ebook img

Piotrowski JS, Okada H, Lu F, Li SC, Hinchman L, Ranjan A, Smith DL, Higbee AJ PDF

14 Pages·2015·2.29 MB·English
by  
Save to my drive
Quick download
Download
Most books are stored in the elastic cloud where traffic is expensive. For this reason, we have a limit on daily download.

Preview Piotrowski JS, Okada H, Lu F, Li SC, Hinchman L, Ranjan A, Smith DL, Higbee AJ

Plant-derived antifungal agent poacic acid β targets -1,3-glucan JeffS.Piotrowskia,1,2,HirokiOkadab,1,FachuangLua,SheenaC.Lic,LiHinchmana,AshishRanjand,DamonL.Smithd, AlanJ.Higbeee,ArneUlbriche,JoshuaJ.Coone,RaameshDeshpandef,YuryV.Bukhmana,SeanMcIlwaina, IreneM.Onga,ChadL.Myersf,CharlesBoonec,g,RobertLandicka,JohnRalpha,MehdiKabbaged,andYoshikazuOhyab,2 aGreatLakesBioenergyResearchCenter,UniversityofWisconsin–Madison,Madison,WI53703;DepartmentsofdPlantPathologyandeChemistry,University ofWisconsin–Madison,Madison,WI53706;bDepartmentofIntegratedBiosciences,GraduateSchoolofFrontierSciences,UniversityofTokyo,Kashiwa, Chiba,Japan277-8561;cRIKENCenterforSustainableResourceScience,Wako,Saitama,Japan351-0198;fDepartmentofComputerScience andEngineering,UniversityofMinnesota–TwinCities,Minneapolis,MN55455;andgTerrenceDonnellyCentreforCellularandBiomolecularResearch, UniversityofToronto,Toronto,ON,CanadaM5S3E1 EditedbyDitervonWettstein,WashingtonStateUniversity,Pullman,WA,andapprovedFebruary11,2015(receivedforreviewJune13,2014) Ariseinresistancetocurrentantifungalsnecessitatesstrategiesto but new compounds continue to be discovered (15). One under- identifyalternativesourcesofeffectivefungicides.Wereportthe studied class of compounds found in grasses and their hydro- discoveryofpoacicacid,apotentantifungalcompoundfoundin lysates is the dehydrodiferulates and compounds derived from lignocellulosic hydrolysates of grasses. Chemical genomics using them (hereafter all simply termed diferulates) (16, 17). In Saccharomyces cerevisiae showed that loss of cell wall synthesis grasses, diferulates are produced by radical dimerization of fer- and maintenance genes conferred increased sensitivity to poacic ulatesthatacylatearabinoxylanpolysaccharidesandfunctionas acid.Morphologicalanalysisrevealedthatcellstreatedwithpoacic powerfulcellwallcross-linkers(16);derivativesofdiferulatesare acidbehavedsimilarlytocellstreatedwithothercellwall-target- releasedduringthehydrolysisofbiomass(16,18,19).Atpresent, ing drugs and mutants with deletions in genes involved in pro- thestructuresofarangeofdiferulateshavebeendescribed(16, cessesrelatedtocellwallbiogenesis.Poacicacidcausesrapidcell 18), but the biological activities of isolated diferulates (beyond lysisandissynergisticwithcaspofunginandfluconazole.Thecel- their function in the plant cell wall) have not been explored. lulartargetwasidentified;poacicacidlocalizedtothecellwalland Diferulatesmaybeexpectedtohaveeffectsonorganismsother inhibitedβ-1,3-glucansynthesisinvivoandinvitro,apparentlyby than plants. One study found a negative correlation between directly binding β-1,3-glucan. Through its activity on the glucan diferulateconcentrationandcolonizationbycorn-boringinsects layer,poacicacidinhibitsgrowthofthefungiSclerotiniasclerotiorum (20), but a direct effect of diferulates is unknown. Despite the andAlternariasolaniaswellastheoomycetePhytophthorasojae.A well-documented antifungal activity of ferulic acid and its singleapplicationofpoacicacidtoleavesinfectedwiththebroad- derivatives(13,21,22),nostudiesontheantifungalpropertiesof range fungal pathogen S. sclerotiorum substantially reduced le- diferulates havebeendescribed. siondevelopment.Thediscoveryofpoacicacidasanaturalanti- Wescreenedacollectionofdiferulatesfoundinlignocellulosic fungal agent targeting β-1,3-glucan highlights the potential side hydrolysatesforantifungalactivityusingtheyeastSaccharomyces use of products generated in the processing of renewable bio- cerevisiaeasadiscoverysystemforantifungalagents.Wefocused mass toward biofuels as a source of valuable bioactive com- on the diferulate 8–5-DC (16) derived during hydrolysis from pounds and further clarifies the nature and mechanism of a major diferulate in grasses; we name this compound here as fermentationinhibitorsfoundinlignocellulosichydrolysates. | | Significance chemicalgenomics high-dimensionalmorphometrics | | lignocellulosichydrolysates fungalcellwall Saccharomycescerevisiae Thesearchfornewantifungalcompoundsisstrugglingtokeep Lignocellulosics are a potential sugar feedstock for biofuels pace with emerging fungicide resistance. Through chemo- andbio-basedchemicals.Before plantmaterialscan becon- prospectingofanuntappedreservoirofinhibitorycompounds, vertedtobiofuelsbyfermentation,theircellwallpolysaccharides lignocellulosic hydrolysates, we have identified a previously mustbehydrolyzedtosugarmonomersformicrobialconversion undescribedantifungalagent,poacicacid.Usingbothchemical (1).Thehydrolysisprocessesgenerates,inadditiontothesugars, genomicsandmorphologicalanalysistogetherforthefirsttime, small acids, furans, and other compounds that affect microbial toourknowledge,weidentifiedthecellulartargetofpoacicacid growthand inhibit fermentation (2–5). Theinhibitory effects of asβ-1,3-glucan.Throughitsactionontheglucanlayeroffungal these compounds represent a challenge to efficient microbial cellwalls,poacicacidisanaturalantifungalagentagainsteco- bioconversion. The primary focus of lignocellulosic-derived in- nomicallysignificantfungiandoomyceteplantpathogens.This hibitor research has been to understand, evolve, and engineer work highlights the chemical diversity within lignocellulosic tolerance in fermentative microbes (2). However, as natural hydrolysatesasasourceofpotentiallyvaluablechemicals. antimicrobialagents,lignocellulosicfermentationinhibitorsoffer anuntappedreservoir ofbioactivecompounds. Authorcontributions:J.S.P.,H.O.,A.R.,D.L.S.,R.L.,M.K.,andY.O.designedresearch;J.S.P., H.O.,F.L.,L.H.,A.R.,A.J.H.,A.U.,andM.K.performedresearch;J.S.P.,H.O.,F.L.,S.C.L., Oneincreasinglyimportantpotentialuseoftheseinhibitorsisas D.L.S.,J.J.C.,C.L.M.,C.B.,J.R.,M.K.,andY.O.contributednewreagents/analytictools;J.S.P., antifungal agents. Worldwide, fungicide-resistant pathogens pose H.O.,A.J.H.,R.D.,Y.V.B.,S.M.,I.M.O.,andC.L.M.analyzeddata;andJ.S.P.,H.O.,C.B.,R.L., a threat to agricultural sustainability. Pathogen resistance to con- J.R.,andY.O.wrotethepaper. ventional fungicides affects multiple crops (6, 7). Copper-based Theauthorsdeclarenoconflictofinterest. fungicidesareeffectiveinorganicagriculturebutfacingrestrictions ThisarticleisaPNASDirectSubmission. because of copper accumulation in soils (8, 9). Furthermore, cli- FreelyavailableonlinethroughthePNASopenaccessoption. matechangeisalteringtheglobaldistributionoffungalpathogens 1J.S.P.andH.O.contributedequallytothiswork. (10, 11). New sources of fungicides are a necessity to keep pace 2Towhomcorrespondencemaybeaddressed.Email:[email protected]@ withtheevolutionofresistantstrainsandemergingpathogens(12). k.u-tokyo.ac.jp. Theantifungalactivitiesofmanyoftheinhibitors(e.g.,ferulic Thisarticlecontainssupportinginformationonlineatwww.pnas.org/lookup/suppl/doi:10. acid and furfural) in hydrolysates have been described (13, 14), 1073/pnas.1410400112/-/DCSupplemental. E1490–E1497 | PNAS | PublishedonlineMarch9,2015 www.pnas.org/cgi/doi/10.1073/pnas.1410400112 S U L P S A N poacic acid, because it is found primarily in grasses (Poaceae). A P By applying both chemical genomics and morphological ana- lysis, we predicted and confirmed that poacic acid binds to cell wall β-1,3-glucan. We showed its biological activity against not only yeast but also, the economically important fungal and oomycete pathogens Sclerotinia sclerotiorum, Alternaria solani, and Phytophthora sojae. Results DiferulateswithAntifungalActivity.Wetestedacollectionofnine diferulates known to occur in hydrolysates from corn stover for their effects on S. cerevisiae growth (Fig. 1A and Table S1, de- tailed nomenclature). Of these, only two had detectable bio- activity at the tested concentration of 1 mg/mL. In particular, poacic acid had the greatest antifungal activity, with an IC of 50 111 μg/mL (324 μM) against our control yeast (Fig. 1B). This inhibition is comparable with that of the widely used fungicides picoxystrobin(IC50of308μM)andpolyoxinD(IC50of340μM) B andsubstantiallylowerthanthatoftheprimaryfungicideusedin organicagriculture,coppersulfate(IC of2.4mM) (23–25). 50 ChemicalGenomicsPredictThatPoacicAcidTargetstheFungalCell Wall. To gain insight into the mode of action and the cellular targetofpoacicacid,weconductedchemicalgenomicanalysis,a method that uses genome-wide collections of viable gene- deletion mutants to identify genes with deletions that confer sensitivity or resistance to bioactive compounds (26, 27). The resulting set of sensitive and resistant gene-deletion mutants associatedwitharesponseyieldsfunctionalinsightintothemode ofaction(26).Wechallengedapooledmixtureof∼4,000different Fig.1. Bioactivityofdiferulates.Thebioactivityofninediferulatesagainst yeast gene-deletion mutants with either poacic acid or a solvent S.cerevisiaeat1mg/mLwastested.Poacicacid(8–5-DC)had(A)thehighest ceonnabtrloeld(uDsMtoSdOe)c.ipSheeqruthenecrienlgatiovfesftitrnaeinss-sopfeceiaficchDyeNasAtmbuatracnotdeins bioactivityand(B)anIC50of111μg/mL(mean±SE). LOGY O thepresenceofthedrugrelativetothesolventcontrol(28). BI O Deletion mutants for genes involved in cell wall and glyco- (Fig. 2C). Additionally, some other cell wall-related gene mu- R C sylation-related processes were present in the top significantly tants were resistant to poacic acid. A deletion mutant of NBP2 MI sensitive and resistant strains (Fig. 2A, blue circles and Table (Nap1 binding protein) was resistant to poacic acid (Fig. 2A); S2).Amongthetop10deletionmutantssensitivetopoacicacid, Nbp2p down-regulates cell wall biogenesis through an interac- wedetectedenrichmentforgenesinvolvedinthegeneontology tionwithBck1p,whichisactivatedbyPkc1p(Fig.2B)(31).Thus, (GO) category fungal-type cell wall organization (P < 0.01). it seems that defects in the PKC pathway confer sensitivity to These mutants included deletion alleles of BCK1 (bypass of C poacic acid, whereas activation of the PKC pathway confers re- kinase), which encodes an MAPKKK in the Pkc1p (protein ki- sistance.AdeletionmutantofDFG5(defectiveforfilamentous nase C) cell wall integrity signaling pathway (PKC pathway); growth)alsowasresistanttopoacicacid;DFG5encodesaGPI- CWH43,whichencodesamembraneproteininvolvedincellwall anchored protein involved in cell wall biogenesis that also has biogenesisanditsnullmutationissyntheticallylethalwithPKC1 agenetic interaction withBck1p(32,33). mutants(29);ROM2(Rho1multicopysuppressor),aGDP/GTP Because the chemical inhibitor of a gene product tends to exchangefactorforRho1pandanothercomponentofthePKC mimictheloss-of-functionphenotypeofamutantthatinactivates pathway;andACK1,whichseemstoencodeanupstreamactivator thegene,thechemical–genomicprofileforabioactivecompound ofPkc1pandhasaphysicalinteractionwithRom2p.Overall,the can resemble the genetic interaction profile for the target (34). PKC pathway was the most sensitive pathway to poacic acid PKC1 has a genetic interaction profile that is most significantly (Pathwayzscore=−7.85)(Fig.2B).Thisprofileissimilartothe correlatedtothechemicalgenomicprofileofpoacicacid(Fig.2B) chemicalgenomicprofilesofotheragentsthattargetthecellwall (P < 0.0001). PKC1 is an essential gene required for growth andrelatedprocesses(e.g.,caspofungin)(26).Deletionmutantsof and response to cell wall stress, and it has been implicated as BCK1arehypersensitivetoagentsthatcompromiseglycosylation a key mediator of cell wall-targeting drugs, such as the echi- (tunicamycin) and cell wall β-1,3-glucan biosynthesis (caspo- nocandins (35). Together, these data narrowed our target fungin)(26).Weconfirmedthesensitivityoftheindividualbck1Δ searchtothefungalcellwall.Poacicacidcoulddirectlydamage mutant and found a sixfold reduction in the IC against poacic thecellwall,inhibitakeycellwallsynthesisenzyme,ordisrupt 50 acidcomparedwiththecontrolstrain(WT)(Fig.2C). the PKC pathway. Among the top significantly resistant strains, we detected significantenrichmentfordeletionsofgenesinvolvedintheGO MorphologicalAnalysisRevealedThatPoacicAcidAffectstheFungal categoryglycosphingolipidbiosyntheticprocess(P<0.01)driven Cell Wall. We investigated the morphological changes induced by csg2Δ (calcium sensitive growth) and sur1Δ (suppressor of by poacic acid using high-dimensional microscopy (36, 37). Re- Rvs161andrvs167mutations)(Fig.2A,yellowcirclesandTable cently, two morphological features (a wide neck and morpho- S2). Deletion of glycosphingolipid genes has been shown to ac- logical heterogeneity) were reported as common phenotypes in tivate the PKC pathway and cell wall biogenesis (Fig. 2B) (30). cells treated with agents known to affect the cell wall (38). The Involvement of SUR1 in poacic acid sensitivity was confirmed morphologies of cells exposed to poacic acid had both features when we isolated a spontaneous chain-termination mutant in (Fig.3AandB);theydisplayeddose-dependentincreaseinbud SUR1abletoformcoloniesonagarwith500μgpoacicacid/mL neck size (Fig. 3C) and heterogeneous morphologies (Fig. 3D). Piotrowskietal. PNAS | PublishedonlineMarch9,2015 | E1491 A B C Fig.2. Chemicalgenomicsofpoacicacid.(A)Treatmentoftheyeastdeletioncollectionwithpoacicacidrevealedthatmutantsinvolvedincellwallbio- synthesisandglycosylationweresensitiveandresistanttothecompound(blue).Mutantsingenesinvolvedinglycosphingolipidbiogenesiswereamongthe mostresistant(yellow).(B)ThePKCpathway,whichgovernscellwallintegritysignaling,wasthemostsensitivepathway(Pathwayzscore=−7.85),withmany membersandinteractinggenesshowingsensitivitytopoacicacid(chemicalgeneticinteractionscoreinparentheses).Comparisonwiththeyeastgeneticin- teractionnetworkindicatedthatthegeneticinteractionprofileoftheessentialgenePKC1wasmostsignificantlycorrelatedtothechemicalgenomicprofileof poacicacid(P<0.001).(C)MutantsofthegeneencodingthecellwallsignalingkinaseBCK1weresixfoldmoresensitivetopoacicacid,whereasapoacicacid- resistantmutant(PAr)withanSNPinSUR1hadincreasedresistancecomparedwiththecontrolstrain(meanandSEbarswereremovedforclarity). Because mutants displaying a high correlation with a drug phe- bypoacicacidistothatinducedbyothercellwall-affectingdrugs, notypecanhelpidentifytargetedprocesses,wenextcomparedthe we compared their morphological profiles. Two echinocandins morphology of poacic acid-treated cells with the individual mor- (micafungin and echinocandin B), both of which bind Fks1p, had phologiesof4,718yeastdeletionmutants(37,39,40).Forty-three morphologicalprofilesthatwerehighlycorrelatedwitheachother, deletion mutants had morphological profiles statistically similar whereas poacic acid-treated cells had lower morphological corre- (P<0.01)tothoseofpoacicacid(Fig.3EandTableS3).Within lationswiththeseandothercellwall-affectingcompounds(Fig.4C). thetopcorrelations,wefoundsignificantenrichmentofgenesin Thus, although there is some morphological similarity with other theGOcategorytransferaseactivity,transferringhexosylgroups cellwallagents,themorphologicalresponsetopoacicacidsuggests (P < 0.001). This GO category contained genes responsible for thatitmayhaveamodeofactionthatisdifferentfromthatofother key processes in the cell wall biogenesis, such as OCH1, which cellwall-targetingagents. encodesamannosyltransferasethatinitiatespolymannoseouter chain elongation, and FKS1 (FK506 sensitivity), which encodes PoacicAcid CausesRapid Cell Leakage.Cell wall-targeting agents, a catalytic subunit of β-1,3-glucan synthase. Taken together, such as echinocandins, can lead to compromised cell integrity thesedatafurtherindicatethatpoacicacidaffectstheyeastcell and ultimately, cell lysis from turgor pressure (45). We inves- wall,consistentwiththe chemicalgenomicanalysis. tigatedwhetherpoacicacidcausedcelllysisinasimilarway.We testedtheextentofcellpermeabilityafter4hoftreatmentwith Poacic Acid Is Synergistic with Drugs That Target the Cell Wall and poacic acid, caspofungin, methyl methanesulfonate (MMS), or MembraneIntegrity.Giventhatpoacicacidmaydirectlytargetthe DMSO using a propidium iodide dye that is taken up only by cellwallortheintegritysignalingpathway,wetestedwhetherthe cells with compromised cell integrity. MMS, an agent that does mode of action of poacic acid differed from that of the echino- not cause rapid cell wall damage, was included as a negative candincaspofungin.Echinocandinsdamagetheyeastcellwallby control.Wefoundthatbothcaspofunginandpoacicacidcaused noncompetitive bindingof theβ-1,3-glucan synthasecomplex at rapidcellleakage,whereasMMSandDMSOdidnot(Fig.4D). the Fks1p subunit (41, 42). Synergistic interactions occur with When growth was arrested by depriving cells of a carbon drugs targeting the same or a functionally related pathway but source,theeffectofcaspofunginwasdiminished,supportingthe through different targets (43). We found significant synergistic known mode of action of echinocandins, which inhibit glucan effects(Fig.4A)betweenpoacicacidandcaspofungin(P<0.05). synthesis.Theeffectsofpoacicacidwerereducedinnonactively Thisinteractionsuggeststhatpoacicacidtargetsthecellwallbut growingcells,butleakagewasstillsignificantlygreater(P<0.05) doessothroughamechanismdistinctfromthatofcaspofungin. thaninallothertreatments(Fig.4D).Themechanismbywhich Becauseechinocandinsarealsosynergisticwithantifungalazoles poacic acid causes leakage is lessened without active growth, that target ergosterol biosynthesis and compromise membrane showingthatthecompoundcanstillcauseleakageinnonactively integrity (44), we tested and determined that poacic acid also growing cells, unlike with echinocandins. This result could in- displayedsignificantsynergywithfluconazole(Fig.4B)(P<0.01). dicate a general disruption of cell wall integrity rather than an enzymatic targetandthus,adifferentmode ofaction. PoacicAcid-InducedMorphologiesAreUniqueComparedwithThose from Other Cell Wall-Targeting Agents. Compounds that induce PoacicAcidLocalizestotheCellSurfaceandTargetsβ-1,3-Glucan.We similar morphological responses can be indicative of similar next sought to determine to which cell wall component poacic modesofaction.Todeterminehowsimilarthemorphologyinduced acidbindsbylocalizingthecompoundintreatedcells.Asaferulate E1492 | www.pnas.org/cgi/doi/10.1073/pnas.1410400112 Piotrowskietal. S U L P S A N A B P C D E Y G O L Fig.3. Morphologicalcharacteristicsofpoacicacid-treatedcells.Poacicacidtreatmentcaused(AandB)abnormalcellmorphologyand(CandD)mor- O BI phologicalcharacteristicssimilartothosecausedbyothercellwall-targetingagents.Poacicacid-treatedcellshad(C)adose-dependentincreasedbudneck O R sizeand(D)heterogeneityofcellmorphology.(E)Thephenotypeofpoacicacid-treatedcellswashighlycorrelatedwiththephenotypesofmutantsingenes C involvedintransferaseactivitytransferringhexosylgroups(P<0.001).TheredlineindicatesanadjustedPvalueof<0.01. MI derivative, poacic acid is fluorescent, enabling us to visualize buds,itdoesnotchangemannoproteinstainingwithfluorescent its accumulation at the cell surface (Fig. 5A). Based on this dye-conjugatedConA(Fig.S1),whichsuggeststhatpoacicacid result together with poacic acid’s chemical genomic profile, acts primarilyonthe formation of theglucan fibrils rather than morphological profile, phenotypic similarity to fks1Δ, and ability byinhibiting mannoprotein assemblyinthecell wall. tocausecellleakage,wehypothesizedthatpoacicacidtargetsthe β-1,3-glucan layer and thus, rapidly compromises cell integrity, PoacicAcidIsanInhibitorofFungalandOomycetePlantPathogens. leadingtocelllysiswhenturgorpressureburststheweakenedcell As a plant-derived natural product, poacic acid may have a po- wall. The absence of chitin-related genes in both the chemical tential use in organic agriculture, which is presently lacking in genomic and morphological profiles and the low correlation be- fungicide diversity beyond copper sulfate mixtures. We initially tween the morphological profile of poacic acid and the chitin testedtheeffectsofpoacicacidonS.sclerotiorum,anascomycete targetingcompoundnikkomycinZledustobelievethatthechitin fungalpathogenwithanextremelybroadhostplantrange(>400 layer is not the cell wall target of poacic acid. Furthermore, species)andworldwidedistribution.Insoybeans,S.sclerotiorum theuniformstainingpatternwithpoacicacidisdifferentfromthe causes Sclerotinia stem rot or white mold of soybean. The in- calcofluorwhitestainingofchitin,whichpreferentiallybindsthe corporationofpoacicacidintoculturemediacausedasignificant budneck andbudscar. (P < 0.01) dose-dependent decrease in fungal growth both on Wesuggestthatthemodeofpoacicacidactionisdistinctfrom agar plates and in liquid cultures, which was evidenced by that of the echinocandins, acting through direct binding to the decreasesincolonyradialgrowthandfungalmass(Fig.6A).We glucan fibrils rather thaninhibition ofglucan synthase. This hy- further investigated whether poacic acid could inhibit lesion de- pothesis is supported by observations that poacic acid does not velopmentinplantaondetachedsoybeanleaves.Solvent(DMSO) localizespecificallytothesiteofbudgrowth,likeFks1p(46),but control or poacic acid (500 μg/mL) solutions were applied to de- rather,bindsacrosstheentirecellsurface(Fig.5A).Poacicacid caninhibitβ-1,3-glucansynthesisinvivoasshownbysignificantly tachedleavesbeforeinoculationwithagarplugscontainingactively decreased glucan staining in buds (Fig. 5B and Fig. S1) and growingmyceliaofS.sclerotiorum.Lesiondevelopmentwasmoni- significantlyless14C-glucoseincorporationintotheβ-1,3-glucan tored daily up to 120 h postinoculation. Poacic acid treatment layer after poacic acid treatment (Fig. 5C). We also observed markedlyreducedlesiondevelopmentoverthistimecoursecom- an in vitro inhibition of β-1,3-glucan synthesis after poacic acid paredwiththecontrol(Fig.6BandC).Wealsofoundpoacicacid treatment (Fig. 5D). By incubating purified yeast glucan with to be similarly effective against the ascomycete A. solani, which poacic acid and observing fluorescence, we found that poacic causesearlyblightintomatoandpotatocrops(Fig.S2).Thesedata aciddirectlybindsβ-1,3-glucan(Fig.5E).Furthermore,although showthatpoacicacidinhibitsfungalgrowthinvitroandinplanta, poacic acid can reduce aniline blue staining of β-1,3-glucan in withpromisingagriculturalapplications. Piotrowskietal. PNAS | PublishedonlineMarch9,2015 | E1493 A B and therefore, represents a previously undescribed compound tar- getingβ-1,3-glucan.Althoughwefoundno effects of poacic acid on mannoprotein assembly, direct binding of glucan fibrils outsidetheplasmamembranemayalsoresultininhibitionofcell wall assemblages, such as the glucan-transglycolase Gas1p and thechitintransglycosylasesCrh1pandUtr2p,thatconnectchitin chainstoglucans,whichrequireglucanasasubstrateorcosubstrate. In nature, diferulates strengthen monocot cell walls by cross- linking polysaccharides (arabinoxylans) to each other and poly- saccharides to lignin—in both cases by radical coupling mecha- nisms (16, 17). It remains unknown if poacic acid has a similar interaction with the fungal cell wall polysaccharides. However, this work raises the question about whether diferulates may C D haveadualroleasantifungalsecondarymetabolitesthatconfer protectiontocertainplants.Otherdiferulatesmayhavethesame potential to bind glucan, but the size/physical properties of poacicacidmaycontribute toitsmoreoptimalbioactivity. Against yeast, the bioactivity of poacic acid is similar to the widelyusedfungicidepicoxystrobin(IC of308μM),lowerthan 50 thiabendazole (IC of 607 μM), and considerably more toxic 50 thancoppersulfate(IC of2.4mM)(25).Poacicacidmayalso 50 have potential to be used combined with agricultural azoles throughitsdocumentedsynergismtoslowthedevelopmentof azole resistance. Although there are conventional fungicides effective at lower doses(e.g., captan at IC of 19μMand pro- 50 chloraz at IC of 132 μM) (25), most conventional agents are 50 Fig. 4. Synergisms and the mode of action of poacic acid. (A) Poacic specific to either the Eumycota or Oomycota, whereas poacic acid(125μg/mL)issignificantlysynergisticwithcaspofungin(12.5ng/mL). acid affects both. Options for organic agriculture are limited to (B)Poacicacid(125μg/mL)isalsosynergisticwithfluconazole(3.8μg/mL). copper-basedfungicides,whicharefacingincreasingrestrictions (C)Morphologicalsimilaritybetweenpoacicacidandothercellwall-affecting because of copper accumulation in soil ecosystems (8, 9). Fur- agentswasmeasuredbasedonthecorrelationcoefficientvalue(R)oftheir morphological profiles. (D) Poacic acid causes cell leakage within 4 h of thermore, as a natural plant-based phenolic acid, poacic acid treatment, similar to the cell wall-targeting compound caspofungin. The would likely be rapidly broken down in the soil and would not leakage is most apparent in actively growingcells [yeast extract peptone accumulate (50). Field trials, application strategies (e.g., seed dextrose(YPD)]comparedwithcellsarrestedwithoutacarbonsource[yeast treatmentvs.foliarspray),andmorediversepathogentestswill extractpeptone(YP)].DMSOandMMSwereincludedascontrolagentsthat be necessary to determine its performance as an agricultural donot directly affect cellwallintegrity. Inarrested cells, poacic acidhad significantlygreatercellleakagethanothertreatments.One-wayANOVA andTukey’stestwereusedtocalculatethedifferencesbetweentreatments (mean±SE).PA,poacicacid. A B C Fungi generally have 30–80% glucan in their cell walls (47); similarly,oomyceteshaveacellwallcontainingβ-1,3-glucanand β-1,6-glucan,butunlikefungi,oomycetewallscontainacellulose layer rather than chitin. Oomycetes are broadly distributed, economicallysignificantpathogens,andanaturalfungicidethat couldaffectbothtruefungiandoomycetes bydisruption ofthe glucan layer could be of high value. We found that poacic acid significantlyreducescolonygrowthoftheoomyceteP.sojae(Fig. D E 6D)(P<0.01),awidespreadpathogenthatcausesrootandstem rot of soybeans. Given its effectiveness against both fungi and oomycetes, poacic acid may have potential as a plant-derived fungicidewithbroadaction. Discussion Through chemoprospecting of lignocellulosic hydrolysates, we have identified a promising antifungal agent. Combining chem- ical genomic and morphological analyses, we determined that poacic acid targets β-1,3-glucan within fungal cell walls. In- hibition of glucan synthesis in vivo and in vitro and cell-wide Fig.5. Poacicacidtargetsβ-1,3-glucan.(A)Poacicacidisfluorescentand localization and direct binding of purified glucan indicate that accumulates on the cell wall. Poacic acid inhibits β-1,3-glucan in vivo as the compound can bind to β-1,3-glucans in the growing glucan shownby(B)thedecreaseinsignalfromanilinebluestaining(arrowheads) fibrilsaswellasthematurewall.ThecellwalldyeCongoredmay and(C)theincorporationof14C-labeledglucoseintotheβ-1,3-glucanlayer ofthecellwall(P<0.05).Concentrationsofpoacicacid,echinocandinB,and also bind growing glucan fibrils (48), but it also binds to chitin, hydroxyureawere250μg/mL,4μg/mL,and30mM,respectively.(D)Poacic and biochemical evidence indicates that the primary target of Congo red is chitin (49). Poacic acid targets the β-1,3-glucan (aEc)idPoinahciibciatsciβd-1d,i3r-egcltulycabninsdysntphuarsifeieadctyiveiatystignluvictarno.wStituhdeannt’IsC5t0teosft3w1aμsgu/mseLd. layeroffungalcellwallsinamannerdistinctfromthatofother to determine significant differences (mean ± SD). DIC, differential inter- cell wall-affecting agents (e.g., caspofungin and nikkomycin Z) ferencecontrast. E1494 | www.pnas.org/cgi/doi/10.1073/pnas.1410400112 Piotrowskietal. S U L P S A N A B Methods P DetailedmethodsareprovidedinSIMethods. Chemical Genomic Analysis. Chemical genomic analysis of poacic acid was performed asdescribed previously (26, 53). Thetested yeast deletion col- lection had ∼4,000 strains using the genetic background described by Andrusiak (54). The optimal inhibitory concentration of poacic acid for chemical genomic profiling [70–80% growth vs. solvent control in yeast extract-peptone-galactosemediumafter24hofgrowth]wasdetermined using an eight-point dose curve. A concentration of 88 μg/mL inhibited C D growthwithinthisrange;200-μLculturesofthepooleddeletioncollection ofS.cerevisiaeweregrownwith88μg/mLpoacicacid(n=3)oraDMSO controlintriplicatefor48hat30°C.GenomicDNAwasextractedusingthe EpicentreMasterPureYeastDNAPurificationKit.Mutant-specificmolecular barcodeswereamplifiedwithspeciallydesignedmultiplexprimers(28).The barcodesweresequencedusinganIlluminaMiSeq.Replicatesofeachcon- dition,poacicacid(n=3)orDMSO(n=2),weresequenced.Thebarcode countsforeachyeastdeletionmutantinthepresenceofpoacicacidwere compared to the DMSO control conditions to determine sensitivity or re- sistanceofindividualstrains(thechemicalgeneticinteractionscore)(26).To determineaPvalueforeachtopsensitiveandresistantmutant,weusedthe EdgeRpackage(55,56).ABonferroni-correctedhypergeometricdistribution testwasusedtosearchforsignificantenrichmentofGOtermsamongthe Fig. 6. Poacic acid inhibits the growth of fungal and oomycete plant top 10 sensitive and resistant deletion mutants (57). To understand the pathogens. (A) Colony growth on plates and mycelia weight of S. scle- pathwaysthatweremostaffectedbypoacicacid,wedevelopedaprotein rotiorum(strain1980)inliquidcultureweresignificantlyinhibitedbypoacic complex/pathwayscorebasedonthesummationofthezscores foreach acidinadose-dependentmanner.(BandC)Asingleaerosoltreatmentof poacicacid(500μg/mL)beforeinoculationinhibitedwhitemoldlesionde- complex/pathway (Pathway z score). Correlation of the chemical genomic profileofpoacicacidwiththeyeastgeneticinteractionnetworkwasper- velopment on soybean leaves in planta. (D) Representative photographs weretaken96hpostinoculation.Poacicacidsignificantlyinhibitedcolony formedaspreviouslydescribed(34). growthofP.sojae(fieldisolate7dofgrowth).DashedcirclesinInsetin- dicatethemyceliumfrontafter2dofgrowth.One-wayANOVAandTukey’s Multivariate Morphological Analysis. Cells of budding yeast S. cerevisiae testwereusedtocalculatethedifferencebetweendrugtreatmentsamong (BY4741his3Δ::KanMX;hereafter,his3Δ)wereculturedin2mL1%Bacto treatments(mean±SE). yeastextract(BDBiosciences),2%Bactopeptone(BDBiosciences),and2% glucose(YPD)with0,25,50,75,100,or125μg/mLpoacicacidoraDMSO Y G controlat25°Cfor16huntiltheearlylogphase.Themaximumconcen- O L fungicideandevaluateitspersistenceintheenvironment,butit trationofthedrug(125μg/mL)wasdeterminedbasedonthegrowthin- BIO may also provide an important and abundant lead compound hibitionrates(10%).Cellfixation,staining,andobservationwereperformed RO thatcouldbemodified forincreasedefficacy. (n=5)asdescribedpreviously(37).Imagesofcellshape,actin,andnuclear MIC Although it was identified from lignocellulosic hydrolysates DNAwereanalyzedusingtheimageprocessingsoftwareCalMorph(version 1.2),whichextractedatotalof501morphologicalquantitativevaluesfrom for bioethanol fermentation, poacic acid alone is not likely to atleast200individualcellsineachexperiment(37).Imageswereprocessed beaprimaryinhibitoraffectingfermentation givenitsrelatively usingPhotoshopCS2(AdobeSystems)forillustrativepurposes. low concentration (0.1 μM) (Table S4). However, it could be To assess the morphological similarity between the cells treated with synergisticwithotherinhibitoryagents(e.g.,furfuralandphenolics) poacicacidandnonessentialdeletionmutantsorcellstreatedwithothercell or other diferulates (e.g., 8–5-C) (2, 16). The combined effects of wall-affecting drugs, their morphological profiles were compared as de- diferulates with other inhibitors could significantly affect lignocel- scribed previously (36). To identify functional gene clusters, the most sig- lulosic biofuel synthesis by fungi, but such combinatorial effects nificant similar mutants (43 genes, P < 0.01 after Bonferroni correction, haveyettobetested. ttest)wereselectedasaqueryforGOtermanalysis(GOtermfinderinthe The complementary profiling methodologies that we applied Saccharomycesgenomedatabase). to the analysis of poacic acid’s effects, including chemical ge- Toextractindependentandcharacteristicfeaturesofmorphologyinduced nomic profiling and morphological profiling, are powerful and bypoacicacid,atwo-stepprincipalcomponentanalysiswasperformedas can provide high-resolution predictions of targeted processes; described previously (39). Tocompare phenotypic noise in the yeast pop- thisworkhighlightsthepowerofthecombinedapproach.Given ulation(poacicacidvs.DMSO),thenoisescorewascalculatedasdescribed previously(58). theincreasingthroughputofbothtechniquesthankstoadvances in next generation sequencing and automated microscopy, the MeasurementofinVivoβ-1,3-GlucanSynthesis.Inhibitionofinvivoβ-1,3-glucan useofbothgeneticandmorphologicalapproachesinlarge-scale synthesis was measured as described previously with slight modification screeningofdruglibrariesmayallowunbiasedwhole-celltarget (n=3)(59).Yeastcells(his3Δ)weregrowninYPDtoearlylogphaseat25°C. identification with less reliance on target-centric high-through- Theculturedcellsweredilutedto1×107cellsper1mLwith1mLof putscreening methods. YPDmediumcontainingone-tenththeglucosecontaining23.125kBq Thisstudywasdesignedtoidentifynovelbioactivecompounds [14C]glucose (ARC0122; American Radiolabeled Chemicals) and test com- from lignocellulosic hydrolysates. Given the goal of cellulosic pounds[250μg/mLforpoacicacid,4μg/mLforechinocandinB,30mMfor ethanol production [60 billion L/y by 2022, requiring 0.6–1.2 hydroxyurea(negativecontrol),or0.4%(vol/vol)DMSOasasolventcontrol]. trillion L hydrolysate/y assuming 5–10% (vol/vol) ethanol before Thecellswereradiolabeledbyculturingat25°Cfor2h.Thelabeledcells distillation] (51, 52), even low-abundance compounds within wereharvestedandincubatedwith1NNaOHat80°Cfor30min.Theinsoluble hydrolysates could be available in significant quantities. We have pellets were resuspended in 10 mM Tris·HCl, pH 7.5, containing 5 mg/mL zymolyase100T(Seikagaku)andincubatedat37°Cfor18h.Afterdigestion, detected monomeric ferulate in hydrolysates at markedly higher thezymolyase-resistantmaterialwasremovedbycentrifugation(15,000×g levels (up to 1.7 mM in alkaline H O -treated corn stover). If 2 2 for15min),andthezymolyase-degradationproduct(mostlyβ-1,3-glucan) synthesizedfromtherecoveredferulatecomponentposthydrolysis, waspurifiedbyultrafiltrationwithacentrifugalfiltermembrane(Amicon poacicacidmayconfergreatervaluetolignocellulosicconversion. Ultra0.5mL;molecularweightcutoffis10,000;Millipore).Theflow-through Our results highlight the chemical diversity within lignocellulosic fractionwasmixedwithscintillationmixture(UltimaGold;PerkinElmer),and hydrolysatesasasourceofpotentiallyvaluablechemicals. radioactivitywasmeasuredbyascintillationcounter(LSC-6100;Aloka).The Piotrowskietal. PNAS | PublishedonlineMarch9,2015 | E1495 differencesintheincorporationratesinsampleswerenormalizedbyΔOD600 was measured. Inhibition of S. sclerotiorum in planta was tested by in- measuredbeforeandafterthelabelingperiod. oculating detached soybean leaves of the commercial variety Williams 82 withanagarplugofactivelygrowingS.sclerotiorummycelia.Leaveswere Measurement of β-1,3-Glucan Synthase Activity in the Membrane Fraction. treatedonetimebeforeinoculationwitheitheranaerosolsprayofwater After the membrane fraction was prepared from S. cerevisiae BY4741 as withDMSO(control)ora500μg/mLsolutionofpoacicacid.Leaveswerein- describedpreviously(n=3)(60),β-1,3-glucansynthaseactivitywasmeasured cubatedinamoistenvironment,andlesiondevelopmentwasmonitoredupto asdescribedpreviously(n=3)(61)withslightmodification.Briefly,20μL 120hpostinoculation.FieldstrainsofP.sojaeandA.solaniweregrownon membranefraction(∼70μgtotalprotein)wasaddedtoareactionmixture cornmealandpotatodextroseagarplates,respectively,atroomtemperature (finalvolumeof100μL)containing50mMTris·HCl,pH7.5,10mMpotassium for 7 and5 d,respectively,beforemeasurement. The growthinhibition of fluoride, 1 mM EDTA, 0.2 mM UDP-Glc (with 89 Bq UDP-[Glucose-14C]; poacicacidwasassessedat0,500,1,000,and1,500μg/mLinreplicateplates NEC403;PerkinElmer),anddifferentconcentrationsofpoacicacid(1.25,2.5, (n=3).Agarplugsfromactivelygrowingcultureswereplacedatthecenterof 5,10,20,40,80,160,or320μg/mL).Thereactionmixturewasincubatedat theplatesandallowedtogrowatroomtemperature.Colonydiameterwas 25°Cfor30minandstoppedbytheadditionofethanol.Totrapreaction monitoredforeachtreatmentinatime-courseexperiment.One-wayANOVA product(β-1,3-glucanpolymer),thereactionmixturewasfilteredthrough andTukey’stestwereusedtocalculatethedifferencesbetweendrugtreat- themembranefilter(mixedcelluloseesters;0.2μminporesize;ADVANTEC), mentsamongtreatments. washedonetimewith2mLdistilledwater,anddriedatroomtemperature. Afteradditionofscintillationmixture(Econofluor-2;PerkinElmer),radioac- ACKNOWLEDGMENTS. We thank T. K. Sato, E. Hendel, S. Morford, and tivitywasmeasuredbyascintillationcounter(LSC-6100;Aloka).Inhibition N. Keller for critical discussions of the manuscript. J.S.P., F.L., L.H., A.J.H., A.U., J.J.C., S.M., I.M.O., and J.R. are funded by Department of Energy curves and IC50 values were determined using R software (ver. 3.0.1) by (DOE)GreatLakesBioenergyResearchCenterDOEBiologicalandEnviron- sigmoidalcurvefittingwiththeglmfunction. mentalResearchOfficeofScienceGrantDE-FC02-07ER64494.J.S.P.,F.L.,and M.K. are supported by Wisconsin Alumni Research Foundation Award Growth Inhibition of Plant Pathogens. To test inhibition in liquid culture, MSN178899.H.O.isaresearchfellowoftheJapanSocietyforthePromotion adosecurveof0,125,250,and500μg/mLpoacicacidin100mLpotato ofScience.S.C.L.issupportedbyaRIKENForeignPostdoctoralFellowship. dextrosebroth(n=3)wasused.CulturesofS.sclerotiorumstrain1980were A.R.,D.L.S.,andM.K.aresupportedbyWisconsinSoybeanMarketingBoard inoculatedwith100μLhomogenizedmyceliaandgrownat25°Cfor48h. GrantMSN172403.R.D.andC.L.M.aresupportedbyNationalInstitutesof HealthGrants1R01HG005084-01A1,1R01GM104975-01,andR01HG005853 Themyceliaintheliquidmediaweredriedandweighed.Thegrowthin- and National Science Foundation Grant DBI 0953881. C.L.M. and C.B. are hibition of poacic acid on solid agar cultures (potato dextrose agar) was supportedbytheCanadianInstituteforAdvancedResearchGeneticNetworks assessedbygeneratingreplicateplates(n=3)containing0,125,250,and Program.M.K.issupportedbyUnitedSoybeanBoardGrantMSN143317.Y.O. 500μg/mLpoacicacid.Plateswereinoculatedwithanactivelygrowingplug issupportedbyMinistryofEducation,Culture,Sports,ScienceandTechnology, ofS.sclerotiorumandgrownat25°C.Themycelialradialgrowthafter48h JapanGrantforScientificResearch24370002. 1. SunY,ChengJ(2002)Hydrolysisoflignocellulosicmaterialsforethanolproduction:A 19.BunzelM,RalphJ,MaritaJM,HatfieldRD,SteinhartH(2001)Diferulatesasstructural review.BioresourTechnol83(1):1–11. componentsinsoluble and insolublecereal dietaryfibre.J Sci FoodAgric 81(7): 2. PiotrowskiJS,etal.(2014)Deathbyathousandcuts:Thechallengesanddiverse 653–660. landscapeoflignocellulosichydrolysateinhibitors.FrontMicrobiol5(2014):90. 20.SantiagoR,etal.(2006)Diferulatecontentofmaizesheathsisassociatedwithre- 3. PalmqvistE,Hahn-HägerdalB(2000)Fermentationoflignocellulosichydrolysates.II: sistancetotheMediterraneancornborerSesamianonagrioides(Lepidoptera:Noctuidae). Inhibitorsandmechanismsofinhibition.BioresourTechnol74(1):25–33. JAgricFoodChem54(24):9140–9144. 4. SkerkerJM,etal.(2013)Dissectingacomplexchemicalstress:Chemogenomicpro- 21.BaranowskiJD,DavidsonPM,NagelCW,BranenAL(1980)InhibitionofSaccharo- filingofplanthydrolysates.MolSystBiol9:674. mycescerevisiaebynaturallyoccurringhydroxycinnamates.JFoodSci45(3):592–594. 5. FitzPatrick M, Champagne P, Cunningham MF, Whitney RA (2010) A biorefinery 22.PiotrowskiJS,MorfordSL,RilligMC(2008)Inhibitionofcolonizationbyanativear- processingperspective:Treatmentoflignocellulosicmaterialsfortheproductionof buscularmycorrhizalfungalcommunityviaPopulustrichocarpalitter,litterextract, value-addedproducts.BioresourTechnol101(23):8915–8922. andsolublephenoliccompounds.SoilBiolBiochem40(3):709–717. 23.JoWJ,etal.(2008)IdentificationofgenesinvolvedinthetoxicresponseofSaccha- 6. AvenotHF,SellamA,KaraoglanidisG,MichailidesTJ(2008)Characterizationofmu- romycescerevisiaeagainstironandcopperoverloadbyparallelanalysisofdeletion tations in the iron-sulphur subunit of succinate dehydrogenase correlating with mutants.ToxicolSci101(1):140–151. BoscalidresistanceinAlternariaalternatafromCaliforniapistachio.Phytopathology 24.RogersB,etal.(2001)ThepleitropicdrugABCtransportersfromSaccharomyces 98(6):736–742. cerevisiae.JMolMicrobiolBiotechnol3(2):207–214. 7. LerochM,KretschmerM,HahnM(2011)FungicideresistancephenotypesofBotrytis 25.FaiPB,GrantA(2009)Arapidresazurinbioassayforassessingthetoxicityoffungi- cinereaisolatesfromcommercialvineyardsinsouthwestGermany.JPhytopathol cides.Chemosphere74(9):1165–1170. 159(1):63–65. 26.ParsonsAB,etal.(2006)Exploringthemode-of-actionofbioactivecompoundsby 8. WightwickAM,SalzmanSA,ReichmanSM,AllinsonG,MenziesNW(2013)Effectsof chemical-geneticprofilinginyeast.Cell126(3):611–625. copperfungicideresiduesonthemicrobialfunctionofvineyardsoils.EnvironSci 27.HoCH,etal.(2011)Combiningfunctionalgenomicsandchemicalbiologytoidentify PollutResInt20(3):1574–1585. targetsofbioactivecompounds.CurrOpinChemBiol15(1):66–78. 9. MackieKA,MüllerT,ZikeliS,KandelerE(2013)Long-termcopperapplicationinan 28.Smith AM, et al. (2009) Quantitative phenotyping via deep barcodesequencing. organicvineyardmodifiesspatialdistributionofsoilmicro-organisms.SoilBiolBio- GenomeRes19(10):1836–1842. chem65(2013):245–253. 29.Martin-Yken H, et al. (2001) Saccharomyces cerevisiae YCRO17c/CWH43 encodes 10.AltizerS,OstfeldRS,JohnsonPTJ,KutzS,HarvellCD(2013)Climatechangeandin- aputativesensor/transporterproteinupstreamoftheBCK2branchofthePKC1- fectious diseases: From evidence to a predictive framework. Science 341(6145): dependentcellwallintegritypathway.Yeast18(9):827–840. 514–519. 30.JeschSA,GasparML,StefanCJ,AregullinMA,HenrySA(2010)Interruptionofinositol 11.GarrettKA,DendySP,FrankEE,RouseMN,TraversSE(2006)Climatechangeeffects sphingolipidsynthesistriggersStt4p-dependentprotein kinaseC signaling.J Biol onplantdisease:Genomestoecosystems.AnnuRevPhytopathol44(1):489–509. Chem285(53):41947–41960. 12.AlexanderBD,PerfectJR(1997)Antifungalresistancetrendstowardstheyear2000. 31.OhkuniK,OkudaA,KikuchiA(2003)YeastNap1-bindingproteinNbp2pisrequired Implicationsfortherapyandnewapproaches.Drugs54(5):657–678. formitoticgrowthathightemperaturesandforcellwallintegrity.Genetics165(2): 13.SarmaBK,SinghUP(2003)FerulicacidmaypreventinfectionofCicerarietinumby 517–529. Sclerotiumrolfsii.WorldJMicrobiolBiotechnol19(2):123–127. 32.KitagakiH,WuH,ShimoiH,ItoK(2002)Twohomologousgenes,DCW1(YKL046c) andDFG5,areessentialforcellgrowthandencodeglycosylphosphatidylinositol(GPI)- 14.HeerD,SauerU(2008)Identificationoffurfuralasakeytoxininlignocellulosichy- drolysatesandevolutionofatolerantyeaststrain.MicrobBiotechnol1(6):497–506. acenrcehvoisrieade.mMeomlbMriacnroebpiorlo4te6i(n4s):1r0e1q1u–ir1e0d22f.or cell wall biogenesis in Saccharomyces 15.JayakodyLN,HayashiN,KitagakiH(2011)Identificationofglycolaldehydeasthekey 33.SurmaMA,etal.(2013)AlipidE-MAPidentifiesUbx2asacriticalregulatoroflipid inhibitorofbioethanolfermentationbyyeastandgenome-wideanalysisofitstoxicity. saturationandlipidbilayerstress.MolCell51(4):519–530. BiotechnolLett33(2):285–292. 34.CostanzoM,etal.(2010)Thegeneticlandscapeofacell.Science327(5964):425–431. 16.RalphJ,QuideauS,GrabberJH,HatfieldRD(1994)Identificationandsynthesisofnew 35.Reinoso-MartínC,SchüllerC,Schuetzer-MuehlbauerM,KuchlerK(2003)Theyeast ferulicaciddehydrodimerspresentingrasscellwalls.JChemSoc23:3485–3498. proteinkinaseCcellintegritypathwaymediatestolerancetotheantifungaldrug 17.RalphJ,etal.(1998)Cellwallcross-linkingingrassesbyferulatesanddiferulates. caspofunginthroughactivationofSlt2pmitogen-activatedproteinkinasesignaling. LigninandLignanBiosynthesis,ACSSymposiumSeries,edsLewisNG,SarkanenS EukaryotCell2(6):1200–1210. (AmericanChemicalSociety,Washington,DC),pp209–236. 36.OhnukiS,OkaS,NogamiS,OhyaY(2010)High-content,image-basedscreeningfor 18.VismehR,etal.(2013)Profilingofdiferulates(plantcellwallcross-linkers)usingultrahigh- drugtargetsinyeast.PLoSONE5(4):e10177. performanceliquidchromatography-tandemmassspectrometry.Analyst(Lond)138(21): 37.OhyaY,etal.(2005)High-dimensionalandlarge-scalephenotypingofyeastmutants. 6683–6692. ProcNatlAcadSciUSA102(52):19015–19020. E1496 | www.pnas.org/cgi/doi/10.1073/pnas.1410400112 Piotrowskietal. S U L P S A N 38.OkadaH,OhnukiS,RonceroC,KonopkaJB,OhyaY(2014)Distinctrolesofcellwall 50.Chen L, et al. (2011) Trichoderma harzianum SQR-T037 rapidly degrades allelo- P biogenesisinyeastmorphogenesisasrevealedbymultivariateanalysisofhigh- chemicalsinrhizospheresofcontinuouslycroppedcucumbers.ApplMicrobiolBio- dimensionalmorphometricdata.MolBiolCell25(2):222–233. technol89(5):1653–1663. 39.OhnukiS,etal.(2012)Analysisofthebiologicalactivityofanovel24-membered 51.WestbrookJ,BarterGE,ManleyDK,WestTH(2014)Aparametricanalysisoffuture macrolideJBIR-19inSaccharomycescerevisiaebythemorphologicalimagingpro- ethanoluseinthelight-dutytransportationsector:CantheUSmeetitsRenewableFuel gramCalMorph.FEMSYeastRes12(3):293–304. Standardgoalswithoutanenforcementmechanism?EnergyPolicy65(2014):419–431. 40.IwakiA,OhnukiS,SugaY,IzawaS,OhyaY(2013)Vanillininhibitstranslationand 52.LauMW,DaleBE(2009)CellulosicethanolproductionfromAFEX-treatedcornstover inducesmessengerribonucleoprotein(mRNP)granuleformationinsaccharomyces using Saccharomyces cerevisiae 424A(LNH-ST). Proc Natl Acad Sci USA 106(5): cerevisiae:Applicationandvalidationofhigh-content,image-basedprofiling.PLoS 1368–1373. ONE8(4):e61748. 53.FungS-Y,etal.(2014)Unbiasedscreeningofmarinespongeextractsforanti- 41.BalashovSV,ParkS,PerlinDS(2006)Assessingresistancetotheechinocandinanti- inflammatoryagentscombinedwithchemicalgenomicsidentifiesgirollineasanin- fungaldrugcaspofungininCandidaalbicansbyprofilingmutationsinFKS1.Anti- hibitorofproteinsynthesis.ACSChemBiol9(1):247–257. microbAgentsChemother50(6):2058–2063. 54.Andrusiak K (2012) Adapting S. cerevisiae chemical genomics for identifyingthe 42.JohnsonME,EdlindTD(2012)TopologicalandmutationalanalysisofSaccharomyces cerevisiaeFks1.EukaryotCell11(7):952–960. modesofactionofnaturalcompounds.Mastersthesis(UniversityofToronto,Toronto). 55.RobinsonDG,ChenW,StoreyJD,GreshamD(2014)DesignandanalysisofBar-seq 43.CokolM,etal.(2011)Systematicexplorationofsynergisticdrugpairs.MolSystBiol experiments.G3(Bethesda)4(1):11–18. 7(2011):544. 56.RobinsonMD,McCarthyDJ,SmythGK(2010)edgeR:ABioconductorpackagefor 44.KirazN,etal.(2010)Synergisticactivitiesofthreetriazoleswithcaspofunginagainst differentialexpressionanalysisofdigitalgeneexpressiondata.Bioinformatics26(1): Candidaglabrataisolatesdeterminedbytime-kill,Etest,anddiskdiffusionmethods. AntimicrobAgentsChemother54(5):2244–2247. 139–140. 45.CassoneA,MasonRE,KerridgeD(1981)Lysisofgrowingyeast-formcellsofCandida 57.Boyle EI, et al. (2004) GO:TermFinder—open source software for accessing Gene albicansbyechinocandin:Acytologicalstudy.Sabouraudia19(2):97–110. OntologyinformationandfindingsignificantlyenrichedGeneOntologytermsasso- 46.UtsugiT,etal.(2002)Movementofyeast1,3-β-glucansynthaseisessentialforuni- ciatedwithalistofgenes.Bioinformatics20(18):3710–3715. formcellwallsynthesis.GenesCells7(1):1–9. 58.YvertG,etal.(2013)Single-cellphenomicsrevealsintra-speciesvariationofpheno- 47.FreeSJ(2013)Fungalcellwallorganizationandbiosynthesis.AdvGenet81:33–82. typicnoiseinyeast.BMCSystBiol7(1):54. 48.KopeckáM,GabrielM(1992)Theinfluenceofcongoredonthecellwalland(1----3)- 59.AbeM,QadotaH,HirataA,OhyaY(2003)LackofGTP-boundRho1pinsecretory beta-D-glucan microfibril biogenesis in Saccharomyces cerevisiae. Arch Microbiol vesiclesofSaccharomycescerevisiae.JCellBiol162(1):85–97. 158(2):115–126. 60.AbeM,etal.(2001)Yeast1,3-β-glucansynthaseactivityisinhibitedbyphytosphingosine 49.ImaiK,NodaY,AdachiH,YodaK(2005)Anovelendoplasmicreticulummembrane localizedtotheendoplasmicreticulum.JBiolChem276(29):26923–26930. proteinRcr1regulateschitindepositioninthecellwallofSaccharomycescerevisiae. 61.InoueSB,etal.(1995)Characterizationandgenecloningof1,3-β-D-glucansynthase JBiolChem280(9):8275–8284. fromSaccharomycescerevisiae.EurJBiochem231(3):845–854. Y G O L O BI O R C MI Piotrowskietal. PNAS | PublishedonlineMarch9,2015 | E1497 Supporting Information Piotrowski et al. 10.1073/pnas.1410400112 SIMethods 0.6.2) (3) using the default parameters, with the exception of Compounds,InitialScreening,andGrowth.Thediferulatecompounds thefractionofmissingalignmentsthreshold,whichwassetat tested were synthesized as described by Lu et al. (1) and resus- 0.08(–ninbwaaln).SNPandindeldetectionwereperformed pended in DMSO. Caspofungin, nikkomycin Z, and MMS were with the Genome Analysis Toolkit (GATK version 1.4) (4) purchased from Sigma-Aldrich. Echinocandin B was a gift from followingtheirbestpracticevariant-callingworkflow(https://www. O.Kondo(ChugaiPharmaceuticals,Tokyo,Japan).Micafungin broadinstitute.org/gatk/).Duplicatereadsweremarkedfollowedby was provided by Astellas Pharma. Diferulates were initially base quality recalibration using a single nucleotide polymorphism screenedataconcentrationof1mg/mLtodeterminebioactivity. databasedesignedfor S.cerevisiae.Tominimizefalse-positivevar- CellsofSaccharomycescerevisiae(MATαpdr1Δ::natMXpdr3Δ:: iant calls, stringent parameters were used: namely, the minimum KI.URA3snq2Δ::KI.LEU2can1Δ::STE2pr-Sp_his5lyp1Δhis3Δ1 base quality required to consider a base for calling was 30, and leu2Δ0ura3Δ0met15Δ0),referredtoasthecontrolstrain,were the minimum phred-scaled confidence threshold for genotype grownin200-μLculturesat30°CinYPDwithadrugorDMSO calling was 50 (–mbq and –scc in the UnifiedGenotyper tool). CustomPerlscriptswereusedtofurtherfiltercallsonthebasis control.PlateswerereadonaTECANM1000overa48-hgrowth of read depth, mapping quality, and strand bias. This analysis period.ThespecificgrowthratewascalculatedusingGCATanal- revealedanSNPinthegeneSUR1(glutamate>stopcodon)in ysis software (https://gcat3-pub.glbrc.org/) (2). When presented, the poacicacid-resistant mutant. IC values for growth rate inhibition were calculated from trip- 50 licate eight-point dose curves and SigmaPlot 12.0. When pre- sented,errorbarsaremeans±SEsofatleastthreereplicates. CellLeakageAssays.AFungaLight CellViability Assay(L34952; Invitrogen)usingaGuavaFlowCytometer(Millipore)wasused to determine if poacic acid caused membrane damage. The DeterminingtheMostSensitivePathwayThroughChemicalGenomics. populationofstainedcells(damagedintegrity)vs.nonstained A complex/pathway score based on chemical genomic data to cellscanbedeterminedbyflowcytometry.Caspofungin(50ng/mL) identifyproteincomplexesorpathwayswasdevelopedbasedon wasincludedasapositivecontrol.MMSandDMSOwereincluded which members showed significant deviation in their chemical asanoncellwall-targetingcontrolandasolventcontrol,respectively. genetic interactions in the presence of a compound. For each To test the effects of the compounds on both active and arrested complex,thechemicalgeneticinteractionscoreofthegenesinthe cells,log-phasecultureswerewashedwith1×PBSandresuspended complex with the compound was summed.To determine signif- toanOD ineitherYPDmediumorYP(nocarbonsource)inthe icance,theexpectationsforsuchasumforrandomsetsofgenesof 0.5 presenceofthedrugs(n=3)for4hat30°C.Thecellswerethen equal size were calculated. The random sets of equal size were stainedandimmediatelyreadbyflowcytometry.One-wayANOVA expectedtohavemeansequaltothebackgroundmeanandSDs andTukey’stestwereusedtocalculatethedifferencebetweendrug equal to the background SD/sqrt(n). With this information, a z treatmentsamongcellswitharrestedgrowth. score (number of SDs from the expected mean) for each com- plexor pathwaycan be computed: SynergyScreening.To test for synergy, a 6 × 6-dose matrix was initially used to identify potentially synergistic dose combina- Pathway z score=ðΣ=n−μÞ=ðσ×sqrtðnÞÞ; tions, and these points were then confirmed in triplicate. Cul- tures (200 μL) were grown with combinations of poacic acid where Σ = sum of the chemical genetic interaction scores of (125μg/mL),caspofungin(12.5ng/mL),andfluconazole(3.8μg/mL), genesinthecomplex,μ=meanofthechemicalgeneticinterac- and the ODs of relevant single-agent and solvent controls were tion scores of the compounds with all genes studies, σ = SD of measured after 24h. Synergy was determined by comparing ac- chemical genetic interaction scores of the compounds with all tual OD in the presence of compound combinations with an genesinthe study, andn =size of the complex. expected value calculated using the multiplicative hypothesis. Thismethodassumesthat,intheabsenceofaninteraction,each Isolation, Sequencing, and Evaluation of Drug-Resistant Mutants. compound would decrease the OD of the cell culture by the Agar containing 500 μg/mL poacic acid was inoculated with ∼1 samefractioninthepresenceoftheothercompoundasitdoes million cells of yeast (control strain). After 1 wk, two colonies when applied alone (that is, E = A × B/C, where E is the ex- were found growing on the agar. Single-colony isolates were pected OD, A is OD when compound A is applied alone, B is obtained and found to be resistant to poacic acid. For whole- OD when compound B is applied alone, and C is OD of the genome sequencing, single-colony isolates of poacic acid- control culture (DMSO). In the presence of synergy, the actual resistant mutant, the caspofungin-resistant mutant, and the con- OD value is lower than the expected OD. A paired t test was trol strain (WT) were grown in triplicate 200-μL cultures and usedtoconfirmstatisticalsignificanceofthisdifferenceinthree pooled for genomic DNA extraction (Epicentre MasterPure replicates ofthe experiment. Yeast Kit; MPY80200). The genomic DNA was prepared for Illuminawhole-genomesequencingusingtheIlluminaTruSeqKkit Staining of Cells with Poacic Acid. Log-phase yeast cells (his3Δ) (FC-121-3001) and sequenced by 150-bp paired-end reads on the were harvested by centrifugation, washed two times with PBS, MiSeqplatform. sonicatedmildly,andthen,incubatedwith0.25%(wt/vol)poacic To determine mutations in the drug-resistant mutants, read acid for 5 min. A small aliquot of the cells was mounted on qualityanalysiswasperformedusingFastQC(www.bioinformatics. a glass slide and observed under an Axioimager M1 Fluores- babraham.ac.uk/projects/fastqc/). Short reads were examined for cenceMicroscope (CarlZeiss)using theXF09FilterSet(Opto quality and trimmed at the 3′ end when average base quality in a Science; excitation wavelength, 340–390 nm; emission wave- 3-nt window fell below Q30. Short reads were mapped to the length,517.5–552.5 nm). standard S. cerevisiae reference genome, strain S288c (obtained from the National Center for Biotechnology Information RefSeq MannoproteinandGlucanStaining.β-1,3-Glucan was stained with repository),usingBurrows-WheelerAlignment(BWAversion anilineblue(016-21302;WakoChemicals)asdescribedpreviously Piotrowskietal.www.pnas.org/cgi/content/short/1410400112 1of6 (5)withslightmodification.Briefly,log-phaseyeastcells(his3Δ) and observed under a fluorescent microscope using a regular were cultured in YPD with poacic acid (125 μg/mL) at 25 °C. DAPI filter set (Carl Zeiss). Then,cellswerecollectedat0,2,4,and6haftertreatmentand stainedwithanilinebluewithoutfixationasdescribedpreviously Determination of Ferulate and Diferulates by Reverse-Phase HPLC– (6).CellsmountedonaglassslidewereexposedtoUVfor30s High-Resolution/AccurateMSinHydrolysates.Ammoniafiberexpan- to bleach out poacic acid fluorescence before acquiring images. sion treated corn stover hydrolysates samples were diluted 1:10, Staining of chitin or mannoproteins with calcofluor white (F3543; and20-μLsampleswereanalyzedbyreverse-phase(C18)HPLC– Sigma-Aldrich)orAlexa594-ConA(C11253;LifeTechnologies), high-resolution/accurate MS. Peak areas of peaks matching in respectively, was performed as described previously (6). For retentiontimeandaccuratemass±10ppmofauthenticreference cell-free glucan staining, yeast glucan (G0331; Tokyo Chem- standards were used to calculate concentrations by comparison ical Industry) was suspended to 0.125% (wt/vol) poacic acid withanexternalstandardcurve. 1.LuF,WeiL,AzarpiraA,RalphJ(2012)Rapidsynthesesofdehydrodiferulatesviabiomimetic 4.DePristoMA,etal.(2011)Aframeworkforvariationdiscoveryandgenotypingusing radicalcouplingreactionsofethylferulate.JAgricFoodChem60(34):8272–8277. next-generationDNAsequencingdata.NatGenet43(5):491–498. 2.SatoTK,etal.(2014)HarnessinggeneticdiversityinSaccharomycescerevisiaeforim- 5.WatanabeD,AbeM,OhyaY(2001)YeastLrg1pactsasaspecializedRhoGAPregu- provedfermentationofxyloseinhydrolysatesofalkalinehydrogenperoxidepre- lating1,3-β-glucansynthesis.Yeast18(10):943–951. treatedbiomass.ApplEnvironMicrobiol80(2):540–554. 6.OkadaH,OhyaY(2015)ColdSpringHarborProtocols(ColdSpringHarborLabPress, 3.LiH,DurbinR(2009)FastandaccurateshortreadalignmentwithBurrows-Wheeler Plainview,NY). transform.Bioinformatics25(14):1754–1760. Fig.S1. Poacicacidtreatmentreducesglucanstainingwithanilinebluebuthasnoeffectonmannoproteinstraining.Thecontrolstrainyeastcells(his3Δ) weregrowninYPDat25°Cuntilearlylogphase,transferredtofreshYPDmediumcontainingpoacicacid(125μg/mL)orDMSO[0.125%(vol/vol)]asasolvent control,andculturedfor6h.Thecellswerecollected,andthecellwallcomponentsmannoproteinswerestainedwithAlexa594-conjugatedConAfollowedby β-1,3-glucanstainingwithanilineblue.Thecellswereobservedunderafluorescentmicroscope,andover150buddingcellswerecountedaccordingtothe stainingsignalfromthreeindependentexperiments.AStudent’sttestwasusedtodeterminesignificantdifferences(mean±SE;n=3). Piotrowskietal.www.pnas.org/cgi/content/short/1410400112 2of6

Description:
velopment in planta on detached soybean leaves. Solvent .. pellets were resuspended in 10 mM Tris·HCl, pH 7.5, containing 5 mg/mL zymolyase 100T . tations in the iron-sulphur subunit of succinate dehydrogenase correlating with .. expected to have means equal to the background mean and SDs.
See more

The list of books you might like

Most books are stored in the elastic cloud where traffic is expensive. For this reason, we have a limit on daily download.