16 research outputs found

    Kinetic Characterization and Allosteric Inhibition of the <i>Yersinia pestis</i> 1-Deoxy-D-Xylulose 5-Phosphate Reductoisomerase (MEP Synthase)

    No full text
    <div><p>The methylerythritol phosphate (MEP) pathway found in many bacteria governs the synthesis of isoprenoids, which are crucial lipid precursors for vital cell components such as ubiquinone. Because mammals synthesize isoprenoids via an alternate pathway, the bacterial MEP pathway is an attractive target for novel antibiotic development, necessitated by emerging antibiotic resistance as well as biodefense concerns. The first committed step in the MEP pathway is the reduction and isomerization of 1-deoxy-D-xylulose-5-phosphate (DXP) to methylerythritol phosphate (MEP), catalyzed by MEP synthase. To facilitate drug development, we cloned, expressed, purified, and characterized MEP synthase from <i>Yersinia pestis</i>. Enzyme assays indicate apparent kinetic constants of K<sub>M</sub><sup>DXP</sup> = 252 µM and K<sub>M</sub><sup>NADPH</sup> = 13 µM, IC<sub>50</sub> values for fosmidomycin and FR900098 of 710 nM and 231 nM respectively, and K<sub>i</sub> values for fosmidomycin and FR900098 of 251 nM and 101 nM respectively. To ascertain if the <i>Y. pestis</i> MEP synthase was amenable to a high-throughput screening campaign, the Z-factor was determined (0.9) then the purified enzyme was screened against a pilot scale library containing rationally designed fosmidomycin analogs and natural product extracts. Several hit molecules were obtained, most notably a natural product allosteric affector of MEP synthase and a rationally designed bisubstrate derivative of FR900098 (able to associate with both the NADPH and DXP binding sites in MEP synthase). It is particularly noteworthy that allosteric regulation of MEP synthase has not been described previously. Thus, our discovery implicates an alternative site (and new chemical space) for rational drug development.</p></div

    Dose-response plot of the <i>Y. pestis</i> MEP synthase when preincubated with the inhibitor.

    No full text
    <p>Assays were performed by combining the enzyme with either A) compound <b>15</b> or B) compound <b>16</b> and preincubating at 37°C for 10 min before addition of NADPH and DXP. All assays were performed in duplicate. Activity of the enzyme is relative to an uninhibited control.</p

    Cation specificity of <i>Y. pestis</i> MEP synthase.

    No full text
    <p>Enzyme assays were performed with fixed NADPH (150 µM), DXP (400 µM), and divalent cation (25 mM) concentration. <i>Y. pestis</i> MEP synthase has comparable activity with either Mg<sup>2+</sup> or Mn<sup>2+</sup>. Assays were performed in duplicate.</p

    Purification of recombinant <i>Y. pestis</i> MEP synthase.

    No full text
    <p>A Coomassie stained SDS-PAGE shows two lanes of purified His-tagged MEP synthase alongside a molecular weight marker (MW). His-tagged MEP synthase has a predicted molecular weight of 46.7 kDa. The typical yield of purified protein averaged 30 mg per 1 L shake flask.</p

    The substrate dependent catalytic activity of <i>Y. pestis</i> MEP synthase.

    No full text
    <p>Shown are the Michaelis-Menten plots of reaction velocity as a function of A) DXP concentration and B) NADPH concentration. Least-squares best fit of the data to the Michaelis-Menten equation produces the kinetic parameters listed in <a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0106243#pone-0106243-t001" target="_blank">Table 1</a>. The R<sup>2</sup> value for each plot is indicated. All assays were performed in duplicate.</p

    The MVA and MEP biosynthetic pathways.

    No full text
    <p>A) The MVA pathway is utilized by humans and other eukaryotes, archaebacteria, and certain eubacteria to produce IPP and DMAPP, the building blocks of isoprenoids. The pathway is initiated by the enzymatic condensation of 3 molecules of acetyl-CoA (1) to form 3-hydroxy-3-methylglutaryl CoA (HMG-CoA) (3), which is then reduced to MVA by HMG-CoA reductase (4) <a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0106243#pone.0106243-M1" target="_blank">[54]</a><a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0106243#pone.0106243-Miziorko1" target="_blank">[55]</a> Subsequent phosphorylation and decarboxylation yield IPP (7) <a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0106243#pone.0106243-Amdur1" target="_blank">[56]</a><a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0106243#pone.0106243-BLOCH1" target="_blank">[57]</a><a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0106243#pone.0106243-DhePaganon1" target="_blank">[58]</a> which is converted to DMAPP (8) by an isomerase <a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0106243#pone.0106243-Agranoff1" target="_blank">[59]</a>. B) The MEP pathway is used by higher plants, the plastids of algae, apicomplexan protozoa, and many eubacteria, including numerous human pathogens. Pyruvate (9) is condensed with glyceraldehyde 3-phosphate (10) to yield 1-deoxy-D-xylulose 5-phosphate (DXP; (11)) <a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0106243#pone.0106243-Lange1" target="_blank">[60]</a>, a branch point intermediate with a role in <i>E. coli</i> vitamin B1 and B6 biosynthesis <a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0106243#pone.0106243-Julliard1" target="_blank">[61]</a><a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0106243#pone.0106243-Julliard2" target="_blank">[62]</a><a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0106243#pone.0106243-Hill1" target="_blank">[63]</a><a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0106243#pone.0106243-Hill2" target="_blank">[64]</a> as well as isoprene biosynthesis. In the first committed step of the <i>E. coli</i> MEP pathway, 1-Deoxy-D-xylulose 5-phosphate reductoisomerase (also called MEP synthase, Dxr or IspC) catalyzes the reduction and rearrangement of 11 to yield MEP (12) <a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0106243#pone.0106243-AndrewTKoppisch1" target="_blank">[28]</a>. CDP-ME synthase then converts MEP into 4-(cytidine 5′-diphospho)-2-C-methyl-D-erythritol (CDP-ME; (13)). CDP-ME kinase phosphorylates CDP-ME, which is subsequently cyclized (coupled with the loss of CMP) by cMEPP synthase to yield 2-C-methyl-D-erythritol 2,4-cyclodiphosphate (15) <a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0106243#pone.0106243-Rohdich2" target="_blank">[65]</a><a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0106243#pone.0106243-Kuzuyama2" target="_blank">[66]</a><a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0106243#pone.0106243-Kuzuyama3" target="_blank">[67]</a><a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0106243#pone.0106243-Lttgen1" target="_blank">[68]</a><a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0106243#pone.0106243-Herz1" target="_blank">[69]</a>. A reductive ring opening of 15 produces 1-hydroxy-2-methyl-2-butenyl diphosphate (HMBPP; (16)) <a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0106243#pone.0106243-Altincicek2" target="_blank">[70]</a><a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0106243#pone.0106243-Campos1" target="_blank">[71]</a><a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0106243#pone.0106243-Hecht1" target="_blank">[72]</a><a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0106243#pone.0106243-Kollas1" target="_blank">[73]</a><a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0106243#pone.0106243-Rohdich3" target="_blank">[74]</a>, which is then reduced to both IPP and DMAPP in a ∼5:1 ratio <a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0106243#pone.0106243-Rohdich1" target="_blank">[8]</a><a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0106243#pone.0106243-Altincicek3" target="_blank">[75]</a><a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0106243#pone.0106243-Cunningham1" target="_blank">[76]</a><a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0106243#pone.0106243-Altincicek4" target="_blank">[77]</a><a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0106243#pone.0106243-McAteer1" target="_blank">[78]</a><a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0106243#pone.0106243-Rohdich4" target="_blank">[79]</a><a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0106243#pone.0106243-Adam1" target="_blank">[80]</a>. C) The reaction catalyzed by MEP synthase. The intermediate 2-C-methyl-D-erythrose 4-phosphate (18), produced by isomerization via cleavage of the bond between C3 and C4 and formation of a new bond between C2 and C4 <a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0106243#pone.0106243-Arigoni1" target="_blank">[81]</a><a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0106243#pone.0106243-Putra1" target="_blank">[82]</a>, is subsequently reduced to yield MEP (12).</p

    Mechanism of inhibition by e29.

    No full text
    <p>A) Relative to NADPH, e29 is an uncompetitive inhibitor of the purified <i>Y. pestis</i> MEP synthase. B) Relative to DXP, e29 is a noncompetitive inhibitor. C) A model of e29 inhibition. MEP synthase (E) undergoes a conformational change (E*) upon binding of NADPH (N), exposing an allosteric site to which the inhibitor (I) binds. As the inhibitor is noncompetitive with respect to DXP (D), I may bind the E*N or E*ND complex, thereby inhibiting the enzyme.</p

    MEP synthase inhibitors.

    No full text
    <p>The structures of fosmidomycin, FR900098, and select rationally designed amide-linked and O-linked inhibitors are shown, including lipophilic prodrug analogs of compound <b>15</b>.</p

    Growth inhibition assay with liquid cultures of <i>Y. pestis</i>.

    No full text
    <p><i>Y. pestis</i> A1122 was cultured in the presence of either 100 µM or 500 µM of the indicated inhibitor. Bacterial growth is relative to an uninhibited culture. All assays were performed in triplicate. At 500 µM, compounds <b>15</b>, <b>16</b>, <b>51</b>, and <b>52</b> have inhibitory activity comparable to FR900098, however relatively poor inhibitory activity is observed at 100 µM. See text for further discussion.</p
    corecore