594 research outputs found

    Differential regulation of Ota and Otb, two primary glycine betaine transporters in the methanogenic archaeon Methanosarcina mazei go1

    Get PDF
    Methanogenic archaea accumulate glycine betaine in response to hypersalinity, but the regulation of proteins involved, their mechanism of activation and regulation of the corresponding genes are largely unknown. Methanosarcina mazei differs from most other methanoarchaea in having two gene clusters both encoding a potential glycine betaine transporter, Ota and Otb. Western blot as well as quantitative real-time PCR revealed that Otb is not regulated by osmolarity. On the other hand, cellular levels of Ota increased with increasing salt concentrations. A maximum was reached at 300-500 m M NaCl. Ota concentrations reached a maximum 4 h after an osmotic upshock. Hyperosmolarity also caused an increase in cellular Ota concentrations. In addition to osmolarity Ota expression was regulated by the growth phase. Expression of Ota as well as transport of betaine was downregulated in the presence of glycine betaine. Copyright (c) 2007 S. Karger AG, Basel

    Iterative design and optimization of initially inactive Proteolysis Targeting Chimeras (PROTACs) identify VZ185 as a potent, fast and selective von Hippel-Lindau (VHL)-based dual degrader probe of BRD9 and BRD7

    Get PDF
    Developing PROTACs to redirect the ubiquitination activity of E3 ligases and potently degrade a target protein within cells can be a lengthy and unpredictable process, and it remains unclear whether any combination of E3 and target might be productive for degradation. We describe a probe-quality degrader for a ligase-target pair deemed unsuitable: the von Hippel-Lindau (VHL) and BRD9, a bromodomain-containing subunit of the SWI/SNF chromatin remodeling complex BAF. VHL-based degraders could be optimized from suboptimal compounds in two rounds by systematically varying conjugation patterns and linkers, and monitoring cellular degradation activities, kinetic profiles, and ubiquitination, as well as ternary complex formation thermodynamics. The emerged structure-activity relationships guided the discovery of VZ185, a potent, fast and selective degrader of BRD9 and of its close homolog BRD7. Our findings qualify a new chemical tool for BRD7/9 knockdown, and provide a roadmap for PROTAC development against seemingly incompatible target-ligase combinations

    Low postseroconversion CD4 count and rapid decrease of CD4 density identify HIV+ fast progressors

    Full text link
    CD4 expression in HIV replication is paradoxical: HIV entry requires high cell-surface CD4 densities, but replication requires CD4 down-modulation. However, is CD4 density in HIV+ patients affected over time? Do changes in CD4 density correlate with disease progression? Here, we examined the role of CD4 density for HIV disease progression by longitudinally quantifying CD4 densities on CD4+ T cells and monocytes of ART-naive HIV+ patients with different disease progression rates. This was a retrospective study. We defined three groups of HIV+ patients by their rate of CD4+ T cell loss, calculated by the time between infection and reaching a CD4 level of 200 cells/microl: fast (12 years). Mathematical modeling permitted us to determine the maximum CD4+ T cell count after HIV seroconversion (defined as "postseroconversion CD4 count") and longitudinal profiles of CD4 count and density. CD4 densities were quantified on CD4+ T cells and monocytes from these patients and from healthy individuals by flow cytometry. Fast progressors had significantly lower postseroconversion CD4 counts than other progressors. CD4 density on T cells was lower in HIV+ patients than in healthy individuals and decreased more rapidly in fast than in slow progressors. Antiretroviral therapy (ART) did not normalize CD4 density. Thus, postseroconversion CD4 counts define individual HIV disease progression rates that may help to identify patients who might benefit most from early ART. Early discrimination of slow and fast progressors suggests that critical events during primary infection define long-term outcome. A more rapid CD4 density decrease in fast progressors might contribute to progressive functional impairments of the immune response in advanced HIV infection. The lack of an effect of ART on CD4 density implies a persistent dysfunctional immune response by uncontrolled HIV infection

    Two proteolytic pathways regulate DNA repair by cotargeting the Mgt1 alkylguanine transferase

    Get PDF
    O^6-methylguanine (O^6meG) and related modifications of guanine in double-stranded DNA are functionally severe lesions that can be produced by many alkylating agents, including N-methyl-N′-nitro-N-nitrosoguanidine (MNNG), a potent carcinogen. O^6meG is repaired through its demethylation by the O^6-alkylguanine-DNA alkyltransferase (AGT). This protein is called Mgmt (or MGMT) in mammals and Mgt1 in the yeast Saccharomyces cerevisiae. AGT proteins remove methyl and other alkyl groups from an alkylated O^6 in guanine by transferring the adduct to an active-site cysteine residue. The resulting S-alkyl-Cys of AGT is not restored back to Cys, so repair proteins of this kind can act only once. We report here that S. cerevisiae Mgt1 is cotargeted for degradation, through a degron near its N terminus, by 2 ubiquitin-mediated proteolytic systems, the Ubr1/Rad6-dependent N-end rule pathway and the Ufd4/Ubc4-dependent ubiquitin fusion degradation (UFD) pathway. The cotargeting of Mgt1 by these pathways is synergistic, in that it increases not only the yield of polyubiquitylated Mgt1, but also the processivity of polyubiquitylation. The N-end rule and UFD pathways comediate both the constitutive and MNNG-accelerated degradation of Mgt1. Yeast cells lacking the Ubr1 and Ufd4 ubiquitin ligases were hyperresistant to MNNG but hypersensitive to the toxicity of overexpressed Mgt1. We consider ramifications of this discovery for the control of DNA repair and mechanisms of substrate targeting by the ubiquitin system

    The SARS-Coronavirus-Host Interactome: Identification of Cyclophilins as Target for Pan-Coronavirus Inhibitors

    Get PDF
    Coronaviruses (CoVs) are important human and animal pathogens that induce fatal respiratory, gastrointestinal and neurological disease. The outbreak of the severe acute respiratory syndrome (SARS) in 2002/2003 has demonstrated human vulnerability to (Coronavirus) CoV epidemics. Neither vaccines nor therapeutics are available against human and animal CoVs. Knowledge of host cell proteins that take part in pivotal virus-host interactions could define broad-spectrum antiviral targets. In this study, we used a systems biology approach employing a genome-wide yeast-two hybrid interaction screen to identify immunopilins (PPIA, PPIB, PPIH, PPIG, FKBP1A, FKBP1B) as interaction partners of the CoV non-structural protein 1 (Nsp1). These molecules modulate the Calcineurin/NFAT pathway that plays an important role in immune cell activation. Overexpression of NSP1 and infection with live SARS-CoV strongly increased signalling through the Calcineurin/NFAT pathway and enhanced the induction of interleukin 2, compatible with late-stage immunopathogenicity and long-term cytokine dysregulation as observed in severe SARS cases. Conversely, inhibition of cyclophilins by cyclosporine A (CspA) blocked the replication of CoVs of all genera, including SARS-CoV, human CoV-229E and -NL-63, feline CoV, as well as avian infectious bronchitis virus. Non-immunosuppressive derivatives of CspA might serve as broad-range CoV inhibitors applicable against emerging CoVs as well as ubiquitous pathogens of humans and livestock

    Die hyperosmotische Stressantwort von Escherichia coli-von der Proteomanalyse zu einzelnen Komponenten

    Get PDF
    Ein Habitat von Escherichia coli ist der Gastrointestinaltrakt von Säugetieren, der sich durch anaerobe Bedingungen und eine hohe Osmolalität auszeichnet. E. coli ist aber auch freilebend in Gegenwart von Sauerstoff in der Umwelt bei variierenden Osmolalitäten nachzuweisen. Eine Adaptation an diese ständig wechselnden Umweltbedingungen ist entscheidend für Wachstum und Überleben. In dieser Arbeit wurde der Adaptationsprozess an erhöhte Osmolalitäten durch globale Proteomanalysen untersucht. Zusätzlich wurden verschiedene Aspekte des Prozesses im Detail analysiert, um weitere regulatorische Komponenten aufzudecken. Es wurden globale Proteomveränderungen im pI-Bereich 4-7 nach osmotischem Stress unter aeroben Bedingungen zeitabhängig visualisiert. Es konnte eine verstärkte Produktion von 12 Proteinen nachgewiesen werden. 11 zusätzliche Proteine akkumulierten in Zellen, die einem osmotischen Stress ausgesetzt waren, der durch Zugabe des Salzes NaCl ausgelöst wurde. Der Großteil der durch Massenspektrometrie identifizierten Proteine waren Proteine mit allgemeiner Schutzfunktion, die auf transkriptioneller Ebene vom globalen Stressregulator RpoS reguliert werden. Der Vergleich von aeroben und anaeroben Bedingungen ergab eine Überlappung der akkumulierten Proteine von 50 %. Durch ergänzende Proteomanalysen mit alternativen Gelsystemen konnten zwei weitere Proteine identifiziert werden, die an der Osmostressantwort beteiligt sind. Die Zugabe des kompatiblen Soluts Glycinbetain resultierte in einer verminderten Akkumulation von 9 RpoS-regulierten Proteinen bei Salzstress unter aeroben Bedingungen. Für mindestens zwei Proteine konnte eine gegenläufige Regulation nachgewiesen werden. Unter anaeroben Bedingungen verminderte Glycinbetain die Akkumulation eines Proteins (ProX) nach Zugabe von NaCl. Es wurden Proteomanalysen einer K+-Aufnahmemutante im Vergleich zum Wildtyp bei hyperosmotischem Stress erstellt, um den Einfluss der erhöhten intrazellulären K+-Konzentration auf die nachfolgende Stressantwort zu untersuchen. Es konnte gezeigt werden, dass die Regulation von zwei Proteinen (ProX und TnaA) von der K+-Akkumulation abhängig ist. Das Regulationsmuster weiterer Proteine, insbesondere metabolischer Enzyme, war durch die fehlende Akkumulation von K+ in der Mutante beeinflusst. Es wurde eine Methode entwickelt, um Veränderungen der Proteininteraktionen direkt nach Salzstress aufzuzeigen. Durch Fixierung der Zellen mit Formaldehyd und anschließender Fraktionierung der Proteine konnten umfassende Veränderungen im Interaktionsmuster periplasmatischer Proteine nachgewiesen werden. Eine Bildung von Sauerstoffradikalen bei hyperosmotischem Stress konnte unter Verwendung eines Fluoreszenzfarbstoffes erstmalig in E. coli nachgewiesen werden. Die Inhibierung der Radikalbildung durch Inkubation mit Natriumascorbat führte zu einer verminderten Überlebenswahrscheinlichkeit der Zellen bei sehr hohen NaCl-Konzentrationen. Zellen, die in Gegenwart von Natriumascorbat einem Salzstress ausgesetzt waren, wiesen verminderte Mengen bestimmter Osmostress-involvierter Proteine auf. Für E. coli Stämme, denen Sauerstoffradikal-abbauende Enzyme wie Katalase und Superoxiddismutase fehlten, wurde eine erhöhte Salzstressresistenz gezeigt. Die phänotypische Analyse einer hdhA Mutante ergab verminderte Wachstumsraten bei erhöhten Osmolalitäten. Die Mutante war im Vergleich zum Wildtyp durch reduzierte Biofilmbildung und Beweglichkeit sowie Veränderungen im Proteom nach hyperosmotischem Stress gekennzeichnet. Die osmotisch induzierte, cytoplasmatische Trehalase TreF reguliert die intrazelluläre Trehalosekonzentration bei Salzstress unter aeroben Bedingungen. Unter anaeroben Bedingungen konnten keine Unterschiede in den Trehalosekonzentrationen in einer treF-Mutante im Vergleich zum Wildtyp beobachtet werden. Die Zugabe des kompatiblen Solutes Glycinbetain führte unabhängig von der Sauerstoffverfügbarkeit zur verstärkten Produktion von TreF

    BAF complex vulnerabilities in cancer demonstrated via structure-based PROTAC design

    Get PDF
    Targeting subunits of BAF/PBAF chromatin remodeling complexes has been proposed as an approach to exploit cancer vulnerabilities. Here, we develop proteolysis targeting chimera (PROTAC) degraders of the BAF ATPase subunits SMARCA2 and SMARCA4 using a bromodomain ligand and recruitment of the E3 ubiquitin ligase VHL. High-resolution ternary complex crystal structures and biophysical investigation guided rational and efficient optimization toward ACBI1, a potent and cooperative degrader of SMARCA2, SMARCA4 and PBRM1. ACBI1 induced anti-proliferative effects and cell death caused by SMARCA2 depletion in SMARCA4 mutant cancer cells, and in acute myeloid leukemia cells dependent on SMARCA4 ATPase activity. These findings exemplify a successful biophysics- and structure-based PROTAC design approach to degrade high profile drug targets, and pave the way toward new therapeutics for the treatment of tumors sensitive to the loss of BAF complex ATPases.</p

    A systematic analysis of host factors reveals a Med23-interferon-λ regulatory axis against herpes simplex virus type 1 replication

    Get PDF
    Herpes simplex virus type 1 (HSV-1) is a neurotropic virus causing vesicular oral or genital skin lesions, meningitis and other diseases particularly harmful in immunocompromised individuals. To comprehensively investigate the complex interaction between HSV-1 and its host we combined two genome-scale screens for host factors (HFs) involved in virus replication. A yeast two-hybrid screen for protein interactions and a RNA interference (RNAi) screen with a druggable genome small interfering RNA (siRNA) library confirmed existing and identified novel HFs which functionally influence HSV-1 infection. Bioinformatic analyses found the 358 HFs were enriched for several pathways and multi-protein complexes. Of particular interest was the identification of Med23 as a strongly anti-viral component of the largely pro-viral Mediator complex, which links specific transcription factors to RNA polymerase II. The anti-viral effect of Med23 on HSV-1 replication was confirmed in gain-of-function gene overexpression experiments, and this inhibitory effect was specific to HSV-1, as a range of other viruses including Vaccinia virus and Semliki Forest virus were unaffected by Med23 depletion. We found Med23 significantly upregulated expression of the type III interferon family (IFN-λ) at the mRNA and protein level by directly interacting with the transcription factor IRF7. The synergistic effect of Med23 and IRF7 on IFN-λ induction suggests this is the major transcription factor for IFN-λ expression. Genotypic analysis of patients suffering recurrent orofacial HSV-1 outbreaks, previously shown to be deficient in IFN-λ secretion, found a significant correlation with a single nucleotide polymorphism in the IFN-λ3 (IL28b) promoter strongly linked to Hepatitis C disease and treatment outcome. This paper describes a link between Med23 and IFN-λ, provides evidence for the crucial role of IFN-λ in HSV-1 immune control, and highlights the power of integrative genome-scale approaches to identify HFs critical for disease progression and outcome
    corecore