Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters










Publication year range
1.
Mol Cell ; 74(6): 1189-1204.e6, 2019 06 20.
Article in English | MEDLINE | ID: mdl-31226278

ABSTRACT

RNA-binding proteins (RBPs) regulate post-transcriptional gene expression by recognizing short and degenerate sequence motifs in their target transcripts, but precisely defining their binding specificity remains challenging. Crosslinking and immunoprecipitation (CLIP) allows for mapping of the exact protein-RNA crosslink sites, which frequently reside at specific positions in RBP motifs at single-nucleotide resolution. Here, we have developed a computational method, named mCross, to jointly model RBP binding specificity while precisely registering the crosslinking position in motif sites. We applied mCross to 112 RBPs using ENCODE eCLIP data and validated the reliability of the discovered motifs by genome-wide analysis of allelic binding sites. Our analyses revealed that the prototypical SR protein SRSF1 recognizes clusters of GGA half-sites in addition to its canonical GGAGGA motif. Therefore, SRSF1 regulates splicing of a much larger repertoire of transcripts than previously appreciated, including HNRNPD and HNRNPDL, which are involved in multivalent protein assemblies and phase separation.


Subject(s)
Heterogeneous-Nuclear Ribonucleoprotein D/chemistry , Models, Molecular , RNA/chemistry , Serine-Arginine Splicing Factors/chemistry , Base Sequence , Binding Sites , Cross-Linking Reagents/chemistry , Gene Expression , HeLa Cells , Hep G2 Cells , Heterogeneous Nuclear Ribonucleoprotein D0 , Heterogeneous-Nuclear Ribonucleoprotein D/genetics , Heterogeneous-Nuclear Ribonucleoprotein D/metabolism , Humans , K562 Cells , Nucleic Acid Conformation , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , RNA/genetics , RNA/metabolism , Sequence Alignment , Sequence Homology, Nucleic Acid , Serine-Arginine Splicing Factors/genetics , Serine-Arginine Splicing Factors/metabolism
2.
RNA Biol ; 16(7): 960-971, 2019 07.
Article in English | MEDLINE | ID: mdl-30951406

ABSTRACT

The RNA-binding protein AUF1 regulates post-transcriptional gene expression by affecting the steady state and translation levels of numerous target RNAs. Remodeling of RNA structures by the largest isoform AUF1 p45 was recently demonstrated in the context of replicating RNA viruses, and involves two RNA remodeling activities, i.e. an RNA chaperone and an RNA annealing activity. AUF1 contains two non-identical RNA recognition motifs (RRM) and one RGG/RG motif located in the C-terminus. In order to determine the functional significance of each motif to AUF1's RNA-binding and remodeling activities we performed a comprehensive mutagenesis study and characterized the wildtype AUF1, and several variants thereof. We demonstrate that each motif contributes to efficient RNA binding and remodeling by AUF1 indicating a tight cooperation of the RRMs and the RGG/RG motif. Interestingly, the data identify two distinct roles for the arginine residues of the RGG/RG motif for each RNA remodeling activity. First, arginine-mediated stacking interactions promote AUF1's helix-destabilizing RNA chaperone activity. Second, the electropositive character of the arginine residues is the major driving force for the RNA annealing activity. Thus, we provide the first evidence that arginine residues of an RGG/RG motif contribute to the mechanism of RNA annealing and RNA chaperoning.


Subject(s)
Heterogeneous-Nuclear Ribonucleoprotein D/chemistry , Heterogeneous-Nuclear Ribonucleoprotein D/metabolism , RNA/metabolism , Amino Acid Motifs , Arginine/metabolism , Base Sequence , Heterogeneous Nuclear Ribonucleoprotein D0 , Protein Binding , Protein Isoforms/chemistry , Protein Isoforms/metabolism , RNA/chemistry , RNA/genetics , Structure-Activity Relationship , Thermodynamics
3.
J Virol ; 92(6)2018 03 15.
Article in English | MEDLINE | ID: mdl-29263261

ABSTRACT

In previous studies, we showed that the cellular RNA-binding protein AUF1 supports the replication process of the flavivirus West Nile virus. Here we demonstrate that the protein also enables effective proliferation of dengue virus and Zika virus, indicating that AUF1 is a general flavivirus host factor. Further studies demonstrated that the AUF1 isoform p45 significantly stimulates the initiation of viral RNA replication and that the protein's RNA chaperone activity enhances the interactions of the viral 5'UAR and 3'UAR genome cyclization sequences. Most interestingly, we observed that AUF1 p45 destabilizes not only the 3'-terminal stem-loop (3'SL) but also 5'-terminal stem-loop B (SLB) of the viral genome. RNA structure analyses revealed that AUF1 p45 increases the accessibility of defined nucleotides within the 3'SL and SLB and, in this way, exposes both UAR cyclization elements. Conversely, AUF1 p45 does not modulate the fold of stem-loop A (SLA) at the immediate genomic 5' end, which is proposed to function as a promoter of the viral RNA-dependent RNA polymerase (RdRp). These findings suggest that AUF1 p45, by destabilizing specific stem-loop structures within the 5' and 3' ends of the flaviviral genome, assists genome cyclization and concurrently enables the RdRp to initiate RNA synthesis. Our study thus highlights the role of a cellular RNA-binding protein inducing a flaviviral RNA switch that is crucial for viral replication.IMPORTANCE The genus Flavivirus within the Flaviviridae family includes important human pathogens, such as dengue, West Nile, and Zika viruses. The initiation of replication of the flaviviral RNA genome requires a transformation from a linear to a cyclized form. This involves considerable structural reorganization of several RNA motifs at the genomic 5' and 3' ends. Specifically, it needs a melting of stem structures to expose complementary 5' and 3' cyclization elements to enable their annealing during cyclization. Here we show that a cellular RNA chaperone, AUF1 p45, which supports the replication of all three aforementioned flaviviruses, specifically rearranges stem structures at both ends of the viral genome and in this way permits 5'-3' interactions of cyclization elements. Thus, AUF1 p45 triggers the RNA switch in the flaviviral genome that is crucial for viral replication. These findings represent an important example of how cellular (host) factors promote the propagation of RNA viruses.


Subject(s)
Flavivirus/physiology , Genome, Viral , Heterogeneous-Nuclear Ribonucleoprotein D/metabolism , Nucleic Acid Conformation , RNA, Viral/metabolism , Virus Replication/physiology , Heterogeneous Nuclear Ribonucleoprotein D0 , Heterogeneous-Nuclear Ribonucleoprotein D/chemistry , Heterogeneous-Nuclear Ribonucleoprotein D/genetics , Humans , RNA, Viral/chemistry , RNA, Viral/genetics , RNA-Dependent RNA Polymerase/chemistry , RNA-Dependent RNA Polymerase/genetics , RNA-Dependent RNA Polymerase/metabolism , Viral Proteins/chemistry , Viral Proteins/genetics , Viral Proteins/metabolism
4.
Biomed Res Int ; 2016: 3286191, 2016.
Article in English | MEDLINE | ID: mdl-27437398

ABSTRACT

AU-rich element binding/degradation factor 1 (AUF1) plays a role in destabilizing mRNAs by forming complexes with AU-rich elements (ARE) in the 3'-untranslated regions. Multiple AUF1-ARE complexes regulate the translation of encoded products related to the cell cycle, apoptosis, and inflammation. AUF1 contains two tandem RNA recognition motifs (RRM) and a Gln- (Q-) rich domain in their C-terminal region. To observe how the two RRMs are involved in recognizing ARE, we obtained the AUF1-p37 protein covering the two RRMs. However, only N-terminal RRM (RRM1) was crystallized and its structure was determined at 1.7 Å resolution. It appears that the RRM1 and RRM2 separated before crystallization. To demonstrate which factors affect the separate RRM1-2, we performed limited proteolysis using trypsin. The results indicated that the intact proteins were cleaved by unknown proteases that were associated with them prior to crystallization. In comparison with each of the monomers, the conformations of the ß2-ß3 loops were highly variable. Furthermore, a comparison with the RRM1-2 structures of HuR and hnRNP A1 revealed that a dimer of RRM1 could be one of the possible conformations of RRM1-2. Our data may provide a guidance for further structural investigations of AUF1 tandem RRM repeat and its mode of ARE binding.


Subject(s)
Heterogeneous-Nuclear Ribonucleoprotein D/chemistry , Heterogeneous-Nuclear Ribonucleoprotein D/genetics , 3' Untranslated Regions , Apoptosis , Crystallography, X-Ray , ELAV-Like Protein 1/chemistry , Escherichia coli/metabolism , Heterogeneous Nuclear Ribonucleoprotein D0 , Heterogeneous-Nuclear Ribonucleoproteins/chemistry , Humans , Inflammation , Protein Domains , Protein Structure, Secondary , Proteolysis , RNA Stability , Ribonucleases/chemistry , Tandem Repeat Sequences
5.
J Biol Chem ; 289(45): 31433-47, 2014 Nov 07.
Article in English | MEDLINE | ID: mdl-25261470

ABSTRACT

miR-141 and miR-146b-5p are two important tumor suppressor microRNAs, which control several cancer-related genes and processes. In the present report, we have shown that these microRNAs bind specific sites at the 3'-untranslated region (UTR) of the mRNA-binding protein AUF1, leading to its down-regulation. This inverse correlation between the levels of these microRNAs and AUF1 has been identified in various osteosarcoma cell lines. Additionally, we present clear evidence that AUF1 promotes mesenchymal features in osteosarcoma cells and that miR-141 and miR-146b-5p suppress this prometastatic process through AUF1 repression. Indeed, both microRNAs suppressed the invasion/migration and proliferation abilities of osteosarcoma cells through inhibiting the AKT protein kinase in an AUF1-dependent manner. We have also shown that AUF1 binds to and stabilizes the mRNA of the AKT activator phosphoinositide-dependent kinase-1 (PDK1). Furthermore, miR-141 and miR-146b-5p positively regulate the epithelial markers (E-cadherin and Epcam) and repress the mesenchymal markers (N-cadherin, Vimentin, Twist2, and ZEB1). These effects were mediated via the repression of the epithelial-to-mesenchymal inducer ZEB1 through targeting AUF1, which binds the 3'-UTR of the ZEB1 mRNA and reduces its turnover. These results indicate that at least some tumor suppressor functions of miR-141 and miR-146b-5p are mediated through the repression of the oncogenic potentials of AUF1. Therefore, these 3'-UTR-directed post-transcriptional gene expression regulators constitute promising new targets for diagnostic and/or therapeutic interventions.


Subject(s)
Heterogeneous-Nuclear Ribonucleoprotein D/chemistry , Homeodomain Proteins/metabolism , MicroRNAs/physiology , Proto-Oncogene Proteins c-akt/metabolism , Transcription Factors/metabolism , 3' Untranslated Regions , Base Sequence , Cell Proliferation , Epithelial-Mesenchymal Transition , Gene Expression Regulation, Neoplastic , Heterogeneous Nuclear Ribonucleoprotein D0 , Heterogeneous-Nuclear Ribonucleoprotein D/physiology , Humans , Mesoderm/cytology , Molecular Sequence Data , Neoplasm Metastasis , Neoplasms/metabolism , Prognosis , Protein Binding , Sequence Homology, Nucleic Acid , Zinc Finger E-box-Binding Homeobox 1
6.
J Biol Chem ; 288(39): 28034-48, 2013 Sep 27.
Article in English | MEDLINE | ID: mdl-23940053

ABSTRACT

AU-rich element RNA-binding protein 1 (AUF1) regulates the stability and/or translational efficiency of diverse mRNA targets, including many encoding products controlling the cell cycle, apoptosis, and inflammation by associating with AU-rich elements residing in their 3'-untranslated regions. Previous biochemical studies showed that optimal AUF1 binding requires 33-34 nucleotides with a strong preference for U-rich RNA despite observations that few AUF1-associated cellular mRNAs contain such extended U-rich domains. Using the smallest AUF1 isoform (p37(AUF1)) as a model, we employed fluorescence anisotropy-based approaches to define thermodynamic parameters describing AUF1 ribonucleoprotein (RNP) complex formation across a panel of RNA substrates. These data demonstrated that 15 nucleotides of AU-rich sequence were sufficient to nucleate high affinity p37(AUF1) RNP complexes within a larger RNA context. In particular, p37(AUF1) binding to short AU-rich RNA targets was significantly stabilized by interactions with a 3'-purine residue and largely base-independent but non-ionic contacts 5' of the AU-rich site. RNP stabilization by the upstream RNA domain was associated with an enhanced negative change in heat capacity consistent with conformational changes in protein and/or RNA components, and fluorescence resonance energy transfer-based assays demonstrated that these contacts were required for p37(AUF1) to remodel local RNA structure. Finally, reporter mRNAs containing minimal high affinity p37(AUF1) target sequences associated with AUF1 and were destabilized in a p37(AUF1)-dependent manner in cells. These findings provide a mechanistic explanation for the diverse population of AUF1 target mRNAs but also suggest how AUF1 binding could regulate protein and/or microRNA binding events at adjacent sites.


Subject(s)
Heterogeneous-Nuclear Ribonucleoprotein D/chemistry , RNA, Messenger/chemistry , Allosteric Site , Binding Sites , Cell Nucleus/metabolism , Fluorescence Resonance Energy Transfer , Heterogeneous Nuclear Ribonucleoprotein A1 , Heterogeneous Nuclear Ribonucleoprotein D0 , Heterogeneous-Nuclear Ribonucleoprotein Group A-B/chemistry , Humans , Nucleic Acid Conformation , Protein Binding , Protein Isoforms , RNA, Messenger/metabolism , Thermodynamics
7.
Mol Cell Biol ; 33(11): 2315-26, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23530064

ABSTRACT

Activation of the mitogen-activated protein (MAP) pathway kinases p38 and MK2 induces phosphorylation of the chaperone Hsp27 and stabilization of mRNAs containing AU-rich elements (AREs) (ARE-mRNAs). Likewise, expression of phosphomimetic mutant forms of Hsp27 also stabilizes ARE-mRNAs. It appears to perform this function by promoting degradation of the ARE-mRNA decay factor AUF1 by proteasomes. In this study, we examined the molecular mechanism linking Hsp27 phosphorylation to AUF1 degradation by proteasomes. AUF1 is a target of ß-TrCP, the substrate recognition subunit of the E3 ubiquitin ligase Skp1-cullin-F-box protein complex, SCF(ß-TrCP). Depletion of ß-TrCP stabilized AUF1. In contrast, overexpression of ß-TrCP enhanced ubiquitination and degradation of AUF1 and led to stabilization of reporter mRNAs containing cytokine AREs. Enhanced AUF1 degradation required expression of phosphomimetic mutant forms of both Hsp27 and AUF1. Our results suggest that a signaling axis composed of p38 MAP kinase-MK2-Hsp27-ß-TrCP may promote AUF1 degradation by proteasomes and stabilization of cytokine ARE-mRNAs.


Subject(s)
HSP27 Heat-Shock Proteins/metabolism , Heterogeneous-Nuclear Ribonucleoprotein D/metabolism , beta-Transducin Repeat-Containing Proteins/metabolism , AU Rich Elements , Gene Expression Regulation , Gene Knockdown Techniques , HSP27 Heat-Shock Proteins/chemistry , HSP27 Heat-Shock Proteins/genetics , HeLa Cells , Heat-Shock Proteins , Heterogeneous Nuclear Ribonucleoprotein D0 , Heterogeneous-Nuclear Ribonucleoprotein D/chemistry , Heterogeneous-Nuclear Ribonucleoprotein D/genetics , Humans , Molecular Chaperones , Mutation , Proteasome Endopeptidase Complex/metabolism , Protein Stability , Signal Transduction , Ubiquitination , beta-Transducin Repeat-Containing Proteins/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
8.
RNA ; 18(11): 2073-82, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23012480

ABSTRACT

Epstein-Barr virus (EBV)-infected cells express two noncoding RNAs called EBV-encoded RNA (EBER) 1 and EBER2. Despite their high abundance in the nucleus (about 10(6) copies), the molecular function of these noncoding RNAs has remained elusive. Here, we report that the insertion into EBER1 of an RNA aptamer that binds the bacteriophage MS2 coat protein allows the isolation of EBER1 and associated protein partners. By combining MS2-mediated selection with stable isotope labeling of amino acids in cell culture (SILAC) and analysis by mass spectrometry, we identified AUF1 (AU-rich element binding factor 1)/hnRNP D (heterogeneous nuclear ribonucleoprotein D) as an interacting protein of EBER1. AUF1 exists as four isoforms generated by alternative splicing and is best known for its role in destabilizing mRNAs upon binding to AU-rich elements (AREs) in their 3' untranslated region (UTR). Using UV crosslinking, we demonstrate that predominantly the p40 isoform of AUF1 interacts with EBER1 in vivo. Electrophoretic mobility shift assays show that EBER1 can compete for the binding of the AUF1 p40 isoform to ARE-containing RNA. Given the high abundance of EBER1 in EBV-positive cells, EBER1 may disturb the normal homeostasis between AUF1 and ARE-containing mRNAs or compete with other AUF1-interacting targets in cells latently infected by EBV.


Subject(s)
Herpesvirus 4, Human/genetics , Heterogeneous-Nuclear Ribonucleoprotein D/metabolism , RNA, Viral/metabolism , 3' Untranslated Regions , AU Rich Elements , Aptamers, Nucleotide/genetics , Binding, Competitive , Cell Line, Tumor , Herpesvirus 4, Human/physiology , Heterogeneous Nuclear Ribonucleoprotein D0 , Heterogeneous-Nuclear Ribonucleoprotein D/chemistry , Host-Pathogen Interactions , Humans , Immunoprecipitation , Mutagenesis, Insertional , Protein Binding , Protein Isoforms/metabolism , RNA Stability , RNA, Viral/chemistry , RNA, Viral/genetics
9.
J Biochem ; 151(4): 423-37, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22368252

ABSTRACT

The stability of RNAs bearing AU-rich elements in their 3'-UTRs, and thus the level of expression of their protein products, is regulated by interactions with cytoplasmic RNA-binding proteins. Binding by HuR generally leads to mRNA stabilization and increased protein production, whereas binding by AUF1 isoforms generally lead to rapid degradation of the mRNA and reduced protein production. The exact nature of the interplay between these and other RNA-binding proteins remains unclear, although recent studies have shown close interactions between them and even suggested competition between the two for binding to their cognate recognition sequences. Other recent reports have suggested that the sequences recognized by the two proteins are different. We therefore performed a detailed in vitro analysis of the binding site(s) for HuR and AUF1 present in androgen receptor mRNA to define their exact target sequences, and show that the same sequence is contacted by both proteins. Furthermore, we analysed a proposed HuR target within the 3'-UTR of MTA1 mRNA, and show that the contacted bases lie outside of the postulated motif and are a better match to a classical ARE than the postulated motif. The defining features of these HuR binding sites are their U-richness and single strandedness.


Subject(s)
ELAV Proteins/chemistry , Heterogeneous-Nuclear Ribonucleoprotein D/chemistry , RNA, Messenger/chemistry , 3' Untranslated Regions , Base Sequence , Binding Sites , Cell Line, Tumor , Electrophoretic Mobility Shift Assay , Heterogeneous Nuclear Ribonucleoprotein D0 , Humans , Inverted Repeat Sequences , Molecular Sequence Data , Nucleic Acid Conformation , Protein Binding , Proto-Oncogene Proteins c-fos/genetics , RNA Stability , Receptors, Androgen/genetics , Tumor Necrosis Factor-alpha/genetics
10.
PLoS One ; 6(7): e21111, 2011.
Article in English | MEDLINE | ID: mdl-21799732

ABSTRACT

BACKGROUND: The cyclin-D/CDK4,6/p16(INK4a)/pRB/E2F pathway, a key regulator of the critical G1 to S phase transition of the cell cycle, is universally disrupted in human cancer. However, the precise function of the different members of this pathway and their functional interplay are still not well defined. METHODOLOGY/PRINCIPAL FINDINGS: We have shown here that the tumor suppressor p16(INK4a) protein positively controls the expression of cyclin D1 and E2F1 in both human and mouse cells. p16(INK4a) stabilizes the mRNAs of the corresponding genes through negative regulation of the mRNA decay-promoting AUF1 protein. Immunoprecipitation of AUF1-associated RNAs followed by RT-PCR indicated that endogenous AUF1 binds to the cyclin D1 and E2F1 mRNAs. Furthermore, AUF1 down-regulation increased the expression levels of these genes, while concurrent silencing of AUF1 and p16(INK4a), using specific siRNAs, restored normal expression of both cyclinD1 and E2F1. Besides, we have shown the presence of functional AU-rich elements in the E2F1 3'UTR, which contributed to p16/AUF1-mediated regulation of E2F1 post-transcriptional events in vivo. Importantly, genome-wide gene expression microarray analysis revealed the presence of a large number of genes differentially expressed in a p16(INK4a) -dependent manner, and several of these genes are also members of the AUF1 and E2F1 regulons. We also present evidence that E2F1 mediates p16-dependent regulation of several pro- and anti-apoptotic proteins, and the consequent induction of spontaneous as well as doxorubicin-induced apoptosis. CONCLUSION/SIGNIFICANCE: These findings show that the cyclin-dependent kinase inhibitor p16( INK4a) is also a modulator of transcription and apoptosis through controlling the expression of two major transcription regulators, AUF1 and E2F1.


Subject(s)
Cyclin D1/metabolism , Cyclin-Dependent Kinase Inhibitor p16/metabolism , E2F1 Transcription Factor/metabolism , Heterogeneous-Nuclear Ribonucleoprotein D/metabolism , 3' Untranslated Regions/genetics , Animals , Apoptosis/drug effects , Base Sequence , Cell Line , Cyclin D1/genetics , Cyclin-Dependent Kinase Inhibitor p16/chemistry , Doxorubicin/pharmacology , E2F1 Transcription Factor/genetics , GC Rich Sequence/genetics , Gene Expression Regulation/drug effects , Heterogeneous Nuclear Ribonucleoprotein D0 , Heterogeneous-Nuclear Ribonucleoprotein D/chemistry , Humans , Mice , Models, Molecular , Molecular Sequence Data , Protein Conformation , RNA, Messenger/genetics , RNA, Messenger/metabolism
11.
Front Biosci (Landmark Ed) ; 16(6): 2307-25, 2011 06 01.
Article in English | MEDLINE | ID: mdl-21622178

ABSTRACT

The mRNA-binding protein AUF1 regulates the expression of many key players in cancer including proto-oncogenes, regulators of apoptosis and the cell cycle, and pro-inflammatory cytokines, principally by directing the decay kinetics of their encoded mRNAs. Most studies support an mRNA-destabilizing role for AUF1, although other findings suggest additional functions for this factor. In this review, we explore how changes in AUF1 isoform distribution, subcellular localization, and post-translational protein modifications can influence the metabolism of targeted mRNAs. However, several lines of evidence also support a role for AUF1 in the initiation and/or development of cancer. Many AUF1-targeted transcripts encode products that control pro- and anti-oncogenic processes. Also, overexpression of AUF1 enhances tumorigenesis in murine models, and AUF1 levels are enhanced in some tumors. Finally, signaling cascades that modulate AUF1 function are deregulated in some cancerous tissues. Together, these features suggest that AUF1 may play a prominent role in regulating the expression of many genes that can contribute to tumorigenic phenotypes, and that this post-transcriptional regulatory control point may be subverted by diverse mechanisms in neoplasia.


Subject(s)
Heterogeneous-Nuclear Ribonucleoprotein D/genetics , Heterogeneous-Nuclear Ribonucleoprotein D/metabolism , Neoplasms/genetics , Neoplasms/metabolism , Animals , Apoptosis/genetics , Apoptosis/physiology , Biological Transport, Active , Cell Cycle/genetics , Cell Cycle/physiology , Gene Expression Regulation, Neoplastic , Heterogeneous Nuclear Ribonucleoprotein D0 , Heterogeneous-Nuclear Ribonucleoprotein D/chemistry , Humans , Mice , Neoplasms/etiology , Neoplasms/pathology , Phosphorylation , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA Processing, Post-Transcriptional , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism
12.
Cell Mol Biol Lett ; 16(1): 55-68, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21161418

ABSTRACT

Thrombospondin-1 (TSP-1) is a matricellular protein that participates in numerous normal and pathological tissue processes and is rapidly modulated by different stimuli. The presence of 8 highly-conserved AU rich elements (AREs) within the 3'-untranslated region (3'UTR) of the TSP-1 mRNA suggests that post-transcriptional regulation is likely to represent one mechanism by which TSP-1 gene expression is regulated. We investigated the roles of these AREs, and proteins which bind to them, in the control of TSP-1 mRNA stability. The endogenous TSP-1 mRNA half-life is approximately 2.0 hours in HEK293 cells. Luciferase reporter mRNAs containing the TSP-1 3'UTR show a similar rate of decay, suggesting that the 3'UTR influences the decay rate. Site-directed mutagenesis of individual and adjacent AREs prolonged reporter mRNA halflife to between 2.2 and 4.4 hours. Mutation of all AREs increased mRNA half life to 8.8 hours, suggesting that all AREs have some effect, but that specific AREs may have key roles in stability regulation. A labeled RNA oligonucleotide derived from the most influential ARE was utilized to purify TSP-1 ARE-binding proteins. The AU-binding protein AUF1 was shown to associate with this motif. These studies reveal that AREs in the 3'UTR control TSP-1 mRNA stability and that the RNA binding protein AUF1 participates in this control. These studies suggest that ARE-dependent control of TSP-1 mRNA stability may represent an important component in the control of TSP-1 gene expression.


Subject(s)
3' Untranslated Regions , Gene Expression Regulation , Thrombospondin 1/genetics , Base Sequence , HEK293 Cells , Half-Life , Heterogeneous Nuclear Ribonucleoprotein D0 , Heterogeneous-Nuclear Ribonucleoprotein D/chemistry , Heterogeneous-Nuclear Ribonucleoprotein D/metabolism , Humans , Mutagenesis, Site-Directed , RNA Stability , RNA, Messenger/metabolism , Thrombospondin 1/metabolism
13.
Wiley Interdiscip Rev RNA ; 1(3): 457-73, 2010.
Article in English | MEDLINE | ID: mdl-21956942

ABSTRACT

Messenger ribonucleic acid (mRNA) turnover is a major control point in gene expression. In mammals, many mRNAs encoding inflammatory cytokines, oncoproteins, and G-protein-coupled receptors are destabilized by the presence of AU-rich elements (AREs) in their 3'-untranslated regions. Association of ARE-binding proteins (AUBPs) with these mRNAs promotes rapid mRNA degradation. ARE/poly(U)-binding/degradation factor 1 (AUF1), one of the best-characterized AUBPs, binds to many ARE-mRNAs and assembles other factors necessary to recruit the mRNA degradation machinery. These factors include translation initiation factor eIF4G, chaperones hsp27 and hsp70, heat-shock cognate protein hsc70, lactate dehydrogenase, poly(A)-binding protein, and other unidentified proteins. Numerous signaling pathways alter the composition of this AUF1 complex of proteins to effect changes in ARE-mRNA degradation rates. This review briefly describes the roles of mRNA decay in gene expression in general and ARE-mediated decay (AMD) in particular, with a focus on AUF1 and the different modes of regulation that govern AUF1 involvement in AMD.


Subject(s)
Heterogeneous-Nuclear Ribonucleoprotein D/physiology , RNA Stability/genetics , Amino Acid Sequence , Animals , Gene Expression Regulation/genetics , Heterogeneous Nuclear Ribonucleoprotein D0 , Heterogeneous-Nuclear Ribonucleoprotein D/chemistry , Heterogeneous-Nuclear Ribonucleoprotein D/genetics , Heterogeneous-Nuclear Ribonucleoprotein D/metabolism , Humans , Models, Biological , Protein Binding/genetics , Protein Processing, Post-Translational , RNA Processing, Post-Transcriptional/genetics , RNA Processing, Post-Transcriptional/physiology , RNA Stability/physiology , Regulatory Sequences, Ribonucleic Acid/genetics , Regulatory Sequences, Ribonucleic Acid/physiology
14.
Arthritis Rheum ; 58(2): 511-20, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18240226

ABSTRACT

OBJECTIVE: To investigate which members of the heterogeneous nuclear RNP (hnRNP) family are targeted by autoantibodies from patients with systemic rheumatic diseases. METHODS: Using a semipurified preparation of natural hnRNP proteins, 365 sera from patients with rheumatic diseases and control subjects were screened by immunoblotting for the presence of autoantibodies. Bacterially expressed recombinant hnRNP D (AUF1) proteins were used for confirming the data obtained. Binding of RNA and autoantibody to AUF1 was investigated by gel retardation assays. Expression of AUF1 in cultivated cells and synovial tissue was analyzed by indirect immunofluorescence and immunohistochemistry. RESULTS: Autoantibodies to AUF1 proteins were detected in 33% of patients with systemic lupus erythematosus, 20% of patients with rheumatoid arthritis, 17% of patients with mixed connective tissue disease, and <10% of patients with other rheumatic disorders. Epitope mapping studies showed the autoantibodies to be directed to conformational epitopes in the N-terminal RNA-binding part of AUF1. However, autoantibody binding did not interfere with RNA binding as assessed by gel-shift assays. Immunohistochemical studies revealed AUF1 to be expressed in the cytoplasm of RA synovial tissue as compared with nuclear staining in osteoarthritis and normal synovium, particularly in macrophages of the lining layer and in fibroblasts of the sublining areas. CONCLUSION: These data identify AUF1 proteins as novel autoantigens in SLE and related autoimmune disorders. Because AUF1 proteins are major components of messenger RNA stability complexes, our findings suggest that these complexes form a novel macromolecular target structure for autoantibodies in rheumatic autoimmune diseases.


Subject(s)
Autoantibodies/immunology , Heterogeneous-Nuclear Ribonucleoprotein D/immunology , RNA Stability/immunology , Rheumatic Diseases/immunology , Tumor Necrosis Factor-alpha/genetics , Arthritis, Rheumatoid/immunology , Autoantibodies/pharmacology , Autoantigens/chemistry , Autoantigens/immunology , Binding Sites/immunology , Epitope Mapping , HeLa Cells , Heterogeneous Nuclear Ribonucleoprotein D0 , Heterogeneous-Nuclear Ribonucleoprotein D/chemistry , Humans , Lupus Erythematosus, Systemic/immunology , Mixed Connective Tissue Disease/immunology , Protein Structure, Tertiary , Synovial Membrane/immunology
15.
Mol Cell ; 28(1): 57-67, 2007 Oct 12.
Article in English | MEDLINE | ID: mdl-17936704

ABSTRACT

Transportin 1 (Trn1) is a transport receptor that transports substrates from the cytoplasm to the nucleus through nuclear pore complexes by recognizing nuclear localization signals (NLSs). Here we describe four crystal structures of human Trn1 in a substrate-free form as well as in the complex with three NLSs (hnRNP D, JKTBP, and TAP, respectively). Our data have revealed that (1) Trn1 has two sites for binding NLSs, one with high affinity (site A) and one with low affinity (site B), and NLS interaction at site B controls overall binding affinity for Trn1; (2) Trn1 recognizes the NLSs at site A followed by conformational change at site B to interact with the NLSs; and (3) a long flexible loop, characteristic of Trn1, interacts with site B, thereby displacing transport substrate in the nucleus. These studies provide deep understanding of substrate recognition and dissociation by Trn1 in import pathways.


Subject(s)
Protein Structure, Tertiary , beta Karyopherins/chemistry , beta Karyopherins/metabolism , ATP Binding Cassette Transporter, Subfamily B, Member 2 , ATP-Binding Cassette Transporters/chemistry , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Active Transport, Cell Nucleus/physiology , Amino Acid Sequence , Binding Sites , Crystallography, X-Ray , Heterogeneous-Nuclear Ribonucleoprotein D/chemistry , Heterogeneous-Nuclear Ribonucleoprotein D/genetics , Heterogeneous-Nuclear Ribonucleoprotein D/metabolism , Humans , Models, Molecular , Molecular Sequence Data , Mutation , Nuclear Localization Signals/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Ribonucleoproteins/chemistry , Ribonucleoproteins/genetics , Ribonucleoproteins/metabolism , Sequence Alignment , Substrate Specificity , beta Karyopherins/genetics
16.
RNA ; 13(9): 1453-68, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17626845

ABSTRACT

A number of highly regulated gene classes are regulated post-transcriptionally at the level of mRNA stability. A central feature in these mRNAs is the presence of A+U-rich elements (ARE) within their 3' UTRs. Two ARE binding proteins, HuR and AUF1, are associated with mRNA stabilization and destabilization, respectively. Previous studies have demonstrated homomultimerization of each protein and the capacity to bind simultaneous or competitively to a single ARE. To investigate this possibility further, cell biological and biophysical approaches were undertaken. Protein-protein interaction was monitored by fluorescence resonance energy transfer (FRET) and by immunocytochemistry in live and fixed cells using fluorescently labeled CFP/YFP fusion proteins of HuR and p37AUF1. Strong nuclear FRET between HuR/HuR and AUF1/AUF1 homodimers as well as HuR/AUF1 heterodimers was observed. Treatment with the MAP kinase activator, anisomycin, which commonly stabilizes ARE-containing mRNAs, caused rapid nuclear to cytoplasmic shuttling of HuR. AUF1 also underwent shuttling, but on a longer time scale. After shuttling, HuR/HuR, AUF1/AUF1, and HuR/AUF1, FRET was also observed in the cytoplasm. In further studies, arsenite rapidly induced the formation of stress granules containing HuR and TIA-1 but not AUF1. The current studies demonstrate that two mRNA binding proteins, HuR and AUF1, are colocalized and are capable of functional interaction in both the nucleus and cytoplasm. FRET-based detection of AUF1/HuR interaction may serve as a basis of opening up new dimensions in delineating the functional interaction of mRNA binding proteins with RNA turnover.


Subject(s)
Antigens, Surface/metabolism , Fluorescence Resonance Energy Transfer , Heterogeneous-Nuclear Ribonucleoprotein D/metabolism , Protein Interaction Mapping , RNA-Binding Proteins/metabolism , Animals , Antigens, Surface/biosynthesis , Antigens, Surface/chemistry , Cell Line , Cell Nucleus/chemistry , Cell Nucleus/metabolism , Cricetinae , Cytoplasm/chemistry , Cytoplasm/metabolism , ELAV Proteins , ELAV-Like Protein 1 , Heterogeneous Nuclear Ribonucleoprotein D0 , Heterogeneous-Nuclear Ribonucleoprotein D/biosynthesis , Heterogeneous-Nuclear Ribonucleoprotein D/chemistry , Humans , Protein Binding/physiology , RNA, Messenger/chemistry , RNA, Messenger/metabolism , RNA-Binding Proteins/biosynthesis , RNA-Binding Proteins/chemistry
17.
Arch Biochem Biophys ; 465(1): 274-81, 2007 Sep 01.
Article in English | MEDLINE | ID: mdl-17603996

ABSTRACT

In vertebrates, mRNAs containing a 5'-terminal oligopyrimidine (TOP) motif are coordinately post-transcriptionally regulated. Binding of specific proteins to this element has been proposed to downregulate expression of TOP mRNAs at the level of translational initiation. We previously reported that rapamycin induces binding activity to the TOP element of ribosomal protein (r-protein) L32 mRNA. In this study, we adapt DEAE-cellulose/oligo dT-cellulose tandem column chromatography to purify TOP element-binding proteins from bovine submaxillary lymph nodes (SLN). We also show by northwestern blot analysis that two proteins of molecular weight 47kDa (47BP) and 43kDa (43BP) specifically bind to a (32)P-labeled riboprobe containing TOP regulatory element of the r-protein L32. Microsequencing of the purified 47BP revealed an internal sequence of 15 amino acids identical to the consensus sequence of the 2x RBD-Gly family. Western blot analysis of the cytoplasm fractions using an AUF1 antibody revealed that these two proteins are p45 AUF1 and p42 AUF1. Increases of the four isoforms of AUF1 protein were observed in 100,000g supernatant fractions of rapamycin-administered rat SLN. Furthermore, decreases of p45 AUF1 and p42 and/or p40 AUF1 were observed in the polysomal fractions of BJAB cells in which translation of TOP mRNAs was selectively suppressed by rapamycin treatment. Taken together, these results suggest that AUF1 is a TOP mRNA-binding protein that may participate in the translational suppression of TOP mRNAs resulting from rapamycin treatment.


Subject(s)
Heterogeneous-Nuclear Ribonucleoprotein D/chemistry , Pyrimidines/chemistry , RNA, Messenger/chemistry , Tacrolimus Binding Proteins/chemistry , Amino Acid Sequence , Binding Sites , Heterogeneous Nuclear Ribonucleoprotein D0 , Molecular Sequence Data , Protein Binding
18.
Cell Calcium ; 40(4): 329-46, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16765440

ABSTRACT

Specific sequences (cis-acting elements) in the 3'-untranslated region (UTR) of RNA, together with stabilizing and destabilizing proteins (trans-acting factors), determine the mRNA stability, and consequently, the level of expression of several proteins. Such interactions were discovered initially for short-lived mRNAs encoding cytokines and early genes like c-jun and c-myc. However, they may also determine the fate of more stable mRNAs in a tissue and disease-dependent manner. The interactions between the cis-acting elements and the trans-acting factors may also be modulated by Ca(2+) either directly or via a control of the phosphorylation status of the trans-acting factors. We focus initially on the basic concepts in mRNA stability with the trans-acting factors AUF1 (destabilizing) and HuR (stabilizing). Sarco/endoplasmic reticulum Ca(2+) pumps, SERCA2a (cardiac and slow twitch muscles) and SERCA2b (most cells including smooth muscle cells), are pivotal in Ca(2+) mobilization during signal transduction. SERCA2a and SERCA2b proteins are encoded by relatively stable mRNAs that contain cis-acting stability determinants in their 3'-regions. We present several pathways where 3'-UTR mediated mRNA decay is key to Ca(2+) signalling: SERCA2a and beta-adrenergic receptors in heart failure, renin-angiotensin system, and parathyroid hormones. Other examples discussed include cytokines vascular endothelial growth factor, endothelin and endothelial nitric oxide synthase. Roles of Ca(2+) and Ca(2+)-binding proteins in mRNA stability are also discussed. We anticipate that these novel modes of control of protein expression will form an emerging area of research that may explore the central role of Ca(2+) in cell function during development and in disease.


Subject(s)
Calcium Signaling/physiology , Gene Expression Regulation , Proteins , RNA Stability , 3' Untranslated Regions , Animals , Calcium/metabolism , Cardiac Output, Low , ELAV Proteins/chemistry , ELAV Proteins/genetics , ELAV Proteins/metabolism , Heterogeneous Nuclear Ribonucleoprotein D0 , Heterogeneous-Nuclear Ribonucleoprotein D/chemistry , Heterogeneous-Nuclear Ribonucleoprotein D/genetics , Heterogeneous-Nuclear Ribonucleoprotein D/metabolism , Nucleic Acid Conformation , Protein Modification, Translational , Proteins/genetics , Proteins/metabolism , RNA, Messenger/chemistry , RNA, Messenger/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases/genetics , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism
19.
J Cell Biochem ; 98(5): 1257-70, 2006 Aug 01.
Article in English | MEDLINE | ID: mdl-16514630

ABSTRACT

Alpha-fetoprotein (AFP) is one of the major serum proteins in the early life of mammals. We have previously identified a novel cis-acting element designated as DAS at the 5'-flanking region of the AFP gene and demonstrated that the DAS sequence can be specifically recognized by nuclear protein DAP-II in AFP-producing hepatoma cells and retinoic acid (RA)-induced AFP-producing F9 cells. In this study, we used DNA affinity chromatography to purify the DAP-II proteins from the nuclear extracts (NE) of RA-treated F9 cells. The purified DAP-II complex mainly contained five proteins, with molecular weights of 45, 42, 32, 30, and 20 kDa, respectively. The identification of these proteins was determined by MALDI-TOF mass spectrometric analysis and a database search. These proteins were found to belong to the AUF1 RNA-binding protein family. Protein (30 kDa), one of five proteins in an isolated DAP-II complex, was matched with amino acid sequence highly similar to muAUF1-3. The expression of this protein is inducible by RA, and the pattern of the protein expression is the same as DAP-II proteins in F9 cells after treatment with RA during differentiation. Our results suggest that the 30-kDa protein is a novel isoform of AUF1 family and is the main component of the DAP-II complex that binds to the DAS sequence.


Subject(s)
Gene Expression Regulation/genetics , Heterogeneous-Nuclear Ribonucleoprotein D/metabolism , Response Elements/genetics , alpha-Fetoproteins/genetics , Amino Acid Sequence , Animals , Base Sequence , Cell Differentiation/drug effects , Cell Line , Chromatography, Affinity , Conserved Sequence , DNA/genetics , DNA/metabolism , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/isolation & purification , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/metabolism , Electrophoresis, Gel, Two-Dimensional , Heterogeneous Nuclear Ribonucleoprotein D0 , Heterogeneous-Nuclear Ribonucleoprotein D/chemistry , Heterogeneous-Nuclear Ribonucleoprotein D/classification , Humans , Mice , Molecular Sequence Data , Protein Binding , Sequence Alignment , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Tretinoin/pharmacology , alpha-Fetoproteins/metabolism
20.
RNA ; 12(5): 883-93, 2006 May.
Article in English | MEDLINE | ID: mdl-16556936

ABSTRACT

An AU-rich element (ARE) located in the 3'-untranslated region of many short-lived mRNAs functions as an instability determinant for these transcripts. AUF1/hnRNP D, an ARE-binding protein family consisting of four isoforms, promotes rapid decay of ARE-mRNAs. The mechanism by which AUF1 promotes rapid decay of ARE-mRNA is unclear. AUF1 has been shown to form an RNase-resistant complex in cells with the cap-initiation complex and heat shock proteins Hsp70 and Hsc70, as well as other unidentified factors. To understand the function of the AUF1 complex, we have biochemically investigated the association of AUF1 with the components of the translation initiation complex. We used purified recombinant proteins and a synthetic ARE RNA oligonucleotide to determine the hierarchy of protein interactions in vitro and the effect of AUF1 binding to the ARE on the formation of protein complexes. We demonstrate that all four AUF1 protein isoforms bind directly and strongly to initiation factor eIF4G at a C-terminal site regardless of AUF1 interaction with the ARE. AUF1 is shown to directly interact with poly(A) binding protein (PABP), both independently of eIF4G and in a complex with eIF4G. AUF1-PABP interaction is opposed by AUF1 binding to the ARE or Hsp70 heat shock protein. In vivo, AUF1 interaction with PABP does not alter PABP stability. Based on these and other data, we propose a model for the molecular interactions of AUF1 that involves translation-dependent displacement of AUF1-PABP complexes from ARE-mRNAs with possible unmasking of the poly(A) tail.


Subject(s)
Eukaryotic Initiation Factor-4G/metabolism , Heterogeneous-Nuclear Ribonucleoprotein D/metabolism , Poly(A)-Binding Proteins/metabolism , Protein Biosynthesis , RNA, Messenger/metabolism , 3' Untranslated Regions , Animals , Binding Sites , CHO Cells , Cricetinae , Glutathione Transferase/metabolism , HSP70 Heat-Shock Proteins/metabolism , Heterogeneous Nuclear Ribonucleoprotein D0 , Heterogeneous-Nuclear Ribonucleoprotein D/chemistry , Heterogeneous-Nuclear Ribonucleoprotein D/genetics , Histidine/chemistry , In Vitro Techniques , Protein Binding , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Structure, Tertiary , Protein Synthesis Inhibitors/pharmacology , RNA, Messenger/chemistry , Recombinant Fusion Proteins/isolation & purification , Recombinant Fusion Proteins/metabolism , Sequence Deletion
SELECTION OF CITATIONS
SEARCH DETAIL
...