Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
bioRxiv ; 2023 Nov 02.
Article in English | MEDLINE | ID: mdl-37961595

ABSTRACT

Huntington's disease (HD) is a neurodegenerative disorder caused by a CAG repeat expansion in the first exon of the HTT gene encoding huntingtin. Prior reports have established a correlation between CAG expanded HTT and altered gene expression. However, the mechanisms leading to disruption of RNA processing in HD remain unclear. Here, our analysis of the reported HTT protein interactome identifies interactions with known RNA-binding proteins (RBPs). Total, long-read sequencing and targeted RASL-seq of RNAs from cortex and striatum of the HD mouse model R6/2 reveals increased exon skipping which is confirmed in Q150 and Q175 knock-in mice and in HD human brain. We identify the RBP TDP-43 and the N6-methyladenosine (m6A) writer protein methyltransferase 3 (METTL3) to be upstream regulators of exon skipping in HD. Along with this novel mechanistic insight, we observe decreased nuclear localization of TDP-43 and cytoplasmic accumulation of phosphorylated TDP-43 in HD mice and human brain. In addition, TDP-43 co-localizes with HTT in human HD brain forming novel nuclear aggregate-like bodies distinct from mutant HTT inclusions or previously observed TDP-43 pathologies. Binding of TDP-43 onto RNAs encoding HD-associated differentially expressed and aberrantly spliced genes is decreased. Finally, m6A RNA modification is reduced on RNAs abnormally expressed in striatum from HD R6/2 mouse brain, including at clustered sites adjacent to TDP-43 binding sites. Our evidence supports TDP-43 loss of function coupled with altered m6A modification as a novel mechanism underlying alternative splicing/unannotated exon usage in HD and highlights the critical nature of TDP-43 function across multiple neurodegenerative diseases.

2.
Cell Rep ; 36(10): 109685, 2021 09 07.
Article in English | MEDLINE | ID: mdl-34496257

ABSTRACT

Persistent cytoplasmic aggregates containing RNA binding proteins (RBPs) are central to the pathogenesis of late-onset neurodegenerative disorders such as amyotrophic lateral sclerosis (ALS). These aggregates share components, molecular mechanisms, and cellular protein quality control pathways with stress-induced RNA granules (SGs). Here, we assess the impact of stress on the global mRNA localization landscape of human pluripotent stem cell-derived motor neurons (PSC-MNs) using subcellular fractionation with RNA sequencing and proteomics. Transient stress disrupts subcellular RNA and protein distributions, alters the RNA binding profile of SG- and ALS-relevant RBPs and recapitulates disease-associated molecular changes such as aberrant splicing of STMN2. Although neurotypical PSC-MNs re-establish a normal subcellular localization landscape upon recovery from stress, cells harboring ALS-linked mutations are intransigent and display a delayed-onset increase in neuronal cell death. Our results highlight subcellular molecular distributions as predictive features and underscore the utility of cellular stress as a paradigm to study ALS-relevant mechanisms.


Subject(s)
Amyotrophic Lateral Sclerosis/metabolism , Cell Death/physiology , Motor Neurons/metabolism , RNA, Messenger/metabolism , Amyotrophic Lateral Sclerosis/genetics , Cell Death/genetics , Cytoplasmic Granules/metabolism , Cytoplasmic Ribonucleoprotein Granules/metabolism , Cytoplasmic Ribonucleoprotein Granules/pathology , DNA-Binding Proteins/metabolism , Humans , Mutation/genetics , RNA-Binding Proteins/metabolism
3.
J Clin Invest ; 131(12)2021 06 15.
Article in English | MEDLINE | ID: mdl-33945510

ABSTRACT

Chronic cellular stress associated with neurodegenerative disease can result in the persistence of stress granule (SG) structures, membraneless organelles that form in response to cellular stress. In Huntington's disease (HD), chronic expression of mutant huntingtin generates various forms of cellular stress, including activation of the unfolded protein response and oxidative stress. However, it has yet to be determined whether SGs are a feature of HD neuropathology. We examined the miRNA composition of extracellular vesicles (EVs) present in the cerebrospinal fluid (CSF) of patients with HD and show that a subset of their target mRNAs were differentially expressed in the prefrontal cortex. Of these targets, SG components were enriched, including the SG-nucleating Ras GTPase-activating protein-binding protein 1 (G3BP1). We investigated localization and levels of G3BP1 and found a significant increase in the density of G3BP1-positive granules in the cortex and hippocampus of R6/2 transgenic mice and in the superior frontal cortex of the brains of patients with HD. Intriguingly, we also observed that the SG-associated TAR DNA-binding protein 43 (TDP43), a nuclear RNA/DNA binding protein, was mislocalized to the cytoplasm of G3BP1 granule-positive HD cortical neurons. These findings suggest that G3BP1 SG dynamics may play a role in the pathophysiology of HD.


Subject(s)
Cytoplasmic Granules/metabolism , DNA Helicases/metabolism , DNA-Binding Proteins/metabolism , Hippocampus/metabolism , Huntington Disease/metabolism , Neurons/metabolism , Poly-ADP-Ribose Binding Proteins/metabolism , Prefrontal Cortex/metabolism , RNA Helicases/metabolism , RNA Recognition Motif Proteins/metabolism , Animals , Cytoplasmic Granules/pathology , DNA Helicases/genetics , DNA-Binding Proteins/genetics , Female , Hippocampus/pathology , Humans , Huntington Disease/genetics , Huntington Disease/pathology , Male , Mice , MicroRNAs/genetics , MicroRNAs/metabolism , Neurons/pathology , Poly-ADP-Ribose Binding Proteins/genetics , Prefrontal Cortex/pathology , Protein Transport/genetics , RNA Helicases/genetics , RNA Recognition Motif Proteins/genetics
5.
Nature ; 583(7818): 711-719, 2020 07.
Article in English | MEDLINE | ID: mdl-32728246

ABSTRACT

Many proteins regulate the expression of genes by binding to specific regions encoded in the genome1. Here we introduce a new data set of RNA elements in the human genome that are recognized by RNA-binding proteins (RBPs), generated as part of the Encyclopedia of DNA Elements (ENCODE) project phase III. This class of regulatory elements functions only when transcribed into RNA, as they serve as the binding sites for RBPs that control post-transcriptional processes such as splicing, cleavage and polyadenylation, and the editing, localization, stability and translation of mRNAs. We describe the mapping and characterization of RNA elements recognized by a large collection of human RBPs in K562 and HepG2 cells. Integrative analyses using five assays identify RBP binding sites on RNA and chromatin in vivo, the in vitro binding preferences of RBPs, the function of RBP binding sites and the subcellular localization of RBPs, producing 1,223 replicated data sets for 356 RBPs. We describe the spectrum of RBP binding throughout the transcriptome and the connections between these interactions and various aspects of RNA biology, including RNA stability, splicing regulation and RNA localization. These data expand the catalogue of functional elements encoded in the human genome by the addition of a large set of elements that function at the RNA level by interacting with RBPs.


Subject(s)
RNA-Binding Proteins/chemistry , RNA-Binding Proteins/metabolism , Transcriptome/genetics , Alternative Splicing/genetics , Base Sequence , Binding Sites , Cell Line , Chromatin/genetics , Chromatin/metabolism , Databases, Genetic , Female , Gene Knockdown Techniques , Humans , Intracellular Space/genetics , Male , Protein Binding , RNA, Messenger/chemistry , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA-Binding Proteins/genetics , Substrate Specificity
6.
Genome Biol ; 21(1): 90, 2020 04 06.
Article in English | MEDLINE | ID: mdl-32252787

ABSTRACT

BACKGROUND: A critical step in uncovering rules of RNA processing is to study the in vivo regulatory networks of RNA binding proteins (RBPs). Crosslinking and immunoprecipitation (CLIP) methods enable mapping RBP targets transcriptome-wide, but methodological differences present challenges to large-scale analysis across datasets. The development of enhanced CLIP (eCLIP) enabled the mapping of targets for 150 RBPs in K562 and HepG2, creating a unique resource of RBP interactomes profiled with a standardized methodology in the same cell types. RESULTS: Our analysis of 223 eCLIP datasets reveals a range of binding modalities, including highly resolved positioning around splicing signals and mRNA untranslated regions that associate with distinct RBP functions. Quantification of enrichment for repetitive and abundant multicopy elements reveals 70% of RBPs have enrichment for non-mRNA element classes, enables identification of novel ribosomal RNA processing factors and sites, and suggests that association with retrotransposable elements reflects multiple RBP mechanisms of action. Analysis of spliceosomal RBPs indicates that eCLIP resolves AQR association after intronic lariat formation, enabling identification of branch points with single-nucleotide resolution, and provides genome-wide validation for a branch point-based scanning model for 3' splice site recognition. Finally, we show that eCLIP peak co-occurrences across RBPs enable the discovery of novel co-interacting RBPs. CONCLUSIONS: This work reveals novel insights into RNA biology by integrated analysis of eCLIP profiling of 150 RBPs with distinct functions. Further, our quantification of both mRNA and other element association will enable further research to identify novel roles of RBPs in regulating RNA processing.


Subject(s)
RNA Processing, Post-Transcriptional , RNA-Binding Proteins/metabolism , Binding Sites , Hep G2 Cells , Humans , Immunoprecipitation , Introns , K562 Cells , RNA/metabolism , RNA Splicing , RNA, Ribosomal/metabolism , Repetitive Sequences, Nucleic Acid , Retroelements , Spliceosomes/metabolism
7.
Nat Neurosci ; 22(1): 25-36, 2019 01.
Article in English | MEDLINE | ID: mdl-30559470

ABSTRACT

Transcriptomic analyses of postmortem brains have begun to elucidate molecular abnormalities in autism spectrum disorder (ASD). However, a crucial pathway involved in synaptic development, RNA editing, has not yet been studied on a genome-wide scale. Here we profiled global patterns of adenosine-to-inosine (A-to-I) editing in a large cohort of postmortem brains of people with ASD. We observed a global bias for hypoediting in ASD brains, which was shared across brain regions and involved many synaptic genes. We show that the Fragile X proteins FMRP and FXR1P interact with RNA-editing enzymes (ADAR proteins) and modulate A-to-I editing. Furthermore, we observed convergent patterns of RNA-editing alterations in ASD and Fragile X syndrome, establishing this as a molecular link between these related diseases. Our findings, which are corroborated across multiple data sets, including dup15q (genomic duplication of 15q11.2-13.1) cases associated with intellectual disability, highlight RNA-editing dysregulation in ASD and reveal new mechanisms underlying this disorder.


Subject(s)
Autistic Disorder/metabolism , Brain/metabolism , RNA Editing , Adenosine Deaminase/genetics , Adenosine Deaminase/metabolism , Autistic Disorder/genetics , Fragile X Mental Retardation Protein/genetics , Fragile X Mental Retardation Protein/metabolism , Gene Expression Profiling , Humans , Neurons/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism
9.
Methods ; 126: 29-37, 2017 08 15.
Article in English | MEDLINE | ID: mdl-28790018

ABSTRACT

Crosslinking and immunoprecipitation (CLIP) followed by high-throughput sequencing identifies the binding sites of RNA binding proteins on RNAs. The covalent RNA-amino acid adducts produced by UV irradiation can cause premature reverse transcription termination and deletions (referred to as crosslink-induced mutation sites (CIMS)), which may decrease overall cDNA yield but are exploited in state-of-the-art CLIP methods to identify these crosslink sites at single-nucleotide resolution. Here, we show the ratio of both crosslinked base deletions and read-through versus termination are highly dependent on the identity of the reverse transcriptase enzyme as well as on buffer conditions used. AffinityScript and TGIRT showed a lack of deletion of the crosslinked base with other enzymes showing variable rates, indicating that utilization and interpretation of CIMS analysis requires knowledge of the reverse transcriptase enzyme used. Commonly used enzymes, including Superscript III and AffinityScript, show high termination rates in standard magnesium buffer conditions, but show a single base difference in the position of termination for TARDBP motifs. In contrast, manganese-containing buffer promoted read-through at the adduct site. These results validate the use of standard enzymes and also propose alternative enzyme and buffer choices for particularly challenging samples that contain extensive RNA adducts or other modifications that inhibit standard reverse transcription.


Subject(s)
RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Reverse Transcription/physiology , Sequence Analysis, RNA/methods , Base Sequence/physiology , Binding Sites/physiology , Humans
10.
Methods Mol Biol ; 1648: 177-200, 2017.
Article in English | MEDLINE | ID: mdl-28766298

ABSTRACT

Profiling of RNA binding protein targets in vivo provides critical insights into the mechanistic roles they play in regulating RNA processing. The enhanced crosslinking and immunoprecipitation (eCLIP) methodology provides a framework for robust, reproducible identification of transcriptome-wide protein-RNA interactions, with dramatically improved efficiency over previous methods. Here we provide a step-by-step description of the eCLIP method, along with insights into optimal performance of critical steps in the protocol. In particular, we describe improvements to the adaptor strategy that enables single-end enhanced CLIP (seCLIP), which removes the requirement for paired-end sequencing of eCLIP libraries. Further, we describe the observation of contaminating RNA present in standard nitrocellulose membrane suppliers, and present options with significantly reduced contamination for sensitive applications. These notes further refine the eCLIP methodology, simplifying robust RNA binding protein studies for all users.


Subject(s)
Cross-Linking Reagents/chemistry , Gene Expression Profiling/methods , Gene Library , Immunoprecipitation/methods , RNA-Binding Proteins/chemistry , Animals , Humans
11.
Nat Methods ; 13(6): 508-14, 2016 06.
Article in English | MEDLINE | ID: mdl-27018577

ABSTRACT

As RNA-binding proteins (RBPs) play essential roles in cellular physiology by interacting with target RNA molecules, binding site identification by UV crosslinking and immunoprecipitation (CLIP) of ribonucleoprotein complexes is critical to understanding RBP function. However, current CLIP protocols are technically demanding and yield low-complexity libraries with high experimental failure rates. We have developed an enhanced CLIP (eCLIP) protocol that decreases requisite amplification by ∼1,000-fold, decreasing discarded PCR duplicate reads by ∼60% while maintaining single-nucleotide binding resolution. By simplifying the generation of paired IgG and size-matched input controls, eCLIP improves specificity in the discovery of authentic binding sites. We generated 102 eCLIP experiments for 73 diverse RBPs in HepG2 and K562 cells (available at https://www.encodeproject.org), demonstrating that eCLIP enables large-scale and robust profiling, with amplification and sample requirements similar to those of ChIP-seq. eCLIP enables integrative analysis of diverse RBPs to reveal factor-specific profiles, common artifacts for CLIP and RNA-centric perspectives on RBP activity.


Subject(s)
Gene Expression Profiling/methods , Immunoprecipitation/methods , RNA-Binding Proteins/genetics , Transcriptome , Binding Sites , Cross-Linking Reagents/chemistry , Hep G2 Cells , Humans , K562 Cells , Photochemical Processes , Ultraviolet Rays
12.
Circulation ; 131(14): 1278-1290, 2015 Apr 07.
Article in English | MEDLINE | ID: mdl-25739401

ABSTRACT

BACKGROUND: Long noncoding RNAs (lncRNAs) have emerged as critical epigenetic regulators with important functions in development and disease. Here, we sought to identify and functionally characterize novel lncRNAs critical for vertebrate development. METHODS AND RESULTS: By relying on human pluripotent stem cell differentiation models, we investigated lncRNAs differentially regulated at key steps during human cardiovascular development with a special focus on vascular endothelial cells. RNA sequencing led to the generation of large data sets that serve as a gene expression roadmap highlighting gene expression changes during human pluripotent cell differentiation. Stage-specific analyses led to the identification of 3 previously uncharacterized lncRNAs, TERMINATOR, ALIEN, and PUNISHER, specifically expressed in undifferentiated pluripotent stem cells, cardiovascular progenitors, and differentiated endothelial cells, respectively. Functional characterization, including localization studies, dynamic expression analyses, epigenetic modification monitoring, and knockdown experiments in lower vertebrates, as well as murine embryos and human cells, confirmed a critical role for each lncRNA specific for each analyzed developmental stage. CONCLUSIONS: We have identified and functionally characterized 3 novel lncRNAs involved in vertebrate and human cardiovascular development, and we provide a comprehensive transcriptomic roadmap that sheds new light on the molecular mechanisms underlying human embryonic development, mesodermal commitment, and cardiovascular specification.


Subject(s)
Cardiovascular System/growth & development , Endothelial Cells/chemistry , Gene Expression Regulation, Developmental/genetics , Myocytes, Cardiac/chemistry , Pluripotent Stem Cells/chemistry , RNA, Long Noncoding/isolation & purification , Vertebrates/genetics , Animals , Cardiovascular System/metabolism , Cell Differentiation , Cell Lineage , Chromosome Mapping , Embryonic Development/genetics , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Fetal Heart/metabolism , Human Umbilical Vein Endothelial Cells , Humans , Mice , Molecular Sequence Data , Morpholinos/pharmacokinetics , Myocytes, Cardiac/cytology , RNA, Long Noncoding/physiology , Sequence Analysis, RNA , Transcriptome , Vertebrates/growth & development , Zebrafish/embryology
SELECTION OF CITATIONS
SEARCH DETAIL
...