1
|
Wang Z, Qiao D, Chen H, Zhang S, Zhang B, Zhang J, Hu X, Wang C, Cui H, Wang X, Li S. Effects of Fmr1 Gene Mutations on Sex Differences in Autism-Like Behavior and Dendritic Spine Development in Mice and Transcriptomic Studies. Neuroscience 2023; 534:16-28. [PMID: 37852411 DOI: 10.1016/j.neuroscience.2023.10.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Revised: 10/01/2023] [Accepted: 10/04/2023] [Indexed: 10/20/2023]
Abstract
Fragile X syndrome (FXS) is the most common single gene disorder contributing to autism spectrum disorder (ASD). Although significant sex differences are observed in FXS, few studies have focused on the phenotypic characteristics as well as the differences in brain pathological changes and gene expression in FXS by sex. Therefore, we analyzed sex differences in autism-like behavior and dendritic spine development in two-month-old male and female Fmr1 KO and C57 mice and evaluated the mechanisms at transcriptome level. Results suggest that Fmr1 KO mice display sex differences in autism-like behavior and dendritic spine density. Compared to females, male had more severe effects on anxiety, repetitive stereotype-like behaviors, and socializing, with higher dendritic spine density. Furthermore, two male-biased and five female-biased expressed genes were screened based on KEGG pathway enrichment and protein-protein interaction (PPI) analyses. In conclusion, our findings show mutations in the Fmr1 gene lead to aberrant expression of related genes and affect the sex-differentiated behavioral phenotypes of Fmr1 KO mice by affecting brain development and functional architecture, and suggest future studies should focus on including female subjects to comprehensively reflect the differentiation of FXS in both sexes and develop more precise and effective therapeutic strategies.
Collapse
Affiliation(s)
- Zhao Wang
- Department of Anatomy, Hebei Medical University, Shijiazhuang, China
| | - Dan Qiao
- Department of Anatomy, Hebei Medical University, Shijiazhuang, China
| | - Huan Chen
- Department of Anatomy, Hebei Medical University, Shijiazhuang, China; Neuroscience Research Center, Hebei Medical University, Shijiazhuang, China; Hebei Key Laboratory of Neurodegenerative Disease Mechanism, Shijiazhuang, China
| | - Shihua Zhang
- Grade 2018, Basic Medicine, Hebei Medical University, Shijiazhuang, China
| | - Bohan Zhang
- Department of Anatomy, Hebei Medical University, Shijiazhuang, China
| | - Jingbao Zhang
- Grade 2020, Basic Medicine, Hebei Medical University, Shijiazhuang, China
| | - Xiangting Hu
- Grade 2020, Basic Medicine, Hebei Medical University, Shijiazhuang, China
| | - Chang Wang
- Department of Anatomy, Hebei Medical University, Shijiazhuang, China; Neuroscience Research Center, Hebei Medical University, Shijiazhuang, China; Hebei Key Laboratory of Neurodegenerative Disease Mechanism, Shijiazhuang, China
| | - Huixian Cui
- Department of Anatomy, Hebei Medical University, Shijiazhuang, China; Neuroscience Research Center, Hebei Medical University, Shijiazhuang, China; Hebei Key Laboratory of Neurodegenerative Disease Mechanism, Shijiazhuang, China
| | - Xia Wang
- Child Health (Psychological Behavior) Department, Children's Hospital of Hebei Province, Shijiazhuang, China.
| | - Sha Li
- Department of Anatomy, Hebei Medical University, Shijiazhuang, China; Neuroscience Research Center, Hebei Medical University, Shijiazhuang, China; Hebei Key Laboratory of Neurodegenerative Disease Mechanism, Shijiazhuang, China; The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, China.
| |
Collapse
|
2
|
Cording KR, Bateup HS. Altered motor learning and coordination in mouse models of autism spectrum disorder. Front Cell Neurosci 2023; 17:1270489. [PMID: 38026686 PMCID: PMC10663323 DOI: 10.3389/fncel.2023.1270489] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 09/25/2023] [Indexed: 12/01/2023] Open
Abstract
Autism spectrum disorder (ASD) is a complex neurodevelopmental disorder with increasing prevalence. Over 1,000 risk genes have now been implicated in ASD, suggesting diverse etiology. However, the diagnostic criteria for the disorder still comprise two major behavioral domains - deficits in social communication and interaction, and the presence of restricted and repetitive patterns of behavior (RRBs). The RRBs associated with ASD include both stereotyped repetitive movements and other motor manifestations including changes in gait, balance, coordination, and motor skill learning. In recent years, the striatum, the primary input center of the basal ganglia, has been implicated in these ASD-associated motor behaviors, due to the striatum's role in action selection, motor learning, and habit formation. Numerous mouse models with mutations in ASD risk genes have been developed and shown to have alterations in ASD-relevant behaviors. One commonly used assay, the accelerating rotarod, allows for assessment of both basic motor coordination and motor skill learning. In this corticostriatal-dependent task, mice walk on a rotating rod that gradually increases in speed. In the extended version of this task, mice engage striatal-dependent learning mechanisms to optimize their motor routine and stay on the rod for longer periods. This review summarizes the findings of studies examining rotarod performance across a range of ASD mouse models, and the resulting implications for the involvement of striatal circuits in ASD-related motor behaviors. While performance in this task is not uniform across mouse models, there is a cohort of models that show increased rotarod performance. A growing number of studies suggest that this increased propensity to learn a fixed motor routine may reflect a common enhancement of corticostriatal drive across a subset of mice with mutations in ASD-risk genes.
Collapse
Affiliation(s)
- Katherine R. Cording
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA, United States
| | - Helen S. Bateup
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA, United States
- Molecular and Cell Biology Department, University of California, Berkeley, Berkeley, CA, United States
- Chan Zuckerberg Biohub, San Francisco, CA, United States
| |
Collapse
|
3
|
Chen H, Qiao D, Wang C, Zhang B, Wang Z, Tang L, Wang Y, Zhang R, Zhang Y, Song L, Zuo H, Guo F, Wang X, Li S, Cui H. Fragile X Mental Retardation Protein Mediates the Effects of Androgen on Hippocampal PSD95 Expression and Dendritic Spines Density/Morphology and Autism-Like Behaviors Through miR-125a. Front Cell Neurosci 2022; 16:872347. [PMID: 35530178 PMCID: PMC9074813 DOI: 10.3389/fncel.2022.872347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 03/30/2022] [Indexed: 12/02/2022] Open
Abstract
Dysregulated synaptic plasticity is a key feature of neurodevelopmental disorders, including autism. This study investigated whether Fragile X mental retardation protein (FMRP), a selective RNA-binding protein that regulates synaptic protein expression by interacting with miRNAs, mediates the effects of androgens that play an important role in regulating the synaptic plasticity in the hippocampus. Experiments using mouse hippocampal neuron HT22 cells demonstrated that dihydrotestosterone (DHT) increased the expression of postsynaptic density protein 95 (PSD95) by inhibiting FMRP expression. Administration of miR-125a inhibitor upregulated the PSD95 expression and significantly increased the DHT-induced upregulation of PSD95. FMRP knockdown in HT22 cells reduced the expression of miR-125a. Moreover, miR-125a inhibitor upregulated the PSD95 expression in the DHT-treated HT22 cells with FMRP knockdown. Subsequently, the effects of androgen-mediated via FMRP in regulating neural behaviors and PSD95 expression and dendritic spines density/morphology were investigated using Fmr1 knockout (KO) and wild-type littermate (WT) mice. The castration of WT mice reduced the androgen levels, aggravated anxiety and depression, and impaired learning and memory and sociability of mice. DHT supplementation post-castration reversed the alterations in density and maturity of dendritic spines of hippocampal neurons and behavioral disorders in WT mice; however, it did not reveal such effects in Fmr1 KO mice. Further, immunohistochemical staining and western blotting analyses after knocking down miR-125a revealed similar effects of castration and post-castration DHT supplementation on PSD95 protein expression. These findings clarified that FMRP mediated the effects of DHT through miR-125a in regulating the expression of hippocampal synaptic protein PSD95. This study provides evidence for the neuroprotective mechanism of androgen in PSD95 expression and dendritic spines density/morphology and suggests that treatment interventions with androgen could be helpful for the management of synaptic plasticity disorders.
Collapse
Affiliation(s)
- Huan Chen
- Department of Anatomy, Hebei Medical University, Shijiazhuang, China
- Neuroscience Research Center, Hebei Medical University, Shijiazhuang, China
| | - Dan Qiao
- Department of Anatomy, Hebei Medical University, Shijiazhuang, China
- Neuroscience Research Center, Hebei Medical University, Shijiazhuang, China
| | - Chang Wang
- Department of Anatomy, Hebei Medical University, Shijiazhuang, China
- Neuroscience Research Center, Hebei Medical University, Shijiazhuang, China
- Hebei Key Laboratory of Neurodegenerative Disease Mechanism, Shijiazhuang, China
| | - Bohan Zhang
- Department of Anatomy, Hebei Medical University, Shijiazhuang, China
- Neuroscience Research Center, Hebei Medical University, Shijiazhuang, China
| | - Zhao Wang
- Department of Anatomy, Hebei Medical University, Shijiazhuang, China
- Neuroscience Research Center, Hebei Medical University, Shijiazhuang, China
| | - Longmei Tang
- Department of Epidemiology and Statistics, Hebei Medical University, Shijiazhuang, China
| | - Yibo Wang
- Clinical Medicine, Hebei Medical University, Shijiazhuang, China
| | - Ran Zhang
- Clinical Medicine, Hebei Medical University, Shijiazhuang, China
| | - Yizhou Zhang
- Department of Anatomy, Hebei Medical University, Shijiazhuang, China
- Neuroscience Research Center, Hebei Medical University, Shijiazhuang, China
- Hebei Key Laboratory of Neurodegenerative Disease Mechanism, Shijiazhuang, China
| | - Leigang Song
- Department of Anatomy, Hebei Medical University, Shijiazhuang, China
- Neuroscience Research Center, Hebei Medical University, Shijiazhuang, China
| | - Hongchun Zuo
- Department of Anatomy, Hebei Medical University, Shijiazhuang, China
- Neuroscience Research Center, Hebei Medical University, Shijiazhuang, China
| | - Fangzhen Guo
- Department of Anatomy, Hebei Medical University, Shijiazhuang, China
- Neuroscience Research Center, Hebei Medical University, Shijiazhuang, China
| | - Xia Wang
- Department of Child Health (Psychological Behavior), Children’s Hospital of Hebei Province, Shijiazhuang, China
| | - Sha Li
- Department of Anatomy, Hebei Medical University, Shijiazhuang, China
- Neuroscience Research Center, Hebei Medical University, Shijiazhuang, China
- Hebei Key Laboratory of Neurodegenerative Disease Mechanism, Shijiazhuang, China
| | - Huixian Cui
- Department of Anatomy, Hebei Medical University, Shijiazhuang, China
- Neuroscience Research Center, Hebei Medical University, Shijiazhuang, China
- Hebei Key Laboratory of Neurodegenerative Disease Mechanism, Shijiazhuang, China
| |
Collapse
|
4
|
Hayashi Y, Ford LK, Fioriti L, McGurk L, Zhang M. Liquid-Liquid Phase Separation in Physiology and Pathophysiology of the Nervous System. J Neurosci 2021; 41:834-844. [PMID: 33472825 PMCID: PMC7880275 DOI: 10.1523/jneurosci.1656-20.2020] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 12/17/2020] [Accepted: 12/18/2020] [Indexed: 12/12/2022] Open
Abstract
Molecules within cells are segregated into functional domains to form various organelles. While some of those organelles are delimited by lipid membranes demarcating their constituents, others lack a membrane enclosure. Recently, liquid-liquid phase separation (LLPS) revolutionized our view of how segregation of macromolecules can produce membraneless organelles. While the concept of LLPS has been well studied in the areas of soft matter physics and polymer chemistry, its significance has only recently been recognized in the field of biology. It occurs typically between macromolecules that have multivalent interactions. Interestingly, these features are present in many molecules that exert key functions within neurons. In this review, we cover recent topics of LLPS in different contexts of neuronal physiology and pathology.
Collapse
Affiliation(s)
- Yasunori Hayashi
- Department of Pharmacology, Kyoto University Graduate School of Medicine, Kyoto 606-8501, Japan
| | - Lenzie K Ford
- Zuckerman Mind, Brain, Behavior Institute, Columbia University, New York, New York 10027
| | - Luana Fioriti
- Department of Neuroscience, Mario Negri Institute for Pharmacological Research, Istituto Di Ricovero e Cura a Carattere Scientifico, Milan 20156, Italy
| | - Leeanne McGurk
- Cell and Developmental Biology, School of Life Sciences, University of Dundee, Dundee DD1 5EH, United Kingdom
| | - Mingjie Zhang
- Division of Life Science, Hong Kong University of Science and Technology, Kowloon, Hong Kong, China
- School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
| |
Collapse
|
5
|
Nie PY, Tong L, Li MD, Fu CH, Peng JB, Ji LL. miR-142 downregulation alleviates rat PTSD-like behaviors, reduces the level of inflammatory cytokine expression and apoptosis in hippocampus, and upregulates the expression of fragile X mental retardation protein. J Neuroinflammation 2021; 18:17. [PMID: 33407653 PMCID: PMC7788709 DOI: 10.1186/s12974-020-02064-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 12/16/2020] [Indexed: 11/30/2022] Open
Abstract
Background FMRP is a selective mRNA-binding protein that regulates protein synthesis at synapses, and its loss may lead to the impairment of trace fear memory. Previously, we found that FMRP levels in the hippocampus of rats with post-traumatic stress disorder (PTSD) were decreased. However, the mechanism underlying these changes remains unclear. Methods Forty-eight male Sprague-Dawley rats were randomly divided into four groups. The experimental groups were treated with the single-prolonged stress (SPS) procedure and injected with a lentivirus-mediated inhibitor of miR-142-5p. Behavior test as well as morphology and molecular biology experiments were performed to detect the effect of miR-142 downregulation on PTSD, which was further verified by in vitro experiments. Results We found that silence of miRNA-142 (miR-142), an upstream regulator of FMRP, could alleviate PTSD-like behaviors of rats exposed to the SPS paradigm. MiR-142 silence not only decreased the levels of proinflammatory mediators, such as interleukin-1β, interleukin-6, and tumor necrosis factor-α, but also increased the expressive levels of synaptic proteins including PSD95 and synapsin I in the hippocampus, which was one of the key brain regions associated with PTSD. We further detected that miR-142 silence also downregulated the transportation of nuclear factor kappa-B (NF-κB) into the nuclei of neurons and might further affect the morphology of neurons. Conclusions The results revealed miR-142 downregulation could alleviate PTSD-like behaviors through attenuating neuroinflammation in the hippocampus of SPS rats by binding to FMRP.
Collapse
Affiliation(s)
- Peng-Yin Nie
- Department of Anatomy, College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Lei Tong
- Department of Anatomy, College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Ming-Da Li
- Department of 1st Clinical Medicine, China Medical University, Shenyang, China
| | - Chang-Hai Fu
- Department of Anatomy, College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Jun-Bo Peng
- Department of Anatomy, College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Li-Li Ji
- Department of Anatomy, College of Basic Medical Sciences, China Medical University, Shenyang, China.
| |
Collapse
|
6
|
Ford LK, Fioriti L. Coiled-Coil Motifs of RNA-Binding Proteins: Dynamicity in RNA Regulation. Front Cell Dev Biol 2020; 8:607947. [PMID: 33330512 PMCID: PMC7710910 DOI: 10.3389/fcell.2020.607947] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 10/30/2020] [Indexed: 01/29/2023] Open
Abstract
Neuronal granules are biomolecular condensates that concentrate high quantities of RNAs and RNA-related proteins within neurons. These dense packets of information are trafficked from the soma to distal sites rich in polysomes, where local protein synthesis can occur. Movement of neuronal granules to distal sites, and local protein synthesis, play a critical role in synaptic plasticity. The formation of neuronal granules is intriguing; these granules lack a membrane and instead phase separate due to protein and RNA interactions. Low complexity motifs and RNA binding domains are highly prevalent in these proteins. Here, we introduce the role that coiled-coil motifs play in neuronal granule proteins, and investigate the structure-function relationship of coiled-coil proteins in RNA regulation. Interestingly, low complexity domains and coiled-coil motifs are highly dynamic, allowing for increased functional response to environmental influences. Finally, biomolecular condensates have been suggested to drive the formation of toxic, neurodegenerative proteins such as TDP-43 and tau. Here, we review the conversion of coiled-coil motifs to amyloid structures, and speculate a role that neuronal granules play in coiled-coil to amyloid conversions of neurodegenerative proteins.
Collapse
Affiliation(s)
- Lenzie K Ford
- Department of Neuroscience, Zuckerman Institute, Columbia University, New York, NY, United States
| | - Luana Fioriti
- Laboratory of Molecular Mechanisms of Polyglutamine Disorders, Department of Neuroscience, Dulbecco Telethon Institute, Istituto di Ricerche Farmacologiche Mario Negri (IRCCS), Milan, Italy
| |
Collapse
|
7
|
Zhang J, Wang G, He WW, Losh M, Berry-Kravis E, Funk WE. Expression and Characterization of Human Fragile X Mental Retardation Protein Isoforms and Interacting Proteins in Human Cells. PROTEOMICS INSIGHTS 2019; 10:1178641818825268. [PMID: 30853789 PMCID: PMC6399764 DOI: 10.1177/1178641818825268] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Accepted: 12/21/2018] [Indexed: 11/16/2022]
Abstract
Fragile X mental retardation protein is an mRNA-binding protein associated with phenotypic manifestations of fragile X syndrome, an X-linked disorder caused by mutation in the FMR1 gene that is the most common inherited cause of intellectual disability. Despite the well-studied genetic mechanism of the disease, the proteoforms of fragile X mental retardation protein have not been thoroughly characterized. Here, we report the expression and mass spectrometric characterization of human fragile X mental retardation protein. FMR1 cDNA clone was transfected into human HEK293 cells to express the full-length human fragile X mental retardation protein. Purified fragile X mental retardation protein was subjected to trypsin digestion and characterized by mass spectrometry. Results show 80.5% protein sequence coverage of fragile X mental retardation protein (Q06787, FMR1_HUMAN) including both the N- and C-terminal peptides, indicating successful expression of the full-length protein. Identified post-translational modifications include N-terminal acetylation, phosphorylation (Ser600), and methylation (Arg290, 471, and 474). In addition to the full-length fragile X mental retardation protein isoform (isoform 6), two endogenous fragile X mental retardation protein alternative splicing isoforms (isoforms 4 and 7), as well as fragile X mental retardation protein interacting proteins, were also identified in the co-purified samples, suggesting the interaction network of the human fragile X mental retardation protein. Quantification was performed at the peptide level, and this information provides important reference for the future development of a targeted assay for quantifying fragile X mental retardation protein in clinical samples. Collectively, this study provides the first comprehensive report of human fragile X mental retardation protein proteoforms and may help advance the mechanistic understanding of fragile X syndrome and related phenotypes associated with the FMR1 mutation.
Collapse
Affiliation(s)
- Jiang Zhang
- Department of Preventive Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | | | - Wei-Wu He
- OriGene Technology, Inc., Rockville, MD, USA
| | - Molly Losh
- Department of Communication Sciences and Disorders, Northwestern University, Evanston, IL, USA
| | - Elizabeth Berry-Kravis
- Departments of Biochemistry, Neurological Sciences and Pediatrics, Rush University, Chicago, IL, USA
| | - William E Funk
- Department of Preventive Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| |
Collapse
|
8
|
Hessl D, Harvey D, Sansone S, Crestodina C, Chin J, Joshi R, Hagerman RJ, Berry‐Kravis E. Effects of mavoglurant on visual attention and pupil reactivity while viewing photographs of faces in Fragile X Syndrome. PLoS One 2019; 14:e0209984. [PMID: 30653533 PMCID: PMC6336311 DOI: 10.1371/journal.pone.0209984] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Accepted: 12/14/2018] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Numerous preclinical studies have supported the theory that enhanced activation of mGluR5 signaling, due to the absence or reduction of the FMR1 protein, contributes to cognitive and behavioral deficits in patients with fragile X syndrome (FXS). However multiple phase 2 controlled trials in patients with FXS have failed to demonstrate efficacy of compounds that negatively modulate mGluR5, including two phase 2b randomized controlled trials (RCT) of mavoglurant (AFQ056, Novartis Pharma AG), when the primary measures of interest were behavioral ratings. This has cast some doubt onto the translation of the mGluR5 theory from animal models to humans with the disorder. METHODS We evaluated social gaze behavior-a key phenotypic feature of the disorder-and sympathetic nervous system influence on pupil size using a previously-validated eye tracking paradigm as a biobehavioral probe, in 57 adolescent or adult patients with FXS at baseline and following three months of blinded treatment with one of three doses of mavoglurant or placebo, within the context of the AFQ056 RCTs. RESULTS Patients with FXS treated with mavoglurant demonstrated increased total absolute looking time and number of fixations to the eye region while viewing human faces relative to baseline, and compared to those treated with placebo. In addition, patients had greater pupil reactivity to faces relative to baseline following mavoglurant treatment compared to placebo. DISCUSSION The study shows that negative modulation of mGluR5 activity improves eye gaze behavior and alters sympathetically-driven reactivity to faces in patients with FXS, providing preliminary evidence of this drug's impact on behavior in humans with the disorder.
Collapse
Affiliation(s)
- David Hessl
- MIND Institute, University of California Davis Medical Center, Sacramento, California, United States of America
- Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, California, United States of America
| | - Danielle Harvey
- Division of Biostatistics, Department of Public Health Sciences, University of California Davis School of Medicine, Davis, CA, United States of America
| | - Stephanie Sansone
- MIND Institute, University of California Davis Medical Center, Sacramento, California, United States of America
- Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, California, United States of America
| | - Crystal Crestodina
- Department of Pediatrics, Rush University Medical Center, Chicago, IL, United States of America
| | - Jamie Chin
- Department of Pediatrics, Rush University Medical Center, Chicago, IL, United States of America
| | - Reshma Joshi
- Department of Pediatrics, Rush University Medical Center, Chicago, IL, United States of America
| | - Randi J. Hagerman
- MIND Institute, University of California Davis Medical Center, Sacramento, California, United States of America
- Department of Pediatrics, University of California Davis School of Medicine, Sacramento, California, United States of America
| | - Elizabeth Berry‐Kravis
- Department of Pediatrics, Rush University Medical Center, Chicago, IL, United States of America
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, United States of America
- Department of Biochemistry, Rush University Medical Center, Chicago, IL, United States of America
| |
Collapse
|
9
|
Saré RM, Song A, Loutaev I, Cook A, Maita I, Lemons A, Sheeler C, Smith CB. Negative Effects of Chronic Rapamycin Treatment on Behavior in a Mouse Model of Fragile X Syndrome. Front Mol Neurosci 2018; 10:452. [PMID: 29375310 PMCID: PMC5770365 DOI: 10.3389/fnmol.2017.00452] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Accepted: 12/22/2017] [Indexed: 01/08/2023] Open
Abstract
Fragile X syndrome (FXS), the most common form of inherited intellectual disability, is also highly associated with autism spectrum disorders (ASD). It is caused by expansion of a CGG repeat sequence on the X chromosome resulting in silencing of the FMR1 gene. This is modeled in the mouse by deletion of Fmr1 (Fmr1 KO). Fmr1 KO mice recapitulate many of the behavioral features of the disorder including seizure susceptibility, hyperactivity, impaired social behavior, sleep problems, and learning and memory deficits. The mammalian target of rapamycin pathway (mTORC1) is upregulated in Fmr1 KO mice and is thought to be important for the pathogenesis of this disorder. We treated Fmr1 KO mice chronically with an mTORC1 inhibitor, rapamycin, to determine if rapamycin treatment could reverse behavioral phenotypes. We performed open field, zero maze, social behavior, sleep, passive avoidance, and audiogenic seizure testing. We found that pS6 was upregulated in Fmr1 KO mice and normalized by rapamycin treatment, but, except for an anxiogenic effect, it did not reverse any of the behavioral phenotypes examined. In fact, rapamycin treatment had an adverse effect on sleep and social behavior in both control and Fmr1 KO mice. These results suggest that targeting the mTOR pathway in FXS is not a good treatment strategy and that other pathways should be considered.
Collapse
Affiliation(s)
- Rachel M Saré
- Section on Neuroadaptation and Protein Metabolism, National Institute of Mental Health, National Institutes of Health, United States Department of Health and Human Services, Bethesda, MD, United States
| | - Alex Song
- Section on Neuroadaptation and Protein Metabolism, National Institute of Mental Health, National Institutes of Health, United States Department of Health and Human Services, Bethesda, MD, United States
| | - Inna Loutaev
- Section on Neuroadaptation and Protein Metabolism, National Institute of Mental Health, National Institutes of Health, United States Department of Health and Human Services, Bethesda, MD, United States
| | - Anna Cook
- Section on Neuroadaptation and Protein Metabolism, National Institute of Mental Health, National Institutes of Health, United States Department of Health and Human Services, Bethesda, MD, United States
| | - Isabella Maita
- Section on Neuroadaptation and Protein Metabolism, National Institute of Mental Health, National Institutes of Health, United States Department of Health and Human Services, Bethesda, MD, United States
| | - Abigail Lemons
- Section on Neuroadaptation and Protein Metabolism, National Institute of Mental Health, National Institutes of Health, United States Department of Health and Human Services, Bethesda, MD, United States
| | - Carrie Sheeler
- Section on Neuroadaptation and Protein Metabolism, National Institute of Mental Health, National Institutes of Health, United States Department of Health and Human Services, Bethesda, MD, United States
| | - Carolyn B Smith
- Section on Neuroadaptation and Protein Metabolism, National Institute of Mental Health, National Institutes of Health, United States Department of Health and Human Services, Bethesda, MD, United States
| |
Collapse
|
10
|
Dear ML, Shilts J, Broadie K. Neuronal activity drives FMRP- and HSPG-dependent matrix metalloproteinase function required for rapid synaptogenesis. Sci Signal 2017; 10:eaan3181. [PMID: 29114039 PMCID: PMC5743058 DOI: 10.1126/scisignal.aan3181] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Matrix metalloproteinase (MMP) functions modulate synapse formation and activity-dependent plasticity. Aberrant MMP activity is implicated in fragile X syndrome (FXS), a disease caused by the loss of the RNA-binding protein FMRP and characterized by neurological dysfunction and intellectual disability. Gene expression studies in Drosophila suggest that Mmps cooperate with the heparan sulfate proteoglycan (HSPG) glypican co-receptor Dally-like protein (Dlp) to restrict trans-synaptic Wnt signaling and that synaptogenic defects in the fly model of FXS are alleviated by either inhibition of Mmp or genetic reduction of Dlp. We used the Drosophila neuromuscular junction (NMJ) glutamatergic synapse to test activity-dependent Dlp and Mmp intersections in the context of FXS. We found that rapid, activity-dependent synaptic bouton formation depended on secreted Mmp1. Acute neuronal stimulation reduced the abundance of Mmp2 but increased that of both Mmp1 and Dlp, as well as enhanced the colocalization of Dlp and Mmp1 at the synapse. Dlp function promoted Mmp1 abundance, localization, and proteolytic activity around synapses. Dlp glycosaminoglycan (GAG) chains mediated this functional interaction with Mmp1. In the FXS fly model, activity-dependent increases in Mmp1 abundance and activity were lost but were restored by reducing the amount of synaptic Dlp. The data suggest that neuronal activity-induced, HSPG-dependent Mmp regulation drives activity-dependent synaptogenesis and that this is impaired in FXS. Thus, exploring this mechanism further may reveal therapeutic targets that have the potential to restore synaptogenesis in FXS patients.
Collapse
Affiliation(s)
- Mary L Dear
- Department of Biological Sciences, Vanderbilt University, Nashville, TN 37232, USA
| | - Jarrod Shilts
- Department of Biological Sciences, Vanderbilt University, Nashville, TN 37232, USA
| | - Kendal Broadie
- Department of Biological Sciences, Vanderbilt University, Nashville, TN 37232, USA.
- Department of Cell and Developmental Biology, Vanderbilt University and Medical School, Nashville, TN 37232, USA
- Vanderbilt Brain Institute, Vanderbilt University and Medical School, Nashville, TN 37232, USA
| |
Collapse
|
11
|
Zhang K, Li YJ, Guo Y, Zheng KY, Yang Q, Yang L, Wang XS, Song Q, Chen T, Zhuo M, Zhao MG. Elevated progranulin contributes to synaptic and learning deficit due to loss of fragile X mental retardation protein. Brain 2017; 140:3215-3232. [DOI: 10.1093/brain/awx265] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Accepted: 08/20/2017] [Indexed: 01/09/2023] Open
Affiliation(s)
- Kun Zhang
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi’an, 710032l, China
| | - Yu-jiao Li
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi’an, 710032l, China
| | - Yanyan Guo
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi’an, 710032l, China
| | - Kai-yin Zheng
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi’an, 710032l, China
| | - Qi Yang
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi’an, 710032l, China
| | - Le Yang
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi’an, 710032l, China
| | - Xin-shang Wang
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi’an, 710032l, China
| | - Qian Song
- Center for Neuron and Disease, Frontier Institutes of Life Science and of Science and Technology, Xi’an Jiaotong University, Xi’an, 710032, China
| | - Tao Chen
- Center for Neuron and Disease, Frontier Institutes of Life Science and of Science and Technology, Xi’an Jiaotong University, Xi’an, 710032, China
- Department of Anatomy, Histology and Embryology and K.K. Leung Brain Research Center, Fourth Military Medical University, Xi’an, 710032, China
| | - Min Zhuo
- Center for Neuron and Disease, Frontier Institutes of Life Science and of Science and Technology, Xi’an Jiaotong University, Xi’an, 710032, China
| | - Ming-gao Zhao
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi’an, 710032l, China
- Center for Neuron and Disease, Frontier Institutes of Life Science and of Science and Technology, Xi’an Jiaotong University, Xi’an, 710032, China
| |
Collapse
|
12
|
Kang JY, Chadchankar J, Vien TN, Mighdoll MI, Hyde TM, Mather RJ, Deeb TZ, Pangalos MN, Brandon NJ, Dunlop J, Moss SJ. Deficits in the activity of presynaptic γ-aminobutyric acid type B receptors contribute to altered neuronal excitability in fragile X syndrome. J Biol Chem 2017; 292:6621-6632. [PMID: 28213518 PMCID: PMC5399111 DOI: 10.1074/jbc.m116.772541] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2016] [Revised: 02/07/2017] [Indexed: 11/06/2022] Open
Abstract
The behavioral and anatomical deficits seen in fragile X syndrome (FXS) are widely believed to result from imbalances in the relative strengths of excitatory and inhibitory neurotransmission. Although modified neuronal excitability is thought to be of significance, the contribution that alterations in GABAergic inhibition play in the pathophysiology of FXS are ill defined. Slow sustained neuronal inhibition is mediated by γ-aminobutyric acid type B (GABAB) receptors, which are heterodimeric G-protein-coupled receptors constructed from R1a and R2 or R1b and R2 subunits. Via the activation of Gi/o, they limit cAMP accumulation, diminish neurotransmitter release, and induce neuronal hyperpolarization. Here we reveal that selective deficits in R1a subunit expression are seen in Fmr1 knock-out mice (KO) mice, a widely used animal model of FXS, but the levels of the respective mRNAs were unaffected. Similar trends of R1a expression were seen in a subset of FXS patients. GABAB receptors (GABABRs) exert powerful pre- and postsynaptic inhibitory effects on neurotransmission. R1a-containing GABABRs are believed to mediate presynaptic inhibition in principal neurons. In accordance with this result, deficits in the ability of GABABRs to suppress glutamate release were seen in Fmr1-KO mice. In contrast, the ability of GABABRs to suppress GABA release and induce postsynaptic hyperpolarization was unaffected. Significantly, this deficit contributes to the pathophysiology of FXS as the GABABR agonist (R)-baclofen rescued the imbalances between excitatory and inhibitory neurotransmission evident in Fmr1-KO mice. Collectively, our results provided evidence that selective deficits in the activity of presynaptic GABABRs contribute to the pathophysiology of FXS.
Collapse
Affiliation(s)
- Ji-Yong Kang
- From the AstraZeneca Tufts Laboratory for Basic and Translational Neuroscience, Tufts University School of Medicine, Boston, Massachusetts 02111
| | - Jayashree Chadchankar
- From the AstraZeneca Tufts Laboratory for Basic and Translational Neuroscience, Tufts University School of Medicine, Boston, Massachusetts 02111
| | - Thuy N Vien
- From the AstraZeneca Tufts Laboratory for Basic and Translational Neuroscience, Tufts University School of Medicine, Boston, Massachusetts 02111
| | | | - Thomas M Hyde
- the Lieber Institute for Brain Development and
- Departments of Neurology and Psychiatry and Behavioral Sciences, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
| | - Robert J Mather
- From the AstraZeneca Tufts Laboratory for Basic and Translational Neuroscience, Tufts University School of Medicine, Boston, Massachusetts 02111
- Neuroscience, Innovative Medicines and Early Development, AstraZeneca, Waltham, Massachusetts 02451
| | - Tarek Z Deeb
- From the AstraZeneca Tufts Laboratory for Basic and Translational Neuroscience, Tufts University School of Medicine, Boston, Massachusetts 02111
| | - Menelas N Pangalos
- Innovative Medicines and Early Development, AstraZeneca, Melbourn Science Park, Cambridge Road, Royston Herts SG8 6EE, United Kingdom, and
| | - Nicholas J Brandon
- From the AstraZeneca Tufts Laboratory for Basic and Translational Neuroscience, Tufts University School of Medicine, Boston, Massachusetts 02111
- Neuroscience, Innovative Medicines and Early Development, AstraZeneca, Waltham, Massachusetts 02451
| | - John Dunlop
- From the AstraZeneca Tufts Laboratory for Basic and Translational Neuroscience, Tufts University School of Medicine, Boston, Massachusetts 02111
- Neuroscience, Innovative Medicines and Early Development, AstraZeneca, Waltham, Massachusetts 02451
| | - Stephen J Moss
- From the AstraZeneca Tufts Laboratory for Basic and Translational Neuroscience, Tufts University School of Medicine, Boston, Massachusetts 02111,
- Neuroscience, Innovative Medicines and Early Development, AstraZeneca, Waltham, Massachusetts 02451
- the Department of Neuroscience, Physiology and Pharmacology, University College, London WC1E 6BT, United Kingdom
| |
Collapse
|
13
|
Martin GE, Barstein J, Hornickel J, Matherly S, Durante G, Losh M. Signaling of noncomprehension in communication breakdowns in fragile X syndrome, Down syndrome, and autism spectrum disorder. JOURNAL OF COMMUNICATION DISORDERS 2017; 65:22-34. [PMID: 28161297 PMCID: PMC5340195 DOI: 10.1016/j.jcomdis.2017.01.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Revised: 01/13/2017] [Accepted: 01/19/2017] [Indexed: 05/15/2023]
Abstract
UNLABELLED The ability to indicate a failure to understand a message is a critical pragmatic (social) language skill for managing communication breakdowns and supporting successful communicative exchanges. The current study examined the ability to signal noncomprehension across different types of confusing message conditions in children and adolescents with fragile X syndrome (FXS), Down syndrome (DS), autism spectrum disorder (ASD), and typical development (TD). Controlling for nonverbal mental age and receptive vocabulary skills, youth with comorbid FXS and ASD and those with DS were less likely than TD controls to signal noncomprehension of confusing messages. Youth with FXS without ASD and those with idiopathic ASD did not differ from controls. No sex differences were detected in any group. Findings contribute to current knowledge of pragmatic profiles in different forms of genetically-based neurodevelopmental disorders associated with intellectual disability, and the role of sex in the expression of such profiles. LEARNING OUTCOMES Upon completion of this article, readers will have learned about: (1) the social-communicative profiles of youth with FXS, DS, and ASD, (2) the importance of signaling noncomprehension in response to a confusing message, and (3) the similarities and differences in noncomprehension signaling in youth with FXS (with and without ASD), DS, idiopathic ASD, and TD.
Collapse
Affiliation(s)
- Gary E Martin
- Department of Communication Sciences and Disorders, St. John's University, Staten Island, NY, USA.
| | - Jamie Barstein
- Roxelyn and Richard Pepper Department of Communication Sciences and Disorders, Northwestern University, Evanston, IL, USA
| | - Jane Hornickel
- Roxelyn and Richard Pepper Department of Communication Sciences and Disorders, Northwestern University, Evanston, IL, USA
| | - Sara Matherly
- Frank Porter Graham Child Development Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Genna Durante
- Frank Porter Graham Child Development Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Molly Losh
- Roxelyn and Richard Pepper Department of Communication Sciences and Disorders, Northwestern University, Evanston, IL, USA
| |
Collapse
|
14
|
Gomis-González M, Busquets-Garcia A, Matute C, Maldonado R, Mato S, Ozaita A. Possible Therapeutic Doses of Cannabinoid Type 1 Receptor Antagonist Reverses Key Alterations in Fragile X Syndrome Mouse Model. Genes (Basel) 2016; 7:E56. [PMID: 27589806 PMCID: PMC5042387 DOI: 10.3390/genes7090056] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Revised: 08/20/2016] [Accepted: 08/22/2016] [Indexed: 12/29/2022] Open
Abstract
Fragile X syndrome (FXS) is the most common monogenetic cause of intellectual disability. The cognitive deficits in the mouse model for this disorder, the Fragile X Mental Retardation 1 (Fmr1) knockout (KO) mouse, have been restored by different pharmacological approaches, among those the blockade of cannabinoid type 1 (CB1) receptor. In this regard, our previous study showed that the CB1 receptor antagonist/inverse agonist rimonabant normalized a number of core features in the Fmr1 knockout mouse. Rimonabant was commercialized at high doses for its anti-obesity properties, and withdrawn from the market on the bases of mood-related adverse effects. In this study we show, by using electrophysiological approaches, that low dosages of rimonabant (0.1 mg/kg) manage to normalize metabotropic glutamate receptor dependent long-term depression (mGluR-LTD). In addition, low doses of rimonabant (from 0.01 mg/kg) equally normalized the cognitive deficit in the mouse model of FXS. These doses of rimonabant were from 30 to 300 times lower than those required to reduce body weight in rodents and to presumably produce adverse effects in humans. Furthermore, NESS0327, a CB1 receptor neutral antagonist, was also effective in preventing the novel object-recognition memory deficit in Fmr1 KO mice. These data further support targeting CB1 receptors as a relevant therapy for FXS.
Collapse
Affiliation(s)
- Maria Gomis-González
- Laboratory of Neuropharmacology-NeuroPhar, Department of Experimental and Health Sciences, Program of Genetics and Neurosciences, University Pompeu Fabra, Barcelona 08003, Spain.
| | - Arnau Busquets-Garcia
- Laboratory of Neuropharmacology-NeuroPhar, Department of Experimental and Health Sciences, Program of Genetics and Neurosciences, University Pompeu Fabra, Barcelona 08003, Spain.
| | - Carlos Matute
- Department of Neurosciences, University of the Basque Country UPV/EHU, Leioa 48940, Spain.
- Achucarro Basque Center for Neuroscience, Zamudio 48170, Spain.
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid 28031, Spain.
| | - Rafael Maldonado
- Laboratory of Neuropharmacology-NeuroPhar, Department of Experimental and Health Sciences, Program of Genetics and Neurosciences, University Pompeu Fabra, Barcelona 08003, Spain.
| | - Susana Mato
- Department of Neurosciences, University of the Basque Country UPV/EHU, Leioa 48940, Spain.
- Achucarro Basque Center for Neuroscience, Zamudio 48170, Spain.
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid 28031, Spain.
| | - Andrés Ozaita
- Laboratory of Neuropharmacology-NeuroPhar, Department of Experimental and Health Sciences, Program of Genetics and Neurosciences, University Pompeu Fabra, Barcelona 08003, Spain.
| |
Collapse
|
15
|
Wang L, Wang Y, Zhou S, Yang L, Shi Q, Li Y, Zhang K, Yang L, Zhao M, Yang Q. Imbalance between Glutamate and GABA in Fmr1 Knockout Astrocytes Influences Neuronal Development. Genes (Basel) 2016; 7:genes7080045. [PMID: 27517961 PMCID: PMC4999833 DOI: 10.3390/genes7080045] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Revised: 07/16/2016] [Accepted: 07/25/2016] [Indexed: 01/21/2023] Open
Abstract
Fragile X syndrome (FXS) is a form of inherited mental retardation that results from the absence of the fragile X mental retardation protein (FMRP), the product of the Fmr1 gene. Numerous studies have shown that FMRP expression in astrocytes is important in the development of FXS. Although astrocytes affect neuronal dendrite development in Fmr1 knockout (KO) mice, the factors released by astrocytes are still unclear. We cultured wild type (WT) cortical neurons in astrocyte-conditioned medium (ACM) from WT or Fmr1 KO mice. Immunocytochemistry and Western blotting were performed to detect the dendritic growth of both WT and KO neurons. We determined glutamate and γ-aminobutyric acid (GABA) levels using high-performance liquid chromatography (HPLC). The total neuronal dendritic length was reduced when cultured in the Fmr1 KO ACM. This neurotoxicity was triggered by an imbalanced release of glutamate and GABA from Fmr1 KO astrocytes. We found increased glutaminase and GABA transaminase (GABA-T) expression and decreased monoamine oxidase B expression in Fmr1 KO astrocytes. The elevated levels of glutamate contributed to oxidative stress in the cultured neurons. Vigabatrin (VGB), a GABA-T inhibitor, reversed the changes caused by glutamate and GABA release in Fmr1 KO astrocytes and the abnormal behaviors in Fmr1 KO mice. Our results indicate that the imbalance in the astrocytic glutamate and GABA release may be involved in the neuropathology and the underlying symptoms of FXS, and provides a therapeutic target for treatment.
Collapse
Affiliation(s)
- Lu Wang
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, China.
| | - Yan Wang
- Department of Gastroenterology and Endoscopy Center, No. 323 Hospital of PLA, Xi'an 710054, China.
| | - Shimeng Zhou
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, China.
| | - Liukun Yang
- Fifth Company, Second Battalion, Cadet Brigade, Fourth Military Medical University, Xi'an 710032, China.
| | - Qixin Shi
- Fifth Company, Second Battalion, Cadet Brigade, Fourth Military Medical University, Xi'an 710032, China.
| | - Yujiao Li
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, China.
| | - Kun Zhang
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, China.
| | - Le Yang
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, China.
| | - Minggao Zhao
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, China.
| | - Qi Yang
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, China.
| |
Collapse
|
16
|
Sen A, Cingolani P, Senut MC, Land S, Mercado-Garcia A, Tellez-Rojo MM, Baccarelli AA, Wright RO, Ruden DM. Lead exposure induces changes in 5-hydroxymethylcytosine clusters in CpG islands in human embryonic stem cells and umbilical cord blood. Epigenetics 2016; 10:607-21. [PMID: 26046694 DOI: 10.1080/15592294.2015.1050172] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Prenatal exposure to neurotoxicants such as lead (Pb) may cause stable changes in the DNA methylation (5mC) profile of the fetal genome. However, few studies have examined its effect on the DNA de-methylation pathway, specifically the dynamic changes of the 5-hydroxymethylcytosine (5hmC) profile. Therefore, in this study, we investigate the relationship between Pb exposure and 5mC and 5hmC modifications during early development. To study the changes in the 5hmC profile, we use a novel modification of the Infinium™ HumanMethylation450 assay (Illumina, Inc.), which we named HMeDIP-450K assay, in an in vitro human embryonic stem cell model of Pb exposure. We model Pb exposure-associated 5hmC changes as clusters of correlated, adjacent CpG sites, which are co-responding to Pb. We further extend our study to look at Pb-dependent changes in high density 5hmC regions in umbilical cord blood DNA from 48 mother-infant pairs from the Early Life Exposure in Mexico to Environmental Toxicants (ELEMENT) cohort. For our study, we randomly selected umbilical cord blood from 24 male and 24 female children from the 1st and 4th quartiles of Pb levels. Our data show that Pb-associated changes in the 5hmC and 5mC profiles can be divided into sex-dependent and sex-independent categories. Interestingly, differential 5mC sites are better markers of Pb-associated sex-dependent changes compared to differential 5hmC sites. In this study we identified several 5hmC and 5mC genomic loci, which we believe might have some potential as early biomarkers of prenatal Pb exposure.
Collapse
Key Words
- 5-hydroxymethylcytosine
- 5-methylcytosine
- A-clustering approach for grouping HM450K CpG sites into clusters based on correlations
- Aclust
- CpG island, CpG islands are usually in the promoter and 5' untranslated regions of genes
- CpG islands
- CpG, sites, CG dinucleotide methylation site in the genome corresponding with probes on HM450K
- DMRs, Differentially methylated regions (clusters) identified by GEE
- DhMRs, Differentially Hydroxymethylated regions (clusters) identified by GEE
- ELEMENT, Early Life Exposure in Mexico to Environmental Toxicants
- GEE, Generalized Estimating Equations, used to measure the estimated change in DNA methylation in co-regulated regions of the genome
- HM450K
- HM450K, Infinium Human Methylation 450K BeadChip array from Illumina
- HMeDIP-450K
- HMeDIP-450K, Infinium Human Methylation 450K BeadChip array from Illumina coupled with immunoprecipitation with 5hmC antibodies
- LINE-1, Long Interspersed Element 1, a common retrotransposon in humans
- MRs, Methylation regions (clusters) identified by A-clustering
- clusters
- hMRs, Hydroxymethylated regions (clusters) identified by A-clustering
- lead
- umbilical cord blood
Collapse
Affiliation(s)
- Arko Sen
- a Institute of Environmental Health Sciences; Wayne State University ; Detroit , MI , USA
| | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Senut MC, Zhang Y, Liu F, Sen A, Ruden DM, Mao G. Size-Dependent Toxicity of Gold Nanoparticles on Human Embryonic Stem Cells and Their Neural Derivatives. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2016; 12:631-46. [PMID: 26676601 PMCID: PMC5033512 DOI: 10.1002/smll.201502346] [Citation(s) in RCA: 97] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Revised: 10/08/2015] [Indexed: 05/17/2023]
Abstract
This study explores the use of human embryonic stem cells (hESCs) for assessing nanotoxicology, specifically, the effect of gold nanoparticles (AuNPs) of different core sizes (1.5, 4, and 14 nm) on the viability, pluripotency, neuronal differentiation, and DNA methylation of hESCs. The hESCs exposed to 1.5 nm thiolate-capped AuNPs exhibit loss of cohesiveness and detachment suggesting ongoing cell death at concentrations as low as 0.1 μg mL(-1). The cells exposed to 1.5 nm AuNPs at this concentration do not form embryoid bodies but rather disintegrate into single cells within 48 h. Cell death caused by 1.5 nm AuNPs also occur in hESC-derived neural progenitor cells. None of the other nanoparticles exhibit toxic effects on the hESCs at concentrations as high as 10 μg mL(-1) during a 19 d neural differentiation period. Thiolate-capped 4 nm AuNPs at 10 μg mL(-1) cause a dramatic decrease in global DNA methylation (5 mC) and a corresponding increase in global DNA hydroxymethylation (5 hmC) of the hESC's DNA in only 24 h. This work identifies a type of AuNPs highly toxic to hESCs and demonstrates the potential of hESCs in predicting nanotoxicity and characterizing their ability to alter the DNA methylation and hydroxymethylation patterns in the cells.
Collapse
Affiliation(s)
- Marie-Claude Senut
- Institute of Environmental Health and Sciences, Wayne State University, 275 East Hancock Road, Detroit, Michigan 48201, USA
| | - Yanhua Zhang
- Department of Chemical Engineering and Materials Science, Wayne State University, 5050 Anthony Wayne Drive, Detroit, Michigan 48202, USA
| | - Fangchao Liu
- Department of Chemical Engineering and Materials Science, Wayne State University, 5050 Anthony Wayne Drive, Detroit, Michigan 48202, USA
| | - Arko Sen
- Institute of Environmental Health and Sciences, Wayne State University, 275 East Hancock Road, Detroit, Michigan 48201, USA
| | - Douglas M. Ruden
- Institute of Environmental Health and Sciences, Wayne State University, 275 East Hancock Road, Detroit, Michigan 48201, USA
| | - Guangzhao Mao
- Department of Chemical Engineering and Materials Science, Wayne State University, 5050 Anthony Wayne Drive, Detroit, Michigan 48202, USA
| |
Collapse
|
18
|
Srivastava A, Srivastava N, Mittal B. Genetics of Obesity. Indian J Clin Biochem 2015; 31:361-71. [PMID: 27605733 DOI: 10.1007/s12291-015-0541-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Accepted: 12/08/2015] [Indexed: 12/29/2022]
Abstract
Numerous classical genetic studies have proved that genes are contributory factors for obesity. Genes are directly responsible for obesity associated disorders such as Bardet-Biedl and Prader-Willi syndromes. However, both genes as well as environment are associated with obesity in the general population. Genetic epidemiological approaches, particularly genome-wide association studies, have unraveled many genes which play important roles in human obesity. Elucidation of their biological functions can be very useful for understanding pathobiology of obesity. In the near future, further exploration of obesity genetics may help to develop useful diagnostic and predictive tests for obesity treatment.
Collapse
Affiliation(s)
- Apurva Srivastava
- Department of Medical Genetics, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Rae Bareli Road, Lucknow, Uttar Pradesh 226014 India ; Department of Physiology, King George's Medical University, Chowk, Lucknow, Uttar Pradesh 226003 India
| | - Neena Srivastava
- Department of Physiology, King George's Medical University, Chowk, Lucknow, Uttar Pradesh 226003 India
| | - Balraj Mittal
- Department of Medical Genetics, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Rae Bareli Road, Lucknow, Uttar Pradesh 226014 India
| |
Collapse
|
19
|
Zhang Y, Gaetano CM, Williams KR, Bassell GJ, Mihailescu MR. FMRP interacts with G-quadruplex structures in the 3'-UTR of its dendritic target Shank1 mRNA. RNA Biol 2015; 11:1364-74. [PMID: 25692235 DOI: 10.1080/15476286.2014.996464] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
Abstract
Fragile X syndrome (FXS), the most common cause of inherited intellectual disability, is caused by the loss of expression of the fragile X mental retardation protein (FMRP). FMRP, which regulates the transport and translation of specific mRNAs, uses its RGG box domain to bind mRNA targets that form G-quadruplex structures. One of the FMRP in vivo targets, Shank1 mRNA, encodes the master scaffold proteins of the postsynaptic density (PSD) which regulate the size and shape of dendritic spines because of their capacity to interact with many different PSD components. Due to their effect on spine morphology, altered translational regulation of Shank1 transcripts may contribute to the FXS pathology. We hypothesized that the FMRP interactions with Shank1 mRNA are mediated by the recognition of the G quadruplex structure, which has not been previously demonstrated. In this study we used biophysical techniques to analyze the Shank1 mRNA 3'-UTR and its interactions with FMRP and its phosphorylated mimic FMRP S500D. We found that the Shank1 mRNA 3 ' -UTR adopts two very stable intramolecular G-quadruplexes which are bound specifically and with high affinity by FMRP both in vitro and in vivo. These results suggest a role of G-quadruplex RNA motif as a structural element in the common mechanism of FMRP regulation of its dendritic mRNA targets.
Collapse
Affiliation(s)
- Yang Zhang
- a Graduate School of Pharmaceutical Sciences; Mylan School of Pharmacy ; Duquesne University ; Pittsburgh , PA USA
| | | | | | | | | |
Collapse
|
20
|
Sen A, Heredia N, Senut MC, Land S, Hollocher K, Lu X, Dereski MO, Ruden DM. Multigenerational epigenetic inheritance in humans: DNA methylation changes associated with maternal exposure to lead can be transmitted to the grandchildren. Sci Rep 2015; 5:14466. [PMID: 26417717 PMCID: PMC4586440 DOI: 10.1038/srep14466] [Citation(s) in RCA: 110] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Accepted: 07/30/2015] [Indexed: 12/26/2022] Open
Abstract
We report that the DNA methylation profile of a child’s neonatal whole blood can be significantly influenced by his or her mother’s neonatal blood lead levels (BLL). We recruited 35 mother-infant pairs in Detroit and measured the whole blood lead (Pb) levels and DNA methylation levels at over 450,000 loci from current blood and neonatal blood from both the mother and the child. We found that mothers with high neonatal BLL correlate with altered DNA methylation at 564 loci in their children’s neonatal blood. Our results suggest that Pb exposure during pregnancy affects the DNA methylation status of the fetal germ cells, which leads to altered DNA methylation in grandchildren’s neonatal dried blood spots. This is the first demonstration that an environmental exposure in pregnant mothers can have an epigenetic effect on the DNA methylation pattern in the grandchildren.
Collapse
Affiliation(s)
- Arko Sen
- Institute of Environmental Health Sciences, Wayne State University, Detroit, MI 48201.,Department of Pharmacology, Wayne State University, Detroit, MI 48201
| | - Nicole Heredia
- Institute of Environmental Health Sciences, Wayne State University, Detroit, MI 48201
| | - Marie-Claude Senut
- Institute of Environmental Health Sciences, Wayne State University, Detroit, MI 48201
| | - Susan Land
- C. S. Mott Centre for Human Growth and Development, Wayne State University, Detroit, MI 48201.,Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI 48201
| | | | - Xiangyi Lu
- Institute of Environmental Health Sciences, Wayne State University, Detroit, MI 48201
| | - Mary O Dereski
- Department of Biomedical Sciences, Oakland University William Beaumont School of Medicine, Rochester, MI 48309
| | - Douglas M Ruden
- Institute of Environmental Health Sciences, Wayne State University, Detroit, MI 48201.,C. S. Mott Centre for Human Growth and Development, Wayne State University, Detroit, MI 48201.,Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI 48201
| |
Collapse
|
21
|
Sen A, Heredia N, Senut MC, Hess M, Land S, Qu W, Hollacher K, Dereski MO, Ruden DM. Early life lead exposure causes gender-specific changes in the DNA methylation profile of DNA extracted from dried blood spots. Epigenomics 2015; 7:379-93. [PMID: 26077427 DOI: 10.2217/epi.15.2] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
AIMS In this paper, we tested the hypothesis that early life lead (Pb) exposure associated DNA methylation (5 mC) changes are dependent on the sex of the child and can serve as biomarkers for Pb exposure. METHODS In this pilot study, we measured the 5mC profiles of DNA extracted from dried blood spots (DBS) in a cohort of 43 children (25 males and 18 females; ages from 3 months to 5 years) from Detroit. Result & Discussion: We found that the effect of Pb-exposure on the 5-mC profiles can be separated into three subtypes: affected methylation loci which are conserved irrespective of the sex of the child (conserved); affected methylation loci unique to males (male-specific); and affected methylation loci unique to females (female-specific).
Collapse
Affiliation(s)
- Arko Sen
- Institute of Environmental Health Sciences, Wayne State University, 2727 Second Avenue, Detroit, MI 48201, USA.,Department of Pharmacology, Wayne State University, Room 4000, 2727 Second Avenue, Detroit, MI 48201, USA
| | - Nicole Heredia
- Department of Obstetrics & Gynecology, Wayne State University, 3750 Woodward Avenue, Detroit, MI 48201, USA
| | - Marie-Claude Senut
- Institute of Environmental Health Sciences, Wayne State University, 2727 Second Avenue, Detroit, MI 48201, USA
| | - Matthew Hess
- Department of Obstetrics & Gynecology, Wayne State University, 3750 Woodward Avenue, Detroit, MI 48201, USA.,CS Mott Center for Human Growth & Development, Wayne State University, 275 E Hancock St, Detroit, MI 48201, USA
| | - Susan Land
- Department of Obstetrics & Gynecology, Wayne State University, 3750 Woodward Avenue, Detroit, MI 48201, USA
| | - Wen Qu
- Department of Pharmacology, Wayne State University, Room 4000, 2727 Second Avenue, Detroit, MI 48201, USA
| | | | - Mary O Dereski
- Department of Obstetrics & Gynecology, Wayne State University, 3750 Woodward Avenue, Detroit, MI 48201, USA
| | - Douglas M Ruden
- Institute of Environmental Health Sciences, Wayne State University, 2727 Second Avenue, Detroit, MI 48201, USA.,Union College, 807 Union St, Schenectady, NY 12308, USA
| |
Collapse
|
22
|
Tejada-Simon MV. Modulation of actin dynamics by Rac1 to target cognitive function. J Neurochem 2015; 133:767-79. [PMID: 25818528 DOI: 10.1111/jnc.13100] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Revised: 03/11/2015] [Accepted: 03/14/2015] [Indexed: 12/14/2022]
Abstract
The small GTPase Rac1 is well known for regulating actin cytoskeleton reorganization in cells. Formation of extensions at the surface of the cell is required for migration and even for cell invasion and metastases. Because an elevated level and hyperactivation of this protein has been associated with metastasis in cancer, direct regulators of Rac1 are currently envisioned as a potential strategy to treat certain cancers. Less research, however, has been done regarding the role of this small GTP-binding protein in brain development, where it has an important role in dendritic spine morphogenesis through the regulation of actin. Alteration of dendritic development and spinogenesis has been often associated with mental disorders. Rac1 is associated with and required for learning and the formation of memories in the brain. Rac1 appears to be dysregulated in certain neurodevelopmental disorders that present all these three alterations: mental retardation, atypical synaptic plasticity and aberrant spine morphology. Thus, to develop novel therapies for rescuing cognitive impairment, a reasonable approach might be to target this protein, Rac1, which plays a pivotal role in directing signals that regulate actin dynamics, which in turn might have an effect in spine cytoarchitecture and synaptic function. It is possible that novel drugs that regulate Rac1 activation and function could modulate actin cytoskeleton and spine dynamics, representing potential candidates to repair intellectual disability in disorders associated with spine abnormalities. Herein, we present a list of the current Rac1 inhibitors that might fulfill this role together with a summary of the latest findings concerning their function as they relate to neuronal studies. While the small GTPase Rac1 is well known for regulating actin cytoskeleton reorganization in different type of cells, it appears to be also required for learning and the formation of memories in the brain. Abnormal regulation of this protein has been associated with cognitive disabilities, atypical synaptic plasticity and abnormal morphology of dendritic spines in certain neurodevelopmental disorders. Thus, modulation of Rac1 activity using novel inhibitors might be a strategy to reestablish cognitive function.
Collapse
Affiliation(s)
- Maria V Tejada-Simon
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston, Houston, Texas, USA.,Department of Biology, University of Houston, Houston, Texas, USA.,Department of Psychology, University of Houston, Houston, Texas, USA.,Biology of Behavior Institute (BoBI), University of Houston, Houston, Texas, USA
| |
Collapse
|
23
|
Moskowitz LJ, Jones EA. Uncovering the evidence for behavioral interventions with individuals with fragile X syndrome: a systematic review. RESEARCH IN DEVELOPMENTAL DISABILITIES 2015; 38:223-241. [PMID: 25575286 DOI: 10.1016/j.ridd.2014.12.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Revised: 12/09/2014] [Accepted: 12/09/2014] [Indexed: 06/04/2023]
Abstract
Fragile X syndrome (FXS) is associated with a wide range of cognitive, emotional, and behavioral difficulties. Although there is considerable research on the behavioral phenotype of FXS, few empirical studies of behavioral interventions with this population have been identified. Through a hand search of 34 behavioral journals, we examined the evidence base for behavioral interventions with individuals with FXS and in light of the current state of knowledge regarding the FXS behavioral phenotype. Systematic review procedures were used to identify behavioral intervention studies that included at least one participant with FXS, extract and summarize the data on several relevant dimensions, and rate the methodological quality of the studies. Results revealed 31 intervention studies with a small number of participants with FXS. Overall, results suggest a behavioral approach to intervention with individuals with FXS shows promise. Future research focused on individuals with FXS will be necessary to continue to examine differences in response to intervention and interventions that specifically address phenotypic characteristics.
Collapse
Affiliation(s)
- Lauren J Moskowitz
- Department of Psychology, St John's University, Queens, NY 11439, United States.
| | - Emily A Jones
- Queens College, The Graduate Center of the City University of New York, Queens, NY, United States
| |
Collapse
|
24
|
Yang S, Yang S, Park JS, Kirkwood A, Bao S. Failed stabilization for long-term potentiation in the auditory cortex of FMR1 knockout mice. PLoS One 2014; 9:e104691. [PMID: 25115962 PMCID: PMC4130563 DOI: 10.1371/journal.pone.0104691] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2014] [Accepted: 07/10/2014] [Indexed: 01/27/2023] Open
Abstract
Fragile X syndrome is a developmental disorder that affects sensory systems. A null mutation of the Fragile X Mental Retardation protein 1 (Fmr1) gene in mice has varied effects on developmental plasticity in different sensory systems, including normal barrel cortical plasticity, altered ocular dominance plasticity and grossly impaired auditory frequency map plasticity. The mutation also has different effects on long-term synaptic plasticity in somatosensory and visual cortical neurons, providing insights on how it may differentially affect the sensory systems. Here we present evidence that long-term potentiation (LTP) is impaired in the developing auditory cortex of the Fmr1 knockout (KO) mice. This impairment of synaptic plasticity is consistent with impaired frequency map plasticity in the Fmr1 KO mouse. Together, these results suggest a potential role of LTP in sensory map plasticity during early sensory development.
Collapse
Affiliation(s)
- Sungchil Yang
- Helen Wills Neuroscience Institute, University of California, Berkeley, California, United States of America
| | - Sunggu Yang
- Department of Neuroscience, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Jae-Sung Park
- Department of Neuroscience, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Alfredo Kirkwood
- Department of Neuroscience, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Shaowen Bao
- Helen Wills Neuroscience Institute, University of California, Berkeley, California, United States of America
- * E-mail:
| |
Collapse
|
25
|
Loss of functional A-type potassium channels in the dendrites of CA1 pyramidal neurons from a mouse model of fragile X syndrome. J Neurosci 2014; 33:19442-50. [PMID: 24336711 DOI: 10.1523/jneurosci.3256-13.2013] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Despite the critical importance of voltage-gated ion channels in neurons, very little is known about their functional properties in Fragile X syndrome: the most common form of inherited cognitive impairment. Using three complementary approaches, we investigated the physiological role of A-type K(+) currents (I(KA)) in hippocampal CA1 pyramidal neurons from fmr1-/y mice. Direct measurement of I(KA) using cell-attached patch-clamp recordings revealed that there was significantly less I(KA) in the dendrites of CA1 neurons from fmr1-/y mice. Interestingly, the midpoint of activation for A-type K(+) channels was hyperpolarized for fmr1-/y neurons compared with wild-type, which might partially compensate for the lower current density. Because of the rapid time course for recovery from steady-state inactivation, the dendritic A-type K(+) current in CA1 neurons from both wild-type and fmr1-/y mice is likely mediated by K(V)4 containing channels. The net effect of the differences in I(KA) was that back-propagating action potentials had larger amplitudes producing greater calcium influx in the distal dendrites of fmr1-/y neurons. Furthermore, CA1 pyramidal neurons from fmr1-/y mice had a lower threshold for LTP induction. These data suggest that loss of I(KA) in hippocampal neurons may contribute to dendritic pathophysiology in Fragile X syndrome.
Collapse
|
26
|
Zhuo M. Long-term potentiation in the anterior cingulate cortex and chronic pain. Philos Trans R Soc Lond B Biol Sci 2013; 369:20130146. [PMID: 24298148 PMCID: PMC3843878 DOI: 10.1098/rstb.2013.0146] [Citation(s) in RCA: 130] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Glutamate is the primary excitatory transmitter of sensory transmission and perception in the central nervous system. Painful or noxious stimuli from the periphery ‘teach’ humans and animals to avoid potentially dangerous objects or environments, whereas tissue injury itself causes unnecessary chronic pain that can even last for long periods of time. Conventional pain medicines often fail to control chronic pain. Recent neurobiological studies suggest that synaptic plasticity taking place in sensory pathways, from spinal dorsal horn to cortical areas, contributes to chronic pain. Injuries trigger long-term potentiation of synaptic transmission in the spinal cord dorsal horn and anterior cingulate cortex, and such persistent potentiation does not require continuous neuronal activity from the periphery. At the synaptic level, potentiation of excitatory transmission caused by injuries may be mediated by the enhancement of glutamate release from presynaptic terminals and potentiated postsynaptic responses of AMPA receptors. Preventing, ‘erasing’ or reducing such potentiation may serve as a new mechanism to inhibit chronic pain in patients in the future.
Collapse
Affiliation(s)
- Min Zhuo
- Center for Neuron and Disease, Frontier Institutes of Life Science, Science and Technology, Xi'an Jiaotong University, , Xi'an 710049, People's Republic of China
| |
Collapse
|
27
|
Abstract
Fragile X syndrome, the most common form of heritable mental retardation, is a developmental disorder with known effects within sensory systems. Altered developmental plasticity has been reported in the visual and somatosensory systems in Fmr1 knock-out (KO) mice. Behavioral studies have revealed maladaptive auditory responses in fragile X syndrome patients and Fmr1 KO mice, suggesting that adaptive plasticity may also be impaired in the auditory system. Here we show that, whereas tonotopic frequency representation develops normally in Fmr1 KO mice, developmental plasticity in primary auditory cortex is grossly impaired. This deficit can be rescued by pharmacological blockade of mGluR5 receptors. These results support the mGluR hypothesis of fragile X mental retardation and suggest that deficient developmental plasticity may contribute to maladaptive auditory processing in fragile X syndrome.
Collapse
|
28
|
Yang Q, Feng B, Zhang K, Guo YY, Liu SB, Wu YM, Li XQ, Zhao MG. Excessive astrocyte-derived neurotrophin-3 contributes to the abnormal neuronal dendritic development in a mouse model of fragile X syndrome. PLoS Genet 2012; 8:e1003172. [PMID: 23300470 PMCID: PMC3531466 DOI: 10.1371/journal.pgen.1003172] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2012] [Accepted: 10/31/2012] [Indexed: 11/25/2022] Open
Abstract
Fragile X syndrome (FXS) is a form of inherited mental retardation in humans that results from expansion of a CGG repeat in the Fmr1 gene. Recent studies suggest a role of astrocytes in neuronal development. However, the mechanisms involved in the regulation process of astrocytes from FXS remain unclear. In this study, we found that astrocytes derived from a Fragile X model, the Fmr1 knockout (KO) mouse which lacks FMRP expression, inhibited the proper elaboration of dendritic processes of neurons in vitro. Furthermore, astrocytic conditioned medium (ACM) from KO astrocytes inhibited proper dendritic growth of both wild-type (WT) and KO neurons. Inducing expression of FMRP by transfection of FMRP vectors in KO astrocytes restored dendritic morphology and levels of synaptic proteins. Further experiments revealed elevated levels of the neurotrophin-3 (NT-3) in KO ACM and the prefrontal cortex of Fmr1 KO mice. However, the levels of nerve growth factor (NGF), brain-derived neurotrophic factor (BDNF), glial cell-derived neurotrophic factor (GDNF), and ciliary neurotrophic factor (CNTF) were normal. FMRP has multiple RNA–binding motifs and is involved in translational regulation. RNA–binding protein immunoprecipitation (RIP) showed the NT-3 mRNA interacted with FMRP in WT astrocytes. Addition of high concentrations of exogenous NT-3 to culture medium reduced the dendrites of neurons and synaptic protein levels, whereas these measures were ameliorated by neutralizing antibody to NT-3 or knockdown of NT-3 expression in KO astrocytes through short hairpin RNAs (shRNAs). Prefrontal cortex microinjection of WT astrocytes or NT-3 shRNA infected KO astrocytes rescued the deficit of trace fear memory in KO mice, concomitantly decreased the NT-3 levels in the prefrontal cortex. This study indicates that excessive NT-3 from astrocytes contributes to the abnormal neuronal dendritic development and that astrocytes could be a potential therapeutic target for FXS. Fragile X syndrome is a form of inherited mental retardation in humans that results from expansion of a CGG repeat in the Fmr1 gene. Recent studies suggest that astrocytes play a role in neuronal growth. In this study, we find that astrocytes derived from a Fragile X model, the Fmr1 knockout (KO) mouse, inhibit the proper elaboration of dendritic processes of neurons in vitro. Excessive neurotrophin-3 (NT-3) is released in the astrocytes from Fmr1 KO mice. Blockage of NT-3 by neutralizing antibodies and knockdown of NT-3 by using short hairpin RNAs (shRNAs) in Fmr1 KO astrocytes can rescue the neuronal dendritic development. In vivo experiments show that prefrontal cortex microinjection of WT astrocytes or NT-3 shRNA–infected KO astrocytes rescues the deficit of trace fear memory in KO mice. This study provides the evidence that a lack of FMRP leads to an overexpression of NT-3, which reduces dendritic growth in neurons.
Collapse
Affiliation(s)
- Qi Yang
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, China
| | - Bin Feng
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, China
| | - Kun Zhang
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, China
| | - Yan-yan Guo
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, China
| | - Shui-bing Liu
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, China
| | - Yu-mei Wu
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, China
| | - Xiao-qiang Li
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, China
| | - Ming-gao Zhao
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, China
- * E-mail:
| |
Collapse
|
29
|
Sansone SM, Widaman KF, Hall SS, Reiss AL, Lightbody A, Kaufmann WE, Berry-Kravis E, Lachiewicz A, Brown EC, Hessl D. Psychometric study of the Aberrant Behavior Checklist in Fragile X Syndrome and implications for targeted treatment. J Autism Dev Disord 2012; 42:1377-92. [PMID: 21972117 DOI: 10.1007/s10803-011-1370-2] [Citation(s) in RCA: 123] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Animal studies elucidating the neurobiology of fragile X syndrome (FXS) have led to multiple controlled trials in humans, with the Aberrant Behavior Checklist-Community (ABC-C) commonly adopted as a primary outcome measure. A multi-site collaboration examined the psychometric properties of the ABC-C in 630 individuals (ages 3-25) with FXS using exploratory and confirmatory factor analysis. Results support a six-factor structure, with one factor unchanged (Inappropriate Speech), four modified (Irritability, Hyperactivity, Lethargy/Withdrawal, and Stereotypy), and a new Social Avoidance factor. A comparison with ABC-C data from individuals with general intellectual disability and a list of commonly endorsed items are also reported. Reformulated ABC-C scores based on this FXS-specific factor structure may provide added outcome measure specificity and sensitivity in FXS clinical trials.
Collapse
Affiliation(s)
- Stephanie M Sansone
- Medical Investigation of Neurodevelopmental Disorders Institute, University of California Davis Medical Center, 2825 50th Street, Sacramento, CA 95817, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Losh M, Martin GE, Klusek J, Hogan-Brown AL, Sideris J. Social communication and theory of mind in boys with autism and fragile x syndrome. Front Psychol 2012; 3:266. [PMID: 22934085 PMCID: PMC3422728 DOI: 10.3389/fpsyg.2012.00266] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2012] [Accepted: 07/09/2012] [Indexed: 11/13/2022] Open
Abstract
Impairments in the social use of language, or pragmatics, constitute a core characteristic of autism. Problems with pragmatic language have also been documented in fragile X syndrome (FXS), a monogenic condition that is the most common known genetic cause of autism. Evidence suggests that social cognitive ability, or theory of mind, may also be impaired in both conditions, and in autism, may importantly relate to pragmatic language ability. Given the substantial overlap observed in autism and FXS, this study aimed to better define those social-communicative phenotypes that overlap in these two conditions by comparing pragmatic language ability and theory of mind in children with idiopathic autism and children with FXS, with and without autism, as well as children with Down syndrome and typically developing controls. We further examined correlations between these cognitive-behavioral phenotypes and molecular genetic variation related to the Fragile X Mental Retardation-1 gene (FMR1) in the FXS group. Results indicated that children with idiopathic autism and those with FXS and autism performed comparably on direct-assessment measures of pragmatic language and theory of mind, whereas those with FXS only did not differ from controls. Theory of mind was related to pragmatic language ability in all groups. Pragmatic language and theory of mind also correlated with genetic variation at the FMR1 locus (Cytosine-Guanine-Guanine repeats and percent methylation). These results point toward substantial overlap in the social and language phenotypes in autism and FXS and suggest a molecular genetic basis to these phenotypic profiles.
Collapse
Affiliation(s)
- Molly Losh
- Roxelyn and Richard Pepper Department of Communication Sciences and Disorders, Northwestern University Evanston, IL, USA
| | | | | | | | | |
Collapse
|
31
|
Wang H, Morishita Y, Miura D, Naranjo JR, Kida S, Zhuo M. Roles of CREB in the regulation of FMRP by group I metabotropic glutamate receptors in cingulate cortex. Mol Brain 2012; 5:27. [PMID: 22867433 PMCID: PMC3478997 DOI: 10.1186/1756-6606-5-27] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2012] [Accepted: 08/02/2012] [Indexed: 12/04/2022] Open
Abstract
Background Fragile X syndrome is caused by lack of fragile X mental retardation protein (FMRP) due to silencing of the FMR1 gene. The metabotropic glutamate receptors (mGluRs) in the central nervous system contribute to higher brain functions including learning/memory, mental disorders and persistent pain. The transcription factor cyclic AMP-responsive element binding protein (CREB) is involved in important neuronal functions, such as synaptic plasticity and neuronal survival. Our recent study has shown that stimulation of Group I mGluRs upregulated FMRP and activated CREB in anterior cingulate cortex (ACC), a key region for brain cognitive and executive functions, suggesting that activation of Group I mGluRs may upregulate FMRP through CREB signaling pathway. Results In this study, we demonstrate that CREB contributes to the regulation of FMRP by Group I mGluRs. In ACC neurons of adult mice overexpressing dominant active CREB mutant, the upregulation of FMRP by stimulating Group I mGluR is enhanced compared to wild-type mice. However, the regulation of FMRP by Group I mGluRs is not altered by overexpression of Ca2+-insensitive mutant form of downstream regulatory element antagonist modulator (DREAM), a transcriptional repressor involved in synaptic transmission and plasticity. Conclusion Our study has provided further evidence for CREB involvement in regulation of FMRP by Group I mGluRs in ACC neurons, and may help to elucidate the pathogenesis of fragile X syndrome.
Collapse
Affiliation(s)
- Hansen Wang
- Department of Physiology, Faculty of Medicine, University of Toronto, 1 King's College Circle, Toronto, ON, M5S 1A8, Canada
| | | | | | | | | | | |
Collapse
|
32
|
Xu ZH, Yang Q, Feng B, Liu SB, Zhang N, Xing JH, Li XQ, Wu YM, Gao GD, Zhao MG. Group I mGluR antagonist rescues the deficit of D1-induced LTP in a mouse model of fragile X syndrome. Mol Neurodegener 2012; 7:24. [PMID: 22640474 PMCID: PMC3467183 DOI: 10.1186/1750-1326-7-24] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2011] [Accepted: 04/17/2012] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Fragile X syndrome (FXS) is caused by the absence of the mRNA-binding protein Fragile X mental retardation protein (FMRP), encoded by the Fmr1 gene. Overactive signaling by group 1 metabotropic glutamate receptor (Grp1 mGluR) could contribute to slowed synaptic development and other symptoms of FXS. Our previous study has identified that facilitation of synaptic long-term potentiation (LTP) by D1 receptor is impaired in Fmr1 knockout (KO) mice. However, the contribution of Grp1 mGluR to the facilitation of synaptic plasticity by D1 receptor stimulation in the prefrontal cortex has been less extensively studied. RESULTS Here we demonstrated that DL-AP3, a Grp1 mGluR antagonist, rescued LTP facilitation by D1 receptor agonist SKF81297 in Fmr1KO mice. Grp1 mGluR inhibition restored the GluR1-subtype AMPA receptors surface insertion by D1 activation in the cultured Fmr1KO neurons. Simultaneous treatment of Grp1 mGluR antagonist with D1 agonist recovered the D1 receptor signaling by reversing the subcellular redistribution of G protein-coupled receptor kinase 2 (GRK2) in the Fmr1KO neurons. Treatment of SKF81297 alone failed to increase the phosphorylation of NR2B-containing N-methyl D-aspartate receptors (NMDARs) at Tyr-1472 (p-NR2B-Tyr1472) in the cultures from KO mice. However, simultaneous treatment of DL-AP3 could rescue the level of p-NR2B-Tyr1472 by SKF81297 in the cultures from KO mice. Furthermore, behavioral tests indicated that simultaneous treatment of Grp1 mGluR antagonist with D1 agonist inhibited hyperactivity and improved the learning ability in the Fmr1KO mice. CONCLUSION The findings demonstrate that mGluR1 inhibition is a useful strategy to recover D1 receptor signaling in the Fmr1KO mice, and combination of Grp1 mGluR antagonist and D1 agonist is a potential drug therapy for the FXS.
Collapse
Affiliation(s)
- Zhao-Hui Xu
- Department of Neurosurgery, Tangdu Hospital, Fourth Military Medical University, 17 Changle West Road, Xi'an, 710032, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
|
34
|
Deshpande PS, Coffey DBJ. Fragile X syndrome and attention-deficit/hyperactivity disorder symptoms. J Child Adolesc Psychopharmacol 2011; 21:639-42. [PMID: 22196318 PMCID: PMC3279710 DOI: 10.1089/cap.2011.2164] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Affiliation(s)
- Presenter: Swapna Deshpande
- Children's Hospital, Department of Psychiatry and Behavioral Services, University of Oklahoma Health Sciences Center, Oklahoma City
| | - Discussant: Barbara J. Coffey
- New York University Child Study Center, New York, N.Y. and Nathan S. Kline Institute for Psychiatric Research, Orangeburg, N.Y
| |
Collapse
|
35
|
Guo W, Murthy AC, Zhang L, Johnson EB, Schaller EG, Allan AM, Zhao X. Inhibition of GSK3β improves hippocampus-dependent learning and rescues neurogenesis in a mouse model of fragile X syndrome. Hum Mol Genet 2011; 21:681-91. [PMID: 22048960 DOI: 10.1093/hmg/ddr501] [Citation(s) in RCA: 126] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Fragile X syndrome (FXS), a common inherited form of intellectual disability with learning deficits, results from a loss of fragile X mental retardation protein (FMRP). Despite extensive research, treatment options for FXS remain limited. Since FMRP is known to play an important role in adult hippocampal neurogenesis and hippocampus-dependent learning and FMRP regulates the adult neural stem cell fate through the translational regulation of glycogen synthase kinase 3β (GSK3β), we investigated the effects of a GSK3β inhibitor, SB216763, on Fmr1 knockout mice (Fmr1 KO). We found that the inhibition of GSK3β could reverse the hippocampus-dependent learning deficits and rescue adult hippocampal neurogenesis at multiple stages in Fmr1 KO mice. Our results point to GSK3β inhibition as a potential treatment for the learning deficits seen in FXS.
Collapse
Affiliation(s)
- Weixiang Guo
- Waisman Center and Department of Neuroscience, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI 53705, USA
| | | | | | | | | | | | | |
Collapse
|
36
|
Moskowitz LJ, Carr EG, Durand VM. Behavioral intervention for problem behavior in children with fragile X syndrome. AMERICAN JOURNAL ON INTELLECTUAL AND DEVELOPMENTAL DISABILITIES 2011; 116:457-478. [PMID: 22126659 DOI: 10.1352/1944-7558-116.6.457] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
Parents and professionals typically report problem behavior as a significant concern for children with fragile X syndrome. In the present study, the authors explored whether behaviorally based interventions would result in a reduction in problem behavior and an improvement in quality of life for 3 children with fragile X syndrome and their families. A multiple baseline design was used to demonstrate intervention effects for specific high-priority contexts (i.e., bedtime, running errands, and toileting). A multicomponent intervention plan was developed to teach the parents and child to effectively cope with the particular context. After intervention, there were substantial improvements in problem behavior and family quality of life within the given contexts. Results of this study demonstrated the effectiveness of behavioral intervention for children with fragile X syndrome.
Collapse
|
37
|
Roy S, Zhao Y, Allensworth M, Farook MF, LeDoux MS, Reiter LT, Heck DH. Comprehensive motor testing in Fmr1-KO mice exposes temporal defects in oromotor coordination. Behav Neurosci 2011; 125:962-9. [PMID: 22004265 DOI: 10.1037/a0025920] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Fragile X syndrome (FXS; MIM #300624), a well-recognized form of inherited human mental retardation is caused, in most cases, by a CGG trinucleotide repeat expansion in the 5'-untranslated region of FMR1, resulting in reduced expression of the fragile X mental retardation protein (FMRP). Clinical features include macroorchidism, anxiety, mental retardation, motor coordination, and speech articulation deficits. The Fmr1 knockout (Fmr1-KO) mouse, a mouse model for FXS, has been shown to replicate the macroorchidism, cognitive deficits, and neuroanatomical abnormalities found in human FXS. Here we asked whether Fmr1-KO mice also display appendicular and oromotor deficits comparable to the ataxia and dysarthric speech seen in FXS patients. We employed standard motor tests for balance and appendicular motor coordination, and used a novel long-term fluid-licking assay to investigate oromotor function in Fmr1-KO mice and their wild-type (WT) littermates. Fmr1-KO mice performed equally well as their WT littermates on standard motor tests, with the exception of a raised-beam task. However, Fmr1-KO mice had a significantly slower licking rhythm than their WT littermates. Deficits in rhythmic fluid-licking in Fmr1-KO mice have been linked to cerebellar pathologies. It is believed that balance and motor coordination deficits in FXS patients are caused by cerebellar neurophathologies. The neuronal bases of speech articulation deficits in FXS patients are currently unknown. It is yet to be established whether similar neuronal circuits control rhythmic fluid-licking pattern in mice and speech articulation movement in humans.
Collapse
Affiliation(s)
- Snigdha Roy
- Department of Anatomy and Neurobiology, University of Tennessee Health Science Center, Memphis, Tennessee 38163, USA
| | | | | | | | | | | | | |
Collapse
|
38
|
Loss of Tsc1 in vivo impairs hippocampal mGluR-LTD and increases excitatory synaptic function. J Neurosci 2011; 31:8862-9. [PMID: 21677170 DOI: 10.1523/jneurosci.1617-11.2011] [Citation(s) in RCA: 164] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The autism spectrum disorder tuberous sclerosis complex (TSC) is caused by mutations in the Tsc1 or Tsc2 genes, whose protein products form a heterodimeric complex that negatively regulates mammalian target of rapamycin-dependent protein translation. Although several forms of synaptic plasticity, including metabotropic glutamate receptor (mGluR)-dependent long-term depression (LTD), depend on protein translation at the time of induction, it is unknown whether these forms of plasticity require signaling through the Tsc1/2 complex. To examine this possibility, we postnatally deleted Tsc1 in vivo in a subset of hippocampal CA1 neurons using viral delivery of Cre recombinase in mice. We found that hippocampal mGluR-LTD was abolished by loss of Tsc1, whereas a protein synthesis-independent form of NMDA receptor-dependent LTD was preserved. Additionally, AMPA and NMDA receptor-mediated EPSCs and miniature spontaneous EPSC frequency were enhanced in Tsc1 KO neurons. These changes in synaptic function occurred in the absence of alterations in spine density, morphology, or presynaptic release probability. Our findings indicate that signaling through Tsc1/2 is required for the expression of specific forms of hippocampal synaptic plasticity as well as the maintenance of normal excitatory synaptic strength. Furthermore, these data suggest that perturbations of synaptic signaling may contribute to the pathogenesis of TSC.
Collapse
|
39
|
Pirozzi F, Tabolacci E, Neri G. The FRAXopathies: definition, overview, and update. Am J Med Genet A 2011; 155A:1803-16. [PMID: 21739597 DOI: 10.1002/ajmg.a.34113] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2011] [Accepted: 04/22/2011] [Indexed: 01/12/2023]
Abstract
The fragile X syndrome, fragile X tremor ataxia syndrome, and premature ovarian insufficiency are conditions related to the X chromosome folate-sensitive fragile site FRAXA. Therefore, we propose that they are considered as a family of disorders under the general designation of FRAXopathies. The present review will outline the main clinical and molecular features of these disorders, with special emphasis on the pathogenic mechanisms that lead to distinct phenotypes, starting from related mutations. The understanding of these mechanisms is already generating promising therapeutic approaches.
Collapse
|
40
|
Yan X, Denman RB. Conformational-dependent and independent RNA binding to the fragile x mental retardation protein. J Nucleic Acids 2011; 2011:246127. [PMID: 21772992 PMCID: PMC3136132 DOI: 10.4061/2011/246127] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2010] [Accepted: 03/16/2011] [Indexed: 01/13/2023] Open
Abstract
The interaction between the fragile X mental retardation protein (FMRP) and BC1 RNA has been the subject of controversy. We probed the parameters of RNA binding to FMRP in several ways. Nondenaturing agarose gel analysis showed that BC1 RNA transcripts produced by in vitro transcription contain a population of conformers, which can be modulated by preannealing. Accordingly, FMRP differentially binds to the annealed and unannealed conformer populations. Using partial RNase digestion, we demonstrate that annealed BC1 RNA contains a unique conformer that FMRP likely binds. We further demonstrate that this interaction is 100-fold weaker than that the binding of eEF-1A mRNA and FMRP, and that preannealing is not a general requirement for FMRP's interaction with RNA. In addition, binding does not require the N-terminal 204 amino acids of FMRP, methylated arginine residues and can be recapitulated by both fragile X paralogs. Altogether, our data continue to support a model in which BC1 RNA functions independently of FMRP.
Collapse
Affiliation(s)
- Xin Yan
- CSI/IBR Center for Developmental Neuroscience, College of Staten Island, City University of New York, Staten Island, NY 10314, USA
| | - Robert B. Denman
- Biochemical Molecular Neurobiology Laboratory, Department of Molecular Biology, New York State Institute for Basic Research in Developmental Disabilities, 1050 Forest Hill Road, Staten Island, NY 10314, USA
| |
Collapse
|
41
|
Bongmba OYN, Martinez LA, Elhardt ME, Butler K, Tejada-Simon MV. Modulation of dendritic spines and synaptic function by Rac1: a possible link to Fragile X syndrome pathology. Brain Res 2011; 1399:79-95. [PMID: 21645877 DOI: 10.1016/j.brainres.2011.05.020] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2011] [Revised: 04/25/2011] [Accepted: 05/10/2011] [Indexed: 01/28/2023]
Abstract
Rac1, a protein of the Rho GTPase subfamily, has been implicated in neuronal and spine development as well as the formation of synapses with appropriate partners. Dendrite and spine abnormalities have been implicated in several psychiatric disorders such as Fragile X syndrome, where neurons show a high density of long, thin, and immature dendritic spines. Although abnormalities in dendrites and spines have been correlated with impaired cognitive abilities in mental retardation, the causes of these malformations are not yet well understood. Fragile X syndrome is the most common type of inherited mental retardation caused by the absence of FMRP protein, a RNA-binding protein implicated in the regulation of mRNA translation and transport, leading to protein synthesis. We suggest that FMRP might act as a negative regulator on the synthesis of Rac1. Maintaining an optimal level of Rac1 and facilitating the reorganization of the cytoskeleton likely leads to normal neuronal morphology during activity-dependent plasticity. In our study, we first demonstrated that Rac1 is not only associated but necessary for normal spine development and long-term synaptic plasticity. We further showed that, in Fmr1 knockout mice, lack of FMRP induces an overactivation of Rac1 in the mouse brain and other organs that have been shown to be altered in Fragile X syndrome. In those animals, pharmacological manipulation of Rac1 partially reverses their altered long-term plasticity. Thus, regulation of Rac1 may provide a functional link among deficient neuronal morphology, aberrant synaptic plasticity and cognition impairment in Fragile X syndrome.
Collapse
Affiliation(s)
- Odelia Y N Bongmba
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston, 521 Science and Research Bldg 2, Houston, TX 77204, USA
| | | | | | | | | |
Collapse
|
42
|
Downregulation of GABA(A) β subunits is transcriptionally controlled by Fmr1p. J Mol Neurosci 2011; 46:272-5. [PMID: 21547530 DOI: 10.1007/s12031-011-9531-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2011] [Accepted: 04/25/2011] [Indexed: 10/18/2022]
Abstract
Fragile X mental retardation syndrome is caused by the transcriptional silence of FMR1. Here, a quantitative PCR technique was used to examine the effect of Fmr1p on the expression of GABA(A) β subunits in different mouse brain regions. Our results demonstrated the reduction of GABA(A) β2 mRNA in all brain regions assessed, and the reduction of GABA(A) β3 mRNA in the cortex, suggesting that the expression of GABA(A) β subunits is transcriptionally regulated by Fmr1p. This finding may help to establish the link between the transcriptional profile of the GABAergic inhibitory system and the development of fragile X mental retardation syndrome.
Collapse
|
43
|
Guo W, Allan AM, Zong R, Zhang L, Johnson EB, Schaller EG, Murthy AC, Goggin SL, Eisch AJ, Oostra BA, Nelson DL, Jin P, Zhao X. Ablation of Fmrp in adult neural stem cells disrupts hippocampus-dependent learning. Nat Med 2011; 17:559-65. [PMID: 21516088 DOI: 10.1038/nm.2336] [Citation(s) in RCA: 181] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2010] [Accepted: 02/23/2011] [Indexed: 12/14/2022]
Abstract
Deficiency in fragile X mental retardation protein (FMRP) results in fragile X syndrome (FXS), an inherited form of intellectual disability. Despite extensive research, it is unclear how FMRP deficiency contributes to the cognitive deficits in FXS. Fmrp-null mice show reduced adult hippocampal neurogenesis. As Fmrp is also enriched in mature neurons, we investigated the function of Fmrp expression in neural stem and progenitor cells (aNSCs) and its role in adult neurogenesis. Here we show that ablation of Fmrp in aNSCs by inducible gene recombination leads to reduced hippocampal neurogenesis in vitro and in vivo, as well as markedly impairing hippocampus-dependent learning in mice. Conversely, restoration of Fmrp expression specifically in aNSCs rescues these learning deficits in Fmrp-deficient mice. These data suggest that defective adult neurogenesis may contribute to the learning impairment seen in FXS, and these learning deficits can be rectified by delayed restoration of Fmrp specifically in aNSCs.
Collapse
Affiliation(s)
- Weixiang Guo
- Department of Neurosciences, University of New Mexico School of Medicine, Albuquerque, New Mexico, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Greco CM, Navarro CS, Hunsaker MR, Maezawa I, Shuler JF, Tassone F, Delany M, Au JW, Berman RF, Jin LW, Schumann C, Hagerman PJ, Hagerman RJ. Neuropathologic features in the hippocampus and cerebellum of three older men with fragile X syndrome. Mol Autism 2011; 2:2. [PMID: 21303513 PMCID: PMC3045897 DOI: 10.1186/2040-2392-2-2] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2010] [Accepted: 02/08/2011] [Indexed: 12/22/2022] Open
Abstract
Background Fragile X syndrome (FXS) is the most common inherited form of intellectual disability, and is the most common single-gene disorder known to be associated with autism. Despite recent advances in functional neuroimaging and our understanding of the molecular pathogenesis, only limited neuropathologic information on FXS is available. Methods Neuropathologic examinations were performed on post-mortem brain tissue from three older men (aged 57, 64 and 78 years) who had received a clinical or genetic diagnosis of FXS. In each case, physical and cognitive features were typical of FXS, and one man was also diagnosed with autism. Guided by reports of clinical and neuroimaging abnormalities of the limbic system and cerebellum of individuals with FXS, the current analysis focused on neuropathologic features present in the hippocampus and the cerebellar vermis. Results Histologic and immunologic staining revealed abnormalities in both the hippocampus and cerebellar vermis. Focal thickening of hippocampal CA1 and irregularities in the appearance of the dentate gyrus were identified. All lobules of the cerebellar vermis and the lateral cortex of the posterior lobe of the cerebellum had decreased numbers of Purkinje cells, which were occasionally misplaced, and often lacked proper orientation. There were mild, albeit excessive, undulations of the internal granular cell layer, with patchy foliar white matter axonal and astrocytic abnormalities. Quantitative analysis documented panfoliar atrophy of both the anterior and posterior lobes of the vermis, with preferential atrophy of the posterior lobule (VI to VII) compared with age-matched normal controls. Conclusions Significant morphologic changes in the hippocampus and cerebellum in three adult men with FXS were identified. This pattern of pathologic features supports the idea that primary defects in neuronal migration, neurogenesis and aging may underlie the neuropathology reported in FXS.
Collapse
Affiliation(s)
- Claudia M Greco
- MIND Institute, University of California-Davis Medical Center, Sacramento, CA, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Abstract
RNA localisation is an important mode of delivering proteins to their site of function. Cis-acting signals within the RNAs, which can be thought of as zip-codes, determine the site of localisation. There are few examples of fully characterised RNA signals, but the signals are thought to be defined through a combination of primary, secondary, and tertiary structures. In this chapter, we describe a selection of computational methods for predicting RNA secondary structure, identifying localisation signals, and searching for similar localisation signals on a genome-wide scale. The chapter is aimed at the biologist rather than presenting the details of each of the individual methods.
Collapse
|
46
|
Ahmed F, Benedito VA, Zhao PX. Mining Functional Elements in Messenger RNAs: Overview, Challenges, and Perspectives. FRONTIERS IN PLANT SCIENCE 2011; 2:84. [PMID: 22639614 PMCID: PMC3355573 DOI: 10.3389/fpls.2011.00084] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2011] [Accepted: 11/03/2011] [Indexed: 05/03/2023]
Abstract
Eukaryotic messenger RNA (mRNA) contains not only protein-coding regions but also a plethora of functional cis-elements that influence or coordinate a number of regulatory aspects of gene expression, such as mRNA stability, splicing forms, and translation rates. Understanding the rules that apply to each of these element types (e.g., whether the element is defined by primary or higher-order structure) allows for the discovery of novel mechanisms of gene expression as well as the design of transcripts with controlled expression. Bioinformatics plays a major role in creating databases and finding non-evident patterns governing each type of eukaryotic functional element. Much of what we currently know about mRNA regulatory elements in eukaryotes is derived from microorganism and animal systems, with the particularities of plant systems lagging behind. In this review, we provide a general introduction to the most well-known eukaryotic mRNA regulatory motifs (splicing regulatory elements, internal ribosome entry sites, iron-responsive elements, AU-rich elements, zipcodes, and polyadenylation signals) and describe available bioinformatics resources (databases and analysis tools) to analyze eukaryotic transcripts in search of functional elements, focusing on recent trends in bioinformatics methods and tool development. We also discuss future directions in the development of better computational tools based upon current knowledge of these functional elements. Improved computational tools would advance our understanding of the processes underlying gene regulations. We encourage plant bioinformaticians to turn their attention to this subject to help identify novel mechanisms of gene expression regulation using RNA motifs that have potentially evolved or diverged in plant species.
Collapse
Affiliation(s)
- Firoz Ahmed
- Bioinformatics Laboratory, Plant Biology Division, Samuel Roberts Noble FoundationArdmore, OK, USA
| | - Vagner A. Benedito
- Genetics and Developmental Biology, Plant and Soil Sciences Division, West Virginia UniversityMorgantown, WV, USA
| | - Patrick Xuechun Zhao
- Bioinformatics Laboratory, Plant Biology Division, Samuel Roberts Noble FoundationArdmore, OK, USA
- *Correspondence: Patrick Xuechun Zhao, Bioinformatics Laboratory, Plant Biology Division, Samuel Roberts Noble Foundation, 2510 Sam Noble Parkway, Ardmore, OK 73401, USA e-mail:
| |
Collapse
|
47
|
Paribello C, Tao L, Folino A, Berry-Kravis E, Tranfaglia M, Ethell IM, Ethell DW. Open-label add-on treatment trial of minocycline in fragile X syndrome. BMC Neurol 2010; 10:91. [PMID: 20937127 PMCID: PMC2958860 DOI: 10.1186/1471-2377-10-91] [Citation(s) in RCA: 163] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2010] [Accepted: 10/11/2010] [Indexed: 11/10/2022] Open
Abstract
Background Fragile X syndrome (FXS) is a disorder characterized by a variety of disabilities, including cognitive deficits, attention-deficit/hyperactivity disorder, autism, and other socio-emotional problems. It is hypothesized that the absence of the fragile X mental retardation protein (FMRP) leads to higher levels of matrix metallo-proteinase-9 activity (MMP-9) in the brain. Minocycline inhibits MMP-9 activity, and alleviates behavioural and synapse abnormalities in fmr1 knockout mice, an established model for FXS. This open-label add-on pilot trial was conducted to evaluate safety and efficacy of minocycline in treating behavioural abnormalities that occur in humans with FXS. Methods Twenty individuals with FXS, ages 13-32, were randomly assigned to receive 100 mg or 200 mg of minocycline daily. Behavioural evaluations were made prior to treatment (baseline) and again 8 weeks after daily minocycline treatment. The primary outcome measure was the Aberrant Behaviour Checklist-Community Edition (ABC-C) Irritability Subscale, and the secondary outcome measures were the other ABC-C subscales, clinical global improvement scale (CGI), and the visual analog scale for behaviour (VAS). Side effects were assessed using an adverse events checklist, a complete blood count (CBC), hepatic and renal function tests, and antinuclear antibody screen (ANA), done at baseline and at 8 weeks. Results The ABC-C Irritability Subscale scores showed significant improvement (p < 0.001), as did the VAS (p = 0.003) and the CGI (p < 0.001). The only significant treatment-related side effects were minor diarrhea (n = 3) and seroconversion to a positive ANA (n = 2). Conclusions Results from this study demonstrate that minocycline provides significant functional benefits to FXS patients and that it is well-tolerated. These findings are consistent with the fmr1 knockout mouse model results, suggesting that minocycline modifies underlying neural defects that account for behavioural abnormalities. A placebo-controlled trial of minocycline in FXS is warranted. Trial registration ClinicalTrials.gov Open-Label Trial NCT00858689.
Collapse
|
48
|
Abstract
A growing and interdisciplinary translational neuroscience research effort for neurodevelopmental disorders (NDDs) is investigating the mechanisms of dysfunction and testing effective treatment strategies in animal models and, when possible, in the clinic. NDDs with a genetic basis have received particular attention. Transgenic animals that mimic genetic insults responsible for disease in man have provided insight about mechanisms of dysfunction, and, surprisingly, have shown that cognitive deficits can be addressed in adult animals. This review will present recent translational research based on animal models of genetic NDDs, as well as pharmacotherapeutic strategies under development to address deficits of brain function for Down syndrome, fragile X syndrome, Rett syndrome, neurofibromatosis-1, tuberous sclerosis, and autism. Although these disorders vary in underlying causes and clinical presentation, common pathways and mechanisms for dysfunction have been observed. These include abnormal gene dosage, imbalance among neurotransmitter systems, and deficits in the development, maintenance and plasticity of neuronal circuits. NDDs affect multiple brain systems and behaviors that may be amenable to drug therapies that target distinct deficits. A primary goal of translational research is to replace symptomatic and supportive drug therapies with pharmacotherapies based on a principled understanding of the causes of dysfunction. Based on this principle, several recently developed therapeutic strategies offer clear promise for clinical development in man.
Collapse
|
49
|
Recombinant bacterial expression and purification of human fragile X mental retardation protein isoform 1. Protein Expr Purif 2010; 74:242-7. [PMID: 20541608 DOI: 10.1016/j.pep.2010.06.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2010] [Revised: 06/04/2010] [Accepted: 06/04/2010] [Indexed: 01/30/2023]
Abstract
The loss of expression of the fragile X mental retardation protein (FMRP) leads to fragile X syndrome. FMRP has two types of RNA binding domains, two K-homology domains and an arginine-glycine-glycine box domain, and it is proposed to act as a translation regulator of specific messenger RNA. The interest to produce sufficient quantities of pure recombinant FMRP for biochemical and biophysical studies is high. However, the recombinant bacterial expression of FMRP has had limited success, and subsequent recombinant eukaryotic and in vitro expression has also resulted in limited success. In addition, the in vitro and eukaryotic expression systems may produce FMRP which is posttranslationally modified, as phosphorylation and arginine methylation have been shown to occur on FMRP. In this study, we have successfully isolated the conditions for recombinant expression, purification and long-term storage of FMRP using Escherichia coli, with a high yield. The expression of FMRP using E. coli renders the protein devoid of the posttranslational modifications of phosphorylation and arginine methylation, allowing the study of the direct effects of these modifications individually and simultaneously. In order to assure that FMRP retained activity throughout the process, we used fluorescence spectroscopy to assay the binding activity of the FMRP arginine-glycine-glycine box for the semaphorin 3F mRNA and confirmed that FMRP remained active.
Collapse
|
50
|
Anthony K, Gallo JM. Aberrant RNA processing events in neurological disorders. Brain Res 2010; 1338:67-77. [DOI: 10.1016/j.brainres.2010.03.008] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2010] [Revised: 02/24/2010] [Accepted: 03/03/2010] [Indexed: 12/12/2022]
|