Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
Nat Commun ; 14(1): 8499, 2023 Dec 21.
Article in English | MEDLINE | ID: mdl-38129387

ABSTRACT

Heterozygous deletions in the ANKS1B gene cause ANKS1B neurodevelopmental syndrome (ANDS), a rare genetic disease characterized by autism spectrum disorder (ASD), attention deficit/hyperactivity disorder, and speech and motor deficits. The ANKS1B gene encodes for AIDA-1, a protein that is enriched at neuronal synapses and regulates synaptic plasticity. Here we report an unexpected role for oligodendroglial deficits in ANDS pathophysiology. We show that Anks1b-deficient mouse models display deficits in oligodendrocyte maturation, myelination, and Rac1 function, and recapitulate white matter abnormalities observed in ANDS patients. Selective loss of Anks1b from the oligodendrocyte lineage, but not from neuronal populations, leads to deficits in social preference and sensory reactivity previously observed in a brain-wide Anks1b haploinsufficiency model. Furthermore, we find that clemastine, an antihistamine shown to increase oligodendrocyte precursor cell maturation and central nervous system myelination, rescues deficits in social preference in 7-month-old Anks1b-deficient mice. Our work shows that deficits in social behaviors present in ANDS may originate from abnormal Rac1 activity within oligodendrocytes.


Subject(s)
Autism Spectrum Disorder , Animals , Humans , Infant , Mice , Autism Spectrum Disorder/genetics , Intracellular Signaling Peptides and Proteins , Neurons , Oligodendroglia , Social Behavior
2.
Front Aging Neurosci ; 15: 1152562, 2023.
Article in English | MEDLINE | ID: mdl-37255534

ABSTRACT

Specific and effective treatments for autism spectrum disorder (ASD) are lacking due to a poor understanding of disease mechanisms. Here we test the idea that similarities between diverse ASD mouse models are caused by deficits in common molecular pathways at neuronal synapses. To do this, we leverage the availability of multiple genetic models of ASD that exhibit shared synaptic and behavioral deficits and use quantitative mass spectrometry with isobaric tandem mass tagging (TMT) to compare their hippocampal synaptic proteomes. Comparative analyses of mouse models for Fragile X syndrome (Fmr1 knockout), cortical dysplasia focal epilepsy syndrome (Cntnap2 knockout), PTEN hamartoma tumor syndrome (Pten haploinsufficiency), ANKS1B syndrome (Anks1b haploinsufficiency), and idiopathic autism (BTBR+) revealed several common altered cellular and molecular pathways at the synapse, including changes in oxidative phosphorylation, and Rho family small GTPase signaling. Functional validation of one of these aberrant pathways, Rac1 signaling, confirms that the ANKS1B model displays altered Rac1 activity counter to that observed in other models, as predicted by the bioinformatic analyses. Overall similarity analyses reveal clusters of synaptic profiles, which may form the basis for molecular subtypes that explain genetic heterogeneity in ASD despite a common clinical diagnosis. Our results suggest that ASD-linked susceptibility genes ultimately converge on common signaling pathways regulating synaptic function and propose that these points of convergence are key to understanding the pathogenesis of this disorder.

4.
Sci Adv ; 8(16): eabh2675, 2022 Apr 22.
Article in English | MEDLINE | ID: mdl-35442745

ABSTRACT

Stress is the most common trigger among episodic neurologic disorders. In episodic ataxia type 2 (EA2), physical or emotional stress causes episodes of severe motor dysfunction that manifest as ataxia and dystonia. We used the tottering (tg/tg) mouse, a faithful animal model of EA2, to dissect the mechanisms underlying stress-induced motor attacks. We find that in response to acute stress, activation of α1-adrenergic receptors (α1-Rs) on Purkinje cells by norepinephrine leads to their erratic firing and consequently motor attacks. We show that norepinephrine induces erratic firing of Purkinje cells by disrupting their spontaneous intrinsic pacemaking via a casein kinase 2 (CK2)-dependent signaling pathway, which likely reduces the activity of calcium-dependent potassium channels. Moreover, we report that disruption of this signaling cascade at a number of nodes prevents stress-induced attacks in the tottering mouse. Together, our results suggest that norepinephrine and CK2 are required for the initiation of stress-induced attacks in EA2 and provide previously unidentified targets for therapeutic intervention.

5.
J Virol ; 96(6): e0217821, 2022 03 23.
Article in English | MEDLINE | ID: mdl-35045266

ABSTRACT

The assembly and egress of alphaherpesviruses, including herpes simplex virus 1 (HSV-1) and pseudorabies virus (PRV), within neurons is poorly understood. A key unresolved question is the structure of the viral particle that moves by anterograde transport along the axon, and two alternative mechanisms have been described. In the "married" model, capsids acquire their envelopes in the cell body and then traffic along axons as enveloped virions within a bounding organelle. In the "separate" model, nonenveloped capsids travel from the cell body into and along the axon, eventually encountering their envelopment organelles at a distal site, such as the nerve cell terminal. Here, we describe an "envelopment trap" to test these models using the dominant negative terminal endosomal sorting complex required for transport (ESCRT) component VPS4-EQ. Green fluorescent protein (GFP)-tagged VPS4-EQ was used to arrest HSV-1 or PRV capsid envelopment, inhibit downstream trafficking, and GFP-label envelopment intermediates. We found that GFP-VPS4-EQ inhibited trafficking of HSV-1 capsids into and along the neurites and axons of mouse CAD cells and rat embryonic primary cortical neurons, consistent with egress via the married pathway. In contrast, transport of HSV-1 capsids was unaffected in the neurites of human SK-N-SH neuroblastoma cells, consistent with the separate mechanism. Unexpectedly, PRV (generally thought to utilize the married pathway) also appeared to employ the separate mechanism in SK-N-SH cells. We propose that apparent differences in the methods of HSV-1 and PRV egress are more likely a reflection of the host neuron in which transport is studied rather than true biological differences between the viruses themselves. IMPORTANCE Alphaherpesviruses, including herpes simplex virus 1 (HSV-1) and pseudorabies virus (PRV), are pathogens of the nervous system. They replicate in the nerve cell body and then travel great distances along axons to reach nerve termini and spread to adjacent epithelial cells; however, key aspects of how these viruses travel along axons remain controversial. Here, we test two alternative mechanisms for transport, the married and separate models, by blocking envelope assembly, a critical step in viral egress. When we arrest formation of the viral envelope using a mutated component of the cellular ESCRT apparatus, we find that entry of viral particles into axons is blocked in some types of neurons but not others. This approach allows us to determine whether envelope assembly occurs prior to entry of viruses into axons or afterwards and, thus, to distinguish between the alternative models for viral transport.


Subject(s)
Alphaherpesvirinae , Endosomal Sorting Complexes Required for Transport , Herpesvirus 1, Human , Herpesvirus 1, Suid , Neurons , Alphaherpesvirinae/metabolism , Animals , Axons/virology , Cell Line, Tumor , Cells, Cultured , Endosomal Sorting Complexes Required for Transport/metabolism , Herpesvirus 1, Human/physiology , Herpesvirus 1, Suid/physiology , Humans , Mice , Neurons/virology , Rats , Virus Assembly/physiology , Virus Internalization
6.
Elife ; 92020 09 09.
Article in English | MEDLINE | ID: mdl-32902378

ABSTRACT

Long-lasting forms of postsynaptic plasticity commonly involve protein synthesis-dependent structural changes of dendritic spines. However, the relationship between protein synthesis and presynaptic structural plasticity remains unclear. Here, we investigated structural changes in cannabinoid-receptor 1 (CB1)-mediated long-term depression of inhibitory transmission (iLTD), a form of presynaptic plasticity that involves a protein-synthesis-dependent long-lasting reduction in GABA release. We found that CB1-iLTD in acute rat hippocampal slices was associated with protein synthesis-dependent presynaptic structural changes. Using proteomics, we determined that CB1 activation in hippocampal neurons resulted in increased ribosomal proteins and initiation factors, but decreased levels of proteins involved in regulation of the actin cytoskeleton, such as ARPC2 and WASF1/WAVE1, and presynaptic release. Moreover, while CB1-iLTD increased ubiquitin/proteasome activity, ubiquitination but not proteasomal degradation was critical for structural and functional presynaptic CB1-iLTD. Thus, CB1-iLTD relies on both protein synthesis and ubiquitination to elicit structural changes that underlie long-term reduction of GABA release.


Subject(s)
Long-Term Synaptic Depression/physiology , Protein Biosynthesis/physiology , Receptor, Cannabinoid, CB1/metabolism , Ubiquitination/physiology , Actin-Related Protein 2-3 Complex/metabolism , Animals , Female , Hippocampus/chemistry , Hippocampus/cytology , Hippocampus/metabolism , Male , Rats , Rats, Sprague-Dawley , Wiskott-Aldrich Syndrome Protein Family/metabolism
7.
Cell Rep ; 29(7): 1789-1799.e6, 2019 11 12.
Article in English | MEDLINE | ID: mdl-31722197

ABSTRACT

The transport and translation of dendritic mRNAs by RNA-binding proteins (RBPs) allows for spatially restricted gene expression in neuronal processes. Although local translation in neuronal dendrites is now well documented, there is little evidence for corresponding effects on local synaptic function. Here, we report that the RBP Sam68 promotes the localization and translation of Arc mRNA preferentially in distal dendrites of rodent hippocampal CA1 pyramidal neurons. Consistent with Arc function in translation-dependent synaptic plasticity, we find that Sam68 knockout (KO) mice display impaired metabotropic glutamate-receptor-dependent long-term depression (mGluR-LTD) and impaired structural plasticity exclusively at distal Schaffer-collateral synapses. Moreover, by using quantitative proteomics, we find that the Sam68 interactome contains numerous regulators of mRNA translation and synaptic function. This work identifies an important player in Arc expression, provides a general framework for Sam68 regulation of protein synthesis, and uncovers a mechanism that enables the precise spatiotemporal expression of long-term plasticity throughout neurons.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , CA1 Region, Hippocampal/metabolism , Dendrites/metabolism , Long-Term Synaptic Depression , Protein Biosynthesis , Pyramidal Cells/metabolism , RNA-Binding Proteins/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , CA1 Region, Hippocampal/cytology , Female , Mice , Mice, Knockout , Pyramidal Cells/cytology , RNA-Binding Proteins/genetics
8.
Nat Commun ; 10(1): 3529, 2019 08 06.
Article in English | MEDLINE | ID: mdl-31388001

ABSTRACT

Neurodevelopmental disorders, including autism spectrum disorder, have complex polygenic etiologies. Single-gene mutations in patients can help define genetic factors and molecular mechanisms underlying neurodevelopmental disorders. Here we describe individuals with monogenic heterozygous microdeletions in ANKS1B, a predicted risk gene for autism and neuropsychiatric diseases. Affected individuals present with a spectrum of neurodevelopmental phenotypes, including autism, attention-deficit hyperactivity disorder, and speech and motor deficits. Neurons generated from patient-derived induced pluripotent stem cells demonstrate loss of the ANKS1B-encoded protein AIDA-1, a brain-specific protein highly enriched at neuronal synapses. A transgenic mouse model of Anks1b haploinsufficiency recapitulates a range of patient phenotypes, including social deficits, hyperactivity, and sensorimotor dysfunction. Identification of the AIDA-1 interactome using quantitative proteomics reveals protein networks involved in synaptic function and the etiology of neurodevelopmental disorders. Our findings formalize a link between the synaptic protein AIDA-1 and a rare, previously undefined genetic disease we term ANKS1B haploinsufficiency syndrome.


Subject(s)
Haploinsufficiency , Intracellular Signaling Peptides and Proteins/genetics , Neurodevelopmental Disorders/genetics , Animals , Behavior, Animal , Cells, Cultured , Child , Child, Preschool , Disease Models, Animal , Female , Hippocampus/pathology , Humans , Induced Pluripotent Stem Cells , Intracellular Signaling Peptides and Proteins/metabolism , Male , Mice , Mice, Knockout , Neurodevelopmental Disorders/pathology , Neurons , Primary Cell Culture , Protein Interaction Mapping , Protein Interaction Maps/genetics , Synapses/pathology , Syndrome , Exome Sequencing
9.
Alzheimers Dement (N Y) ; 5: 906-917, 2019.
Article in English | MEDLINE | ID: mdl-31890855

ABSTRACT

INTRODUCTION: Women are at increased risk for Alzheimer's disease (AD), but the reason why remains unknown. One hypothesis is that low estrogen levels at menopause increases vulnerability to AD, but this remains unproven. METHODS: We compared neuronal genes upregulated by estrogen in ovariectomized female rhesus macaques with a database of >17,000 diverse gene sets and applied a rare variant burden test to exome sequencing data from 1208 female AD patients with the age of onset < 75 years and 2162 female AD controls. RESULTS: We found a striking overlap between genes upregulated by estrogen in macaques and genes downregulated in the human postmortem AD brain, and we found that estrogen upregulates the APOE gene and that progesterone acts antagonistically to estrogen genome-wide. We also found that female patients with AD have excess rare mutations in the early menopause gene MCM8. DISCUSSION: We show with genomic data that the menopausal loss of estrogen could underlie the increased risk for AD in women.

10.
Neuron ; 92(2): 479-492, 2016 Oct 19.
Article in English | MEDLINE | ID: mdl-27764673

ABSTRACT

Long-term changes of neurotransmitter release are critical for proper brain function. However, the molecular mechanisms underlying these changes are poorly understood. While protein synthesis is crucial for the consolidation of postsynaptic plasticity, whether and how protein synthesis regulates presynaptic plasticity in the mature mammalian brain remain unclear. Here, using paired whole-cell recordings in rodent hippocampal slices, we report that presynaptic protein synthesis is required for long-term, but not short-term, plasticity of GABA release from type 1 cannabinoid receptor (CB1)-expressing axons. This long-term depression of inhibitory transmission (iLTD) involves cap-dependent protein synthesis in presynaptic interneuron axons, but not somata. Translation is required during the induction, but not maintenance, of iLTD. Mechanistically, CB1 activation enhances protein synthesis via the mTOR pathway. Furthermore, using super-resolution STORM microscopy, we revealed eukaryotic ribosomes in CB1-expressing axon terminals. These findings suggest that presynaptic local protein synthesis controls neurotransmitter release during long-term plasticity in the mature mammalian brain.


Subject(s)
Axons/metabolism , Interneurons/metabolism , Long-Term Synaptic Depression , Neuronal Plasticity/physiology , Presynaptic Terminals/metabolism , Protein Biosynthesis , Receptor, Cannabinoid, CB1/metabolism , gamma-Aminobutyric Acid/metabolism , Animals , Benzoxazines/pharmacology , Cell Body/metabolism , Hippocampus/cytology , Hippocampus/metabolism , Image Processing, Computer-Assisted , Imaging, Three-Dimensional , Mice , Mice, Inbred C57BL , Microscopy , Molecular Dynamics Simulation , Morpholines/pharmacology , Naphthalenes/pharmacology , Neural Inhibition , Neurons/metabolism , Optical Imaging , Patch-Clamp Techniques , Piperidines/pharmacology , Pyramidal Cells/metabolism , Pyrazoles/pharmacology , Rats , Rats, Sprague-Dawley , Receptor, Cannabinoid, CB1/agonists , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Ribosomes/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism
11.
PLoS One ; 11(9): e0162863, 2016.
Article in English | MEDLINE | ID: mdl-27657535

ABSTRACT

Transmembrane adaptor proteins (TRAPs) are important organisers for the transduction of immunoreceptor-mediated signals. Prr7 is a TRAP that regulates T cell receptor (TCR) signalling and potently induces cell death when overexpressed in human Jurkat T cells. Whether endogenous Prr7 has a similar functional role is currently unknown. To address this issue, we analysed the development and function of the immune system in Prr7 knockout mice. We found that loss of Prr7 partially impairs development of single positive CD4+ T cells in the thymus but has no effect on the development of other T cell subpopulations, B cells, NK cells, or NKT cells. Moreover, Prr7 does not affect the TCR signalling pathway as T cells derived from Prr7 knockout and wild-type animals and stimulated in vitro express the same levels of the activation marker CD69, and retain their ability to proliferate and activate induced cell death programs. Importantly, Prr7 knockout mice retained the capacity to mount a protective immune response when challenged with Listeria monocytogenes infection in vivo. In addition, T cell effector functions (activation, migration, and reactivation) were normal following induction of experimental autoimmune encephalomyelitis (EAE) in Prr7 knockout mice. Collectively, our work shows that loss of Prr7 does not result in a major immune system phenotype and suggests that Prr7 has a dispensable function for TCR signalling.

12.
EMBO J ; 35(17): 1923-34, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27458189

ABSTRACT

Elevated c-Jun levels result in apoptosis and are evident in neurodegenerative disorders such as Alzheimer's disease and dementia and after global cerebral insults including stroke and epilepsy. NMDA receptor (NMDAR) antagonists block c-Jun upregulation and prevent neuronal cell death following excitotoxic insults. However, the molecular mechanisms regulating c-Jun abundance in neurons are poorly understood. Here, we show that the synaptic component Proline rich 7 (PRR7) accumulates in the nucleus of hippocampal neurons following NMDAR activity. We find that PRR7 inhibits the ubiquitination of c-Jun by E3 ligase SCF(FBW) (7) (FBW7), increases c-Jun-dependent transcriptional activity, and promotes neuronal death. Microarray assays show that PRR7 abundance is directly correlated with transcripts associated with cellular viability. Moreover, PRR7 knockdown attenuates NMDAR-mediated excitotoxicity in neuronal cultures in a c-Jun-dependent manner. Our results show that PRR7 links NMDAR activity to c-Jun function and provide new insights into the molecular processes that underlie NMDAR-dependent excitotoxicity.


Subject(s)
JNK Mitogen-Activated Protein Kinases/metabolism , Membrane Proteins/metabolism , Neurons/physiology , Protein Processing, Post-Translational , Animals , Cell Survival , Cells, Cultured , Excitatory Amino Acid Agonists/metabolism , Hippocampus/pathology , Humans , Microarray Analysis , N-Methylaspartate/metabolism , Rats , Ubiquitination
14.
Neural Plast ; 2016: 3857934, 2016.
Article in English | MEDLINE | ID: mdl-26904297

ABSTRACT

Decades of research have demonstrated that rapid alterations in protein abundance are required for synaptic plasticity, a cellular correlate for learning and memory. Control of protein abundance, known as proteostasis, is achieved across a complex neuronal morphology that includes a tortuous axon as well as an extensive dendritic arbor supporting thousands of individual synaptic compartments. To regulate the spatiotemporal synthesis of proteins, neurons must efficiently coordinate the transport and metabolism of mRNAs. Among multiple levels of regulation, transacting RNA binding proteins (RBPs) control proteostasis by binding to mRNAs and mediating their transport and translation in response to synaptic activity. In addition to synthesis, protein degradation must be carefully balanced for optimal proteostasis, as deviations resulting in excess or insufficient abundance of key synaptic factors produce pathologies. As such, mutations in components of the proteasomal or translational machinery, including RBPs, have been linked to the pathogenesis of neurological disorders such as Fragile X Syndrome (FXS), Fragile X Tremor Ataxia Syndrome (FXTAS), and Autism Spectrum Disorders (ASD). In this review, we summarize recent scientific findings, highlight ongoing questions, and link basic molecular mechanisms to the pathogenesis of common neuropsychiatric disorders.


Subject(s)
Brain/metabolism , Mental Disorders/metabolism , Neuronal Plasticity , Neurons/metabolism , RNA-Binding Proteins/metabolism , Synapses/metabolism , Animals , Autism Spectrum Disorder/metabolism , Dendrites/metabolism , Fragile X Syndrome/metabolism , Homeostasis , Humans , Protein Biosynthesis , RNA, Messenger/metabolism
15.
J Neurosci ; 35(24): 8986-96, 2015 Jun 17.
Article in English | MEDLINE | ID: mdl-26085624

ABSTRACT

NMDA receptors (NMDARs) are key mediators of glutamatergic transmission and synaptic plasticity, and their dysregulation has been linked to diverse neuropsychiatric and neurodegenerative disorders. While normal NMDAR function requires regulated expression and trafficking of its different subunits, the molecular mechanisms underlying these processes are not fully understood. Here we report that the amyloid precursor protein intracellular domain associated-1 protein (AIDA-1), which associates with NMDARs and is encoded by ANKS1B, a gene recently linked to schizophrenia, regulates synaptic NMDAR subunit composition. Forebrain-specific AIDA-1 conditional knock-out (cKO) mice exhibit reduced GluN2B-mediated and increased GluN2A-mediated synaptic transmission, and biochemical analyses show AIDA-1 cKO mice have low GluN2B and high GluN2A protein levels at isolated hippocampal synaptic junctions compared with controls. These results are corroborated by immunocytochemical and electrophysiological analyses in primary neuronal cultures following acute lentiviral shRNA-mediated knockdown of AIDA-1. Moreover, hippocampal NMDAR-dependent but not metabotropic glutamate receptor-dependent plasticity is impaired in AIDA-1 cKO mice, further supporting a role for AIDA-1 in synaptic NMDAR function. We also demonstrate that AIDA-1 preferentially associates with GluN2B and with the adaptor protein Ca(2+)/calmodulin-dependent serine protein kinase and kinesin KIF17, which regulate the transport of GluN2B-containing NMDARs from the endoplasmic reticulum (ER) to synapses. Consistent with this function, GluN2B accumulates in ER-enriched fractions in AIDA-1 cKO mice. These findings suggest that AIDA-1 regulates NMDAR subunit composition at synapses by facilitating transport of GluN2B from the ER to synapses, which is critical for NMDAR plasticity. Our work provides an explanation for how AIDA-1 dysfunction might contribute to neuropsychiatric conditions, such as schizophrenia.


Subject(s)
Carrier Proteins/physiology , Hippocampus/physiology , Receptors, N-Methyl-D-Aspartate/physiology , Synapses/physiology , Synaptic Transmission/physiology , Animals , Cells, Cultured , Endoplasmic Reticulum/chemistry , Endoplasmic Reticulum/physiology , Female , Hippocampus/chemistry , Intracellular Signaling Peptides and Proteins , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Protein Subunits/analysis , Protein Subunits/physiology , Rats , Rats, Sprague-Dawley , Receptors, N-Methyl-D-Aspartate/analysis , Synapses/chemistry
16.
EMBO J ; 34(8): 984-6, 2015 Apr 15.
Article in English | MEDLINE | ID: mdl-25762591

ABSTRACT

Decades of research has shown that long-term changes in synaptic function ultimately require changes in gene expression.Recent work has focused on nuclear signaling by calcium and protein messengers initiated at postsynaptic sites. In this issue of The EMBO Journal, Ivanova and colleagues show that shuttling of CtBP-1 between presynaptic areas and nuclei regulates gene expression, which reminds us that presynaptic zones should not be ignored when considering synapse-to nucleus signaling.


Subject(s)
Carrier Proteins/physiology , Cytoskeletal Proteins/metabolism , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Neuropeptides/metabolism , Synapses/metabolism , Transcription Factors/physiology , Animals , Humans
17.
Cell Rep ; 10(9): 1459-1466, 2015 Mar 10.
Article in English | MEDLINE | ID: mdl-25753412

ABSTRACT

Dendritic protein homeostasis is crucial for most forms of long-term synaptic plasticity, and its dysregulation is linked to a wide range of brain disorders. Current models of metabotropic glutamate receptor mediated long-term depression (mGluR-LTD) suggest that rapid, local synthesis of key proteins is necessary for the induction and expression of LTD. Here, we find that mGluR-LTD can be induced in the absence of translation if the proteasome is concurrently inhibited. We report that enhanced proteasomal degradation during the expression of mGluR-LTD depletes dendritic proteins and inhibits subsequent inductions of LTD. Moreover, proteasome inhibition can rescue mGluR-LTD in mice null for the RNA binding protein Sam68, which we show here lack mGluR-dependent translation and LTD. Our study provides mechanistic insights for how changes in dendritic protein abundance regulate mGluR-LTD induction. We propose that Sam68-mediated translation helps to counterbalance degradation, ensuring that protein levels briefly remain above a permissive threshold during LTD induction.

18.
Proc Natl Acad Sci U S A ; 110(8): 3125-30, 2013 Feb 19.
Article in English | MEDLINE | ID: mdl-23382180

ABSTRACT

Proper synaptic function requires the spatial and temporal compartmentalization of RNA metabolism via transacting RNA-binding proteins (RBPs). Loss of RBP activity leads to abnormal posttranscriptional regulation and results in diverse neurological disorders with underlying deficits in synaptic morphology and transmission. Functional loss of the 68-kDa RBP Src associated in mitosis (Sam68) is associated with the pathogenesis of the neurological disorder fragile X tremor/ataxia syndrome. Sam68 binds to the mRNA of ß-actin (actb), an integral cytoskeletal component of dendritic spines. We show that Sam68 knockdown or disruption of the binding between Sam68 and its actb mRNA cargo in primary hippocampal cultures decreases the amount of actb mRNA in the synaptodendritic compartment and results in fewer dendritic spines. Consistent with these observations, we find that Sam68-KO mice have reduced levels of actb mRNA associated with synaptic polysomes and diminished levels of synaptic actb protein, suggesting that Sam68 promotes the translation of actb mRNA at synapses in vivo. Moreover, genetic knockout of Sam68 or acute knockdown in vivo results in fewer excitatory synapses in the hippocampal formation as assessed morphologically and functionally. Therefore, we propose that Sam68 regulates synapse number in a cell-autonomous manner through control of postsynaptic actb mRNA metabolism. Our research identifies a role for Sam68 in synaptodendritic posttranscriptional regulation of actb and may provide insight into the pathophysiology of fragile X tremor/ataxia syndrome.


Subject(s)
Actins/genetics , Adaptor Proteins, Signal Transducing/physiology , Dendrites/metabolism , RNA, Messenger/metabolism , RNA-Binding Proteins/physiology , Synapses , Adaptor Proteins, Signal Transducing/genetics , Animals , Mice , Mice, Knockout , RNA-Binding Proteins/genetics , Rats , Rats, Sprague-Dawley
19.
Alcohol ; 46(6): 569-76, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22703994

ABSTRACT

Recent studies have identified synaptic proteins that undergo synapse-to-nucleus translocation in response to neuronal activity that modulate protein synthesis. One such translational regulatory protein of the postsynaptic density (PSD) is AIDA-1d, which binds to PSD-95 via its C-terminus. Activation of synaptic NMDA receptors induces the cleavage of AIDA-1d, and the N-terminus is then shuttled to nuclear Cajal bodies where it plays a role in regulating global protein synthesis. Chronic ethanol exposure has been shown to increase the synaptic clustering of NMDA receptors and PSD-95. Here, we tested the hypothesis that AIDA-1d regulates chronic ethanol-induced increases in synaptic NMDA receptor expression. As reported, we found that AIDA-1 was highly enriched in dendritic spines and co-localized with PSD-95. Acute NMDA treatment increased AIDA-1 colocalization with p80 coilin, a marker of Cajal bodies. Chronic treatment (4 day) of cultures with ethanol (25-100 mM) or with the NMDA receptor antagonist AP-V (50 µM) enhanced the clustering of AIDA-1 at synaptic sites. However, chronic ethanol treatment (50 mM) in the presence of the NMDA receptor agonist NMDA (2.5 µM) prevented this increase. Surprisingly, PSD-95 did not seem to play a role in the synaptic distribution of AIDA-1 as this distribution was not affected by declustering PSD-95 from synapses in response to inhibition of palmitoylation. We found that lentiviral knockdown of AIDA-1d did not affect protein expression levels of NMDA receptor subunits GluN1, GluN1 C2', or GluN2B. The results of this study demonstrate that synaptic AIDA-1 expression is enhanced by chronic ethanol exposure that can be prevented by concurrent stimulation of NMDA receptors. In addition, we found that the association of AIDA-1 with PSD-95 is not required for its localization to the PSD. Moreover, we found that AIDA-1 does not regulate protein expression levels or alternative splicing of the GluN1 subunit of NMDA receptors.


Subject(s)
Ethanol/pharmacology , Hippocampus/metabolism , Neurons/metabolism , Animals , Disks Large Homolog 4 Protein , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , N-Methylaspartate/pharmacology , Nuclear Proteins/metabolism , Rats , Receptors, N-Methyl-D-Aspartate/biosynthesis , Receptors, N-Methyl-D-Aspartate/drug effects , Receptors, N-Methyl-D-Aspartate/metabolism , Up-Regulation
20.
J Neurosci ; 32(2): 599-609, 2012 Jan 11.
Article in English | MEDLINE | ID: mdl-22238095

ABSTRACT

Neuronal activity elicits changes in synaptic composition that play an important role in experience-dependent plasticity (Choquet and Triller, 2003; Lisman and Raghavachari, 2006; Bourne and Harris, 2008; Holtmaat and Svoboda, 2009). We used a modified version of stable isotope labeling by amino acids in cell culture to identify activity-dependent modifications in the composition of postsynaptic densities (PSDs) isolated from rat primary neuronal cultures. We found that synaptic activity altered ∼2% of the PSD proteome, which included an increase in diverse RNA binding proteins (RNABPs). Indeed, 12 of the 37 identified proteins whose levels changed with synaptic activity were RNABPs and included the heterogeneous nuclear ribonucleoproteins (hnRNPs) G, A2/B1, M, and D. Knockdown of hnRNPs M and G using shRNAs resulted in altered numbers of dendritic spines, suggesting a crucial role for these proteins in spine density. Synaptic activity also resulted in a concomitant increase in dendritic and synaptic poly(A) mRNA. However, this increase was not affected by knockdown of hnRNPs M or G. Our results suggest that hnRNP proteins regulate dendritic spine density and may play a role in synaptodendritic mRNA metabolism.


Subject(s)
Neurons/physiology , Post-Synaptic Density/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Synaptic Transmission/physiology , Animals , Brain-Derived Neurotrophic Factor/pharmacology , Cells, Cultured , Dendritic Spines/metabolism , Female , Heterogeneous-Nuclear Ribonucleoproteins/genetics , Heterogeneous-Nuclear Ribonucleoproteins/metabolism , Lentivirus/genetics , Long-Term Potentiation/drug effects , Long-Term Potentiation/physiology , Male , Neurons/cytology , Neurons/virology , Pregnancy , Proteome/genetics , Proteome/metabolism , Rats , Synaptic Transmission/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...