33 research outputs found

    Regulation of MMP2 and MMP9 metalloproteinases by FSH and growth factors in bovine granulosa cells

    Get PDF
    Matrix metalloproteinases (MMP) are key enzymes involved in tissue remodeling. Within the ovary, they are believed to play a major role in ovulation, and have been linked to follicle atresia. To gain insight into the regulation of MMPs, we measured the effect of hormones and growth factors on MMP2 and MMP9 mRNA levels in non-luteinizing granulosa cells in serum-free culture. FSH and IGF1 both stimulated estradiol secretion and inhibited MMP2 and MMP9 mRNA abundance. In contrast, EGF and FGF2 both inhibited estradiol secretion but had no effect on MMP expression. At physiological doses, none of these hormones altered the proportion of dead cells. Although we cannot link MMP expression with apoptosis, the specific down regulation by the gonadotropic hormones FSH and IGF1 in vitro suggests that excess MMP2 and MMP9 expression is neither required nor desired for follicle development

    Database for mRNA Half-Life of 19 977 Genes Obtained by DNA Microarray Analysis of Pluripotent and Differentiating Mouse Embryonic Stem Cells

    Get PDF
    Degradation of mRNA is one of the key processes that control the steady-state level of gene expression. However, the rate of mRNA decay for the majority of genes is not known. We successfully obtained the rate of mRNA decay for 19 977 non-redundant genes by microarray analysis of RNA samples obtained from mouse embryonic stem (ES) cells. Median estimated half-life was 7.1 h and only <100 genes, including Prdm1, Myc, Gadd45 g, Foxa2, Hes5 and Trib1, showed half-life less than 1 h. In general, mRNA species with short half-life were enriched among genes with regulatory functions (transcription factors), whereas mRNA species with long half-life were enriched among genes related to metabolism and structure (extracellular matrix, cytoskeleton). The stability of mRNAs correlated more significantly with the structural features of genes than the function of genes: mRNA stability showed the most significant positive correlation with the number of exon junctions per open reading frame length, and negative correlation with the presence of PUF-binding motifs and AU-rich elements in 3′-untranslated region (UTR) and CpG di-nucleotides in the 5′-UTR. The mRNA decay rates presented in this report are the largest data set for mammals and the first for ES cells

    The Deubiquitinase USP47 Stabilizes MAPK by Counteracting the Function of the N-end Rule ligase POE/UBR4 in <i>Drosophila</i>

    No full text
    <div><p>RAS-induced MAPK signaling is a central driver of the cell proliferation apparatus. Disruption of this pathway is widely observed in cancer and other pathologies. Consequently, considerable effort has been devoted to understanding the mechanistic aspects of RAS-MAPK signal transmission and regulation. While much information has been garnered on the steps leading up to the activation and inactivation of core pathway components, comparatively little is known on the mechanisms controlling their expression and turnover. We recently identified several factors that dictate <i>Drosophila</i> MAPK levels. Here, we describe the function of one of these, the deubiquitinase (DUB) USP47. We found that USP47 acts post-translationally to counteract a proteasome-mediated event that reduces MAPK half-life and thereby dampens signaling output. Using an RNAi-based genetic interaction screening strategy, we identified UBC6, POE/UBR4, and UFD4, respectively, as E2 and E3 enzymes that oppose USP47 activity. Further characterization of POE-associated factors uncovered KCMF1 as another key component modulating MAPK levels. Together, these results identify a novel protein degradation module that governs MAPK levels. Given the role of UBR4 as an N-recognin ubiquitin ligase, our findings suggest that RAS-MAPK signaling in <i>Drosophila</i> is controlled by the N-end rule pathway and that USP47 counteracts its activity.</p></div

    <i>Usp47</i> RNAi impacts RTK-RAS signaling downstream of MEK.

    No full text
    <p>(A) Schematic representation of the USP47 protein product with the position of the USP domain (ubiquitin specific protease catalytic domain) represented along with the amino acid length. (B) Epistasis analysis in <i>Drosophila</i> S2 cells employing constitutively active forms of RAS, RAF, and MEK to induce MAPK activation [<a href="http://www.plosbiology.org/article/info:doi/10.1371/journal.pbio.1002539#pbio.1002539.ref008" target="_blank">8</a>]. Phosphorylated MAPK was measured by quantitative microscopy and normalized to a <i>GFP</i> dsRNA control [<a href="http://www.plosbiology.org/article/info:doi/10.1371/journal.pbio.1002539#pbio.1002539.ref007" target="_blank">7</a>]. (C) MAPK activation is induced by three different receptor tyrosine kinases (RTKs) acting upstream: InR through insulin stimulation; EGFR-expressing cells stimulated with the Spitz ligand; or a heat-shock inducible constitutively activated form of Sevenless (Sev<sup>S11</sup>). JNK pathway activation induced by RAC1<sup>V12</sup> is used as a negative control. (B-C) A dsRNA targeting the Exon Junction Complex (EJC) component <i>mago</i>, a factor also positioned downstream of MEK and known to reduce MAPK expression [<a href="http://www.plosbiology.org/article/info:doi/10.1371/journal.pbio.1002539#pbio.1002539.ref007" target="_blank">7</a>], is shown for comparison. The <i>cnk</i> dsRNA is a control for a factor known to act at the level of RAF [<a href="http://www.plosbiology.org/article/info:doi/10.1371/journal.pbio.1002539#pbio.1002539.ref024" target="_blank">24</a>]. (D) The <i>sal</i><sup><i>EPv</i></sup><i>-Gal4</i> drives expression in the wing pouch, which corresponds to a segment of the wing blade extending from the L2 wing vein to the L4-5 intervein with a weaker expression area extending in the periphery of this region [<a href="http://www.plosbiology.org/article/info:doi/10.1371/journal.pbio.1002539#pbio.1002539.ref025" target="_blank">25</a>,<a href="http://www.plosbiology.org/article/info:doi/10.1371/journal.pbio.1002539#pbio.1002539.ref026" target="_blank">26</a>]. A <i>mapk</i> gain of function <i>Sevenmaker</i> (<i>Sem</i>) mutant induced the production of extra wing vein material. Two different <i>Usp47</i> RNAi lines suppressing the extra wing vein material generated by <i>mapk</i><sup><i>Sem</i></sup> are shown (these correspond to the following fly lines: <i>Usp47</i> (1), VDRC line GD26027; <i>Usp47</i> (2), NIG line 5486R-3). Raw data for (B and C) can be found in <a href="http://www.plosbiology.org/article/info:doi/10.1371/journal.pbio.1002539#pbio.1002539.s001" target="_blank">S1 Data</a>.</p

    An RNAi screen to identify factors that modify the impact of USP47 on MAPK levels.

    No full text
    <p>(A) The rescue effect of <i>Uba1</i> depletion can be robustly measured in a plate-based quantitative microscopy assay suitable for large-scale screening. <i>Usp47</i> and <i>Uba1</i> co-depletion significantly restored MAPK levels (***: <i>p</i> < 0.001) compared to a <i>Usp47</i> single depletion (**: <i>p</i> < 0.01). <i>Uba1</i> single depletion did not significantly alter MAPK levels compared to <i>GFP</i> dsRNA treated control cells (used for normalization). In this experiment, S2 cells were pre-incubated with either <i>GFP</i> dsRNA (first and third sample) or with <i>Usp47</i> dsRNA (second and fourth sample) for 3 d. The cells were then distributed in 384 well plates containing the indicated dsRNAs and incubated for another 3 d. Following dsRNA treatment, anti-MAPK stained cells were imaged and analyzed by high-content microscopy. (B) Screening strategy for a large-scale RNAi screen focused on ubiquitin-proteasome associated factors. RNAi treatment and MAPK quantification for the ubiquitin-proteasome dsRNA set was performed as in A. Each condition was tested in quadruplicate. (C) Histogram showing the distribution of results from the <i>Usp47</i> RNAi screen. The frequency (number of dsRNAs) is displayed on the <i>y</i>-axis. MAPK levels (<i>x</i>-axis) were normalized to GFP dsRNA treated cells. The <i>Usp47</i> co-depleted cells show a clear shift towards a reduction in MAPK levels. The <i>Uba1</i> dsRNA control stands out from the lot, as it completely counteracts the <i>Usp47</i> dsRNA. (D) Distribution of <i>Usp47</i> genetic interaction scores (Δ<i>m</i>; <i>x</i>-axis) for the candidate dsRNAs (frequency displayed on <i>y</i>-axis) tested in the <i>Usp47</i> RNAi screen. The negative Δ<i>m</i> obtained for Uba1 is consistent with the observed alleviation of the MAPK level reduction. Raw data for (A) can be found in <a href="http://www.plosbiology.org/article/info:doi/10.1371/journal.pbio.1002539#pbio.1002539.s001" target="_blank">S1 Data</a>. Numerical data for (C and D) can be found in <a href="http://www.plosbiology.org/article/info:doi/10.1371/journal.pbio.1002539#pbio.1002539.s012" target="_blank">S1 Table</a>.</p

    Models for regulation of MAPK stability by USP47.

    No full text
    <p>(A) Direct ubiquitination of MAPK. In this model, UBC6, POE, UFD4, and KCMF1 would participate in the direct ubiquitination of MAPK. Ubiquitinated MAPK could then be targeted to the proteasome for degradation. In the presence of USP47, MAPK would be stabilized as USP47 acts to deubiquitinate MAPK, thus counteracting the activity of the ubiquitin ligases. (B) Indirect regulation of MAPK stability. In this alternative model, UBC6, the E3s, and USP47 act on a yet unidentified factor whose proposed role is to stabilize MAPK. In this scenario, the unidentified factor is the direct target of (de)ubiquitination, whereas MAPK is destabilized through ubiquitin-independent means. The unidentified factor may be degraded by the proteasome following its ubiquitination. MAPK might be degraded through a process that does not implicate the proteasome. Alternatively, even though it is not directly ubiquitinated, MAPK might be degraded by the proteasome through ubiquitin-independent degradation. One possibility in this latter case would be that the unidentified factor acts as a chaperone, bringing MAPK to the proteasome. (A,B) Arrow colors are used to indicate positive (green) or negative (red) regulatory impact on MAPK.</p

    <i>Ubc6</i>, <i>poe</i>, and <i>Ufd4</i> dsRNAs alleviate <i>Usp47</i>’s impact on MAPK levels.

    No full text
    <p>(A) Selected hits from the <i>Usp47</i> co-depletion RNAi screen (qPCR validation in <a href="http://www.plosbiology.org/article/info:doi/10.1371/journal.pbio.1002539#pbio.1002539.s016" target="_blank">S5 Table</a>). After validation with independent dsRNA reagents, our set of candidates included one E2 conjugating enzyme, <i>Ubc6</i>, and two E3 ligases, <i>poe</i> and <i>Ufd4</i>. The set also included proteasome components such as <i>Rpn2</i> (FBgn0028692) and ubiquitin genes such as <i>Ubi-p5E</i> (FBgn0086558). <i>Ufd1-like</i>, a proteasome-associated factor also linked to endoplasmic-reticulum-associated protein degradation (ERAD) was also present in our set (see <a href="http://www.plosbiology.org/article/info:doi/10.1371/journal.pbio.1002539#pbio.1002539.s013" target="_blank">S2 Table</a> for full validation screen data). Data shown here is from the RNAi validation experiment. All MAPK levels are normalized to <i>GFP</i> dsRNA controls. The numerical data presented in this panel can be found in <a href="http://www.plosbiology.org/article/info:doi/10.1371/journal.pbio.1002539#pbio.1002539.s013" target="_blank">S2 Table</a>. (B) Schematic representations of the UBC6, UFD4, and POE protein products drawn to scale with the position of identifiable domains and regions (UBCc: ubiquitin-conjugating enzyme E2 catalytic domain; ANK: ankyrin repeat motif; HECT: E3 ligase domain [HECT stands for “homologous to the E6-AP carboxyl terminus”]; UBR: ubiquitin protein ligase E3 component n-recognin domain [also known as UBR box motif]; CRD: cysteine rich domain). Amino acid lengths are also shown. (C) Western blot experiments confirm that <i>Ubc6</i>, <i>poe</i> and <i>Ufd4</i> depletion rescue endogenous MAPK levels in <i>Usp47</i> depleted cells. The rescue mediated by <i>Ufd4</i> depletion is weaker, possibly due to comparatively moderate depletion efficiency (see <a href="http://www.plosbiology.org/article/info:doi/10.1371/journal.pbio.1002539#pbio.1002539.s016" target="_blank">S5 Table</a>). (D) Co-depletion experiments conducted in third instar <i>Drosophila</i> wing discs show that <i>poe</i> and <i>Ubc6</i> can rescue <i>Usp47</i> depletion in vivo. The <i>engrailed-gal4</i> driver was used to drive expression of RNAi in the posterior segment (GFP-positive) of the disc. <i>Ubc6</i> RNAi expression caused extensive larval lethality. Those wing discs that could be recovered were of reduced size (cell lethality was also problematic in confirming <i>Ubc6</i> depletion by qPCR (see <a href="http://www.plosbiology.org/article/info:doi/10.1371/journal.pbio.1002539#pbio.1002539.s016" target="_blank">S5 Table</a>). The RNAi lines used in this experiment correspond to the following VDRC lines: <i>Usp47</i>, GD26027; <i>poe</i> (1), KK108296; <i>poe</i> (2), GD17648; <i>Ubc6</i>, GD23229. The depletion of the <i>Usp47</i>, <i>poe</i>, and <i>Ubc6</i> transcripts were also measured by qPCR (see <a href="http://www.plosbiology.org/article/info:doi/10.1371/journal.pbio.1002539#pbio.1002539.s016" target="_blank">S5 Table</a>). (E) The extra wing vein material induced by <i>mapk</i><sup><i>Sem</i></sup> expression under the control of <i>sal</i><sup><i>EPv</i></sup>-<i>Gal4</i> is suppressed by <i>Usp47</i> RNAi (see <a href="http://www.plosbiology.org/article/info:doi/10.1371/journal.pbio.1002539#pbio.1002539.g001" target="_blank">Fig 1D</a>). Co-expression of <i>poe</i> dsRNA counteracts <i>Usp47</i> RNAi and restores the extra wing vein phenotype. The RNAi lines used here are the same as in C. (F) Pre-translational downregulation of mapk expression is not rescued by <i>poe</i> RNAi. Knocking down the EJC component <i>eIF4AIII</i> reduces MAPK levels due to altered splicing of the <i>mapk</i> transcript. This, unlike the depletion produced by <i>Usp47</i> dsRNA, is not rescued by co-depleting POE. (G) Exogenous MAPK levels measured by immunoblot are rescued by co-depletion of <i>Usp47</i> with <i>poe</i> or <i>Ubc6</i>. An <i>HA-mapk</i> stable cell line was treated with the indicated dsRNA for 4 d. A rescue effect was observed on both endogenous and exogenous MAPK. (C, F, and G) Densitometry quantifications are provided for MAPK levels (normalized to the AKT loading controls). All experiments were performed in triplicate or more, and <i>p</i>-values were calculated (paired two tailed Student’s <i>t</i> test) comparing values to the <i>Rluc</i> control (*: <i>p</i> < 0.05; **: <i>p</i> < 0.01). <i>t</i> tests performed on other samples are shown in red. “ns” denotes not significant. Numerical data for (A) can be found in <a href="http://www.plosbiology.org/article/info:doi/10.1371/journal.pbio.1002539#pbio.1002539.s013" target="_blank">S2 Table</a>. Raw data for (C, F, and G) can be found in <a href="http://www.plosbiology.org/article/info:doi/10.1371/journal.pbio.1002539#pbio.1002539.s001" target="_blank">S1 Data</a>.</p
    corecore