Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
1.
Cell ; 170(6): 1209-1223.e20, 2017 Sep 07.
Article in English | MEDLINE | ID: mdl-28823556

ABSTRACT

Fragile X syndrome (FXS) is a leading genetic cause of intellectual disability and autism. FXS results from the loss of function of fragile X mental retardation protein (FMRP), which represses translation of target transcripts. Most of the well-characterized target transcripts of FMRP are synaptic proteins, yet targeting these proteins has not provided effective treatments. We examined a group of FMRP targets that encode transcriptional regulators, particularly chromatin-associated proteins. Loss of FMRP in mice results in widespread changes in chromatin regulation and aberrant gene expression. To determine if targeting epigenetic factors could reverse phenotypes associated with the disorder, we focused on Brd4, a BET protein and chromatin reader targeted by FMRP. Inhibition of Brd4 function alleviated many of the phenotypes associated with FXS. We conclude that loss of FMRP results in significant epigenetic misregulation and that targeting transcription via epigenetic regulators like Brd4 may provide new treatments for FXS.


Subject(s)
Azepines/pharmacology , Fragile X Mental Retardation Protein/metabolism , Fragile X Syndrome/drug therapy , Fragile X Syndrome/metabolism , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/metabolism , Transcription Factors/antagonists & inhibitors , Transcription Factors/metabolism , Triazoles/pharmacology , Animals , Cells, Cultured , Epigenesis, Genetic , Gene Expression/drug effects , Gene Expression Regulation/drug effects , Histones/metabolism , Mice , Mice, Knockout , Naphthyridines/pharmacology , Neurons/metabolism , Phenazines , Transcription, Genetic
2.
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
3.
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
4.
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
5.
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
6.
Elife ; 62017 07 13.
Article in English | MEDLINE | ID: mdl-28703706

ABSTRACT

Dopamine neurons in the ventral tegmental area use glutamate as a cotransmitter. To elucidate the behavioral role of the cotransmission, we targeted the glutamate-recycling enzyme glutaminase (gene Gls1). In mice with a dopamine transporter (Slc6a3)-driven conditional heterozygous (cHET) reduction of Gls1 in their dopamine neurons, dopamine neuron survival and transmission were unaffected, while glutamate cotransmission at phasic firing frequencies was reduced, enabling a selective focus on the cotransmission. The mice showed normal emotional and motor behaviors, and an unaffected response to acute amphetamine. Strikingly, amphetamine sensitization was reduced and latent inhibition potentiated. These behavioral effects, also seen in global GLS1 HETs with a schizophrenia resilience phenotype, were not seen in mice with an Emx1-driven forebrain reduction affecting most brain glutamatergic neurons. Thus, a reduction in dopamine neuron glutamate cotransmission appears to mediate significant components of the GLS1 HET schizophrenia resilience phenotype, and glutamate cotransmission appears to be important in attribution of motivational salience.


Subject(s)
Behavior, Animal , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/physiology , Glutamic Acid/metabolism , Ventral Tegmental Area/physiology , Action Potentials , Animals , Gene Knockdown Techniques , Glutaminase/genetics , Mice
7.
Elife ; 52016 Feb 19.
Article in English | MEDLINE | ID: mdl-26894958

ABSTRACT

Neuronal ELAV-like (nELAVL) RNA binding proteins have been linked to numerous neurological disorders. We performed crosslinking-immunoprecipitation and RNAseq on human brain, and identified nELAVL binding sites on 8681 transcripts. Using knockout mice and RNAi in human neuroblastoma cells, we showed that nELAVL intronic and 3' UTR binding regulates human RNA splicing and abundance. We validated hundreds of nELAVL targets among which were important neuronal and disease-associated transcripts, including Alzheimer's disease (AD) transcripts. We therefore investigated RNA regulation in AD brain, and observed differential splicing of 150 transcripts, which in some cases correlated with differential nELAVL binding. Unexpectedly, the most significant change of nELAVL binding was evident on non-coding Y RNAs. nELAVL/Y RNA complexes were specifically remodeled in AD and after acute UV stress in neuroblastoma cells. We propose that the increased nELAVL/Y RNA association during stress may lead to nELAVL sequestration, redistribution of nELAVL target binding, and altered neuronal RNA splicing.


Subject(s)
Brain/physiology , ELAV Proteins/metabolism , Gene Expression Regulation , RNA, Messenger/metabolism , RNA, Untranslated/metabolism , Alzheimer Disease/pathology , Animals , Cell Line, Tumor , Humans , Mice, Knockout , Neurons/physiology , Protein Binding , RNA Interference , RNA Splicing
8.
Nat Neurosci ; 18(10): 1464-73, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26301327

ABSTRACT

Precise regulation of transcription is crucial for the cellular mechanisms underlying memory formation. However, the link between neuronal stimulation and the proteins that directly interact with histone modifications to activate transcription in neurons remains unclear. Brd4 is a member of the bromodomain and extra-terminal domain (BET) protein family, which binds acetylated histones and is a critical regulator of transcription in many cell types, including transcription in response to external cues. Small molecule BET inhibitors are in clinical trials, yet almost nothing is known about Brd4 function in the brain. Here we show that Brd4 mediates the transcriptional regulation underlying learning and memory. The loss of Brd4 function affects critical synaptic proteins, which results in memory deficits in mice but also decreases seizure susceptibility. Thus Brd4 provides a critical link between neuronal activation and the transcriptional responses that occur during memory formation.


Subject(s)
Memory/physiology , Neurons/metabolism , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Transcriptional Activation/physiology , Animals , Azepines/pharmacology , Blotting, Western , Cells, Cultured , Chromatin Immunoprecipitation , Female , Immunohistochemistry , Male , Memory/drug effects , Mice , Mice, Inbred C57BL , Microscopy, Confocal , Neurons/drug effects , Polymerase Chain Reaction , RNA, Small Interfering , Seizures , Transcriptional Activation/drug effects , Transfection , Triazoles/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL