Show simple item record

Files in this item

Thumbnail

Item metadata

dc.contributor.authorLeung, J.M.
dc.contributor.authorTran, F.
dc.contributor.authorPathak, R.B.
dc.contributor.authorPoupart, S.
dc.contributor.authorHeaslip, A.T.
dc.contributor.authorBallif, B.A.
dc.contributor.authorWestwood, N.J.
dc.contributor.authorWard, G.E.
dc.date.accessioned2014-07-16T14:01:05Z
dc.date.available2014-07-16T14:01:05Z
dc.date.issued2014-06-03
dc.identifier.citationLeung , J M , Tran , F , Pathak , R B , Poupart , S , Heaslip , A T , Ballif , B A , Westwood , N J & Ward , G E 2014 , ' Identification of T. gondii myosin light chain-1 as a direct target of TachypleginA-2, a small-molecule inhibitor of parasite motility and invasion ' , PLoS One , vol. 9 , no. 6 , e98056 . https://doi.org/10.1371/journal.pone.0098056en
dc.identifier.issn1932-6203
dc.identifier.otherPURE: 132228444
dc.identifier.otherPURE UUID: 272a2e33-78ad-4230-834a-875d8154a0fc
dc.identifier.otherScopus: 84902504034
dc.identifier.otherORCID: /0000-0003-0630-0138/work/56424183
dc.identifier.otherWOS: 000336911400031
dc.identifier.urihttps://hdl.handle.net/10023/5025
dc.descriptionThis work was supported by US Public Health Service grant AI054961 (GEW/NJW), a University Research Fellowship from the Royal Society (NJW) and funding for the mass spectrometry analysis was provided by the Vermont Genetics Network/NIH Grant 8P20GM103449 from the INBRE program of the NIGMS.en
dc.description.abstractMotility of the protozoan parasite Toxoplasma gondii plays an important role in the parasite's life cycle and virulence within animal and human hosts. Motility is driven by a myosin motor complex that is highly conserved across the Phylum Apicomplexa. Two key components of this complex are the class XIV unconventional myosin, TgMyoA, and its associated light chain, TgMLC1. We previously showed that treatment of parasites with a small-molecule inhibitor of T. gondii invasion and motility, tachypleginA, induces an electrophoretic mobility shift of TgMLC1 that is associated with decreased myosin motor activity. However, the direct target(s) of tachypleginA and the molecular basis of the compound-induced TgMLC1 modification were unknown. We show here by ''click'' chemistry labelling that TgMLC1 is a direct and covalent target of an alkyne-derivatized analogue of tachypleginA. We also show that this analogue can covalently bind to model thiol substrates. The electrophoretic mobility shift induced by another structural analogue, tachypleginA-2, was associated with the formation of a 225.118 Da adduct on S57 and/or C58, and treatment with deuterated tachypleginA-2 confirmed that the adduct was derived from the compound itself. Recombinant TgMLC1 containing a C58S mutation (but not S57A) was refractory to click labelling and no longer exhibited a mobility shift in response to compound treatment, identifying C58 as the site of compound binding on TgMLC1. Finally, a knock-in parasite line expressing the C58S mutation showed decreased sensitivity to compound treatment in a quantitative 3D motility assay. These data strongly support a model in which tachypleginA and its analogues inhibit the motility of T. gondii by binding directly and covalently to C58 of TgMLC1, thereby causing a decrease in the activity of the parasite's myosin motor. 
dc.format.extent13
dc.language.isoeng
dc.relation.ispartofPLoS Oneen
dc.rights© 2014 Leung et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.en
dc.subjectQH301 Biologyen
dc.subject.lccQH301en
dc.titleIdentification of T. gondii myosin light chain-1 as a direct target of TachypleginA-2, a small-molecule inhibitor of parasite motility and invasionen
dc.typeJournal articleen
dc.description.versionPublisher PDFen
dc.contributor.institutionUniversity of St Andrews. School of Chemistryen
dc.contributor.institutionUniversity of St Andrews. EaSTCHEMen
dc.contributor.institutionUniversity of St Andrews. Biomedical Sciences Research Complexen
dc.identifier.doihttps://doi.org/10.1371/journal.pone.0098056
dc.description.statusPeer revieweden


This item appears in the following Collection(s)

Show simple item record