Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Clin Mol Hepatol ; 30(2): 177-190, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38163441

ABSTRACT

BACKGROUND/AIMS: New prognostic markers are needed to identify patients with hepatocellular carcinoma (HCC) who carry a worse prognosis. Ultra-low-pass whole-genome sequencing (ULP-WGS) (≤0.5× coverage) of cell-free DNA (cfDNA) has emerged as a low-cost promising tool to assess both circulating tumor DNA (ctDNA) fraction and large structural genomic alterations. Here, we studied the performance of ULP-WGS of plasma cfDNA to infer prognosis in patients with HCC. METHODS: Plasma samples were obtained from patients with HCC prior to surgery, locoregional or systemic therapy, and were analyzed by ULP-WGS of cfDNA to an average genome-wide fold coverage of 0.3x. ctDNA and copy number alterations (CNA) were estimated using the software package ichorCNA. RESULTS: Samples were obtained from 73 HCC patients at different BCLC stages (BCLC 0/A: n=37, 50.7%; BCLC B/C: n=36, 49.3%). ctDNA was detected in 18 out of 31 patients who received systemic treatment. Patients with detectable ctDNA showed significantly worse overall survival (median, 13.96 months vs not reached). ctDNA remained an independent predictor of prognosis after adjustment by clinical-pathologic features and type of systemic treatment (hazard ratio 7.69; 95%, CI 2.09-28.27). Among ctDNA-positive patients under systemic treatments, the loss of large genomic regions in 5q and 16q arms was associated with worse prognosis after multivariate analysis. CONCLUSION: ULP-WGS of cfDNA provides clinically relevant information about the tumor biology. The presence of ctDNA and the loss of 5q and 16q arms in ctDNA-positive patients are independent predictors of worse prognosis in patients with advanced HCC receiving systemic therapy.


Subject(s)
Carcinoma, Hepatocellular , Cell-Free Nucleic Acids , Circulating Tumor DNA , Liver Neoplasms , Humans , Carcinoma, Hepatocellular/diagnosis , Carcinoma, Hepatocellular/genetics , Circulating Tumor DNA/genetics , Prognosis , Liver Neoplasms/diagnosis , Liver Neoplasms/genetics , Mutation , Biomarkers, Tumor
2.
Front Cell Infect Microbiol ; 13: 1110467, 2023.
Article in English | MEDLINE | ID: mdl-36761900

ABSTRACT

Background: The main objective was to evaluate the efficacy of intranasal photodynamic therapy (PDT) in SARS-CoV-2 mildly symptomatic carriers on decreasing the infectivity period. SARS-CoV-2-specific immune-stimulating effects and safety were also analysed. Methods: We performed a randomized, placebo-controlled, clinical trial in a tertiary hospital (NCT05184205). Patients with a positive SARS-CoV-2 PCR in the last 48 hours were recruited and aleatorily assigned to PDT or placebo. Patients with pneumonia were excluded. Participants and investigators were masked to group assignment. The primary outcome was the reduction in in vitro infectivity of nasopharyngeal samples at days 3 and 7. Additional outcomes included safety assessment and quantification of humoral and T-cell immune-responses. Findings: Patients were recruited between December 2021 and February 2022. Most were previously healthy adults vaccinated against COVID-19 and most carried Omicron variant. 38 patients were assigned to placebo and 37 to PDT. Intranasal PDT reduced infectivity at day 3 post-treatment when compared to placebo with a ß-coefficient of -812.2 (CI95%= -478660 - -1.3, p<0.05) infectivity arbitrary units. The probability of becoming PCR negative (ct>34) at day 7 was higher on the PDT-group, with an OR of 0.15 (CI95%=0.04-0.58). There was a decay in anti-Spike titre and specific SARS-CoV-2 T cell immunity in the placebo group 10 and 20 weeks after infection, but not in the PDT-group. No serious adverse events were reported. Interpretation: Intranasal-PDT is safe in pauci-symptomatic COVID-19 patients, it reduces SARS-CoV-2 infectivity and decelerates the decline SARS-CoV-2 specific immune-responses.


Subject(s)
COVID-19 , SARS-CoV-2 , Adult , Humans , T-Lymphocytes , Nose
3.
J Cell Biol ; 219(9)2020 09 07.
Article in English | MEDLINE | ID: mdl-32858747

ABSTRACT

Cancer is characterized by genomic instability leading to deletion or amplification of oncogenes or tumor suppressors. However, most of the altered regions are devoid of known cancer drivers. Here, we identify lncRNAs frequently lost or amplified in cancer. Among them, we found amplified lncRNA associated with lung cancer-1 (ALAL-1) as frequently amplified in lung adenocarcinomas. ALAL-1 is also overexpressed in additional tumor types, such as lung squamous carcinoma. The RNA product of ALAL-1 is able to promote the proliferation and tumorigenicity of lung cancer cells. ALAL-1 is a TNFα- and NF-κB-induced cytoplasmic lncRNA that specifically interacts with SART3, regulating the subcellular localization of the protein deubiquitinase USP4 and, in turn, its function in the cell. Interestingly, ALAL-1 expression inversely correlates with the immune infiltration of lung squamous tumors, while tumors with ALAL-1 amplification show lower infiltration of several types of immune cells. We have thus unveiled a pro-oncogenic lncRNA that mediates cancer immune evasion, pointing to a new target for immune potentiation.


Subject(s)
DNA Copy Number Variations/genetics , Immune Evasion/genetics , Lung Neoplasms/genetics , RNA, Long Noncoding/genetics , A549 Cells , Adenocarcinoma of Lung/genetics , Antigens, Neoplasm/genetics , Carcinoma, Squamous Cell/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic/genetics , Humans , NF-kappa B/genetics , Oncogenes/genetics , Ubiquitin-Specific Proteases/genetics
4.
Nat Commun ; 11(1): 936, 2020 02 18.
Article in English | MEDLINE | ID: mdl-32071317

ABSTRACT

LncRNAs have been shown to be direct players in chromatin regulation, but little is known about their role at active genomic loci. We investigate the role of lncRNAs in gene activation by profiling the RNA interactome of SMARCB1-containing SWI/SNF complexes in proliferating and senescent conditions. The isolation of SMARCB1-associated transcripts, together with chromatin profiling, shows prevalent association to active regions where SMARCB1 differentially binds locally transcribed RNAs. We identify SWINGN, a lncRNA interacting with SMARCB1 exclusively in proliferating conditions, exerting a pro-oncogenic role in some tumor types. SWINGN is transcribed from an enhancer and modulates the activation of GAS6 oncogene as part of a topologically organized region, as well as a larger network of pro-oncogenic genes by favoring SMARCB1 binding. Our results indicate that SWINGN influences the ability of the SWI/SNF complexes to drive epigenetic activation of specific promoters, suggesting a SWI/SNF-RNA cooperation to achieve optimal transcriptional activation.


Subject(s)
Gene Expression Regulation, Neoplastic , Neoplasms/genetics , Promoter Regions, Genetic/genetics , RNA, Long Noncoding/metabolism , SMARCB1 Protein/metabolism , Animals , Apoptosis/genetics , Carcinogenesis , Cell Proliferation/genetics , Datasets as Topic , Female , Gene Regulatory Networks , HCT116 Cells , HEK293 Cells , Humans , Intercellular Signaling Peptides and Proteins/genetics , Mice , Neoplasms/pathology , RNA, Long Noncoding/genetics , RNA, Small Interfering/metabolism , RNA-Seq , Transcriptional Activation , Xenograft Model Antitumor Assays
6.
J Exp Clin Cancer Res ; 38(1): 172, 2019 04 24.
Article in English | MEDLINE | ID: mdl-31014355

ABSTRACT

BACKGROUND: Thousands of long noncoding RNAs (lncRNAs) are aberrantly expressed in various types of cancers, however our understanding of their role in the disease is still very limited. METHODS: We applied RNAseq analysis from patient-derived data with validation in independent cohort of patients. We followed these studies with gene regulation analysis as well as experimental dissection of the role of the identified lncRNA by multiple in vitro and in vivo methods. RESULTS: We analyzed RNA-seq data from tumors of 456 CRC patients compared to normal samples, and identified SNHG15 as a potentially oncogenic lncRNA that encodes a snoRNA in one of its introns. The processed SNHG15 is overexpressed in CRC tumors and its expression is highly correlated with poor survival of patients. Interestingly, SNHG15 is more highly expressed in tumors with high levels of MYC expression, while MYC protein binds to two E-box motifs on SNHG15 sequence, indicating that SNHG15 transcription is directly regulated by the oncogene MYC. The depletion of SNHG15 by siRNA or CRISPR-Cas9 inhibits cell proliferation and invasion, decreases colony formation as well as the tumorigenic capacity of CRC cells, whereas its overexpression leads to opposite effects. Gene expression analysis performed upon SNHG15 inhibition showed changes in multiple relevant genes implicated in cancer progression, including MYC, NRAS, BAG3 or ERBB3. Several of these genes are functionally related to AIF, a protein that we found to specifically interact with SNHG15, suggesting that the SNHG15 acts, at least in part, by regulating the activity of AIF. Interestingly, ROS levels, which are directly regulated by AIF, show a significant reduction in SNHG15-depleted cells. Moreover, knockdown of SNHG15 increases the sensitiveness of the cells to 5-FU, while its overexpression renders them more resistant to the chemotherapeutic drug. CONCLUSION: Altogether, these results describe an important role of SNHG15 in promoting colon cancer and mediating drug resistance, suggesting its potential as prognostic marker and target for RNA-based therapies.


Subject(s)
Apoptosis Inducing Factor/genetics , Colorectal Neoplasms/genetics , Proto-Oncogene Proteins c-myc/genetics , RNA, Long Noncoding/genetics , Animals , CRISPR-Cas Systems/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Drug Resistance, Neoplasm/genetics , Female , Fluorouracil/administration & dosage , Gene Expression Regulation, Neoplastic/drug effects , Humans , Kaplan-Meier Estimate , Male , Mice , Middle Aged , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , RNA, Small Nucleolar/genetics , Sequence Analysis, RNA , Xenograft Model Antitumor Assays
7.
Cell ; 173(6): 1318-1319, 2018 05 31.
Article in English | MEDLINE | ID: mdl-29856950

ABSTRACT

The role of the noncoding genome in cancer biology is continually expanding. Cho et al. reveal a new and unexpected mechanism for the regulation of MYC expression mediated by the promoter sequence of its neighbor gene PVT1. This DNA element acts as a promoter-enhancer competitor and a candidate tumor suppressor.


Subject(s)
Promoter Regions, Genetic , RNA, Long Noncoding , Cell Line, Tumor , DNA, Neoplasm
8.
Genome Biol ; 18(1): 206, 2017 10 31.
Article in English | MEDLINE | ID: mdl-29084573

ABSTRACT

A major shift in our understanding of genome regulation has emerged recently. It is now apparent that the majority of cellular transcripts do not code for proteins, and many of them are long noncoding RNAs (lncRNAs). Increasingly, studies suggest that lncRNAs regulate gene expression through diverse mechanisms. We review emerging mechanistic views of lncRNAs in gene regulation in the cell nucleus. We discuss the functional interactions that lncRNAs establish with other molecules as well as the relationship between lncRNA transcription and function. While some of these mechanisms are specific to lncRNAs, others might be shared with other types of genes.


Subject(s)
Gene Expression Regulation , RNA, Long Noncoding/metabolism , Animals , Chromatin/metabolism , DNA/chemistry , DNA/metabolism , DNA-Binding Proteins/metabolism , Enhancer Elements, Genetic , RNA-Binding Proteins/metabolism , Transcription, Genetic
10.
Mol Cell ; 63(3): 397-407, 2016 08 04.
Article in English | MEDLINE | ID: mdl-27477908

ABSTRACT

Long noncoding RNAs (lncRNAs) are involved in diverse cellular processes through multiple mechanisms. Here, we describe a previously uncharacterized human lncRNA, CONCR (cohesion regulator noncoding RNA), that is transcriptionally activated by MYC and is upregulated in multiple cancer types. The expression of CONCR is cell cycle regulated, and it is required for cell-cycle progression and DNA replication. Moreover, cells depleted of CONCR show severe defects in sister chromatid cohesion, suggesting an essential role for CONCR in cohesion establishment during cell division. CONCR interacts with and regulates the activity of DDX11, a DNA-dependent ATPase and helicase involved in DNA replication and sister chromatid cohesion. These findings unveil a direct role for an lncRNA in the establishment of sister chromatid cohesion by modulating DDX11 enzymatic activity.


Subject(s)
Chromatids/metabolism , DNA Replication , DNA, Neoplasm/biosynthesis , Neoplasms/metabolism , RNA, Long Noncoding/metabolism , A549 Cells , Animals , Apoptosis , Cell Proliferation , Chromatids/genetics , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/metabolism , DNA Helicases/genetics , DNA Helicases/metabolism , DNA, Neoplasm/genetics , Female , Gene Expression Regulation, Neoplastic , HCT116 Cells , HeLa Cells , Humans , Mice, Inbred BALB C , Mice, Transgenic , Neoplasms/genetics , Neoplasms/pathology , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , RNA Interference , RNA, Long Noncoding/genetics , Time Factors , Transcription, Genetic , Transcriptional Activation , Transfection , Tumor Burden , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
11.
Sci Rep ; 5: 7615, 2015 Jan 05.
Article in English | MEDLINE | ID: mdl-25556859

ABSTRACT

Ubiquitin fusion degradation (UFD) substrates are delivered at the proteasome by a handover mechanism involving the ubiquitin-selective chaperone Cdc48 and the ubiquitin shuttle factor Rad23. Here, we show that introduction of a 20 amino acid peptide extension not only rendered degradation independent of Cdc48, in line with the model that this chaperone is involved in early unfolding events of tightly folded substrates, but at the same time relieved the need for efficient polyubiquitylation and the ubiquitin shuttle factor Rad23. Removal of the ubiquitylation sites in the N-terminal UFD signal made the degradation of this substrate strictly dependent on the peptide extension and also on Cdc48 and, importantly the presence of a functional ubiquitylation machinery. This suggests that the extension in the absence of N-terminal ubiquitylation sites is not properly positioned to engage the unfoldase machinery of the proteasome. Thus the need for efficient ubiquitylation and Cdc48 in facilitating proteasomal degradation are tightly linked but can be bypassed in the context of UFD substrates by the introduction of an unstructured extension. Our data suggest that polyubiquitin-binding complexes acting upstream of the proteasome, rather than the proteasome itself, can be primary determinants for the level of ubiquitylation required for protein degradation.


Subject(s)
Adenosine Triphosphatases/metabolism , Cell Cycle Proteins/metabolism , Proteasome Endopeptidase Complex/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Adenosine Triphosphatases/genetics , Cell Cycle Proteins/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Mutation , Protein Structure, Tertiary , Proteolysis , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Substrate Specificity , Ubiquitin/metabolism , Ubiquitination , Valosin Containing Protein
12.
Nat Commun ; 5: 5812, 2014 Dec 19.
Article in English | MEDLINE | ID: mdl-25524025

ABSTRACT

Despite the inarguable relevance of p53 in cancer, genome-wide studies relating endogenous p53 activity to the expression of lncRNAs in human cells are still missing. Here, by integrating RNA-seq with p53 ChIP-seq analyses of a human cancer cell line under DNA damage, we define a high-confidence set of 18 lncRNAs that are p53 transcriptional targets. We demonstrate that two of the p53-regulated lncRNAs are required for the efficient binding of p53 to some of its target genes, modulating the p53 transcriptional network and contributing to apoptosis induction by DNA damage. We also show that the expression of p53-lncRNAs is lowered in colorectal cancer samples, constituting a tumour suppressor signature with high diagnostic power. Thus, p53-regulated lncRNAs establish a positive regulatory feedback loop that enhances p53 tumour suppressor activity. Furthermore, the signature defined by p53-regulated lncRNAs supports their potential use in the clinic as biomarkers and therapeutic targets.


Subject(s)
Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Gene Regulatory Networks , RNA, Long Noncoding/metabolism , Tumor Suppressor Protein p53/metabolism , DNA Damage , Gene Expression Regulation, Neoplastic , Genes, Tumor Suppressor , HCT116 Cells , Humans , Protein Binding , RNA, Long Noncoding/genetics , Tumor Suppressor Protein p53/genetics
13.
Blood ; 124(24): 3646-55, 2014 Dec 04.
Article in English | MEDLINE | ID: mdl-25293769

ABSTRACT

Tissue factor (TF) (CD142) is a 47 kDa transmembrane cell surface glycoprotein that triggers the extrinsic coagulation cascade and links thrombosis with inflammation. Although macrophage TF expression is known to be regulated at the RNA level, very little is known about the mechanisms involved. Poly(adenosine 5'-diphosphate [ADP]-ribose)-polymerase (PARP)-14 belongs to a family of intracellular proteins that generate ADP-ribose posttranslational adducts. Functional screening of PARP-14-deficient macrophages mice revealed that PARP-14 deficiency leads to increased TF expression and functional activity in macrophages after challenge with bacterial lipopolysaccharide. This was related to an increase in TF messenger RNA (mRNA) stability. Ribonucleoprotein complex immunoprecipitation and biotinylated RNA pull-down assays demonstrated that PARP-14 forms a complex with the mRNA-destabilizing protein tristetraprolin (TTP) and a conserved adenylate-uridylate-rich element in the TF mRNA 3' untranslated region. TF mRNA regulation by PARP-14 was selective, as tumor necrosis factor (TNF)α mRNA, which is also regulated by TTP, was not altered in PARP-14 deficient macrophages. Consistent with the in vitro data, TF expression and TF activity, but not TNFα expression, were increased in Parp14(-/-) mice in vivo. Our study provides a novel mechanism for the posttranscriptional regulation of TF expression, indicating that this is selectively regulated by PARP-14.


Subject(s)
Gene Expression Regulation , Macrophages/metabolism , Poly(ADP-ribose) Polymerases/metabolism , Thromboplastin/biosynthesis , Tristetraprolin/metabolism , 3' Untranslated Regions/physiology , Animals , Lipopolysaccharides/pharmacology , Mice , Mice, Knockout , Poly(ADP-ribose) Polymerases/genetics , RNA Stability/drug effects , RNA Stability/physiology , Thromboplastin/genetics , Tristetraprolin/genetics , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/genetics
14.
Epigenetics ; 9(1): 21-6, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24335342

ABSTRACT

The emergence of long non-coding RNAs (lncRNAs) has shaken up our conception of gene expression regulation, as lncRNAs take prominent positions as components of cellular networks. Several cellular processes involve lncRNAs, and a significant number of them have been shown to function in cooperation with chromatin modifying enzymes to promote epigenetic activation or silencing of gene expression. Different model mechanisms have been proposed to explain how lncRNAs achieve regulation of gene expression by interacting with the epigenetic machinery. Here we describe these models in light of the current knowledge of lncRNAs, such as Xist and HOTAIR, and discuss recent literature on the role of the three-dimensional structure of the genome in the mechanism of action of lncRNAs and chromatin modifiers.


Subject(s)
Chromatin/metabolism , DNA Restriction-Modification Enzymes/metabolism , Epigenesis, Genetic/physiology , RNA, Long Noncoding/metabolism , Animals , DNA-Binding Proteins , Gene Expression , Humans , Potassium Channels, Voltage-Gated/metabolism , RNA, Long Noncoding/genetics , X Chromosome/genetics
15.
Genome Biol ; 14(9): R104, 2013.
Article in English | MEDLINE | ID: mdl-24070194

ABSTRACT

BACKGROUND: The p53 transcription factor is located at the core of a complex wiring of signaling pathways that are critical for the preservation of cellular homeostasis. Only recently it has become clear that p53 regulates the expression of several long intergenic noncoding RNAs (lincRNAs). However, relatively little is known about the role that lincRNAs play in this pathway. RESULTS: Here we characterize a lincRNA named Pint (p53 induced noncoding transcript). We show that Pint is aubiquitously expressed lincRNA that is finely regulated by p53. In mouse cells, Pint promotes cell proliferation and survival by regulating the expression of genes of the TGF-b, MAPK and p53 pathways. Pint is a nuclear lincRNA that directly interacts with the Polycomb repressive complex 2 (PRC2), and is required for PRC2 targeting of specific genes for H3K27 tri-methylation and repression. Furthermore, Pint functional activity is highly dependent on PRC2 expression. We have also identified Pint human ortholog (PINT), which presents suggestive analogies with the murine lincRNA. PINT is similarly regulated by p53, and its expression significantly correlates with the same cellular pathways as the mouse ortholog, including the p53 pathway. Interestingly, PINT is downregulated in colon primary tumors, while its overexpression inhibits the proliferation of tumor cells, suggesting a possible role as tumor suppressor. CONCLUSIONS: Our results reveal a p53 autoregulatory negative mechanism where a lincRNA connects p53 activation with epigenetic silencing by PRC2. Additionally, we show analogies and differences between the murine and human orthologs, identifying a novel tumor suppressor candidate lincRNA.


Subject(s)
Colonic Neoplasms/genetics , Epigenesis, Genetic , Histones/genetics , Polycomb Repressive Complex 2/genetics , RNA, Long Noncoding/genetics , Tumor Suppressor Protein p53/genetics , Animals , Cell Proliferation , Cell Survival , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Histones/metabolism , Humans , Mice , Mitogen-Activated Protein Kinases/genetics , Mitogen-Activated Protein Kinases/metabolism , NIH 3T3 Cells , Polycomb Repressive Complex 2/metabolism , RNA, Long Noncoding/antagonists & inhibitors , RNA, Long Noncoding/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , Tumor Suppressor Protein p53/metabolism
16.
J Biol Chem ; 285(36): 27590-600, 2010 Sep 03.
Article in English | MEDLINE | ID: mdl-20595389

ABSTRACT

Tristetraprolin (TTP) directs its target AU-rich element (ARE)-containing mRNAs for degradation by promoting removal of the poly(A) tail. The p38 MAPK pathway regulates mRNA stability via the downstream kinase MAPK-activated protein kinase 2 (MAPKAP kinase 2 or MK2), which phosphorylates and prevents the mRNA-destabilizing function of TTP. We show that deadenylation of endogenous ARE-containing tumor necrosis factor mRNA is inhibited by p38 MAPK. To investigate whether phosphorylation of TTP by MK2 regulates TTP-directed deadenylation of ARE-containing mRNAs, we used a cell-free assay that reconstitutes the mechanism in vitro. We find that phosphorylation of Ser-52 and Ser-178 of TTP by MK2 results in inhibition of TTP-directed deadenylation of ARE-containing RNA. The use of 14-3-3 protein antagonists showed that regulation of TTP-directed deadenylation by MK2 is independent of 14-3-3 binding to TTP. To investigate the mechanism whereby TTP promotes deadenylation, it was necessary to identify the deadenylases involved. The carbon catabolite repressor protein (CCR)4.CCR4-associated factor (CAF)1 complex was identified as the major source of deadenylase activity in HeLa cells responsible for TTP-directed deadenylation. CAF1a and CAF1b were found to interact with TTP in an RNA-independent fashion. We find that MK2 phosphorylation reduces the ability of TTP to promote deadenylation by inhibiting the recruitment of CAF1 deadenylase in a mechanism that does not involve sequestration of TTP by 14-3-3. Cyclooxygenase-2 mRNA stability is increased in CAF1-depleted cells in which it is no longer p38 MAPK/MK2-regulated.


Subject(s)
Intracellular Signaling Peptides and Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Ribonucleases/metabolism , Transcription Factors/metabolism , Tristetraprolin/metabolism , 14-3-3 Proteins/metabolism , Animals , Base Composition , Cyclooxygenase 2/genetics , HeLa Cells , Humans , MAP Kinase Signaling System , Mice , Phosphorylation , RNA Stability , RNA, Messenger/chemistry , RNA, Messenger/metabolism , Receptors, CCR4/metabolism , Tumor Necrosis Factor-alpha/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
17.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...