15 research outputs found

    The Escherichia coli RhaS Transcriptional Activator: Transcriptional Activation by the DNA-Binding Domain, The Interdomain Effector Response, and Negative Autoregulation

    Get PDF
    The chapters herein are the accepted manuscript versions of articles that were published independently in scholarly research journals. They have been combined and submitted in fulfillment of the thesis requirement for a Master of Arts degree in Microbiology from the University of Kansas Department of Molecular Biosciences. In addition to the work presented here, my graduate work included two additional projects: a high-throughput screen to identify inhibitors of the Escherichia coli RhaS protein, and a site-directed mutagenesis screen to better understand the molecular mechanisms of the L-rhamnose response in RhaS. The high-throughput screen identified a compound that inhibits DNA binding by RhaS, the related E. coli RhaR protein and the virulence activators Rns from enterotoxigenic E. coli and VirF from Shigella flexneri, but by neither E. coli LacI nor CRP. It appears that this compound may have broad, specific inhibitory activity against AraC-family proteins, making it a candidate for development into an antimicrobial drug that functions by blocking the expression of certain bacterial virulence factors that require an AraC-family activator for expression. The compound likely binds in a pocket between the two helix-turn- helix motifs of the conserved AraC-family DNA-binding domain, thereby sterically prohibiting the protein from binding DNA. In order to better understand the molecular mechanism by which the L-rhamnose signal is transmitted through RhaS from the N-terminal effector-binding domain to the C-terminal DNA-binding domain to regulate DNA binding in response to effector, I constructed a library of several dozen site-directed RhaS mutants. The goal of this work was to identify amino acids key to interdomain signaling by identifying point mutants with phenotypes consistent with defects in signaling. I focused my mutagenesis on regions of the protein predicted to be important in signaling, based on molecular modeling and similarities with related proteins. I isolated mutants in the DNA-binding domain with nearly wild-type activity (-)L-rhamnose and reduced activity (+)L-rhamnose, consistent with a decreased ability to stimulate activity (+)L-rhamnose, at positions Asn174 and Leu175. We conclude that these two residues are likely important in the signal transduction pathway; future work will identify the region of the N-terminal domain involved in this interaction

    Identification of a Small Molecule Inhibitor of Bacterial AraC Family Activators

    Get PDF
    Protein members of the AraC family of bacterial transcriptional activators have great promise as targets for the development of novel antibacterial agents. Here, we describe an in vivo high throughput screen to identify inhibitors of the AraC family activator protein RhaS. The screen used two E. coli reporter fusions; one to identify potential RhaS inhibitors, and a second to eliminate non-specific inhibitors from consideration. One compound with excellent selectivity, OSSL_051168, was chosen for further study. OSSL_051168 inhibited in vivo transcription activation by the RhaS DNA-binding domain to the same extent as the full-length protein, indicating that this domain was the target of its inhibition. Growth curves showed that OSSL_051168 did not impact bacterial cell growth at the concentrations used in this study. In vitro DNA binding assays with purified protein suggest that OSSL_051168 inhibits DNA binding by RhaS. In addition, we found that it inhibits DNA binding by a second AraC family protein, RhaR, which shares 30% amino acid identity with RhaS. OSSL_051168 did not have a significant impact on DNA binding by the non-AraC family proteins CRP and LacI, suggesting that the inhibition is likely specific for RhaS, RhaR, and possibly additional AraC family activator proteins

    Small-Molecule Inhibitor of the Shigella flexneri Master Virulence Regulator VirF

    Get PDF
    This is the publisher's version, also available electronically from http://iai.asm.org/content/81/11/4220VirF is an AraC family transcriptional activator that is required for the expression of virulence genes associated with invasion and cell-to-cell spread by Shigella flexneri, including multiple components of the type three secretion system (T3SS) machinery and effectors. We tested a small-molecule compound, SE-1 (formerly designated OSSL_051168), which we had identified as an effective inhibitor of the AraC family proteins RhaS and RhaR, for its ability to inhibit VirF. Cell-based reporter gene assays with Escherichia coli and Shigella, as well as in vitro DNA binding assays with purified VirF, demonstrated that SE-1 inhibited DNA binding and transcription activation (likely by blocking DNA binding) by VirF. Analysis of mRNA levels using real-time quantitative reverse transcription-PCR (qRT-PCR) further demonstrated that SE-1 reduced the expression of the VirF-dependent virulence genes icsA, virB, icsB, and ipaB in Shigella. We also performed eukaryotic cell invasion assays and found that SE-1 reduced invasion by Shigella. The effect of SE-1 on invasion required preincubation of Shigella with SE-1, in agreement with the hypothesis that SE-1 inhibited the expression of VirF-activated genes required for the formation of the T3SS apparatus and invasion. We found that the same concentrations of SE-1 had no detectable effects on the growth or metabolism of the bacterial cells or the eukaryotic host cells, respectively, indicating that the inhibition of invasion was not due to general toxicity. Overall, SE-1 appears to inhibit transcription activation by VirF, exhibits selectivity toward AraC family proteins, and has the potential to be developed into a novel antibacterial agent

    Marching towards Making Evolution-Proof Antibiotics

    No full text

    The AraC/XylS Family Activator RhaS Negatively Autoregulates rhaSR Expression by Preventing Cyclic AMP Receptor Protein Activation▿

    No full text
    The Escherichia coli RhaR protein activates expression of the rhaSR operon in the presence of its effector, l-rhamnose. The resulting RhaS protein (plus l-rhamnose) activates expression of the l-rhamnose catabolic and transport operons, rhaBAD and rhaT, respectively. Here, we further investigated our previous finding that rhaS deletion resulted in a threefold increase in rhaSR promoter activity, suggesting RhaS negative autoregulation of rhaSR. We found that RhaS autoregulation required the cyclic AMP receptor protein (CRP) binding site at rhaSR and that RhaS was able to bind to the RhaR binding site at rhaSR. In contrast to the expected repression, we found that in the absence of both RhaR and the CRP binding site at the rhaSR promoter, RhaS activated expression to a level comparable with RhaR activation of the same promoter. However, when the promoter included the RhaR and CRP binding sites, the level of activation by RhaS and CRP was much lower than that by RhaR and CRP, suggesting that CRP could not fully coactivate with RhaS. Taken together, our results indicate that RhaS negative autoregulation involves RhaS competition with RhaR for binding to the RhaR binding site at rhaSR. Although RhaS and RhaR activate rhaSR transcription to similar levels, CRP cannot effectively coactivate with RhaS. Therefore, once RhaS reaches a relatively high protein concentration, presumably sufficient to saturate the RhaS-activated promoters, there will be a decrease in rhaSR transcription. We propose a model in which differential DNA bending by RhaS and RhaR may be the basis for the difference in CRP coactivation

    Transcription Activation by the DNA-Binding Domain of the AraC Family Protein RhaS in the Absence of Its Effector-Binding Domain▿

    No full text
    The Escherichia coli l-rhamnose-responsive transcription activators RhaS and RhaR both consist of two domains, a C-terminal DNA-binding domain and an N-terminal dimerization domain. Both function as dimers and only activate transcription in the presence of l-rhamnose. Here, we examined the ability of the DNA-binding domains of RhaS (RhaS-CTD) and RhaR (RhaR-CTD) to bind to DNA and activate transcription. RhaS-CTD and RhaR-CTD were both shown by DNase I footprinting to be capable of binding specifically to the appropriate DNA sites. In vivo as well as in vitro transcription assays showed that RhaS-CTD could activate transcription to high levels, whereas RhaR-CTD was capable of only very low levels of transcription activation. As expected, RhaS-CTD did not require the presence of l-rhamnose to activate transcription. The upstream half-site at rhaBAD and the downstream half-site at rhaT were found to be the strongest of the known RhaS half-sites, and a new putative RhaS half-site with comparable strength to known sites was identified. Given that cyclic AMP receptor protein (CRP), the second activator required for full rhaBAD expression, cannot activate rhaBAD expression in a ΔrhaS strain, it was of interest to test whether CRP could activate transcription in combination with RhaS-CTD. We found that RhaS-CTD allowed significant activation by CRP, both in vivo and in vitro, although full-length RhaS allowed somewhat greater CRP activation. We conclude that RhaS-CTD contains all of the determinants necessary for transcription activation by RhaS
    corecore