Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Proc Natl Acad Sci U S A ; 116(23): 11285-11290, 2019 06 04.
Article in English | MEDLINE | ID: mdl-31113881

ABSTRACT

AUF1 promotes rapid decay of mRNAs containing 3' untranslated region (3'UTR) AU-rich elements (AREs). AUF1 depletion in mice accelerates muscle loss and causes limb girdle muscular dystrophy. Here, we demonstrate that the selective, targeted degradation by AUF1 of key muscle stem cell fate-determining checkpoint mRNAs regulates each stage of muscle development and regeneration by reprogramming each myogenic stage. Skeletal muscle stem (satellite) cell explants show that Auf1 transcription is activated with satellite cell activation by stem cell regulatory factor CTCF. AUF1 then targets checkpoint ARE-mRNAs for degradation, progressively reprogramming the transcriptome through each stage of myogenesis. Transition steps in myogenesis, from stem cell proliferation to differentiation to muscle fiber development, are each controlled by fate-determining checkpoint mRNAs, which, surprisingly, were found to be controlled in their expression by AUF1-targeted mRNA decay. Checkpoint mRNAs targeted by AUF1 include Twist1, decay of which promotes myoblast development; CyclinD1, decay of which blocks myoblast proliferation and initiates differentiation; and RGS5, decay of which activates Sonic Hedgehog (SHH) pathway-mediated differentiation of mature myotubes. AUF1 therefore orchestrates muscle stem cell proliferation, self-renewal, myoblast differentiation, and ultimately formation of muscle fibers through targeted, staged mRNA decay.


Subject(s)
Heterogeneous Nuclear Ribonucleoprotein D0/metabolism , Muscle Development/physiology , RNA, Messenger/metabolism , Regeneration/physiology , 3' Untranslated Regions/physiology , AU Rich Elements/physiology , Animals , Cell Differentiation/physiology , Cell Line , Cell Proliferation/physiology , Female , Hedgehog Proteins/metabolism , Male , Mice , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/metabolism , Myoblasts/metabolism , RGS Proteins/metabolism , RNA Stability/physiology , Stem Cells/metabolism
2.
J Biol Chem ; 294(19): 7558-7565, 2019 05 10.
Article in English | MEDLINE | ID: mdl-30962286

ABSTRACT

Replication-dependent histone (RDH) mRNAs have a nonpolyadenylated 3'-UTR that ends in a highly conserved stem-loop structure. Nonetheless, a subset of RDH mRNAs has a poly(A) tail under physiological conditions. The biological meaning of poly(A)-containing (+) RDH mRNAs and details of their biosynthesis remain elusive. Here, using HeLa cells and Western blotting, qRT-PCR, and biotinylated RNA pulldown assays, we show that poly(A)+ RDH mRNAs are post-transcriptionally regulated via adenylate- and uridylate-rich element-mediated mRNA decay (AMD). We observed that the rapid degradation of poly(A)+ RDH mRNA is driven by butyrate response factor 1 (BRF1; also known as ZFP36 ring finger protein-like 1) under normal conditions. Conversely, cellular stresses such as UV C irradiation promoted BRF1 degradation, increased the association of Hu antigen R (HuR; also known as ELAV-like RNA-binding protein 1) with the 3'-UTR of poly(A)+ RDH mRNAs, and eventually stabilized the poly(A)+ RDH mRNAs. Collectively, our results provide evidence that AMD surveils poly(A)+ RDH mRNAs via BRF1-mediated degradation under physiological conditions.


Subject(s)
AU Rich Elements/physiology , Histones/biosynthesis , RNA Stability/physiology , RNA, Messenger/metabolism , TATA-Binding Protein Associated Factors/metabolism , HeLa Cells , Histones/genetics , Humans , RNA, Messenger/genetics
3.
Cell Rep ; 20(5): 1187-1200, 2017 08 01.
Article in English | MEDLINE | ID: mdl-28768202

ABSTRACT

Pat1 RNA-binding proteins, enriched in processing bodies (P bodies), are key players in cytoplasmic 5' to 3' mRNA decay, activating decapping of mRNA in complex with the Lsm1-7 heptamer. Using co-immunoprecipitation and immunofluorescence approaches coupled with RNAi, we provide evidence for a nuclear complex of Pat1b with the Lsm2-8 heptamer, which binds to the spliceosomal U6 small nuclear RNA (snRNA). Furthermore, we establish the set of interactions connecting Pat1b/Lsm2-8/U6 snRNA/SART3 and additional U4/U6.U5 tri-small nuclear ribonucleoprotein particle (tri-snRNP) components in Cajal bodies, the site of snRNP biogenesis. RNA sequencing following Pat1b depletion revealed the preferential upregulation of mRNAs normally found in P bodies and enriched in 3' UTR AU-rich elements. Changes in >180 alternative splicing events were also observed, characterized by skipping of regulated exons with weak donor sites. Our data demonstrate the dual role of a decapping enhancer in pre-mRNA processing as well as in mRNA decay via distinct nuclear and cytoplasmic Lsm complexes.


Subject(s)
DNA-Binding Proteins/metabolism , Multiprotein Complexes/metabolism , N-Terminal Acetyltransferase C/metabolism , Proto-Oncogene Proteins/metabolism , RNA Precursors/metabolism , RNA Processing, Post-Transcriptional/physiology , RNA-Binding Proteins/metabolism , Ribonucleoprotein, U4-U6 Small Nuclear/metabolism , Ribonucleoproteins, Small Nuclear/metabolism , AU Rich Elements/physiology , DNA-Binding Proteins/genetics , HEK293 Cells , Humans , Multiprotein Complexes/genetics , N-Terminal Acetyltransferase C/genetics , Proto-Oncogene Proteins/genetics , RNA Precursors/genetics , RNA-Binding Proteins/genetics , Ribonucleoprotein, U4-U6 Small Nuclear/genetics , Ribonucleoproteins, Small Nuclear/genetics
4.
Inflammation ; 40(2): 645-656, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28124257

ABSTRACT

We have shown previously that bacterial lipopolysaccharide (LPS)-mediated suppression of phospholipase-Cß-2 (PLCß-2) expression is involved in M1 (inflammatory) to M2-like (wound healing) phenotypic switching of macrophages triggered by adenosine. This suppression is mediated post-transcriptionally by destabilization of PLCß-2 mRNA (messenger ribonucleic acid). To investigate the mechanism of this LPS-mediated destabilization, we examined the roles of RNA-binding agents including microRNAs and RNA-binding proteins that are involved in regulating stability of mRNAs encoding growth factors, inflammatory mediators, and proto-oncogenes. Adenylate and uridylate (AU)-rich elements (AREs) in 3'UTRs are specific recognition sites for RNA-binding proteins including tristetraprolin (TTP), HuR, and AUF1 and for microRNAs that are involved in regulating mRNA stability. In this study, we investigated the role of TTP and AREs in regulating PLCß-2 mRNA stability. The 3'UTR of the PLCß-2 gene was inserted into the pLightswitch luciferase reporter plasmid and transfected into RAW264.7 cells. LPS suppressed luciferase expression from this reporter. Luciferase expression from mutant 3'UTR constructs lacking AREs was similarly downregulated, suggesting that these regions are not required for LPS-mediated suppression of PLCß-2. TTP was rapidly upregulated in both primary murine macrophages and RAW264.7 cells in response to LPS. Suppression of PLCß-2 by LPS was examined using macrophages from mice lacking TTP (TTP-/-). LPS suppressed PLCß-2 expression to the same extent in wild type (WT) and TTP-/- macrophages. Also, the rate of decay of PLCß-2 mRNA in LPS-treated macrophages following transcriptional blockade was similar in WT and TTP-/- macrophages, clearly indicating that TTP is not involved in LPS-mediated destabilization of PLCß-2 mRNA in macrophages.


Subject(s)
AU Rich Elements/physiology , Macrophages/metabolism , Phospholipase C beta/genetics , RNA Stability/drug effects , Tristetraprolin/physiology , 3' Untranslated Regions/genetics , Animals , Cells, Cultured , Diabetes Mellitus, Experimental , Lipopolysaccharides/pharmacology , Mice , RAW 264.7 Cells , RNA-Binding Proteins
5.
RNA Biol ; 11(2): 124-33, 2014.
Article in English | MEDLINE | ID: mdl-24525793

ABSTRACT

The mRNAs of most inflammatory mediators are short-lived due to AU-rich elements (AREs) in their 3'-untranslated regions. AREs ensure a low basal level of expression during homeostasis and a transient nature of expression during the inflammatory response. Here, we report that the mRNA of the pro-inflammatory chemokine IL-8, which contains an archetypal ARE, is unexpectedly constitutively abundant and highly stable in primary human monocytes and macrophages. Using the pre-monocyte-like THP-1 cell line that can differentiate into macrophage-like cells, we show that a low level of unstable IL-8 mRNA in undifferentiated cells (half-life<30 min) becomes constitutively elevated and the mRNA is dramatically stabilized in differentiated THP-1 cells with a half-life of more than 15 h similar to primary monocytes and macrophages. In contrast, the level and stability of TNF-α mRNA also containing an ARE is only slightly affected by differentiation; it remains low and unstable in primary macrophages and differentiated THP-1 cells with an estimated half-life of less than 20 min. This differentiation-dependent stabilization of IL-8 mRNA is p38 MAPK-independent and is probably coupled with reduced protein translation. Reporter assays in THP-1 cells suggest that the ARE alone is not sufficient for the constitutive stabilization in macrophage-like cells and imply an effect of the natural biogenesis of the transcript on the stabilization of the mature form. We present a novel, cell type-dependent sustained stabilization of an ARE-containing mRNA with similarities to situations found in disease.


Subject(s)
Interleukin-8/genetics , Interleukin-8/metabolism , Macrophages/metabolism , Monocytes/metabolism , RNA, Messenger/metabolism , Tumor Necrosis Factor-alpha/metabolism , AU Rich Elements/physiology , Cell Differentiation , Cell Line, Tumor , Dactinomycin/pharmacology , HeLa Cells , Humans , MAP Kinase Signaling System/drug effects , Nucleic Acid Synthesis Inhibitors/pharmacology , Phorbol Esters/pharmacology , RNA Stability , Tumor Necrosis Factor-alpha/genetics
6.
Cell Mol Life Sci ; 70(12): 2031-44, 2013 Jun.
Article in English | MEDLINE | ID: mdl-22968342

ABSTRACT

Members of the tristetraprolin (TTP/TIS11) family are important RNA-binding proteins initially characterized as mediators of mRNA degradation. They act via their interaction with AU-rich elements present in the 3'UTR of regulated transcripts. However, it is progressively appearing that the different steps of mRNA processing and fate including transcription, splicing, polyadenylation, translation, and degradation are coordinately regulated by multifunctional integrator proteins that possess a larger panel of functions than originally anticipated. Tristetraprolin and related proteins are very good examples of such integrators. This review gathers the present knowledge on the functions of this family of RNA-binding proteins, including their role in AU-rich element-mediated mRNA decay and focuses on recent advances that support the concept of their broader involvement in distinct steps of mRNA biogenesis and degradation.


Subject(s)
Gene Expression Regulation/physiology , Multigene Family/genetics , Protein Biosynthesis/physiology , RNA Stability/physiology , RNA, Messenger/biosynthesis , RNA-Binding Proteins/metabolism , Signal Transduction/physiology , Tristetraprolin/metabolism , 3' Untranslated Regions/genetics , 3' Untranslated Regions/physiology , AU Rich Elements/genetics , AU Rich Elements/physiology , Amino Acid Sequence , Animals , Gene Components , Gene Expression Regulation/genetics , Mice , Models, Biological , Molecular Sequence Data , Phenotype , Protein Biosynthesis/genetics , RNA, Messenger/metabolism , RNA-Binding Proteins/genetics , Signal Transduction/genetics , Tristetraprolin/genetics
7.
Atherosclerosis ; 226(1): 95-101, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23116706

ABSTRACT

OBJECTIVE: 5-Aminoimidazole-4-carboxamide ribonucleoside (AICAR) has pleiotropic and beneficial effects on metabolic disorders. However, the effects of AICAR on low density lipoprotein (LDL) metabolism are poorly understood. METHODS AND RESULTS: AICAR induces increased LDLR mRNA levels and increased LDLR protein production in hepatocytes. The AICAR-dependent LDLR mRNA increase was partially mediated by mRNA stabilization in an extracellular signal-regulated kinase1/2 (ERK1/2)-dependent manner, but not by the AMP-activated protein kinase (AMPK) activation. Reporter assays using a variety of constructs harboring the 3'-untranslated region (UTR) of human LDLR mRNA revealed that the most upstream AU-rich element (ARE) was critical for these AICAR effects. Using UV cross-linking assays, we found increased binding of three cytoplasmic proteins to this ARE region in response to AICAR and identified a 35-kDa protein as Human antigen R (HuR). Blocking ERK signaling pathway activation resulted in attenuated HuR binding. Silencing HuR expression by RNA interference hindered AICAR-induced LDLR mRNA stability, whereas its overexpression stabilized this mRNA. CONCLUSIONS: AICAR-dependent LDLR mRNA stabilization is mediated, at least in part, by HuR binding to the ARE1 region. Given that AICAR enhanced LDL uptake in hepatocytes, our findings warrant further studies using animal models to develop a novel LDL-cholesterol lowering agent as a possible strategy to treat atherosclerosis-related cardiovascular diseases.


Subject(s)
AU Rich Elements/physiology , Aminoimidazole Carboxamide/analogs & derivatives , ELAV Proteins/metabolism , Hepatocytes , MAP Kinase Signaling System/physiology , RNA, Messenger/physiology , Receptors, LDL/genetics , Ribonucleosides/physiology , Cells, Cultured , Humans , Protein Binding
8.
J Biol Chem ; 287(42): 35527-35538, 2012 Oct 12.
Article in English | MEDLINE | ID: mdl-22932903

ABSTRACT

The destabilization of AU-rich element (ARE)-containing mRNAs mediated by proteins of the TIS11 family is conserved among eukaryotes including Drosophila. Previous studies have demonstrated that Tristetraprolin, a human protein of the TIS11 family, induces the degradation of ARE-containing mRNAs through a large variety of mechanisms including deadenylation, decapping, and P-body targeting. We have previously shown that the degradation of the mRNA encoding the antimicrobial peptide Cecropin A1 (CecA1) is controlled by the TIS11 protein (dTIS11) in Drosophila cells. In this study, we used CecA1 mRNA as a model to investigate the molecular mechanism of dTIS11-mediated mRNA decay. We observed that during the biphasic deadenylation and decay process of this mRNA, dTIS11 enhances deadenylation performed by the CCR4-CAF-NOT complex while the mRNA is still associated with ribosomes. Sequencing of mRNA degradation intermediates revealed that the complete deadenylation of the mRNA triggers its decapping and decay in both the 5'-3' and the 3'-5' directions. Contrary to the observations made for its mammalian homologs, overexpression of dTIS11 does not promote the localization of ARE-containing mRNAs in P-bodies but rather decreases the accumulation of CecA1 mRNA in these structures by enhancing the degradation process. Therefore, our results suggest that proteins of the TIS11 family may have acquired additional functions in the course of evolution from invertebrates to mammals.


Subject(s)
AU Rich Elements/physiology , Drosophila Proteins/metabolism , RNA Stability/physiology , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism , Ribonucleases/metabolism , Tristetraprolin/metabolism , Animals , Antimicrobial Cationic Peptides/biosynthesis , Antimicrobial Cationic Peptides/genetics , Cell Line , Drosophila Proteins/genetics , Drosophila melanogaster , Evolution, Molecular , Humans , RNA, Messenger/genetics , RNA-Binding Proteins/genetics , Ribonucleases/genetics , Tristetraprolin/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...