20 research outputs found

    Potential limitations of the "Sleeping Beauty" transposon use in gene expression studies

    Get PDF

    Construction of a set of novel transposon vectors for efficient silencing of protein and lncRNA genes via CRISPR interference

    Get PDF
    In recent years, CRISPR interference (CRISPRi) technology of gene silencing has emerged as a promising alternative to RNA interference (RNAi) surpassing the latter in terms of efficiency and accuracy. Here, we describe the construction of a set of transposon vectors suitable for constitutive or tetracycline (doxycycline)-inducible silencing of genes of interest via CRISPRi method and conferring three different antibiotic resistances, using vectors available via Addgene repository. We have analyzed the performance of the new vectors in the silencing of mouse Adam10 and human lncRNA, NORAD. The empty vector variants can be used to efficiently silence any genes of interest

    EGF activates TTP expression by activation of ELK-1 and EGR-1 transcription factors

    Get PDF
    <p>Abstract</p> <p>Background</p> <p>Tristetraprolin (TTP) is a key mediator of processes such as inflammation resolution, the inhibition of autoimmunity and in cancer. It carries out this role by the binding and degradation of mRNA transcripts, thereby decreasing their half-life. Transcripts modulated by TTP encode proteins such as cytokines, pro-inflammatory agents and immediate-early response proteins. TTP can also modulate neoplastic phenotypes in many cancers. TTP is induced and functionally regulated by a spectrum of both pro- and anti-inflammatory cytokines, mitogens and drugs in a MAPK-dependent manner. So far the contribution of p38 MAPK to the regulation of TTP expression and function has been best described.</p> <p>Results</p> <p>Our results demonstrate the induction of the gene coding TTP (<it>ZFP36</it>) by EGF through the ERK1/2-dependent pathway and implicates the transcription factor ELK-1 in this process. We show that ELK-1 regulates <it>ZFP36 </it>expression by two mechanisms: by binding the <it>ZFP36 </it>promoter directly through ETS-binding site (+ 883 to +905 bp) and by inducing expression of EGR-1, which in turn increases <it>ZFP36 </it>expression through sequences located between -111 and -103 bp.</p> <p>Conclusions</p> <p>EGF activates TTP expression via ELK-1 and EGR-1 transcription factors.</p

    The molecular basis of tRNA selectivity by human pseudouridine synthase 3

    Get PDF
    Pseudouridine (Ψ), the isomer of uridine, is ubiquitously found in RNA, including tRNA, rRNA, and mRNA. Human pseudouridine synthase 3 (PUS3) catalyzes pseudouridylation of position 38/39 in tRNAs. However, the molecular mechanisms by which it recognizes its RNA targets and achieves site specificity remain elusive. Here, we determine single-particle cryo-EM structures of PUS3 in its apo form and bound to three tRNAs, showing how the symmetric PUS3 homodimer recognizes tRNAs and positions the target uridine next to its active site. Structure-guided and patient-derived mutations validate our structural findings in complementary biochemical assays. Furthermore, we deleted PUS1 and PUS3 in HEK293 cells and mapped transcriptome-wide Ψ sites by Pseudo-seq. Although PUS1-dependent sites were detectable in tRNA and mRNA, we found no evidence that human PUS3 modifies mRNAs. Our work provides the molecular basis for PUS3-mediated tRNA modification in humans and explains how its tRNA modification activity is linked to intellectual disabilities

    The role of NF-κB and Elk-1 in the regulation of mouse ADAM17 expression

    No full text
    ADAM17 is a cell membrane metalloproteinase responsible for the release of ectodomains of numerous proteins from the cell surface. Although ADAM17 is often overexpressed in tumours and at sites of inflammation, little is known about the regulation of its expression. Here we investigate the role of NF-κB and Elk-1 transcription factors and upstream signalling pathways, NF-κB and ERK1/2 in ADAM17 expression in mouse brain endothelial cells stimulated with pro-inflammatory factors (TNF, IL-1β, LPS) or a phorbol ester (PMA), a well-known stimulator of ADAM17 activity. Notably, NF-κB inhibitor, IKK VII, interfered with the IL-1β- and LPS-mediated stimulation of ADAM17 expression. Furthermore, Adam17 promoter contains an NF-κB binding site occupied by p65 subunit of NF-κB. The transient increase in Adam17 mRNA in response to PMA was strongly reduced by an inhibitor of ERK1/2 phosphorylation, U0126. Luciferase reporter assay with vectors encoding the ERK1/2 substrate, Elk-1, fused with constitutively activating or repressing domains, indicated Elk-1 involvement in Adam17 expression. The site-directed mutagenesis of potential Elk-1 binding sites pointed to four functional Elk-1 binding sites in Adam17 promoter. All in all, our results indicate that NF-κB and Elk-1 transcription factors via NF-κB and ERK1/2 signalling pathways contribute to the regulation of mouse Adam17 expression

    IF-combined smRNA FISH reveals interaction of MCPIP1 protein with IER3 mRNA

    Get PDF
    MCPIP1 and IER3 are recently described proteins essential for maintenance of immune homeostasis. IER3 is involved in the regulation of apoptosis and differentiation and has been shown lately to protect activated T cells and macrophages from apoptosis. MCPIP1 is an RNase critical for controlling inflammation-related mRNAs. MCPIP1 interacts with and degrades a set of stem-loop-containing mRNAs (including IL-6). Our results demonstrate the involvement of MCPIP1 in the regulation of IER3 mRNA levels. A dual luciferase assay revealed that over-expression of MCPIP1 resulted in a decrease of luciferase activity in the samples co-transfected with constructs containing luciferase CDS attached to IER3 3′UTR. We identified a stem-loop structure similar to that described to be important for destabilization of the IL-6 mRNA by MCPIP1. Examination of IER3 3′UTR sequence, structure and evolutionary conservation revealed that the identified stem-loop is buried within a bigger element. Deletion of this fragment abolished the regulation of IER3 3′UTR-containing transcript by MCPIP1. Finally, using immunofluorescence-combined single-molecule RNA FISH we have shown that the MCPIP1 protein co-localizes with IER3 mRNA. By this method we also proved that the presence of the wild-type NYN/PIN-like domain of MCPIP1 correlated with the decreased level of IER3 mRNA. RNA immunoprecipitation further confirmed the interaction of MCPIP1 with IER3 transcripts in vivo
    corecore