Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Nat Neurosci ; 27(5): 822-835, 2024 May.
Article in English | MEDLINE | ID: mdl-38589584

ABSTRACT

Learning and memory require activity-induced changes in dendritic translation, but which mRNAs are involved and how they are regulated are unclear. In this study, to monitor how depolarization impacts local dendritic biology, we employed a dendritically targeted proximity labeling approach followed by crosslinking immunoprecipitation, ribosome profiling and mass spectrometry. Depolarization of primary cortical neurons with KCl or the glutamate agonist DHPG caused rapid reprogramming of dendritic protein expression, where changes in dendritic mRNAs and proteins are weakly correlated. For a subset of pre-localized messages, depolarization increased the translation of upstream open reading frames (uORFs) and their downstream coding sequences, enabling localized production of proteins involved in long-term potentiation, cell signaling and energy metabolism. This activity-dependent translation was accompanied by the phosphorylation and recruitment of the non-canonical translation initiation factor eIF4G2, and the translated uORFs were sufficient to confer depolarization-induced, eIF4G2-dependent translational control. These studies uncovered an unanticipated mechanism by which activity-dependent uORF translational control by eIF4G2 couples activity to local dendritic remodeling.


Subject(s)
Dendrites , Eukaryotic Initiation Factor-4G , Neurons , Open Reading Frames , Protein Biosynthesis , Animals , Dendrites/metabolism , Eukaryotic Initiation Factor-4G/metabolism , Protein Biosynthesis/physiology , Neurons/metabolism , Open Reading Frames/genetics , Rats , Mice , Cells, Cultured , Potassium Chloride/pharmacology
2.
Elife ; 102021 12 23.
Article in English | MEDLINE | ID: mdl-34939924

ABSTRACT

Neurons rely on translation of synaptic mRNAs in order to generate activity-dependent changes in plasticity. Here, we develop a strategy combining compartment-specific crosslinking immunoprecipitation (CLIP) and translating ribosome affinity purification (TRAP) in conditionally tagged mice to precisely define the ribosome-bound dendritic transcriptome of CA1 pyramidal neurons. We identify CA1 dendritic transcripts with differentially localized mRNA isoforms generated by alternative polyadenylation and alternative splicing, including many that have altered protein-coding capacity. Among dendritic mRNAs, FMRP targets were found to be overrepresented. Cell-type-specific FMRP-CLIP and TRAP in microdissected CA1 neuropil revealed 383 dendritic FMRP targets and suggests that FMRP differentially regulates functionally distinct modules in CA1 dendrites and cell bodies. FMRP regulates ~15-20% of mRNAs encoding synaptic functions and 10% of chromatin modulators, in the dendrite and cell body, respectively. In the absence of FMRP, dendritic FMRP targets had increased ribosome association, consistent with a function for FMRP in synaptic translational repression. Conversely, downregulation of FMRP targets involved in chromatin regulation in cell bodies suggests a role for FMRP in stabilizing mRNAs containing stalled ribosomes in this compartment. Together, the data support a model in which FMRP regulates the translation and expression of synaptic and nuclear proteins within different compartments of a single neuronal cell type.


The brain has over 100 billion neurons that together form vast networks to relay electrical signals. A neuron receives electrical signals from other neurons via branch-like structures known as dendrites. The signals then travel into the cell body of the neuron. If their sum reaches a threshold, they fire a new signal through a single outgoing projection known as the axon, which is connected to the dendrites of other neurons. A single neuron has thousands of dendrites that each receive inputs from different axons, and it is thought that the strengthening and weakening of these dendritic connections enables us to learn and store memories. Dendrites are filled with molecules known as messenger ribonucleic acids (mRNAs) that act as templates to make proteins. Axonal signals reaching the dendrites can trigger these mRNAs to make new proteins that strengthen or weaken the connections between the two neurons, which is believed to be necessary for generating long-term memories. A protein called FMRP is found in both the cell body and dendrites and is able to bind to and regulate the ability of mRNAs to make proteins. A loss of the gene encoding FMRP is the most common cause of inherited intellectual disability and autism in humans, but it remains unclear precisely what role this protein plays in learning and memory. Hale et al. used genetic and bioinformatics approaches to specifically study mRNAs in the dendrites and the cell body of a specific type of neuron involved in memory in mice. The experiments revealed that FMRP played different roles in the dendrites and cell body. In the dendrites, FMRP interacted with mRNAs encoding proteins that can change how the neuron responds to a signal from a neighboring neuron and may alter how strong the connections between the neurons are. On the other hand, FMRP in the cell body modulated the activities of mRNAs encoding proteins that in turn regulate the activities of genes. These findings change the way we think about how memory may work by suggesting that groups of mRNAs encoding proteins with certain activities are found in distinct parts of a single neuron. These observations offer new ways to approach intellectual disabilities and autism spectrum disorder.


Subject(s)
Cell Body/physiology , Dendrites/physiology , Fragile X Mental Retardation Protein/genetics , Gene Expression Regulation , Pyramidal Cells/physiology , RNA, Messenger/genetics , Animals , Female , Male , Mice , Mice, Inbred C57BL , Neuronal Plasticity/physiology , Pyramidal Cells/classification , Transcriptome
3.
Elife ; 82019 12 20.
Article in English | MEDLINE | ID: mdl-31860442

ABSTRACT

Loss of the RNA binding protein FMRP causes Fragile X Syndrome (FXS), the most common cause of inherited intellectual disability, yet it is unknown how FMRP function varies across brain regions and cell types and how this contributes to disease pathophysiology. Here we use conditional tagging of FMRP and CLIP (FMRP cTag CLIP) to examine FMRP mRNA targets in hippocampal CA1 pyramidal neurons, a critical cell type for learning and memory relevant to FXS phenotypes. Integrating these data with analysis of ribosome-bound transcripts in these neurons revealed CA1-enriched binding of autism-relevant mRNAs, and CA1-specific regulation of transcripts encoding circadian proteins. This contrasted with different targets in cerebellar granule neurons, and was consistent with circadian defects in hippocampus-dependent memory in Fmr1 knockout mice. These findings demonstrate differential FMRP-dependent regulation of mRNAs across neuronal cell types that may contribute to phenotypes such as memory defects and sleep disturbance associated with FXS.


Subject(s)
Autistic Disorder/metabolism , CA1 Region, Hippocampal/metabolism , Fragile X Mental Retardation Protein/genetics , Fragile X Syndrome/genetics , Memory Disorders/genetics , Pyramidal Cells/metabolism , Animals , Autistic Disorder/genetics , Autistic Disorder/physiopathology , CA1 Region, Hippocampal/cytology , Cerebellum/cytology , Cerebellum/metabolism , Circadian Clocks/genetics , Circadian Clocks/physiology , Disease Models, Animal , Fragile X Mental Retardation Protein/metabolism , Fragile X Syndrome/metabolism , Fragile X Syndrome/physiopathology , Gene Expression Regulation , Humans , Memory Disorders/metabolism , Memory Disorders/physiopathology , Mice, Inbred C57BL , Mice, Knockout , Neurons/metabolism
4.
Cell Rep ; 28(4): 979-991.e6, 2019 07 23.
Article in English | MEDLINE | ID: mdl-31340158

ABSTRACT

Post-transcriptional regulation by microRNAs (miRNAs) is essential for complex molecular responses to physiological insult and disease. Although many disease-associated miRNAs are known, their global targets and culminating network effects on pathophysiology remain poorly understood. We applied Argonaute (AGO) crosslinking immunoprecipitation (CLIP) to systematically elucidate altered miRNA-target interactions in brain following ischemia and reperfusion (I/R) injury. Among 1,190 interactions identified, the most prominent was the cumulative loss of target regulation by miR-29 family members. Integration of translational and time-course RNA profiles revealed a dynamic mode of miR-29 target de-regulation, led by acute translational activation and a later increase in RNA levels, allowing rapid proteomic changes to take effect. These functional regulatory events rely on canonical and non-canonical miR-29 binding and engage glutamate reuptake signals, such as glial glutamate transporter (GLT-1), to control local glutamate levels. These results uncover a miRNA target network that acts acutely to maintain brain homeostasis after ischemic stroke.


Subject(s)
Argonaute Proteins/metabolism , Brain Ischemia/metabolism , Brain/metabolism , Cross-Linking Reagents/chemistry , Glutamic Acid/metabolism , Homeostasis , Stroke/metabolism , Animals , Base Sequence , Brain Ischemia/complications , Brain Ischemia/genetics , Down-Regulation/genetics , Gene Regulatory Networks , Glucose/deficiency , Humans , Immunoprecipitation , Mice, Inbred C57BL , MicroRNAs/genetics , MicroRNAs/metabolism , Models, Biological , Neuroglia/metabolism , Oxygen , Polymorphism, Genetic , Signal Transduction , Stroke/complications , Stroke/genetics , Time Factors
5.
Nat Genet ; 51(6): 973-980, 2019 06.
Article in English | MEDLINE | ID: mdl-31133750

ABSTRACT

We address the challenge of detecting the contribution of noncoding mutations to disease with a deep-learning-based framework that predicts the specific regulatory effects and the deleterious impact of genetic variants. Applying this framework to 1,790 autism spectrum disorder (ASD) simplex families reveals a role in disease for noncoding mutations-ASD probands harbor both transcriptional- and post-transcriptional-regulation-disrupting de novo mutations of significantly higher functional impact than those in unaffected siblings. Further analysis suggests involvement of noncoding mutations in synaptic transmission and neuronal development and, taken together with previous studies, reveals a convergent genetic landscape of coding and noncoding mutations in ASD. We demonstrate that sequences carrying prioritized mutations identified in probands possess allele-specific regulatory activity, and we highlight a link between noncoding mutations and heterogeneity in the IQ of ASD probands. Our predictive genomics framework illuminates the role of noncoding mutations in ASD and prioritizes mutations with high impact for further study, and is broadly applicable to complex human diseases.


Subject(s)
Autism Spectrum Disorder/genetics , Deep Learning , Genetic Predisposition to Disease , Genome, Human , Genomics , Mutation , RNA, Untranslated , Algorithms , Alleles , Autism Spectrum Disorder/diagnosis , Computational Biology/methods , Gene Expression , Gene Expression Regulation , Genes, Reporter , Genetic Association Studies , Genomics/methods , Humans , Phenotype , RNA Processing, Post-Transcriptional , Transcription, Genetic
6.
Neuron ; 101(4): 707-720.e5, 2019 02 20.
Article in English | MEDLINE | ID: mdl-30638744

ABSTRACT

RNA-binding proteins (RBPs) regulate genetic diversity, but the degree to which they do so in individual cell types in vivo is unknown. We developed NOVA2 cTag-crosslinking and immunoprecipitation (CLIP) to generate functional RBP-RNA maps from different neuronal populations in the mouse brain. Combining cell type datasets from Nova2-cTag and Nova2 conditional knockout mice revealed differential NOVA2 regulatory actions on alternative splicing (AS) on the same transcripts expressed in different neurons. This includes functional differences in transcripts expressed in cortical and cerebellar excitatory versus inhibitory neurons, where we find NOVA2 is required for, respectively, development of laminar structure, motor coordination, and synapse formation. We also find that NOVA2-regulated AS is coupled to NOVA2 regulation of intron retention in hundreds of transcripts, which can sequester the trans-acting splicing factor PTBP2. In summary, cTag-CLIP complements single-cell RNA sequencing (RNA-seq) studies by providing a means for understanding RNA regulation of functional cell diversity.


Subject(s)
Alternative Splicing , Antigens, Neoplasm/genetics , Cerebellum/embryology , Cerebral Cortex/embryology , Neurogenesis , Neurons/metabolism , RNA-Binding Proteins/genetics , Animals , Antigens, Neoplasm/metabolism , Cells, Cultured , Cerebellum/cytology , Cerebellum/physiology , Cerebral Cortex/cytology , Excitatory Postsynaptic Potentials , Female , Inhibitory Postsynaptic Potentials , Male , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neuro-Oncological Ventral Antigen , Neurons/cytology , Neurons/physiology , Polypyrimidine Tract-Binding Protein/genetics , Polypyrimidine Tract-Binding Protein/metabolism , RNA-Binding Proteins/metabolism
7.
Elife ; 72018 05 31.
Article in English | MEDLINE | ID: mdl-29848443

ABSTRACT

Dynamic post-transcriptional control of RNA expression by RNA-binding proteins (RBPs) is critical during immune response. ZFP36 RBPs are prominent inflammatory regulators linked to autoimmunity and cancer, but functions in adaptive immunity are less clear. We used HITS-CLIP to define ZFP36 targets in mouse T cells, revealing unanticipated actions in regulating T-cell activation, proliferation, and effector functions. Transcriptome and ribosome profiling showed that ZFP36 represses mRNA target abundance and translation, notably through novel AU-rich sites in coding sequence. Functional studies revealed that ZFP36 regulates early T-cell activation kinetics cell autonomously, by attenuating activation marker expression, limiting T cell expansion, and promoting apoptosis. Strikingly, loss of ZFP36 in vivo accelerated T cell responses to acute viral infection and enhanced anti-viral immunity. These findings uncover a critical role for ZFP36 RBPs in restraining T cell expansion and effector functions, and suggest ZFP36 inhibition as a strategy to enhance immune-based therapies.


Subject(s)
Antiviral Agents/metabolism , Immunity , Lymphocyte Activation , RNA-Binding Proteins/metabolism , T-Lymphocytes/metabolism , Tristetraprolin/metabolism , Animals , Base Sequence , Bone Marrow/virology , CD4-Positive T-Lymphocytes/metabolism , Cell Lineage , Kinetics , Lymphocytic choriomeningitis virus/physiology , Mice, Inbred C57BL , Mice, Knockout , Protein Biosynthesis , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA-Binding Proteins/genetics , Ribosomes/metabolism , Transcriptome/genetics , Tristetraprolin/genetics
8.
Elife ; 72018 03 26.
Article in English | MEDLINE | ID: mdl-29578408

ABSTRACT

Alternative polyadenylation (APA) regulates mRNA translation, stability, and protein localization. However, it is unclear to what extent APA regulates these processes uniquely in specific cell types. Using a new technique, cTag-PAPERCLIP, we discovered significant differences in APA between the principal types of mouse cerebellar neurons, the Purkinje and granule cells, as well as between proliferating and differentiated granule cells. Transcripts that differed in APA in these comparisons were enriched in key neuronal functions and many differed in coding sequence in addition to 3'UTR length. We characterize Memo1, a transcript that shifted from expressing a short 3'UTR isoform to a longer one during granule cell differentiation. We show that Memo1 regulates granule cell precursor proliferation and that its long 3'UTR isoform is targeted by miR-124, contributing to its downregulation during development. Our findings provide insight into roles for APA in specific cell types and establish a platform for further functional studies.


Subject(s)
3' Untranslated Regions , Neurons/physiology , Polyadenylation , Protein Biosynthesis , RNA Stability , RNA, Messenger/metabolism , Animals , Cell Differentiation , Cell Proliferation , Cerebellum/cytology , Mice
9.
Neuron ; 95(6): 1334-1349.e5, 2017 Sep 13.
Article in English | MEDLINE | ID: mdl-28910620

ABSTRACT

Alternative polyadenylation (APA) is increasingly recognized to regulate gene expression across different cell types, but obtaining APA maps from individual cell types typically requires prior purification, a stressful procedure that can itself alter cellular states. Here, we describe a new platform, cTag-PAPERCLIP, that generates APA profiles from single cell populations in intact tissues; cTag-PAPERCLIP requires no tissue dissociation and preserves transcripts in native states. Applying cTag-PAPERCLIP to profile four major cell types in the mouse brain revealed common APA preferences between excitatory and inhibitory neurons distinct from astrocytes and microglia, regulated in part by neuron-specific RNA-binding proteins NOVA2 and PTBP2. We further identified a role of APA in switching Araf protein isoforms during microglia activation, impacting production of downstream inflammatory cytokines. Our results demonstrate the broad applicability of cTag-PAPERCLIP and a previously undiscovered role of APA in contributing to protein diversity between different cell types and cellular states within the brain.


Subject(s)
Brain/cytology , Microglia/metabolism , Neurons/metabolism , Polyadenylation , Protein Serine-Threonine Kinases/metabolism , Animals , Antigens, Neoplasm/physiology , Astrocytes/metabolism , Brain/metabolism , Cells, Cultured , Female , Humans , Male , Mice , Microglia/cytology , Nerve Tissue Proteins/physiology , Neuro-Oncological Ventral Antigen , Organ Specificity , Polypyrimidine Tract-Binding Protein/physiology , Protein Isoforms/metabolism , RNA-Binding Proteins/physiology
10.
Genes Dev ; 31(10): 990-1006, 2017 05 15.
Article in English | MEDLINE | ID: mdl-28637692

ABSTRACT

Understanding the biologic role of N6-methyladenosine (m6A) RNA modifications in mRNA requires an understanding of when and where in the life of a pre-mRNA transcript the modifications are made. We found that HeLa cell chromatin-associated nascent pre-mRNA (CA-RNA) contains many unspliced introns and m6A in exons but very rarely in introns. The m6A methylation is essentially completed upon the release of mRNA into the nucleoplasm. Furthermore, the content and location of each m6A modification in steady-state cytoplasmic mRNA are largely indistinguishable from those in the newly synthesized CA-RNA or nucleoplasmic mRNA. This result suggests that quantitatively little methylation or demethylation occurs in cytoplasmic mRNA. In addition, only ∼10% of m6As in CA-RNA are within 50 nucleotides of 5' or 3' splice sites, and the vast majority of exons harboring m6A in wild-type mouse stem cells is spliced the same in cells lacking the major m6A methyltransferase Mettl3. Both HeLa and mouse embryonic stem cell mRNAs harboring m6As have shorter half-lives, and thousands of these mRNAs have increased half-lives (twofold or more) in Mettl3 knockout cells compared with wild type. In summary, m6A is added to exons before or soon after exon definition in nascent pre-mRNA, and while m6A is not required for most splicing, its addition in the nascent transcript is a determinant of cytoplasmic mRNA stability.


Subject(s)
Cytoplasm/metabolism , RNA Precursors/metabolism , RNA Splicing , RNA, Messenger/metabolism , Animals , Chromatin/metabolism , Embryonic Stem Cells , Exons/genetics , Gene Knockout Techniques , HeLa Cells , Humans , Introns/genetics , Methylation , Methyltransferases/genetics , Methyltransferases/metabolism , Mice
11.
Cell Rep ; 18(5): 1312-1323, 2017 01 31.
Article in English | MEDLINE | ID: mdl-28147283

ABSTRACT

How type I and II interferons prevent periodic reemergence of latent pathogens in tissues of diverse cell types remains unknown. Using homogeneous neuron cultures latently infected with herpes simplex virus 1, we show that extrinsic type I or II interferon acts directly on neurons to induce unique gene expression signatures and inhibit the reactivation-specific burst of viral genome-wide transcription called phase I. Surprisingly, interferons suppressed reactivation only during a limited period early in phase I preceding productive virus growth. Sensitivity to type II interferon was selectively lost if viral ICP0, which normally accumulates later in phase I, was expressed before reactivation. Thus, interferons suppress reactivation by preventing initial expression of latent genomes but are ineffective once phase I viral proteins accumulate, limiting interferon action. This demonstrates that inducible reactivation from latency is only transiently sensitive to interferon. Moreover, it illustrates how latent pathogens escape host immune control to periodically replicate by rapidly deploying an interferon-resistant state.


Subject(s)
DNA Replication/immunology , Gene Expression/immunology , Virus Latency/immunology , Virus Replication/immunology , Animals , Cells, Cultured , Genome, Viral/immunology , Herpesvirus 1, Human/immunology , Interferon Type I/immunology , Interferon-gamma/immunology , Neurons/immunology , Neurons/virology , Rats , Rats, Sprague-Dawley , Transcription, Genetic/immunology , Viral Proteins/immunology , Virus Activation/immunology
12.
Elife ; 52016 05 25.
Article in English | MEDLINE | ID: mdl-27223325

ABSTRACT

The neuron specific RNA-binding proteins NOVA1 and NOVA2 are highly homologous alternative splicing regulators. NOVA proteins regulate at least 700 alternative splicing events in vivo, yet relatively little is known about the biologic consequences of NOVA action and in particular about functional differences between NOVA1 and NOVA2. Transcriptome-wide searches for isoform-specific functions, using NOVA1 and NOVA2 specific HITS-CLIP and RNA-seq data from mouse cortex lacking either NOVA isoform, reveals that NOVA2 uniquely regulates alternative splicing events of a series of axon guidance related genes during cortical development. Corresponding axonal pathfinding defects were specific to NOVA2 deficiency: Nova2-/- but not Nova1-/- mice had agenesis of the corpus callosum, and axonal outgrowth defects specific to ventral motoneuron axons and efferent innervation of the cochlea. Thus we have discovered that NOVA2 uniquely regulates alternative splicing of a coordinate set of transcripts encoding key components in cortical, brainstem and spinal axon guidance/outgrowth pathways during neural differentiation, with severe functional consequences in vivo.


Subject(s)
Antigens, Neoplasm/metabolism , Axon Guidance , Cerebral Cortex/embryology , Gene Expression Regulation, Developmental , Neurons/physiology , RNA-Binding Proteins/metabolism , RNA/metabolism , Animals , Mice , Mice, Knockout , Neuro-Oncological Ventral Antigen
13.
Cell Rep ; 15(2): 423-35, 2016 Apr 12.
Article in English | MEDLINE | ID: mdl-27050522

ABSTRACT

Accurate and precise annotation of 3' UTRs is critical for understanding how mRNAs are regulated by microRNAs (miRNAs) and RNA-binding proteins (RBPs). Here, we describe a method, poly(A) binding protein-mediated mRNA 3' end retrieval by crosslinking immunoprecipitation (PAPERCLIP), that shows high specificity for mRNA 3' ends and compares favorably with existing 3' end mapping methods. PAPERCLIP uncovers a previously unrecognized role of CstF64/64tau in promoting the usage of a selected group of non-canonical poly(A) sites, the majority of which contain a downstream GUKKU motif. Furthermore, in the mouse brain, PAPERCLIP discovers extended 3' UTR sequences harboring functional miRNA binding sites and reveals developmentally regulated APA shifts, including one in Atp2b2 that is evolutionarily conserved in humans and results in the gain of a functional binding site of miR-137. PAPERCLIP provides a powerful tool to decipher post-transcriptional regulation of mRNAs through APA in vivo.


Subject(s)
Immunoprecipitation/methods , MicroRNAs/metabolism , Poly A/metabolism , tau Proteins/metabolism , 3' Untranslated Regions/genetics , Animals , Base Sequence , Brain/metabolism , Cells, Cultured , Conserved Sequence , Evolution, Molecular , HEK293 Cells , HeLa Cells , Humans , Mice, Inbred C57BL , Nucleotide Motifs/genetics , Polyadenylation , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transcriptome/genetics
14.
Nat Commun ; 6: 8864, 2015 Nov 25.
Article in English | MEDLINE | ID: mdl-26602609

ABSTRACT

microRNAs (miRNAs) act as sequence-specific guides for Argonaute (AGO) proteins, which mediate posttranscriptional silencing of target messenger RNAs. Despite their importance in many biological processes, rules governing AGO-miRNA targeting are only partially understood. Here we report a modified AGO HITS-CLIP strategy termed CLEAR (covalent ligation of endogenous Argonaute-bound RNAs)-CLIP, which enriches miRNAs ligated to their endogenous mRNA targets. CLEAR-CLIP mapped ∼130,000 endogenous miRNA-target interactions in mouse brain and ∼40,000 in human hepatoma cells. Motif and structural analysis define expanded pairing rules for over 200 mammalian miRNAs. Most interactions combine seed-based pairing with distinct, miRNA-specific patterns of auxiliary pairing. At some regulatory sites, this specificity confers distinct silencing functions to miRNA family members with shared seed sequences but divergent 3'-ends. This work provides a means for explicit biochemical identification of miRNA sites in vivo, leading to the discovery that miRNA 3'-end pairing is a general determinant of AGO binding specificity.


Subject(s)
Argonaute Proteins/metabolism , Chimera/metabolism , MicroRNAs/metabolism , RNA Interference , RNA, Messenger/metabolism , Animals , Base Pairing , Binding Sites , Blotting, Northern , Blotting, Western , Brain/metabolism , Cell Line, Tumor , Electrophoresis, Polyacrylamide Gel , Flow Cytometry , Humans , Immunoprecipitation , Mice , Reverse Transcriptase Polymerase Chain Reaction
15.
Genes Dev ; 29(19): 2037-53, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26404942

ABSTRACT

We adapted UV CLIP (cross-linking immunoprecipitation) to accurately locate tens of thousands of m(6)A residues in mammalian mRNA with single-nucleotide resolution. More than 70% of these residues are present in the 3'-most (last) exons, with a very sharp rise (sixfold) within 150-400 nucleotides of the start of the last exon. Two-thirds of last exon m(6)A and >40% of all m(6)A in mRNA are present in 3' untranslated regions (UTRs); contrary to earlier suggestions, there is no preference for location of m(6)A sites around stop codons. Moreover, m(6)A is significantly higher in noncoding last exons than in next-to-last exons harboring stop codons. We found that m(6)A density peaks early in the 3' UTR and that, among transcripts with alternative polyA (APA) usage in both the brain and the liver, brain transcripts preferentially use distal polyA sites, as reported, and also show higher proximal m(6)A density in the last exons. Furthermore, when we reduced m6A methylation by knocking down components of the methylase complex and then examined 661 transcripts with proximal m6A peaks in last exons, we identified a set of 111 transcripts with altered (approximately two-thirds increased proximal) APA use. Taken together, these observations suggest a role of m(6)A modification in regulating proximal alternative polyA choice.


Subject(s)
3' Untranslated Regions/genetics , Adenosine/metabolism , DNA Methylation/genetics , Exons/genetics , Gene Expression Regulation , RNA, Messenger/chemistry , Animals , Brain/cytology , Brain/metabolism , Cell Line , Gene Knockdown Techniques , Humans , Liver/cytology , Liver/metabolism , Mice , Polyadenylation , tRNA Methyltransferases/genetics , tRNA Methyltransferases/metabolism
16.
Cell ; 160(6): 1099-110, 2015 Mar 12.
Article in English | MEDLINE | ID: mdl-25768906

ABSTRACT

Hepatitis C virus (HCV) uniquely requires the liver-specific microRNA-122 for replication, yet global effects on endogenous miRNA targets during infection are unexplored. Here, high-throughput sequencing and crosslinking immunoprecipitation (HITS-CLIP) experiments of human Argonaute (AGO) during HCV infection showed robust AGO binding on the HCV 5'UTR at known and predicted miR-122 sites. On the human transcriptome, we observed reduced AGO binding and functional mRNA de-repression of miR-122 targets during virus infection. This miR-122 "sponge" effect was relieved and redirected to miR-15 targets by swapping the miRNA tropism of the virus. Single-cell expression data from reporters containing miR-122 sites showed significant de-repression during HCV infection depending on expression level and site number. We describe a quantitative mathematical model of HCV-induced miR-122 sequestration and propose that such miR-122 inhibition by HCV RNA may result in global de-repression of host miR-122 targets, providing an environment fertile for the long-term oncogenic potential of HCV.


Subject(s)
Hepacivirus/metabolism , Hepatitis C/metabolism , Hepatitis C/virology , MicroRNAs/metabolism , RNA, Viral/metabolism , Argonaute Proteins/metabolism , Base Sequence , Cell Line, Tumor , Eukaryotic Initiation Factors/metabolism , Hepacivirus/genetics , Humans , Liver/metabolism , Liver/virology , Molecular Sequence Data , RNA, Viral/chemistry , Virus Replication
17.
Neuron ; 75(6): 1067-80, 2012 Sep 20.
Article in English | MEDLINE | ID: mdl-22998874

ABSTRACT

The paraneoplastic neurologic disorders target several families of neuron-specific RNA binding proteins (RNABPs), revealing that there are unique aspects of gene expression regulation in the mammalian brain. Here, we used HITS-CLIP to determine robust binding sites targeted by the neuronal Elav-like (nElavl) RNABPs. Surprisingly, nElav protein binds preferentially to GU-rich sequences in vivo and in vitro, with secondary binding to AU-rich sequences. nElavl null mice were used to validate the consequence of these binding events in the brain, demonstrating that they bind intronic sequences in a position dependent manner to regulate alternative splicing and to 3'UTR sequences to regulate mRNA levels. These controls converge on the glutamate synthesis pathway in neurons; nElavl proteins are required to maintain neurotransmitter glutamate levels, and the lack of nElavl leads to spontaneous epileptic seizure activity. The genome-wide analysis of nElavl targets reveals that one function of neuron-specific RNABPs is to control excitation-inhibition balance in the brain.


Subject(s)
Brain/cytology , ELAV Proteins/metabolism , Gene Expression Regulation/physiology , Glutamic Acid/metabolism , Neurons/physiology , RNA Splicing/genetics , 3' Untranslated Regions/physiology , Animals , Animals, Newborn , Brain/metabolism , Computational Biology , Disease Models, Animal , ELAV Proteins/deficiency , Electroencephalography , Epilepsy/genetics , Epilepsy/physiopathology , Gene Expression Regulation/genetics , Glutaminase/genetics , Glutaminase/metabolism , Mice , Mice, Knockout , Microarray Analysis , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , RNA, Messenger/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism
18.
Genes Dev ; 26(14): 1626-42, 2012 Jul 15.
Article in English | MEDLINE | ID: mdl-22802532

ABSTRACT

Two polypyrimidine tract RNA-binding proteins (PTBs), one near-ubiquitously expressed (Ptbp1) and another highly tissue-restricted (Ptbp2), regulate RNA in interrelated but incompletely understood ways. Ptbp1, a splicing regulator, is replaced in the brain and differentiated neuronal cell lines by Ptbp2. To define the roles of Ptbp2 in the nervous system, we generated two independent Ptbp2-null strains, unexpectedly revealing that Ptbp2 is expressed in neuronal progenitors and is essential for postnatal survival. A HITS-CLIP (high-throughput sequencing cross-linking immunoprecipitation)-generated map of reproducible Ptbp2-RNA interactions in the developing mouse neocortex, combined with results from splicing-sensitive microarrays, demonstrated that the major action of Ptbp2 is to inhibit adult-specific alternative exons by binding pyrimidine-rich sequences upstream of and/or within them. These regulated exons are present in mRNAs encoding proteins associated with control of cell fate, proliferation, and the actin cytoskeleton, suggesting a role for Ptbp2 in neurogenesis. Indeed, neuronal progenitors in the Ptbp2-null brain exhibited an aberrant polarity and were associated with regions of premature neurogenesis and reduced progenitor pools. Thus, Ptbp2 inhibition of a discrete set of adult neuronal exons underlies early brain development prior to neuronal differentiation and is essential for postnatal survival.


Subject(s)
Alternative Splicing/physiology , Brain/embryology , Cell Differentiation/physiology , Nerve Tissue Proteins/metabolism , Neural Stem Cells/metabolism , Polypyrimidine Tract-Binding Protein/metabolism , RNA, Messenger/metabolism , Animals , Brain/metabolism , Exons/physiology , Heterogeneous-Nuclear Ribonucleoproteins/genetics , Heterogeneous-Nuclear Ribonucleoproteins/metabolism , Mice , Mice, Mutant Strains , Nerve Tissue Proteins/genetics , Neural Stem Cells/cytology , Polypyrimidine Tract-Binding Protein/genetics , RNA, Messenger/genetics
19.
Cell ; 146(2): 247-61, 2011 Jul 22.
Article in English | MEDLINE | ID: mdl-21784246

ABSTRACT

FMRP loss of function causes Fragile X syndrome (FXS) and autistic features. FMRP is a polyribosome-associated neuronal RNA-binding protein, suggesting that it plays a key role in regulating neuronal translation, but there has been little consensus regarding either its RNA targets or mechanism of action. Here, we use high-throughput sequencing of RNAs isolated by crosslinking immunoprecipitation (HITS-CLIP) to identify FMRP interactions with mouse brain polyribosomal mRNAs. FMRP interacts with the coding region of transcripts encoding pre- and postsynaptic proteins and transcripts implicated in autism spectrum disorders (ASD). We developed a brain polyribosome-programmed translation system, revealing that FMRP reversibly stalls ribosomes specifically on its target mRNAs. Our results suggest that loss of a translational brake on the synthesis of a subset of synaptic proteins contributes to FXS. In addition, they provide insight into the molecular basis of the cognitive and allied defects in FXS and ASD and suggest multiple targets for clinical intervention.


Subject(s)
Autistic Disorder/metabolism , Brain/metabolism , Fragile X Mental Retardation Protein/metabolism , Fragile X Syndrome/metabolism , Ribosomes/metabolism , Synapses/metabolism , Animals , Autistic Disorder/physiopathology , Fragile X Mental Retardation Protein/genetics , Fragile X Syndrome/physiopathology , Humans , Mice , Mice, Knockout , Polyribosomes/metabolism , Protein Biosynthesis , RNA-Binding Proteins , Sequence Analysis, RNA
20.
Science ; 329(5990): 439-43, 2010 Jul 23.
Article in English | MEDLINE | ID: mdl-20558669

ABSTRACT

The control of RNA alternative splicing is critical for generating biological diversity. Despite emerging genome-wide technologies to study RNA complexity, reliable and comprehensive RNA-regulatory networks have not been defined. Here, we used Bayesian networks to probabilistically model diverse data sets and predict the target networks of specific regulators. We applied this strategy to identify approximately 700 alternative splicing events directly regulated by the neuron-specific factor Nova in the mouse brain, integrating RNA-binding data, splicing microarray data, Nova-binding motifs, and evolutionary signatures. The resulting integrative network revealed combinatorial regulation by Nova and the neuronal splicing factor Fox, interplay between phosphorylation and splicing, and potential links to neurologic disease. Thus, we have developed a general approach to understanding mammalian RNA regulation at the systems level.


Subject(s)
Alternative Splicing , Antigens, Neoplasm/metabolism , Brain/metabolism , Gene Regulatory Networks , Nerve Tissue Proteins/metabolism , RNA-Binding Proteins/metabolism , Animals , Artificial Intelligence , Bayes Theorem , Binding Sites , Cell Line , Computational Biology , Evolution, Molecular , Exons , Humans , Introns , Mice , Models, Genetic , Models, Statistical , Nervous System Diseases/genetics , Neuro-Oncological Ventral Antigen , Oligonucleotide Array Sequence Analysis , Phosphorylation , Protein Binding , Proteins/genetics , Proteins/metabolism , RNA/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...