11 research outputs found

    Control of replication initiation by the Sum1/Rfm1/Hst1 histone deacetylase

    Get PDF
    <p>Abstract</p> <p>Background</p> <p>Replication initiation at origins of replication in the yeast genome takes place on chromatin as a template, raising the question how histone modifications, for instance histone acetylation, influence origin firing. Initiation requires binding of the replication initiator, the Origin Recognition Complex (ORC), to a consensus sequence within origins. In addition, other proteins bind to recognition sites in the vicinity of ORC and support initiation. In previous work, we identified Sum1 as an origin-binding protein that contributes to efficient replication initiation. Sum1 is part of the Sum1/Rfm1/Hst1 complex that represses meiotic genes during vegetative growth via histone deacetylation by the histone deacetylase (HDAC) Hst1.</p> <p>Results</p> <p>In this study, we investigated how Sum1 affected replication initiation. We found that it functioned in initiation as a component of the Sum1/Rfm1/Hst1 complex, implying a role for histone deacetylation in origin activity. We identified several origins in the yeast genome whose activity depended on both Sum1 and Hst1. Importantly, <it>sum1</it>Δ or <it>hst1</it>Δ caused a significant increase in histone H4 lysine 5 (H4 K5) acetylation levels, but not other H4 acetylation sites, at those origins. Furthermore, mutation of lysines to glutamines in the H4 tail, which imitates the constantly acetylated state, resulted in a reduction of origin activity comparable to that in the absence of Hst1, showing that deacetylation of H4 was important for full initiation capacity of these origins.</p> <p>Conclusion</p> <p>Taken together, our results demonstrate a role for histone deacetylation in origin activity and reveal a novel aspect of origin regulation by chromatin. These results suggest recruitment of the Sum1/Rfm1/Hst1 complex to a number of yeast origins, where Hst1 deacetylated H4 K5.</p

    A role for Sum1 in HML silencing and replication initiation in Saccharomyces cerevisiae

    Get PDF
    In der eukaryotischen Ontogenese ist die Etablierung differenzierungsspezifischer Genexpression eng an die Unterteilung des Genoms in funktionell getrennte Domänen gekoppelt. Solche Domänen lassen entweder erhöhte transkriptionelle Aktivität zu oder unterdrücken sie und werden Eu- bzw. Heterochromatin genannt. Heterochromatin enthält spezielle Proteine, die zur Ausbildung dieser repressiven Chromatinstruktur beitragen. Eine der Hauptfragen in der Heterochromatinbiologie ist, wie solche Proteine rekrutiert werden. Dieser Prozess ist entscheidend damit einzelnde Regionen im Genom koordiniert zeit- und ortsabhängig reprimiert werden können. In Saccharomyces cerevisiae entsteht Hetero-chromatin an den silent-mating-type Loci HMRa und HMLalpha durch die zielgerichtete Rekrutierung des Sir-Komplexes über eine Gruppe von Proteinen, die an sogenannte silencer-DNA Sequenzen binden. In diese Arbeit wird gezeigt, daß das Protein Sum1, bisher bekannt als Repressor meiotischer Gene im vegetativen Zellzyklus, als Heterochromatin-Rekrutierungsfaktor für HMLalpha fungiert. Sum1 konnte in vitro und in vivo an HMLalpha über ein funktionelles Element innerhalb des HML-E silencers binden und die Deletion von SUM1 verursachte einen Verlust von Repression an HMLalpha. SUM1 beeinflußte außerdem die Fähigkeit von HML-E als Replikationsstartpunkt (origin) zu agieren, was eine Rolle von Sum1 in der Replikation nahelegt. Die Beobachtung, daß orc2-1 und orc5-1 mit sum1Delta synthetisch lethal waren und daß cdc6-1, cdc7-1 oder cdc45-1 mit sum1Delta einen synthetischen Wachstumsdefekt aufwiesen unterstützt die Vermutung, daß SUM1 eine globale Rolle in der Replikationsinitiation besitzt. In einer genomweiten Suche wurden ARS Elemente gefunden, die sowohl Sum1 als auch ORC rekrutieren. Dabei konnte gezeigt werden, daß die Replikationsaktivität dieser ARS Elemente von Sum1 bzw. Sum1 Bindungsstellen abhängig war. Als Repressor von meiosespezifischen Genen interagiert Sum1 oft mit der Histondeacetylase Hst1. In diesem Zusammenhang konnte gezeigt werden, daß SUM1-regulierte origins ebenfalls HST1 zur vollen Aktivität benötigten. Zusammenfassend schlagen wir Sum1 als neuartigen Modulator für die Replikationsinitiation an einer Untergruppe chromosomaler Replikationsstartpunkte vor.The division of eukaryotic chromatin into functionally distinct domains is critical to implement gene expression programs that drive the development of multicellular organisms. Regions termed euchromatin exist in the genome that are generally conducive to transcription, whereas heterochromatin contains specialized chromatin binding proteins that repress transcription in these regions. A central question in heterochromatin biology is how the heterochromatin factors are targeted to specific genomic regions, a process that is crucial to ensure that the designated domains, and only they, are repressed in the appropriate spatial and temporal fashion. In Saccharomyces cerevisiae heterochromatinization at the silent mating-type loci HMRa and HMLalpha is achieved by targeting the Sir complex to these regions via a set of anchor proteins that bind to the silencers. Here, we have identified a novel heterochromatin targeting factor for HMLalpha, the protein Sum1, a repressor of meiotic genes during vegetative growth. Sum1 bound both in vitro and in vivo to HMLalpha via a functional element within the HML-E silencer, and deletion of SUM1 caused HMLalpha derepression. Significantly SUM1 was also required for origin activity of HML-E, suggesting a role of Sum1 in replication initiation. Our observations of a synthetic lethality between orc2-1 or orc5-1 and sum1Delta as well as a synthetic growth defect of cdc6-1, cdc7-1 and cdc45-1 with sum1Delta support the notion that SUM1 has a global role in replication initiation. In a genome-wide search for Sum1-regulated origins, we identified a set of autonomous replicative sequences (ARS elements) that bound both the origin recognition complex and Sum1. Full initiation activity of these origins required Sum1, and their origin activity was decreased upon removal of the Sum1 binding site. In its role as a repressor of meiosis specific genes, Sum1 often works in concert with the histone deacetylase Hst1. We found that SUM1-regulated origins also required HST1 for full activity. Taken together we propose that Sum1 is a novel replication initiation modulator for a subset of chromosomal origins

    Control of replication initiation and heterochromatin formation in Saccharomyces cerevisiae by a regulator of meiotic gene expression

    No full text
    Heterochromatinization at the silent mating-type loci HMR and HML in Saccharomyces cerevisiae is achieved by targeting the Sir complex to these regions via a set of anchor proteins that bind to the silencers. Here, we have identified a novel heterochromatin-targeting factor for HML, the protein Sum1, a repressor of meiotic genes during vegetative growth. Sum1 bound both in vitro and in vivo to HML via a functional element within the HML-E silencer, and sum1Δ caused HML derepression. Significantly, Sum1 was also required for origin activity of HML-E, demonstrating a role of Sum1 in replication initiation. In a genome-wide search for Sum1-regulated origins, we identified a set of autonomous replicative sequences (ARS elements) that bound both the origin recognition complex and Sum1. Full initiation activity of these origins required Sum1, and their origin activity was decreased upon removal of the Sum1-binding site. Thus, Sum1 constitutes a novel global regulator of replication initiation in yeast

    Discovery and Characterization of the Potent and Highly Selective 1,7-Naphthyridine-Based Inhibitors BAY-091 and BAY-297 of the Kinase PIP4K2A

    No full text
    PIP4K2A is an insufficiently studied type II lipid kinase that catalyzes the conversion of phosphatidylinositol-5-phosphate (PI5P) into phosphatidylinositol 4,5-bisphosphate (PI4,5P2). The involvement of PIP4K2A/B in cancer has been suggested, particularly in the context of p53 mutant/null tumors. PIP4K2A/B depletion has been shown to induce tumor growth inhibition, possibly due to hyperactivation of AKT and reactive oxygen species-mediated apoptosis. Herein, we report the identification of the novel potent and highly selective inhibitors BAY-091 and BAY-297 of the kinase PIP4K2A by high-throughput screening and subsequent structure-based optimization. Cellular target engagement of BAY-091 and BAY-297 was demonstrated using cellular thermal shift assay technology. However, inhibition of PIP4K2A with BAY-091 or BAY-297 did not translate into the hypothesized mode of action and antiproliferative activity in p53-deficient tumor cells. Therefore, BAY-091 and BAY-297 serve as valuable chemical probes to study PIP4K2A signaling and its involvement in pathophysiological conditions such as cancer

    Discovery of BAY-985, a Highly Selective TBK1/IKK epsilon Inhibitor

    Full text link
    The serine/threonine kinase TBK1 (TANK-binding kinase 1) and its homologue IKKϵ are noncanonical members of the inhibitor of the nuclear factor κB (IκB) kinase family. These kinases play important roles in multiple cellular pathways and, in particular, in inflammation. Herein, we describe our investigations on a family of benzimidazoles and the identification of the potent and highly selective TBK1/IKKϵ inhibitor BAY-985. BAY-985 inhibits the cellular phosphorylation of interferon regulatory factor 3 and displays antiproliferative efficacy in the melanoma cell line SK-MEL-2 but showed only weak antitumor activity in the SK-MEL-2 human melanoma xenograft model

    Targeting the aryl hydrocarbon receptor (AhR) with BAY 2416964: a selective small molecule inhibitor for cancer immunotherapy

    No full text
    Background The metabolism of tryptophan to kynurenines (KYN) by indoleamine-2,3-dioxygenase or tryptophan-2,3-dioxygenase is a key pathway of constitutive and adaptive tumor immune resistance. The immunosuppressive effects of KYN in the tumor microenvironment are predominantly mediated by the aryl hydrocarbon receptor (AhR), a cytosolic transcription factor that broadly suppresses immune cell function. Inhibition of AhR thus offers an antitumor therapy opportunity via restoration of immune system functions.Methods The expression of AhR was evaluated in tissue microarrays of head and neck squamous cell carcinoma (HNSCC), non-small cell lung cancer (NSCLC) and colorectal cancer (CRC). A structure class of inhibitors that block AhR activation by exogenous and endogenous ligands was identified, and further optimized, using a cellular screening cascade. The antagonistic properties of the selected AhR inhibitor candidate BAY 2416964 were determined using transactivation assays. Nuclear translocation, target engagement and the effect of BAY 2416964 on agonist-induced AhR activation were assessed in human and mouse cancer cells. The immunostimulatory properties on gene and cytokine expression were examined in human immune cell subsets. The in vivo efficacy of BAY 2416964 was tested in the syngeneic ovalbumin-expressing B16F10 melanoma model in mice. Coculture of human H1299 NSCLC cells, primary peripheral blood mononuclear cells and fibroblasts mimicking the human stromal-tumor microenvironment was used to assess the effects of AhR inhibition on human immune cells. Furthermore, tumor spheroids cocultured with tumor antigen-specific MART-1 T cells were used to study the antigen-specific cytotoxic T cell responses. The data were analyzed statistically using linear models.Results AhR expression was observed in tumor cells and tumor-infiltrating immune cells in HNSCC, NSCLC and CRC. BAY 2416964 potently and selectively inhibited AhR activation induced by either exogenous or endogenous AhR ligands. In vitro, BAY 2416964 restored immune cell function in human and mouse cells, and furthermore enhanced antigen-specific cytotoxic T cell responses and killing of tumor spheroids. In vivo, oral application with BAY 2416964 was well tolerated, induced a proinflammatory tumor microenvironment, and demonstrated antitumor efficacy in a syngeneic cancer model in mice.Conclusions These findings identify AhR inhibition as a novel therapeutic approach to overcome immune resistance in various types of cancers

    Novel Class of Potent and Cellularly Active Inhibitors Devalidates MTH1 as Broad-Spectrum Cancer Target

    No full text
    MTH1 is a hydrolase responsible for sanitization of oxidized purine nucleoside triphosphates to prevent their incorporation into replicating DNA. Early tool compounds published in the literature inhibited the enzymatic activity of MTH1 and subsequently induced cancer cell death; however recent studies have questioned the reported link between these two events. Therefore, it is important to validate MTH1 as a cancer dependency with high quality chemical probes. Here, we present BAY-707, a substrate-competitive, highly potent and selective inhibitor of MTH1, chemically distinct compared to those previously published. Despite superior cellular target engagement and pharmacokinetic properties, inhibition of MTH1 with BAY-707 resulted in a clear lack of <i>in vitro</i> or <i>in vivo</i> anticancer efficacy either in mono- or in combination therapies. Therefore, we conclude that MTH1 is dispensable for cancer cell survival
    corecore