Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
1.
Biol Psychiatry ; 95(2): 161-174, 2024 Jan 15.
Article in English | MEDLINE | ID: mdl-37704042

ABSTRACT

BACKGROUND: 15q11.2 deletions and duplications have been linked to autism spectrum disorder, schizophrenia, and intellectual disability. Recent evidence suggests that dysfunctional CYFIP1 (cytoplasmic FMR1 interacting protein 1) contributes to the clinical phenotypes observed in individuals with 15q11.2 deletion/duplication syndrome. CYFIP1 plays crucial roles in neuronal development and brain connectivity, promoting actin polymerization and regulating local protein synthesis. However, information about the impact of single nucleotide variants in CYFIP1 on neurodevelopmental disorders is limited. METHODS: Here, we report a family with 2 probands exhibiting intellectual disability, autism spectrum disorder, spastic tetraparesis, and brain morphology defects and who carry biallelic missense point mutations in the CYFIP1 gene. We used skin fibroblasts from one of the probands, the parents, and typically developing individuals to investigate the effect of the variants on the functionality of CYFIP1. In addition, we generated Drosophila knockin mutants to address the effect of the variants in vivo and gain insight into the molecular mechanism that underlies the clinical phenotype. RESULTS: Our study revealed that the 2 missense variants are in protein domains responsible for maintaining the interaction within the wave regulatory complex. Molecular and cellular analyses in skin fibroblasts from one proband showed deficits in actin polymerization. The fly model for these mutations exhibited abnormal brain morphology and F-actin loss and recapitulated the core behavioral symptoms, such as deficits in social interaction and motor coordination. CONCLUSIONS: Our findings suggest that the 2 CYFIP1 variants contribute to the clinical phenotype in the probands that reflects deficits in actin-mediated brain development processes.


Subject(s)
Autism Spectrum Disorder , Intellectual Disability , Humans , Intellectual Disability/genetics , Actins/genetics , Actins/metabolism , Autism Spectrum Disorder/genetics , Autism Spectrum Disorder/metabolism , Polymerization , Adaptor Proteins, Signal Transducing/genetics , Fragile X Mental Retardation Protein/metabolism
2.
Trends Pharmacol Sci ; 44(12): 963-977, 2023 12.
Article in English | MEDLINE | ID: mdl-37940430

ABSTRACT

Autism spectrum disorder (ASD) is a complex neurodevelopmental condition with uncertain origins. Understanding of the mechanisms underlying ASD remains limited, and treatments are lacking. Genetic diversity complicates drug development. Given the complexity and severity of ASD symptoms and the rising number of diagnoses, exploring novel therapeutic strategies is essential. Here, we focus on shared molecular pathways between ASD and cancer and highlight recent progress on the repurposing of cancer drugs for ASD treatment, such as mTOR inhibitors, histone deacetylase inhibitors, and anti-inflammatory agents. We discuss how to improve trial design considering drug dose and patient age. Lastly, the discussion explores the critical aspects of side effects, commercial factors, and the efficiency of drug-screening pipelines; all of which are essential considerations in the pursuit of repurposing cancer drugs for addressing core features of ASD.


Subject(s)
Antineoplastic Agents , Autism Spectrum Disorder , Neoplasms , Humans , Autism Spectrum Disorder/drug therapy , Autism Spectrum Disorder/diagnosis , Autism Spectrum Disorder/genetics , Drug Repositioning , Drug Development , Drug Evaluation, Preclinical , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Neoplasms/drug therapy
3.
Neuron ; 111(11): 1760-1775.e8, 2023 06 07.
Article in English | MEDLINE | ID: mdl-36996810

ABSTRACT

The proteome of glutamatergic synapses is diverse across the mammalian brain and involved in neurodevelopmental disorders (NDDs). Among those is fragile X syndrome (FXS), an NDD caused by the absence of the functional RNA-binding protein FMRP. Here, we demonstrate how the brain region-specific composition of postsynaptic density (PSD) contributes to FXS. In the striatum, the FXS mouse model shows an altered association of the PSD with the actin cytoskeleton, reflecting immature dendritic spine morphology and reduced synaptic actin dynamics. Enhancing actin turnover with constitutively active RAC1 ameliorates these deficits. At the behavioral level, the FXS model displays striatal-driven inflexibility, a typical feature of FXS individuals, which is rescued by exogenous RAC1. Striatal ablation of Fmr1 is sufficient to recapitulate behavioral impairments observed in the FXS model. These results indicate that dysregulation of synaptic actin dynamics in the striatum, a region largely unexplored in FXS, contributes to the manifestation of FXS behavioral phenotypes.


Subject(s)
Fragile X Syndrome , Animals , Mice , Fragile X Mental Retardation Protein/genetics , Actins/metabolism , Brain/metabolism , Disease Models, Animal , Mice, Knockout , Dendritic Spines/metabolism , Mammals/metabolism
4.
Nat Commun ; 14(1): 763, 2023 02 20.
Article in English | MEDLINE | ID: mdl-36808152

ABSTRACT

Sleep behavior is conserved throughout evolution, and sleep disturbances are a frequent comorbidity of neuropsychiatric disorders. However, the molecular basis underlying sleep dysfunctions in neurological diseases remains elusive. Using a model for neurodevelopmental disorders (NDDs), the Drosophila Cytoplasmic FMR1 interacting protein haploinsufficiency (Cyfip85.1/+), we identify a mechanism modulating sleep homeostasis. We show that increased activity of the sterol regulatory element-binding protein (SREBP) in Cyfip85.1/+ flies induces an increase in the transcription of wakefulness-associated genes, such as the malic enzyme (Men), causing a disturbance in the daily NADP+/NADPH ratio oscillations and reducing sleep pressure at the night-time onset. Reduction in SREBP or Men activity in Cyfip85.1/+ flies enhances the NADP+/NADPH ratio and rescues the sleep deficits, indicating that SREBP and Men are causative for the sleep deficits in Cyfip heterozygous flies. This work suggests modulation of the SREBP metabolic axis as a new avenue worth exploring for its therapeutic potential in sleep disorders.


Subject(s)
Drosophila Proteins , Sterol Regulatory Element Binding Proteins , Animals , Sterol Regulatory Element Binding Proteins/metabolism , NADP/metabolism , Drosophila/metabolism , Sterol Regulatory Element Binding Protein 1/metabolism , Sleep , Sterol Regulatory Element Binding Protein 2/metabolism , Drosophila Proteins/metabolism , Fragile X Mental Retardation Protein
5.
Cell Death Dis ; 13(8): 719, 2022 08 18.
Article in English | MEDLINE | ID: mdl-35982038

ABSTRACT

Converging evidence indicates that the Fragile X Messenger Ribonucleoprotein (FMRP), which absent or mutated in Fragile X Syndrome (FXS), plays a role in many types of cancers. However, while FMRP roles in brain development and function have been extensively studied, its involvement in the biology of brain tumors remains largely unexplored. Here we show, in human glioblastoma (GBM) biopsies, that increased expression of FMRP directly correlates with a worse patient outcome. In contrast, reductions in FMRP correlate with a diminished tumor growth and proliferation of human GBM stem-like cells (GSCs) in vitro in a cell culture model and in vivo in mouse brain GSC xenografts. Consistently, increased FMRP levels promote GSC proliferation. To characterize the mechanism(s) by which FMRP regulates GSC proliferation, we performed GSC transcriptome analyses in GSCs expressing high levels of FMRP, and in these GSCs after knockdown of FMRP. We show that the WNT signalling is the most significantly enriched among the published FMRP target genes and genes involved in ASD. Consistently, we find that reductions in FMRP downregulate both the canonical WNT/ß-Catenin and the non-canonical WNT-ERK1/2 signalling pathways, reducing the stability of several key transcription factors (i.e. ß-Catenin, CREB and ETS1) previously implicated in the modulation of malignant features of glioma cells. Our findings support a key role for FMRP in GBM cancer progression, acting via regulation of WNT signalling.


Subject(s)
Brain Neoplasms , Fragile X Mental Retardation Protein/metabolism , Glioblastoma , Animals , Brain Neoplasms/pathology , Cell Line, Tumor , Glioblastoma/pathology , Humans , Mice , Neoplastic Stem Cells/metabolism , Ribonucleoproteins , Wnt Signaling Pathway/genetics , beta Catenin/metabolism
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.
Neuroscience ; 445: 12-30, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32730949

ABSTRACT

Neurodevelopmental disorders (NDDs) include a large number of conditions such as Fragile  X  syndrome, autism spectrum disorders and Down syndrome, among others. They are characterized by limitations in adaptive and social behaviors, as well as intellectual disability (ID). Whole-exome and whole-genome sequencing studies have highlighted a large number of NDD/ID risk genes. To dissect the genetic causes and underlying biological pathways, in vivo experimental validation of the effects of these mutations is needed. The fruit fly, Drosophila melanogaster, is an ideal model to study NDDs, with highly tractable genetics, combined with simple behavioral and circuit assays, permitting rapid medium-throughput screening of NDD/ID risk genes. Here, we review studies where the use of well-established assays to study mechanisms of learning and memory in Drosophila has permitted insights into molecular mechanisms underlying IDs. We discuss how technologies in the fly model, combined with a high degree of molecular and physiological conservation between flies and mammals, highlight the Drosophila system as an ideal model to study neurodevelopmental disorders, from genetics to behavior.


Subject(s)
Intellectual Disability , Neurodevelopmental Disorders , Animals , Drosophila , Drosophila melanogaster , Intellectual Disability/genetics , Memory
8.
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
9.
J Vis Exp ; (153)2019 11 27.
Article in English | MEDLINE | ID: mdl-31840671

ABSTRACT

In retroviruses, antisense transcription has been described in both human immunodeficiency virus type 1 (HIV-1) and human T-lymphotropic virus 1 (HTLV-1). In HIV-1, the antisense protein ASP gene is located on the negative strand of env, in the reading frame -2, spanning the junction gp120/gp41. In the sense orientation, the 3' end of the ASP open reading frame overlaps with gp120 hypervariable regions V4 and V5. The study of ASP RNA has been thwarted by a phenomenon known as RT-self-priming, whereby RNA secondary structures have the ability to prime RT in absence of the specific primer, generating non-specific cDNAs. The combined use of high RNA denaturation with biotinylated reverse primers in the RT reaction, together with affinity purification of the cDNA onto streptavidin-coated magnetic beads, has allowed us to selectively amplify ASP RNA in CD4+ T cells derived from individuals infected with HIV-1. Our method is relatively low-cost, simple to perform, highly reliable, and easily reproducible. In this respect, it represents a powerful tool for the study of antisense transcription not only in HIV-1 but also in other biological systems.


Subject(s)
HIV-1/genetics , RNA, Antisense/genetics , Reverse Transcriptase Polymerase Chain Reaction/methods , Adult , Humans , Male , Middle Aged , Open Reading Frames/genetics , Young Adult
10.
Nat Commun ; 10(1): 3454, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31371726

ABSTRACT

Copy-number variants of the CYFIP1 gene in humans have been linked to autism spectrum disorders (ASD) and schizophrenia (SCZ), two neuropsychiatric disorders characterized by defects in brain connectivity. Here, we show that CYFIP1 plays an important role in brain functional connectivity and callosal functions. We find that Cyfip1-heterozygous mice have reduced functional connectivity and defects in white matter architecture, similar to phenotypes found in patients with ASD, SCZ and other neuropsychiatric disorders. Cyfip1-deficient mice also present decreased myelination in the callosal axons, altered presynaptic function, and impaired bilateral connectivity. Finally, Cyfip1 deficiency leads to abnormalities in motor coordination, sensorimotor gating and sensory perception, which are also known neuropsychiatric disorder-related symptoms. These results show that Cyfip1 haploinsufficiency compromises brain connectivity and function, which might explain its genetic association to neuropsychiatric disorders.


Subject(s)
Autism Spectrum Disorder/genetics , Autism Spectrum Disorder/metabolism , Brain/metabolism , Genetic Predisposition to Disease/genetics , Nerve Tissue Proteins/metabolism , Schizophrenia/metabolism , Adaptor Proteins, Signal Transducing , Animals , Autism Spectrum Disorder/diagnostic imaging , Axons , Behavior, Animal , Brain/diagnostic imaging , DNA Copy Number Variations , Disease Models, Animal , Genetic Association Studies , Haploinsufficiency , Heterozygote , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nerve Tissue Proteins/genetics , Nervous System/metabolism , Nervous System Physiological Phenomena/genetics , Phenotype , Psychomotor Performance , Schizophrenia/diagnostic imaging , Schizophrenia/genetics , Sensory Gating , White Matter
11.
Neuron ; 103(2): 217-234.e4, 2019 07 17.
Article in English | MEDLINE | ID: mdl-31171447

ABSTRACT

Synapses are fundamental information-processing units of the brain, and synaptic dysregulation is central to many brain disorders ("synaptopathies"). However, systematic annotation of synaptic genes and ontology of synaptic processes are currently lacking. We established SynGO, an interactive knowledge base that accumulates available research about synapse biology using Gene Ontology (GO) annotations to novel ontology terms: 87 synaptic locations and 179 synaptic processes. SynGO annotations are exclusively based on published, expert-curated evidence. Using 2,922 annotations for 1,112 genes, we show that synaptic genes are exceptionally well conserved and less tolerant to mutations than other genes. Many SynGO terms are significantly overrepresented among gene variations associated with intelligence, educational attainment, ADHD, autism, and bipolar disorder and among de novo variants associated with neurodevelopmental disorders, including schizophrenia. SynGO is a public, universal reference for synapse research and an online analysis platform for interpretation of large-scale -omics data (https://syngoportal.org and http://geneontology.org).


Subject(s)
Brain/cytology , Gene Ontology , Proteomics , Software , Synapses/physiology , Animals , Brain/physiology , Databases, Genetic , Humans , Knowledge Bases , Synaptic Potentials/physiology , Synaptosomes
12.
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
13.
Cell Death Dis ; 8(11): e3169, 2017 11 16.
Article in English | MEDLINE | ID: mdl-29144507

ABSTRACT

The fragile X mental retardation protein (FMRP) is lacking or mutated in patients with the fragile X syndrome (FXS), the most frequent form of inherited intellectual disability. FMRP affects metastasis formation in a mouse model for breast cancer. Here we show that FMRP is overexpressed in human melanoma with high Breslow thickness and high Clark level. Furthermore, meta-analysis of the TCGA melanoma data revealed that high levels of FMRP expression correlate significantly with metastatic tumor tissues, risk of relapsing and disease-free survival. Reduction of FMRP in metastatic melanoma cell lines impinges on cell migration, invasion and adhesion. Next-generation sequencing in human melanoma cells revealed that FMRP regulates a large number of mRNAs involved in relevant processes of melanoma progression. Our findings suggest an association between FMRP levels and the invasive phenotype in melanoma and might open new avenues towards the discovery of novel therapeutic targets.


Subject(s)
Fragile X Mental Retardation Protein/metabolism , Melanoma/metabolism , Melanoma/pathology , Fragile X Mental Retardation Protein/genetics , Humans , Neoplasm Invasiveness , Transfection
14.
Acta Neuropathol Commun ; 5(1): 5, 2017 01 11.
Article in English | MEDLINE | ID: mdl-28077174

ABSTRACT

The small heat shock protein HSPB1 (Hsp27) is an ubiquitously expressed molecular chaperone able to regulate various cellular functions like actin dynamics, oxidative stress regulation and anti-apoptosis. So far disease causing mutations in HSPB1 have been associated with neurodegenerative diseases such as distal hereditary motor neuropathy, Charcot-Marie-Tooth disease and amyotrophic lateral sclerosis. Most mutations in HSPB1 target its highly conserved α-crystallin domain, while other mutations affect the C- or N-terminal regions or its promotor. Mutations inside the α-crystallin domain have been shown to enhance the chaperone activity of HSPB1 and increase the binding to client proteins. However, the HSPB1-P182L mutation, located outside and downstream of the α-crystallin domain, behaves differently. This specific HSPB1 mutation results in a severe neuropathy phenotype affecting exclusively the motor neurons of the peripheral nervous system. We identified that the HSPB1-P182L mutant protein has a specifically increased interaction with the RNA binding protein poly(C)binding protein 1 (PCBP1) and results in a reduction of its translational repressive activity. RNA immunoprecipitation followed by RNA sequencing on mouse brain lead to the identification of PCBP1 mRNA targets. These targets contain larger 3'- and 5'-UTRs than average and are enriched in an RNA motif consisting of the CTCCTCCTCCTCC consensus sequence. Interestingly, next to the clear presence of neuronal transcripts among the identified PCBP1 targets we identified known genes associated with hereditary peripheral neuropathies and hereditary spastic paraplegias. We therefore conclude that HSPB1 can mediate translational repression through interaction with an RNA binding protein further supporting its role in neurodegenerative disease.


Subject(s)
Carrier Proteins/metabolism , HSP27 Heat-Shock Proteins/metabolism , Heat-Shock Proteins/metabolism , Heterogeneous-Nuclear Ribonucleoproteins/metabolism , Neoplasm Proteins/metabolism , Animals , Brain/metabolism , Carrier Proteins/genetics , Charcot-Marie-Tooth Disease/genetics , Charcot-Marie-Tooth Disease/metabolism , Consensus Sequence , DNA-Binding Proteins , Fibroblasts/metabolism , Gene Expression Regulation/physiology , HEK293 Cells , HSP27 Heat-Shock Proteins/genetics , HeLa Cells , Heat-Shock Proteins/genetics , Heterogeneous-Nuclear Ribonucleoproteins/genetics , Humans , Mice , Molecular Chaperones , Mutant Proteins/genetics , Mutant Proteins/metabolism , Mutation , Neoplasm Proteins/genetics , Protein Binding , Protein Biosynthesis/physiology , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA-Binding Proteins , Untranslated Regions
15.
Curr Opin Neurobiol ; 39: 146-51, 2016 08.
Article in English | MEDLINE | ID: mdl-27352301

ABSTRACT

Mutations in RNA-binding proteins (RBPs) are often linked to specific neurological disorders, suggesting that each of these RBPs regulates a particular neuronal function. Instead, they recognise many mRNAs and often participate in various post-transcriptional processes. To gain specificity, RBPs bind to RNA in collaboration with other RBPs. This model also explains how an RBP can play diverse roles: many RBPs do not contain an effector domain, which joins the RNA-protein complex as an additional unit. Different complexes, even if anchored on the same RBP, recruit diverse effectors. Therefore, the combination of RBPs determines the fate of an mRNA. We argue that new experimental and bioinformatic paradigms are needed to elucidate the combination of RBPs acting on a given mRNA.


Subject(s)
RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism , Humans , Nervous System Diseases/physiopathology , Protein Binding , Protein Domains , RNA-Binding Proteins/genetics
16.
J Chem Theory Comput ; 11(7): 3401-10, 2015 Jul 14.
Article in English | MEDLINE | ID: mdl-26575774

ABSTRACT

Cytoplasmic FMRP interacting protein 1 (CYFIP1), also known as specifically RAC1 activated protein 1 (Sra1), plays a dual role: together with fragile X mental retardation protein (FMRP) and eIF4E it forms a complex that inhibits mRNA translation, while together with WAVE1, NCKAP1, ABI2, and HSPC300 it forms the WAVE regulatory complex (WRC) that upon RAC1 activation initiates actin polymerization. Here we performed a molecular dynamics (MD) simulation on CYFIP1 extracted from the known WRC structure, which shows that, in the absence of its WRC partners, a butterfly-like motion brings the two ends of CYFIP1 closer together, enabling the interaction with eIF4E. Our MD simulation is supported by available data showing that binding of CYFIP1 to eIF4E and binding to the WRC are mutually exclusive and that there is fluorescence resonance energy transfer between the N- and C-termini of CYFIP1. The differential interaction of RAC1-GTP with the two CYFIP1 structures predicts that RAC1 is directly responsible for the switch between these conformations.


Subject(s)
Adaptor Proteins, Signal Transducing/chemistry , Molecular Docking Simulation , Molecular Dynamics Simulation , Movement , Humans
17.
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
18.
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
20.
Neurobiol Dis ; 73: 275-88, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25315682

ABSTRACT

miR-29 is expressed strongly in the brain and alterations in expression have been linked to several neurological disorders. To further explore the function of this miRNA in the brain, we generated miR-29a/b-1 knockout animals. Knockout mice develop a progressive disorder characterized by locomotor impairment and ataxia. The different members of the miR-29 family are strongly expressed in neurons of the olfactory bulb, the hippocampus and in the Purkinje cells of the cerebellum. Morphological analysis showed that Purkinje cells are smaller and display less dendritic arborisation compared to their wildtype littermates. In addition, a decreased number of parallel fibers form synapses on the Purkinje cells. We identified several mRNAs significantly up-regulated in the absence of the miR-29a/b-1 cluster. At the protein level, however, the voltage-gated potassium channel Kcnc3 (Kv3.3) was significantly up-regulated in the cerebella of the miR-29a/b knockout mice. Dysregulation of KCNC3 expression may contribute to the ataxic phenotype.


Subject(s)
Ataxia/metabolism , Cerebellum/metabolism , MicroRNAs/metabolism , Purkinje Cells/metabolism , Shaw Potassium Channels/metabolism , Animals , Behavior, Animal , Mice , Mice, Inbred C57BL , Mice, Knockout , Motor Activity
SELECTION OF CITATIONS
SEARCH DETAIL
...