Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 106
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 180(6): 1178-1197.e20, 2020 03 19.
Article in English | MEDLINE | ID: mdl-32200800

ABSTRACT

Social impairment is frequently associated with mitochondrial dysfunction and altered neurotransmission. Although mitochondrial function is crucial for brain homeostasis, it remains unknown whether mitochondrial disruption contributes to social behavioral deficits. Here, we show that Drosophila mutants in the homolog of the human CYFIP1, a gene linked to autism and schizophrenia, exhibit mitochondrial hyperactivity and altered group behavior. We identify the regulation of GABA availability by mitochondrial activity as a biologically relevant mechanism and demonstrate its contribution to social behavior. Specifically, increased mitochondrial activity causes gamma aminobutyric acid (GABA) sequestration in the mitochondria, reducing GABAergic signaling and resulting in social deficits. Pharmacological and genetic manipulation of mitochondrial activity or GABA signaling corrects the observed abnormalities. We identify Aralar as the mitochondrial transporter that sequesters GABA upon increased mitochondrial activity. This study increases our understanding of how mitochondria modulate neuronal homeostasis and social behavior under physiopathological conditions.


Subject(s)
Calcium-Binding Proteins/metabolism , Drosophila Proteins/metabolism , Mitochondria/metabolism , gamma-Aminobutyric Acid/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Animals, Genetically Modified , Aspartic Acid/metabolism , Calcium/metabolism , Calcium-Binding Proteins/physiology , Drosophila Proteins/physiology , Drosophila melanogaster/metabolism , Glucose/metabolism , Homeostasis , Humans , Male , Mitochondria/genetics , Mitochondrial Membrane Transport Proteins/genetics , Mitochondrial Proteins/metabolism , Neurons/metabolism , Social Behavior , Synaptic Transmission , gamma-Aminobutyric Acid/genetics
2.
Cell ; 178(4): 769-771, 2019 08 08.
Article in English | MEDLINE | ID: mdl-31398332

ABSTRACT

Autism spectrum disorder (ASD) is prevalent, complex, and heterogeneous, and currently there is no cure. Identifying shared mechanisms across the ASD spectrum is of utmost importance for therapeutic intervention. Orefice et al. show that tackling the GABAA receptor pathway in the peripheral somatosensory system in various ASD mouse models rescues core ASD-like phenotypes.


Subject(s)
Autism Spectrum Disorder , Animals , Interneurons , Mice , Phenotype , Receptors, GABA-A
3.
Cell ; 159(1): 218-218.e1, 2014 Sep 25.
Article in English | MEDLINE | ID: mdl-25259928

ABSTRACT

The Fragile X syndrome, caused by the absence or mutation of fragile X mental retardation protein, FMRP, is a the common component of inherited intellectual disability and autism. This SnapShot surveys the protein interaction partners of FMRP, focusing on the cellular pathways in which they are involved.


Subject(s)
Fragile X Mental Retardation Protein/metabolism , Animals , Fragile X Syndrome/metabolism , Humans , Protein Interaction Maps , RNA, Messenger/metabolism
4.
Cell ; 158(6): 1446-1446.e1, 2014 Sep 11.
Article in English | MEDLINE | ID: mdl-25215498

ABSTRACT

FMRP, or fragile X mental retardation protein is an RNA-binding protein. Mutations in the FMRP protein have been associated with neurological disease as have a number of its mRNA-binding targets. This SnapShot presents 40 bona fide FMRP targets for which mRNA binding and protein regulation have been robustly reported in mammals along with the diseases with which they have been associated.


Subject(s)
Brain Diseases/genetics , Fragile X Mental Retardation Protein/metabolism , Mental Disorders/genetics , RNA, Messenger/metabolism , Animals , Brain Diseases/metabolism , Humans , Mental Disorders/metabolism , Neoplasms/genetics , Neoplasms/metabolism
5.
EMBO J ; 39(20): e104467, 2020 10 15.
Article in English | MEDLINE | ID: mdl-32706158

ABSTRACT

Nucleoporins (Nups) build highly organized nuclear pore complexes (NPCs) at the nuclear envelope (NE). Several Nups assemble into a sieve-like hydrogel within the central channel of the NPCs. In the cytoplasm, the soluble Nups exist, but how their assembly is restricted to the NE is currently unknown. Here, we show that fragile X-related protein 1 (FXR1) can interact with several Nups and facilitate their localization to the NE during interphase through a microtubule-dependent mechanism. Downregulation of FXR1 or closely related orthologs FXR2 and fragile X mental retardation protein (FMRP) leads to the accumulation of cytoplasmic Nup condensates. Likewise, models of fragile X syndrome (FXS), characterized by a loss of FMRP, accumulate Nup granules. The Nup granule-containing cells show defects in protein export, nuclear morphology and cell cycle progression. Our results reveal an unexpected role for the FXR protein family in the spatial regulation of nucleoporin condensation.


Subject(s)
Cell Nucleus/metabolism , Fragile X Mental Retardation Protein/metabolism , Fragile X Syndrome/metabolism , Microtubules/metabolism , Nuclear Envelope/metabolism , Nuclear Pore Complex Proteins/metabolism , RNA-Binding Proteins/metabolism , Acrylates/pharmacology , Animals , Cell Line , Cytoplasm/drug effects , Cytoplasm/metabolism , Down-Regulation , Fragile X Mental Retardation Protein/genetics , Fragile X Syndrome/genetics , G1 Phase Cell Cycle Checkpoints/genetics , Humans , In Situ Hybridization, Fluorescence , Interphase/genetics , Mice , Microscopy, Electron, Transmission , Microtubules/drug effects , Microtubules/ultrastructure , Myoblasts/drug effects , Myoblasts/metabolism , Nuclear Envelope/drug effects , Nuclear Envelope/ultrastructure , Nuclear Pore Complex Proteins/genetics , RNA, Small Interfering , RNA-Binding Proteins/genetics
6.
EMBO Rep ; 22(4): e51404, 2021 04 07.
Article in English | MEDLINE | ID: mdl-33779029

ABSTRACT

Status epilepticus (SE) is a condition in which seizures are not self-terminating and thereby pose a serious threat to the patient's life. The molecular mechanisms underlying SE are likely heterogeneous and not well understood. Here, we reveal a role for the RNA-binding protein Fragile X-Related Protein 2 (FXR2P) in SE. Fxr2 KO mice display reduced sensitivity specifically to kainic acid-induced SE. Immunoprecipitation of FXR2P coupled to next-generation sequencing of associated mRNAs shows that FXR2P targets are enriched in genes that encode glutamatergic post-synaptic components. Of note, the FXR2P target transcriptome has a significant overlap with epilepsy and SE risk genes. In addition, Fxr2 KO mice fail to show sustained ERK1/2 phosphorylation induced by KA and present reduced burst activity in the hippocampus. Taken together, our findings show that the absence of FXR2P decreases the expression of glutamatergic proteins, and this decrease might prevent self-sustained seizures.


Subject(s)
Kainic Acid , Status Epilepticus , Animals , Hippocampus/metabolism , Kainic Acid/toxicity , Mice , Mice, Inbred C57BL , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Seizures/chemically induced , Seizures/genetics , Status Epilepticus/chemically induced , Status Epilepticus/genetics
7.
Cell ; 134(6): 1042-54, 2008 Sep 19.
Article in English | MEDLINE | ID: mdl-18805096

ABSTRACT

Strong evidence indicates that regulated mRNA translation in neuronal dendrites underlies synaptic plasticity and brain development. The fragile X mental retardation protein (FMRP) is involved in this process; here, we show that it acts by inhibiting translation initiation. A binding partner of FMRP, CYFIP1/Sra1, directly binds the translation initiation factor eIF4E through a domain that is structurally related to those present in 4E-BP translational inhibitors. Brain cytoplasmic RNA 1 (BC1), another FMRP binding partner, increases the affinity of FMRP for the CYFIP1-eIF4E complex in the brain. Levels of proteins encoded by known FMRP target mRNAs are increased upon reduction of CYFIP1 in neurons. Translational repression is regulated in an activity-dependent manner because BDNF or DHPG stimulation of neurons causes CYFIP1 to dissociate from eIF4E at synapses, thereby resulting in protein synthesis. Thus, the translational repression activity of FMRP in the brain is mediated, at least in part, by CYFIP1.


Subject(s)
Brain/metabolism , Fragile X Mental Retardation Protein/metabolism , Nerve Tissue Proteins/metabolism , Protein Biosynthesis , Adaptor Proteins, Signal Transducing , Amino Acid Sequence , Animals , Brain/embryology , Cells, Cultured , Eukaryotic Initiation Factor-4E/genetics , Eukaryotic Initiation Factor-4E/metabolism , Fragile X Mental Retardation Protein/chemistry , Fragile X Mental Retardation Protein/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Molecular , Molecular Sequence Data , Nerve Tissue Proteins/genetics , Neurons/metabolism , Sequence Alignment , Synapses
8.
J Neurosci ; 40(9): 1819-1833, 2020 02 26.
Article in English | MEDLINE | ID: mdl-31964717

ABSTRACT

Dendritic arborization is highly regulated and requires tight control of dendritic growth, branching, cytoskeletal dynamics, and ion channel expression to ensure proper function. Abnormal dendritic development can result in altered network connectivity, which has been linked to neurodevelopmental disorders, including autism spectrum disorders (ASDs). How neuronal growth control programs tune dendritic arborization to ensure function is still not fully understood. Using Drosophila dendritic arborization (da) neurons as a model, we identified the conserved Ste20-like kinase Tao as a negative regulator of dendritic arborization. We show that Tao kinase activity regulates cytoskeletal dynamics and sensory channel localization required for proper sensory function in both male and female flies. We further provide evidence for functional conservation of Tao kinase, showing that its ASD-linked human ortholog, Tao kinase 2 (Taok2), could replace Drosophila Tao and rescue dendritic branching, dynamic microtubule alterations, and behavioral defects. However, several ASD-linked Taok2 variants displayed impaired rescue activity, suggesting that Tao/Taok2 mutations can disrupt sensory neuron development and function. Consistently, we show that Tao kinase activity is required in developing and as well as adult stages for maintaining normal dendritic arborization and sensory function to regulate escape and social behavior. Our data suggest an important role for Tao kinase signaling in cytoskeletal organization to maintain proper dendritic arborization and sensory function, providing a strong link between developmental sensory aberrations and behavioral abnormalities relevant for Taok2-dependent ASDs.SIGNIFICANCE STATEMENT Autism spectrum disorders (ASDs) are linked to abnormal dendritic arbors. However, the mechanisms of how dendritic arbors develop to promote functional and proper behavior are unclear. We identified Drosophila Tao kinase, the ortholog of the ASD risk gene Taok2, as a regulator of dendritic arborization in sensory neurons. We show that Tao kinase regulates cytoskeletal dynamics, controls sensory ion channel localization, and is required to maintain somatosensory function in vivo Interestingly, ASD-linked human Taok2 mutations rendered it nonfunctional, whereas its WT form could restore neuronal morphology and function in Drosophila lacking endogenous Tao. Our findings provide evidence for a conserved role of Tao kinase in dendritic development and function of sensory neurons, suggesting that aberrant sensory function might be a common feature of ASDs.


Subject(s)
Cytoskeleton/physiology , Dendrites/physiology , Drosophila Proteins/genetics , Drosophila Proteins/physiology , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/physiology , Sensation/physiology , Actins/metabolism , Animals , Animals, Genetically Modified , Cytoskeleton/ultrastructure , Dendrites/ultrastructure , Drosophila , Escape Reaction , Female , Humans , Male , Mechanoreceptors/physiology , Mutation/genetics , Social Behavior
9.
Hum Mol Genet ; 27(12): 2039-2051, 2018 06 15.
Article in English | MEDLINE | ID: mdl-29590342

ABSTRACT

Fragile X syndrome (FXS) is a monogenic form of intellectual disability and autism spectrum disorder caused by the absence of the fragile X mental retardation protein (FMRP). In biological models for the disease, this leads to upregulated mRNA translation and as a consequence, deficits in synaptic architecture and plasticity. Preclinical studies revealed that pharmacological interventions restore those deficits, which are thought to mediate the FXS cognitive and behavioral symptoms. Here, we characterized the de novo rate of protein synthesis in patients with FXS and their relationship with clinical severity. We measured the rate of protein synthesis in fibroblasts derived from 32 individuals with FXS and from 17 controls as well as in fibroblasts and primary neurons of 27 Fmr1 KO mice and 20 controls. Here, we show that levels of protein synthesis are increased in fibroblasts of individuals with FXS and Fmr1 KO mice. However, this cellular phenotype displays a broad distribution and a proportion of fragile X individuals and Fmr1 KO mice do not show increased levels of protein synthesis, having measures in the normal range. Because the same Fmr1 KO animal measures in fibroblasts predict those in neurons we suggest the validity of this peripheral biomarker. Our study offers a potential explanation for the comprehensive drug development program undertaken thus far yielding negative results and suggests that a significant proportion, but not all individuals with FXS, may benefit from the reduction of excessive levels of protein synthesis.


Subject(s)
Autism Spectrum Disorder/genetics , Fragile X Mental Retardation Protein/genetics , Fragile X Syndrome/genetics , Adolescent , Adult , Aged , Animals , Autism Spectrum Disorder/physiopathology , Child , Disease Models, Animal , Female , Fibroblasts/metabolism , Fibroblasts/pathology , Fragile X Mental Retardation Protein/biosynthesis , Fragile X Syndrome/physiopathology , Hippocampus/metabolism , Hippocampus/physiopathology , Humans , Male , Mice , Mice, Knockout , Middle Aged , Neurons/metabolism , Neurons/pathology , Young Adult
10.
Annu Rev Genomics Hum Genet ; 18: 115-142, 2017 08 31.
Article in English | MEDLINE | ID: mdl-28859574

ABSTRACT

The Ras-MAPK and PI3K-AKT-mTOR signaling cascades were originally identified as cancer regulatory pathways but have now been demonstrated to be critical for synaptic plasticity and behavior. Neurodevelopmental disorders arising from mutations in these pathways exhibit related neurological phenotypes, including cognitive dysfunction, autism, and intellectual disability. The downstream targets of these pathways include regulation of transcription and protein synthesis. Other disorders that affect protein translation include fragile X syndrome (an important cause of syndromal autism), and other translational regulators are now also linked to autism. Here, we review how mechanisms of synaptic plasticity have been revealed by studies of mouse models for Ras-MAPK, PI3K-AKT-mTOR, and translation regulatory pathway disorders. We discuss the face validity of these mouse models and review current progress in clinical trials directed at ameliorating cognitive and behavioral symptoms.


Subject(s)
Cognitive Dysfunction/physiopathology , Intellectual Disability/metabolism , Intellectual Disability/psychology , Neuronal Plasticity , Signal Transduction , Animals , Cognitive Dysfunction/metabolism , Disease Models, Animal , Humans , Intellectual Disability/physiopathology , Mice , TOR Serine-Threonine Kinases
11.
J Gen Virol ; 100(5): 863-876, 2019 05.
Article in English | MEDLINE | ID: mdl-30896385

ABSTRACT

The detection of antisense RNA is hampered by reverse transcription (RT) non-specific priming, due to the ability of RNA secondary structures to prime RT in the absence of specific primers. The detection of antisense RNA by conventional RT-PCR does not allow assessment of the polarity of the initial RNA template, causing the amplification of non-specific cDNAs. In this study we have developed a modified protocol for the detection of human immunodeficiency virus type 1 (HIV-1) antisense protein (ASP) RNA. Using this approach, we have identified ASP transcripts in CD4+ T cells isolated from five HIV-infected individuals, either untreated or under suppressive therapy. We show that ASP RNA can be detected in stimulated CD4+ T cells from both groups of patients, but not in unstimulated cells. We also show that in untreated patients, the patterns of expression of ASP and env are very similar, with the levels of ASP RNA being markedly lower than those of env. Treatment of cells from one viraemic patient with α-amanitin greatly reduces the rate of ASP RNA synthesis, suggesting that it is associated with RNA polymerase II, the central enzyme in the transcription of protein-coding genes. Our data represent the first nucleotide sequences obtained in patients for ASP, demonstrating that its transcription indeed occurs in those HIV-1 lineages in which the ASP open reading frame is present.


Subject(s)
HIV Infections/virology , HIV-1/genetics , RNA, Antisense/genetics , RNA, Viral/genetics , Adult , Base Sequence/genetics , CD4-Positive T-Lymphocytes/virology , Gene Expression Regulation, Viral/genetics , Humans , Male , Middle Aged , Open Reading Frames/genetics , Virus Replication/genetics , Young Adult
12.
J Cell Sci ; 128(9): 1787-99, 2015 May 01.
Article in English | MEDLINE | ID: mdl-25788698

ABSTRACT

A common feature of non-coding repeat expansion disorders is the accumulation of RNA repeats as RNA foci in the nucleus and/or cytoplasm of affected cells. These RNA foci can be toxic because they sequester RNA-binding proteins, thus affecting various steps of post-transcriptional gene regulation. However, the precise step that is affected by C9orf72 GGGGCC (G4C2) repeat expansion, the major genetic cause of amyotrophic lateral sclerosis (ALS), is still poorly defined. In this work, we set out to characterise these mechanisms by identifying proteins that bind to C9orf72 RNA. Sequestration of some of these factors into RNA foci was observed when a (G4C2)31 repeat was expressed in NSC34 and HeLa cells. Most notably, (G4C2)31 repeats widely affected the distribution of Pur-alpha and its binding partner fragile X mental retardation protein 1 (FMRP, also known as FMR1), which accumulate in intra-cytosolic granules that are positive for stress granules markers. Accordingly, translational repression is induced. Interestingly, this effect is associated with a marked accumulation of poly(A) mRNAs in cell nuclei. Thus, defective trafficking of mRNA, as a consequence of impaired nuclear mRNA export, might affect translation efficiency and contribute to the pathogenesis of C9orf72 ALS.


Subject(s)
Amyotrophic Lateral Sclerosis/metabolism , Cell Nucleus/metabolism , Models, Biological , Protein Biosynthesis , Proteins/metabolism , Trinucleotide Repeat Expansion , Amyotrophic Lateral Sclerosis/pathology , Animals , C9orf72 Protein , DNA-Binding Proteins , Eukaryotic Initiation Factor-2/metabolism , Fragile X Mental Retardation Protein/metabolism , HeLa Cells , Humans , Intracellular Space/metabolism , Mice , Motor Neurons/metabolism , Phosphorylation , Poly(A)-Binding Proteins/metabolism , Protein Binding , RNA Splicing/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transcription Factors
13.
RNA Biol ; 14(11): 1580-1591, 2017 11 02.
Article in English | MEDLINE | ID: mdl-28640668

ABSTRACT

The fragile X syndrome (FXS), the most common form of inherited intellectual disability, is due to the absence of FMRP, a protein regulating RNA metabolism. Recently, an unexpected function of FMRP in modulating the activity of Adenosine Deaminase Acting on RNA (ADAR) enzymes has been reported both in Drosophila and Zebrafish. ADARs are RNA-binding proteins that increase transcriptional complexity through a post-transcriptional mechanism called RNA editing. To evaluate the ADAR2-FMRP interaction in mammals we analyzed several RNA editing re-coding sites in the fmr1 knockout (KO) mice. Ex vivo and in vitro analysis revealed that absence of FMRP leads to an increase in the editing levels of brain specific mRNAs, indicating that FMRP might act as an inhibitor of editing activity. Proximity Ligation Assay (PLA) in mouse primary cortical neurons and in non-neuronal cells revealed that ADAR2 and FMRP co-localize in the nucleus. The ADAR2-FMRP co-localization was further observed by double-immunogold Electron Microscopy (EM) in the hippocampus. Moreover, ADAR2-FMRP interaction appeared to be RNA independent. Because changes in the editing pattern are associated with neuropsychiatric and neurodevelopmental disorders, we propose that the increased editing observed in the fmr1-KO mice might contribute to the FXS molecular phenotypes.


Subject(s)
Adenosine Deaminase/genetics , Fragile X Mental Retardation Protein/genetics , Fragile X Syndrome/genetics , Neurons/metabolism , RNA Editing , RNA, Messenger/genetics , RNA-Binding Proteins/genetics , Adenosine Deaminase/metabolism , Animals , Cell Nucleus/metabolism , Cell Nucleus/ultrastructure , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Disease Models, Animal , Fragile X Mental Retardation Protein/metabolism , Fragile X Syndrome/metabolism , Fragile X Syndrome/pathology , Gene Deletion , Hippocampus/metabolism , Hippocampus/pathology , Humans , Male , Mice , Mice, Knockout , Neurons/pathology , Phenotype , Primary Cell Culture , Protein Binding , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism
14.
Proc Natl Acad Sci U S A ; 111(36): 13205-10, 2014 Sep 09.
Article in English | MEDLINE | ID: mdl-25157163

ABSTRACT

Protein ubiquitination is a core regulatory determinant of neural development. Previous studies have indicated that the Nedd4-family E3 ubiquitin ligases Nedd4-1 and Nedd4-2 may ubiquitinate phosphatase and tensin homolog (PTEN) and thereby regulate axonal growth in neurons. Using conditional knockout mice, we show here that Nedd4-1 and Nedd4-2 are indeed required for axonal growth in murine central nervous system neurons. However, in contrast to previously published data, we demonstrate that PTEN is not a substrate of Nedd4-1 and Nedd4-2, and that aberrant PTEN ubiquitination is not involved in the impaired axon growth upon deletion of Nedd4-1 and Nedd4-2. Rather, PTEN limits Nedd4-1 protein levels by modulating the activity of mTORC1, a protein complex that controls protein synthesis and cell growth. Our data demonstrate that Nedd4-family E3 ligases promote axonal growth and branching in the developing mammalian brain, where PTEN is not a relevant substrate. Instead, PTEN controls neurite growth by regulating Nedd4-1 expression.


Subject(s)
Endosomal Sorting Complexes Required for Transport/metabolism , Multiprotein Complexes/metabolism , Neurites/metabolism , PTEN Phosphohydrolase/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Axons/metabolism , Cerebral Cortex/cytology , Hippocampus/cytology , Mechanistic Target of Rapamycin Complex 1 , Mice, Knockout , Models, Biological , Morphogenesis , Nedd4 Ubiquitin Protein Ligases , Polyubiquitin/metabolism , Protein Biosynthesis , Ubiquitination
15.
J Neurosci ; 35(25): 9402-8, 2015 Jun 24.
Article in English | MEDLINE | ID: mdl-26109663

ABSTRACT

In brain, specific RNA-binding proteins (RBPs) associate with localized mRNAs and function as regulators of protein synthesis at synapses exerting an indirect control on neuronal activity. Thus, the Fragile X Mental Retardation protein (FMRP) regulates expression of the scaffolding postsynaptic density protein PSD95, but the mode of control appears to be different from other FMRP target mRNAs. Here, we show that the fragile X mental retardation-related protein 2 (FXR2P) cooperates with FMRP in binding to the 3'-UTR of mouse PSD95/Dlg4 mRNA. Absence of FXR2P leads to decreased translation of PSD95/Dlg4 mRNA in the hippocampus, implying a role for FXR2P as translation activator. Remarkably, mGluR-dependent increase of PSD95 synthesis is abolished in neurons lacking Fxr2. Together, these findings show a coordinated regulation of PSD95/Dlg4 mRNA by FMRP and FXR2P that ultimately affects its fine-tuning during synaptic activity.


Subject(s)
Gene Expression Regulation/physiology , Guanylate Kinases/biosynthesis , Membrane Proteins/biosynthesis , Neuronal Plasticity/physiology , Neurons/metabolism , RNA-Binding Proteins/metabolism , Animals , Blotting, Western , Disks Large Homolog 4 Protein , Guanylate Kinases/genetics , Immunohistochemistry , Immunoprecipitation , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Protein Biosynthesis/physiology , RNA-Binding Proteins/genetics
16.
Neurobiol Dis ; 96: 201-215, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27616423

ABSTRACT

Fragile X syndrome (FXS) is a common inherited form of intellectual disability caused by the absence or reduction of the fragile X mental retardation protein (FMRP) encoded by the FMR1 gene. In humans, one symptom of FXS is hypersensitivity to sensory stimuli, including touch. We used a mouse model of FXS (Fmr1 KO) to study sensory processing of tactile information conveyed via the whisker system. In vivo electrophysiological recordings in somatosensory barrel cortex showed layer-specific broadening of the receptive fields at the level of layer 2/3 but not layer 4, in response to whisker stimulation. Furthermore, the encoding of tactile stimuli at different frequencies was severely affected in layer 2/3. The behavioral effect of this broadening of the receptive fields was tested in the gap-crossing task, a whisker-dependent behavioral paradigm. In this task the Fmr1 KO mice showed differences in the number of whisker contacts with platforms, decrease in the whisker sampling duration and reduction in the whisker touch-time while performing the task. We propose that the increased excitability in the somatosensory barrel cortex upon whisker stimulation may contribute to changes in the whisking strategy as well as to other observed behavioral phenotypes related to tactile processing in Fmr1 KO mice.


Subject(s)
Afferent Pathways/pathology , Fragile X Mental Retardation Protein/metabolism , Fragile X Syndrome/pathology , Somatosensory Cortex/pathology , Touch/physiology , Vibrissae/innervation , Animals , Disease Models, Animal , Fragile X Mental Retardation Protein/genetics , Fragile X Syndrome/complications , Fragile X Syndrome/genetics , Humans , Locomotion/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Reaction Time/genetics
18.
Learn Mem ; 21(10): 543-55, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25227249

ABSTRACT

The Fragile X syndrome (FXS) is the most frequent form of inherited mental disability and is considered a monogenic cause of autism spectrum disorder. FXS is caused by a triplet expansion that inhibits the expression of the FMR1 gene. The gene product, the Fragile X Mental Retardation Protein (FMRP), regulates mRNA metabolism in brain and nonneuronal cells. During brain development, FMRP controls the expression of key molecules involved in receptor signaling, cytoskeleton remodeling, protein synthesis and, ultimately, spine morphology. Symptoms associated with FXS include neurodevelopmental delay, cognitive impairment, anxiety, hyperactivity, and autistic-like behavior. Twenty years ago the first Fmr1 KO mouse to study FXS was generated, and several years later other key models including the mutant Drosophila melanogaster, dFmr1, have further helped the understanding of the cellular and molecular causes behind this complex syndrome. Here, we review to which extent these biological models are affected by the absence of FMRP, pointing out the similarities with the observed human dysfunction. Additionally, we discuss several potential treatments under study in animal models that are able to partially revert some of the FXS abnormalities.


Subject(s)
Disease Models, Animal , Drosophila Proteins/genetics , Fragile X Mental Retardation Protein/genetics , Fragile X Syndrome/genetics , Fragile X Syndrome/physiopathology , Learning/physiology , Animals , Anxiety/genetics , Circadian Rhythm/genetics , Drosophila melanogaster , Humans , Mice , Mice, Knockout , Sensory Gating/genetics , Signal Transduction/genetics , Social Behavior
19.
J Neurosci ; 33(46): 18234-41, 2013 Nov 13.
Article in English | MEDLINE | ID: mdl-24227732

ABSTRACT

Activity-dependent protein synthesis at synapses is dysregulated in the Fragile X syndrome (FXS). This process contributes to dendritic spine dysmorphogenesis and synaptic dysfunction in FXS. Matrix Metalloproteinase 9 (MMP-9) is an enzyme involved in activity-dependent reorganization of dendritic spine architecture and was shown to regulate spine morphology in a mouse model of FXS, the Fmr1 knock-out mice. Here we show that MMP-9 mRNA is part of the FMRP complex and colocalizes in dendrites. In the absence of FMRP MMP-9 mRNA translation is increased at synapses, suggesting that this mechanism contributes to the increased metalloproteinase level at synapses of Fmr1 knock-out mice. We propose that such a local effect can contribute to the aberrant dendritic spine morphology observed in the Fmr1 knock-out mice and in patients with FXS.


Subject(s)
Fragile X Mental Retardation Protein/physiology , Matrix Metalloproteinase 9/biosynthesis , RNA, Messenger/biosynthesis , Synapses/enzymology , Animals , Dendrites/enzymology , Dendrites/genetics , Female , Hippocampus/enzymology , Matrix Metalloproteinase 9/genetics , Mice , Mice, Knockout , RNA, Messenger/genetics , Rats , Synapses/genetics
20.
Hum Mol Genet ; 21(10): 2143-56, 2012 May 15.
Article in English | MEDLINE | ID: mdl-22328088

ABSTRACT

Fragile X syndrome (FXS) is the most common inherited form of intellectual disability and results from the loss of the fragile X mental retardation protein (FMRP). Many fragile X-related cognitive and behavioral features emerge during childhood and are associated with abnormal synaptic and cellular organization of the cerebral cortex. Identifying the roles of FMRP in cortical development will provide a basis for understanding the pathogenesis of the syndrome. However, how the loss of FMRP influences the developmental trajectory of cortical maturation remains unclear. We took advantage of the stereotyped and well-characterized development of the murine primary somatosensory cortex to examine cortical maturation during a time-window that corresponds to late embryonic and early postnatal development in the human. In the Fmr1 knockout mouse, we find a delay in somatosensory map formation, alterations in the morphology profile of dendrites and spines of layer 4 neurons and a decrease in the synaptic levels of proteins involved in glutamate receptor signaling at times corresponding to the highest levels of FMRP expression. In contrast, cortical arealization, synaptic density in layer 4 and early postnatal regulation of mRNAs encoding synaptic proteins are not altered in Fmr1 knockout mice. The specificity of the developmental delay in Fmr1 knockout mice indicates that the loss of FMRP does not result in a general stalling of cerebral cortex maturation. Instead, our results suggest that inaccurate timing of developmental processes caused by the loss of FMRP may lead to alterations in neural circuitry that underlie behavioral and cognitive dysfunctions associated with FXS.


Subject(s)
Fragile X Mental Retardation Protein/genetics , Fragile X Syndrome/metabolism , Somatosensory Cortex/metabolism , Animals , Disease Models, Animal , Fragile X Mental Retardation Protein/metabolism , Fragile X Syndrome/genetics , Mice , Mice, Knockout , Microscopy, Electron , RNA, Messenger/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL