1
|
Tsotsokou G, Miliou A, Trompoukis G, Leontiadis LJ, Papatheodoropoulos C. Region-Related Differences in Short-Term Synaptic Plasticity and Synaptotagmin-7 in the Male and Female Hippocampus of a Rat Model of Fragile X Syndrome. Int J Mol Sci 2024; 25:6975. [PMID: 39000085 PMCID: PMC11240911 DOI: 10.3390/ijms25136975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 06/22/2024] [Accepted: 06/24/2024] [Indexed: 07/16/2024] Open
Abstract
Fragile X syndrome (FXS) is an intellectual developmental disorder characterized, inter alia, by deficits in the short-term processing of neural information, such as sensory processing and working memory. The primary cause of FXS is the loss of fragile X messenger ribonucleoprotein (FMRP), which is profoundly involved in synaptic function and plasticity. Short-term synaptic plasticity (STSP) may play important roles in functions that are affected by FXS. Recent evidence points to the crucial involvement of the presynaptic calcium sensor synaptotagmin-7 (Syt-7) in STSP. However, how the loss of FMRP affects STSP and Syt-7 have been insufficiently studied. Furthermore, males and females are affected differently by FXS, but the underlying mechanisms remain elusive. The aim of the present study was to investigate possible changes in STSP and the expression of Syt-7 in the dorsal (DH) and ventral (VH) hippocampus of adult males and females in a Fmr1-knockout (KO) rat model of FXS. We found that the paired-pulse ratio (PPR) and frequency facilitation/depression (FF/D), two forms of STSP, as well as the expression of Syt-7, are normal in adult KO males, but the PPR is increased in the ventral hippocampus of KO females (6.4 ± 3.7 vs. 18.3 ± 4.2 at 25 ms in wild type (WT) and KO, respectively). Furthermore, we found no gender-related differences, but did find robust region-dependent difference in the STSP (e.g., the PPR at 50 ms: 50.0 ± 5.5 vs. 17.6 ± 2.9 in DH and VH of WT male rats; 53.1 ± 3.6 vs. 19.3 ± 4.6 in DH and VH of WT female rats; 48.1 ± 2.3 vs. 19.1 ± 3.3 in DH and VH of KO male rats; and 51.2 ± 3.3 vs. 24.7 ± 4.3 in DH and VH of KO female rats). AMPA receptors are similarly expressed in the two hippocampal segments of the two genotypes and in both genders. Also, basal excitatory synaptic transmission is higher in males compared to females. Interestingly, we found more than a twofold higher level of Syt-7, not synaptotagmin-1, in the dorsal compared to the ventral hippocampus in the males of both genotypes (0.43 ± 0.1 vs. 0.16 ± 0.02 in DH and VH of WT male rats, and 0.6 ± 0.13 vs. 0.23 ± 0.04 in DH and VH of KO male rats) and in the WT females (0.97 ± 0.23 vs. 0.31 ± 0.09 in DH and VH). These results point to the susceptibility of the female ventral hippocampus to FMRP loss. Importantly, the different levels of Syt-7, which parallel the higher score of the dorsal vs. ventral hippocampus on synaptic facilitation, suggest that Syt-7 may play a pivotal role in defining the striking differences in STSP along the long axis of the hippocampus.
Collapse
Affiliation(s)
| | | | | | | | - Costas Papatheodoropoulos
- Lab of Physiology-Neurophysiology, Department of Medicine, University of Patras, 265 04 Patras, Greece; (G.T.); (A.M.); (G.T.); (L.J.L.)
| |
Collapse
|
2
|
Gredell M, Lu J, Zuo Y. The effect of single-cell knockout of Fragile X Messenger Ribonucleoprotein on synaptic structural plasticity. Front Synaptic Neurosci 2023; 15:1135479. [PMID: 37035256 PMCID: PMC10076639 DOI: 10.3389/fnsyn.2023.1135479] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Accepted: 03/07/2023] [Indexed: 04/11/2023] Open
Abstract
Fragile X Syndrome (FXS) is the best-known form of inherited intellectual disability caused by the loss-of-function mutation in a single gene. The FMR1 gene mutation abolishes the expression of Fragile X Messenger Ribonucleoprotein (FMRP), which regulates the expression of many synaptic proteins. Cortical pyramidal neurons in postmortem FXS patient brains show abnormally high density and immature morphology of dendritic spines; this phenotype is replicated in the Fmr1 knockout (KO) mouse. While FMRP is well-positioned in the dendrite to regulate synaptic plasticity, intriguing in vitro and in vivo data show that wild type neurons embedded in a network of Fmr1 KO neurons or glia exhibit spine abnormalities just as neurons in Fmr1 global KO mice. This raises the question: does FMRP regulate synaptic morphology and dynamics in a cell-autonomous manner, or do the synaptic phenotypes arise from abnormal pre-synaptic inputs? To address this question, we combined viral and mouse genetic approaches to delete FMRP from a very sparse subset of cortical layer 5 pyramidal neurons (L5 PyrNs) either during early postnatal development or in adulthood. We then followed the structural dynamics of dendritic spines on these Fmr1 KO neurons by in vivo two-photon microscopy. We found that, while L5 PyrNs in adult Fmr1 global KO mice have abnormally high density of thin spines, single-cell Fmr1 KO in adulthood does not affect spine density, morphology, or dynamics. On the contrary, neurons with neonatal FMRP deletion have normal spine density but elevated spine formation at 1 month of age, replicating the phenotype in Fmr1 global KO mice. Interestingly, these neurons exhibit elevated thin spine density, but normal total spine density, by adulthood. Together, our data reveal cell-autonomous FMRP regulation of cortical synaptic dynamics during adolescence, but spine defects in adulthood also implicate non-cell-autonomous factors.
Collapse
Affiliation(s)
| | | | - Yi Zuo
- Department of Molecular, Cell and Developmental Biology, University of California, Santa Cruz, Santa Cruz, CA, United States
| |
Collapse
|
3
|
Berry-Kravis E. Disease-Targeted Treatment Translation in Fragile X Syndrome as a Model for Neurodevelopmental Disorders. J Child Neurol 2022; 37:797-812. [PMID: 35791522 DOI: 10.1177/08830738221089740] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Fragile X syndrome (FXS), the most common monogenic cause of intellectual disability and autism spectrum disorder, has been one of the first neurodevelopmental disorders in which molecular and neuronal mechanisms of disease were identified, leading to the concept of targeting the underlying disease to reverse symptoms. Translating findings in basic science and animal models to humans with FXS has proven difficult. These challenges have prompted the FXS field to organize to build interlocking projects and initiatives to improve consistency of supportive care, make clinical research accessible to families, generate collaborative research on natural history, outcome measures and biomarkers, and create clinical trial consortia and novel trial designs. This work has resulted in improved success in recent clinical trials, providing key steps toward regulatory approval of disease-targeted treatments for FXS. Progress in the FXS field has informed translation of transformative new disease-targeted therapies for other monogenic neurodevelopmental disorders.
Collapse
Affiliation(s)
- Elizabeth Berry-Kravis
- Departments of Pediatrics, Neurological Sciences, Anatomy and Cell Biology, Rush University Medical Center, Chicago, IL, USA
| |
Collapse
|
4
|
Chen Y, Guo L, Han M, Zhang S, Chen Y, Zou J, Bai S, Cheng G, Zeng Y. Cerebellum Neuropathology and Motor Skill Deficits in Fragile X Syndrome. Int J Dev Neurosci 2022; 82:557-568. [DOI: 10.1002/jdn.10217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 07/11/2022] [Accepted: 07/14/2022] [Indexed: 11/06/2022] Open
Affiliation(s)
- Yu‐shan Chen
- Brain Science and Advanced Technology Institute, School of Medicine Wuhan University of Science and Technology Wuhan China
| | - Liu Guo
- Brain Science and Advanced Technology Institute, School of Medicine Wuhan University of Science and Technology Wuhan China
| | - Man Han
- Brain Science and Advanced Technology Institute, School of Medicine Wuhan University of Science and Technology Wuhan China
| | - Si‐ming Zhang
- Brain Science and Advanced Technology Institute, School of Medicine Wuhan University of Science and Technology Wuhan China
| | - Yi‐qi Chen
- Brain Science and Advanced Technology Institute, School of Medicine Wuhan University of Science and Technology Wuhan China
| | - Jia Zou
- Brain Science and Advanced Technology Institute, School of Medicine Wuhan University of Science and Technology Wuhan China
| | - Shu‐yuan Bai
- Brain Science and Advanced Technology Institute, School of Medicine Wuhan University of Science and Technology Wuhan China
| | - Gui‐rong Cheng
- Brain Science and Advanced Technology Institute, School of Medicine Wuhan University of Science and Technology Wuhan China
| | - Yan Zeng
- Brain Science and Advanced Technology Institute, School of Medicine Wuhan University of Science and Technology Wuhan China
| |
Collapse
|
5
|
Effects of clonidine on progressive ratio schedule performance in Fmr1 knockout mice. Psychopharmacology (Berl) 2021; 238:1133-1140. [PMID: 33449137 DOI: 10.1007/s00213-021-05760-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 01/06/2021] [Indexed: 10/22/2022]
Abstract
RATIONALE Fragile X syndrome (FXS), the most prevalent genetic form of intellectual disability, is characterized by intellectual impairment, impaired sociability, aggression, self-injury, hyperactivity, and attention deficits. A consequence of the hyperactivity and attention deficits is that individuals with FXS are frequently diagnosed with attention deficit hyperactivity disorder (ADHD) and treated with medications approved for ADHD (e.g., the α2-agonist clonidine). The pharmacotherapy of FXS is often accompanied with behavioral therapies that rely on positive reinforcement and other operant principles. Despite the commonplace mixture of drug and behavioral therapy, little attention has been paid to the observation that clonidine or other psychotropic drugs may alter operant processes. OBJECTIVES In the present progressive ratio study, we used a knockout mouse model to test the effects of the fragile X genotype, the α2-agonist clonidine, and the fragile X genotype and clonidine together on operant processes in a positive reinforcement task. RESULTS We found that clonidine decreased the progressive ratio breakpoint, increased the length of post-reinforcement pauses, and slowed the run rate. None of these effects varied by genotype. The effect on breakpoint suggests that clonidine alters motivation, but analysis using mathematical principles of reinforcement (MPR) did not rule out motor parameters as a contributor. CONCLUSIONS Our findings show that clonidine alters operant behavior and serve as a caution for combining clonidine with behavioral therapies that rely on positive reinforcement. Further research using different murine behaviors (e.g., touchscreen tasks) or different animal models (e.g., knockout rats) is needed to explore the interaction between pharmaco- and behavioral therapy.
Collapse
|
6
|
Gandhi T, Lee CC. Neural Mechanisms Underlying Repetitive Behaviors in Rodent Models of Autism Spectrum Disorders. Front Cell Neurosci 2021; 14:592710. [PMID: 33519379 PMCID: PMC7840495 DOI: 10.3389/fncel.2020.592710] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 12/09/2020] [Indexed: 12/15/2022] Open
Abstract
Autism spectrum disorder (ASD) is comprised of several conditions characterized by alterations in social interaction, communication, and repetitive behaviors. Genetic and environmental factors contribute to the heterogeneous development of ASD behaviors. Several rodent models display ASD-like phenotypes, including repetitive behaviors. In this review article, we discuss the potential neural mechanisms involved in repetitive behaviors in rodent models of ASD and related neuropsychiatric disorders. We review signaling pathways, neural circuits, and anatomical alterations in rodent models that display robust stereotypic behaviors. Understanding the mechanisms and circuit alterations underlying repetitive behaviors in rodent models of ASD will inform translational research and provide useful insight into therapeutic strategies for the treatment of repetitive behaviors in ASD and other neuropsychiatric disorders.
Collapse
Affiliation(s)
- Tanya Gandhi
- Department of Comparative Biomedical Sciences, Louisiana State University School of Veterinary Medicine, Baton Rouge, LA, United States
| | | |
Collapse
|
7
|
Feuge J, Scharkowski F, Michaelsen-Preusse K, Korte M. FMRP Modulates Activity-Dependent Spine Plasticity by Binding Cofilin1 mRNA and Regulating Localization and Local Translation. Cereb Cortex 2020; 29:5204-5216. [PMID: 30953439 DOI: 10.1093/cercor/bhz059] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Revised: 02/14/2019] [Accepted: 02/27/2019] [Indexed: 11/13/2022] Open
Abstract
Multiple variants of intellectual disability, e.g., the Fragile X Syndrome are associated with alterations in dendritic spine morphology, thereby pointing to dysregulated actin dynamics during development and processes of synaptic plasticity. Surprisingly, although the necessity of spine actin remodeling was demonstrated repeatedly, the importance and precise role of actin regulators is often undervalued. Here, we provide evidence that structural and functional plasticity are severely impaired after NMDAR-dependent LTP in the hippocampus of Fmr1 KO mice. We can link these defects to an aberrant activity-dependent regulation of Cofilin 1 (cof1) as activity-dependent modulations of local cof1 mRNA availability, local cof1 translation as well as total cof1 expression are impaired in the absence of FMRP. Finally, we can rescue activity-dependent structural plasticity in KO neurons by mimicking the regulation of cof1 observed in WT cells, thereby illustrating the potential of actin modulators to provide novel treatment strategies for the Fragile X Syndrome.
Collapse
Affiliation(s)
- Jonas Feuge
- Division of Cellular Neurobiology, Zoological Institute, TU Braunschweig, Germany
| | | | | | - Martin Korte
- Division of Cellular Neurobiology, Zoological Institute, TU Braunschweig, Germany.,Helmholtz Center for Infection Research, Research group Neuroinflammation and Neurodegeneration, Braunschweig, Germany
| |
Collapse
|
8
|
Kim T, Shin H, Song B, Won C, Yoshida H, Yamaguchi M, Cho KS, Lee I. Overexpression of
H3K36
methyltransferase
NSD
in glial cells affects brain development in
Drosophila. Glia 2020; 68:2503-2516. [DOI: 10.1002/glia.23867] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 04/21/2020] [Accepted: 05/16/2020] [Indexed: 11/09/2022]
Affiliation(s)
- Taejoon Kim
- Department of Biological Sciences, CHANS Research Center Konkuk University Seoul South Korea
| | - Hyewon Shin
- Department of Biological Sciences, CHANS Research Center Konkuk University Seoul South Korea
| | - Bokyeong Song
- Department of Biological Sciences, CHANS Research Center Konkuk University Seoul South Korea
| | - Chihyun Won
- Department of Biological Sciences, CHANS Research Center Konkuk University Seoul South Korea
| | - Hideki Yoshida
- Department of Applied Biology Kyoto Institute of Technology Kyoto Japan
| | | | - Kyoung Sang Cho
- Department of Biological Sciences, CHANS Research Center Konkuk University Seoul South Korea
| | - Im‐Soon Lee
- Department of Biological Sciences, CHANS Research Center Konkuk University Seoul South Korea
| |
Collapse
|
9
|
Michaelsen‐Preusse K, Feuge J, Korte M. Imbalance of synaptic actin dynamics as a key to fragile X syndrome? J Physiol 2018; 596:2773-2782. [PMID: 29380377 PMCID: PMC6046079 DOI: 10.1113/jp275571] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Accepted: 01/09/2018] [Indexed: 11/08/2022] Open
Abstract
Our experiences and memories define who we are, and evidence has accumulated that memory formation is dependent on functional and structural adaptations of synaptic structures in our brain. Especially dendritic spines, the postsynaptic compartments of synapses show a strong structure-to-function relationship and a high degree of structural plasticity. Although the molecular mechanisms are not completely understood, it is known that these modifications are highly dependent on the actin cytoskeleton, the major cytoskeletal component of the spine. Given the crucial involvement of actin in these mechanisms, dysregulations of spine actin dynamics (reflected by alterations in dendritic spine morphology) can be found in a variety of neurological disorders ranging from schizophrenia to several forms of autism spectrum disorders such as fragile X syndrome (FXS). FXS is caused by a single mutation leading to an inactivation of the X-linked fragile X mental retardation 1 gene and loss of its gene product, the RNA-binding protein fragile X mental retardation protein 1 (FMRP), which normally can be found both pre- and postsynaptically. FMRP is involved in mRNA transport as well as regulation of local translation at the synapse, and although hundreds of FMRP-target mRNAs could be identified only a very few interactions between FMRP and actin-regulating proteins have been reported and validated. In this review we give an overview of recent work by our lab and others providing evidence that dysregulated actin dynamics might indeed be at the very base of a deeper understanding of neurological disorders ranging from cognitive impairment to the autism spectrum.
Collapse
Affiliation(s)
- Kristin Michaelsen‐Preusse
- Zoological Institute, Division of Cellular NeurobiologyTU BraunschweigSpielmannstr. 7Braunschweig38106Germany
| | - Jonas Feuge
- Zoological Institute, Division of Cellular NeurobiologyTU BraunschweigSpielmannstr. 7Braunschweig38106Germany
| | - Martin Korte
- Zoological Institute, Division of Cellular NeurobiologyTU BraunschweigSpielmannstr. 7Braunschweig38106Germany
- Helmholtz Centre for Infection ResearchAG NINDInhoffenstr. 7Braunschweig38124Germany
| |
Collapse
|
10
|
Wallingford J, Scott AL, Rodrigues K, Doering LC. Altered Developmental Expression of the Astrocyte-Secreted Factors Hevin and SPARC in the Fragile X Mouse Model. Front Mol Neurosci 2017; 10:268. [PMID: 28900386 PMCID: PMC5581809 DOI: 10.3389/fnmol.2017.00268] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Accepted: 08/09/2017] [Indexed: 11/26/2022] Open
Abstract
Astrocyte dysfunction has been indicated in many neurodevelopmental disorders, including Fragile X Syndrome (FXS). FXS is caused by a deficiency in fragile X mental retardation protein (FMRP). FMRP regulates the translation of numerous mRNAs and its loss disturbs the composition of proteins important for dendritic spine and synapse development. Here, we investigated whether the astrocyte-derived factors hevin and SPARC, known to regulate excitatory synapse development, have altered expression in FXS. Specifically, we analyzed the expression of these factors in wild-type (WT) mice and in fragile X mental retardation 1 (Fmr1) knock-out (KO) mice that lack FMRP expression. Samples were collected from the developing cortex and hippocampus (regions of dendritic spine abnormalities in FXS) of Fmr1 KO and WT pups. Hevin and SPARC showed altered expression patterns in Fmr1 KO mice compared to WT, in a brain-region specific manner. In cortical tissue, we found a transient increase in the level of hevin in postnatal day (P)14 Fmr1 KO mice, compared to WT. Additionally, there were modest decreases in Fmr1 KO cortical levels of SPARC at P7 and P14. In the hippocampus, hevin expression was much lower in P7 Fmr1 KO mice than in WT. At P14, hippocampal hevin levels were similar between genotypes, and by P21 Fmr1 KO hevin expression surpassed WT levels. These findings imply aberrant astrocyte signaling in FXS and suggest that the altered expression of hevin and SPARC contributes to abnormal synaptic development in FXS.
Collapse
Affiliation(s)
- Jessica Wallingford
- McMaster Integrative Neuroscience Discovery and Study (MiNDS), McMaster UniversityHamilton, ON, Canada
| | - Angela L Scott
- Department of Pathology and Molecular Medicine, McMaster UniversityHamilton, ON, Canada
| | - Kelly Rodrigues
- Department of Pathology and Molecular Medicine, McMaster UniversityHamilton, ON, Canada
| | - Laurie C Doering
- McMaster Integrative Neuroscience Discovery and Study (MiNDS), McMaster UniversityHamilton, ON, Canada.,Department of Pathology and Molecular Medicine, McMaster UniversityHamilton, ON, Canada
| |
Collapse
|
11
|
Aging-Resilient Associations between the Arcuate Fasciculus and Vocabulary Knowledge: Microstructure or Morphology? J Neurosci 2017; 36:7210-22. [PMID: 27383595 DOI: 10.1523/jneurosci.4342-15.2016] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Accepted: 05/27/2016] [Indexed: 11/21/2022] Open
Abstract
UNLABELLED Vocabulary knowledge is one of the few cognitive functions that is relatively preserved in older adults, but the reasons for this relative preservation have not been well delineated. We tested the hypothesis that individual differences in vocabulary knowledge are influenced by arcuate fasciculus macrostructure (i.e., shape and volume) properties that remain stable during the aging process, rather than white matter microstructure that demonstrates age-related declines. Vocabulary was not associated with age compared to pronounced age-related declines in cognitive processing speed across 106 healthy adults (19.92-88.29 years) who participated in this neuroimaging experiment. Fractional anisotropy in the left arcuate fasciculus was significantly related to individual variability in vocabulary. This effect was present despite marked age-related differences in a T1-weighted/T2-weighted ratio (T1w/T2w) estimate of myelin that were observed throughout the left arcuate fasciculus and associated with age-related differences in cognitive processing speed. However, atypical patterns of arcuate fasciculus morphology or macrostructure were associated with decreased vocabulary knowledge. These results suggest that deterioration of tissue in the arcuate fasciculus occurs with normal aging, while having limited impact on tract organization that underlies individual differences in the acquisition and retrieval of lexical and semantic information. SIGNIFICANCE STATEMENT Vocabulary knowledge is resilient to widespread age-related declines in brain structure that limit other cognitive functions. We tested the hypothesis that arcuate fasciculus morphology, which supports the development of reading skills that bolster vocabulary, could explain this relative preservation. We disentangled (1) the effects of age-related declines in arcuate microstructure (mean diffusivity; myelin content estimate) that predicted cognitive processing speed but not vocabulary, from (2) relatively stable arcuate macrostructure (shape/volume) that explained significant variance in an age-independent association between fractional anisotropy and vocabulary. This latter result may reflect differences in fiber trajectory and organization that are resilient to aging. We propose that developmental sculpting of the arcuate fasciculus determines acquisition, storage, and access of lexical information across the adult lifespan.
Collapse
|
12
|
Entezari A, Khaniani MS, Bahrami T, Derakhshan SM, Darvish H. Screening for intermediate CGG alleles of FMR1 gene in male Iranian patients with Parkinsonism. Neurol Sci 2016; 38:123-128. [DOI: 10.1007/s10072-016-2723-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Accepted: 09/01/2016] [Indexed: 02/01/2023]
|
13
|
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: 1.9] [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
|
14
|
Hippocampal neuronal subtypes develop abnormal dendritic arbors in the presence of Fragile X astrocytes. Neuroscience 2016; 324:202-17. [DOI: 10.1016/j.neuroscience.2016.03.011] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Revised: 02/02/2016] [Accepted: 03/04/2016] [Indexed: 12/18/2022]
|
15
|
Wrenn CC, Heitzer AM, Roth AK, Nawrocki L, Valdovinos MG. Effects of clonidine and methylphenidate on motor activity in Fmr1 knockout mice. Neurosci Lett 2014; 585:109-13. [PMID: 25433180 DOI: 10.1016/j.neulet.2014.11.035] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2014] [Revised: 11/04/2014] [Accepted: 11/24/2014] [Indexed: 01/27/2023]
Abstract
Fragile X syndrome (FXS), a disorder caused by a mutation in the FMR1 gene, is often associated with Attention Deficit Hyperactivity Disorder (ADHD). Common treatments for the hyperactivity often seen in ADHD involve the use of stimulants and α2-adrenergic agonists. The Fmr1 knockout (KO) mouse has been found to be a valid model for FXS both biologically and behaviorally. Of particular interest to our research, the Fmr1 KO mouse has been demonstrated to show increased locomotion in comparison to wild type (WT) littermates. In the present study, we assessed the effects of clonidine (0.05 mg/kg) and methylphenidate (5 mg/kg) on motor activity in Fmr1 KO mice and their WT littermates in the open field test. Results showed that methylphenidate increased motor activity in both genotypes. Clonidine decreased motor activity in both genotypes, but the effect was delayed in the Fmr1 KO mice.
Collapse
Affiliation(s)
- Craige C Wrenn
- College of Pharmacy & Health Sciences, Drake University, 2507 University Avenue, Des Moines, IA 50311, USA
| | - Andrew M Heitzer
- Department of Psychology, Drake University, 2507 University Avenue, Des Moines, IA 50311, USA
| | - Alexandra K Roth
- Department of Psychology, Drake University, 2507 University Avenue, Des Moines, IA 50311, USA
| | - Lauren Nawrocki
- Neuroscience Program, Drake University, 2507 University Avenue, Des Moines, IA 50311, USA
| | - Maria G Valdovinos
- Department of Psychology, Drake University, 2507 University Avenue, Des Moines, IA 50311, USA.
| |
Collapse
|
16
|
Ryan SJ, Ehrlich DE, Rainnie DG. Morphology and dendritic maturation of developing principal neurons in the rat basolateral amygdala. Brain Struct Funct 2014; 221:839-54. [PMID: 25381464 DOI: 10.1007/s00429-014-0939-x] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2014] [Accepted: 11/01/2014] [Indexed: 02/08/2023]
Abstract
The basolateral nucleus of the amygdala (BLA) assigns emotional valence to sensory stimuli, and many amygdala-dependent behaviors undergo marked development during postnatal life. We recently showed principal neurons in the rat BLA undergo dramatic changes to their electrophysiological properties during the first postnatal month, but no study to date has thoroughly characterized changes to morphology or gene expression that may underlie the functional development of this neuronal population. We addressed this knowledge gap with reconstructions of biocytin-filled principal neurons in the rat BLA at postnatal days 7 (P7), 14, 21, 28, and 60. BLA principal neurons underwent a number of morphological changes, including a twofold increase in soma volume from P7 to P21. Dendritic arbors expanded significantly during the first postnatal month and achieved a mature distribution around P28, in terms of total dendritic length and distance from soma. The number of primary dendrites and branch points were consistent with age, but branch points were found farther from the soma in older animals. Dendrites of BLA principal neurons at P7 had few spines, and spine density increased nearly fivefold by P21. Given the concurrent increase in dendritic material, P60 neurons had approximately 17 times as many total spines as P7 neurons. Together, these developmental transitions in BLA principal neuron morphology help explain a number of concomitant electrophysiological changes during a critical period in amygdala development.
Collapse
Affiliation(s)
- Steven J Ryan
- Department of Psychiatry and Behavioral Sciences, Division of Behavioral Neuroscience and Psychiatric Disorders, Yerkes Research Center, Emory University School of Medicine, 954 Gatewood Rd., Atlanta, GA, 30033, USA
| | - David E Ehrlich
- Department of Psychiatry and Behavioral Sciences, Division of Behavioral Neuroscience and Psychiatric Disorders, Yerkes Research Center, Emory University School of Medicine, 954 Gatewood Rd., Atlanta, GA, 30033, USA
| | - Donald G Rainnie
- Department of Psychiatry and Behavioral Sciences, Division of Behavioral Neuroscience and Psychiatric Disorders, Yerkes Research Center, Emory University School of Medicine, 954 Gatewood Rd., Atlanta, GA, 30033, USA.
| |
Collapse
|
17
|
Molnár K, Kéri S. Bigger is better and worse: on the intricate relationship between hippocampal size and memory. Neuropsychologia 2014; 56:73-8. [PMID: 24423661 DOI: 10.1016/j.neuropsychologia.2014.01.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2013] [Revised: 12/14/2013] [Accepted: 01/03/2014] [Indexed: 10/25/2022]
Abstract
The structure-function relationship between the hippocampal region and memory is a debated topic in the literature. It has been suggested that larger hippocampi are associated with less effective memory performance in healthy young adults because of a partial synaptic pruning. Here, we tested this hypothesis in individuals with Fragile X Syndrome (FXS) with known abnormal pruning and IQ- and age-matched individuals with hypoxic brain injury, preterm birth, and obstetric complications. Results revealed larger normalized hippocampal volume in FXS compared with neurotypical controls, whereas individuals with hypoxic injury had smaller hippocampi. In neurotypical controls and individuals with hypoxic injury, better general memory, as indexed by the Wechsler Memory Scale-Revised, was associated with larger hippocampus. In contrast, in FXS we observed the opposite relationship: larger hippocampus was associated with worse general memory. Caudate volume did not correlate with memory in either group. These results suggest that incomplete pruning in young healthy adults may not contribute to less efficient memory capacity, and hippocampal size is positively associated with memory performance. However, abnormally large and poorly pruned hippocampus may indeed be less effective in FXS.
Collapse
Affiliation(s)
- Katalin Molnár
- Nyírő Gyula Hospital, National Institute of Psychiatry and Addictions, Budapest, Hungary; University of Szeged, Faculty of Medicine, Department of Physiology, Szeged, Hungary; Budapest University of Technology and Economics, Department of Cognitive Science, Budapest, Hungary
| | - Szabolcs Kéri
- Nyírő Gyula Hospital, National Institute of Psychiatry and Addictions, Budapest, Hungary; University of Szeged, Faculty of Medicine, Department of Physiology, Szeged, Hungary; Budapest University of Technology and Economics, Department of Cognitive Science, Budapest, Hungary.
| |
Collapse
|
18
|
Abstract
The central nervous system is comprised of numerous cell types that work in concert to facilitate proper function and homeostasis. Disruption of these carefully orchestrated networks results in neuronal dysfunction, manifesting itself in a variety of neurologic disorders. Although neuronal dysregulation is causative of symptoms that manifest in the clinic, the etiology of these disorders is often more complex than simply a loss of neurons or intrinsic dysregulation of their function. In the adult brain, astrocytes comprise the most abundant cell type and play key roles in central nervous system physiology; therefore, it stands to reason that dysregulation of normal astrocyte function contributes to the etiology and progression of varied neurologic disorders. We review here some neurologic disorders associated with an astrocyte factor and discuss how the related astrocyte dysfunction contributes to the etiology or progression of these disorders or both.
Collapse
|
19
|
Clarke LE, Barres BA. Emerging roles of astrocytes in neural circuit development. Nat Rev Neurosci 2013; 14:311-21. [PMID: 23595014 DOI: 10.1038/nrn3484] [Citation(s) in RCA: 675] [Impact Index Per Article: 61.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Astrocytes are now emerging as key participants in many aspects of brain development, function and disease. In particular, new evidence shows that astrocytes powerfully control the formation, maturation, function and elimination of synapses through various secreted and contact-mediated signals. Astrocytes are also increasingly being implicated in the pathophysiology of many psychiatric and neurological disorders that result from synaptic defects. A better understanding of how astrocytes regulate neural circuit development and function in the healthy and diseased brain might lead to the development of therapeutic agents to treat these diseases.
Collapse
Affiliation(s)
- Laura E Clarke
- Department of Neurobiology, Stanford University School of Medicine, Stanford, California 94305, USA.
| | | |
Collapse
|
20
|
What's hAPPening at synapses? The role of amyloid β-protein precursor and β-amyloid in neurological disorders. Mol Psychiatry 2013; 18:425-34. [PMID: 22925831 DOI: 10.1038/mp.2012.122] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Accumulating evidence suggests that dysregulated levels of amyloid β-protein precursor (APP) and its catabolites contribute to the impaired synaptic plasticity and seizure incidence observed in several neurological disorders, including Alzheimer's disease, fragile X syndrome, Down's syndrome, autism, epilepsy and Parkinson's disease as well as in brain injury. This review article summarizes what is known regarding the synaptic synthesis, processing and function of APP and amyloid-beta (Aβ), as well as discusses how these proteins could contribute to the altered synaptic plasticity and pathology of the aforementioned disorders. In addition, APP and its proteolytic fragments are emerging as biomarkers for neurological health, and pharmacological interventions that modulate their levels, such as secretase inhibitors, passive immunotherapy against Aβ and mGluR5 antagonists, are reviewed.
Collapse
|
21
|
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: 45] [Impact Index Per Article: 3.8] [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
|
22
|
Telias M, Segal M, Ben-Yosef D. Neural differentiation of Fragile X human Embryonic Stem Cells reveals abnormal patterns of development despite successful neurogenesis. Dev Biol 2012; 374:32-45. [PMID: 23219959 DOI: 10.1016/j.ydbio.2012.11.031] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2012] [Revised: 11/24/2012] [Accepted: 11/28/2012] [Indexed: 12/23/2022]
Abstract
Fragile X Syndrome (FXS) is the most common form of inherited intellectual disability, caused by developmentally regulated inactivation of FMR1, leading to the absence of its encoded protein FMRP. We have previously shown that undifferentiated Fragile X human Embryonic Stem Cells (FX-hESCs) express FMRP, despite the presence of the full FMR1 mutation (>200 CGG repeats). We describe here, for the first time, in-vitro differentiation of FX-hESCs into neurons progressively inactivating FMR1. Abnormal neurogenesis and aberrant gene expression were found already during early stages of differentiation, leading to poor neuronal maturation and high gliogenic development. Human FX neurons fired action potentials but displayed poor spontaneous synaptic activity and lacked reactivity to glutamate. Our dynamic FX-hESCs model can contribute to the understanding of the sequence of developmental events taking place during neurogenesis and how they are altered in FXS individuals, leading to intellectual disability. Furthermore, it may shed light over the striking phenotypic features characterizing FXS in human.
Collapse
Affiliation(s)
- Michael Telias
- Stem Cell Research Lab, Racine IVF Unit, Lis Maternity Hospital, Tel-Aviv Sourasky Medical Center, 6 Weizmann St., 64239 Tel-Aviv, Israel
| | | | | |
Collapse
|
23
|
Molofsky AV, Krencik R, Krenick R, Ullian EM, Ullian E, Tsai HH, Deneen B, Richardson WD, Barres BA, Rowitch DH. Astrocytes and disease: a neurodevelopmental perspective. Genes Dev 2012; 26:891-907. [PMID: 22549954 DOI: 10.1101/gad.188326.112] [Citation(s) in RCA: 510] [Impact Index Per Article: 42.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Astrocytes are no longer seen as a homogenous population of cells. In fact, recent studies indicate that astrocytes are morphologically and functionally diverse and play critical roles in neurodevelopmental diseases such as Rett syndrome and fragile X mental retardation. This review summarizes recent advances in astrocyte development, including the role of neural tube patterning in specification and developmental functions of astrocytes during synaptogenesis. We propose here that a precise understanding of astrocyte development is critical to defining heterogeneity and could lead advances in understanding and treating a variety of neuropsychiatric diseases.
Collapse
Affiliation(s)
- Anna V Molofsky
- Department of Pediatrics, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California at San Francisco, San Francisco, California 94143, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Human pathologies associated with defective RNA transport and localization in the nervous system. Biol Cell 2012; 99:649-61. [DOI: 10.1042/bc20070045] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
25
|
Withers GS, Wallace CS, Gibbs EM, Emery IR, Applegate ML. Synapses on demand require dendrites at the ready: how defining stages of dendritic development in vitro could inform studies of behaviorally driven information storage in the brain. Dev Psychobiol 2011; 53:443-55. [PMID: 21678392 DOI: 10.1002/dev.20560] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Bill Greenough's work provides a framework for thinking about synaptogenesis not only as a key step in the initial wiring of neural systems according to a species typical plan (i.e., experience-expectant development), but also as a mechanism for storing information based an individual's unique experience over its lifetime (i.e., experience-dependent plasticity). Analysis of synaptic development in vitro brings a new opportunity to test the limits of expectant-expectant development at the level of the individual neuron. We analyzed dendritic growth, synapse formation, and the development of specialized cytoplasmic microdomains during development in cultured hippocampal neurons, to determine if the timing of each of these events is correlated. Taken together, the findings reported here support the hypotheses that (1) dendritic development is rate limiting in synapse formation and (2) synaptic circuits are assembled in a step-wise fashion consistent with a stage-specific shift from genomically pre-programmed to activity-dependent mechanisms.
Collapse
Affiliation(s)
- Ginger S Withers
- Department of Biology, Whitman College, Walla Walla, WA 99362, USA.
| | | | | | | | | |
Collapse
|
26
|
Satb1 ablation alters temporal expression of immediate early genes and reduces dendritic spine density during postnatal brain development. Mol Cell Biol 2011; 32:333-47. [PMID: 22064485 DOI: 10.1128/mcb.05917-11] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Complex behaviors, such as learning and memory, are associated with rapid changes in gene expression of neurons and subsequent formation of new synaptic connections. However, how external signals are processed to drive specific changes in gene expression is largely unknown. We found that the genome organizer protein Satb1 is highly expressed in mature neurons, primarily in the cerebral cortex, dentate hilus, and amygdala. In Satb1-null mice, cortical layer morphology was normal. However, in postnatal Satb1-null cortical pyramidal neurons, we found a substantial decrease in the density of dendritic spines, which play critical roles in synaptic transmission and plasticity. Further, we found that in the cerebral cortex, Satb1 binds to genomic loci of multiple immediate early genes (IEGs) (Fos, Fosb, Egr1, Egr2, Arc, and Bdnf) and other key neuronal genes, many of which have been implicated in synaptic plasticity. Loss of Satb1 resulted in greatly alters timing and expression levels of these IEGs during early postnatal cerebral cortical development and also upon stimulation in cortical organotypic cultures. These data indicate that Satb1 is required for proper temporal dynamics of IEG expression. Based on these findings, we propose that Satb1 plays a critical role in cortical neurons to facilitate neuronal plasticity.
Collapse
|
27
|
Grabrucker AM, Schmeisser MJ, Schoen M, Boeckers TM. Postsynaptic ProSAP/Shank scaffolds in the cross-hair of synaptopathies. Trends Cell Biol 2011; 21:594-603. [PMID: 21840719 DOI: 10.1016/j.tcb.2011.07.003] [Citation(s) in RCA: 187] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2011] [Revised: 07/10/2011] [Accepted: 07/14/2011] [Indexed: 01/31/2023]
Abstract
Intact synaptic homeostasis is a fundamental prerequisite for a healthy brain. Thus, it is not surprising that altered synaptic morphology and function are involved in the molecular pathogenesis of so-called synaptopathies including autism, schizophrenia (SCZ) and Alzheimer's disease (AD). Intriguingly, various recent studies revealed a crucial role of postsynaptic ProSAP/Shank scaffold proteins in all of the aforementioned disorders. Considering these findings, we follow the hypothesis that ProSAP/Shank proteins are key regulators of synaptic development and plasticity with clear-cut isoform-specific roles. We thus propose a model where ProSAP/Shank proteins are in the center of a postsynaptic signaling pathway that is disrupted in several neuropsychiatric disorders.
Collapse
|
28
|
Napoli E, Ross-Inta C, Wong S, Omanska-Klusek A, Barrow C, Iwahashi C, Garcia-Arocena D, Sakaguchi D, Berry-Kravis E, Hagerman R, Hagerman PJ, Giulivi C. Altered zinc transport disrupts mitochondrial protein processing/import in fragile X-associated tremor/ataxia syndrome. Hum Mol Genet 2011; 20:3079-92. [PMID: 21558427 DOI: 10.1093/hmg/ddr211] [Citation(s) in RCA: 86] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Fragile X-associated tremor/ataxia syndrome (FXTAS) is a late-onset neurodegenerative disorder that affects individuals who are carriers of small CGG premutation expansions in the fragile X mental retardation 1 (FMR1) gene. Mitochondrial dysfunction was observed as an incipient pathological process occurring in individuals who do not display overt features of FXTAS (1). Fibroblasts from premutation carriers had lower oxidative phosphorylation capacity (35% of controls) and Complex IV activity (45%), and higher precursor-to-mature ratios (P:M) of nDNA-encoded mitochondrial proteins (3.1-fold). However, fibroblasts from carriers with FXTAS symptoms presented higher FMR1 mRNA expression (3-fold) and lower Complex V (38%) and aconitase activities (43%). Higher P:M of ATPase β-subunit (ATPB) and frataxin were also observed in cortex from patients that died with FXTAS symptoms. Biochemical findings observed in FXTAS cells (lower mature frataxin, lower Complex IV and aconitase activities) along with common phenotypic traits shared by Friedreich's ataxia and FXTAS carriers (e.g. gait ataxia, loss of coordination) are consistent with a defective iron homeostasis in both diseases. Higher P:M, and lower ZnT6 and mature frataxin protein expression suggested defective zinc and iron metabolism arising from altered ZnT protein expression, which in turn impairs the activity of mitochondrial Zn-dependent proteases, critical for the import and processing of cytosolic precursors, such as frataxin. In support of this hypothesis, Zn-treated fibroblasts showed a significant recovery of ATPB P:M, ATPase activity and doubling time, whereas Zn and desferrioxamine extended these recoveries and rescued Complex IV activity.
Collapse
Affiliation(s)
- Eleonora Napoli
- Department of Molecular Biosciences, School of Veterinary Medicine, School of Medicine, University of California Davis, Davis, CA 95616, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Early continuous inhibition of group 1 mGlu signaling partially rescues dendritic spine abnormalities in the Fmr1 knockout mouse model for fragile X syndrome. Psychopharmacology (Berl) 2011; 215:291-300. [PMID: 21181121 DOI: 10.1007/s00213-010-2130-2] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2010] [Accepted: 12/01/2010] [Indexed: 12/17/2022]
Abstract
RATIONALE Abnormal dendritic spine morphology is a significant neuroanatomical defect in fragile X mental retardation. It has been suggested that overactive group 1 metabotropic glutamate receptor (mGlu) signaling is associated with the spine dysmorphology occurring in fragile X syndrome (FXS). Thus, group 1 mGlu became a new therapeutic target for the treatment of FXS. OBJECTIVE The purpose of this study was to identify the effect of inhibition of mGlu signaling in FXS. METHODS We observed the changes in dendritic spines after pharmacological modulation of mGlu signaling in an Fmr1 knockout (KO) mouse model. RESULTS The activation of group 1 mGlu resulted in elongation of dendritic spines in the cultured neurons derived from Fmr1 KO mice and wild-type (WT) mice. Antagonism of group 1 mGlu reduced the average spine length of Fmr1 KO neurons. Furthermore, systemic administration of the selective group 1 mGlu5 antagonist 2-methyl-6-phenylethynyl pyridine (MPEP) reduced the average spine length and density in the cortical neurons of Fmr1 KO mice at developmental age. For the adult mice, MPEP administration was less effective for the restoration of spine length. The percentage of immature spines showed a similar reduction in parallel to the changes of spine length. Temporary MPEP intervention with single-dose treatment did not show any effect. CONCLUSION These results show that MPEP administration could partially rescue the morphological deficits of dendritic spines in Fmr1 KO mice at developmental age.
Collapse
|
30
|
Paul LK. Developmental malformation of the corpus callosum: a review of typical callosal development and examples of developmental disorders with callosal involvement. J Neurodev Disord 2011; 3:3-27. [PMID: 21484594 PMCID: PMC3163989 DOI: 10.1007/s11689-010-9059-y] [Citation(s) in RCA: 155] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2010] [Accepted: 08/13/2010] [Indexed: 12/11/2022] Open
Abstract
This review provides an overview of the involvement of the corpus callosum (CC) in a variety of developmental disorders that are currently defined exclusively by genetics, developmental insult, and/or behavior. I begin with a general review of CC development, connectivity, and function, followed by discussion of the research methods typically utilized to study the callosum. The bulk of the review concentrates on specific developmental disorders, beginning with agenesis of the corpus callosum (AgCC)-the only condition diagnosed exclusively by callosal anatomy. This is followed by a review of several genetic disorders that commonly result in social impairments and/or psychopathology similar to AgCC (neurofibromatosis-1, Turner syndrome, 22q11.2 deletion syndrome, Williams yndrome, and fragile X) and two forms of prenatal injury (premature birth, fetal alcohol syndrome) known to impact callosal development. Finally, I examine callosal involvement in several common developmental disorders defined exclusively by behavioral patterns (developmental language delay, dyslexia, attention-deficit hyperactive disorder, autism spectrum disorders, and Tourette syndrome).
Collapse
Affiliation(s)
- Lynn K Paul
- Division of Humanities and Social Sciences, California Institute of Technology, HSS 228-77, Caltech, Pasadena, CA, 91125, USA,
| |
Collapse
|
31
|
Roullet FI, Crawley JN. Mouse models of autism: testing hypotheses about molecular mechanisms. Curr Top Behav Neurosci 2011; 7:187-212. [PMID: 21225409 PMCID: PMC3396120 DOI: 10.1007/7854_2010_113] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Autism is a neurodevelopmental disorder that is currently diagnosed by the presence of three behavioral criteria (1) qualitative impairments in reciprocal social interactions, (2) deficits in communication, including delayed language and noninteractive conversation, and (3) motor stereotypies, repetitive behaviors, insistence on sameness, and restricted interests. This chapter describes analogous behavioral assays that have been developed for mice, including tests for social approach, reciprocal social interactions, olfactory communication, ultrasonic vocalizations, repetitive and perseverative behaviors, and motor stereotypies. Examples of assay applications to genetic mouse models of autism are provided. Robust endophenotypes that are highly relevant to the core symptoms of autism are enabling the search for the genetic and environmental causes of autism, and the discovery of effective treatments.
Collapse
Affiliation(s)
- Florence I. Roullet
- Laboratory of Behavioral Neuroscience, Intramural Research Program, National Institute of Mental Health, Building 35 Room 1C-903/909, Mail Code 3730, Bethesda, MD 20892-3730, USA
| | - Jacqueline N. Crawley
- Laboratory of Behavioral Neuroscience, Intramural Research Program, National Institute of Mental Health, Building 35 Room 1C-903/909, Mail Code 3730, Bethesda, MD 20892-3730, USA
| |
Collapse
|
32
|
Jacobs S, Nathwani M, Doering LC. Fragile X astrocytes induce developmental delays in dendrite maturation and synaptic protein expression. BMC Neurosci 2010; 11:132. [PMID: 20955577 PMCID: PMC2970604 DOI: 10.1186/1471-2202-11-132] [Citation(s) in RCA: 79] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2010] [Accepted: 10/18/2010] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Fragile X syndrome is the most common inherited form of mental impairment characterized by cognitive impairment, attention deficit and autistic behaviours. The mouse model of Fragile X is used to study the underlying neurobiology associated with behavioral deficiencies. The effect of Fragile X glial cells on the development of neurons has not been studied. We used a co-culture technique in combination with morphometrics on immunostained neurons to investigate the role of astrocytes in the development delays associated with hippocampal neuron development. RESULTS We found that hippocampal neurons grown on Fragile X astrocytes exhibited a significant difference from the neurons grown with normal astrocytes after 7 days in vitro for many parameters including increases in dendritic branching and in area of the cell body. However, after 21 days in culture, the neurons grown on Fragile X astrocytes exhibited morphological characteristics that did not differ significantly from the neurons grown on normal astrocytes. With antibodies to the pre-synaptic protein, synapsin, and to the excitatory post-synaptic protein, PSD-95, we quantified the number of developing excitatory synapses on the dendrites. In addition to the delays in dendritic patterning, the development of excitatory synapses was also delayed in the hippocampal neurons. CONCLUSIONS These experiments are the first to establish a role for astrocytes in the delayed growth characteristics and abnormal morphological features in dendrites and synapses that characterize the Fragile X syndrome.
Collapse
Affiliation(s)
- Shelley Jacobs
- Department of Pathology & Molecular Medicine, McMaster University, 1200 Main Street West, Hamilton, Ontario, L8N 3Z5, Canada
| | | | | |
Collapse
|
33
|
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: 11.6] [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
|
34
|
Evidence of mitochondrial dysfunction in fragile X-associated tremor/ataxia syndrome. Biochem J 2010; 429:545-52. [PMID: 20513237 DOI: 10.1042/bj20091960] [Citation(s) in RCA: 141] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
FXTAS (fragile X-associated tremor/ataxia syndrome) is a late-onset neurodegenerative disorder that affects individuals who are carriers of premutation expansions (55-200 CGG repeats) in the 5' untranslated region of the FMR1 (fragile X mental retardation 1) gene. The role of MD (mitochondrial dysfunction) in FXTAS was evaluated in fibroblasts and brain samples from premutation carriers with and without FXTAS symptoms, with a range of CGG repeats. This study resulted in several important conclusions: (i) decreased NAD- and FAD-linked oxygen uptake rates and uncoupling between electron transport and synthesis of ATP were observed in fibroblasts from premutation carriers; (ii) a lower expression of mitochondrial proteins preceded both in age and in CGG repeats the appearance of overt clinical involvement; (iii) the CGG repeat size required for altered mitochondrial protein expression was also smaller than that required to produce brain intranuclear inclusions from individuals with the premutation who died, suggesting that MD is an incipient pathological process occurring in individuals who do not display overt features of FXTAS; and (iv) on the basis of the CGG repeats, MD preceded the increase in oxidative/nitrative stress damage, indicating that the latter is a late event. MD in carriers of small CGG repeats, even when the allele size is not sufficient to produce FXTAS, may predispose them to other disorders (e.g. Parkinson's disease) that are likely to involve MD, and to environmental stressors, which may trigger the development of FXTAS symptoms. Detection of MD is of critical importance to the management of FXTAS, since it opens up additional treatment options for this disorder.
Collapse
|
35
|
Wang LW, Berry-Kravis E, Hagerman RJ. Fragile X: leading the way for targeted treatments in autism. Neurotherapeutics 2010; 7:264-74. [PMID: 20643379 PMCID: PMC4084556 DOI: 10.1016/j.nurt.2010.05.005] [Citation(s) in RCA: 103] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2010] [Revised: 05/19/2010] [Accepted: 05/22/2010] [Indexed: 10/19/2022] Open
Abstract
Two different mutations in the FMR1 gene may lead to autism. The full mutation, with >200 CGG repeats in the 5' end of FMR1, leads to hypermethylation and transcriptional silencing of FMR1, resulting in absence or deficiency of the protein product, FMRP. Deficiency of FMRP in the brain causes fragile X syndrome (FXS). Autism occurs in approximately 30% of those with FXS, and pervasive developmental disorders-not otherwise specified occur in an additional 30%. FMRP is an RNA binding protein that modulates receptor-mediated dendritic translation; deficiency leads to dysregulation of many proteins important for synaptic plasticity. Group I metabotropic glutamate receptor (mGluR1/5) activated translation is upregulated in FXS, and new targeted treatments that act on this system include mGluR5 antagonists and GABA agonists, which may reverse the cognitive and behavioral deficits in FXS. Matrix metalloproteinase 9 (MMP-9) is one of the proteins elevated in FXS, and minocycline reduces excess MMP-9 activity in the Fmr1 knockout mouse model of FXS. Both minocycline and mGluR5 antagonists are currently being evaluated in patients with FXS through controlled treatment trials. The premutation (55-200 CGG repeats) may also contribute to the mechanism of autism in approximately 10% of males and 2-3% of females. Premutations with <150 repeats exert cellular effects through a different molecular mechanism, one that involves elevated levels of FMR1 mRNA, CGG-mediated toxicity to neurons, early cell death, and fragile X-associated tremor/ataxia syndrome. In those with large premutations (150-200), lowered levels of FMRP also occur.
Collapse
Affiliation(s)
- Lulu W Wang
- Department of Pediatrics, University of California, Davis, School of Medicine, Sacramento, California 95817, USA.
| | | | | |
Collapse
|
36
|
Jacobs S, Doering LC. Astrocytes prevent abnormal neuronal development in the fragile x mouse. J Neurosci 2010; 30:4508-14. [PMID: 20335488 PMCID: PMC6634485 DOI: 10.1523/jneurosci.5027-09.2010] [Citation(s) in RCA: 124] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2009] [Revised: 02/08/2010] [Accepted: 03/01/2010] [Indexed: 11/21/2022] Open
Abstract
Astrocytes are now distinguished as major regulators of neuronal growth and synaptic development. Recently, they have been identified as key players in the progression of a number of developmental disorders; however, in fragile X syndrome (FXS), the role of astrocytes is not known. Using a coculture design, we found that hippocampal neurons exhibited abnormal dendritic morphology and a decreased number of presynaptic and postsynaptic protein aggregates when they were grown on astrocytes from a fragile X mouse. Moreover, we found that normal astrocytes could prevent the development of abnormal dendrite morphology and preclude the reduction of presynaptic and postsynaptic protein clusters in neurons from a fragile X mouse. These experiments are the first to establish a role for astrocytes in the altered neurobiology of FXS. Our results support the notion that astrocytes contribute to abnormal dendrite morphology and the dysregulated synapse development in FXS.
Collapse
Affiliation(s)
- Shelley Jacobs
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario L8N 3Z5, Canada
| | - Laurie C. Doering
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario L8N 3Z5, Canada
| |
Collapse
|
37
|
Sallansonnet-Froment M, De Greslan T, Roux X, Bounolleau P, Ouologuem M, Taillia H, Ricard D, Renard JL. [Tremor/ataxia syndrome related to Fragile X premutation]. Presse Med 2009; 39:187-95. [PMID: 19419833 DOI: 10.1016/j.lpm.2008.12.035] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2008] [Revised: 12/20/2008] [Accepted: 12/31/2008] [Indexed: 11/20/2022] Open
Abstract
The FXTAS syndrome (Fragile X-associated tremor/ataxia syndrome) is a specific neurodegenerative syndrome affecting subjects carrying a premutation of the FMR1 (fragile X mental retardation 1) gene. It affects mainly men with the premutation and aged more than 50 years. This syndrome is separate and distinct from the fragile X syndrome. The FXTAS syndrome remains underestimated today. It should be considered in patients older than 50 years with tremors and cerebellar ataxia, especially when Parkinson disease or cognitive disorders are present or when there is a family history of infertility, early menopause, or mental retardation. In these patients, hyperintense signals of mid-cerebellar peduncle images on T2 and FLAIR MRI justify genetic testing for the FMR1 premutation.
Collapse
Affiliation(s)
- Magali Sallansonnet-Froment
- Hôpital d'instruction des armées du Val-de-Grâce, Service de neurologie, F-75230 Paris Cedex 05, B.P.1-00446 Armées, France.
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Berry-Kravis E, Hessl D, Coffey S, Hervey C, Schneider A, Yuhas J, Hutchison J, Snape M, Tranfaglia M, Nguyen DV, Hagerman R. A pilot open label, single dose trial of fenobam in adults with fragile X syndrome. J Med Genet 2009; 46:266-71. [PMID: 19126569 PMCID: PMC2658751 DOI: 10.1136/jmg.2008.063701] [Citation(s) in RCA: 270] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Objective: A pilot open label, single dose trial of fenobam, an mGluR5 antagonist, was conducted to provide an initial evaluation of safety and pharmacokinetics in adult males and females with fragile X syndrome (FXS). Methods: Twelve subjects, recruited from two fragile X clinics, received a single oral dose of 50–150 mg of fenobam. Blood for pharmacokinetic testing, vital signs and side effect screening was obtained at baseline and numerous time points for 6 h after dosing. Outcome measures included prepulse inhibition (PPI) and a continuous performance test (CPT) obtained before and after dosing to explore the effects of fenobam on core phenotypic measures of sensory gating, attention and inhibition. Results: There were no significant adverse reactions to fenobam administration. Pharmacokinetic analysis showed that fenobam concentrations were dose dependent but variable, with mean (SEM) peak values of 39.7 (18.4) ng/ml at 180 min after the 150 mg dose. PPI met a response criterion of an improvement of at least 20% over baseline in 6 of 12 individuals (4/6 males and 2/6 females). The CPT did not display improvement with treatment due to ceiling effects. Conclusions: Clinically significant adverse effects were not identified in this study of single dose fenobam across the range of dosages utilised. The positive effects seen in animal models of FXS treated with fenobam or other mGluR5 antagonists, the apparent lack of clinically significant adverse effects, and the potential beneficial clinical effects seen in this pilot trial support further study of the compound in adults with FXS.
Collapse
Affiliation(s)
- E Berry-Kravis
- Departments of Pediatrics, Rush University Medical Center, 1725 West Harrison Street, Suite 718, Chicago, IL 60612, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Smit AE, van der Geest JN, Vellema M, Koekkoek SKE, Willemsen R, Govaerts LCP, Oostra BA, De Zeeuw CI, VanderWerf F. Savings and extinction of conditioned eyeblink responses in fragile X syndrome. GENES BRAIN AND BEHAVIOR 2008; 7:770-7. [PMID: 18616611 PMCID: PMC2613242 DOI: 10.1111/j.1601-183x.2008.00417.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
The fragile X syndrome (FRAXA) is the most widespread heritable form of mental retardation caused by the lack of expression of the fragile X mental retardation protein (FMRP). This lack has been related to deficits in cerebellum-mediated acquisition of conditioned eyelid responses in individuals with FRAXA. In the present behavioral study, long-term effects of deficiency of FMRP were investigated by examining the acquisition, savings and extinction of delay eyeblink conditioning in male individuals with FRAXA. In the acquisition experiment, subjects with FRAXA displayed a significantly poor performance compared with controls. In the savings experiment performed at least 6 months later, subjects with FRAXA and controls showed similar levels of savings of conditioned responses. Subsequently, extinction was faster in subjects with FRAXA than in controls. These findings confirm that absence of the FMRP affects cerebellar motor learning. The normal performance in the savings experiment and aberrant performance in the acquisition and extinction experiments of individuals with FRAXA suggest that different mechanisms underlie acquisition, savings and extinction of cerebellar motor learning.
Collapse
Affiliation(s)
- A E Smit
- Department of Neuroscience, Erasmus MC, Rotterdam, The Netherlands
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Berry-Kravis E, Abrams L, Coffey SM, Hall DA, Greco C, Gane LW, Grigsby J, Bourgeois JA, Finucane B, Jacquemont S, Brunberg JA, Zhang L, Lin J, Tassone F, Hagerman PJ, Hagerman RJ, Leehey MA. Fragile X-associated tremor/ataxia syndrome: clinical features, genetics, and testing guidelines. Mov Disord 2008; 22:2018-30, quiz 2140. [PMID: 17618523 DOI: 10.1002/mds.21493] [Citation(s) in RCA: 220] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Fragile X-associated tremor/ataxia syndrome (FXTAS) is a neurodegenerative disorder with core features of action tremor and cerebellar gait ataxia. Frequent associated findings include parkinsonism, executive function deficits and dementia, neuropathy, and dysautonomia. Magnetic Resonance Imaging studies in FXTAS demonstrate increased T2 signal intensity in the middle cerebellar peduncles (MCP sign) in the majority of patients. Similar signal alterations are seen in deep and subependymal cerebral white matter, as is general cortical and subcortical atrophy. The major neuropathological feature of FXTAS is the presence of intranuclear, neuronal, and astrocytic, inclusions in broad distribution throughout the brain and brainstem. FXTAS is caused by moderate expansions (55-200 repeats; premutation range) of a CGG trinucleotide in the fragile X mental retardation 1 (FMR1) gene, the same gene which causes fragile X syndrome when in the full mutation range (200 or greater CGG repeats). The pathogenic mechanism is related to overexpression and toxicity of the FMR1 mRNA per se. Although only recently discovered, and hence currently under-diagnosed, FXTAS is likely to be one of the most common single-gene disorders leading to neurodegeneration in males. In this report, we review information available on the clinical, radiological, and pathological features, and prevalence and management of FXTAS. We also provide guidelines for the practitioner to assist with identifying appropriate patients for DNA testing for FXTAS, as well as recommendations for genetic counseling once a diagnosis of FXTAS is made.
Collapse
|
41
|
Valdovinos MG. Brief review of current research in FXS: implications for treatment with psychotropic medication. RESEARCH IN DEVELOPMENTAL DISABILITIES 2007; 28:539-45. [PMID: 17034990 DOI: 10.1016/j.ridd.2006.09.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2006] [Revised: 08/07/2006] [Accepted: 09/09/2006] [Indexed: 05/12/2023]
Abstract
The purpose of this paper is to provide a brief review of current research in fragile X syndrome (FXS) with regards to the morphology and behavioral phenotype associated with FXS and the use of psychotropic medication for the treatment of behavior problems (e.g., aggression) often seen in FXS (full mutation). The lack of production of the fragile X mental retardation protein (FMRP) is associated with FXS and has been found to result in various neuronal changes such as altered dendritic morphology and function as well as altered neurotransmitter functions. A review of the basic literature on animal models and the relevance of these findings for the use of psychotropic treatment of problem behaviors in FXS will be discussed. Future research directions will be presented.
Collapse
Affiliation(s)
- Maria G Valdovinos
- Drake University, Psychology Department, 2507 University Avenue, Des Moines, IA 50311, United States.
| |
Collapse
|
42
|
Lee AD, Leow AD, Lu A, Reiss AL, Hall S, Chiang MC, Toga AW, Thompson PM. 3D pattern of brain abnormalities in Fragile X syndrome visualized using tensor-based morphometry. Neuroimage 2007; 34:924-38. [PMID: 17161622 PMCID: PMC1995408 DOI: 10.1016/j.neuroimage.2006.09.043] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2006] [Revised: 08/22/2006] [Accepted: 09/21/2006] [Indexed: 11/17/2022] Open
Abstract
Fragile X syndrome (FraX), a genetic neurodevelopmental disorder, results in impaired cognition with particular deficits in executive function and visuo-spatial skills. Here we report the first detailed 3D maps of the effects of the Fragile X mutation on brain structure, using tensor-based morphometry. TBM visualizes structural brain deficits automatically, without time-consuming specification of regions-of-interest. We compared 36 subjects with FraX (age: 14.66+/-1.58 S.D., 18 females/18 males), and 33 age-matched healthy controls (age: 14.67+/-2.2 S.D., 17 females/16 males), using high-dimensional elastic image registration. All 69 subjects' 3D T1-weighted brain MRIs were spatially deformed to match a high-resolution single-subject average MRI scan in ICBM space, whose geometry was optimized to produce a minimal deformation target. Maps of the local Jacobian determinant (expansion factor) were computed from the deformation fields. Statistical maps showed increased caudate (10% higher; p = 0.001) and lateral ventricle volumes (19% higher; p = 0.003), and trend-level parietal and temporal white matter excesses (10% higher locally; p = 0.04). In affected females, volume abnormalities correlated with reduction in systemically measured levels of the Fragile X mental retardation protein (FMRP; Spearman's r < -0.5 locally). Decreased FMRP correlated with ventricular expansion (p = 0.042; permutation test), and anterior cingulate tissue reductions (p = 0.0026; permutation test) supporting theories that FMRP is required for normal dendritic pruning in fronto-striatal-limbic pathways. No sex differences were found; findings were confirmed using traditional volumetric measures in regions of interest. Deficit patterns were replicated by performing statistics after logarithmic transformation, which may be more appropriate for tensor-valued data. Investigation of how these anomalies emerge over time will accelerate our understanding of FraX and its treatment.
Collapse
Affiliation(s)
- Agatha D Lee
- Laboratory of Neuro Imaging, Department of Neurology, UCLA School of Medicine, 635 Charles E. Young Drive South, Suite 225E, Los Angeles, CA 90095-7332, USA
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Jacquemont S, Hagerman RJ, Hagerman PJ, Leehey MA. Fragile-X syndrome and fragile X-associated tremor/ataxia syndrome: two faces of FMR1. Lancet Neurol 2007; 6:45-55. [PMID: 17166801 DOI: 10.1016/s1474-4422(06)70676-7] [Citation(s) in RCA: 170] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Recent advances in our understanding of the clinical and molecular features of the fragile-X mental-retardation 1 gene, FMR1, highlight the importance of single-gene disorders. 15 years after its discovery, FMR1 continues to reveal new and unexpected clinical presentations and molecular mechanisms. Loss of function of FMR1 is a model for neurodevelopmental and behavioural disorders, including mental retardation, autism, anxiety, and mood instability. In addition, overexpression and CNS toxicity of FMR1 mRNA causes a late-onset neurodegenerative disorder, the fragile-X-associated tremor/ataxia syndrome (FXTAS). A similar mechanism is probably involved in premature ovarian failure, which affects up to 20% of female carriers of an altered FMR1 gene.
Collapse
Affiliation(s)
- Sebastien Jacquemont
- Service de Génétique, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland.
| | | | | | | |
Collapse
|
44
|
Belmonte MK, Bourgeron T. Fragile X syndrome and autism at the intersection of genetic and neural networks. Nat Neurosci 2006; 9:1221-5. [PMID: 17001341 DOI: 10.1038/nn1765] [Citation(s) in RCA: 213] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Autism, an entirely behavioral diagnosis with no largely understood etiologies and no population-wide biomarkers, contrasts with fragile X syndrome (FXS), a single-gene disorder with definite alterations of gene expression and neuronal morphology. Nevertheless, the behavioral overlap between autism and FXS suggests some overlapping mechanisms. Understanding how the single-gene alteration in FXS plays out within complex genetic and neural network processes may suggest targets for autism research and illustrate strategies for relating autism to more singular genetic syndromes.
Collapse
Affiliation(s)
- Matthew K Belmonte
- Department of Human Development, Cornell University, Martha Van Rensselaer Hall, Ithaca, New York 14853-4401, USA.
| | | |
Collapse
|
45
|
Berry-Kravis E, Krause SE, Block SS, Guter S, Wuu J, Leurgans S, Decle P, Potanos K, Cook E, Salt J, Maino D, Weinberg D, Lara R, Jardini T, Cogswell J, Johnson SA, Hagerman R. Effect of CX516, an AMPA-modulating compound, on cognition and behavior in fragile X syndrome: a controlled trial. J Child Adolesc Psychopharmacol 2006; 16:525-40. [PMID: 17069542 DOI: 10.1089/cap.2006.16.525] [Citation(s) in RCA: 108] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
A Phase II, 4-week randomized, double-blind, placebo-controlled clinical trial was conducted to evaluate the safety and efficacy of the Ampakine compound CX516 as a potential treatment for the underlying disorder in fragile X syndrome (FXS). After baseline screening, subjects with FXS (n = 49) underwent a 1-week placebo lead-in and then were randomized to study drug or placebo for a 4-week period. Cognitive and behavioral outcome measures were administered prior to treatment, at the end of treatment, and 2 weeks posttreatment. There were minimal side effects, no significant changes in safety parameters, and no serious adverse events. There was a 12.5% frequency of allergic rash in the CX516 group and 1 subject developed a substantial rash. There was also no significant improvement in memory, the primary outcome measure, or in secondary measures of language, attention/executive function, behavior, and overall functioning in CX516-treated subjects compared to placebo. This study did demonstrate that many outcome measures were reproducible in this test-retest setting for the FXS population, yet some were too difficult or variable. Adult subjects with FXS were able to complete an intensive clinical trial, and some valid outcome measures were identified for future FXS trial design. Problems with potency of CX516 in other studies have suggested dosing may have been inadequate for therapeutic effect and thus it remains unclear whether modulation of AMPA-mediated neurotransmission is a viable therapeutic strategy for the treatment of FXS.
Collapse
|
46
|
Restifo LL. Mental retardation genes in drosophila: New approaches to understanding and treating developmental brain disorders. ACTA ACUST UNITED AC 2006; 11:286-94. [PMID: 16240406 DOI: 10.1002/mrdd.20083] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Drosophila melanogaster is emerging as a valuable genetic model system for the study of mental retardation (MR). MR genes are remarkably similar between humans and fruit flies. Cognitive behavioral assays can detect reductions in learning and memory in flies with mutations in MR genes. Neuroanatomical methods, including some at single-neuron resolution, are helping to reveal the cellular bases of faulty brain development caused by MR gene mutations. Drosophila fragile X mental retardation 1 (dfmr1) is the fly counterpart of the human gene whose malfunction causes fragile X syndrome. Research on the fly gene is leading the field in molecular mechanisms of the gene product's biological function and in pharmacological rescue of brain and behavioral phenotypes. Future work holds the promise of using genetic pathway analysis and primary neuronal culture methods in Drosophila as tools for drug discovery for a wide range of MR and related disorders.
Collapse
Affiliation(s)
- Linda L Restifo
- ARL Division of Neurobiology, University of Arizona, and Department of Neurology, Arizona Health Sciences Center, Tucson Arizona 85721-0077, USA.
| |
Collapse
|
47
|
Gantois I, Vandesompele J, Speleman F, Reyniers E, D'Hooge R, Severijnen LA, Willemsen R, Tassone F, Kooy RF. Expression profiling suggests underexpression of the GABA(A) receptor subunit delta in the fragile X knockout mouse model. Neurobiol Dis 2005; 21:346-57. [PMID: 16199166 DOI: 10.1016/j.nbd.2005.07.017] [Citation(s) in RCA: 124] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2005] [Revised: 07/05/2005] [Accepted: 07/28/2005] [Indexed: 11/29/2022] Open
Abstract
It is still unclear why absence of the fragile X protein (FMRP) leads to mental retardation and specific behavioral problems. In neurons, the protein transports specific mRNAs towards the actively translating ribosomes near the synapses. To unravel the mechanism leading to the disorder, we performed global gene expression analysis by means of the differential display method using the fragile X mouse model. To verify differential expression, we used microarray technology and real-time PCR. Three differentially expressed cDNAs showed consistent underexpression in the fragile X knockout mouse, including a GABA(A) receptor subunit delta, a Rho guanine exchange factor 12 and an EST BU563433. In addition, we identified 5 genes that showed differential expression dependent on the sample of RNA analysis. We consider their differential expression as provisional. It is possible that these differentially expressed genes play an important role in the cognitive and behavioral problems observed in the fragile X syndrome.
Collapse
Affiliation(s)
- Ilse Gantois
- Department of Medical Genetics, University of Antwerp, Universiteitsplein 1, 2610 Antwerp, Belgium
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Abstract
PURPOSE OF REVIEW This review will describe recent developments in the neurobiology of fragile X syndrome (FXS), the association between FXS and autism, and involvement in premutation carriers. RECENT FINDINGS Metabotropic glutamate receptor 5 (mGluR5)-coupled pathways are dysregulated in individuals with FXS and this is thought to relate to the FXS phenotype. The mGluR5 model suggests that mGluR5 antagonists, including downstream effectors such as lithium, could be useful for treating FXS. Two forms of clinical involvement associated with the fragile X mental retardation 1 (FMR1) gene, autism and fragile X-associated tremor/ataxia syndrome (FXTAS), have received additional attention during the past year. One study has found that approximately 30% of individuals with FXS have autism; those with autism have lowered cognitive abilities, language problems, and behavioral difficulties compared to those with FXS alone. Furthermore, evidence is mounting that autism also occurs in some young males who have premutation alleles. Finally, males and occasional females with premutation alleles may develop a neurological syndrome with aging that consists of tremor, ataxia, peripheral neuropathy, and cognitive deficits. Significant brain atrophy and white-matter disease is usually seen. This new disorder (FXTAS) is thought to be related to elevated levels of abnormal FMR1 mRNA. SUMMARY Full-mutation forms of the gene (> 200 repeats) can cause autism, learning disabilities, anxiety disorders, and mental retardation. Disorders associated with premutation forms of the gene (55-200 repeats) include, in addition to autism, FXTAS in older males and females, and premature ovarian failure.
Collapse
Affiliation(s)
- Randi J Hagerman
- Department of Pediatrics, University of California at Davis Medical Center, M.I.N.D. Institute, Sacramento, CA 95817, USA.
| | | | | |
Collapse
|
49
|
Dong WK, Greenough WT. Plasticity of nonneuronal brain tissue: roles in developmental disorders. ACTA ACUST UNITED AC 2005; 10:85-90. [PMID: 15362161 DOI: 10.1002/mrdd.20016] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Neuronal and nonneuronal plasticity are both affected by environmental and experiential factors. Remodeling of existing neurons induced by such factors has been observed throughout the brain, and includes alterations in dendritic field dimensions, synaptogenesis, and synaptic morphology. The brain loci affected by these plastic neuronal changes are dependent on the type of experience and learning. Increased neurogenesis in the hippocampal dentate gyrus is a well-documented response to environmental complexity ("enrichment") and learning. Exposure to challenging experiences and learning opportunities also alters existing glial cells (i.e., astrocytes and oligodendrocytes), and up-regulates gliogenesis, in the cerebral cortex and cerebellum. Such glial plasticity often parallels neuronal remodeling in both time and place, and this enhanced morphological synergism may be important for optimizing the functional interaction between glial cells and neurons. Aberrant structural plasticity of nonneuronal elements is a contributing factor, as is aberrant neuron plasticity, to neurological and developmental disorders such as epilepsy, autism, and mental retardation (i.e., fragile X syndrome). Some of these nonneuronal pathologies include abnormal cerebral and cerebellar white matter and myelin-related proteins in autism; abnormal myelin basic protein in fragile X syndrome (FXS); and abnormal astrocytes in autism, FXS, and epilepsy. A number of recent studies demonstrate the possibility of using environmental and experiential intervention to reduce or ameliorate some of the neuronal and nonneuronal abnormalities, as well as behavioral deficits, present in these neurological and developmental disorders.
Collapse
Affiliation(s)
- Willie K Dong
- Neurotech Group, Beckman Institute, University of Illinios, Urbana, Illinois 61801, USA
| | | |
Collapse
|
50
|
Belmonte MK, Allen G, Beckel-Mitchener A, Boulanger LM, Carper RA, Webb SJ. Autism and abnormal development of brain connectivity. J Neurosci 2004; 24:9228-31. [PMID: 15496656 PMCID: PMC6730085 DOI: 10.1523/jneurosci.3340-04.2004] [Citation(s) in RCA: 789] [Impact Index Per Article: 39.5] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2004] [Revised: 09/01/2004] [Accepted: 09/02/2004] [Indexed: 11/21/2022] Open
Affiliation(s)
- Matthew K Belmonte
- Autism Research Centre, Department of Psychiatry, University of Cambridge, Cambridge CB2 2AH, United Kingdom
| | | | | | | | | | | |
Collapse
|