Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 153
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 184(18): 4680-4696.e22, 2021 09 02.
Article in English | MEDLINE | ID: mdl-34380047

ABSTRACT

Mutations causing amyotrophic lateral sclerosis (ALS) often affect the condensation properties of RNA-binding proteins (RBPs). However, the role of RBP condensation in the specificity and function of protein-RNA complexes remains unclear. We created a series of TDP-43 C-terminal domain (CTD) variants that exhibited a gradient of low to high condensation propensity, as observed in vitro and by nuclear mobility and foci formation. Notably, a capacity for condensation was required for efficient TDP-43 assembly on subsets of RNA-binding regions, which contain unusually long clusters of motifs of characteristic types and density. These "binding-region condensates" are promoted by homomeric CTD-driven interactions and required for efficient regulation of a subset of bound transcripts, including autoregulation of TDP-43 mRNA. We establish that RBP condensation can occur in a binding-region-specific manner to selectively modulate transcriptome-wide RNA regulation, which has implications for remodeling RNA networks in the context of signaling, disease, and evolution.


Subject(s)
DNA-Binding Proteins/metabolism , RNA-Binding Proteins/metabolism , RNA/metabolism , 3' Untranslated Regions/genetics , Base Sequence , Cell Nucleus/metabolism , HEK293 Cells , HeLa Cells , Homeostasis , Humans , Mutation/genetics , Nucleotide Motifs/genetics , Phase Transition , Point Mutation/genetics , Poly A/metabolism , Protein Binding , Protein Multimerization , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sequence Deletion
2.
Cell ; 174(5): 1067-1081.e17, 2018 08 23.
Article in English | MEDLINE | ID: mdl-30078707

ABSTRACT

Long mammalian introns make it challenging for the RNA processing machinery to identify exons accurately. We find that LINE-derived sequences (LINEs) contribute to this selection by recruiting dozens of RNA-binding proteins (RBPs) to introns. This includes MATR3, which promotes binding of PTBP1 to multivalent binding sites within LINEs. Both RBPs repress splicing and 3' end processing within and around LINEs. Notably, repressive RBPs preferentially bind to evolutionarily young LINEs, which are located far from exons. These RBPs insulate the LINEs and the surrounding intronic regions from RNA processing. Upon evolutionary divergence, changes in RNA motifs within LINEs lead to gradual loss of their insulation. Hence, older LINEs are located closer to exons, are a common source of tissue-specific exons, and increasingly bind to RBPs that enhance RNA processing. Thus, LINEs are hubs for the assembly of repressive RBPs and also contribute to the evolution of new, lineage-specific transcripts in mammals. VIDEO ABSTRACT.


Subject(s)
Heterogeneous-Nuclear Ribonucleoproteins/chemistry , Long Interspersed Nucleotide Elements , Nuclear Matrix-Associated Proteins/chemistry , Polyadenylation , Polypyrimidine Tract-Binding Protein/chemistry , RNA-Binding Proteins/chemistry , RNA/chemistry , Alternative Splicing , Animals , Binding Sites , Exons , HeLa Cells , Humans , Introns , Mice , Mutation , Nucleotide Motifs , Phylogeny , Protein Binding , Protein Interaction Mapping , RNA Splicing
3.
Nat Rev Mol Cell Biol ; 20(11): 715, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31506602

ABSTRACT

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

4.
Mol Cell ; 83(8): 1206-1207, 2023 04 20.
Article in English | MEDLINE | ID: mdl-37084712

ABSTRACT

RNA looping adds crucial information to understanding the position-dependent regulatory mechanisms of protein-RNA interactions. In this issue, Xue et al.1 present CRIC-seq, which comprehensively identifies RNA loops mediated by specific proteins and demonstrates their value for interpreting disease-causing mutations.


Subject(s)
Polypyrimidine Tract-Binding Protein , RNA Splicing , RNA , Alternative Splicing , Polypyrimidine Tract-Binding Protein/genetics , Polypyrimidine Tract-Binding Protein/metabolism , Proteins/genetics , RNA Splicing/genetics
5.
Mol Cell ; 83(24): 4509-4523.e11, 2023 Dec 21.
Article in English | MEDLINE | ID: mdl-38134885

ABSTRACT

The cytoplasm is highly compartmentalized, but the extent and consequences of subcytoplasmic mRNA localization in non-polarized cells are largely unknown. We determined mRNA enrichment in TIS granules (TGs) and the rough endoplasmic reticulum (ER) through particle sorting and isolated cytosolic mRNAs by digitonin extraction. When focusing on genes that encode non-membrane proteins, we observed that 52% have transcripts enriched in specific compartments. Compartment enrichment correlates with a combinatorial code based on mRNA length, exon length, and 3' UTR-bound RNA-binding proteins. Compartment-biased mRNAs differ in the functional classes of their encoded proteins: TG-enriched mRNAs encode low-abundance proteins with strong enrichment of transcription factors, whereas ER-enriched mRNAs encode large and highly expressed proteins. Compartment localization is an important determinant of mRNA and protein abundance, which is supported by reporter experiments showing that redirecting cytosolic mRNAs to the ER increases their protein expression. In summary, the cytoplasm is functionally compartmentalized by local translation environments.


Subject(s)
Endoplasmic Reticulum , Proteins , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum/metabolism , Proteins/metabolism , Cytosol/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Protein Transport , Protein Biosynthesis
6.
Nat Rev Mol Cell Biol ; 18(5): 331-337, 2017 05.
Article in English | MEDLINE | ID: mdl-28270684

ABSTRACT

Transcription and chromatin function are regulated by proteins that bind to DNA, nucleosomes or RNA polymerase II, with specific non-coding RNAs (ncRNAs) functioning to modulate their recruitment or activity. Unlike ncRNAs, nascent pre-mRNA was considered to be primarily a passive player in these processes. In this Opinion article, we describe recently identified interactions between nascent pre-mRNAs and regulatory proteins, highlight commonalities between the functions of nascent pre-mRNA and nascent ncRNA, and propose that both types of RNA have an active role in transcription and chromatin regulation.


Subject(s)
Chromatin/metabolism , Transcription, Genetic , Animals , Cell Nucleus/metabolism , Gene Expression Regulation , Humans , RNA Splicing , Repressor Proteins/metabolism , Transcription Elongation, Genetic , Transcription Factors
7.
Mol Cell ; 81(19): 4059-4075.e11, 2021 10 07.
Article in English | MEDLINE | ID: mdl-34437837

ABSTRACT

DDX3X is a ubiquitously expressed RNA helicase involved in multiple stages of RNA biogenesis. DDX3X is frequently mutated in Burkitt lymphoma, but the functional basis for this is unknown. Here, we show that loss-of-function DDX3X mutations are also enriched in MYC-translocated diffuse large B cell lymphoma and reveal functional cooperation between mutant DDX3X and MYC. DDX3X promotes the translation of mRNA encoding components of the core translational machinery, thereby driving global protein synthesis. Loss-of-function DDX3X mutations moderate MYC-driven global protein synthesis, thereby buffering MYC-induced proteotoxic stress during early lymphomagenesis. Established lymphoma cells restore full protein synthetic capacity by aberrant expression of DDX3Y, a Y chromosome homolog, the expression of which is normally restricted to the testis. These findings show that DDX3X loss of function can buffer MYC-driven proteotoxic stress and highlight the capacity of male B cell lymphomas to then compensate for this loss by ectopic DDX3Y expression.


Subject(s)
B-Lymphocytes/enzymology , DEAD-box RNA Helicases/metabolism , Lymphoma, B-Cell/enzymology , Minor Histocompatibility Antigens/metabolism , Neoplasm Proteins/biosynthesis , Proto-Oncogene Proteins c-myc/metabolism , Adolescent , Adult , Aged , Aged, 80 and over , Animals , B-Lymphocytes/pathology , Cell Line, Tumor , Child , Child, Preschool , DEAD-box RNA Helicases/genetics , Endoplasmic Reticulum Stress , Female , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Humans , Loss of Function Mutation , Lymphoma, B-Cell/genetics , Lymphoma, B-Cell/pathology , Male , Mice, Transgenic , Middle Aged , Minor Histocompatibility Antigens/genetics , Neoplasm Proteins/genetics , Protein Biosynthesis , Proteome , Proteostasis , Proto-Oncogene Proteins c-myc/genetics , Young Adult
8.
Cell ; 152(3): 453-66, 2013 Jan 31.
Article in English | MEDLINE | ID: mdl-23374342

ABSTRACT

There are ~650,000 Alu elements in transcribed regions of the human genome. These elements contain cryptic splice sites, so they are in constant danger of aberrant incorporation into mature transcripts. Despite posing a major threat to transcriptome integrity, little is known about the molecular mechanisms preventing their inclusion. Here, we present a mechanism for protecting the human transcriptome from the aberrant exonization of transposable elements. Quantitative iCLIP data show that the RNA-binding protein hnRNP C competes with the splicing factor U2AF65 at many genuine and cryptic splice sites. Loss of hnRNP C leads to formation of previously suppressed Alu exons, which severely disrupt transcript function. Minigene experiments explain disease-associated mutations in Alu elements that hamper hnRNP C binding. Thus, by preventing U2AF65 binding to Alu elements, hnRNP C plays a critical role as a genome-wide sentinel protecting the transcriptome. The findings have important implications for human evolution and disease.


Subject(s)
Alu Elements , Heterogeneous-Nuclear Ribonucleoprotein Group C/metabolism , Nuclear Proteins/metabolism , Ribonucleoproteins/metabolism , Transcriptome , Evolution, Molecular , Exons , Gene Expression Profiling , Gene Knockdown Techniques , HeLa Cells , Heterogeneous-Nuclear Ribonucleoprotein Group C/genetics , High-Throughput Nucleotide Sequencing , Humans , Immunoprecipitation , RNA Splice Sites , Sequence Analysis, RNA , Splicing Factor U2AF
9.
Nature ; 609(7928): 829-834, 2022 09.
Article in English | MEDLINE | ID: mdl-36104565

ABSTRACT

RNA splicing, the process of intron removal from pre-mRNA, is essential for the regulation of gene expression. It is controlled by the spliceosome, a megadalton RNA-protein complex that assembles de novo on each pre-mRNA intron through an ordered assembly of intermediate complexes1,2. Spliceosome activation is a major control step that requires substantial protein and RNA rearrangements leading to a catalytically active complex1-5. Splicing factor 3B subunit 1 (SF3B1) protein-a subunit of the U2 small nuclear ribonucleoprotein6-is phosphorylated during spliceosome activation7-10, but the kinase that is responsible has not been identified. Here we show that cyclin-dependent kinase 11 (CDK11) associates with SF3B1 and phosphorylates threonine residues at its N terminus during spliceosome activation. The phosphorylation is important for the association between SF3B1 and U5 and U6 snRNAs in the activated spliceosome, termed the Bact complex, and the phosphorylation can be blocked by OTS964, a potent and selective inhibitor of CDK11. Inhibition of CDK11 prevents spliceosomal transition from the precatalytic complex B to the activated complex Bact and leads to widespread intron retention and accumulation of non-functional spliceosomes on pre-mRNAs and chromatin. We demonstrate a central role of CDK11 in spliceosome assembly and splicing regulation and characterize OTS964 as a highly selective CDK11 inhibitor that suppresses spliceosome activation and splicing.


Subject(s)
Cyclin-Dependent Kinases , Phosphoproteins , RNA Precursors , RNA Splicing , Ribonucleoprotein, U2 Small Nuclear , Spliceosomes , Chromatin/metabolism , Cyclin-Dependent Kinases/antagonists & inhibitors , Cyclin-Dependent Kinases/metabolism , Enzyme Activation/drug effects , Phosphoproteins/chemistry , Phosphoproteins/metabolism , Phosphorylation , Quinolones/pharmacology , RNA Precursors/genetics , RNA Precursors/metabolism , RNA Splicing/drug effects , Ribonucleoprotein, U2 Small Nuclear/chemistry , Ribonucleoprotein, U2 Small Nuclear/metabolism , Spliceosomes/drug effects , Spliceosomes/metabolism , Threonine/metabolism
10.
Nature ; 603(7899): 131-137, 2022 03.
Article in English | MEDLINE | ID: mdl-35197628

ABSTRACT

Variants of UNC13A, a critical gene for synapse function, increase the risk of amyotrophic lateral sclerosis and frontotemporal dementia1-3, two related neurodegenerative diseases defined by mislocalization of the RNA-binding protein TDP-434,5. Here we show that TDP-43 depletion induces robust inclusion of a cryptic exon in UNC13A, resulting in nonsense-mediated decay and loss of UNC13A protein. Two common intronic UNC13A polymorphisms strongly associated with amyotrophic lateral sclerosis and frontotemporal dementia risk overlap with TDP-43 binding sites. These polymorphisms potentiate cryptic exon inclusion, both in cultured cells and in brains and spinal cords from patients with these conditions. Our findings, which demonstrate a genetic link between loss of nuclear TDP-43 function and disease, reveal the mechanism by which UNC13A variants exacerbate the effects of decreased TDP-43 function. They further provide a promising therapeutic target for TDP-43 proteinopathies.


Subject(s)
Amyotrophic Lateral Sclerosis , Frontotemporal Dementia , TDP-43 Proteinopathies , Alternative Splicing , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/metabolism , Codon, Nonsense , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Frontotemporal Dementia/genetics , Frontotemporal Dementia/metabolism , Humans , Nerve Tissue Proteins , Polymorphism, Single Nucleotide/genetics
11.
Trends Genet ; 40(7): 580-586, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38705823

ABSTRACT

Due to their capacity to mediate repetitive protein interactions, intrinsically disordered regions (IDRs) are crucial for the formation of various types of protein-RNA complexes. The functions of IDRs are strongly modulated by post-translational modifications (PTMs). Phosphorylation is the most common and well-studied modification of IDRs, which can alter homomeric or heteromeric interactions of proteins and impact their ability to phase separate. Moreover, phosphorylation can influence the RNA-binding properties of proteins, and recent studies demonstrated its selective impact on the global profiles of protein-RNA binding and regulation. These findings highlight the need for further integrative approaches to understand how signalling remodels protein-RNA networks in cells.


Subject(s)
Intrinsically Disordered Proteins , Protein Binding , Protein Processing, Post-Translational , RNA-Binding Proteins , RNA , Phosphorylation , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , RNA/metabolism , RNA/genetics , Protein Processing, Post-Translational/genetics , Humans , Intrinsically Disordered Proteins/metabolism , Intrinsically Disordered Proteins/genetics , Intrinsically Disordered Proteins/chemistry
12.
EMBO J ; 42(11): e110902, 2023 06 01.
Article in English | MEDLINE | ID: mdl-37039106

ABSTRACT

Oncogenic RAS signaling reprograms gene expression through both transcriptional and post-transcriptional mechanisms. While transcriptional regulation downstream of RAS is relatively well characterized, how RAS post-transcriptionally modulates gene expression to promote malignancy remains largely unclear. Using quantitative RNA interactome capture analysis, we here reveal that oncogenic RAS signaling reshapes the RNA-bound proteomic landscape of pancreatic cancer cells, with a network of nuclear proteins centered around nucleolin displaying enhanced RNA-binding activity. We show that nucleolin is phosphorylated downstream of RAS, which increases its binding to pre-ribosomal RNA (rRNA), boosts rRNA production, and promotes ribosome biogenesis. This nucleolin-dependent enhancement of ribosome biogenesis is crucial for RAS-induced pancreatic cancer cell proliferation and can be targeted therapeutically to inhibit tumor growth. Our results reveal that oncogenic RAS signaling drives ribosome biogenesis by regulating the RNA-binding activity of nucleolin and highlight a crucial role for this mechanism in RAS-mediated tumorigenesis.


Subject(s)
Genes, ras , Pancreatic Neoplasms , Humans , MAP Kinase Signaling System , Proteomics , Phosphoproteins/metabolism , RNA, Ribosomal/metabolism , RNA/metabolism , Cell Transformation, Neoplastic/genetics , Ribosomes/genetics , Ribosomes/metabolism , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Nucleolin
13.
EMBO J ; 42(14): e113168, 2023 07 17.
Article in English | MEDLINE | ID: mdl-37248947

ABSTRACT

Enhanced expression of the cold-shock protein RNA binding motif 3 (RBM3) is highly neuroprotective both in vitro and in vivo. Whilst upstream signalling pathways leading to RBM3 expression have been described, the precise molecular mechanism of RBM3 cold induction remains elusive. To identify temperature-dependent modulators of RBM3, we performed a genome-wide CRISPR-Cas9 knockout screen using RBM3-reporter human iPSC-derived neurons. We found that RBM3 mRNA and protein levels are robustly regulated by several splicing factors, with heterogeneous nuclear ribonucleoprotein H1 (HNRNPH1) being the strongest positive regulator. Splicing analysis revealed that moderate hypothermia significantly represses the inclusion of a poison exon, which, when retained, targets the mRNA for nonsense-mediated decay. Importantly, we show that HNRNPH1 mediates this cold-dependent exon skipping via its thermosensitive interaction with a G-rich motif within the poison exon. Our study provides novel mechanistic insights into the regulation of RBM3 and provides further targets for neuroprotective therapeutic strategies.


Subject(s)
Poisons , Humans , Cold Shock Proteins and Peptides/metabolism , Cold Temperature , RNA, Messenger/genetics , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism
14.
Nat Immunol ; 16(4): 415-25, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25706746

ABSTRACT

Post-transcriptional regulation of mRNA by the RNA-binding protein HuR (encoded by Elavl1) is required in B cells for the germinal center reaction and for the production of class-switched antibodies in response to thymus-independent antigens. Transcriptome-wide examination of RNA isoforms and their abundance and translation in HuR-deficient B cells, together with direct measurements of HuR-RNA interactions, revealed that HuR-dependent splicing of mRNA affected hundreds of transcripts, including that encoding dihydrolipoamide S-succinyltransferase (Dlst), a subunit of the 2-oxoglutarate dehydrogenase (α-KGDH) complex. In the absence of HuR, defective mitochondrial metabolism resulted in large amounts of reactive oxygen species and B cell death. Our study shows how post-transcriptional processes control the balance of energy metabolism required for the proliferation and differentiation of B cells.


Subject(s)
B-Lymphocytes/immunology , ELAV Proteins/immunology , Germinal Center/immunology , Immunity, Humoral , Immunoglobulins/biosynthesis , RNA, Messenger/immunology , Acyltransferases/genetics , Acyltransferases/immunology , Alternative Splicing/immunology , Animals , Antigens/administration & dosage , Antigens/immunology , B-Lymphocytes/cytology , B-Lymphocytes/drug effects , Cell Death , Cell Differentiation , Cell Proliferation , ELAV Proteins/genetics , Erythrocytes/immunology , Germinal Center/cytology , Germinal Center/drug effects , Immunization , Immunoglobulin Class Switching , Lipopolysaccharides/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/genetics , Mitochondria/immunology , RNA, Messenger/genetics , Reactive Oxygen Species/immunology , Reactive Oxygen Species/metabolism , Sheep
15.
Nature ; 594(7861): 117-123, 2021 06.
Article in English | MEDLINE | ID: mdl-34012113

ABSTRACT

The human genome expresses thousands of natural antisense transcripts (NAT) that can regulate epigenetic state, transcription, RNA stability or translation of their overlapping genes1,2. Here we describe MAPT-AS1, a brain-enriched NAT that is conserved in primates and contains an embedded mammalian-wide interspersed repeat (MIR), which represses tau translation by competing for ribosomal RNA pairing with the MAPT mRNA internal ribosome entry site3. MAPT encodes tau, a neuronal intrinsically disordered protein (IDP) that stabilizes axonal microtubules. Hyperphosphorylated, aggregation-prone tau forms the hallmark inclusions of tauopathies4. Mutations in MAPT cause familial frontotemporal dementia, and common variations forming the MAPT H1 haplotype are a significant risk factor in many tauopathies5 and Parkinson's disease. Notably, expression of MAPT-AS1 or minimal essential sequences from MAPT-AS1 (including MIR) reduces-whereas silencing MAPT-AS1 expression increases-neuronal tau levels, and correlate with tau pathology in human brain. Moreover, we identified many additional NATs with embedded MIRs (MIR-NATs), which are overrepresented at coding genes linked to neurodegeneration and/or encoding IDPs, and confirmed MIR-NAT-mediated translational control of one such gene, PLCG1. These results demonstrate a key role for MAPT-AS1 in tauopathies and reveal a potentially broad contribution of MIR-NATs to the tightly controlled translation of IDPs6, with particular relevance for proteostasis in neurodegeneration.


Subject(s)
Protein Biosynthesis/genetics , Proteostasis/genetics , RNA, Antisense/genetics , Tauopathies/genetics , Tauopathies/metabolism , tau Proteins/genetics , tau Proteins/metabolism , Aged , Animals , Binding Sites , Brain/metabolism , Brain/pathology , Case-Control Studies , Cell Differentiation , Disease Progression , Female , Humans , Internal Ribosome Entry Sites/genetics , Male , Mice , Mice, Transgenic , Middle Aged , Neurons/metabolism , Neurons/pathology , Ribosomes/metabolism , tau Proteins/biosynthesis
16.
Mol Cell ; 76(2): 329-345, 2019 10 17.
Article in English | MEDLINE | ID: mdl-31626751

ABSTRACT

High-throughput sequencing-based methods and their applications in the study of transcriptomes have revolutionized our understanding of alternative splicing. Networks of functionally coordinated and biologically important alternative splicing events continue to be discovered in an ever-increasing diversity of cell types in the context of physiologically normal and disease states. These studies have been complemented by efforts directed at defining sequence codes governing splicing and their cognate trans-acting factors, which have illuminated important combinatorial principles of regulation. Additional studies have revealed critical roles of position-dependent, multivalent protein-RNA interactions that direct splicing outcomes. Investigations of evolutionary changes in RNA binding proteins, splice variants, and associated cis elements have further shed light on the emergence, mechanisms, and functions of splicing networks. Progress in these areas has emphasized the need for a coordinated, community-based effort to systematically address the functions of individual splice variants associated with normal and disease biology.


Subject(s)
Alternative Splicing/physiology , Evolution, Molecular , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Humans
17.
Mol Cell ; 74(2): 254-267.e10, 2019 04 18.
Article in English | MEDLINE | ID: mdl-30824372

ABSTRACT

DNA damage response (DDR) involves dramatic transcriptional alterations, the mechanisms of which remain ill defined. Here, we show that following genotoxic stress, the RNA-binding motif protein 7 (RBM7) stimulates RNA polymerase II (Pol II) transcription and promotes cell viability by activating the positive transcription elongation factor b (P-TEFb) via its release from the inhibitory 7SK small nuclear ribonucleoprotein (7SK snRNP). This is mediated by activation of p38MAPK, which triggers enhanced binding of RBM7 with core subunits of 7SK snRNP. In turn, P-TEFb relocates to chromatin to induce transcription of short units, including key DDR genes and multiple classes of non-coding RNAs. Critically, interfering with the axis of RBM7 and P-TEFb provokes cellular hypersensitivity to DNA-damage-inducing agents due to activation of apoptosis. Our work uncovers the importance of stress-dependent stimulation of Pol II pause release, which enables a pro-survival transcriptional response that is crucial for cell fate upon genotoxic insult.


Subject(s)
Positive Transcriptional Elongation Factor B/genetics , RNA Polymerase II/genetics , RNA-Binding Proteins/genetics , Transcription, Genetic , Apoptosis/genetics , Cell Survival/genetics , DNA Damage/genetics , HEK293 Cells , Humans , RNA, Long Noncoding/genetics , Ribonucleoproteins, Small Nuclear/genetics , p38 Mitogen-Activated Protein Kinases/genetics
18.
Mol Cell ; 74(5): 951-965.e13, 2019 06 06.
Article in English | MEDLINE | ID: mdl-31047794

ABSTRACT

RNA-binding proteins (RBPs) and long non-coding RNAs (lncRNAs) are key regulators of gene expression, but their joint functions in coordinating cell fate decisions are poorly understood. Here we show that the expression and activity of the RBP TDP-43 and the long isoform of the lncRNA Neat1, the scaffold of the nuclear compartment "paraspeckles," are reciprocal in pluripotent and differentiated cells because of their cross-regulation. In pluripotent cells, TDP-43 represses the formation of paraspeckles by enhancing the polyadenylated short isoform of Neat1. TDP-43 also promotes pluripotency by regulating alternative polyadenylation of transcripts encoding pluripotency factors, including Sox2, which partially protects its 3' UTR from miR-21-mediated degradation. Conversely, paraspeckles sequester TDP-43 and other RBPs from mRNAs and promote exit from pluripotency and embryonic patterning in the mouse. We demonstrate that cross-regulation between TDP-43 and Neat1 is essential for their efficient regulation of a broad network of genes and, therefore, of pluripotency and differentiation.


Subject(s)
Cell Differentiation/genetics , DNA-Binding Proteins/genetics , Mouse Embryonic Stem Cells/metabolism , RNA, Long Noncoding/genetics , Animals , Cell Nucleus/genetics , Cell Nucleus/metabolism , DNA-Binding Proteins/metabolism , Humans , Mice , MicroRNAs/genetics , Pluripotent Stem Cells/metabolism , Polyadenylation/genetics , RNA, Long Noncoding/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism
19.
Mol Cell ; 69(3): 354-369, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29395060

ABSTRACT

RNA binding proteins (RBPs) regulate all aspects in the life cycle of RNA molecules. To elucidate the elements that guide RNA specificity, regulatory mechanisms, and functions of RBPs, methods that identify direct endogenous protein-RNA interactions are particularly valuable. UV crosslinking and immunoprecipitation (CLIP) purifies short RNA fragments that crosslink to a specific protein and then identifies these fragments by sequencing. When combined with high-throughput sequencing, CLIP can produce transcriptome-wide maps of RNA crosslink sites. The protocol is comprised of several dozen biochemical steps, and improvements made over the last 15 years have increased its resolution, sensitivity, and convenience. Adaptations of CLIP are also emerging in the epitranscriptomic field to map the positions of RNA modifications accurately. Here, we describe the rationale for each step in the protocol and discuss the impact of variations to help users determine the most suitable option.


Subject(s)
Immunoprecipitation/methods , RNA Recognition Motif Proteins/genetics , RNA/genetics , Binding Sites , High-Throughput Nucleotide Sequencing/methods , Protein Binding , RNA-Binding Proteins/metabolism , Sequence Analysis, RNA/methods , Transcriptome
20.
Mol Cell ; 72(3): 496-509.e9, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30388411

ABSTRACT

Recursive splicing (RS) starts by defining an "RS-exon," which is then spliced to the preceding exon, thus creating a recursive 5' splice site (RS-5ss). Previous studies focused on cryptic RS-exons, and now we find that the exon junction complex (EJC) represses RS of hundreds of annotated, mainly constitutive RS-exons. The core EJC factors, and the peripheral factors PNN and RNPS1, maintain RS-exon inclusion by repressing spliceosomal assembly on RS-5ss. The EJC also blocks 5ss located near exon-exon junctions, thus repressing inclusion of cryptic microexons. The prevalence of annotated RS-exons is high in deuterostomes, while the cryptic RS-exons are more prevalent in Drosophila, where EJC appears less capable of repressing RS. Notably, incomplete repression of RS also contributes to physiological alternative splicing of several human RS-exons. Finally, haploinsufficiency of the EJC factor Magoh in mice is associated with skipping of RS-exons in the brain, with relevance to the microcephaly phenotype and human diseases.


Subject(s)
Alternative Splicing/physiology , Exons/physiology , RNA Splice Sites/physiology , Animals , Cell Line , Cell Nucleus , Drosophila , HEK293 Cells , HeLa Cells , Humans , Introns , K562 Cells , Mice , Nuclear Proteins , RNA Precursors/physiology , RNA Splicing/physiology , RNA, Messenger/genetics , RNA-Binding Proteins , Ribonucleoproteins/physiology , Transcriptome/genetics
SELECTION OF CITATIONS
SEARCH DETAIL