Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Sci Rep ; 14(1): 913, 2024 01 09.
Article in English | MEDLINE | ID: mdl-38195703

ABSTRACT

Glucocorticoids (GC) like dexamethasone (Dex) are potent anti-inflammatory agents with diverse cellular functions including the potentiation of the activity of AU-rich elements (AREs). AREs are cis-acting instability sequence elements located in the 3'UTRs of many inflammatory mediator mRNAs. Here, available RNA-seq data were used to investigate the effect of GCs on the ARE-mRNA-transcriptome. At a global scale, ARE-mRNAs had a tendency to be downregulated after GC-treatment of the A549 lung cancer cell-line, but with notable cases of upregulation. mRNA stability experiments indicated that not only the downregulated, but also the upregulated ARE-mRNAs are destabilized by Dex-treatment. Several of the most upregulated ARE-mRNAs code for anti-inflammatory mediators including the established GC targets DUSP1 and ZFP36; both code for proteins that target ARE-containing mRNAs for destruction. GCs are widely used in the treatment of COVID-19 patients; we show that ARE-mRNAs are more likely to regulate in opposite directions between Dex-treatment and SARS-CoV-2 infections compared to non-ARE mRNAs. The effect of GC treatment on ARE-mRNA abundance was also investigated in blood monocytes of COVID-19 patients. The results were heterogeneous; however, in agreement with in vitro observations, ZFP36 and DUSP1 were often amongst the most differentially expressed mRNAs. The results of this study propose a universal destabilization of ARE-mRNAs by GCs, but a diverse overall outcome in vitro likely due to induced transcription or due to the heterogeneity of COVID-19 patient's responses in vivo.


Subject(s)
COVID-19 , Glucocorticoids , Humans , Glucocorticoids/pharmacology , Glucocorticoids/therapeutic use , 3' Untranslated Regions , A549 Cells , COVID-19/genetics , RNA, Messenger/genetics
2.
Hum Genomics ; 16(1): 59, 2022 11 15.
Article in English | MEDLINE | ID: mdl-36380320

ABSTRACT

BACKGROUND: AU-rich elements (AREs) are located in the 3'UTRs of 22% of human mRNAs, including most transiently expressed inflammatory mediators. By default, AREs mark mRNAs for decay and translational inhibition, but this activity can be temporarily inhibited in case of infection to allow the onset of inflammation. Morbidity and mortality in COVID-19 patients have been associated with dysregulated inflammation, a process that may include aberrant ARE activity. RESULTS: RNA-seq data from available transcriptomic studies were analyzed to investigate a possible differential expression of mRNAs that contain AREs in the context of SARS-CoV-2 infections. ARE-mRNAs turned out to be significantly overrepresented among the upregulated mRNAs after SARS-CoV-2 infection (up to 42%). In contrast, ARE-mRNAs were underrepresented (16%) in the downregulated group. Consequently, at a global scale, ARE-mRNAs are significantly more upregulated after SARS-CoV-2 infection compared to non-ARE mRNAs. This observation was apparent in lung cell line models such as A549 and Calu-3 and with infections with other respiratory viruses and cell lines. Most importantly, at the clinical level, the elevated ARE-mRNA response appeared strongest in blood cells of COVID-19 patients with mild disease. It diminished with disease severity and was least apparent in patients in need of intubation and respiratory-related death. Gene function and clustering analysis suggest that the ARE-response is rather global and the upregulated ARE-mRNAs in patients with mild disease do not particularly cluster in specific functional groups. CONCLUSIONS: Compared to the rest of the transcriptome, ARE-containing mRNAs are preferentially upregulated in response to viral infections at a global level. In the context of COVID-19, they are most upregulated in mild disease. Due to their large number, their levels measured by RNA-seq may provide a reliable indication of COVID-19 severity.


Subject(s)
COVID-19 , Humans , RNA, Messenger/genetics , RNA, Messenger/metabolism , COVID-19/genetics , Up-Regulation/genetics , SARS-CoV-2 , Inflammation
3.
Mol Oncol ; 15(8): 2120-2139, 2021 08.
Article in English | MEDLINE | ID: mdl-33411958

ABSTRACT

Dysfunctions in post-transcriptional control are observed in cancer and chronic inflammatory diseases. Here, we employed a kinome inhibitor library (n = 378) in a reporter system selective for 3'-untranslated region-AU-rich elements (ARE). Fifteen inhibitors reduced the ARE-reporter activity; among the targets is the polo-like kinase 1 (PLK1). RNA-seq experiments demonstrated that the PLK1 inhibitor, volasertib, reduces the expression of cytokine and cell growth ARE mRNAs. PLK1 inhibition caused accelerated mRNA decay in cancer cells and was associated with reduced phosphorylation and stability of the mRNA decay-promoting protein, tristetraprolin (ZFP36/TTP). Ectopic expression of PLK1 increased abundance and stability of high molecular weight of ZFP36/TTP likely of the phosphorylated form. PLK1 effect was associated with the MAPK-MK2 pathway, a major regulator of ARE-mRNA stability, as evident from MK2 inhibition, in vitro phosphorylation, and knockout experiments. Mutational analysis demonstrates that TTP serine 186 is a target for PLK1 effect. Treatment of mice with the PLK1 inhibitor reduced both ZFP36/TTP phosphorylation in xenograft tumor tissues, and the tumor size. In cancer patients' tissues, PLK1/ARE-regulated gene cluster was overexpressed in solid tumors and associated with poor survival. The data showed that PLK1-mediated post-transcriptional aberration could be a therapeutic target.


Subject(s)
Cell Cycle Proteins/metabolism , Neoplasms/genetics , Protein Kinase Inhibitors/pharmacology , Protein Kinases/drug effects , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , RNA Processing, Post-Transcriptional , 3' Untranslated Regions , Animals , Humans , Mice , Mice, Nude , Phosphorylation , Pteridines/pharmacology , Tristetraprolin/pharmacology , Xenograft Model Antitumor Assays , Polo-Like Kinase 1
4.
RNA Biol ; 16(3): 309-319, 2019 03.
Article in English | MEDLINE | ID: mdl-30664390

ABSTRACT

AU-rich elements (AREs) are cis-acting instability and translation inhibition elements that are present in the 3'UTR of most inducible inflammatory mRNAs such as TNF and Cxcl2. mRNAs that contain AREs are, by default, repressed and only transiently expressed in response to stimuli. They are targeted by the inducible RNA-binding protein Tristetraprolin (TTP) which blocks their translation and facilitates their decay, thereby contributing to the quick termination of their expression. The exogenous over-expression of TTP in HEK293 cells can unexpectedly lead to the upregulation and extended expression of a nanoLuciferase reporter that contains the ARE of TNF. Here we show that, a moderate downregulation of the highly expressed endogenous TTP after LPS induction by siRNA in macrophages can lead to a reduction in the release of TNF and Cxcl2. We propose that, in contrast to their canonical function, very high levels of induced TTP at the onset of the inflammatory response can enhance the expression of ARE-mRNAs at the post-transcriptional level, independently of phosphorylation status. As the inflammatory response progresses, TTP levels diminish but they continuously regain their ability to reduce the expression of ARE-mRNAs to reach a turning point of 'optimal TTP level' with a maximum ability to repress ARE-mRNA expression. Below this level, a further reduction in TTP levels now leads to the loss of canonical-TTP function resulting in increased ARE-mRNA expression. These novel findings should contribute to the understanding of feedback loops that control the kinetics of the inflammatory response.


Subject(s)
Inflammation/genetics , Inflammation/metabolism , RNA Processing, Post-Transcriptional , Tristetraprolin/genetics , Tristetraprolin/metabolism , 3' Untranslated Regions , AU Rich Elements , Animals , Chemokine CXCL2/metabolism , Down-Regulation , Gene Knockdown Techniques , Genes, Reporter , Humans , Mice , RNA Stability , RNA, Messenger , Tristetraprolin/chemistry , Tumor Necrosis Factor-alpha/metabolism
5.
Front Immunol ; 10: 3050, 2019.
Article in English | MEDLINE | ID: mdl-32010134

ABSTRACT

Cyclic-di-AMP (c-di-AMP) is a bacterial second messenger that is produced by intracellular bacterial pathogens in mammalian host macrophages. Previous reports have shown that c-di-AMP is recognized by intracellular pattern recognition receptors of the innate immune system and stimulate type I interferon response. Here we report that the response to c-di-AMP includes a post-transcriptional component that is involved in the induction of additional inflammatory cytokines including IL-6, CXCL2, CCL3, and CCL4. Their mRNAs contain AU-rich elements (AREs) in their 3' UTR that promote decay and repress translation. We show that c-di-AMP leads to the phosphorylation of p38 MAPK as well as the induction of the ARE-binding protein TTP, both of which are components of a signaling pathway that modulate the expression of ARE-containing mRNAs at the post-transcriptional level. Pharmacological inhibition of p38 reduces the c-di-AMP-dependent release of induced cytokines, while TTP knockdown increases their release and mRNA stability. C-di-AMP can specifically increase the expression of a nano-Luciferase reporter that contains AREs. We propose a non-canonical intracellular mode of activation of the p38 MAPK pathway with the subsequent enhancement in the expression of inflammatory cytokines. C-di-AMP is widely distributed in bacteria, including infectious intracellular pathogens; hence, understanding of its post-transcriptional gene regulatory effect on the host response may provide novel approaches for therapy.


Subject(s)
Bacteria/metabolism , Bacterial Infections/genetics , Bacterial Infections/microbiology , Dinucleoside Phosphates/metabolism , Host-Pathogen Interactions/genetics , RNA Processing, Post-Transcriptional , 3' Untranslated Regions , AU Rich Elements , Animals , Bacteria/immunology , Bacterial Infections/immunology , Bacterial Infections/metabolism , Cytokines/chemistry , Cytokines/metabolism , Gene Expression Regulation , Genes, Reporter , Host-Pathogen Interactions/immunology , Mice , Open Reading Frames , Promoter Regions, Genetic , RAW 264.7 Cells , RNA Stability , Signal Transduction , p38 Mitogen-Activated Protein Kinases
6.
Biochim Biophys Acta Gene Regul Mech ; 1861(2): 167-177, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29413897

ABSTRACT

Adenylate-uridylate (AU)-rich elements (AREs) are sequence instability elements that are known to be located in the 3' untranslated regions (UTR) in thousands of human transcripts. AREs regulate the expression of many genes at the post-transcriptional level, and they are essential for many normal cellular functions. We conducted a transcriptome-wide screen for AREs and found that they are most abundant in introns, with up to 25% of introns containing AREs corresponding to 58% of human genes. Clustering studies of ARE size, complexity, and distribution revealed that, in introns, longer AREs with two or more overlapping repeats are more abundant than in the 3'UTR, and only introns can contain very long AREs with 6-14 overlapping AUUUA pentamers. We found that intronic sites of the ARE binding proteins HuR/ELAVL1, ZFP36/TTP, AUF1, and BRF1/ZFP36L1 overlap with the intronic AREs with HuR being most abundant. Accordingly, RNA-IP experiments demonstrated a specific association of HuR with reporter and endogenous pre-mRNAs that contain intronic AREs. Moreover, HuR knockdown led to a significant general reduction in the mRNA levels of genes that contain intronic AREs and to a specific reduction in the expression of ARE-intronic reporters. The data represent bioinformatics analysis for key RNA-binding proteins interactions with intronic AREs and provide experimental evidence for HuR binding to AREs. The widespread distribution of intronic AREs and their particular association with HuR and HuR binding sites indicates that more than half of human genes can be regulated post-transcriptionally by AREs.


Subject(s)
AU Rich Elements/genetics , ELAV-Like Protein 1/genetics , Gene Expression Regulation , Introns/genetics , Transcriptome/genetics , 3' Untranslated Regions/genetics , Base Sequence , Binding Sites/genetics , ELAV-Like Protein 1/metabolism , HEK293 Cells , Humans , Protein Binding , RNA Interference
7.
Nucleic Acids Res ; 46(D1): D218-D220, 2018 01 04.
Article in English | MEDLINE | ID: mdl-29077946

ABSTRACT

Here we present an updated version of the AU-Rich Element Database (ARED-Plus) that is freely available at http://brp.kfshrc.edu.sa/ared. AREs are conserved sequence elements that were first discovered in the 3'UTR of mammalian transcripts. Over the past years, we compiled a series of ARE databases that revealed the extent and wide distribution of ARE-containing genes. For this update, we adopted an optimized search algorithm with improved specificity and sensitivity in ARE selection. The designation of the different ARE clusters was simplified by directly correlating the number of the ARE cluster to the number of overlapping AUUUA pentamers. Additionally, the new database was expanded to include genes with intronic AREs (pre-mRNAs) and their characteristics since recent observations reported their abundance and biological significance. Several enhancements were incorporated such as customized column view, additional search options and live search functionalities. The new version includes links to AREsite and AREScore, two related ARE assessment algorithms for further evaluation of the ARE characteristics. ARED-Plus now contains an updated repertoire of AREs in the human transcriptome that may be useful in several research fields.


Subject(s)
AU Rich Elements , Databases, Nucleic Acid , RNA, Messenger/genetics , 3' Untranslated Regions , Algorithms , Animals , Humans , Internet , Introns , Search Engine
8.
Cancer Res ; 76(14): 4068-80, 2016 07 15.
Article in English | MEDLINE | ID: mdl-27197193

ABSTRACT

Defects in AU-rich elements (ARE)-mediated posttranscriptional control can lead to several abnormal processes that underlie carcinogenesis. Here, we performed a systematic analysis of ARE-mRNA expression across multiple cancer types. First, the ARE database (ARED) was intersected with The Cancer Genome Atlas databases and others. A large set of ARE-mRNAs was over-represented in cancer and, unlike non-ARE-mRNAs, correlated with the reversed balance in the expression of the RNA-binding proteins tristetraprolin (TTP, ZFP36) and HuR (ELAVL1). Serial statistical and functional enrichment clustering identified a cluster of 11 overexpressed ARE-mRNAs (CDC6, KIF11, PRC1, NEK2, NCAPG, CENPA, NUF2, KIF18A, CENPE, PBK, TOP2A) that negatively correlated with TTP/HuR mRNA ratios and was involved in the mitotic cell cycle. This cluster was upregulated in a number of solid cancers. Experimentally, we demonstrated that the ARE-mRNA cluster is upregulated in a number of tumor breast cell lines when compared with noninvasive and normal-like breast cancer cells. RNA-IP demonstrated the association of the ARE-mRNAs with TTP and HuR. Experimental modulation of TTP or HuR expression led to changes in the mitosis ARE-mRNAs. Posttranscriptional reporter assays confirmed the functionality of AREs. Moreover, TTP augmented mitotic cell-cycle arrest as demonstrated by flow cytometry and histone H3 phosphorylation. We found that poor breast cancer patient survival was significantly associated with low TTP/HuR mRNA ratios and correlated with high levels of the mitotic ARE-mRNA signature. These results significantly broaden the role of AREs and their binding proteins in cancer, and demonstrate that TTP induces an antimitotic pathway that is diminished in cancer. Cancer Res; 76(14); 4068-80. ©2016 AACR.


Subject(s)
Gene Expression Regulation, Neoplastic , Neoplasms/genetics , RNA-Binding Proteins/metabolism , Adenine/analysis , Cell Cycle Checkpoints , Cell Line, Tumor , ELAV-Like Protein 1/genetics , Humans , Polyadenylation , RNA, Messenger/analysis , Tristetraprolin/genetics , Uridine/analysis
9.
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
10.
Front Biosci (Landmark Ed) ; 17(5): 1846-60, 2012 01 01.
Article in English | MEDLINE | ID: mdl-22201840

ABSTRACT

Adenylate-uridylate rich elements (AREs) in the 3'UTRs of many transiently expressed genes regulate mRNA instability and translation. Such ARE-genes are involved in vital biological processes like cellular growth, differentiation, and immunity. Defects in their expression contribute to a variety of disease conditions like cancer, autoimmune diseases, diabetes, and cardiovascular and chronic inflammatory diseases. Over the past two decades, considerable progress has been made in understanding the mode of regulation of AREs containing mRNAs by RNA-binding proteins, miRNAs, and signaling pathways. This review focuses on the less documented sequence variation affecting ARE functions and its relation to disease. We discuss reports describing genetic polymorphisms, alternative polyadenylation, and alternative splicing that can lead to the loss or gain of function of AREs, often with significant implications to disease.


Subject(s)
Genetic Variation , Alternative Splicing , Animals , Disease/genetics , Humans , Polyadenylation , Polymorphism, Genetic
11.
RNA Biol ; 8(4): 681-91, 2011.
Article in English | MEDLINE | ID: mdl-21691152

ABSTRACT

Unlike AU-rich elements (AREs) that are largely present in the 3'UTRs of many unstable mammalian mRNAs, the function and abundance of GU-rich elements (GREs) are poorly understood. We performed a genome-wide analysis and found that at least 5% of human genes contain GREs in their 3'UTRs with functional over-representation in genes involved in transcription, nucleic acid metabolism, developmental processes, and neurogenesis. GREs have similar sequence clustering patterns with AREs such as overlapping GUUUG pentamers and enrichment in 3'UTRs. Functional analysis using T-cell mRNA expression microarray data confirms correlation with mRNA destabilization. Reporter assays show that compared to AREs the ability of GREs to destabilize mRNA is modest and does not increase with the increasing number of overlapping pentamers. Naturally occurring GREs within U-rich contexts were more potent in destabilizing GFP reporter mRNAs than synthetic GREs with perfectly overlapping pentamers. Overall, we find that GREs bear a resemblance to AREs in sequence patterns but they regulate a different repertoire of genes and have different dynamics of mRNA decay. A dedicated resource on all GRE-containing genes of the human, mouse and rat genomes can be found at brp.kfshrc.edu.sa/GredOrg.


Subject(s)
3' Untranslated Regions , Gene Expression Regulation , Genome, Human , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transcriptome , Base Sequence , Cell Line , Guanine/chemistry , HEK293 Cells , Humans , RNA, Messenger/chemistry , Uracil/chemistry
12.
Biochem Pharmacol ; 80(12): 1915-20, 2010 Dec 15.
Article in English | MEDLINE | ID: mdl-20599781

ABSTRACT

Downstream of mitogen-activated protein kinases (MAPKs), three structurally related MAPK-activated protein kinases (MAPKAPKs or MKs) - MK2, MK3 and MK5 - signal to diverse cellular targets. Although there is no known common function for all three MKs, MK2 and MK3 are mainly involved in regulation of gene expression at the post-transcriptional level and are implicated in inflammation and cancer. MK2 and MK3 are phosphorylated and activated by p38(MAPKα,ß) and, in turn phosphorylate various substrates involved in diverse cellular processes. In addition to forwarding of the p38-signal by MK2/3, protein complex formation between MK2/3 and p38 mutually stabilizes these enzymes and affects p38(MAPK) signaling in general. Among the substrates of MK2/3, there are mRNA-AU-rich-element (ARE)-binding proteins, such as tristetraprolin (TTP) and hnRNP A0, which regulate mRNA stability and translation in a phosphorylation-dependent manner. Phosphorylation by MK2 stabilizes TTP, releases ARE-containing mRNAs, such as TNF-mRNA, from default translational repression and inhibits their nucleolytic degradation. Here we demonstrate that MK2/3 also contribute to the de novo synthesis of TTP. Whether this contribution proceeds via transcription factors directly targeted by MK2/3 or via chromatin remodeling by the reported binding of MK2/3 to the polycomb repressive complex is still open. A model is proposed, which demonstrates how this new function of transcriptional activation of TTP by MK2/3 cooperates with the role of MK2/3 in post-transcriptional gene expression to limit the inflammatory response.


Subject(s)
Inflammation/metabolism , Intracellular Signaling Peptides and Proteins/physiology , Protein Serine-Threonine Kinases/physiology , Tristetraprolin/biosynthesis , Tumor Necrosis Factor-alpha/biosynthesis , Animals , Humans , Mitogen-Activated Protein Kinase 14/metabolism , Phosphorylation , RNA, Messenger/biosynthesis , Transcription, Genetic , Tumor Necrosis Factor-alpha/genetics
13.
RNA ; 16(6): 1245-55, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20418359

ABSTRACT

Assessment of post-transcriptional control relies on use of transcriptional inhibitors and is masked by copious and cryptic transcriptional induction. We screened several cellular promoters that are constitutively active yet noninducible to external stimuli. The ribosomal protein RPS30 promoter was chosen; its TATA signal and sp1 site location were optimized. The modified promoter (RPS30M) is selective to post-transcriptional effects of AU-rich elements (ARE) in the 3'UTR, while it is not transcriptionally responsive to a wide variety of agents including pro-inflammatory cytokines and RNA-binding proteins. Specific cis-acting elements can be appended to RPS30M by a cloning-free approach to allow coupled transcriptional/post-transcriptional assessment, as demonstrated with NF-kappaB and beta-catenin/wnt signaling experiments. Moreover, efficient tetracycline-regulated RPS30M was created for quantitative assessment of the half-lives of mRNAs containing AREs. The described approach provides enhanced versatility and suitability for selective post-transcriptional assessment with or without transcriptional induction.


Subject(s)
RNA Processing, Post-Transcriptional , RNA Stability/genetics , RNA, Messenger/genetics , Transcription, Genetic , 3' Untranslated Regions , Cloning, Molecular , Gene Expression Regulation , Humans , NF-kappa B/genetics , Promoter Regions, Genetic , Ribosomal Proteins/genetics , Signal Transduction/genetics
14.
FEBS Lett ; 583(12): 1933-8, 2009 Jun 18.
Article in English | MEDLINE | ID: mdl-19416727

ABSTRACT

p38 mitogen-activated protein kinase (MAPK) stabilises pro-inflammatory mediator mRNAs by inhibiting AU-rich element (ARE)-mediated decay. We show that in bone-marrow derived murine macrophages tristetraprolin (TTP) is necessary for the p38 MAPK-sensitive decay of several pro-inflammatory mRNAs, including cyclooxygenase-2 and the novel targets interleukin (IL)-6, and IL-1alpha. TTP(-/-) macrophages also strongly overexpress IL-10, an anti-inflammatory cytokine that constrains the production of the IL-6 despite its disregulation at the post-transcriptional level. TTP directly controls IL-10 mRNA stability, which is increased and insensitive to inhibition of p38 MAPK in TTP(-/-) macrophages. Furthermore, TTP enhances deadenylation of an IL-10 3'-untranslated region RNA in vitro.


Subject(s)
Inflammation Mediators/metabolism , Interleukin-10/genetics , Macrophages/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Tristetraprolin/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Base Sequence , In Vitro Techniques , Interleukin-10/antagonists & inhibitors , Interleukin-12 Subunit p40/biosynthesis , Interleukin-6/biosynthesis , MAP Kinase Signaling System , Mice , Mice, Inbred C57BL , Mice, Knockout , RNA Stability , Tristetraprolin/deficiency , Tristetraprolin/genetics
15.
Eur J Neurosci ; 28(4): 642-54, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18702688

ABSTRACT

Mitogen-activated protein kinase-activated protein kinase (MK)2 is one of several downstream targets of p38 mitogen-activated protein kinase and has a well documented role in inflammation. Here, we describe a possible new function of MK2. We show that triggering depolarization by potassium chloride or increasing the cellular cAMP by forskolin treatment led to elevated levels of expression and activity of mouse MK2. In both treatments, the kinase inhibitor H89 completely prevented the up-regulation of MK2 at the transcript level. By the use of different cell lines we demonstrated that the induction of MK2 expression is characteristic of neuronal cells and is absent in fibroblasts, macrophages and kidney cells. In vivo, induction of a status epilepticus by systemic administration of the chemoconvulsant kainic acid resulted in markedly reduced neurodegeneration in the pyramidal layer of the hippocampus, dentate gyrus and hilus of MK2-deficient mice compared with wild-type mice. Together, our data suggest a possible role of MK2 in the cellular response after neuronal depolarization, in particular in excitotoxicity.


Subject(s)
Intracellular Signaling Peptides and Proteins/metabolism , Membrane Potentials/physiology , Neurons/physiology , Protein Serine-Threonine Kinases/metabolism , Animals , Cells, Cultured , Colforsin/metabolism , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Cyclic AMP-Dependent Protein Kinases/metabolism , Enzyme Activation , Enzyme Stability , Excitatory Amino Acid Agonists/pharmacology , Hippocampus/cytology , Hippocampus/drug effects , Hippocampus/metabolism , Humans , Intracellular Signaling Peptides and Proteins/genetics , Isoquinolines/metabolism , Kainic Acid/pharmacology , Mice , Mice, Knockout , Neurons/cytology , PC12 Cells , Potassium Chloride/metabolism , Protein Kinase Inhibitors/metabolism , Protein Serine-Threonine Kinases/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Seizures/chemically induced , Signal Transduction/physiology , Sulfonamides/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
16.
J Neurochem ; 105(5): 2039-52, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18298661

ABSTRACT

The inflammatory response in the brain is closely associated with the pathogenesis of degenerative neurological disorders. A role for the p38 stress-activated protein kinase/MAPK-activated protein kinase 2 (MK2) axis in inflammation and apoptosis is well documented. Here, we provide evidence that neurodegeneration can be prevented by eliminating MK2. In primary mesencephalic neuron-glia co-cultures dopaminergic neurons from MK2-deficient (MK2-/-) mice were significantly more resistant to lipopolysaccharide-induced neurotoxicity compared with cells from wild-type mice. This neuroprotection in MK2-deficient cultures was associated with a reduced inflammatory response, especially with reduced production of the inflammatory mediators tumor necrosis factor alpha, keratinocyte-derived chemokine, interleukin-6, and nitric oxide (NO). Interestingly, in primary neuron-enriched cell cultures p38 MAPK, but not MK2, also participates in NO-mediated neuronal cell death. In the MPTP mouse model for Parkinson's disease, MK2-deficient mice show a reduced neuroinflammation and less degeneration of dopaminergic neurons in the substantia nigra after MPTP lesion compared with wild-type mice. In conclusion, our results reveal that MK2 does not directly participate in neuronal cell death, but indirectly contributes to neurodegeneration by the production of neurotoxic substances, such as NO or tumor necrosis factor alpha, from activated glia cells.


Subject(s)
Disease Models, Animal , Intracellular Signaling Peptides and Proteins/deficiency , Neurons/enzymology , Neurons/pathology , Parkinson Disease/enzymology , Parkinson Disease/pathology , Protein Serine-Threonine Kinases/deficiency , Animals , Cell Death/genetics , Cells, Cultured , Inflammation/enzymology , Inflammation/genetics , Inflammation/pathology , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Parkinson Disease/genetics , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/physiology
17.
Mol Cell Biol ; 26(6): 2399-407, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16508014

ABSTRACT

The mitogen-activated protein kinase (MAPK) p38/MAPK-activated protein kinase 2 (MK2) signaling pathway plays an important role in the posttranscriptional regulation of tumor necrosis factor (TNF), which is dependent on the adenine/uridine-rich element (ARE) in the 3' untranslated region of TNF mRNA. After lipopolysaccharide (LPS) stimulation, MK2-deficient macrophages show a 90% reduction in TNF production compared to the wild type. Tristetraprolin (TTP), a protein induced by LPS, binds ARE and destabilizes TNF mRNA. Accordingly, macrophages lacking TTP produce large amounts of TNF. Here, we generated MK2/TTP double knockout mice and show that, after LPS stimulation, bone marrow-derived macrophages produce TNF mRNA and protein levels comparable to those of TTP knockout cells, indicating that in the regulation of TNF biosynthesis TTP is genetically downstream of MK2. In addition, we show that MK2 is essential for the stabilization of TTP mRNA, and phosphorylation by MK2 leads to increased TTP protein stability but reduced ARE affinity. These data suggest that MK2 inhibits the mRNA destabilizing activity of TTP and, in parallel, codegradation of TTP together, with the target mRNA resulting in increased cellular levels of TTP.


Subject(s)
Protein Kinases/metabolism , RNA Stability , RNA, Messenger/metabolism , Tristetraprolin/metabolism , Tumor Necrosis Factor-alpha/genetics , 3' Untranslated Regions , Abnormalities, Multiple/genetics , Abnormalities, Multiple/pathology , Adenine/metabolism , Animals , Cells, Cultured , Down-Regulation , Intracellular Signaling Peptides and Proteins , Macrophages/metabolism , Macrophages/pathology , Mice , Mice, Mutant Strains , Phosphorylation , Protein Biosynthesis , Protein Kinases/genetics , Protein Serine-Threonine Kinases , RNA Processing, Post-Transcriptional , RNA, Messenger/genetics , Tristetraprolin/genetics , Tumor Necrosis Factor-alpha/metabolism , Uridine/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
18.
FEBS Lett ; 574(1-3): 25-30, 2004 Sep 10.
Article in English | MEDLINE | ID: mdl-15358534

ABSTRACT

Xenopus laevis RNA-binding protein A (Xlrbpa) is a highly conserved, ubiquitously expressed hnRNP- and ribosome-associated RNA-binding protein that contains three double stranded RNA-binding domains (dsRBDs) in tandem arrangement. A two-hybrid screen with Xlrbpa as a bait recovered Xlrbpa itself as the strongest interaction partner, indicating multimerization of this protein. To search for regions responsible for the observed interaction, we conducted two-hybrid assays with Xlrbpa deletion constructs and identified the third dsRBD of Xlrbpa as the exclusive interacting domain. Additionally, these results were confirmed by coimmunoprecipitation experiments with truncated proteins expressed both in yeast and Xenopus oocytes. In PACT, the human homologue of Xlrbpa, we could demonstrate that the third dsRBD displays the same multimerization activity. Interestingly, this domain is essential for the activation of the dsRNA-activated protein kinase PKR. Addition of RNAses to coimmunoprecipitation experiments did not affect the dimerization, suggesting that the interaction is independent of RNA-binding. We report here a homomultimerization activity of a type B dsRBD and suggest possible implications that include a model for PKR activation by PACT.


Subject(s)
RNA, Double-Stranded/metabolism , Xenopus Proteins/metabolism , Amino Acid Sequence , Animals , Base Sequence , Biopolymers , Cloning, Molecular , DNA Primers , Molecular Sequence Data , Sequence Homology, Amino Acid , Two-Hybrid System Techniques , Xenopus Proteins/chemistry , Xenopus laevis
SELECTION OF CITATIONS
SEARCH DETAIL
...