associated With NMDA Receptor Hypofunction and Reduced Dendritic Complexity in Mature Dentate Granule Cells. Front Mol Neurosci 2019; 11:495. [PMID: 30705620 PMCID: PMC6344420 DOI: 10.3389/fnmol.2018.00495] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Accepted: 12/27/2018] [Indexed: 12/11/2022] Open
Abstract
Fragile X syndrome (FXS) is the most common form of inherited intellectual disability. It is caused by the overexpansion of cytosine-guanine-guanine (CGG) trinucleotide in Fmr1 gene, resulting in complete loss of the fragile X mental retardation protein (FMRP). Previous studies using Fmr1 knockout (Fmr1 KO) mice have suggested that a N-methyl-D-aspartate receptors (NMDAR) hypofunction in the hippocampal dentate gyrus may partly contribute to cognitive impairments in FXS. Since activation of NMDAR plays an important role in dendritic arborization during neuronal development, we examined whether deficits in NMDAR function are associated with alterations in dendritic complexity in the hippocampal dentate region. The dentate granule cell layer (GCL) presents active postnatal neurogenesis, and consists of a heterogenous neuronal population with gradient ages from the superficial to its deep layer. Here, we show that neurons with multiple primary dendrites that reside in the outer GCL of Fmr1 KO mice display significantly smaller NMDAR excitatory post-synaptic currents (EPSCs) and a higher α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) to NMDA ratio in comparison to their wild-type counterparts. These deficits were associated with a significant decrease in dendritic complexity, with both dendritic length and number of intersections being significantly reduced. In contrast, although neurons with a single primary dendrite resided in the inner GCL of Fmr1 KO mice had a trend toward a reduction in NMDAR EPSCs and a higher AMPA/NMDA ratio, no alterations were found in dendritic complexity at this developmental stage. Our data indicate that the loss of FMRP causes NMDAR deficits and reduced dendritic complexity in granule neurons with multiple primary dendrites which are thought to be more mature in the GCL.
Collapse
Affiliation(s)
- Suk-Yu Yau
- Division of Medical Sciences, Island Medical Program, University of Victoria, Victoria, BC, Canada
| | - Luis Bettio
- Division of Medical Sciences, Island Medical Program, University of Victoria, Victoria, BC, Canada
| | - Jason Chiu
- Division of Medical Sciences, Island Medical Program, University of Victoria, Victoria, BC, Canada
| | - Christine Chiu
- Division of Medical Sciences, Island Medical Program, University of Victoria, Victoria, BC, Canada
| | - Brian R Christie
- Division of Medical Sciences, Island Medical Program, University of Victoria, Victoria, BC, Canada
| |
Collapse
|
15
|
Wang X, Zorio DAR, Schecterson L, Lu Y, Wang Y. Postsynaptic FMRP Regulates Synaptogenesis In Vivo in the Developing Cochlear Nucleus. J Neurosci 2018; 38:6445-6460. [PMID: 29950504 PMCID: PMC6052239 DOI: 10.1523/jneurosci.0665-18.2018] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 06/18/2018] [Accepted: 06/20/2018] [Indexed: 12/29/2022] Open
Abstract
A global loss of the fragile X mental retardation protein (FMRP; encoded by the Fmr1 gene) leads to sensory dysfunction and intellectual disabilities. One underlying mechanism of these phenotypes is structural and functional deficits in synapses. Here, we determined the autonomous function of postsynaptic FMRP in circuit formation, synaptogenesis, and synaptic maturation. In normal cochlea nucleus, presynaptic auditory axons form large axosomatic endbulb synapses on cell bodies of postsynaptic bushy neurons. In ovo electroporation of drug-inducible Fmr1-shRNA constructs produced a mosaicism of FMRP expression in chicken (either sex) bushy neurons, leading to reduced FMRP levels in transfected, but not neighboring nontransfected, neurons. Structural analyses revealed that postsynaptic FMRP reduction led to smaller size and abnormal morphology of individual presynaptic endbulbs at both early and later developmental stages. We further examined whether FMRP reduction affects dendritic development, as a potential mechanism underlying defective endbulb formation. Normally, chicken bushy neurons grow extensive dendrites at early stages and retract these dendrites when endbulbs begin to form. Neurons transfected with Fmr1 shRNA exhibited a remarkable delay in branch retraction, failing to provide necessary somatic surface for timely formation and growth of large endbulbs. Patch-clamp recording verified functional consequences of dendritic and synaptic deficits on neurotransmission, showing smaller amplitudes and slower kinetics of spontaneous and evoked EPSCs. Together, these data demonstrate that proper levels of postsynaptic FMRP are required for timely maturation of somatodendritic morphology, a delay of which may affect synaptogenesis and thus contribute to long-lasting deficits of excitatory synapses.SIGNIFICANCE STATEMENT Fragile X mental retardation protein (FMRP) regulates a large variety of neuronal activities. A global loss of FMRP affects neural circuit development and synaptic function, leading to fragile X syndrome (FXS). Using temporally and spatially controlled genetic manipulations, this study provides the first in vivo report that autonomous FMRP regulates multiple stages of dendritic development, and that selective reduction of postsynaptic FMRP leads to abnormal development of excitatory presynaptic terminals and compromised neurotransmission. These observations demonstrate secondary influence of developmentally transient deficits in neuronal morphology and connectivity to the development of long-lasting synaptic pathology in FXS.
Collapse
Affiliation(s)
- Xiaoyu Wang
- Department of Biomedical Science, Program in Neuroscience, Florida State University College of Medicine, Tallahassee, Florida 32306
| | - Diego A R Zorio
- Department of Biomedical Science, Program in Neuroscience, Florida State University College of Medicine, Tallahassee, Florida 32306
| | - Leslayann Schecterson
- Department of Otolaryngology, Bloedel Hearing Research Center, University of Washington, Seattle, Washington 98195, and
| | - Yong Lu
- Department of Anatomy and Neurobiology, College of Medicine, Northeast Ohio Medical University, Rootstown, Ohio 44272
| | - Yuan Wang
- Department of Biomedical Science, Program in Neuroscience, Florida State University College of Medicine, Tallahassee, Florida 32306,
| |
Collapse
|
16
|
Yau S, Bettio L, Vetrici M, Truesdell A, Chiu C, Chiu J, Truesdell E, Christie B. Chronic minocycline treatment improves hippocampal neuronal structure, NMDA receptor function, and memory processing in Fmr1 knockout mice. Neurobiol Dis 2018; 113:11-22. [DOI: 10.1016/j.nbd.2018.01.014] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Revised: 12/18/2017] [Accepted: 01/19/2018] [Indexed: 10/18/2022] Open
|
17
|
Varghese M, Keshav N, Jacot-Descombes S, Warda T, Wicinski B, Dickstein DL, Harony-Nicolas H, De Rubeis S, Drapeau E, Buxbaum JD, Hof PR. Autism spectrum disorder: neuropathology and animal models. Acta Neuropathol 2017; 134:537-566. [PMID: 28584888 PMCID: PMC5693718 DOI: 10.1007/s00401-017-1736-4] [Citation(s) in RCA: 318] [Impact Index Per Article: 39.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Revised: 05/30/2017] [Accepted: 05/31/2017] [Indexed: 12/13/2022]
Abstract
Autism spectrum disorder (ASD) has a major impact on the development and social integration of affected individuals and is the most heritable of psychiatric disorders. An increase in the incidence of ASD cases has prompted a surge in research efforts on the underlying neuropathologic processes. We present an overview of current findings in neuropathology studies of ASD using two investigational approaches, postmortem human brains and ASD animal models, and discuss the overlap, limitations, and significance of each. Postmortem examination of ASD brains has revealed global changes including disorganized gray and white matter, increased number of neurons, decreased volume of neuronal soma, and increased neuropil, the last reflecting changes in densities of dendritic spines, cerebral vasculature and glia. Both cortical and non-cortical areas show region-specific abnormalities in neuronal morphology and cytoarchitectural organization, with consistent findings reported from the prefrontal cortex, fusiform gyrus, frontoinsular cortex, cingulate cortex, hippocampus, amygdala, cerebellum and brainstem. The paucity of postmortem human studies linking neuropathology to the underlying etiology has been partly addressed using animal models to explore the impact of genetic and non-genetic factors clinically relevant for the ASD phenotype. Genetically modified models include those based on well-studied monogenic ASD genes (NLGN3, NLGN4, NRXN1, CNTNAP2, SHANK3, MECP2, FMR1, TSC1/2), emerging risk genes (CHD8, SCN2A, SYNGAP1, ARID1B, GRIN2B, DSCAM, TBR1), and copy number variants (15q11-q13 deletion, 15q13.3 microdeletion, 15q11-13 duplication, 16p11.2 deletion and duplication, 22q11.2 deletion). Models of idiopathic ASD include inbred rodent strains that mimic ASD behaviors as well as models developed by environmental interventions such as prenatal exposure to sodium valproate, maternal autoantibodies, and maternal immune activation. In addition to replicating some of the neuropathologic features seen in postmortem studies, a common finding in several animal models of ASD is altered density of dendritic spines, with the direction of the change depending on the specific genetic modification, age and brain region. Overall, postmortem neuropathologic studies with larger sample sizes representative of the various ASD risk genes and diverse clinical phenotypes are warranted to clarify putative etiopathogenic pathways further and to promote the emergence of clinically relevant diagnostic and therapeutic tools. In addition, as genetic alterations may render certain individuals more vulnerable to developing the pathological changes at the synapse underlying the behavioral manifestations of ASD, neuropathologic investigation using genetically modified animal models will help to improve our understanding of the disease mechanisms and enhance the development of targeted treatments.
Collapse
Affiliation(s)
- Merina Varghese
- Fishberg Department of Neuroscience, Icahn School of Medicine at Mount Sinai, Box 1639, One Gustave L. Levy Place, New York, NY, 10029, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Neha Keshav
- Fishberg Department of Neuroscience, Icahn School of Medicine at Mount Sinai, Box 1639, One Gustave L. Levy Place, New York, NY, 10029, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Sarah Jacot-Descombes
- Fishberg Department of Neuroscience, Icahn School of Medicine at Mount Sinai, Box 1639, One Gustave L. Levy Place, New York, NY, 10029, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Unit of Psychiatry, Department of Children and Teenagers, University Hospitals and School of Medicine, Geneva, CH-1205, Switzerland
| | - Tahia Warda
- Fishberg Department of Neuroscience, Icahn School of Medicine at Mount Sinai, Box 1639, One Gustave L. Levy Place, New York, NY, 10029, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Bridget Wicinski
- Fishberg Department of Neuroscience, Icahn School of Medicine at Mount Sinai, Box 1639, One Gustave L. Levy Place, New York, NY, 10029, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Dara L Dickstein
- Fishberg Department of Neuroscience, Icahn School of Medicine at Mount Sinai, Box 1639, One Gustave L. Levy Place, New York, NY, 10029, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Pathology, Uniformed Services University of the Health Sciences, Bethesda, MD, 20814, USA
| | - Hala Harony-Nicolas
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Silvia De Rubeis
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Elodie Drapeau
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Joseph D Buxbaum
- Fishberg Department of Neuroscience, Icahn School of Medicine at Mount Sinai, Box 1639, One Gustave L. Levy Place, New York, NY, 10029, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Patrick R Hof
- Fishberg Department of Neuroscience, Icahn School of Medicine at Mount Sinai, Box 1639, One Gustave L. Levy Place, New York, NY, 10029, USA.
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
| |
Collapse
|
18
|
Sakano H, Zorio DAR, Wang X, Ting YS, Noble WS, MacCoss MJ, Rubel EW, Wang Y. Proteomic analyses of nucleus laminaris identified candidate targets of the fragile X mental retardation protein. J Comp Neurol 2017; 525:3341-3359. [PMID: 28685837 DOI: 10.1002/cne.24281] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Revised: 06/23/2017] [Accepted: 07/04/2017] [Indexed: 12/17/2022]
Abstract
The avian nucleus laminaris (NL) is a brainstem nucleus necessary for binaural processing, analogous in structure and function to the mammalian medial superior olive. In chickens (Gallus gallus), NL is a well-studied model system for activity-dependent neural plasticity. Its neurons have bipolar extension of dendrites, which receive segregated inputs from two ears and display rapid and compartment-specific reorganization in response to unilateral changes in auditory input. More recently, fragile X mental retardation protein (FMRP), an RNA-binding protein that regulates local protein translation, has been shown to be enriched in NL dendrites, suggesting its potential role in the structural dynamics of these dendrites. To explore the molecular role of FMRP in this nucleus, we performed proteomic analysis of NL, using micro laser capture and liquid chromatography tandem mass spectrometry. We identified 657 proteins, greatly represented in pathways involved in mitochondria, translation and metabolism, consistent with high levels of activity of NL neurons. Of these, 94 are potential FMRP targets, by comparative analysis with previously proposed FMRP targets in mammals. These proteins are enriched in pathways involved in cellular growth, cellular trafficking and transmembrane transport. Immunocytochemistry verified the dendritic localization of several proteins in NL. Furthermore, we confirmed the direct interaction of FMRP with one candidate, RhoC, by in vitro RNA binding assays. In summary, we provide a database of highly expressed proteins in NL and in particular a list of potential FMRP targets, with the goal of facilitating molecular characterization of FMRP signaling in future studies.
Collapse
Affiliation(s)
- Hitomi Sakano
- Virginia Merrill Bloedel Hearing Research Center, Department of Otolaryngology-Head and Neck Surgery, University of Washington, School of Medicine, Seattle, Washington
| | - Diego A R Zorio
- Department of Biomedical Sciences, Florida State University, Tallahassee, Florida
| | - Xiaoyu Wang
- Department of Biomedical Sciences, Florida State University, Tallahassee, Florida
| | - Ying S Ting
- Department of Genome Sciences, University of Washington, Seattle, Washington
| | - William S Noble
- Department of Genome Sciences, University of Washington, Seattle, Washington
| | - Michael J MacCoss
- Department of Genome Sciences, University of Washington, Seattle, Washington
| | - Edwin W Rubel
- Virginia Merrill Bloedel Hearing Research Center, Department of Otolaryngology-Head and Neck Surgery, University of Washington, School of Medicine, Seattle, Washington
| | - Yuan Wang
- Department of Biomedical Sciences, Florida State University, Tallahassee, Florida.,Program in Neuroscience, Florida State University, Tallahassee, Florida
| |
Collapse
|
19
|
Mottahedin A, Ardalan M, Chumak T, Riebe I, Ek J, Mallard C. Effect of Neuroinflammation on Synaptic Organization and Function in the Developing Brain: Implications for Neurodevelopmental and Neurodegenerative Disorders. Front Cell Neurosci 2017; 11:190. [PMID: 28744200 PMCID: PMC5504097 DOI: 10.3389/fncel.2017.00190] [Citation(s) in RCA: 83] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Accepted: 06/20/2017] [Indexed: 12/27/2022] Open
Abstract
The brain is a plastic organ where both the intrinsic CNS milieu and extrinsic cues play important roles in shaping and wiring neural connections. The perinatal period constitutes a critical time in central nervous system development with extensive refinement of neural connections, which are highly sensitive to fetal and neonatal compromise, such as inflammatory challenges. Emerging evidence suggests that inflammatory cells in the brain such as microglia and astrocytes are pivotal in regulating synaptic structure and function. In this article, we will review the role of glia cells in synaptic physiology and pathophysiology, including microglia-mediated elimination of synapses. We propose that activation of the immune system dynamically affects synaptic organization and function in the developing brain. We will discuss the role of neuroinflammation in altered synaptic plasticity following perinatal inflammatory challenges and potential implications for neurodevelopmental and neurodegenerative disorders.
Collapse
Affiliation(s)
- Amin Mottahedin
- Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of GothenburgGothenburg, Sweden
| | - Maryam Ardalan
- Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of GothenburgGothenburg, Sweden
| | - Tetyana Chumak
- Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of GothenburgGothenburg, Sweden
| | - Ilse Riebe
- Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of GothenburgGothenburg, Sweden
| | - Joakim Ek
- Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of GothenburgGothenburg, Sweden
| | - Carina Mallard
- Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of GothenburgGothenburg, Sweden
| |
Collapse
|
20
|
Budimirovic DB, Berry-Kravis E, Erickson CA, Hall SS, Hessl D, Reiss AL, King MK, Abbeduto L, Kaufmann WE. Updated report on tools to measure outcomes of clinical trials in fragile X syndrome. J Neurodev Disord 2017; 9:14. [PMID: 28616097 PMCID: PMC5467057 DOI: 10.1186/s11689-017-9193-x] [Citation(s) in RCA: 114] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Accepted: 02/22/2017] [Indexed: 12/27/2022] Open
Abstract
OBJECTIVE Fragile X syndrome (FXS) has been the neurodevelopmental disorder with the most active translation of preclinical breakthroughs into clinical trials. This process has led to a critical assessment of outcome measures, which resulted in a comprehensive review published in 2013. Nevertheless, the disappointing outcome of several recent phase III drug trials in FXS, and parallel efforts at evaluating behavioral endpoints for trials in autism spectrum disorder (ASD), has emphasized the need for re-assessing outcome measures and revising recommendations for FXS. METHODS After performing an extensive database search (PubMed, Food and Drug Administration (FDA)/National Institutes of Health (NIH)'s www.ClinicalTrials.gov, etc.) to determine progress since 2013, members of the Working Groups who published the 2013 Report evaluated the available outcome measures for FXS and related neurodevelopmental disorders using the COSMIN grading system of levels of evidence. The latter has also been applied to a British survey of endpoints for ASD. In addition, we also generated an informal classification of outcome measures for use in FXS intervention studies as instruments appropriate to detect shorter- or longer-term changes. RESULTS To date, a total of 22 double-blind controlled clinical trials in FXS have been identified through www.ClinicalTrials.gov and an extensive literature search. The vast majority of these FDA/NIH-registered clinical trials has been completed between 2008 and 2015 and has targeted the core excitatory/inhibitory imbalance present in FXS and other neurodevelopmental disorders. Limited data exist on reliability and validity for most tools used to measure cognitive, behavioral, and other problems in FXS in these trials and other studies. Overall, evidence for most tools supports a moderate tool quality grading. Data on sensitivity to treatment, currently under evaluation, could improve ratings for some cognitive and behavioral tools. Some progress has also been made at identifying promising biomarkers, mainly on blood-based and neurophysiological measures. CONCLUSION Despite the tangible progress in implementing clinical trials in FXS, the increasing data on measurement properties of endpoints, and the ongoing process of new tool development, the vast majority of outcome measures are at the moderate quality level with limited information on reliability, validity, and sensitivity to treatment. This situation is not unique to FXS, since reviews of endpoints for ASD have arrived at similar conclusions. These findings, in conjunction with the predominance of parent-based measures particularly in the behavioral domain, indicate that endpoint development in FXS needs to continue with an emphasis on more objective measures (observational, direct testing, biomarkers) that reflect meaningful improvements in quality of life. A major continuous challenge is the development of measurement tools concurrently with testing drug safety and efficacy in clinical trials.
Collapse
Affiliation(s)
- Dejan B. Budimirovic
- Departments of Psychiatry and Behavioral Sciences, Kennedy Krieger Institute and Child Psychiatry, Johns Hopkins University School of Medicine, 716 N. Broadway, Baltimore, MD 21205 USA
| | - Elizabeth Berry-Kravis
- Departments of Pediatrics, Neurological Sciences, Biochemistry, Rush University Medical Center, 1725 West Harrison, Suite 718, Chicago, IL 60612 USA
| | - Craig A. Erickson
- Division of Child and Adolescent Psychiatry, Cincinnati Children’s Hospital Medical Center and the University of Cincinnati College of Medicine, 3333 Burnet Avenue MLC 4002, Cincinnati, OH 45229 USA
| | - Scott S. Hall
- Division of Interdisciplinary Brain Sciences, Department of Psychiatry and Behavioral Sciences, Stanford University, 401 Quarry Road, Stanford, CA 94305 USA
| | - David Hessl
- MIND Institute and Department of Psychiatry and Behavioral Sciences, University of California Davis Medical Center, 2825 50th Street, Sacramento, CA 95817 USA
| | - Allan L. Reiss
- Division of Interdisciplinary Brain Sciences, Departments of Psychiatry and Behavioral Sciences, Radiology and Pediatrics, Stanford University, 401 Quarry Road, Stanford, CA 94305 USA
| | - Margaret K. King
- Autism & Developmental Medicine Institute, Geisinger Health System, Present address: Novartis Pharmaceuticals Corporation, US Medical, One Health Plaza, East Hanover, NJ 07936 USA
| | - Leonard Abbeduto
- MIND Institute and Department of Psychiatry and Behavioral Sciences, University of California Davis Medical Center, 2825 50th Street, Sacramento, CA 95817 USA
| | - Walter E. Kaufmann
- Center for Translational Research, Greenwood Genetic Center, 113 Gregor Mendel Circle, Greenwood, SC 29646 USA
- Department of Neurology, Boston Children’s Hospital, Boston, MA 02115 USA
| |
Collapse
|
21
|
Martínez-Cerdeño V. Dendrite and spine modifications in autism and related neurodevelopmental disorders in patients and animal models. Dev Neurobiol 2017; 77:393-404. [PMID: 27390186 PMCID: PMC5219951 DOI: 10.1002/dneu.22417] [Citation(s) in RCA: 170] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Revised: 06/29/2016] [Accepted: 07/04/2016] [Indexed: 12/12/2022]
Abstract
Dendrites and spines are the main neuronal structures receiving input from other neurons and glial cells. Dendritic and spine number, size, and morphology are some of the crucial factors determining how signals coming from individual synapses are integrated. Much remains to be understood about the characteristics of neuronal dendrites and dendritic spines in autism and related disorders. Although there have been many studies conducted using autism mouse models, few have been carried out using postmortem human tissue from patients. Available animal models of autism include those generated through genetic modifications and those non-genetic models of the disease. Here, we review how dendrite and spine morphology and number is affected in autism and related neurodevelopmental diseases, both in human, and genetic and non-genetic animal models of autism. Overall, data obtained from human and animal models point to a generalized reduction in the size and number, as well as an alteration of the morphology of dendrites; and an increase in spine densities with immature morphology, indicating a general spine immaturity state in autism. Additional human studies on dendrite and spine number and morphology in postmortem tissue are needed to understand the properties of these structures in the cerebral cortex of patients with autism. © 2016 Wiley Periodicals, Inc. Develop Neurobiol 77: 419-437, 2017.
Collapse
Affiliation(s)
- Verónica Martínez-Cerdeño
- Department of Pathology and Laboratory Medicine, UC Davis, Sacramento, California
- Institute for Pediatric Regenerative Medicine and Shriners Hospitals for Children Northern California, North California, Sacramento, California
- MIND Institute, UC Davis School of Medicine, Sacramento, California
| |
Collapse
|
22
|
Franco LM, Okray Z, Linneweber GA, Hassan BA, Yaksi E. Reduced Lateral Inhibition Impairs Olfactory Computations and Behaviors in a Drosophila Model of Fragile X Syndrome. Curr Biol 2017; 27:1111-1123. [PMID: 28366741 PMCID: PMC5405172 DOI: 10.1016/j.cub.2017.02.065] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Revised: 02/02/2017] [Accepted: 02/28/2017] [Indexed: 01/02/2023]
Abstract
Fragile X syndrome (FXS) patients present neuronal alterations that lead to severe intellectual disability, but the underlying neuronal circuit mechanisms are poorly understood. An emerging hypothesis postulates that reduced GABAergic inhibition of excitatory neurons is a key component in the pathophysiology of FXS. Here, we directly test this idea in a FXS Drosophila model. We show that FXS flies exhibit strongly impaired olfactory behaviors. In line with this, olfactory representations are less odor specific due to broader response tuning of excitatory projection neurons. We find that impaired inhibitory interactions underlie reduced specificity in olfactory computations. Finally, we show that defective lateral inhibition across projection neurons is caused by weaker inhibition from GABAergic interneurons. We provide direct evidence that deficient inhibition impairs sensory computations and behavior in an in vivo model of FXS. Together with evidence of impaired inhibition in autism and Rett syndrome, these findings suggest a potentially general mechanism for intellectual disability. Lack of dFMRP leads to reduced olfactory attraction and aversion in fruit flies Odor selectivity of antennal lobe projection neurons is impaired in dfmr1− flies GABAergic lateral inhibition within the antennal lobe is weaker in dfmr1− flies Deficient lateral inhibition impairs sensory computations and animal behavior
Collapse
Affiliation(s)
- Luis M Franco
- Neuroelectronics Research Flanders (NERF), KU Leuven, Kapeldreef 75, 3001 Leuven, Belgium; VIB Center for the Biology of Disease, KU Leuven, Herestraat 49, 3000 Leuven, Belgium; Center for Human Genetics, KU Leuven, Herestraat 49, 3000 Leuven, Belgium
| | - Zeynep Okray
- VIB Center for the Biology of Disease, KU Leuven, Herestraat 49, 3000 Leuven, Belgium; Center for Human Genetics, KU Leuven, Herestraat 49, 3000 Leuven, Belgium
| | - Gerit A Linneweber
- VIB Center for the Biology of Disease, KU Leuven, Herestraat 49, 3000 Leuven, Belgium; Center for Human Genetics, KU Leuven, Herestraat 49, 3000 Leuven, Belgium
| | - Bassem A Hassan
- VIB Center for the Biology of Disease, KU Leuven, Herestraat 49, 3000 Leuven, Belgium; Center for Human Genetics, KU Leuven, Herestraat 49, 3000 Leuven, Belgium; Institut du Cerveau et de la Moelle Epinière (ICM) - Hôpital Pitié-Salpêtrière, UPMC, Sorbonne Universités, Inserm, CNRS, 47 Boulevard Hôpital, 75013 Paris, France.
| | - Emre Yaksi
- Neuroelectronics Research Flanders (NERF), KU Leuven, Kapeldreef 75, 3001 Leuven, Belgium; Kavli Institute for Systems Neuroscience and Centre for Neural Computation, NTNU, Olav Kyrres gate 9, 7030 Trondheim, Norway.
| |
Collapse
|
23
|
Moody L, Chen H, Pan YX. Early-Life Nutritional Programming of Cognition-The Fundamental Role of Epigenetic Mechanisms in Mediating the Relation between Early-Life Environment and Learning and Memory Process. Adv Nutr 2017; 8:337-350. [PMID: 28298276 PMCID: PMC5347110 DOI: 10.3945/an.116.014209] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The perinatal period is a window of heightened plasticity that lays the groundwork for future anatomic, physiologic, and behavioral outcomes. During this time, maternal diet plays a pivotal role in the maturation of vital organs and the establishment of neuronal connections. However, when perinatal nutrition is either lacking in specific micro- and macronutrients or overloaded with excess calories, the consequences can be devastating and long lasting. The brain is particularly sensitive to perinatal insults, with several neurologic and psychiatric disorders having been linked to a poor in utero environment. Diseases characterized by learning and memory impairments, such as autism, schizophrenia, and Alzheimer disease, are hypothesized to be attributed in part to environmental factors, and evidence suggests that the etiology of these conditions may date back to very early life. In this review, we discuss the role of the early-life diet in shaping cognitive outcomes in offspring. We explore the endocrine and immune mechanisms responsible for these phenotypes and discuss how these systemic factors converge to change the brain's epigenetic landscape and regulate learning and memory across the lifespan. Through understanding the maternal programming of cognition, critical steps may be taken toward preventing and treating diseases that compromise learning and memory.
Collapse
Affiliation(s)
| | - Hong Chen
- Division of Nutritional Sciences,,Department of Food Science and Human Nutrition, and
| | - Yuan-Xiang Pan
- Division of Nutritional Sciences, .,Department of Food Science and Human Nutrition, and.,Illinois Informatics Institute, University of Illinois at Urbana-Champaign, Urbana, IL
| |
Collapse
|
24
|
Zorio DAR, Jackson CM, Liu Y, Rubel EW, Wang Y. Cellular distribution of the fragile X mental retardation protein in the mouse brain. J Comp Neurol 2017; 525:818-849. [PMID: 27539535 PMCID: PMC5558202 DOI: 10.1002/cne.24100] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Revised: 08/10/2016] [Accepted: 08/11/2016] [Indexed: 11/07/2022]
Abstract
The fragile X mental retardation protein (FMRP) plays an important role in normal brain development. Absence of FMRP results in abnormal neuronal morphologies in a selected manner throughout the brain, leading to intellectual deficits and sensory dysfunction in the fragile X syndrome (FXS). Despite FMRP importance for proper brain function, its overall expression pattern in the mammalian brain at the resolution of individual neuronal cell groups is not known. In this study we used FMR1 knockout and isogenic wildtype mice to systematically map the distribution of FMRP expression in the entire mouse brain. Using immunocytochemistry and cellular quantification analyses, we identified a large number of prominent cell groups expressing high levels of FMRP at the subcortical levels, in particular sensory and motor neurons in the brainstem and thalamus. In contrast, many cell groups in the midbrain and hypothalamus exhibit low FMRP levels. More important, we describe differential patterns of FMRP distribution in both cortical and subcortical brain regions. Almost all major brain areas contain high and low levels of FMRP cell groups adjacent to each other or between layers of the same cortical areas. These differential patterns indicate that FMRP expression appears to be specific to individual neuronal cell groups instead of being associated with all neurons in distinct brain regions, as previously considered. Taken together, these findings support the notion of FMRP differential neuronal regulation and strongly implicate the contribution of fundamental sensory and motor processing at subcortical levels to FXS pathology. J. Comp. Neurol. 525:818-849, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Diego A. R. Zorio
- Department of Biomedical Sciences, College of Medicine, Florida State University, Tallahassee, FL 32306, USA
| | - Christine M. Jackson
- Department of Biomedical Sciences, College of Medicine, Florida State University, Tallahassee, FL 32306, USA
| | - Yong Liu
- Department of Biomedical Sciences, College of Medicine, Florida State University, Tallahassee, FL 32306, USA
| | - Edwin W Rubel
- Virginia Merrill Bloedel Hearing Research Center, Department of Otolaryngology-Head and Neck Surgery, University of Washington School of Medicine, Box 357923, Seattle, WA 98195, USA
| | - Yuan Wang
- Department of Biomedical Sciences, College of Medicine, Florida State University, Tallahassee, FL 32306, USA
| |
Collapse
|
25
|
Ripamonti S, Ambrozkiewicz MC, Guzzi F, Gravati M, Biella G, Bormuth I, Hammer M, Tuffy LP, Sigler A, Kawabe H, Nishimori K, Toselli M, Brose N, Parenti M, Rhee J. Transient oxytocin signaling primes the development and function of excitatory hippocampal neurons. eLife 2017; 6. [PMID: 28231043 PMCID: PMC5323041 DOI: 10.7554/elife.22466] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Accepted: 02/06/2017] [Indexed: 12/30/2022] Open
Abstract
Beyond its role in parturition and lactation, oxytocin influences higher brain processes that control social behavior of mammals, and perturbed oxytocin signaling has been linked to the pathogenesis of several psychiatric disorders. However, it is still largely unknown how oxytocin exactly regulates neuronal function. We show that early, transient oxytocin exposure in vitro inhibits the development of hippocampal glutamatergic neurons, leading to reduced dendrite complexity, synapse density, and excitatory transmission, while sparing GABAergic neurons. Conversely, genetic elimination of oxytocin receptors increases the expression of protein components of excitatory synapses and excitatory synaptic transmission in vitro. In vivo, oxytocin-receptor-deficient hippocampal pyramidal neurons develop more complex dendrites, which leads to increased spine number and reduced γ-oscillations. These results indicate that oxytocin controls the development of hippocampal excitatory neurons and contributes to the maintenance of a physiological excitation/inhibition balance, whose disruption can cause neurobehavioral disturbances. DOI:http://dx.doi.org/10.7554/eLife.22466.001
Collapse
Affiliation(s)
- Silvia Ripamonti
- Department of Molecular Neurobiology, Max Planck Institute of Experimental Medicine, Göttingen, Germany.,Department of Medicine and Surgery, University of Milan-Bicocca, Monza, Italy
| | - Mateusz C Ambrozkiewicz
- Department of Molecular Neurobiology, Max Planck Institute of Experimental Medicine, Göttingen, Germany.,Cortical Development, Institute of Cell Biology and Neurobiology, Charité-Universitätsmedizin, Berlin, Germany
| | - Francesca Guzzi
- Department of Medicine and Surgery, University of Milan-Bicocca, Monza, Italy.,NeuroMi - Milan Center for Neuroscience, Monza, Italy
| | - Marta Gravati
- Department of Biology and Biotechnology, University of Pavia, Pavia, Italy
| | - Gerardo Biella
- Department of Biology and Biotechnology, University of Pavia, Pavia, Italy
| | - Ingo Bormuth
- Cortical Development, Institute of Cell Biology and Neurobiology, Charité-Universitätsmedizin, Berlin, Germany
| | - Matthieu Hammer
- Department of Molecular Neurobiology, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Liam P Tuffy
- Department of Molecular Neurobiology, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Albrecht Sigler
- Department of Molecular Neurobiology, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Hiroshi Kawabe
- Department of Molecular Neurobiology, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Katsuhiko Nishimori
- Department of Molecular and Cell Biology, Graduate School of Agricultural Science, Tohoku University, Miyagi, Japan
| | - Mauro Toselli
- Department of Biology and Biotechnology, University of Pavia, Pavia, Italy
| | - Nils Brose
- Department of Molecular Neurobiology, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Marco Parenti
- Department of Medicine and Surgery, University of Milan-Bicocca, Monza, Italy.,NeuroMi - Milan Center for Neuroscience, Monza, Italy
| | - JeongSeop Rhee
- Department of Molecular Neurobiology, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| |
Collapse
|
26
|
Lin YC, Frei JA, Kilander MBC, Shen W, Blatt GJ. A Subset of Autism-Associated Genes Regulate the Structural Stability of Neurons. Front Cell Neurosci 2016; 10:263. [PMID: 27909399 PMCID: PMC5112273 DOI: 10.3389/fncel.2016.00263] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Accepted: 10/28/2016] [Indexed: 12/15/2022] Open
Abstract
Autism spectrum disorder (ASD) comprises a range of neurological conditions that affect individuals’ ability to communicate and interact with others. People with ASD often exhibit marked qualitative difficulties in social interaction, communication, and behavior. Alterations in neurite arborization and dendritic spine morphology, including size, shape, and number, are hallmarks of almost all neurological conditions, including ASD. As experimental evidence emerges in recent years, it becomes clear that although there is broad heterogeneity of identified autism risk genes, many of them converge into similar cellular pathways, including those regulating neurite outgrowth, synapse formation and spine stability, and synaptic plasticity. These mechanisms together regulate the structural stability of neurons and are vulnerable targets in ASD. In this review, we discuss the current understanding of those autism risk genes that affect the structural connectivity of neurons. We sub-categorize them into (1) cytoskeletal regulators, e.g., motors and small RhoGTPase regulators; (2) adhesion molecules, e.g., cadherins, NCAM, and neurexin superfamily; (3) cell surface receptors, e.g., glutamatergic receptors and receptor tyrosine kinases; (4) signaling molecules, e.g., protein kinases and phosphatases; and (5) synaptic proteins, e.g., vesicle and scaffolding proteins. Although the roles of some of these genes in maintaining neuronal structural stability are well studied, how mutations contribute to the autism phenotype is still largely unknown. Investigating whether and how the neuronal structure and function are affected when these genes are mutated will provide insights toward developing effective interventions aimed at improving the lives of people with autism and their families.
Collapse
Affiliation(s)
- Yu-Chih Lin
- Laboratory of Neuronal Connectivity, Program in Neuroscience, Hussman Institute for Autism, Baltimore MD, USA
| | - Jeannine A Frei
- Laboratory of Neuronal Connectivity, Program in Neuroscience, Hussman Institute for Autism, Baltimore MD, USA
| | - Michaela B C Kilander
- Laboratory of Neuronal Connectivity, Program in Neuroscience, Hussman Institute for Autism, Baltimore MD, USA
| | - Wenjuan Shen
- Laboratory of Neuronal Connectivity, Program in Neuroscience, Hussman Institute for Autism, Baltimore MD, USA
| | - Gene J Blatt
- Laboratory of Autism Neurocircuitry, Program in Neuroscience, Hussman Institute for Autism, Baltimore MD, USA
| |
Collapse
|
27
|
Somatosensory map expansion and altered processing of tactile inputs in a mouse model of fragile X syndrome. Neurobiol Dis 2016; 96:201-215. [PMID: 27616423 DOI: 10.1016/j.nbd.2016.09.007] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Revised: 08/30/2016] [Accepted: 09/06/2016] [Indexed: 11/20/2022] Open
Abstract
Fragile X syndrome (FXS) is a common inherited form of intellectual disability caused by the absence or reduction of the fragile X mental retardation protein (FMRP) encoded by the FMR1 gene. In humans, one symptom of FXS is hypersensitivity to sensory stimuli, including touch. We used a mouse model of FXS (Fmr1 KO) to study sensory processing of tactile information conveyed via the whisker system. In vivo electrophysiological recordings in somatosensory barrel cortex showed layer-specific broadening of the receptive fields at the level of layer 2/3 but not layer 4, in response to whisker stimulation. Furthermore, the encoding of tactile stimuli at different frequencies was severely affected in layer 2/3. The behavioral effect of this broadening of the receptive fields was tested in the gap-crossing task, a whisker-dependent behavioral paradigm. In this task the Fmr1 KO mice showed differences in the number of whisker contacts with platforms, decrease in the whisker sampling duration and reduction in the whisker touch-time while performing the task. We propose that the increased excitability in the somatosensory barrel cortex upon whisker stimulation may contribute to changes in the whisking strategy as well as to other observed behavioral phenotypes related to tactile processing in Fmr1 KO mice.
Collapse
|
28
|
Belagodu AP, Zendeli L, Slater BJ, Galvez R. Blocking elevated VEGF‐A attenuates non‐vasculature Fragile X syndrome abnormalities. Dev Neurobiol 2016; 77:14-25. [DOI: 10.1002/dneu.22404] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Revised: 03/15/2016] [Accepted: 05/31/2016] [Indexed: 01/19/2023]
Affiliation(s)
- Amogh P. Belagodu
- Neuroscience Program, University of Illinois at Urbana‐Champaign405 N Mathews AveUrbana Illinois61801
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana‐Champaign405 N Mathews AveUrbana Illinois61801
| | - Liridon Zendeli
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana‐Champaign405 N Mathews AveUrbana Illinois61801
| | - Bernard J. Slater
- Neuroscience Program, University of Illinois at Urbana‐Champaign405 N Mathews AveUrbana Illinois61801
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana‐Champaign405 N Mathews AveUrbana Illinois61801
| | - Roberto Galvez
- Neuroscience Program, University of Illinois at Urbana‐Champaign405 N Mathews AveUrbana Illinois61801
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana‐Champaign405 N Mathews AveUrbana Illinois61801
- Psychology DepartmentUniversity of Illinois at Urbana‐Champaign405 N Mathews AveUrbana Illinois61801
| |
Collapse
|
29
|
Zhang L, Liang Z, Zhu P, Li M, Yi YH, Liao WP, Su T. Altered intrinsic properties and bursting activities of neurons in layer IV of somatosensory cortex from Fmr-1 knockout mice. Exp Neurol 2016; 280:60-9. [PMID: 27048919 DOI: 10.1016/j.expneurol.2016.03.025] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Revised: 03/04/2016] [Accepted: 03/29/2016] [Indexed: 10/22/2022]
Abstract
Neuroadaptations and alterations in neuronal excitability are critical in brain maturation and many neurological diseases. Fragile X syndrome (FXS) is a pervasive neurodevelopmental disorder characterized by extensive synaptic and circuit dysfunction. It is still unclear about the alterations in intrinsic excitability of individual neurons and their link to hyperexcitable circuitry. In this study, whole cell patch-clamp recordings were employed to characterize the membrane and firing properties of layer IV cells in slices of the somatosensory cortex of Fmr-1 knockout (KO) mice. These cells generally exhibited a regular spiking (RS) pattern, while there were significant increases in the number of cells that adopted intrinsic bursting (IB) compared with age-matched wild type (WT) cells. The cells subgrouped according to their firing patterns and maturation differed significantly in membrane and discharge properties between KO and WT. The changes in the intrinsic properties were consistent with highly facilitated discharges in KO cells induced by current injection. Spontaneous activities of RS neurons driven by local network were also increased in the KO cells, especially in neonate groups. Under an epileptiform condition mimicked by omission of Mg(2+) in extracellular solution, these RS neurons from KO mice were more likely to switch to burst discharges. Analysis on bursts revealed that the KO cells tended to form burst discharges and even severe events manifested as seizure-like ictal discharges. These results suggest that alterations in intrinsic properties in individual neurons are involved in the abnormal excitability of cortical circuitry and possibly account for the pathogenesis of epilepsy in FXS.
Collapse
Affiliation(s)
- Linming Zhang
- Institute of Neuroscience and the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China; Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou, China; Department of Neurology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Zhanrong Liang
- Institute of Neuroscience and the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China; Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou, China
| | - Pingping Zhu
- Institute of Neuroscience and the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China; Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou, China
| | - Meng Li
- Institute of Neuroscience and the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China; Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou, China
| | - Yong-Hong Yi
- Institute of Neuroscience and the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China; Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou, China
| | - Wei-Ping Liao
- Institute of Neuroscience and the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China; Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou, China
| | - Tao Su
- Institute of Neuroscience and the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China; Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou, China.
| |
Collapse
|
30
|
Ruby K, Falvey K, Kulesza R. Abnormal neuronal morphology and neurochemistry in the auditory brainstem of Fmr1 knockout rats. Neuroscience 2015; 303:285-98. [DOI: 10.1016/j.neuroscience.2015.06.061] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2015] [Revised: 06/10/2015] [Accepted: 06/27/2015] [Indexed: 01/19/2023]
|
31
|
Wang GX, Smith SJ, Mourrain P. Fmr1 KO and fenobam treatment differentially impact distinct synapse populations of mouse neocortex. Neuron 2015; 84:1273-86. [PMID: 25521380 DOI: 10.1016/j.neuron.2014.11.016] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/18/2014] [Indexed: 02/07/2023]
Abstract
Cognitive deficits in fragile X syndrome (FXS) are attributed to molecular abnormalities of the brain's vast and heterogeneous synapse populations. Unfortunately, the density of synapses coupled with their molecular heterogeneity presents formidable challenges in understanding the specific contribution of synapse changes in FXS. We demonstrate powerful new methods for the large-scale molecular analysis of individual synapses that allow quantification of numerous specific changes in synapse populations present in the cortex of a mouse model of FXS. Analysis of nearly a million individual synapses reveals distinct, quantitative changes in synaptic proteins distributed across over 6,000 pairwise metrics. Some, but not all, of these synaptic alterations are reversed by treatment with the candidate therapeutic fenobam, an mGluR5 antagonist. These patterns of widespread, but diverse synaptic protein changes in response to global perturbation suggest that FXS and its treatment must be understood as a networked system at the synapse level.
Collapse
Affiliation(s)
- Gordon X Wang
- Center for Sleep Sciences and Medicine, Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA; Department of Molecular and Cellular Physiology, Stanford University, Stanford, CA 94305, USA.
| | - Stephen J Smith
- Department of Molecular and Cellular Physiology, Stanford University, Stanford, CA 94305, USA; Allen Institute for Brain Science, Seattle, WA 98103, USA
| | - Philippe Mourrain
- Center for Sleep Sciences and Medicine, Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA; INSERM 1024, Ecole Normale Supérieure Paris, 75005, France
| |
Collapse
|
32
|
Rotschafer SE, Marshak S, Cramer KS. Deletion of Fmr1 alters function and synaptic inputs in the auditory brainstem. PLoS One 2015; 10:e0117266. [PMID: 25679778 PMCID: PMC4332492 DOI: 10.1371/journal.pone.0117266] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2014] [Accepted: 12/21/2014] [Indexed: 01/27/2023] Open
Abstract
Fragile X Syndrome (FXS), a neurodevelopmental disorder, is the most prevalent single-gene cause of autism spectrum disorder. Autism has been associated with impaired auditory processing, abnormalities in the auditory brainstem response (ABR), and reduced cell number and size in the auditory brainstem nuclei. FXS is characterized by elevated cortical responses to sound stimuli, with some evidence for aberrant ABRs. Here, we assessed ABRs and auditory brainstem anatomy in Fmr1-/- mice, an animal model of FXS. We found that Fmr1-/- mice showed elevated response thresholds to both click and tone stimuli. Amplitudes of ABR responses were reduced in Fmr1-/- mice for early peaks of the ABR. The growth of the peak I response with sound intensity was less steep in mutants that in wild type mice. In contrast, amplitudes and response growth in peaks IV and V did not differ between these groups. We did not observe differences in peak latencies or in interpeak latencies. Cell size was reduced in Fmr1-/- mice in the ventral cochlear nucleus (VCN) and in the medial nucleus of the trapezoid body (MNTB). We quantified levels of inhibitory and excitatory synaptic inputs in these nuclei using markers for presynaptic proteins. We measured VGAT and VGLUT immunolabeling in VCN, MNTB, and the lateral superior olive (LSO). VGAT expression in MNTB was significantly greater in the Fmr1-/- mouse than in wild type mice. Together, these observations demonstrate that FXS affects peripheral and central aspects of hearing and alters the balance of excitation and inhibition in the auditory brainstem.
Collapse
Affiliation(s)
- Sarah E. Rotschafer
- Department of Neurobiology and Behavior, University of California Irvine, Irvine, California, 92697, United States of America
| | - Sonya Marshak
- Department of Neurobiology and Behavior, University of California Irvine, Irvine, California, 92697, United States of America
| | - Karina S. Cramer
- Department of Neurobiology and Behavior, University of California Irvine, Irvine, California, 92697, United States of America
- * E-mail:
| |
Collapse
|
33
|
Wang Y, Sakano H, Beebe K, Brown MR, de Laat R, Bothwell M, Kulesza RJ, Rubel EW. Intense and specialized dendritic localization of the fragile X mental retardation protein in binaural brainstem neurons: a comparative study in the alligator, chicken, gerbil, and human. J Comp Neurol 2015; 522:2107-28. [PMID: 24318628 DOI: 10.1002/cne.23520] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2013] [Revised: 12/04/2013] [Accepted: 12/05/2013] [Indexed: 11/09/2022]
Abstract
Neuronal dendrites are structurally and functionally dynamic in response to changes in afferent activity. The fragile X mental retardation protein (FMRP) is an mRNA binding protein that regulates activity-dependent protein synthesis and morphological dynamics of dendrites. Loss and abnormal expression of FMRP occur in fragile X syndrome (FXS) and some forms of autism spectrum disorders. To provide further understanding of how FMRP signaling regulates dendritic dynamics, we examined dendritic expression and localization of FMRP in the reptilian and avian nucleus laminaris (NL) and its mammalian analogue, the medial superior olive (MSO), in rodents and humans. NL/MSO neurons are specialized for temporal processing of low-frequency sounds for binaural hearing, which is impaired in FXS. Protein BLAST analyses first demonstrate that the FMRP amino acid sequences in the alligator and chicken are highly similar to human FMRP with identical mRNA-binding and phosphorylation sites, suggesting that FMRP functions similarly across vertebrates. Immunocytochemistry further reveals that NL/MSO neurons have very high levels of dendritic FMRP in low-frequency hearing vertebrates including alligator, chicken, gerbil, and human. Remarkably, dendritic FMRP in NL/MSO neurons often accumulates at branch points and enlarged distal tips, loci known to be critical for branch-specific dendritic arbor dynamics. These observations support an important role for FMRP in regulating dendritic properties of binaural neurons that are essential for low-frequency sound localization and auditory scene segregation, and support the relevance of studying this regulation in nonhuman vertebrates that use low frequencies in order to further understand human auditory processing disorders.
Collapse
Affiliation(s)
- Yuan Wang
- Virginia Merrill Bloedel Hearing Research Center, Department of Otolaryngology-Head and Neck Surgery, University of Washington School of Medicine, Seattle, WA, 98195-7923
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Abstract
Fragile X Syndrome (FXS) is commonly thought to arise from dysfunction of the synapse, the site of communication between neurons. However, loss of the protein that results in FXS occurs early in embryonic development, while synapses are formed relatively late. Fragile X Syndrome (FXS) is the leading known monogenic form of autism and the most common form of inherited intellectual disability. FXS results from silencing the FMR1 gene during embryonic development, leading to loss of Fragile X Mental Retardation Protein (FMRP), an RNA-binding protein that regulates mRNA transport, stability, and translation. FXS is commonly thought of as a disease of synaptic dysfunction; however, FMRP expression is lost early in embryonic development, well before most synaptogenesis occurs. Recent studies suggest that loss of FMRP results in aberrant neurogenesis, but neurogenic defects have been variable. We investigated whether FMRP affects neurogenesis in Xenopus laevis tadpoles that express a homolog of FMR1. We used in vivo time-lapse imaging of neural progenitor cells and their neuronal progeny to evaluate the effect of acute loss or overexpression of FMRP on neurogenesis in the developing optic tectum. We complimented the time-lapse studies with SYTOX labeling to quantify apoptosis and CldU labeling to measure cell proliferation. Animals with increased or decreased levels of FMRP have significantly decreased neuronal proliferation and survival. They also have increased neuronal differentiation, but deficient dendritic arbor elaboration. The presence and severity of these defects was highly sensitive to FMRP levels. These data demonstrate that FMRP plays an important role in neurogenesis and suggest that endogenous FMRP levels are carefully regulated. These studies show promise in using Xenopus as an experimental system to study fundamental deficits in brain development with loss of FMRP and give new insight into the pathophysiology of FXS.
Collapse
|
35
|
Liu J, Liu B, Zhang X, Yu B, Guan W, Wang K, Yang Y, Gong Y, Wu X, Yanagawa Y, Wu S, Zhao C. Calretinin-positive L5a pyramidal neurons in the development of the paralemniscal pathway in the barrel cortex. Mol Brain 2014; 7:84. [PMID: 25404384 PMCID: PMC4246560 DOI: 10.1186/s13041-014-0084-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2014] [Accepted: 11/04/2014] [Indexed: 01/06/2023] Open
Abstract
Background The rodent barrel cortex has been established as an ideal model for studying the development and plasticity of a neuronal circuit. The barrel cortex consists of barrel and septa columns, which receive various input signals through distinct pathways. The lemniscal pathway transmits whisker-specific signals to homologous barrel columns, and the paralemniscal pathway transmits multi-whisker signals to both barrel and septa columns. The integration of information from both lemniscal and paralemniscal pathways in the barrel cortex is critical for precise object recognition. As the main target of the posterior medial nucleus (POm) in the paralemniscal pathway, layer 5a (L5a) pyramidal neurons are involved in both barrel and septa circuits and are considered an important site of information integration. However, information on L5a neurons is very limited. This study aims to explore the cellular features of L5a neurons and to provide a morphological basis for studying their roles in the development of the paralemniscal pathway and in information integration. Results 1. We found that the calcium-binding protein calretinin (CR) is dynamically expressed in L5a excitatory pyramidal neurons of the barrel cortex, and L5a neurons form a unique serrated pattern similar to the distributions of their presynaptic POm axon terminals. 2. Infraorbital nerve transection disrupts this unique alignment, indicating that it is input dependent. 3. The formation of the L5a neuronal alignment develops synchronously with barrels, which suggests that the lemniscal and paralemniscal pathways may interact with each other to regulate pattern formation and refinement in the barrel cortex. 4. CR is specifically expressed in the paralemniscal pathway, and CR deletion disrupts the unique L5a neuronal pattern, which indicates that CR may be required for the development of the paralemniscal pathway. Conclusions Our results demonstrate that L5a neurons form a unique, input-dependent serrated alignment during the development of cortical barrels and that CR may play an important role in the development of the paralemniscal pathway. Our data provide a morphological basis for studying the role of L5a pyramidal neurons in information integration within the lemniscal and paralemniscal pathways.
Collapse
Affiliation(s)
- Junhua Liu
- Key Laboratory of Developmental Genes and Human Diseases, MOE, Department of Anatomy and Neuroscience, Medical School, Southeast University, Nanjing, 210009, PR China.
| | - Bin Liu
- Key Laboratory of Developmental Genes and Human Diseases, MOE, Department of Anatomy and Neuroscience, Medical School, Southeast University, Nanjing, 210009, PR China.
| | - XiaoYun Zhang
- Key Laboratory of Developmental Genes and Human Diseases, MOE, Department of Anatomy and Neuroscience, Medical School, Southeast University, Nanjing, 210009, PR China.
| | - Baocong Yu
- Key Laboratory of Developmental Genes and Human Diseases, MOE, Department of Anatomy and Neuroscience, Medical School, Southeast University, Nanjing, 210009, PR China.
| | - Wuqiang Guan
- Institute of Neurobiology, Institutes of Brain Science and State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, 200032, PR China.
| | - Kun Wang
- Key Laboratory of Developmental Genes and Human Diseases, MOE, Department of Anatomy and Neuroscience, Medical School, Southeast University, Nanjing, 210009, PR China.
| | - Yang Yang
- Key Laboratory of Developmental Genes and Human Diseases, MOE, Department of Anatomy and Neuroscience, Medical School, Southeast University, Nanjing, 210009, PR China.
| | - Yifan Gong
- Key Laboratory of Developmental Genes and Human Diseases, MOE, Department of Anatomy and Neuroscience, Medical School, Southeast University, Nanjing, 210009, PR China.
| | - Xiaojing Wu
- Key Laboratory of Developmental Genes and Human Diseases, MOE, Department of Anatomy and Neuroscience, Medical School, Southeast University, Nanjing, 210009, PR China.
| | - Yuchio Yanagawa
- Department of Genetic and Behavioral Neuroscience, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, 371-8511, Japan.
| | - Shengxi Wu
- Department of Anatomy, Histology and Embryology, K.K. Leung Brain Research Centre, School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, PR China.
| | - Chunjie Zhao
- Key Laboratory of Developmental Genes and Human Diseases, MOE, Department of Anatomy and Neuroscience, Medical School, Southeast University, Nanjing, 210009, PR China. .,Center of Depression, Beijing Institute for Brain Disorders, Beijing, 100069, PR China.
| |
Collapse
|
36
|
Kazdoba TM, Leach PT, Silverman JL, Crawley JN. Modeling fragile X syndrome in the Fmr1 knockout mouse. Intractable Rare Dis Res 2014; 3:118-33. [PMID: 25606362 PMCID: PMC4298642 DOI: 10.5582/irdr.2014.01024] [Citation(s) in RCA: 174] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Accepted: 11/28/2014] [Indexed: 11/05/2022] Open
Abstract
Fragile X Syndrome (FXS) is a commonly inherited form of intellectual disability and one of the leading genetic causes for autism spectrum disorder. Clinical symptoms of FXS can include impaired cognition, anxiety, hyperactivity, social phobia, and repetitive behaviors. FXS is caused by a CGG repeat mutation which expands a region on the X chromosome containing the FMR1 gene. In FXS, a full mutation (> 200 repeats) leads to hypermethylation of FMR1, an epigenetic mechanism that effectively silences FMR1 gene expression and reduces levels of the FMR1 gene product, fragile X mental retardation protein (FMRP). FMRP is an RNA-binding protein that is important for the regulation of protein expression. In an effort to further understand how loss of FMR1 and FMRP contribute to FXS symptomology, several FXS animal models have been created. The most well characterized rodent model is the Fmr1 knockout (KO) mouse, which lacks FMRP protein due to a disruption in its Fmr1 gene. Here, we review the behavioral phenotyping of the Fmr1 KO mouse to date, and discuss the clinical relevance of this mouse model to the human FXS condition. While much remains to be learned about FXS, the Fmr1 KO mouse is a valuable tool for understanding the repercussions of functional loss of FMRP and assessing the efficacy of pharmacological compounds in ameliorating the molecular and behavioral phenotypes relevant to FXS.
Collapse
Affiliation(s)
- Tatiana M. Kazdoba
- MIND Institute, Department of Psychiatry and Behavioral Sciences, University of California, Davis, School of Medicine, Sacramento, CA, USA
- Address correspondence to: Dr. Tatiana M. Kazdoba, MIND Institute, Department of Psychiatry and Behavioral Sciences, University of California, Davis, School of Medicine, Sacramento, Research II Building 96, 4625 2nd Avenue, Sacramento, CA 95817, USA. E-mail:
| | - Prescott T. Leach
- MIND Institute, Department of Psychiatry and Behavioral Sciences, University of California, Davis, School of Medicine, Sacramento, CA, USA
| | - Jill L. Silverman
- MIND Institute, Department of Psychiatry and Behavioral Sciences, University of California, Davis, School of Medicine, Sacramento, CA, USA
| | - Jacqueline N. Crawley
- MIND Institute, Department of Psychiatry and Behavioral Sciences, University of California, Davis, School of Medicine, Sacramento, CA, USA
| |
Collapse
|
37
|
Beebe K, Wang Y, Kulesza R. Distribution of fragile X mental retardation protein in the human auditory brainstem. Neuroscience 2014; 273:79-91. [PMID: 24838064 DOI: 10.1016/j.neuroscience.2014.05.006] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2014] [Revised: 04/29/2014] [Accepted: 05/02/2014] [Indexed: 01/20/2023]
Abstract
Fragile X mental retardation protein (FMRP) binds select mRNAs, functions in intracellular transport of these mRNAs and represses their translation. FMRP is highly expressed in neurons and lack of FMRP has been shown to result in dendritic dysmorphology and altered synaptic function. FMRP is known to interact with mRNAs for the Kv3.1b potassium channel which is required for neurons to fire action potentials at high rates with remarkable temporal precision. Auditory brainstem neurons are known for remarkably high spike rates and expression of Kv3.1b potassium channels. Fragile X syndrome (FXS) is a genetic disorder caused by a mutation in the fragile X mental retardation 1 gene (Fmr1) resulting in decreased expression of FMRP and subsequent intellectual disability, seizures, attention deficit and hypersensitivity to auditory and other sensory stimuli. We therefore hypothesize that the auditory difficulties in FXS result, at least in part, from dysfunction of auditory brainstem neurons. To examine this hypothesis, we have studied normal human brainstem tissue with immunohistochemical techniques and confocal microscopy. Our results demonstrate that FMRP is widely expressed in cell bodies and dendritic arbors of neurons in the human cochlear nucleus and superior olivary complex and also that coincidence detector neurons of the medial superior olive colocalization of FMRP and Kv3.1b. We interpret these observations to suggest that the lower auditory brainstem is a potential site of dysfunction in FXS.
Collapse
Affiliation(s)
- K Beebe
- Lake Erie College of Osteopathic Medicine, Auditory Research Center, Erie, PA, USA
| | - Y Wang
- Virginia Merrill Bloedel Hearing Research Center, Department of Otolaryngology-Head and Neck Surgery, University of Washington School of Medicine, Seattle, WA, USA
| | - R Kulesza
- Lake Erie College of Osteopathic Medicine, Auditory Research Center, Erie, PA, USA.
| |
Collapse
|
38
|
Chau LS, Prakapenka AV, Zendeli L, Davis AS, Galvez R. Training-dependent associative learning induced neocortical structural plasticity: a trace eyeblink conditioning analysis. PLoS One 2014; 9:e95317. [PMID: 24760074 PMCID: PMC3997347 DOI: 10.1371/journal.pone.0095317] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2013] [Accepted: 03/26/2014] [Indexed: 11/18/2022] Open
Abstract
Studies utilizing general learning and memory tasks have suggested the importance of neocortical structural plasticity for memory consolidation. However, these learning tasks typically result in learning of multiple different tasks over several days of training, making it difficult to determine the synaptic time course mediating each learning event. The current study used trace-eyeblink conditioning to determine the time course for neocortical spine modification during learning. With eyeblink conditioning, subjects are presented with a neutral, conditioned stimulus (CS) paired with a salient, unconditioned stimulus (US) to elicit an unconditioned response (UR). With multiple CS-US pairings, subjects learn to associate the CS with the US and exhibit a conditioned response (CR) when presented with the CS. Trace conditioning is when there is a stimulus free interval between the CS and the US. Utilizing trace-eyeblink conditioning with whisker stimulation as the CS (whisker-trace-eyeblink: WTEB), previous findings have shown that primary somatosensory (barrel) cortex is required for both acquisition and retention of the trace-association. Additionally, prior findings demonstrated that WTEB acquisition results in an expansion of the cytochrome oxidase whisker representation and synaptic modification in layer IV of barrel cortex. To further explore these findings and determine the time course for neocortical learning-induced spine modification, the present study utilized WTEB conditioning to examine Golgi-Cox stained neurons in layer IV of barrel cortex. Findings from this study demonstrated a training-dependent spine proliferation in layer IV of barrel cortex during trace associative learning. Furthermore, findings from this study showing that filopodia-like spines exhibited a similar pattern to the overall spine density further suggests that reorganization of synaptic contacts set the foundation for learning-induced neocortical modifications through the different neocortical layers.
Collapse
Affiliation(s)
- Lily S. Chau
- Psychology Department, University of Illinois at Urbana-Champaign, Champaign, Illinois, United States of America
- * E-mail:
| | - Alesia V. Prakapenka
- Psychology Department, University of Illinois at Urbana-Champaign, Champaign, Illinois, United States of America
| | - Liridon Zendeli
- Psychology Department, University of Illinois at Urbana-Champaign, Champaign, Illinois, United States of America
| | - Ashley S. Davis
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Champaign, Illinois, United States of America
| | - Roberto Galvez
- Psychology Department, University of Illinois at Urbana-Champaign, Champaign, Illinois, United States of America
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Champaign, Illinois, United States of America
- Neuroscience Program, University of Illinois at Urbana-Champaign, Champaign, Illinois, United States of America
| |
Collapse
|
39
|
van der Molen MJW, Stam CJ, van der Molen MW. Resting-state EEG oscillatory dynamics in fragile X syndrome: abnormal functional connectivity and brain network organization. PLoS One 2014; 9:e88451. [PMID: 24523898 PMCID: PMC3921158 DOI: 10.1371/journal.pone.0088451] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2013] [Accepted: 01/13/2014] [Indexed: 12/11/2022] Open
Abstract
Disruptions in functional connectivity and dysfunctional brain networks are considered to be a neurological hallmark of neurodevelopmental disorders. Despite the vast literature on functional brain connectivity in typical brain development, surprisingly few attempts have been made to characterize brain network integrity in neurodevelopmental disorders. Here we used resting-state EEG to characterize functional brain connectivity and brain network organization in eight males with fragile X syndrome (FXS) and 12 healthy male controls. Functional connectivity was calculated based on the phase lag index (PLI), a non-linear synchronization index that is less sensitive to the effects of volume conduction. Brain network organization was assessed with graph theoretical analysis. A decrease in global functional connectivity was observed in FXS males for upper alpha and beta frequency bands. For theta oscillations, we found increased connectivity in long-range (fronto-posterior) and short-range (frontal-frontal and posterior-posterior) clusters. Graph theoretical analysis yielded evidence of increased path length in the theta band, suggesting that information transfer between brain regions is particularly impaired for theta oscillations in FXS. These findings are discussed in terms of aberrant maturation of neuronal oscillatory dynamics, resulting in an imbalance in excitatory and inhibitory neuronal circuit activity.
Collapse
Affiliation(s)
- Melle J. W. van der Molen
- Institute of Psychology, Developmental Psychology Unit, Leiden University, Leiden, the Netherlands
- Leiden Institute for Brain and Cognition. Leiden, the Netherlands
- * E-mail:
| | - Cornelis J. Stam
- Department of Clinical Neurophysiology, VU University Medical Center, Amsterdam, the Netherlands
- Neuroscience Campus Amsterdam, VU University Medical Center, Amsterdam, The Netherlands
| | - Maurits W. van der Molen
- Department of Developmental Psychology, University of Amsterdam, Amsterdam, the Netherlands
- Cognitive Science Center Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
40
|
Rotschafer SE, Razak KA. Auditory processing in fragile x syndrome. Front Cell Neurosci 2014; 8:19. [PMID: 24550778 PMCID: PMC3912505 DOI: 10.3389/fncel.2014.00019] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2013] [Accepted: 01/12/2014] [Indexed: 11/24/2022] Open
Abstract
Fragile X syndrome (FXS) is an inherited form of intellectual disability and autism. Among other symptoms, FXS patients demonstrate abnormalities in sensory processing and communication. Clinical, behavioral, and electrophysiological studies consistently show auditory hypersensitivity in humans with FXS. Consistent with observations in humans, the Fmr1 KO mouse model of FXS also shows evidence of altered auditory processing and communication deficiencies. A well-known and commonly used phenotype in pre-clinical studies of FXS is audiogenic seizures. In addition, increased acoustic startle response is seen in the Fmr1 KO mice. In vivo electrophysiological recordings indicate hyper-excitable responses, broader frequency tuning, and abnormal spectrotemporal processing in primary auditory cortex of Fmr1 KO mice. Thus, auditory hyper-excitability is a robust, reliable, and translatable biomarker in Fmr1 KO mice. Abnormal auditory evoked responses have been used as outcome measures to test therapeutics in FXS patients. Given that similarly abnormal responses are present in Fmr1 KO mice suggests that cellular mechanisms can be addressed. Sensory cortical deficits are relatively more tractable from a mechanistic perspective than more complex social behaviors that are typically studied in autism and FXS. The focus of this review is to bring together clinical, functional, and structural studies in humans with electrophysiological and behavioral studies in mice to make the case that auditory hypersensitivity provides a unique opportunity to integrate molecular, cellular, circuit level studies with behavioral outcomes in the search for therapeutics for FXS and other autism spectrum disorders.
Collapse
Affiliation(s)
- Sarah E Rotschafer
- Graduate Neuroscience Program, Department of Psychology, University of California, Riverside, CA USA
| | - Khaleel A Razak
- Graduate Neuroscience Program, Department of Psychology, University of California, Riverside, CA USA
| |
Collapse
|
41
|
Chau LS, Akhtar O, Mohan V, Kondilis A, Galvez R. Rapid adult experience-dependent anatomical plasticity in layer IV of primary somatosensory cortex. Brain Res 2013; 1543:93-100. [PMID: 24183785 DOI: 10.1016/j.brainres.2013.10.043] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2013] [Revised: 10/21/2013] [Accepted: 10/22/2013] [Indexed: 10/26/2022]
Abstract
Sensory deprivation, such as whisker deprivation, is one of the most common paradigms used to examine experience-dependent plasticity. Many of these studies conducted during development have demonstrated anatomical and synaptic neocortical plasticity with varying lengths of deprivation (for review, see Holtmaat and Svoboda, 2009). However, to date, there have been few studies exploring brief periods of experience-dependent neocortical plasticity in adulthood, similar to that observed from learning and memory paradigms (Siucinska and Kossut, 1996, 2004; Galvez et al., 2006; Chau et al., 2013). Examining both synapsin I and Golgi-Cox stained neurons in primary somatosensory cortex of unilaterally whisker-deprived adult mice, the current study demonstrates that 5 days of whisker deprivation results in more synapses in spared barrels and reduced synapses in deprived barrels. To our knowledge, this is the first study to characterize anatomical changes in layer IV of primary somatosensory cortex after a brief period of sensory deprivation in adulthood. Furthermore, findings from the present study suggest that analyses from prolonged periods of either sensory deprivation or stimulation during adulthood are missing forms of plasticity that could provide better insight into various cognitive processes, such as learning and memory.
Collapse
Affiliation(s)
- Lily S Chau
- Psychology Department University of Illinois at Urbana-Champaign, USA.
| | - Omar Akhtar
- Psychology Department University of Illinois at Urbana-Champaign, USA
| | - Vijay Mohan
- Psychology Department University of Illinois at Urbana-Champaign, USA
| | | | - Roberto Galvez
- Psychology Department University of Illinois at Urbana-Champaign, USA; Beckman Institute for Advanced Science and Technology University of Illinois at Urbana-Champaign, USA; Neuroscience Program University of Illinois at Urbana-Champaign, USA
| |
Collapse
|
42
|
Cook D, Nuro E, Murai KK. Increasing our understanding of human cognition through the study of Fragile X Syndrome. Dev Neurobiol 2013; 74:147-77. [PMID: 23723176 PMCID: PMC4216185 DOI: 10.1002/dneu.22096] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2013] [Accepted: 05/17/2013] [Indexed: 12/16/2022]
Abstract
Fragile X Syndrome (FXS) is considered the most common form of inherited intellectual disability. It is caused by reductions in the expression level or function of a single protein, the Fragile X Mental Retardation Protein (FMRP), a translational regulator which binds to approximately 4% of brain messenger RNAs. Accumulating evidence suggests that FXS is a complex disorder of cognition, involving interactions between genetic and environmental influences, leading to difficulties in acquiring key life skills including motor skills, language, and proper social behaviors. Since many FXS patients also present with one or more features of autism spectrum disorders (ASDs), insights gained from studying the monogenic basis of FXS could pave the way to a greater understanding of underlying features of multigenic ASDs. Here we present an overview of the FXS and FMRP field with the goal of demonstrating how loss of a single protein involved in translational control affects multiple stages of brain development and leads to debilitating consequences on human cognition. We also focus on studies which have rescued or improved FXS symptoms in mice using genetic or therapeutic approaches to reduce protein expression. We end with a brief description of how deficits in translational control are implicated in FXS and certain cases of ASDs, with many recent studies demonstrating that ASDs are likely caused by increases or decreases in the levels of certain key synaptic proteins. The study of FXS and its underlying single genetic cause offers an invaluable opportunity to study how a single gene influences brain development and behavior.
Collapse
Affiliation(s)
- Denise Cook
- Department of Neurology and Neurosurgery, Centre for Research in Neuroscience, The Research Institute of the McGill University Health Centre, Montreal General Hospital, Montreal, Quebec, Canada
| | | | | |
Collapse
|
43
|
Rotschafer S, Razak K. Altered auditory processing in a mouse model of fragile X syndrome. Brain Res 2013; 1506:12-24. [PMID: 23458504 DOI: 10.1016/j.brainres.2013.02.038] [Citation(s) in RCA: 77] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2013] [Accepted: 02/18/2013] [Indexed: 12/30/2022]
Abstract
This study provides the first description of auditory cortical processing in a mouse model of Fragile X Syndrome (FXS). FXS is a genetic cause of intellectual impairment and is an autism spectrum disorder. Human studies with auditory evoked potentials indicate that FXS is associated with abnormal auditory processing. The Fmr1 knock-out (KO) mouse is a useful model for studying FXS. The KO mice show acoustic hypersensitivity and propensity for audiogenic seizures, suggesting altered auditory responses. However, the nature of changes at the neuronal level is not known. Here we conducted in vivo single unit extracellular electrophysiology in the auditory cortex of urethane/xylazine-anesthetized Fmr1 KO mice in response to tones and frequency modulated (FM) sweeps. Using tones as stimuli, we report expanded frequency tuning, enhanced response magnitude, and more variable first spike latencies in Fmr1 KO mice compared to wild-type controls. FM sweep stimuli revealed altered sensitivity to the rate of frequency change indicating abnormal spectrotemporal processing. There was no difference in FM sweep direction selectivity. Consistent with studies of the somatosensory cortex, these data point to hyper-responsiveness of auditory neurons as a key processing abnormality in FXS. Auditory neural responses can serve as outcome measures in preclinical trials of therapeutics for FXS as well as serve as physiological probes to study their mechanisms of action.
Collapse
Affiliation(s)
- Sarah Rotschafer
- Graduate Neuroscience Program and Department of Psychology, University of California, Riverside, CA, USA
| | | |
Collapse
|
44
|
Mechanism of repeat-associated microRNAs in fragile X syndrome. Neural Plast 2012; 2012:104796. [PMID: 22779005 PMCID: PMC3388308 DOI: 10.1155/2012/104796] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2011] [Revised: 02/11/2012] [Accepted: 02/15/2012] [Indexed: 12/14/2022] Open
Abstract
The majority of the human genome is comprised of non-coding DNA, which frequently contains redundant microsatellite-like trinucleotide repeats. Many of these trinucleotide repeats are involved in triplet repeat expansion diseases (TREDs) such as fragile X syndrome (FXS). After transcription, the trinucleotide repeats can fold into RNA hairpins and are further processed by Dicer endoribonuclases to form microRNA (miRNA)-like molecules that are capable of triggering targeted gene-silencing effects in the TREDs. However, the function of these repeat-associated miRNAs (ramRNAs) is unclear. To solve this question, we identified the first native ramRNA in FXS and successfully developed a transgenic zebrafish model for studying its function. Our studies showed that ramRNA-induced DNA methylation of the FMR1 5′-UTR CGG trinucleotide repeat expansion is responsible for both pathological and neurocognitive characteristics linked to the transcriptional FMR1 gene inactivation and the deficiency of its protein product FMRP. FMRP deficiency often causes synapse deformity in the neurons essential for cognition and memory activities, while FMR1 inactivation augments metabotropic glutamate receptor (mGluR)-activated long-term depression (LTD), leading to abnormal neuronal responses in FXS. Using this novel animal model, we may further dissect the etiological mechanisms of TREDs, with the hope of providing insights into new means for therapeutic intervention.
Collapse
|
45
|
Nestor MW, Hoffman DA. Aberrant dendritic excitability: a common pathophysiology in CNS disorders affecting memory? Mol Neurobiol 2012; 45:478-87. [PMID: 22528602 PMCID: PMC3496255 DOI: 10.1007/s12035-012-8265-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2012] [Accepted: 03/29/2012] [Indexed: 01/13/2023]
Abstract
Discovering the etiology of pathophysiologies and aberrant behavior in many central nervous system (CNS) disorders has proven elusive because susceptibility to these diseases can be a product of multiple factors such as genetics, epigenetics, and environment. Advances in molecular biology and wide-scale genomics have shown that a large heterogeneity of genetic mutations are potentially responsible for the neuronal pathologies and dysfunctional behaviors seen in CNS disorders. Despite this seemingly complex array of genetic and physiological factors, many disorders of the CNS converge on common dysfunctions in memory. In this review, we propose that mechanisms underlying the development of many CNS disorders may share an underlying cause involving abnormal dendritic integration of synaptic signals. Through understanding the relationship between molecular genetics and dendritic computation, future research may uncover important links between neuronal physiology at the cellular level and higher-order circuit and network abnormalities observed in CNS disorders, and their subsequent affect on memory.
Collapse
Affiliation(s)
- Michael W. Nestor
- Molecular Neurophysiology and Biophysics Unit, LCSN, NICHD, NIH, 35 Lincoln Drive Room 3C-905, Bethesda, Maryland, 20892
| | - Dax A. Hoffman
- Molecular Neurophysiology and Biophysics Unit, LCSN, NICHD, NIH, 35 Lincoln Drive Room 3C-905, Bethesda, Maryland, 20892
| |
Collapse
|
46
|
Till SM, Wijetunge LS, Seidel VG, Harlow E, Wright AK, Bagni C, Contractor A, Gillingwater TH, Kind PC. Altered maturation of the primary somatosensory cortex in a mouse model of fragile X syndrome. Hum Mol Genet 2012; 21:2143-56. [DOI: 10.1093/hmg/dds030] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
|
47
|
Molecular and Cellular Aspects of Mental Retardation in the Fragile X Syndrome: From Gene Mutation/s to Spine Dysmorphogenesis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2012; 970:517-51. [DOI: 10.1007/978-3-7091-0932-8_23] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
48
|
Meguid NA, Fahim C, Sami R, Nashaat NH, Yoon U, Anwar M, El-Dessouky HM, Shahine EA, Ibrahim AS, Mancini-Marie A, Evans AC. Cognition and lobar morphology in full mutation boys with fragile X syndrome. Brain Cogn 2011; 78:74-84. [PMID: 22070923 DOI: 10.1016/j.bandc.2011.09.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2011] [Revised: 09/13/2011] [Accepted: 09/15/2011] [Indexed: 01/05/2023]
Abstract
The aims of the present study are twofold: (1) to examine cortical morphology (CM) associated with alterations in cognition in fragile X syndrome (FXS); (2) to characterize the CM profile of FXS versus FXS with an autism diagnosis (FXS+Aut) as a preliminary attempt to further elucidate the behavioral distinctions between the two sub-groups. We used anatomical magnetic resonance imaging surface-based morphometry in 21 male children (FXS N=11 and age [2.27-13.3] matched controls [C] N=10). We found (1) increased whole hemispheric and lobar cortical volume, cortical thickness and cortical complexity bilaterally, yet insignificant changes in hemispheric surface area and gyrification index in FXS compared to C; (2) linear regression analyses revealed significant negative correlations between CM and cognition; (3) significant CM differences between FXS and FXS+Aut associated with their distinctive behavioral phenotypes. These findings are critical in understanding the neuropathophysiology of one of the most common intellectual deficiency syndromes associated with altered cognition as they provide human in vivo information about genetic control of CM and cognition.
Collapse
Affiliation(s)
- Nagwa A Meguid
- Department of Research on Children with Special Needs, Medical Genetics Division, The National Research Centre, Cairo, Egypt
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Van der Molen MJW, Van der Molen MW, Ridderinkhof KR, Hamel BCJ, Curfs LMG, Ramakers GJA. Auditory and visual cortical activity during selective attention in fragile X syndrome: a cascade of processing deficiencies. Clin Neurophysiol 2011; 123:720-9. [PMID: 21958658 DOI: 10.1016/j.clinph.2011.08.023] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2011] [Revised: 07/11/2011] [Accepted: 08/20/2011] [Indexed: 01/06/2023]
Abstract
OBJECTIVE This study examined whether attention deficits in fragile X syndrome (FXS) can be traced back to abnormalities in basic information processing. METHOD Sixteen males with FXS and 22 age-matched control participants (mean age 29 years) performed a standard oddball task to examine selective attention in both auditory and visual modalities. Five FXS males were excluded from analysis because they performed below chance level on the auditory task. ERPs were recorded to investigate the N1, P2, N2b, and P3b components. RESULTS N1 and N2b components were significantly enhanced in FXS males to both auditory and visual stimuli. Interestingly, in FXS males, the P3b to auditory stimuli was significantly reduced relative to visual stimuli. These modality differences in information processing corresponded to behavioral results, showing more errors on the auditory than on the visual task. CONCLUSIONS The current findings suggest that attentional impairments in FXS at the behavioral level can be traced back to abnormalities in event-related cortical activity. These information processing abnormalities in FXS may hinder the allocation of attentional resources needed for optimal processing at higher-levels. SIGNIFICANCE These findings demonstrate that auditory information processing in FXS males is critically impaired relative to visual information processing.
Collapse
Affiliation(s)
- M J W Van der Molen
- Department of Developmental Psychology, University of Amsterdam, The Netherlands.
| | | | | | | | | | | |
Collapse
|
50
|
Paluszkiewicz SM, Martin BS, Huntsman MM. Fragile X syndrome: the GABAergic system and circuit dysfunction. Dev Neurosci 2011; 33:349-64. [PMID: 21934270 DOI: 10.1159/000329420] [Citation(s) in RCA: 129] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2010] [Accepted: 03/10/2011] [Indexed: 12/18/2022] Open
Abstract
Fragile X syndrome (FXS) is a neurodevelopmental disorder characterized by intellectual disability, sensory hypersensitivity, and high incidences of autism spectrum disorders and epilepsy. These phenotypes are suggestive of defects in neural circuit development and imbalances in excitatory glutamatergic and inhibitory GABAergic neurotransmission. While alterations in excitatory synapse function and plasticity are well-established in Fmr1 knockout (KO) mouse models of FXS, a number of recent electrophysiological and molecular studies now identify prominent defects in inhibitory GABAergic transmission in behaviorally relevant forebrain regions such as the amygdala, cortex, and hippocampus. In this review, we summarize evidence for GABAergic system dysfunction in FXS patients and Fmr1 KO mouse models alike. We then discuss some of the known developmental roles of GABAergic signaling, as well as the development and refinement of GABAergic synapses as a framework for understanding potential causes of mature circuit dysfunction. Finally, we highlight the GABAergic system as a relevant target for the treatment of FXS.
Collapse
Affiliation(s)
- Scott M Paluszkiewicz
- Center for Neuroscience Research, Children's National Medical Center, Washington, DC 20010, USA
| | | | | |
Collapse
|