Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 53
Filter
1.
PLoS Genet ; 19(3): e1010656, 2023 03.
Article in English | MEDLINE | ID: mdl-36857387

ABSTRACT

DND1 is essential to maintain germ cell identity. Loss of Dnd1 function results in germ cell differentiation to teratomas in some inbred strains of mice or to somatic fates in zebrafish. Using our knock-in mouse line in which a functional fusion protein between DND1 and GFP is expressed from the endogenous locus (Dnd1GFP), we distinguished two male germ cell (MGC) populations during late gestation cell cycle arrest (G0), consistent with recent reports of heterogeneity among MGCs. Most MGCs express lower levels of DND1-GFP (DND1-GFP-lo), but some MGCs express elevated levels of DND1-GFP (DND1-GFP-hi). A RNA-seq time course confirmed high Dnd1 transcript levels in DND1-GFP-hi cells along with 5-10-fold higher levels for multiple epigenetic regulators. Using antibodies against DND1-GFP for RNA immunoprecipitation (RIP)-sequencing, we identified multiple epigenetic and translational regulators that are binding targets of DND1 during G0 including DNA methyltransferases (Dnmts), histone deacetylases (Hdacs), Tudor domain proteins (Tdrds), actin dependent regulators (Smarcs), and a group of ribosomal and Golgi proteins. These data suggest that in DND1-GFP-hi cells, DND1 hosts coordinating mRNA regulons that consist of functionally related and localized groups of epigenetic enzymes and translational components.


Subject(s)
Spermatogonia , Zebrafish , Animals , Female , Male , Mice , Pregnancy , Chromatin/metabolism , Neoplasm Proteins/genetics , RNA-Binding Proteins/genetics , Spermatogonia/metabolism , Zebrafish/genetics , Zebrafish/metabolism
2.
Biol Reprod ; 104(4): 861-874, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33394034

ABSTRACT

In vertebrates, the RNA-binding protein (RBP) dead end 1 (DND1) is essential for primordial germ cell (PGC) survival and maintenance of cell identity. In multiple species, Dnd1 loss or mutation leads to severe PGC loss soon after specification or, in some species, germ cell transformation to somatic lineages. Our investigations into the role of DND1 in PGC specification and differentiation have been limited by the absence of an available antibody. To address this problem, we used CRISPR/Cas9 gene editing to establish a transgenic mouse line carrying a DND1GFP fusion allele. We present imaging analysis of DND1GFP expression showing that DND1GFP expression is heterogeneous among male germ cells (MGCs) and female germ cells (FGCs). DND1GFP was detected in MGCs throughout fetal life but lost from FGCs at meiotic entry. In postnatal and adult testes, DND1GFP expression correlated with classic markers for the premeiotic spermatogonial population. Utilizing the GFP tag for RNA immunoprecipitation (RIP) analysis in MGCs validated this transgenic as a tool for identifying in vivo transcript targets of DND1. The DND1GFP mouse line is a novel tool for isolation and analysis of embryonic and fetal germ cells, and the spermatogonial population of the postnatal and adult testis.


Subject(s)
Germ Cells/metabolism , Green Fluorescent Proteins/genetics , Neoplasm Proteins/genetics , Alleles , Animals , Cell Differentiation/genetics , Embryo, Mammalian , Female , Gene Expression Regulation, Developmental , Genes, Reporter , Germ Cells/physiology , Green Fluorescent Proteins/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neoplasm Proteins/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Recombinant Fusion Proteins/genetics
3.
Development ; 146(19)2019 07 25.
Article in English | MEDLINE | ID: mdl-31253634

ABSTRACT

The adult spermatogonial stem cell population arises from pluripotent primordial germ cells (PGCs) that enter the fetal testis around embryonic day (E)10.5. PGCs undergo rapid mitotic proliferation, then enter prolonged cell cycle arrest (G1/G0), during which they transition to pro-spermatogonia. In mice homozygous for the Ter mutation in the RNA-binding protein Dnd1 (Dnd1Ter/Ter ), many male germ cells (MGCs) fail to enter G1/G0 and instead form teratomas: tumors containing many embryonic cell types. To investigate the origin of these tumors, we sequenced the MGC transcriptome in Dnd1Ter/Ter mutants at E12.5, E13.5 and E14.5, immediately prior to teratoma formation, and correlated this information with DO-RIP-Seq-identified DND1 direct targets. Consistent with previous results, we found DND1 controls downregulation of many genes associated with pluripotency and active cell cycle, including mTor, Hippo and Bmp/Nodal signaling pathway elements. However, DND1 targets also include genes associated with male differentiation, including a large group of chromatin regulators activated in wild-type but not mutant MGCs during the E13.5 and E14.5 transition. Results suggest multiple DND1 functions and link DND1 to initiation of epigenetic modifications in MGCs.


Subject(s)
Cellular Reprogramming/genetics , Epigenesis, Genetic , Germ Cells/cytology , Germ Cells/metabolism , Neoplasm Proteins/metabolism , Pluripotent Stem Cells/metabolism , RNA-Binding Proteins/metabolism , Animals , Apoptosis/genetics , Base Sequence , Cell Cycle/genetics , Chromatin/metabolism , DNA Transposable Elements/genetics , Down-Regulation/genetics , Embryo, Mammalian/cytology , Female , Homozygote , Male , Mice , Mutation/genetics , Repressor Proteins/metabolism , Signal Transduction/genetics , Transcription, Genetic , Up-Regulation/genetics
4.
iScience ; 4: 97-108, 2018 Jun 29.
Article in English | MEDLINE | ID: mdl-30240757

ABSTRACT

Epithelial-to-mesenchymal transition (EMT) is integral to cancer progression, with considerable evidence that EMT has multiple intermediary stages. Understanding the mechanisms of this stepwise activation is of great interest. We recreated a genetically defined model in which primary cells were immortalized, resulting in migratory capacity, and subsequently H-Ras-transformed, causing malignancy and invasion. To determine the mechanisms coordinating stepwise malignancy, we quantified the changes in messenger RNA (mRNA) and protein abundance. During immortalization, we found dramatic changes in mRNA, consistent with EMT, which correlated with protein abundance. Many of these same proteins also changed following Ras transformation, suggesting that pre-malignant cells were primed for malignant conversion. Unexpectedly, changes in protein abundance did not correlate with changes in mRNA following transformation. Importantly, proteins involved in cellular adhesion and cytoskeletal structure decreased during immortalization and decreased further following Ras transformation, whereas their encoding mRNAs only changed during the immortalization step. Thus, Ras induced EMT-associated invasion via post-transcriptional mechanisms in primed pre-malignant cells.

5.
Cell Physiol Biochem ; 48(3): 1215-1229, 2018.
Article in English | MEDLINE | ID: mdl-30045014

ABSTRACT

BACKGROUND/AIMS: Myofibroblasts (MF) derived from quiescent nonfibrogenic hepatic stellate cells (HSC) are the major sources of fibrous matrix in cirrhosis. Because many factors interact to regulate expansion and regression of MF-HSC populations, efforts to prevent cirrhosis by targeting any one factor have had limited success, motivating research to identify mechanisms that integrate these diverse inputs. As key components of RNA regulons, RNA binding proteins (RBPs) may fulfill this function by orchestrating changes in the expression of multiple genes that must be coordinately regulated to affect the complex phenotypic modifications required for HSC transdifferentiation. METHODS: We profiled the transcriptomes of quiescent and MF-HSC to identify RBPs that were differentially-expressed during HSC transdifferentiation, manipulated the expression of the most significantly induced RBP, insulin like growth factor 2 binding protein 3 (Igf2bp3), and evaluated transcriptomic and phenotypic effects. RESULTS: Depleting Igf2bp3 changed the expression of thousands of HSC genes, including multiple targets of TGF-ß signaling, and caused HSCs to reacquire a less proliferative, less myofibroblastic phenotype. RNA immunoprecipitation assays demonstrated that some of these effects were mediated by direct physical interactions between Igf2bp3 and mRNAs that control proliferative activity and mesenchymal traits. Inhibiting TGF-ß receptor-1 signaling revealed a microRNA-dependent mechanism that induces Igf2bp3. CONCLUSIONS: The aggregate results indicate that HSC transdifferentiation is ultimately dictated by Igf2bp3-dependent RNA regulons and thus, can be controlled simply by manipulating Igf2bp3.


Subject(s)
Cell Transdifferentiation , Gene Expression Regulation , Hepatic Stellate Cells/cytology , Myofibroblasts/cytology , RNA-Binding Proteins/genetics , Transcriptome , Animals , Cells, Cultured , Hepatic Stellate Cells/metabolism , Humans , Male , Mice, Inbred C57BL , Myofibroblasts/metabolism , RNA-Binding Proteins/metabolism
6.
Methods ; 126: 193-200, 2017 08 15.
Article in English | MEDLINE | ID: mdl-28529064

ABSTRACT

Post-transcriptional regulation of gene expression by RNA binding proteins (RBPs) and non-coding RNAs plays an important role in global gene expression. Many post-transcriptional regulators are misexpressed and misregulated in cancers, resulting in altered programs of protein biosynthesis that can drive tumor progression. While comparative studies of several RBPs and microRNAs expressed in various cancer types have been reported, a model system that can be used to quantify RBP regulation and functional outcomes during the initiation and early stages of tumorigenesis is lacking. It was previously demonstrated that oncogenic transformation of normal human cells can be induced by expressing hTERT, p53DD, cyclin D1, CDK4R24C, C-MYCT58A and H-RASG12V. Here we describe a user-friendly method for generating this genetically defined model of step-wise tumorigenesis beginning with normal donor-derived human cells. This method immortalizes a donor's normal cells in about a week, reducing the chances of senescence. The entire stable system can be established in less than 12weeks. We then demonstrate the utility of such a system in elucidating the expression of multiple RBPs at an early step of tumor formation. We identify significant changes in the expression levels of transcripts encoding RBPs prior to transformation, suggesting that our described donor-derived isogenic system can provide insight about post-transcriptional regulation during the earliest stages of tumorigenesis in the context of diverse genetic backgrounds.


Subject(s)
Carcinogenesis/genetics , Disease Progression , Neoplasms/genetics , RNA Processing, Post-Transcriptional/physiology , RNA-Binding Proteins/genetics , Carcinogenesis/metabolism , Cell Line, Transformed , HEK293 Cells , Humans , Neoplasms/metabolism , RNA-Binding Proteins/metabolism
7.
RNA ; 23(1): 32-46, 2017 01.
Article in English | MEDLINE | ID: mdl-27742911

ABSTRACT

RNA-binding proteins (RBPs) and noncoding RNAs orchestrate post-transcriptional processes through the recognition of specific sites on targeted transcripts. Thus, understanding the connection between binding to specific sites and active regulation of the whole transcript is essential. Many immunoprecipitation techniques have been developed that identify either whole transcripts or binding sites of RBPs on each transcript using cell lysates. However, none of these methods simultaneously measures the strength of each binding site and quantifies binding to whole transcripts. In this study, we compare current procedures and present digestion optimized (DO)-RIP-seq, a simple method that locates and quantifies RBP binding sites using a continuous metric. We have used the RBP HuR/ELAVL1 to demonstrate that DO-RIP-seq can quantify HuR binding sites with high coverage across the entire human transcriptome, thereby generating metrics of relative RNA binding strength. We demonstrate that this quantitative enrichment of binding sites is proportional to the relative in vitro binding strength for these sites. In addition, we used DO-RIP-seq to quantify and compare HuR's binding to whole transcripts, thus allowing for seamless integration of binding site data with whole-transcript measurements. Finally, we demonstrate that DO-RIP-seq is useful for identifying functional mRNA target sets and binding sites where combinatorial interactions between HuR and AGO-microRNAs regulate the fate of the transcripts. Our data indicate that DO-RIP-seq will be useful for quantifying RBP binding events that regulate dynamic biological processes.


Subject(s)
ELAV-Like Protein 1/metabolism , Gene Expression Profiling/methods , MicroRNAs/genetics , RNA, Messenger/genetics , Sequence Analysis, RNA/methods , Binding Sites , Databases, Genetic , ELAV-Like Protein 1/chemistry , Gene Expression Regulation , HEK293 Cells , High-Throughput Nucleotide Sequencing , Humans , Protein Binding
8.
Methods ; 118-119: 16-23, 2017 04 15.
Article in English | MEDLINE | ID: mdl-27840290

ABSTRACT

Post-transcriptional processes orchestrate gene expression through dynamic protein-RNA interactions. These interactions occur at specific sites determined by RNA sequence, secondary structure, or nucleotide modifications. Methods have been developed either to quantify binding of whole transcripts or to identify the binding sites, but there is none proven to quantify binding at both the whole transcript and binding site levels. Here we describe digestion optimized RNA immunoprecipitation with deep sequencing (DO-RIP-seq) as a method that quantitates at the whole transcript target (RIP-Seq-Like or RSL) level and at the binding site level (BSL) using continuous metrics. DO-RIP-seq methodology was developed using the RBP HuR/ELAVL1 as a test case (Nicholson et al., 2016). DO-RIP-seq employs treatment of cell lysates with a nuclease under optimized conditions to yield partially digested RNA fragments bound by RNA binding proteins, followed by immunoprecipitations that capture the digested RNA-protein complexes and assess non-specific or background interactions. Analyses of sequenced cDNA libraries made from the bound RNA fragments yielded two types of enrichment scores; one for RSL binding events and the other for BSL events (Nicholson et al., 2016). These analyses plus the extensive read coverage of DO-RIP-seq allows seamless integration of binding site and whole transcript information. Therefore, DO-RIP-seq is useful for quantifying RBP binding events that are regulated during dynamic biological processes.


Subject(s)
ELAV-Like Protein 1/genetics , High-Throughput Nucleotide Sequencing/methods , Immunoprecipitation/methods , RNA-Binding Proteins/genetics , RNA/chemistry , Transcriptome , Antibodies/chemistry , Base Sequence , Binding Sites , ELAV-Like Protein 1/metabolism , Gene Library , HEK293 Cells , Humans , Nucleic Acid Conformation , Protein Binding , RNA/genetics , RNA/metabolism , RNA-Binding Proteins/metabolism , Ribonucleases/chemistry , Sequence Analysis, RNA
9.
Proc Natl Acad Sci U S A ; 113(37): 10322-7, 2016 09 13.
Article in English | MEDLINE | ID: mdl-27578869

ABSTRACT

The 18-kb Xist long noncoding RNA (lncRNA) is essential for X-chromosome inactivation during female eutherian mammalian development. Global structural architecture, cell-induced conformational changes, and protein-RNA interactions within Xist are poorly understood. We used selective 2'-hydroxyl acylation analyzed by primer extension and mutational profiling (SHAPE-MaP) to examine these features of Xist at single-nucleotide resolution both in living cells and ex vivo. The Xist RNA forms complex well-defined secondary structure domains and the cellular environment strongly modulates the RNA structure, via motifs spanning one-half of all Xist nucleotides. The Xist RNA structure modulates protein interactions in cells via multiple mechanisms. For example, repeat-containing elements adopt accessible and dynamic structures that function as landing pads for protein cofactors. Structured RNA motifs create interaction domains for specific proteins and also sequester other motifs, such that only a subset of potential binding sites forms stable interactions. This work creates a broad quantitative framework for understanding structure-function interrelationships for Xist and other lncRNAs in cells.


Subject(s)
Nucleic Acid Conformation , RNA, Long Noncoding/genetics , RNA-Binding Proteins/genetics , Acylation/genetics , Animals , Female , Mice , Mutation , RNA, Long Noncoding/chemistry , RNA-Binding Proteins/chemistry , X Chromosome/genetics , X Chromosome Inactivation/genetics
10.
Nucleic Acids Res ; 44(1): 426-36, 2016 Jan 08.
Article in English | MEDLINE | ID: mdl-26490963

ABSTRACT

Global mRNA abundance depends on the balance of synthesis and decay of a population of mRNAs. To account for this balance during activation of T cells, we used metabolic labeling to quantify the contributions of RNA transcription and decay over a 4 h time course during activation of leukemia-derived Jurkat T cells. While prior studies suggested more than half of the changes in mRNA abundance were due to RNA stability, we found a smaller but more interesting population of mRNAs changed stability. These mRNAs clustered into functionally related subpopulations that included replicative histones, ribosomal biogenesis and cell motility functions. We then applied a novel analysis based on integrating global protein-RNA binding with concurrent changes in RNA stability at specific time points following activation. This analysis demonstrated robust stabilization of mRNAs by the HuR RNA-binding protein 4 h after activation. Our unexpected findings demonstrate that the temporal regulation of mRNA stability coordinates vital cellular pathways and is in part controlled by the HuR RNA binding protein in Jurkat T cells following activation.


Subject(s)
ELAV-Like Protein 1/metabolism , Lymphocyte Activation/genetics , RNA Stability , RNA, Messenger/genetics , T-Lymphocytes/metabolism , Histones/genetics , Histones/metabolism , Humans , Jurkat Cells , Lymphocyte Activation/immunology , RNA, Messenger/metabolism , T-Lymphocytes/immunology , Transcription, Genetic
11.
PLoS Pathog ; 11(3): e1004708, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25747802

ABSTRACT

We demonstrate that both Hepatitis C virus (HCV) and Bovine Viral Diarrhea virus (BVDV) contain regions in their 5' UTRs that stall and repress the enzymatic activity of the cellular 5'-3' exoribonuclease XRN1, resulting in dramatic changes in the stability of cellular mRNAs. We used biochemical assays, virus infections, and transfection of the HCV and BVDV 5' untranslated regions in the absence of other viral gene products to directly demonstrate the existence and mechanism of this novel host-virus interaction. In the context of HCV infection, we observed globally increased stability of mRNAs resulting in significant increases in abundance of normally short-lived mRNAs encoding a variety of relevant oncogenes and angiogenesis factors. These findings suggest that non-coding regions from multiple genera of the Flaviviridae interfere with XRN1 and impact post-transcriptional processes, causing global dysregulation of cellular gene expression which may promote cell growth and pathogenesis.


Subject(s)
5' Untranslated Regions , Diarrhea Viruses, Bovine Viral/pathogenicity , Exoribonucleases/metabolism , Hepacivirus/pathogenicity , Host-Parasite Interactions/genetics , Microtubule-Associated Proteins/metabolism , RNA Stability/genetics , Virus Replication/genetics , 5' Untranslated Regions/genetics , Animals , Blotting, Western , Cattle , Cell Line , Diarrhea Viruses, Bovine Viral/genetics , Hepacivirus/genetics , Humans , Polymerase Chain Reaction , RNA, Messenger , Transfection
12.
Semin Cell Dev Biol ; 34: 44-54, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24882724

ABSTRACT

The cellular growth cycle is initiated and maintained by punctual, yet agile, regulatory events involving modifications of cell cycle proteins as well as coordinated gene expression to support cyclic checkpoint decisions. Recent evidence indicates that post-transcriptional partitioning of messenger RNA subsets by RNA-binding proteins help physically localize, temporally coordinate, and efficiently translate cell cycle proteins. This dynamic organization of mRNAs encoding cell cycle components contributes to the overall economy of the cell cycle consistent with the post-transcriptional RNA regulon model of gene expression. This review examines several recent studies demonstrating the coordination of mRNA subsets encoding cell cycle proteins during nuclear export and subsequent coupling to protein synthesis, and discusses evidence for mRNA coordination of p53 targets and the DNA damage response pathway. We consider how these observations may connect to upstream and downstream post-transcriptional coordination and coupling of splicing, export, localization, and translation. Published examples from yeast, nematode, insect, and mammalian systems are discussed, and we consider genetic evidence supporting the conclusion that dysregulation of RNA regulons may promote pathogenic states of growth such as carcinogenesis.


Subject(s)
Cell Cycle , Cell Proliferation , RNA, Messenger/metabolism , Active Transport, Cell Nucleus , Animals , DNA Damage , Gene Expression , Humans , RNA Interference , RNA Transport , RNA, Messenger/genetics , Regulon
13.
J Virol ; 88(17): 9514-28, 2014 Sep 01.
Article in English | MEDLINE | ID: mdl-24899193

ABSTRACT

UNLABELLED: The ability of CD8+ T cells to effectively limit HIV-1 replication and block HIV-1 acquisition is determined by the capacity to rapidly respond to HIV-1 antigens. Understanding both the functional properties and regulation of an effective CD8+ response would enable better evaluation of T cell-directed vaccine strategies and may inform the design of new therapies. We assessed the antigen specificity, cytokine signature, and mechanisms that regulate antiviral gene expression in CD8+ T cells from a cohort of HIV-1-infected virus controllers (VCs) (<5,000 HIV-1 RNA copies/ml and CD4+ lymphocyte counts of >400 cells/µl) capable of soluble inhibition of HIV-1. Gag p24 and Nef CD8+ T cell-specific soluble virus inhibition was common among the VCs and correlated with substantial increases in the abundance of mRNAs encoding the antiviral cytokines macrophage inflammatory proteins MIP-1α, MIP-1αP (CCL3L1), and MIP-1ß; granulocyte-macrophage colony-stimulating factor (GM-CSF); lymphotactin (XCL1); tumor necrosis factor receptor superfamily member 9 (TNFRSF9); and gamma interferon (IFN-γ). The induction of several of these mRNAs was driven through a coordinated response of both increased transcription and stabilization of mRNA, which together accounted for the observed increase in mRNA abundance. This coordinated response allows rapid and robust induction of mRNA messages that can enhance the CD8+ T cells' ability to inhibit virus upon antigen encounter. IMPORTANCE: We show that mRNA stability, in addition to transcription, is key in regulating the direct anti-HIV-1 function of antigen-specific memory CD8+ T cells. Regulation at the level of RNA helps enable rapid recall of memory CD8+ T cell effector functions for HIV-1 inhibition. By uncovering and understanding the mechanisms employed by CD8+ T cell subsets with antigen-specific anti-HIV-1 activity, we can identify new strategies for comprehensive identification of other important antiviral genes. This will, in turn, enhance our ability to inhibit virus replication by informing both cure strategies and HIV-1 vaccine designs that aim to reduce transmission and can aid in blocking HIV-1 acquisition.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cytokines/biosynthesis , Gene Expression Regulation , HIV Antigens/immunology , HIV-1/immunology , Protein Biosynthesis , Transcription, Genetic , Cohort Studies , Cytokines/genetics , Gene Expression Profiling , HIV Long-Term Survivors , Humans , RNA, Messenger/analysis , RNA, Messenger/genetics
14.
Genome Biol ; 15(1): R2, 2014 Jan 07.
Article in English | MEDLINE | ID: mdl-24393468

ABSTRACT

BACKGROUND: Sequence specific RNA binding proteins are important regulators of gene expression. Several related crosslinking-based, high-throughput sequencing methods, including PAR-CLIP, have recently been developed to determine direct binding sites of global protein-RNA interactions. However, no studies have quantitatively addressed the contribution of background binding to datasets produced by these methods. RESULTS: We measured non-specific RNA background in PAR-CLIP data, demonstrating that covalently crosslinked background binding is common, reproducible and apparently universal among laboratories. We show that quantitative determination of background is essential for identifying targets of most RNA-binding proteins and can substantially improve motif analysis. We also demonstrate that by applying background correction to an RNA binding protein of unknown binding specificity, Caprin1, we can identify a previously unrecognized RNA recognition element not otherwise apparent in a PAR-CLIP study. CONCLUSIONS: Empirical background measurements of global RNA-protein crosslinking are a necessary addendum to other experimental controls, such as performing replicates, because covalently crosslinked background signals are reproducible and otherwise unavoidable. Recognizing and quantifying the contribution of background extends the utility of PAR-CLIP and can improve mechanistic understanding of protein-RNA specificity, protein-RNA affinity and protein-RNA association dynamics.


Subject(s)
High-Throughput Nucleotide Sequencing/methods , RNA, Long Noncoding/genetics , RNA, Messenger/genetics , Binding Sites , Chromosome Mapping , HEK293 Cells , Humans , RNA, Long Noncoding/metabolism , RNA, Messenger/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Sequence Analysis, RNA/methods
15.
Natl Sci Rev ; 1(2): 184-186, 2014 Jun.
Article in English | MEDLINE | ID: mdl-28078165
16.
Curr Opin Genet Dev ; 23(1): 35-43, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23312841

ABSTRACT

The 5' and 3' untranslated regions (UTRs) of messenger RNAs (mRNAs) function as platforms that can determine the fate of each mRNA individually and in aggregate. Multiple mRNAs that encode proteins that are functionally related often interact with RNA-binding proteins (RBPs) and noncoding RNAs (ncRNAs) that coordinate their expression in time and space as RNA regulons within the ribonucleoprotein (RNP) infrastructure we term the ribonome. Recent ribonomic methods have emerged that can determine which mRNAs are bound and regulated by RBPs and ncRNAs, some of which act in combination to determine global outcomes. ELAV/Hu proteins bind to AU-rich elements (ARE) in mRNAs and regulate their stability from splicing to translation, and the ubiquitous HuR protein has been implicated in cancerous cell growth. Recent work is focused on mechanistic models of how ELAV/Hu proteins increase mRNA stability and translation by repressing microRNAs (miRs) and the RNA induced silencing complex (RISC) via ARE-based ribonucleosomes that may affect global functions of mRNA regulons.


Subject(s)
3' Untranslated Regions/genetics , 5' Untranslated Regions/genetics , ELAV Proteins/genetics , Regulon , ELAV Proteins/metabolism , Gene Expression Regulation , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , RNA Splicing , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Ribonucleoproteins/genetics , Ribonucleoproteins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
17.
Nucleic Acids Res ; 40(6): 2734-46, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22139917

ABSTRACT

The ubiquitously expressed RNA-binding protein HuR increases the stability and translation of mRNAs encoding growth regulatory proteins that promote proliferation in a variety of cell types. However, the three neuron-specific ELAV/Hu proteins, HuB, HuC and HuD, while binding to the same types of mRNAs, are required instead for neuronal differentiation, and it becomes difficult to reconcile these contrary functions when all four Hu proteins are expressed in the same neuron. HuR mRNA exists as three alternatively polyadenylated variants, a 1.5-kb testes-specific mRNA isoform, a ubiquitous 2.4-kb isoform and a 6.0-kb isoform that we now show is induced during neuronal differentiation and appears to be neuron-specific. This 6.0-kb neuron-specific mRNA isoform is inherently less stable and produces less HuR protein than the ubiquitous 2.4-kb mRNA. Furthermore, we show that neuronal HuB, HuC and HuD, as well as HuR itself, can bind at the 2.4-kb mRNA polyadenylation site, and when overexpressed can affect alternative polyadenylation to generate an extended HuR 3'-UTR that is translationally suppressed. We propose that the regulation of HuR protein expression by alternative polyadenylation allows neurons to post-transcriptionally regulate mRNAs-encoding factors required for proliferation versus differentiation to facilitate neuronal differentiation.


Subject(s)
ELAV Proteins/genetics , Neurogenesis/genetics , Neurons/metabolism , Polyadenylation , 3' Untranslated Regions , Cell Line , ELAV Proteins/biosynthesis , ELAV Proteins/metabolism , Gene Expression Regulation , Humans , Neurons/cytology , Protein Biosynthesis , RNA Isoforms/metabolism , RNA Stability
18.
Genome Biol ; 12(8): R79, 2011 Aug 18.
Article in English | MEDLINE | ID: mdl-21851591

ABSTRACT

Crosslinking and immunoprecipitation (CLIP) protocols have made it possible to identify transcriptome-wide RNA-protein interaction sites. In particular, PAR-CLIP utilizes a photoactivatable nucleoside for more efficient crosslinking. We present an approach, centered on the novel PARalyzer tool, for mapping high-confidence sites from PAR-CLIP deep-sequencing data. We show that PARalyzer delineates sites with a high signal-to-noise ratio. Motif finding identifies the sequence preferences of RNA-binding proteins, as well as seed-matches for highly expressed microRNAs when profiling Argonaute proteins. Our study describes tailored analytical methods and provides guidelines for future efforts to utilize high-throughput sequencing in RNA biology. PARalyzer is available at http://www.genome.duke.edu/labs/ohler/research/PARalyzer/.


Subject(s)
Binding Sites/genetics , Immunoprecipitation/methods , RNA/isolation & purification , Sequence Analysis, RNA/methods , Argonaute Proteins/genetics , Argonaute Proteins/metabolism , Databases, Genetic , High-Throughput Nucleotide Sequencing/methods , Humans , Immunoprecipitation/instrumentation , Linear Models , MicroRNAs/genetics , MicroRNAs/metabolism , RNA/metabolism , Signal-To-Noise Ratio , Transcriptome
19.
Mol Cell ; 43(3): 327-39, 2011 Aug 05.
Article in English | MEDLINE | ID: mdl-21723170

ABSTRACT

RNA-binding proteins coordinate the fates of multiple RNAs, but the principles underlying these global interactions remain poorly understood. We elucidated regulatory mechanisms of the RNA-binding protein HuR, by integrating data from diverse high-throughput targeting technologies, specifically PAR-CLIP, RIP-chip, and whole-transcript expression profiling. The number of binding sites per transcript, degree of HuR association, and degree of HuR-dependent RNA stabilization were positively correlated. Pre-mRNA and mature mRNA containing both intronic and 3' UTR binding sites were more highly stabilized than transcripts with only 3' UTR or only intronic binding sites, suggesting that HuR couples pre-mRNA processing with mature mRNA stability. We also observed HuR-dependent splicing changes and substantial binding of HuR in polypyrimidine tracts of pre-mRNAs. Comparison of the spatial patterns surrounding HuR and miRNA binding sites provided functional evidence for HuR-dependent antagonism of proximal miRNA-mediated repression. We conclude that HuR coordinates gene expression outcomes at multiple interconnected steps of RNA processing.


Subject(s)
Antigens, Surface/metabolism , RNA Precursors/metabolism , RNA Processing, Post-Transcriptional , RNA Stability , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism , Amino Acid Motifs , Antigens, Surface/chemistry , Antigens, Surface/physiology , Binding Sites , Computational Biology , ELAV Proteins , ELAV-Like Protein 1 , Gene Expression Regulation , HEK293 Cells , Humans , MicroRNAs/metabolism , MicroRNAs/physiology , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/physiology , Software
20.
Blood ; 117(8): 2441-50, 2011 Feb 24.
Article in English | MEDLINE | ID: mdl-21209379

ABSTRACT

Maintenance of genomic stability depends on the DNA damage response, a biologic barrier in early stages of cancer development. Failure of this response results in genomic instability and high predisposition toward lymphoma, as seen in patients with ataxia-telangiectasia mutated (ATM) dysfunction. ATM activates multiple cell-cycle checkpoints and DNA repair after DNA damage, but its influence on posttranscriptional gene expression has not been examined on a global level. We show that ionizing radiation modulates the dynamic association of the RNA-binding protein HuR with target mRNAs in an ATM-dependent manner, potentially coordinating the genotoxic response as an RNA operon. Pharmacologic ATM inhibition and use of ATM-null cells revealed a critical role for ATM in this process. Numerous mRNAs encoding cancer-related proteins were differentially associated with HuR depending on the functional state of ATM, in turn affecting expression of encoded proteins. The findings presented here reveal a previously unidentified role of ATM in controlling gene expression posttranscriptionally. Dysregulation of this DNA damage response RNA operon is probably relevant to lymphoma development in ataxia-telangiectasia persons. These novel RNA regulatory modules and genetic networks provide critical insight into the function of ATM in oncogenesis.


Subject(s)
Cell Cycle Proteins/genetics , DNA Damage , DNA-Binding Proteins/genetics , Gene Expression Regulation , Lymphocytes/metabolism , Operon/genetics , Protein Serine-Threonine Kinases/genetics , Tumor Suppressor Proteins/genetics , Antigens, Surface/metabolism , Ataxia Telangiectasia Mutated Proteins , DNA Repair , ELAV Proteins , ELAV-Like Protein 1 , Gene Regulatory Networks , Humans , Lymphoma/etiology , Mutant Proteins , Protein Binding/radiation effects , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism , Radiation, Ionizing
SELECTION OF CITATIONS
SEARCH DETAIL
...