Kinetic characterization and allosteric inhibition of the Yersinia pestis 1-deoxy-D-xylulose 5-phosphate reductoisomerase (MEP synthase).

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 targe...

Full description

Bibliographic Details
Main Authors: Amanda Haymond, Chinchu Johny, Tyrone Dowdy, Brandon Schweibenz, Karen Villarroel, Richard Young, Clark J Mantooth, Trishal Patel, Jessica Bases, Geraldine San Jose, Emily R Jackson, Cynthia S Dowd, Robin D Couch
Format: Article
Language:English
Published: Public Library of Science (PLoS) 2014-01-01
Series:PLoS ONE
Online Access:http://europepmc.org/articles/PMC4149570?pdf=render
id doaj-dd1996804d5d44a7a770cf35a3430aee
record_format Article
spelling doaj-dd1996804d5d44a7a770cf35a3430aee2020-11-25T00:05:48ZengPublic Library of Science (PLoS)PLoS ONE1932-62032014-01-0198e10624310.1371/journal.pone.0106243Kinetic characterization and allosteric inhibition of the Yersinia pestis 1-deoxy-D-xylulose 5-phosphate reductoisomerase (MEP synthase).Amanda HaymondChinchu JohnyTyrone DowdyBrandon SchweibenzKaren VillarroelRichard YoungClark J MantoothTrishal PatelJessica BasesGeraldine San JoseEmily R JacksonCynthia S DowdRobin D CouchThe 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 Yersinia pestis. Enzyme assays indicate apparent kinetic constants of KMDXP = 252 µM and KMNADPH = 13 µM, IC50 values for fosmidomycin and FR900098 of 710 nM and 231 nM respectively, and Ki values for fosmidomycin and FR900098 of 251 nM and 101 nM respectively. To ascertain if the Y. pestis 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.http://europepmc.org/articles/PMC4149570?pdf=render
collection DOAJ
language English
format Article
sources DOAJ
author Amanda Haymond
Chinchu Johny
Tyrone Dowdy
Brandon Schweibenz
Karen Villarroel
Richard Young
Clark J Mantooth
Trishal Patel
Jessica Bases
Geraldine San Jose
Emily R Jackson
Cynthia S Dowd
Robin D Couch
spellingShingle Amanda Haymond
Chinchu Johny
Tyrone Dowdy
Brandon Schweibenz
Karen Villarroel
Richard Young
Clark J Mantooth
Trishal Patel
Jessica Bases
Geraldine San Jose
Emily R Jackson
Cynthia S Dowd
Robin D Couch
Kinetic characterization and allosteric inhibition of the Yersinia pestis 1-deoxy-D-xylulose 5-phosphate reductoisomerase (MEP synthase).
PLoS ONE
author_facet Amanda Haymond
Chinchu Johny
Tyrone Dowdy
Brandon Schweibenz
Karen Villarroel
Richard Young
Clark J Mantooth
Trishal Patel
Jessica Bases
Geraldine San Jose
Emily R Jackson
Cynthia S Dowd
Robin D Couch
author_sort Amanda Haymond
title Kinetic characterization and allosteric inhibition of the Yersinia pestis 1-deoxy-D-xylulose 5-phosphate reductoisomerase (MEP synthase).
title_short Kinetic characterization and allosteric inhibition of the Yersinia pestis 1-deoxy-D-xylulose 5-phosphate reductoisomerase (MEP synthase).
title_full Kinetic characterization and allosteric inhibition of the Yersinia pestis 1-deoxy-D-xylulose 5-phosphate reductoisomerase (MEP synthase).
title_fullStr Kinetic characterization and allosteric inhibition of the Yersinia pestis 1-deoxy-D-xylulose 5-phosphate reductoisomerase (MEP synthase).
title_full_unstemmed Kinetic characterization and allosteric inhibition of the Yersinia pestis 1-deoxy-D-xylulose 5-phosphate reductoisomerase (MEP synthase).
title_sort kinetic characterization and allosteric inhibition of the yersinia pestis 1-deoxy-d-xylulose 5-phosphate reductoisomerase (mep synthase).
publisher Public Library of Science (PLoS)
series PLoS ONE
issn 1932-6203
publishDate 2014-01-01
description 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 Yersinia pestis. Enzyme assays indicate apparent kinetic constants of KMDXP = 252 µM and KMNADPH = 13 µM, IC50 values for fosmidomycin and FR900098 of 710 nM and 231 nM respectively, and Ki values for fosmidomycin and FR900098 of 251 nM and 101 nM respectively. To ascertain if the Y. pestis 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.
url http://europepmc.org/articles/PMC4149570?pdf=render
work_keys_str_mv AT amandahaymond kineticcharacterizationandallostericinhibitionoftheyersiniapestis1deoxydxylulose5phosphatereductoisomerasemepsynthase
AT chinchujohny kineticcharacterizationandallostericinhibitionoftheyersiniapestis1deoxydxylulose5phosphatereductoisomerasemepsynthase
AT tyronedowdy kineticcharacterizationandallostericinhibitionoftheyersiniapestis1deoxydxylulose5phosphatereductoisomerasemepsynthase
AT brandonschweibenz kineticcharacterizationandallostericinhibitionoftheyersiniapestis1deoxydxylulose5phosphatereductoisomerasemepsynthase
AT karenvillarroel kineticcharacterizationandallostericinhibitionoftheyersiniapestis1deoxydxylulose5phosphatereductoisomerasemepsynthase
AT richardyoung kineticcharacterizationandallostericinhibitionoftheyersiniapestis1deoxydxylulose5phosphatereductoisomerasemepsynthase
AT clarkjmantooth kineticcharacterizationandallostericinhibitionoftheyersiniapestis1deoxydxylulose5phosphatereductoisomerasemepsynthase
AT trishalpatel kineticcharacterizationandallostericinhibitionoftheyersiniapestis1deoxydxylulose5phosphatereductoisomerasemepsynthase
AT jessicabases kineticcharacterizationandallostericinhibitionoftheyersiniapestis1deoxydxylulose5phosphatereductoisomerasemepsynthase
AT geraldinesanjose kineticcharacterizationandallostericinhibitionoftheyersiniapestis1deoxydxylulose5phosphatereductoisomerasemepsynthase
AT emilyrjackson kineticcharacterizationandallostericinhibitionoftheyersiniapestis1deoxydxylulose5phosphatereductoisomerasemepsynthase
AT cynthiasdowd kineticcharacterizationandallostericinhibitionoftheyersiniapestis1deoxydxylulose5phosphatereductoisomerasemepsynthase
AT robindcouch kineticcharacterizationandallostericinhibitionoftheyersiniapestis1deoxydxylulose5phosphatereductoisomerasemepsynthase
_version_ 1725423379168624640