ArticlePDF Available

HNMT Upregulation Induces Cancer Stem Cell Formation and Confers Protection against Oxidative Stress through Interaction with HER2 in Non‐Small‐Cell Lung Cancer

Authors:
  • Translational Research Laboratory Shuang-Ho Hospital, Taipei Medical University

Abstract

Background: The treatment of non-small-cell lung cancer (NSCLC) involves platinum-based chemotherapy. It is typically accompanied by chemoresistance resulting from antioxidant properties conferred by cancer stem cells (CSCs). Human epidermal growth factor receptor 2 (HER2) enhances CSCs and antioxidant properties in cancers, including NSCLC. Methods: Here, we elucidated the role of histamine N-methyltransferase (HNMT), a histamine metabolism enzyme significantly upregulated in NSCLC and coexpressed with HER2. HNMT expression in lung cancer tissues was determined using quantitative reverse transcription PCR (RT-qPCR). A publicly available dataset was used to determine HNMT's potential as an NSCLC target molecule. Immunohistochemistry and coimmunoprecipitation were used to determine HNMT-HER2 correlations and interactions, respectively. HNMT shRNA and overexpression plasmids were used to explore HNMT functions in vitro and in vivo. We also examined miRNAs that may target HNMT and investigated HNMT/HER2's role on NSCLC cells' antioxidant properties. Finally, how HNMT loss affects NSCLC cells' sensitivity to cisplatin was investigated. Results: HNMT was significantly upregulated in human NSCLC tissues, conferred a worse prognosis, and was coexpressed with HER2. HNMT depletion and overexpression respectively decreased and increased cell proliferation, colony formation, tumorsphere formation, and CSCs marker expression. Coimmunoprecipitation analysis indicated that HNMT directly interacts with HER2. TARGETSCAN analysis revealed that HNMT is a miR-223 and miR-3065-5p target. TBHp treatment increased HER2 expression, whereas shHNMT disrupted the Nuclear factor erythroid 2-related factor 2 (Nrf2)/ hemeoxygenase-1 (HO-1)/HER2 axis and increased reactive oxygen species accumulation in NSCLC cells. Finally, shHNMT sensitized H441 cells to cisplatin treatment in vitro and in vivo. Conclusions: Therefore, HNMT upregulation in NSCLC cells may upregulate HER2 expression, increasing tumorigenicity and chemoresistance through CSCs maintenance and antioxidant properties. This newly discovered regulatory axis may aid in retarding NSCLC progression and chemoresistance.
Int.J.Mol.Sci.2022,23,1663.https://doi.org/10.3390/ijms23031663www.mdpi.com/journal/ijms
Article
HNMTUpregulationInducesCancerStemCellFormationand
ConfersProtectionagainstOxidativeStressthrough
InteractionwithHER2inNonSmallCellLungCancer
KuangTaiKuo
1,2
,ChengHsinLin
3,4,5
,ChunHuaWang
6,7
,NarpatiWesaPikatan
8
,VijeshKumarYadav
9
,
IatHangFong
9
,ChiTaiYeh
9,10,
*,WeiHwaLee
11
andWenChienHuang
12,13,
*
1 DivisionofThoracicSurgery,DepartmentofSurgery,SchoolofMedicine,CollegeofMedicine ,
TaipeiMedicalUniversity,Taipei110,Taiwan;ktkuo@tmu.edu.tw
2 DivisionofThoracicSurgery,DepartmentofSurgery,ShuangHoHospital,TaipeiMedicalUniversity,
NewTaipeiCity235,Taiwan
3 TaipeiHeartInstitute,TaipeiMedicalUniversity,Taipei110,Taiwan;chlin99025@tmu.edu.tw
4 DivisionofCardiovascularSurgery,DepartmentofSurgery,ShuangHoHospital,
TaipeiMedicalUniversity,NewTaipeiCity235,Taiwan
5 DivisionofCardiovascularSurgery,DepartmentofSurgery,SchoolofMedicine,CollegeofMedicine,
TaipeiMedicalUniversity,Taipei110,Taiwan
6 DepartmentofDermatology,TaipeiTzuChiHospital,BuddhistTzuChiMedicalFoundation,
NewTaipeiCity231,Taiwan;10205@s.tmu.edu.tw
7 SchoolofMedicine,BuddhistTzuChiUniversity,Hualien970,Taiwan
8 DivisionofUrology,DepartmentofSurgery,FacultyofMedicine,UniversitasGadjahMada/Dr.Sardjito
Hospital,Yogyakarta55281,Indonesia;narpatiwp@gmail.com
9 DepartmentofMedicalResearch&Education,ShuangHoHospital,TaipeiMedicalUniversity,
NewTaipeiCity235,Taiwan;vijeshp2@gmail.com(V.K.Y.);impossiblewasnothing@hotmail.com(I.H.F.)
10 DepartmentofMedicalLaboratoryScienceandBiotechnology,YuanpeiUniversityofMedicalTechnology,
Hsinchu300,Taiwan
11 DepartmentofPathology,TaipeiMedicalUniversity—ShuangHoHospital,NewTaipeiCity235,Taiwan;
whlpath97616@s.tmu.edu.tw
12 DepartmentofMedicine,MacKayMedicalCollege,NewTaipeiCity252,Taiwan
13 DivisionofThoracicSurgery,DepartmentofSurgery,MacKayMemorialHospital,Taipei104,Taiwan
*Correspondence:ctyeh@s.tmu.edu.tw(C.T.Y.);wjhuang0@gmail.com(W.C.H.);
Tel.:+88622490088(ext.8881)(C.T.Y.);+88622490088(ext.2919)(W.C.H.);
Fax:+886222480900(C.T.Y.);+886222480900(W.C.H.)
Abstract:Background:Thetreatmentofnonsmallcelllungcancer(NSCLC)involvesplatinum
basedchemotherapy.Itistypicallyaccompaniedbychemoresistanceresultingfromantioxidant
propertiesconferredbycancerstemcells(CSCs).Humanepidermalgrowthfactorreceptor2
(HER2)enhancesCSCsandantioxidantpropertiesincancers,includingNSCLC.Methods:Here,
weelucidatedtheroleofhistamineNmethyltransferase(HNMT),ahistaminemetabolismenzyme
significantlyupregulatedinNSCLCandcoexpressedwithHER2.HNMTexpressioninlungcancer
tissueswasdeterminedusingquantitativereversetranscriptionPCR(RTqPCR).Apubliclyavail
abledatasetwasusedtodetermineHNMT’spotentialasanNSCLCtargetmolecule.Immunohisto
chemistryandcoimmunoprecipitationwereusedtodetermineHNMT–HER2correlationsandin
teractions,respectively.HNMTshRNAandoverexpressionplasmidswereusedtoexploreHNMT
functionsinvitroandinvivo.WealsoexaminedmiRNAsthatmaytargetHNMTandinvestigated
HNMT/HER2sroleonNSCLCcells’antioxidantproperties.Finally,howHNMTlossaffects
NSCLCcells’sensitivitytocisplatinwasinvestigated.Results:HNMTwassignificantlyupregu
latedinhumanNSCLCtissues,conferredaworseprognosis,andwascoexpressedwithHER2.
HNMTdepletionandoverexpressionrespectivelydecreasedandincreasedcellproliferation,col
onyformation,tumorsphereformation,andCSCsmarkerexpression.Coimmunoprecipitationanal
ysisindicatedthatHNMTdirectlyinteractswithHER2.TARGETSCANanalysisrevealedthat
HNMTisamiR223andmiR30655ptarget.TBHptreatmentincreasedHER2expression,whereas
shHNMTdisruptedtheNuclearfactorerythroid2relatedfactor2(Nrf2)/hemeoxygenase1(HO
Citation:Kuo,K.T.;Lin,C.H.;
Wang,C.H.;Pikatan,N.W.;
Yadav,V.K.;Fong,I.H.;Yeh,C.T.;
Lee,W.H.;Huang,W.C.HNMT
HNMTUpregulationInduces
CancerStemCellFormationand
ConfersProtectionagainstOxidative
StressthroughInteractionwith
HER2inNonSmallCellLung
Cancer.Int.J.Mol.Sci.2022,23,1663.
https://doi.org/10.3390/ijms23031663
AcademicEditors:PinarUysal
OnganerandRichardW.E.
Clarkson
Received:22December2021
Accepted:28January2022
Published:31January2022
Publisher’sNote:MDPIstaysneu
tralwithregardtojurisdictional
claimsinpublishedmapsandinstitu
tionalaffiliations.
Copyright:©2022bytheauthors.Li
censeeMDPI,Basel,Switzerland.
Thisarticleisanopenaccessarticle
distributedunderthetermsandcon
ditionsoftheCreativeCommonsAt
tribution(CCBY)license(https://cre
ativecommons.org/licenses/by/4.0/).
Int.J.Mol.Sci.2022,23,16632of21
1)/HER2axisandincreasedreactiveoxygenspeciesaccumulationinNSCLCcells.Finally,shHNMT
sensitizedH441cellstocisplatintreatmentinvitroandinvivo.Conclusions:Therefore,HNMTup
regulationinNSCLCcellsmayupregulateHER2expression,increasingtumorigenicityandchemo
resistancethroughCSCsmaintenanceandantioxidantproperties.Thisnewlydiscoveredregulatory
axismayaidinretardingNSCLCprogressionandchemoresistance.
Keywords:nonsmallcelllungcancer;cancerstemcells;humanepidermalgrowthfactorreceptor
2;NRF2/HO1/HER2axis;HNMT/HER2srole
1.Introduction
Lungcancerisoneofthemostcommoncausesofcancerassociatedmortalityworld
wide,accountingforalmost1.76milliondeathcasesperyear.Nonsmallcelllungcancer
(NSCLC)islinkedwithapproximately80%ofalllungcancercases[1].Radiotherapy
aloneorwithchemotherapyandadjuvantdurvalumabarethemostcommontreatments
forpatientswithlocallyadvancedNSCLC.Platinumbasedchemotherapyremainsthe
standardfirstlinedefensetreatmentformetastaticNSCLCbutisfrequentlyaccompanied
bychemoresistance.Manyotherchemotherapeuticsdrugshavebeentriedasasecond
linetreatmentbutthepandrugresistanceacquiredbymostNSCLCresultsintreatment
failureanduninhibiteddiseaseprogression.Therefore,developingpredictivebiomarkers
toidentifypatientsandtargetedtherapiestotreatpatientslikelytobenefitfromthese
therapiesiscritical[2].
Tumormetastasisandchemoresistancehavebeeninvestigatedseparatelyinthepast,
butthesearefrequentlyobservedtogetherclinicallyandarelinkedbiologically.Inastudy
onbreastcancer,theinteractionbetweenthecancercellsandthehostmicroenvironment
wascharacterized,andtheconnectingchemotherapyfailurewithmetastaticrelapsewas
observed[3].Nevertheless,thekeymolecularmechanismsinvolvedintheassociationbe
tweenmetastasisandchemoresistance—thatmightdifferincancertypesandclinicalset
tings—remainunknown.Cancerstemcells(CSCs)contributetotumorrelapse,cancercell
propagation,andchemo–radioresistance[4].Theyalsoconferprotectiveantioxidant
propertiestocancers,leadingtochemoresistance[5].
Reactiveoxygenspecies(ROS),includingdifferentspeciesofsecondarymessengers,
suchashydrogenperoxide(H2O2),superoxideanion(O2),hypochlorousacid(HOCl),
singletoxygen(1O2),andhydroxylradical(OH),areinvolvedincellsignalingforvarious
biologicalprocessesinbothnorms.Wealandcancercells[6].ROSregulatecriticalrecep
tortyrosinekinasesignalingtargets,includinghumanepidermalgrowthfactorreceptor
2(HER2),whichalsodirectlyinteractswithNRF2toregulateDNAbindingactivity.Fur
thermore,HER2downstreameffectorsincludePI3K,andmitogenactivatedproteinki
nasesignalingpromotestheNrf2bindingtoDNAandresultsinthetranscriptionalacti
vationoftargetgenes[7,8].
Histamine,throughitsinteractionswithH1–4receptorsubtypes,stimulatesvarious
signalingpathways.AllthesereceptorsaremembersoftheheptahelicalGproteincou
pledreceptor(GPCR)family[9–11].HighhistamineNmethyltransferase(HNMT)ex
pressionhasbeennotedinpatientswithductalbreastcancercomparedwithhealthycon
trols[12].AnotherstudyreportedthatahistamineH3receptor(H3R)antagonistinhibited
proliferationandtriggeredcaspasedependentapoptosisinbothestrogenreceptorposi
tiveand‐negativebreastcancercells[13].However,theexactmechanismunderlyingthis
overexpressionanditseffectinNSCLCremainsunexplored.
Inthisstudy,bigdata,clinicalpatients,andinvitroandinvivoapproacheswereused
toinvestigatetheroleofHNMTinNSCLC.WeevaluatedHNMTexpressioninNSCLC
anditsinteractionorcoexpressionwithHER2.Ourfindingsprovidethefirstevidence
thattheCSCspopulationinNSCLChasalowermiR3065/223expression,resultingina
higherexpressionoftheirtargetgene,HNMT.This,inturn,resultsinafeedbackloop
Int.J.Mol.Sci.2022,23,16633of21
mechanisminwhichHNMTupregulationaffectsHER2expressionandsubsequentlyin
creaseschemoresistanceinNSCLCthroughtumorsphereformationandtheantioxidant
responsesystem.
2.Results
2.1.HNMTUpregulationinNSCLCTissuesIsRelatedtoWorsenedPrognosisand
SignificantlyCoexpressedwithHER2
ToassesstheroleofHNMTintheprogressionanddevelopmentofhumanNSCLC,
wefirstexaminedHNMTmRNAexpressionbyusingabioinformaticsapproach.HNMT
mRNAlevelsincreasedsignificantlyinpatientswithNSCLCcomparedwithpairednor
maladjacenttissue(Figure1A).Wethenextrapolatedourpreviousfindingtoevaluateits
prognosticsignificancebyusingthePrognoscandatabase[14].Weobservedthatpatients
withhigherHNMTexpression(n=74)hadworseprognosesthandidpatientswithlower
HNMTexpression(n=37;Figure1B).HER2mutationwasnotedtoleadtotheworst
prognosisinNSCLC.Inaddition,otherstudieshaveindicatedapotentialinteractionbe
tweenHNMTandHER2[15].Therefore,weinvestigatedtheinteractionbetweenthemin
NSCLC.WeusedtheR2database[16]andthreedatasets—namelyPeitsch(n=121),EXPO
(n=150),andBild(n=114)—anddetectedthatHNMTandHER2weresignificantlyco
expressed(Figure1C–E).WeverifiedHER2coexpressionwithHNMTintissuesobtained
frompatientswithNSCLC(Figure1F).Moreover,HER2wasnotmoresignificantlyex
pressedinNSCLCtumortissues,whereasHNMTwasstronglyexpressedinthetumor
tissuecomparedwiththenormaladjacenttissue(Figure1G).Notably,accordingtoour
bioinformaticsfindings,bothHER2andHNMTweresignificantlycoexpressedintheclin
icalsamples(Figure1H).Furthermore,wevalidatedthestandardtocomparetheclinico
pathologicparameters,asshowninTable1(below),wefoundthattumordifferentiation,
tumorsize,lymphnodemetastasis(LMN)status,andpathologicalclinicalstagere
mainedsignificantlydifferentbetweenHNMThighandlowexpressiongroup.Addition
ally,thecelltypewasalsoasignificantfactor,withHNMThighexpressionlessfrequent
inNSCLC.
Subsequentunivariate(UA)andmultivariateanalysis(MA)wasusedtoevaluatethe
significanceoftheexpressionofHNMTandHer2,age,gender,tumorsize,LNM,clinical
stage,subtypesonNSCLCspecificsurvival.OverexpressionofHNMT(hazardratio
0.294,95%confidenceinterval:0.108to0.806,p<0.017forUAandhazardratio0.152,95%
confidenceinterval:0.047to0.495;p<0.002forMA,Table2)couldbeconsideredasinde
pendentprognosticfactorsinNSCLCpatients.Takentogether,theseresultsimplythat
HNMTisassociatedwithHER2expressionandisapotentialtherapeutictargetinNSCLC.
Int.J.Mol.Sci.2022,23,16634of21
Figure1.AssociationofHNMTwithworseprognosisinNSCLCanditscorrelationwithHER2
expression.(A)HNMTmRNAexpressionwasevaluatedusingRTqPCR.Samplesweregrouped
asNSCLCandpairedwithnormaladjacent.(B)Kaplan–Meiersurvivalanalysisofpatientswith
NSCLCexpressingHNMTbasedonthePrognoscandatabase(GSE3141).HNMTandHER2
(ERBB2)genecorrelationanalysisdeterminedusingR2:GenomicsAnalysisandVisualizationPlat
form(http://r2.amc.nl;accessedon21Nov2021)fromthedataset(C)Peitsch(n=121),(D)EXPO(n
=150),and(E)Bild(n=114).(F)RepresentativeimmunohistochemistrystainingfromthreeNSCLC
cases.(G)JscorecomparisonbetweennontumorandtumortissuestakenfromNSCLCclinicaltis
suesamples.(H)HNMTandHER2proteinexpressioncorrelationanalysisinNSCLCclinicaltissue
samples.Scalebar:100μm.
Int.J.Mol.Sci.2022,23,16635of21
Table1.ThesummaryofclinicopathologicalfeaturesanddistributionofHNMTstatusin60pa
tientswithNSCLCpatients.
ClinicopathologicalVariables
 HNMT
N=60HighLowx2pValue
ExpressionExpression
Age,years
602911181.1330.287
>60311615
Gender    
Male3718190.5190.471
Female23914
Differentiation
Well/Moderately3813254.8750.027
Poor22148
TumorSize(cm)    
53912279.1180.003
>521156
Lymphnodemetastasis
N03611257.5870.006
N1N224168
ClinicalStage    
Earlystage(III)3072311.38<0.001
Latestage(IIIIV)302010
Subtypes
Adeno3720173.1970.074
Squamous23716
HER2Qscore    
Low3511246.2510.012
High25169
Table2.UnivariateandmultivariateanalysisofHNMTexpressionintheNSCLCcohort.
ParameterUnivariateMultivariate
HR95%CIpHR95%CIp
Gender(Femalevs.male)0.6790.2741.6830.4030.5940.1791.9710.395
age_60(<60vs.>60)0.8570.3602.0370.7260.6240.2111.8400.392
tumor_50(<50vs.>50)0.7210.2971.7530.4710.2960.0541.6320.162
LN_12(1,2vs.0)0.6750.2781.6370.3840.4270.1251.4580.174
Subtype_c(squavs.adeno)0.9990.4132.4120.9980.7380.2242.4310.618
stage(latevs.early)1.1400.4792.7110.7681.0110.3163.2350.986
Differentiation_c(well,moderatelyvs.
poor)0.9520.4002.2670.9110.4720.0992.2550.347
HNMT(lowvs.high)0.2940.1080.8060.0170.1520.0470.4950.002
HER2(lowvs.high)1.0610.4422.5460.8941.3460.4084.4450.626
2.2.HNMTInteractswithHER2andAffectsNSCLCCellLineDevelopment
WenextexaminedHNMTexpressioninsixNSCLCcelllines(BEAS2B,CL10,CL1
5,H838,A549,andH441)throughWesternblotting.TheresultsrevealedthatHNMTpro
teinlevelsweresignificantlyupregulatedinA549andH441cellsbutweremostlyunder
expressedinotherNSCLCcelllines.Notably,HNMTexpressionwasaccompaniedby
HER2andHER3expression.ThecellswithhigherHNMTexpressionalsohadhighervi
mentinbutlowerNcadherinexpression,implyingHNMT’sroleinEMTinNSCLC
Int.J.Mol.Sci.2022,23,16636of21
(Figure2A).Therefore,weemployedH441andCL10cellsforlossoffunctionandgain
offunctionteststoelucidatetheroleofHNMTexpressioninNSCLC.Figure2B,Cdepicts
theeffectivenessoftheclonesusedforHNMTknockdownoroverexpressioninH441and
CL10cells,respectively.BecausewedetectedHER2coexpressionwithHNMTinNSCLC
celllines,weinvestigatedtheassociationbetweenthetwo.Reciprocalimmunoprecipita
tionwithHNMTorHER2antibodiesindicatedapowerfulinteractionbetweenendoge
nousHNMTandHER2intheH441cells(Figure2D).HNMTalsoaffectedNSCLCcell
proliferation(Figure2E,F);itsknockdownwasassociatedwithreducedcellproliferation,
anditsoverexpressionwasassociatedwithanincreasedcellproliferationrate.Rawdata
ofwesternblotwithfullsizeblotshowninsupplementaryFigureS1.Theresultsofthese
experimentsfurtherconfirmtheinteractionbetweenHNMTandHER2andtheirrolein
NSCLCcelldevelopment.
Figure2.MolecularmanipulationeffectivenessofHNMTinlungcancercelllines.(A)Westernblot
tingofNSCLCcelllines.CelllysatesfromsixNSCLCcelllineshavebeensubjecttoWesternblot
ting.HNMT,HER2,HER3,vimentin,andNcadweredetectedusingtheircorrespondingantibod
ies.TheexperimentswerecarriedoutaftercalibrationusingGAPDHasaninternalcontrol.(B)
WesternblottingfortheHNMTandHER2expressioninH441cellstransfectedwithshScramble,
shHNMT#1,orshHNMT#2.(C)WesternblottingforHNMTandHER2expressionofCL10cells
transfectedwithcontrolvector,HNMToverexpressingplasmid#1(pCMV6HNMT,HNMT#1),or
Int.J.Mol.Sci.2022,23,16637of21
HNMToverexpressingplasmid#2(pCMVHNMTN1,HNMT#2).Densitometryanalysisforthe
proteinexpressionofbothcelllinesisprovided.(D)Coimmunoprecipitationwasusedtodetect
HNMT–HER2interactionandβactinwereusedasahousekeepingcontrol.(E)Cellproliferation
growthcurvecomparisonofH441cellstransfectedwithshScramble,shHNMT#1,orshHNMT#2,
and(F)CL10cellstransfectedwithvectorcontrolorcontrolvector,HNMToverexpressingplasmid
#1(HNMT#1),orHNMToverexpressingplasmid#2(HNMT#2).***p<0.001.
2.3.HNMTPromotesCSCDevelopmentinNSCLCCellLines
ToevaluatewhetherHNMTalsoplayedaroleintheoriginofcancerstemnessin
NSCLC,wefirstassessedthecolonyformationabilityofH441withHNMTknockdown
andofCL10withHNMToverexpression.WeobservedthatHNMTknockdownnega
tivelyinfluencedH441colonyformation.Itsoverexpression,however,significantlyin
creasedthecolonyformationabilityofCL10cells(Figure3A).Similarly,HNMTknock
downandoverexpressionresultedinreducedandincreased,respectively,tumorsphere
formationabilityinH441andCL10cellslines(Figure3B).HNMTmanipulationalsoaf
fectedstemnessmarkerexpression,whichwasevaluatedthroughWesternblotting.In
H441cells,shHNMTledtosignificantdownregulationofKLF4,NANOG,OCT4,and
CD133comparedwithshScramble.HNMToverexpression,inturn,increasedKLF4,
NANOG,OCT4,andCD133expressioninCL10cells(Figure3C),rawdataofwestern
blotwithfullsizeblotillustratedinsupplementaryFigureS2.
Int.J.Mol.Sci.2022,23,16638of21
Figure3.EffectsofHNMTonCSCsmaintenanceinlungcancercelllines.(A)H441andCL10cells
transfectedwithknockdownandexpressionvectorswereseededon6wellplates.Subsequentto2
weeks,thecolonieswerestainedwithcrystalviolet,andthenumberofcolonieswascounted.(B)
TumorsphereformationpotentialofNSCLCcells.BothH441andCL10cellscouldformtu
morspheres;HNMTknockdownandoverexpressionsignificantlyreducedandincreasedtheirtu
morsphereformingpotential,respectively.(C)CelllysatesfromtheH441andCL10cellswith
HNMTknockdownandoverexpressionweresubjectedtoWesternblotting,respectively.HNMT,
HER2,HER3,vimentin,andNcadweredetectedusingtheirrespectiveantibodies.Theexperiments
wereconductedfollowingcalibrationusingβ‐actinastheinternalhousekeepingcontrol.Theexper
imentswereanalyzedandnormalizedacrossthreeseparateexperiments.**p<0.01,***p<0.001.
Scalebar:100μm.
2.4.HNMTIstheGeneTargetofMiR223andMiR30655p
WenextexaminedwhethermiRNAmaytargetHNMTusingTARGETSCAN(ver
sion6.2)andobservedthatHNMTcontainedtwocomplementary7mertargetsitesfor
miR223andonesiteformiR30655p(Figure4A).WeevaluatedmiR223andmiR3065
5pexpressioninNSCLCtumorspheresandfoundbothexpressionsweresignificantly
Int.J.Mol.Sci.2022,23,16639of21
decreasedinNSCLCtumorspherescomparedwiththatincontroladherentcells(Figure
4B).Furthermore,theH441cellsweretransfectedwiththemimicsofmiR223,miR3065
5p,orboth.ThecellswereprobedforHNMTandHER2expressionanalysisthroughim
munofluorescence.Asexpected,asinglemiRNAmimicforeithermiR223ormiR3065
5pdecreasedHNMTexpressionandcombiningboththemimicsfurtherdecreased
HNMTexpression(Figure4C).Next,wetransfectedH441cellswiththewildtype(WT)
ormutated(mut)HNMT3UTRdirectedluciferasereporter,andwethencotransfected
thecellswithmiR223,miR30655pmimic,orboth.Throughthisexperiment,wedemon
stratedthatWTHNMT3UTRluciferaseactivitywassuppressedwithmiRNAmimics
cotransfection,butmutHNMT3UTRdidnotsubstantiallyaffectmiRNAmimics.This
resultfurtheraffirmedthatHNMTisthetargetgeneofmiR223ormiR30655p(Figure
4D).WesternblottingalsoyieldedsimilarresultsforHNMT,andHER2expressionsig
nificantlydecreasedwitheithermiR223ormiR30655pmimictransfection,withfurther
reductionwithtransfectionofbothmimics(Figure4E),Rawdataofwesternblotwithfull
sizeblotdisplayedinsupplementaryFigureS3.MiR223andmiR30655pmimictrans
fectionalsonegativelyinfluencedH441colonyformation,whichfurtherreduced
shHNMTcotransfection(Figure4F).
Int.J.Mol.Sci.2022,23,166310of21
Figure4.HNMTisoneofthekeytargetgenesofmiR3065andmiR223inlungcancer.(A)Com
plementarysitesformiR30655pandmiR223inthe3UTRregionofHNMTaccordingtoTAR
GETSCAN(version6.2).(B)miR30655pandmiR223expressioninadherentversussphereH441
cells.RNU62wasusedascontrol.BothqPCRresultswerenormalizedtoadherentH441value.(C)
ImmunofluorescencestainingofHNMTandHER2inH441cellstransfectedwithNCormiR3065
and/ormiR223inhibitordemonstrateddifferentiallocalization.DAPIstaining(blue)wasusedto
labelDNAinallcells.Scalebar=100μm.(D)H441cellswerecotransfectedwithmiR3065,miR
223,orcontrolmimic(miRNC)andwildtype(WT)ormutated(mut)HNMT3UTRdirectedlucif
erasereporter.Luciferaseactivitywasmeasuredusingdualluciferasereporterassays.(E)Western
blottingofHNMTandHER2differentialproteinexpressioninH441cellstransfectedwithNCor
miR3065and/ormiR223inhibitor.(F)H441cellstransfectedwithNCormiR3065and/ormiR223
Int.J.Mol.Sci.2022,23,166311of21
mimicsshowingdifferentialcolonyformationpotential.Graphbarsaremean±SDsofthreeinde
pendentexperiments.*p<0.05,**p<0.01,***p<0.001.Scalebar:100μm.
2.5.HNMTRegulatestheAntioxidativeStressPathwayinNSCLCCells
ROSaccumulationhasbeenemployedasacancerkillingmechanisminchemother
apeuticdevelopment.HER2expressionhasbeenshowntoconferprotectionagainstROS
damageincancercells;thus,targetingHER2activityincancercellsmayincreasetheir
susceptibilitytochemotherapy.WefirstobservedincreasedHER2promoteractivityafter
treatmentwithvariousdosagesofTBHp(50,100,or200μM).Next,wetransfectedH441
cellswithshHNMTandtreatedthemwith200μMTBHp.Asexpected,HER2promoter
activitysignificantlydecreasedincellswithHNMTknockdown(Figure5A).Next,tocon
firmthatHER2andHER3expressionincreasesunderoxidativestressconditions,we
treatedH441cellswith200μMTBHpandfoundthatTBHpincreasedHER2andHER3
expressioncomparedwithuntreatedcells(Figure5B).Wethenevaluatedintracellular
ROSlevelsbyusingDCFDA,afluorescenceprobe,andfoundthatHNMTknockdown
significantlyincreasedROSgeneration(Figure5C).HNMTknockdownalsosignificantly
disruptedtheactivityofHER2viaNRF2/HO1signalingasanalyzedthroughWestern
blotting(Figure5D),rawdataofwesternfullsizeblotshowninsupplementaryFigure
S4.
Figure5.ThenegativeinfluenceofHNMTknockdownonHER2dependentantioxidantresponse
mechanisminlungcancercells.(A)tBHQinducedHER2promotertranscriptionalactivity
Int.J.Mol.Sci.2022,23,166312of21
inductioninaconcentrationdependentmanner.TheH441celllinewastransfectedandthentreated
for4hwithvariousconcentrationsoftBHQ,asindicated.(B)FollowingtBHqtreatment,immunob
lotanalysisshowedtotalproteininductionofbothHER2andHER3.Cellsweretreatedwith200
μMtBHQfor4h,lysedtoextracttheprotein,andanalyzedthroughWesternblotting.(C)Flow
cytometricanalysisoftBHQinducedROSformationinH441cellswithorwithoutshHNMTtrans
fectionusingtheROSsensitivefluorometricprobeDCFDA.RelativeROSproductioninallthe
testedcellswasnormalizedtotheTBH1+shHNMTgroup.(D)Immunoblotanalysisofcellstreated
with200μMtBHqwithorwithoutshHNMTtransfection.NRF2,HO1,pHER2,andHER2were
detectedwiththeirrespectiveantibodies.*p<0.05,**p<0.01,***p<0.001.
2.6.HNMTInhibitionSensitizesNSCLCtoCisplatinChemotherapyBothatInVivoandIn
Vitro
OurinvitroassaysindicatedthatHNMT/HER2signalingplaysavitalroleincancer
stemnessandantioxidantpathways.Thus,wehypothesizedthattargetingHNMTexpres
sionsensitizesNSCLCcellstoconservativechemotherapy.Wefirstcomparedtheviabil
ityofH441cellstransfectedwithshScrambleandshHNMTandtreatedwithanincreasing
cisplatindosage.HNMTknockdownsensitizedH441cellstocisplatintreatment(Figure
6A).WethenevaluatedtheapoptosisofcellsbyusingAnnexinV/PIstainingandnoted
thatH441cellstransfectedwithshHNMThadahighercellapoptosisratecomparedwith
thosetransfectedwithshScramble(Figure6B).Subsequently,theH441cellstransfected
withshHNMTandtreatedwith4μMcisplatinhadgreatlyreducedcolonyformationabil
itycomparedwitheithercisplatinonlyorshHNMTonlycells(Figure6C).Next,weex
trapolatedoursinvitroresultstotheconductedinvivotrials.Afterall,miceweresacrificed
onday22itwasdiscoveredthatshHNMTsensitizedthetumortocisplatintreatment
(Figure6D).However,weidentifiednosignificantweightdifferencebetweenthegroups,
implyingthatshHNMTknockdownwas,atleastpartially,safe(Figure6E).Wetheneval
uatedthetumortissuethroughtheTUNELassay.WenotedthattumorswithHNMT
knockdownandtreatedwithcisplatinhadanincreasedlevelofTUNELstaining,suggest
inganincreasedapoptoticrate(Figure6F).Combined,thesedatasuggestthatshHNMT
hasavitalroletoplayinconferringchemoresistancetoNSCLCcells.
Int.J.Mol.Sci.2022,23,166313of21
Figure6.AssociationofHNMTwithcisplatinresistanceinlungcancer.(A)Cellviability(SRB)as
sayperformedonA549celllinewithorwithoutshHNMTtransfectionfollowedcloselybytreat
mentwithvariousdosagesofcisplatinfor48h.(B)AnnexinVdetectionofapoptosisinH441cells
Int.J.Mol.Sci.2022,23,166314of21
transfectedwithshScrambleorshHNMTandincubatedwithorwithout4μMcisplatinfor48h.
Livecells(lowerleftsquare),deadcells(upperleftsquare),cellsinearlyapoptosis(lowerright
square),andlateapoptoticcells(upperrightsquare).(C)ColonyformationassayofH441cellstrans
fectedwitheithershScrambleorshHNMTandincubatedwithorwithout1μMcisplatinfor10days.
Cellswerethenstainedwithcrystalvioletsolution.(D)Tumorvolumeofmicexenograftedwith
H441cellswitheithershScrambleorshHNMTwasevaluatedevery3daysafterday8oftumor
implantationtoevaluatecisplatintreatmenteffectiveness.(E)Mousebodyweighthasbeenevalu
atedevery3days.(F)RepresentativeimagesoftheTUNELassay(redfluorescenceforapoptotic
cellsandbluefluorescenceforcellnucleihavebeendetectedusingafluorescencemicroscope;mag
nification,400×).Theapoptosisrateisdepictedinthebottompanel.*p<0.05,**p<0.01,***p<
0.001.Scalebar:100μm.
3.Discussion
Here,wediscoveredthattheHNMTwassubstantiallyupregulatedinNSCLCtis
sues,inaccordancewithourfindingsobtainedthroughtheuseofthePrognoscandata
basedata.Furthermore,patientswithhighHNMTexpressionhadtheworstprognosis.
WeexaminedthreedatasetsusingtheR2platform,andwenotedthatHNMTwassignif
icantlyassociatedwithHER2expression.WeconfirmedthisfindingintheclinicalNSCLC
samples,whichexhibitedhigherHNMTandHER2expressioninthetumortissuethanin
thenontumortissue.TofurtherelucidatetheroleofHNMTinNSCLCtumorigenesis,we
employedNSCLCcelllinesforlossof‐andgainoffunctionstudies.Ourcoimmunopre
cipitationdataindicatedthatHNMTinteractedwithHER2,anditsmanipulationaffected
NSCLCcellgrowthrates.IdentifyingtheroleofHNMTintheCSCssubpopulationin
NSCLC,weassessedthecolonyformationabilityofNSCLCcelllineswithHNMTknock
downoroverexpression.HNMTlossorgainsignificantlyreducedorincreasedNSCLC
colonyformation,respectively.Consistently,thecelllines’tumorsphereformationwas
similarlyaffectedbyHNMTloss/gain.CSCsmarkers,suchasKLF4,NANOG,OCT4,and
CD133,wereincreasedanddecreasedfollowingHNMTknockdownandoverexpression,
respectively.Next,weexploredtheregulationofHNMTexpressioninNSCLCbyusing
TARGETSCANdatatodeterminewhichmiRNAmaytargetthismolecule.Resultsindi
catethatthe3UTRregionofHNMThasbeenacomplementarysiteofmiR30655pand
miR223.Notably,miR3065andmiR223hadrelativelylowexpressionintheNSCLC
tumorspherepopulation.UsinginhibitorsandmimicsofbothmiR3065andmiR223,we
observedthatHNMTwasthetargetofbothmiRNAs.
HER2endowscancercellswithrobustantioxidantproperties[8,17].Giventhe
HNMT–HER2interaction,weassessedtheeffectofTBHpinducedoxidativedamageon
NSCLCcellswithHNMTknockdown.Ourluciferasereporterassayconfirmedthatvari
ousTBHpdosagesincreasedHER2andHER3signalintensity,butHNMTknockdown
significantlyreducedtheirintensity.WealsodetectedahigherrateofcellswithDCFDA
staining,implyinghigherROSaccumulation,inH441cellswithHNMTknockdown.Fi
nally,ourWesternblottingresultsindicatedthatshHNMTreducedtheHER2/NRF2/HO
1signalingpathwayactivity.CSCsandcancercells’antioxidativepropertiescontributeto
chemoresistance.HavingproventhatHNMTplaysasignificantroleinCSCsmaintenance
andoxidativestressresponse,wesubsequentlytreatedWTandshHNMTH441cellswith
varieddosagesofcisplatinandevaluatedtheirresponse.HNMTknockdownincreased
H441cells’sensitivitytocisplatintreatment,asevidencedbytheirlowerviabilityand
higherapoptosisrate.Inaddition,cisplatinsignificantlyreducedshHNMTH441colony
formationcomparedwithshScrambleH441.Wethenextrapolatedtheseinvitrofindings
bytransfectingshScrambleandshHNMTH441cellsintoNOD/SCIDmiceandevaluating
theirresponsetocisplatintreatment.MicebearingshHNMTH441cellsweremoresensi
tivetocisplatintreatmentasindicatedbyadelayedgrowthrateandincreasedTUNEL
stainingintensitycomparedwiththeshScramblegroup.
Histaminehasnumerousbiologicalactivities,includingrolesincellproliferationand
differentiation,gastrointestinalfunctionregulation,andimmuneresponsemodulation
[18].ItisalowmolecularweightaminegeneratedsolelybyLHDCandisfoundin
Int.J.Mol.Sci.2022,23,166315of21
variouscellsinthebody,includingthegastricmucosaandparietalandmastcells[19].
HistamineactsbyinteractingwithitsGPCRs—namelyH1HR,H2HR,H3HR,andH4HR.
Whenthesereceptorsareactivatedorinhibited,downstreamsignalingpathwaysaretrig
gered,elicitingimmunemodulatoryandproinflammatorycellresponses[20].Afterhis
tamineisproducedbyHDC,itisrapidlystoredordegradedbyHNMTandmonoamine
oxidaseB[21,22].
TheroleofHNMTincancerremainsunclear.Amicroarraybasedstudyonesopha
gealsquamouscellcarcinomadeterminedthatHNMTisoneofthecrucialplayersincon
trollingsignaltransductionnetworks[23].Inpediatricacutelymphoblasticleukaemia,75
keygeneswereidentified,allofwhichwereconsiderablyenrichedin25GOfunctions
andthechronicmyeloidleukaemiapathway.Subsequently,27diseaserisksubpathways
wereidentifiedwithHNMTasakeygeneenrichedinthesesubpathways[24].B[a]Pisa
wellknownpolycyclicaromatichydrocarbonandacommonpollutantintheatmosphere
thatcancausecancerinbothanimalsandhumans.AstudyconductedinHepG2cells
revealedthatHNMTgeneexpressionsignificantlyincreasedwithB[a]Ptreatment;how
ever,itspreciseroleincarcinogenesisremainsunclear[25].
Inthisstudy,weelucidatedtheroleofHNMTincancer,specificallyinNSCLCcar
cinogenesis.HistaminemetabolismmayplayamajorroleinNSCLCcarcinogenesis
[26,27].ThedirectinteractionofHNMTwithHER2atleastpartiallyexplainsthecarcino
genicroleofHNMT.HER2overexpressionhasbeenobservedinpatientswithmanysolid
cancers,includingNSCLC.AlthoughtheexactmechanismunderlyingHER2–HNMTin
teractionremainsunclear,HNMTmaybeassociatedwithHER2homodimerization,as
indicatedbyYoshiokaetal.[28].TheauthorsalsodemonstratedthatSETandMYND
domaincontainingprotein3(SMYD3),aproteinlysinemethyltransferase,canaffect
HER2homodimerizationandtheactivationofitsdownstreampathwaysbyinducingthe
trimethylationoflysine175residuesinHER2[28].
HER2isawellknownprognosticandpredictivefactorinbreastcancer;however,its
functioninlungcancerwarrantsfurtherclarification[15].HER2mutationscanbefound
inasmallpercentageofpatientswithlungcancer.Theseshiftscanbeinterpretedason
cogenicdriversaswellasamechanismofacquiredresistancefollowingtargetedtherapy.
Similarchangeshavebeenobservedinothercancers,suchasbreastandgastriccancer,
andtheyhavebeenlinkedtopoorprognosisandshortoverallsurvival[29–32].Inaddi
tion,HER2alteredstageIVNSCLCleadstoarelativelyshortoverallsurvival,presuma
blyduetointrinsicresistancetochemotherapy[33].Consistentwithourfindings,HER2
expressionhasalsobeenimplicatedinCSCs’development.Accordingtopreviousdata,
HER2mayenhancecarcinogenesis,invasion,andmetastasisinHER2positivebreastcan
cers,atleastinpart,bysustainingandincreasingCSCs[34,35].Honkanenetal.found
that,inpatientswithNSCLC,HER2modulatestheCSLCphenotypeinALKtranslocated
lungcancers,andthismodulationisprimarilyorchestratedbyHER2/HER3heterodimers
[36].HER2wasalsoimplicatedinCSCsinducedchemoresistance[37].Wangetal.[37]
investigatedtheeffectofHER2ontheinductionofCSCsandthedrugsusceptibilityof
ovariancancercelllines.HER2expressionwascorrelatedwithtumorsphereformation
efficiency,andNF‐κβwasresponsibleforthemediationofHER2inducedCSCs.Further
more,HER2inhibitionsignificantlyincreasedthesensitivityofovariancancercells[37].
Inaddition,CSCsmayconferantioxidantpropertiesthatmayinterferewithchemothera
peuticresponsesincancers[38].Asmentionedabove,weobservedthatHNMTknock
downdisruptstheHER2/NRF2/HO1signalingaxis.Thisisinaccordancewithprevious
studiesthatdemonstratedHER2–NRF2interactioninducesoxaliplatinresistanceincolon
andbreastcancercells[5,8].
CSCscanundergosignificantchangesthatmayaltertheirbiologicalsignature,in
cludingtheirmiRNAexpression.MiRNAsareoftenfoundtobedysregulated,andthey
maystronglyaffecttumorigenicity.Inourstudy,miR223andmiR3065weresignifi
cantlydownregulatedinH441CSCspopulations.Thisfindingvalidatedourpreliminary
findingsbecausewealsodemonstratedthroughTARGETSCANdatabaseminingthat
Int.J.Mol.Sci.2022,23,166316of21
thesetwomiRNAswerehighlycorrelatedwithHNMT.Therefore,thesedata—atleast
partially—explainedwhyHNMTismorehighlyexpressedinsphereenrichedH441cells.
TheroleofmiRNAsinHNMTregulationremainsunderexplored.Astudydemonstrated
thatmiR223mightindirectlyupregulateHNMTexpressioninatopicdermatitispatho
genesis[39].Bycontrast,weobservedthatmiR223inhibitedHNMTexpression.Arecent
reviewelaboratedthecomplexitiesofthismiRNA:itcanactaseitheranoncomiroron
cosuppressor[40].UnlikemiR223,dataconcerningtheroleofmiR3065incancerhave
beenrelativelylimited;however,miR3065wasrecentlyfoundtobeapotentialpredictor
ofdiseaseseverityinovariancancer,alongwithitstargetADH7[41].MiR3065isalso
associatedwithrenalcancercelltumorigenicity[42].Ourstudyaddstoexistingevidence
foramiR3065sroleasanoncosuppressorthatinteractswithHNMTinlungcancer.
4.Methods
4.1.ClinicalSamples
Themicroarraygeneexpressiondatasetsofthelungcancerpatientswereanalyzed
byusingtheonlinePrognoscandatabase[Duke(n=111)]togeneratetheKaplan–Meier
survivalcurve[14].GenecorrelationanalysisofHNMTandHER2wasanalyzedusingR2
GenomicAnalysisandVisualizationPlatform(https://hgserver1.amc.nl/cgi
bin/r2/main.cgi;accessedon21Nov2021)byusingthePeitsch(n=121),EXPO(n=150),
andBild(n=114)lungcancerdatasets.Furthermore,thepatients’clinicalsampleswere
alsocollectedfromMacKayMemorialHospital,TaipeiCity,Taiwan.Allpatientspro
videdwrittenconsentfortheirtissuetobeusedforscientificresearch.Thestudyofpa
tients’sampleswasapprovedbytheMacKayMemorialHospital(Approvalno.:
IRB:20MMHIS500e)andcompliedwiththerecommendationsoftheDeclarationofHel
sinkiforBiomedicalResearch.Inallpatients,lungcancertissuesandnormaltissues>3
cmawayfromthecancerwereobtained.TissuearraysofNSCLCsamplesweresubjected
toimmunohistologicalanalysisafterincubatingwithprimaryantibodiesagainstthe
HNMT(1:100dilution,SC81159;SantaCruzBiotechnology,USA)andHER2(1:100dilu
tion,SC81159;SantaCruzBiotechnology,USA)at4°Covernight.HRPandDABstaining
withhematoxylincounterstainingwereperformedasperthestandardimmunohisto
chemistryprotocolwerefollowedbyimagingandestimationoftheexpressionofthepro
tein.
4.2.CellsandCultureMedium
Thelungcancercelllines,suchasBEAS2B,CL10,CL15,H838,A549,andH441,
werepurchasedfromtheAmericanTypeCultureCollection(ATCC,Manassas,VA,
USA).ThecellswereculturedinDulbecco’smodifiedEagle’smedium(#12491023;
GIBCO,LifeTechnologies,Carlsbad,CA,USA)supplementedwith10%fetalbovinese
rum(GIBCO,LifeTechnologies,Carlsbad,CA,USA),penicillin(100IU/mL),andstrepto
mycin(100g/mL;#15140122,GIBCO,LifeTechnologies,Carlsbad,CA,USA)growninthe
humidified,5%CO2incubatorat37°C.
4.3.CellStableTransfection
TheinformationoftheHNMTexpressionplasmid(OriGeneRC204676)wasusedto
designpolymerasechainreaction(PCR)primers.OligonucleotidesHindIIIHNMT‐F(5′‐
AATTAAGCTTATGGCATCTTCCATGAGGAG3)andMluIHNMTR(5′‐AAT
TACGCGTTGCCTCAATCTCTATG3)weredesignedforPCRamplificationof
HNMTgenesequences.ThesegmentofHNMTwasborneonanemptyplasmid(pCMV
MCSN1EmptyvectorcontrolplasmidDNA,GenBankaccessionU55762).Cell
transfectionwasperformedusingLipofectamine2000(Invitrogen),followingthe
manufacturer’sprotocols.
Eightmicrogramsofemptyplasmid(pCMV6EntryvectorcontrolplasmidDNA,
OriGeneaccessionPS100001)orHNMTexpressionplasmid(pCMV6HNMT,OriGene
Int.J.Mol.Sci.2022,23,166317of21
RC204676,HNMT#1;pCMVHNMTN1,GenBankaccessionU55762,HNMT#2)were
used.TheDNAlipofectaminereagentcomplexesstayatroomtemperaturefor30min.
Themixturewasaddedtothewellandmixedgentlybyrockingtheplatebackandforth.
Reagentcomplexesdidnothavetoberemovedfollowingtransfection.Thecellswere
incubatedat37inaCO2incubatorfor48handassayedfortransgeneexpression.
4.4.CoImmunoprecipitation(CoIP)
CoimmunoprecipitationwasusedtodetectHNMT–HER2interactioninvitro.The
standardCoIPprotocolwasthesameasthatdescribedforIP.Nondenaturinglysis
buffer(20mMTrisHClpH8,137mMNaCl,1%NonidetP40,2mMEDTA)wasstored
at4°C,andimmediatelybeforeuse,proteaseinhibitorswereadded.Thecellculturewas
placedinadishonice,andthecellswerewashedwithicecoldPBS.Thenicecoldlysis
bufferwasadded.Afterthat,cellswerecompletelylysedundernondenaturingcondi
tions,andproteinsthatboundtogetherwerekept.Irrelevant,nonbindingproteins,anti
gens,andanyproteinsthatwereboundwereelutedbyaseriesofwashes.Then,the
boundproteinswhichelutedwereanalyzedbySDSPAGE/immunoblotting.
4.5.WesternBlotting
Lungcancercellswereextractedandlysedaftertrypsinization.Afterthetotalpro
teinslysateswereextractedandthesamplewasprepared,itwasseparatedusingtheSDS
PAGEusingMiniProteanIIIsystem(BioRad,Taiwan)andtransferredontoPVDFmem
branesusingTransBlotTurboTransferSystem(BioRad,Taiwan).Membraneswerein
cubatedovernightat4°CintheprimaryantibodiesshowninSupplementaryTableS1.
SecondaryantibodieswerepurchasedfromSantaCruzBiotechnology(SantaCruz,CA,
USA),andanECLdetectionkitwasusedforthedetectionoftheproteinofinterest.Im
ageswerecapturedandanalyzedusingtheUVPBioDocItsystem(Upland,CA,USA).
4.6.TotalRNAIsolationandQunatitiaveReverseTranscriptionPolymeraseChainReaction
(qRTPCR)
ThetotalRNAwasisolatedandpurifiedusingTRIzolbasedprotocol(Invitrogen,
ThermoFisherScientific,Waltham,MA,USA)accordingtotheprotocolprovidedbythe
manufacturer.TwomicrogramoftotalRNAwasreversetranscribedusingQIAGEN
OneStepRTPCRKit(QIAGEN,Taiwan),andthePCRreactionwasperformedusinga
RotorGeneSYBRGreenPCRKit(400,QIAGEN,Taiwan).HNMTmRNAexpressionwas
detectedinlungcancerandnormaltissues.Theprimersequencesusedwereasfollows:
HNMTamplification(452bp),5′‐TACGTCCAAGGTCGGGCAGGAAGA3;upstream,
5′‐CACTGATAGGCAGTTCTC;downstream,5′‐GGTTCTCAGTTGGTGCTTC.Glycer
aldehyde3phosphatedehydrogenase(GAPDH)wasusedasaninternalreferencetode
tectHNMTmRNAexpressionlevels.TherelativeexpressionlevelofHNMTmRNAwas
calculatedusing2−∆∆Cqformulae.
4.7.ClonogenicAssay
Fortheassessmentofthesensitivityofcancercellstowardsanytreatment,the“Clon
ogenicAssay”isagoldstandard.Atotalof2.7×104cellsperwellwereseededina6well
plateandincubatedat37°Cfor2days.Further,thecellswereculturedforanadditional
24hinmediaandincubatedat37°Cfor2daysin5%CO2treated.Thecellswerethen
subculturedandseededat350cellsperwellintonew6wellplatesandkeptforincubation
for10daysat37°Cinahumidifiedincubatorwith5%CO2.Thecellswerefixedanddried
afterbeingsetandstainedwith0.1%crystalviolet.Theexperimentswereconductedin
triplicate.
Int.J.Mol.Sci.2022,23,166318of21
4.8.SulforhodamineBAssay
CellularviabilitywasdeterminedusingthesulforhodamineB(SRB)assayasperthe
protocolsuggestedbyourlabprotocol.Briefly,lungcancercellswereseededin96well
plates(3.5×105cells/well),followedbyincubationat37°C,inahumidified5%CO2incu
bator.Thecellswerefixedwiththegentleadditionof50mLofcold10%w/vtricarboxylic
acid(TCA)andincubatedat4°Cfor60min.Next,50μLof0.4%w/vSBRsolutionin1%
CH3COOHwasaddedtoeachwell,followedbyincubationatroomtemperaturefor20
min.Unbounddyewasrecoveredafterstaining,andresidualdyewasremovedbythor
oughlywashingwellplateswith1%CH3COOHandairdrying.Theboundstainwas
dissolvedin10mMTrizmabase,andtheabsorbancewasmeasuredat515nmonan
ELISAplatereader(690nmreferencewavelength).
4.9.ImmunofluorescenceAssay
H441cellswereculturedonglasscoverslipsbeforebeingtransfected,asmentioned
previously.Afterincubation,cellswerefixedfor15minat4°Cwith4%formaldehyde,
permeabilizedfor5minwith0.01%TritonX100andblockedfor30minatroomtemper
aturewith1%bovineserumalbumin.Thecellswerethenincubatedfor24hat4°Cwith
primaryantibodiesagainstGSK3(#12456,1:100,CellSignalingTechnology,Danvers,
MA,USA)andβ‐catenin(#8814,1:100,CellSignalingTechnology).Thecellswerestained
withanisotypespecificsecondaryantibody(AlexaFluor594AffiniPuredonkeyanti
rabbitIgG;JacksonImmunoResearch,WestGrove,PA,USA)for1hthefollowingday.
4.10.ROSProductionMeasurement
ForROSproductionevaluation,cancercellswereplatedin96wellplatesat20,000
cellsperwellinafinalvolumeof80μLofthemedium.Next,10μLof50μMDCFDAwas
addedtoeachwell,andthecellswereincubatedfor30min.Onamicroplatefluorometer
(Tecan,Seestrasse,Männedorf,Switzerland),fluorescencewasmeasuredwithanexcita
tionfiltersetat488nmandanemissionfiltersetat530nm.
4.11.TumorXenograftStudy
FourtosixweekoldfemaleNOD/SCIDmice(meanweight=17.4±2.1g)werepur
chasedfromBioLASCOTaiwan(Taipei,Taiwan).Theinvivostudieswereapprovedby
theInstitutionalAnimalCareandUseCommittee(IACUC)oftheMacKayMemorialHos
pital(Approvalno.:MMHAS10910).Themice(n=10each)wererandomlyassigned
totheshScramble,shScramble+cisplatin,shHNMT,orshHNMT+cisplatingroupsafter
receivinganinjectionof2×106H441cellswithscrambleshRNA(shScramble)orHNMT
shRNA(shHNMT)intheirhindflanks.Whenthetumorswerepalpableonday8,2mg/kg
cisplatinwasadministeredintraperitoneallyevery72hfor12days.Tumorsizeswerede
terminedwithcallipersevery3daysondays6,9,12,15,and18,andtumorvolumes(v)
wereestimatedusingtheformulalength(l)×(width(w))2×0.5.Thetumorbearingmice
werehumanelysacrificedattheendofthetrialonday18,andthetumorswereremoved,
tested,photographed,andweighedagain.
4.12.StatisticalAnalysis
Themeansandstandarddeviations(SDs)wereusedtopresentallresults.Formul
tiplecomparisonsorrepeatedmeasurements,Student’sttestwasused.Formultiplecom
parisonsorrepeatedmeasurements,ANOVAorrepeatedANOVAaccompaniedby
Tukey’sposthoctestwasused.Statisticalsignificancewasdefinedasp<0.05.GraphPad
Prism(version7;GraphPadSoftware,SanDiego,CA,USA)wasusedforallstatistical
analyses.
Int.J.Mol.Sci.2022,23,166319of21
5.Conclusions
OurresultsrevealedthatHNMTupregulationinNSCLCcellsleadstoHER2upreg
ulation,whichinturnincreasestumorigenicityandchemoresistancethroughCSCs
maintenanceandantioxidantproperties.ThisCSCsmaydownregulatemiR30655pand
miR223expression,thusreducingtheinhibitionoftheirtargetgene,HNMT,therebyre
sultinginafeedbackloopthatmayaidinmaintainingtheCSCspopulationofNSCLC
andconferringchemoresistance(Figure7).Theseresultsprovidenovelinsightintothe
rolesandinteractionsofHNMT,HER2,andmiRNAsinNSCLCpathogenesisandbehav
iour.TargetingthisnewlydiscoveredregulatoryaxismayaidinretardingNSCLCpro
gressionandcombatingchemoresistance.
Figure7.Graphicalsummary.HNMTinteractswithHER2toinduceCSCsphenotypedevelopment
andenhanceNSCLCantioxidantproperties,whichinturnattenuatemiR223/3065transcription
thatloopsbacktoincreasedHNMTexpression.HNMT/HER2inducedCSCsandantioxidanten
hancementsubsequentlyresultedincisplatinchemoresistanceinNSCLCs.
SupplementaryMaterials:Thefollowingsupportinginformationcanbedownloadedat:
www.mdpi.com/article/10.3390/ijms23031663/s1.SupplementaryTableS1.primaryantibodiesof
Westernblots.WesternBlotRawData:SupplementaryFigureS1.FullsizeblotsofFigure2.A,B,
C,andD,SupplementaryFigureS2.FullsizeblotsofFigure3C,SupplementaryFigureS3.Fullsize
blotsofFigure4E,SupplementaryFigureS4.FullsizeblotsofFigure5B,andD.
AuthorContributions:K.T.K.,C.H.L.,C.H.W.,N.W.P.,andV.K.Y.:Studyconceptionanddesign,
collection,andassemblyofdata,dataanalysisandinterpretation,andmanuscriptwriting.I.H.F.:
Dataanalysisandinterpretation.C.T.Y.,W.H.L.,andW.C.H.:Studyconceptionanddesign,data
analysisandinterpretation,finalmanuscriptapproval.Allauthorshavereadandagreedtothepub
lishedversionofthemanuscript.
Int.J.Mol.Sci.2022,23,166320of21
Funding:Pleaseadd:ThisresearchreceivednoexternalfundingorThisresearchwasfundedby
Dr.KuangTaiKuograntnumber[MinistryofScienceandTechnology:MOST1082314B038‐114
MY3].
InstitutionalReviewBoardStatement:Allenrolledlungcancerpatientsprovidedwrittenin
formedconsentfortheirtissuestobeusedforscientificresearch.Thestudywasapprovedbythe
MacKayMemorialHospital(Approvalno.:IRB:20MMHIS500e)andcompliedwiththerecommen
dationsoftheDeclarationofHelsinkiforBiomedicalResearch.Theinvivostudieswereapproved
bytheInstitutionalAnimalCareandUseCommittee(IACUC)oftheMacKayMemorialHospital
(Approvalno.:MMHAS10910).
InformedConsentStatement:Anyresearcharticledescribingastudyinvolvinghumansshould
containthisstatement.Writteninformedconsenthasbeenobtainedfromthepatient(s)topublish
thispaper.
DataAvailabilityStatement:Thedatasetsusedandanalyzedinthecurrentstudyarepubliclyac
cessible,asindicatedinthemanuscript.
Acknowledgments:TheauthorsthankallresearchassistantsoftheCancerTranslationalResearch
LaboratoryandCoreFacilityCenter,TaipeiMedicalUniversityShuangHoHospital,fortheirassis
tancewiththemolecularandcellbasedassays.
ConflictsofInterest:Theauthorsdeclarethattheyhavenopotentialfinancialcompetinginterests.
References
1. Subramanian,J.;Morgensztern,D.;Goodgame,B.;Baggstrom,M.Q.;Gao,F.;Piccirillo,J.;Govindan,R.Distinctive
characteristicsofnonsmallcelllungcancer(NSCLC)intheyoung:Asurveillance,epidemiology,andendresults(SEER)
analysis.J.Thorac.Oncol.2010,5,23–28.https://doi.org/10.1097/JTO.0b013e3181c41e8d.
2. Chang,A.Chemotherapy,chemoresistanceandthechangingtreatmentlandscapeforNSCLC.LungCancer2011,71,3–10.
https://doi.org/10.1016/j.lungcan.2010.08.022.
3. Zhong,G.;Qin,S.;Townsend,D.;Schulte,B.A.;Tew,K.D.;Wang,G.Y.Oxidativestressinducessenescenceinbreastcancer
stemcells.Biochem.Biophys.Res.Commun.2019,514,1204–1209.https://doi.org/10.1016/j.bbrc.2019.05.098.
4. Kharkar,P.S.CancerStemCell(CSC)InhibitorsinOncologyAPromiseforaBetterTherapeuticOutcome:StateoftheArtand
FuturePerspectives.J.Med.Chem.2020,63,15279–15307.https://doi.org/10.1021/acs.jmedchem.0c01336.
5. PirpourTazehkand,A.;Akbarzadeh,M.;Velaie,K.;Sadeghi,M.R.;Samadi,N.TheroleofHer2Nrf2axisininductionof
oxaliplatinresistanceincoloncancercells.Biomed.Pharm.2018,103,755–766.https://doi.org/10.1016/j.biopha.2018.04.105.
6. Chio,I.I.C.;Tuveson,D.A.ROSinCancer:TheBurningQuestion.TrendsMol.Med.2017,23,411–429.
https://doi.org/10.1016/j.molmed.2017.03.004.
7. Zipper,L.M.;Mulcahy,R.T.InhibitionofERKandp38MAPkinasesinhibitsbindingofNrf2andinductionofGCSgenes.
Biochem.Biophys.Res.Commun.2000,278,484–492.https://doi.org/10.1006/bbrc.2000.3830.
8. Kang,H.J.;Yi,Y.W.;Hong,Y.B.;Kim,H.J.;Jang,Y.J.;Seong,Y.S.;Bae,I.HER2confersdrugresistanceofhumanbreastcancer
cellsthroughactivationofNRF2bydirectinteraction.Sci.Rep.2014,4,7201.https://doi.org/10.1038/srep07201.
9. Brown,R.E.;Stevens,D.R.;Haas,H.L.Thephysiologyofbrainhistamine.Prog.Neurobiol.2001,63,637–672.
https://doi.org/10.1016/s03010082(00)000393.
10. Haas,H.L.;Panula,P.Histaminereceptors.Neuropharmacology2016,106,1–2.https://doi.org/10.1016/j.neuropharm.2016.04.007.
11. Leurs,R.;Smit,M.J.;Timmerman,H.Molecularpharmacologicalaspectsofhistaminereceptors.Pharmacol.Ther.1995,66,413–
463.https://doi.org/10.1016/01637258(95)000063.
12. vonMachSzczypinski,J.;Stanosz,S.;Sieja,K.;Stanosz,M.Histamineanditsmetabolizingenzymesintissuesofprimaryductal
breastcancer.Eur.J.Gynaecol.Oncol.2009,30,509–511.
13. Tanaka,S.;Sakaguchi,M.;Yoneyama,H.;Usami,Y.;Harusawa,S.HistamineH3receptorantagonistOUP186attenuatesthe
proliferationofculturedhumanbreastcancercelllines.Biochem.Biophys.Res.Commun.2016,480,479–485.
https://doi.org/10.1016/j.bbrc.2016.10.077.
14. Mizuno,H.;Kitada,K.;Nakai,K.;Sarai,A.PrognoScan:Anewdatabaseformetaanalysisoftheprognosticvalueofgenes.
BMCMed.Genom.2009,2,18.https://doi.org/10.1186/17558794218.
15. Ricciardi,G.R.;Russo,A.;Franchina,T.;Ferraro,G.;Zanghi,M.;Picone,A.;Scimone,A.;Adamo,V.NSCLCandHER2:Between
lightsandshadows.J.Thorac.Oncol.2014,9,1750–1762.https://doi.org/10.1097/JTO.0000000000000379.
16. Koster,J.R2:GenomicsAnalysisandVisualizationPlatform.Availabeonline:http://r2.amc.nl;21Nov2021.
17. Guda,R.;Kumar,G.;Korra,R.;Balaji,S.;Dayakar,G.;Palabindela,R.;Myadaraveni,P.;Yellu,N.R.;Kasula,M.EGFR,HER2
targetbasedmoleculardockinganalysis,invitroscreeningof2,4,5trisubstitutedimidazolederivativesaspotentialanti
oxidantandcytotoxicagents.J.Photochem.Photobiol.B2017,176,69–80.https://doi.org/10.1016/j.jphotobiol.2017.09.010.
18. PinoAngeles,A.;ReyesPalomares,A.;Melgarejo,E.;SanchezJimenez,F.Histamine:Anundercoveragentinmultiplerare
diseases?J.Cell.Mol.Med.2012,16,1947–1960.https://doi.org/10.1111/j.15824934.2012.01566.x.
Int.J.Mol.Sci.2022,23,166321of21
19. Francis,H.;DeMorrow,S.;Venter,J.;Onori,P.;White,M.;Gaudio,E.;Francis,T.;Greene,J.F.,Jr.;Tran,S.;Meininger,C.J.;et
al.Inhibitionofhistidinedecarboxylaseablatestheautocrinetumorigeniceffectsofhistamineinhumancholangiocarcinoma.
Gut2012,61,753–764.https://doi.org/10.1136/gutjnl2011300007.
20. Shan,L.;Bao,A.M.;Swaab,D.F.ChangesinHistidineDecarboxylase,HistamineNMethyltransferaseandHistamineReceptors
inNeuropsychiatricDisorders.Handb.Exp.Pharmacol.2017,241,259–276.https://doi.org/10.1007/164_2016_125.
21. Krystal,A.D.;Richelson,E.;Roth,T.ReviewofthehistaminesystemandtheclinicaleffectsofH1antagonists:Basisforanew
modelforunderstandingtheeffectsofinsomniamedications.SleepMed.Rev.2013,17,263–272.
https://doi.org/10.1016/j.smrv.2012.08.001.
22. Schwelberger,H.G.;Feurle,J.;Houen,G.MappingofthebindingsitesofhumanhistamineNmethyltransferase(HNMT)
monoclonalantibodies.Inflamm.Res.2017,66,1021–1029.https://doi.org/10.1007/s0001101710867.
23. Xu,C.Q.;Zhu,S.T.;Wang,M.;Guo,S.L.;Sun,X.J.;Cheng,R.;Xing,J.;Wang,W.H.;Shao,L.L.;Zhang,S.T.Pathwayanalysisof
differentiallyexpressedgenesinhumanesophagealsquamouscellcarcinoma.Eur.Rev.Med.Pharmacol.Sci.2015,19,1652–1661.
24. Gao,H.Y.;Luo,X.G.;Chen,X.;Wang,J.H.Identificationofkeygenesaffectingdiseasefreesurvivaltimeofpediatricacute
lymphoblasticleukemiabasedonbioinformaticanalysis.BloodCellsMol.Dis.2015,54,38–43.
https://doi.org/10.1016/j.bcmd.2014.08.002.
25. Roh,T.;Kwak,M.Y.;Kwak,E.H.;Kim,D.H.;Han,E.Y.;Bae,J.Y.;Bangdu,Y.;Lim,D.S.;Ahn,I.Y.;Jang,D.E.;etal.
Chemopreventivemechanismsofmethionineoninhibitionofbenzo(a)pyreneDNAadductsformationinhuman
hepatocellularcarcinomaHepG2cells.Toxicol.Lett.2012,208,232–238.https://doi.org/10.1016/j.toxlet.2011.11.013.
26. Xue,L.F.;Cheng,J.;Wang,J.W.;Song,Y.;Gu,S.X.Histamineandlungcancer.II.Humanlungcancercelllineandhistamine.
ZhonghuaZhongLiuZaZhi1988,10,19–22.
27. Kondratenko,T.Y.;Zacharova,I.V.;Katukov,V.;Kuzina,N.V.;Severin,E.S.;Kornilova,Z.;Perelman,M.I.Thestudyof
histamineH1‐andH2receptorsinhumanlungcancer.Biochem.Mol.Biol.Int.1993,31,399–404.
28. Yoshioka,Y.;Suzuki,T.;Matsuo,Y.;Tsurita,G.;Watanabe,T.;Dohmae,N.;Nakamura,Y.;Hamamoto,R.Proteinlysine
methyltransferaseSMYD3isinvolvedintumorigenesisthroughregulationofHER2homodimerization.CancerMed.2017,6,
1665–1672.https://doi.org/10.1002/cam4.1099.
29. Slamon,D.J.;Clark,G.M.;Wong,S.G.;Levin,W.J.;Ullrich,A.;McGuire,W.L.Humanbreastcancer:Correlationofrelapseand
survivalwithamplificationoftheHER2/neuoncogene.Science1987,235,177–182.https://doi.org/10.1126/science.3798106.
30. Nakamura,H.;Kawasaki,N.;Taguchi,M.;Kabasawa,K.AssociationofHER2overexpressionwithprognosisinnonsmallcell
lungcarcinoma:Ametaanalysis.Cancer2005,103,1865–1873.https://doi.org/10.1002/cncr.20957.
31. Liu,L.;Shao,X.;Gao,W.;Bai,J.;Wang,R.;Huang,P.;Yin,Y.;Liu,P.;Shu,Y.Theroleofhumanepidermalgrowthfactor
receptor2asaprognosticfactorinlungcancer:Ametaanalysisofpublisheddata.J.Thorac.Oncol.2010,5,1922–1932.
https://doi.org/10.1097/jto.0b013e3181f26266.
32. Shitara,K.;Yatabe,Y.;Matsuo,K.;Sugano,M.;Kondo,C.;Takahari,D.;Ura,T.;Tajika,M.;Ito,S.;Muro,K.Prognosisofpatients
withadvancedgastriccancerbyHER2statusandtrastuzumabtreatment.GastricCancer2013,16,261–267.
https://doi.org/10.1007/s1012001201799.
33. Mazieres,J.;Barlesi,F.;Filleron,T.;Besse,B.;Monnet,I.;BeauFaller,M.;Peters,S.;Dansin,E.;Fruh,M.;Pless,M.;etal.Lung
cancerpatientswithHER2mutationstreatedwithchemotherapyandHER2targeteddrugs:ResultsfromtheEuropean
EUHER2cohort.Ann.Oncol.2016,27,281–286.https://doi.org/10.1093/annonc/mdv573.
34. Korkaya,H.;Paulson,A.;Iovino,F.;Wicha,M.S.HER2regulatesthemammarystem/progenitorcellpopulationdriving
tumorigenesisandinvasion.Oncogene2008,27,6120–6130.https://doi.org/10.1038/onc.2008.207.
35. Magnifico,A.;Albano,L.;Campaner,S.;Delia,D.;Castiglioni,F.;Gasparini,P.;Sozzi,G.;Fontanella,E.;Menard,S.;Tagliabue,
E.TumorinitiatingcellsofHER2positivecarcinomacelllinesexpressthehighestoncoproteinlevelsandaresensitiveto
trastuzumab.Clin.CancerRes.2009,15,2010–2021.https://doi.org/10.1158/10780432.CCR081327.
36. Honkanen,T.;Wilenius,E.;Koivunen,P.;Koivunen,J.P.HER2regulatescancerstemlikecellphenotypeinALKtranslocated
NSCLC.Int.J.Oncol.2017,51,599–606.https://doi.org/10.3892/ijo.2017.4048.
37. Wang,W.;Gao,Y.;Hai,J.;Yang,J.;Duan,S.HER2decreasesdrugsensitivityofovariancancercellsviainducingstemcelllike
propertyinanNFkappaBdependentway.Biosci.Rep.2019,39.https://doi.org/10.1042/BSR20180829.
38. Suzuki,S.;Okada,M.;Shibuya,K.;Seino,M.;Sato,A.;Takeda,H.;Seino,S.;Yoshioka,T.;Kitanaka,C.JNKsuppressionof
chemotherapeuticagentsinducedROSconferschemoresistanceonpancreaticcancerstemcells.Oncotarget2015,6,458–470.
https://doi.org/10.18632/oncotarget.2693.
39. Jia,H.Z.;Liu,S.L.;Zou,Y.F.;Chen,X.F.;Yu,L.;Wan,J.;Zhang,H.Y.;Chen,Q.;Xiong,Y.;Yu,B.;etal.MicroRNA223isinvolved
inthepathogenesisofatopicdermatitisbyaffectinghistamineNmethyltransferase.CellMol.Biol.2018,64,103–107.
https://doi.org/10.14715/cmb/2018.64.3.17.
40. Favero,A.;Segatto,I.;Perin,T.;Belletti,B.ThemanyfacetsofmiR223incancer:Oncosuppressor,oncogenicdriver,therapeutic
target,andbiomarkerofresponse.WileyInterdiscip.Rev.RNA2021,e1659.https://doi.org/10.1002/wrna.1659.
41. Ding,S.;Huang,X.;Zhu,J.;Xu,B.;Xu,L.;Gu,D.;Zhang,W.ADH7,miR3065andLINC01133areassociatedwithcervical
cancerprogressionindifferentagegroups.Oncol.Lett.2020,19,2326–2338.https://doi.org/10.3892/ol.2020.11348.
42. Muller,S.;Nowak,K.ExploringthemiRNAmRNAregulatorynetworkinclearcellrenalcellcarcinomasbynextgeneration
sequencingexpressionprofiles.BioMedRes.Int.2014,2014,948408.https://doi.org/10.1155/2014/948408.
... Recent work suggests that histamine N-methyltransferase (HNMT), which is regulated by ROS, is co-expressed with human epidermal growth factor receptor (HER2) [95]. The expression of both HER2 and HNMT is increased in breast and lung cancer [95,96]. ...
... Recent work suggests that histamine N-methyltransferase (HNMT), which is regulated by ROS, is co-expressed with human epidermal growth factor receptor (HER2) [95]. The expression of both HER2 and HNMT is increased in breast and lung cancer [95,96]. HNMT directly interacts with HER2 to regulate NRF2 DNA binding activity and target gene expression, which is in part mediated by phosphatidylinositol-4,5-bisphosphate 3-kinase (PI3K) [95]. ...
... The expression of both HER2 and HNMT is increased in breast and lung cancer [95,96]. HNMT directly interacts with HER2 to regulate NRF2 DNA binding activity and target gene expression, which is in part mediated by phosphatidylinositol-4,5-bisphosphate 3-kinase (PI3K) [95]. In non-small-cell lung cancer (NSCLC) CSCs, expression levels of the HNMTinhibiting miRs, miR3065/223, are low, leading to elevated expression/activity of NRF2. ...
Article
Full-text available
Cancer stem cells (CSCs) represent a small subset of slowly dividing cells with tumor-initiating ability. They can self-renew and differentiate into all the distinct cell populations within a tumor. CSCs are naturally resistant to chemotherapy or radiotherapy. CSCs, thus, can repopulate a tumor after therapy and are responsible for recurrence of disease. Stemness manifests itself through, among other things, the expression of stem cell markers, the ability to induce sphere formation and tumor growth in vivo, and resistance to chemotherapeutics and irradiation. Stemness is maintained by keeping levels of reactive oxygen species (ROS) low, which is achieved by enhanced activity of antioxidant pathways. Here, cellular sources of ROS, antioxidant pathways employed by CSCs, and underlying mechanisms to overcome resistance are discussed.
... Although several therapies such as chemotherapy and surgical resection are used to decrease mortality, there are still dilemmas including late detection for approximately 75% of LUAD patients, as well as high recurrence rates and poor prognoses [5,6]. Therefore, it is imperative to explore potential biomarkers and develop novel targeted cancer therapies for future application [7][8][9][10][11]. ...
Article
Full-text available
The complexity of lung adenocarcinoma (LUAD), the development of which involves many interacting biological processes, makes it difficult to find therapeutic biomarkers for treatment. FK506-binding proteins (FKBPs) are composed of 12 members classified as conservative intracellular immunophilin family proteins, which are often connected to cyclophilin structures by tetratricopeptide repeat domains and have peptidyl prolyl isomerase activity that catalyzes proline from residues and turns the trans form into the cis form. Since FKBPs belong to chaperone molecules and promote protein folding, previous studies demonstrated that FKBP family members significantly contribute to the degradation of damaged, misfolded, abnormal, and foreign proteins. However, transcript expressions of this gene family in LUAD still need to be more fully investigated. In this research, we adopted high-throughput bioinformatics technology to analyze FKBP family genes in LUAD to provide credible information to clinicians and promote the development of novel cancer target drugs in the future. The current data revealed that the messenger (m)RNA levels of FKBP2, FKBP3, FKBP4, FKBP10, FKBP11, and FKBP14 were overexpressed in LUAD, and FKBP10 had connections to poor prognoses among LUAD patients in an overall survival (OS) analysis. Based on the above results, we selected FKBP10 to further conduct a comprehensive analysis of the downstream pathway and network. Through a DAVID analysis, we found that FKBP10 was involved in mitochondrial electron transport, NADH to ubiquinone transport, mitochondrial respiratory chain complex I assembly, etc. The MetaCore pathway analysis also indicated that FKBP10 was involved in "Ubiquinone metabolism", "Translation_(L)-selenoaminoacid incorporation in proteins during translation", and "Transcription_Negative regulation of HIF1A function". Collectively, this study revealed that FKBP family members are both significant prognostic biomarkers for lung cancer progression and promising clinical therapeutic targets, thus providing new targets for treating LUAD patients.
... In addition, HNMT upregulation Frontiers in Nutrition 13 frontiersin.org causes cancer stem cell formation and protect it from oxidative stress by interaction with HER2 in NSCLC (41). TFEB (Transcription Factor EB) enables DNA-binding transcription factor activity. ...
Article
Full-text available
Background Amino acid (AA) metabolism plays a crucial role in cancer. However, its role in acute myeloid leukemia (AML) is still unavailable. We screened out AA metabolic genes, which related to prognosis, and analyzed their correlation with tumor immune microenvironment in AML. Methods We evaluated 472 amino acid metabolism-related genes in 132 AML patients. The predictive risk model was developed according to differentially expressed genes, univariate Cox and LASSO analyses. We validated the risk signature by survival analysis and independence tests. Single-sample gene set enrichment analysis (ssGSEA), tumor immune microenvironment (TME), tumor mutation burden (TMB), functional enrichment, and the IC50 of drugs were assessed to explore the correlations among the risk model, immunity, and drug sensitivity of AML. Results Six amino acid metabolism-related genes were confirmed to develop the risk model, including TRH, HNMT, TFEB, SDSL, SLC43A2, and SFXN3. The high-risk subgroup had an immune “hot” phenotype and was related to a poor prognosis. The high-risk group was also associated with more activity of immune cells, such as Tregs, had higher expression of some immune checkpoints, including PD1 and CTLA4, and might be more susceptible to immunotherapy. Xenobiotic metabolism, the reactive oxygen species (ROS) pathway, fatty acid metabolism, JAK/STAT3, and the inflammatory response were active in the high-risk subgroup. Furthermore, the high-risk subgroup was sensitive to sorafenib, selumetinib, and entospletinib. ssGSEA discovered that the processes of glutamine, arginine, tryptophan, cysteine, histidine, L-serine, isoleucine, threonine, tyrosine, and L-phenylalanine metabolism were more active in the high-risk subgroup. Conclusion This study revealed that AA metabolism-related genes were correlated with the immune microenvironment of AML patients and could predict the prognosis and immunotherapy response of AML patients.
... Zhong and their colleagues revealed that the TM6SF1 was related to the NSCLC tumor Microenvironment (34). Kuo and their colleagues found that the upregulation of HNMT could induce tumor stemness in NSCLC (35). MRO and CAPZA2 have not been reported in NSCLC. ...
Article
Full-text available
Background The progression process of lung cancer can be accelerated by M2 macrophages. However, genes that affect M2 macrophage polarization remain unidentified. Methods The Cancer Genome Atlas, Gene Expression Omnibus, and Arrayexpress databases were used to obtain open-access data. The analysis of public data was mostly performed with R studio. The RNA levels of specific genes were detected using quantitative real-time PCR. The proliferation ability of the cells was assessed by CCK8, colony formation, and EdU assays. Results Based on the multiple datasets, we noticed a poor prognosis in patients with high M2 macrophage infiltration. There were 114 genes differentially expressed between high and low M2 macrophages infiltrated samples, regarded as M2 macrophage-related genes. Subsequently, a prognosis prediction signature consisting of ABHD5, HS3ST2, TM6SF1, CAPZA2, LEPROT, HNMT, and MRO was identified and presented a satisfactory performance. The pathway enrichment results revealed a positive correlation between riskscore and enrichment scores for most immunotherapy-related positive terms. Also, there might be an increase in genomic instability among patients at high risk. Interestingly, low risk patients are most likely to benefit from PD-1 therapy, while high risk patients may benefit from CTLA-4 therapy. Meanwhile, the estimated IC50 of seven drugs differs significantly between two risk groups, including Cisplatin, Docetaxel, Doxorubicin, Gefitinib, Paclitaxel, Sunitinib and Vinorelbine. Moreover, further experiments indicated that HNMT was overexpressed and can enhance the proliferation ability in lung cancer cells. Conclusions In summary, our study identified the molecules significantly affecting M2 macrophage infiltration and identified a prognosis signature that robustly indicated patients prognosis. Moreover, we validated the cancer-promoting effect of HNMT using in vitro experiments.
... Once cell-cell adhesion is disturbed, adhesionrelated pathways are subsequently interfered with. Disruption of cadherin signaling has determining influence on tumor progression and tumor immune responses [99][100][101][102][103][104]. ...
Article
Full-text available
Breast cancer is one of the leading deaths in all kinds of malignancies; therefore, it is important for early detection. At the primary tumor site, tumor cells could take on mesenchymal properties, termed the epithelial-to-mesenchymal transition (EMT). This process is partly regulated by members of the cadherin (CDH) family of genes, and it is an essential step in the formation of metastases. There has been a lot of study of the roles of some of the CDH family genes in cancer; however, a holistic approach examining the roles of distinct CDH family genes in the development of breast cancer remains largely unexplored. In the present study, we used a bioinformatics approach to examine expression profiles of CDH family genes using the Oncomine, Gene Expression Profiling Interactive Analysis 2 (GEPIA2), cBioPortal, MetaCore, and Tumor IMmune Estimation Resource (TIMER) platforms. We revealed that CDH1/2/4/11/12/13 messenger (m)RNA levels are overexpressed in breast cancer cells compared to normal cells and were correlated with poor prognoses in breast cancer patients' distant metastasis-free survival. An enrichment analysis showed that high expressions of CDH1/2/4/11/12/13 were significantly correlated with cell adhesion, the extracellular matrix remodeling process, the EMT, WNT/beta-catenin, and interleukin-mediated immune responses. Collectively, CDH1/2/4/11/12/13 are thought to be potential biomarkers for breast cancer progression and metastasis.
... Moreover, HNMT has a positive relationship with HER2 that could improve the features of CSCs. The knockdown of HNMT could decrease the tumorsphere formation ability, and reduce the expression of CSC markers such as NANOG, CD133, OCT4, and KLF4 through the Nrf2/HO-1/ HER2 signaling pathway increasing the accumulation of reactive oxygen species (ROS) (79). The stemness markers ALDH and CD133 are well-verified in LCSCs; p53 is a cancer suppressor, the mutation which is found in 47% of NSCLC cases, and the knockdown of the three genes could reduce the CSC characteristics and prolong the survival of NSCLC patients (80). ...
Article
Full-text available
Despite improved methods of diagnosis and the development of different treatments, mortality from lung cancer remains surprisingly high. Non-small cell lung cancer (NSCLC) accounts for the large majority of lung cancer cases. Therefore, it is important to review current methods of diagnosis and treatments of NSCLC in the clinic and preclinic. In this review, we describe, as a guide for clinicians, current diagnostic methods and therapies (such as chemotherapy, chemoradiotherapy, targeted therapy, antiangiogenic therapy, immunotherapy, and combination therapy) for NSCLC.
Article
Full-text available
Despite the treatment of lung adenocarcinoma (LUAD) having partially improved in recent years, LUAD patients still have poor prognosis rates. Therefore, it is especially important to explore effective biomarkers and exploit novel therapeutic developments. High-throughput technologies are widely used as systematic approaches to explore differences in expressions of thousands of genes for both biological and genomic systems. Recently, using big data analyses in biomedicine research by integrating several high-throughput databases and tools, including The Cancer Genome Atlas (TCGA), cBioportal, Oncomine, and Kaplan–Meier plotter, is an important strategy to identify novel biomarkers for cancer therapy. Here, we used two different comprehensive bioinformatics analysis and revealed protein tyrosine phosphatase non-receptor type (PTPN) family genes, especially PTPN1 and PTPN22, were downregulated in lung cancer tissue in comparison with normal samples. The survival curves indicated that LUAD patients with high transcription levels of PTPN5 were significantly associated with a good prognosis. Meanwhile, Gene Ontology (GO) and MetaCore analyses indicated that co-expression of the PTPN1, PTPN5, and PTPN21 genes was significantly enriched in cancer development-related pathways, including GTPase activity, regulation of small GTPase-mediated signal transduction, response to mechanical stimuli, vasculogenesis, organ morphogenesis, regulation of stress fiber assembly, mitogen-activated protein kinase (MAPK) cascade, cell migration, and angiogenesis. Collectively, this study revealed that PTPN family members are both significant prognostic biomarkers for lung cancer progression and promising clinical therapeutic targets, which provide new targets for treating LUAD patients.
Article
Full-text available
The period (PER) and cryptochrome (CRY) families play critical roles in circadian rhythms. The imbalance of circadian factors may lead to the occurrence of cancer. Expressions of PER and CRY family members decrease in various cancers. Nevertheless, expression levels, genetic variations, and molecular mechanisms of PER and CRY family members in lung adenocarcinoma (LUAD) and their correlations with prognoses and immune infiltration in LUAD patients are still unclear. In this study, to identify their biological functions in LUAD development, comprehensive high-throughput techniques were applied to analyze the relationships of expressions of PER and CRY family members with genetic variations, molecular mechanisms, and immune infiltration. The present results showed that transcription levels of PER1 and CRY2 in LUAD were significantly downregulated. High expression levels of PER2, PER3, CRY1, and CRY2 indicated longer overall survival. Some cancer signaling pathways were related to PER and CRY family members, such as cell-cycle, histidine metabolism, and progesterone-mediated oocyte maturation pathways. Expressions of PER and CRY family members significantly affected the infiltration of different immune cells. In conclusion, our findings may help better understand the molecular basis of LUAD, and provide new perspectives of PER and CRY family members as novel biomarkers for LUAD.
Article
GEC is one of the most common cancers and has become a significant health burden worldwide. HER-2 is a proto-oncogene, amplified or overexpressed in different tumors, and associated with a worse prognosis. Trastuzumab plus chemotherapy is the current standard treatment for advanced HER-2 positive GEC. However, it still does not benefit well all patients. HER-2 targeted therapy can up-regulate the expression of PD-1, CTL-4, and TAMs in the tumor microenvironment, strengthen the ADCC process, and enhance the efficacy of immunotherapy. While immune checkpoint inhibitors could reduce drug resistance to anti-HER-2 drugs. Moreover, immunotherapy combined with HER-2 targeted therapy in HER-2 positive GEC has shown perceptible efficacy and acceptable side effects in clinical trials and is regarded as a new therapeutic strategy for GEC. However, there remain significant challenges and requires more research to improve the survival benefit of this therapeutic approach.
Article
Alkylresorcinols (ARs) are phenolic lipids present in the bran part of whole grain wheat and rye, which possess antioxidant, anti-inflammatory, anti-cancer and anti-tumor properties. The physiological activities of ARs have been proven to be diverse; however, the specific molecular mechanisms are still unclear. In this study, reverse virtual screening and network pharmacology were used to explore the potential molecular mechanisms of the physiological function of ARs and their endogenous metabolites. The Metascape database was used for GO enrichment and KEGG pathway analysis. Furthermore, molecular docking was used to investigate the interactions between active compounds and potential targets. The results showed that the bioavailability of most ARs and their endogenous metabolites was 0.55 and 0.56, while the bioavailability of certain endogenous metabolites was only 0.11. Multiplex analysis was used to screen 73 important targets and 4 core targets (namely, HSP90AA1, EP300, HSP90AB1 and ERBB2) out of the 163 initial targets. The important targets involved in the key KEGG pathway were pathways in cancer (hsa05200), lipid and atherosclerosis (hsa05417), Th17 cell differentiation (hsa04659), chemical carcinogenesis-receptor activation (hsa05207), and prostate cancer (hsa05215). The compounds involved in the core targets were AR-C21, AR-C19, AR-C17, 3,5-DHPHTA-S, 3,5-DHPHTA-G, 3,5-DHPPTA, 3,5-DHPPTA-S, 3,5-DHPPTA-G, 3,5-DHPPTA-Gly and 3,5-DHPPA-G. The interaction force between them was mainly related to hydrogen bonds and van der Waals. Overall, the physiological activities of ARs are not only related to their multiple targets, but may also be related to the synergistic effect of their endogenous metabolites.
Article
Full-text available
Given their intrinsic pleiotropism, microRNAs (miR) play complex biological roles, in both normal and pathological conditions. Often the same miR can act as oncogene or oncosuppressor, depending on the biological process dysregulated in each specific tissue. miR‐223 does not represent an exception to this rule and its functions greatly differ in different contexts. miR‐223 has been widely studied in the hematopoietic compartment, where it plays a central role in innate immune response, regulating myeloid differentiation and granulocytes function. Accordingly, dysregulated expression of miR‐223 has been associated to different inflammatory disorders and tumors arising from the immune compartment. Most carcinomas, breast cancer being the most studied, display loss of miR‐223. However, in gastro‐esophageal cancers miR‐223 is frequently overexpressed and correlates with worse prognosis. A link between miR‐223 and response to CDK4/6‐inhibitors has been recently proposed, suggesting a role as biomarker of therapeutic response. The notion that one of the most commonly mutated protein in cancer, mutant p53, binds the promoter of miR‐223 and suppresses its transcription, adds a further level of complexity to the full understanding of miR‐223 in cancer. In this review, we will summarize the current knowledge on the molecular networks that alter or are altered by miR‐223, in different cancer types. We will discuss if the times are ready for the exploitation of miR‐223 as predictive biomarker of treatment response or, even, as therapeutic target, in specific settings. Finally, we will suggest which could be the next steps to be taken for a realistic clinical application of miR‐223. This article is categorized under: RNA in Disease and Development > RNA in Disease
Article
Full-text available
The aim of the present study was to identify potential therapeutic targets that serve crucial roles in the progression of cervical cancer. Clinical data, RNA sequencing (RNAseq)-counts and micro (mi)RNA data regarding cervical squamous cell carcinoma were retrieved from The Cancer Genome Atlas, and analyses were performed using the University of California Santa Cruz database. RNAseq and miRNA data were stratified into 3 groups (according to the patients' age), and genes were re-annotated and preprocessed prior to Mfuzz time clustering analysis. Subsequently, enrichment analyses were performed in order to identify differentially expressed mRNAs (DEmRNAs) and a protein-protein interaction analysis network was constructed. miRNA-gene, miRNA-lncRNA, and long non-coding (lnc)RNA-mRNA pairs were collected and the lncRNA-miRNA-mRNA competing endogenous (ce)RNA network was established. Further enrichment analyses were performed in order to identify crucial mRNAs in the ceRNA network. Finally, survival and drug association analyses were implemented. A total of 269 DEmRNAs [including alcohol dehydrogenase 7 (ADH7), vestigial-like family member 3 (VGLL3) and cytochrome P450, family 26, subfamily B, polypeptide 1 (CYP26B1)], 274 DElncRNAs (including LINC01133) and 16 DEmiRNAs (including miR-3065 and miR-330) were identified. There were 102 lncRNAs, 15 miRNAs, 15 mRNAs and 522 interaction pairs in the ceRNA network. In particular, ADH7 was regulated by miR-3065, and miR-3065 interacted with LINC01133 in the ceRNA network. Furthermore, ADH7 and CYP26B1 were enriched in the retinoic acid metabolic process and the retinol metabolism pathway. ADH7 and VGLL3 were significantly associated with the cervical cancer survival rate. ADH7, VGLL3, CYP26B1, miR-3065, miR-330, miR-499a and LINC01133 play pivotal roles in the progression of cervical cancer in different age groups.
Article
Full-text available
Increasing evidence shows that cancer stem cells are responsible for drug resistance and relapse of tumors. In breast cancer, human epidermal growth factor receptor 2 (HER2) induces Herceptin resistance by inducing cancer stem cells. In the present study, we explored the effect of HER2 on cancer stem cells induction and drug sensitivity of ovarian cancer cell lines. First, we found that HER2 overexpression (HER2 OE) induced, while HER2 knockdown (HER2 KD) decreased CD44+/CD24- population. Consistently, HER2 expression was closely correlated with the sphere formation efficiency (SFE) of ovarian cancer cells. Second, we found that NFκB inhibition by specific inhibitor JSH23 or siRNA targetting subunit p65 dramatically impaired the induction of ovarian cancer stem cells by HER2, indicating that NFκB mediated HER2-induced ovarian cancer stem cells. Third, we found that HER2 KD significantly attenuated the tumorigenicity of ovarian cancer cells. Further, we found that HER2 inhibition increased drastically the sensitivity of ovarian cancer cells to doxorubicin (DOX) or paclitaxel (PTX). Finally, we examined the correlation between HER2 status and stem cell-related genes expression in human ovarian tumor tissues, and found that expressions of OCT4, COX2, and Nanog were higher in HER2 positive tumors than in HER2 negative tumors. Consistently, the 5-year tumor-free survival rate of HER2 positive patients was dramatically lower than HER2 negative patients. Taken together, our data indicate that HER2 decreases drug sensitivity of ovarian cancer cells via inducing stem cell-like property.
Article
Full-text available
Objective: Recently, we characterized mouse monoclonal antibodies that allow the specific and sensitive detection of human histamine N-methyltransferase (HNMT). To understand differences in binding characteristics and recognition of enzyme variants we mapped the antibody binding sites. Methods: Fragments of human HNMT were expressed as glutathione S-transferase fusion proteins that were used for testing antibody binding on immunoblots. Combined information from species cross-reactivity, sequence comparison, protein structure, and binding site prediction software were used to localize the epitope recognized by each antibody. Results: All eight monoclonal HNMT antibodies bound to linear epitopes in the C-terminal domain of the 292 amino acid protein. Of the five antibodies cross-reacting with HNMT from other species, one bound region L(182)-T(223), three region M(224)-E(261), and one region L(262)-A(292). All three antibodies recognising only human HNMT bound the C-terminal region L(262)-A(292) that contains residues present only in the human protein. Conclusions: Our HNMT monoclonal antibodies bind in three different regions of the protein and those binding the same putative epitope exhibit similar binding characteristics and species cross-reactivity. Antibodies binding non-overlapping epitopes will facilitate analyses of all clinically relevant variants described for HNMT.
Article
Full-text available
HER2 is a receptor tyrosine kinase, which is amplified and overexpressed in a subset of human cancers including breast and gastric cancers, and is indicated in its involvement in progression of cancer. Although its specific ligand(s) has not been detected, HER2 homodimerization, which is critical for its activation, is considered to be dependent on its expression levels. Here, we demonstrate a significant role of HER2 methylation by protein lysine methyltransferase SMYD3 in HER2 homodimerization. We found that SMYD3 trimethylates HER2 protein at lysine 175. HER2 homodimerization was enhanced in the presence of SMYD3, and substitution of lysine 175 of HER2 with alanine (HER2-K175A) reduced the formation of HER2 homodimers. Furthermore, HER2-K175A revealed lower level of autophosphorylation than wild-type HER2. We also identified that knockdown of SMYD3 attenuated this autophosphorylation in breast cancer cells. Our results imply that SMYD3-mediated methylation of HER2 at Lysine 175 may regulate the formation of HER2 homodimer and subsequent autophosphorylation and suggest that the SMYD3-mediated methylation pathway seems to be a good target for development of novel anti-cancer therapy.
Article
Cancer stem cells (CSCs), a subpopulation of cancer cells endowed with self-renewal, tumorigenicity, pluripotency, chemoresistance, differentiation, invasive ability, and plasticity, reside in specialized tumor niches and are responsible for tumor maintenance, metastasis, therapy resistance, and tumor relapse. The new-age "hierarchical or CSC"model of tumor heterogeneity is based on the concept of eradicating CSCs to prevent tumor relapse and therapy resistance. Small-molecular entities and biologics acting on various stemness signaling pathways, surface markers, efflux transporters, or components of complex tumor microenvironment are under intense investigation as potential anti-CSC agents. In addition, smart nanotherapeutic tools have proved their utility in achieving CSC targeting. Several CSC inhibitors in clinical development have shown promise, either as mono- or combination therapy, in refractory and difficult-to-treat cancers. Clinical investigations with CSC marker follow-up as a measure of clinical efficacy are needed to turn the "hype"into the "hope"these new-age oncology therapeutics have to offer.
Article
Cancer stem cells (CSCs) have been shown to be resistant to current anticancer therapies and the induction of oxidative stress is an important mechanism of action for many anticancer agents. However, it is still largely unknown how CSCs respond to hydrogen peroxide (H2O2)-induced oxidative stress. Here, we show that the levels of reactive oxygen species (ROS) are markedly lower in breast CSCs (BCSCs) than that in non-cancer stem cells (NCSCs). A transient exposure of breast cancer cells to sublethal doses of H2O2 resulted in a dose-dependent increase of the epithelium-specific antigen (ESA)⁺/CD44⁺/CD24⁻ subpopulations, a known phenotype for BCSCs. Although BCSCs survived sublethal doses of H2O2 treatment, they lost the ability to form tumor spheres and failed to generate colonies as demonstrated by mammosphere-formation and clonogenic assays, respectively. Mechanistic studies revealed that H2O2 treatment led to a marked increase of senescence-associated β-galactosidase activity but only minimal apoptotic cell death in BCSCs. Furthermore, H2O2 triggers p53 activation and promotes p21 expression, indicating a role for the p53/p21 signaling pathway in oxidative stress-induced senescence in BCSCs. Taken together, these results demonstrate that the maintenance of a lower level of ROS is critical for CSCs to avoid oxidative stress and H2O2-induced BCSC loss of function is likely attributable to oxidative stress-triggered senescence induction, suggesting that ROS-generating drugs may have the therapeutic potential to eradicate drug-resistant CSCs via induction of premature senescence.
Article
Atopic dermatitis (AD) is one of the most prevalent skin diseases around the world. Excessive histamine plays a critical role as an inflammatory factor in the pathogenesis of AD. Deregulated microRNAs (miRNAs) were involved in atopic dermatitis by targeting various genes. MiR-223 had been reported to play a vital role in hematopoiesis. In this study, we identified upregulated miR-223 in the whole blood cells of a large group of AD patients. What's more, we found for the first time that one of the major histamine degradation enzymes, histamine-N-methyltransferase (HNMT), was increased in AD patients and AD model mice. Although there was one miR-223 binding site in the 3'- untranslated region of the HNMT gene, HNMT were not inhibited by miR-223. Taken together, it suggested that miR-223 participates in AD through upregulating HNMT indirectly to degrade the excessive histamine.
Article
In our endeavor towards the development of potent molecules for cancer diseases, we have designed and synthesized a series of 2,4,5-trisubstituted imidazole derivatives (B1-B24) and characterized by using various spectroscopic techniques. All these compounds are further evaluated for their in vitro anti-cancer, anti-oxidant activities and molecular docking studies against EGFR, HER2 protein receptors. The in vitro anti-cancer activity analysis reveals that compounds B11 and B16 were found to be effective scaffolds against the tested human cancer cell lines IMR-32, A549 and HeLa. Particularly, B16 and B11 showed effective cytotoxicity against A549 and IMR-32 with IC50 values of 09.521±0.54μM and 10.294±0.43μM, respectively. Moreover, compounds B17, B18 and B23 showed potent activity towards the anti-oxidant screening with IC50 values of 5.87±1.73μM, 6.29±1.27μM and 4.95±1.81μM, respectively compared to standard ascorbic acid. Molecular docking was performed against the EGFR, HER2 protein receptors to provide more insight into their mechanism of interaction by comparing with standard EGFR, HER2 inhibitors like Gefitinib (EFGR), Lapatanib (EGFR), Afitinib (HER2) and Canertinib (HER2). Compounds B15, B16, B11 and B10 were exhibiting their minimum binding energies. Out of the aforementioned docked molecules, B15 and B16 showed the best binding energies of -11.15kcalmol(-1), -10.70kcalmol(-1) and -10.49kcalmol(-1), -10.12kcalmol(-1) against EGFR and HER2 protein receptors, respectively. The molecular docking results are well corroborated with the in vitro anti-cancer activity finding.