ArticlePDF Available

Abstract and Figures

Sugars are ubiquitous in organisms and well-known cosmetic ingredients for moisturizing skin with minimal side-effects. Glucose, a simple sugar used as an energy source by living cells, is often used in skin care products. Several reports have demonstrated that sugar and sugar-related compounds have anti-melanogenic effects on melanocytes. However, the underlying molecular mechanism by which glucose inhibits melanin synthesis is unknown, even though glucose is used as a whitening as well as moisturizing ingredient in cosmetics. Herein, we found that glucose significantly reduced the melanin content of α-melanocyte-stimulating hormone (MSH)-stimulated B16 cells and darkly pigmented normal human melanocytes with no signs of cytotoxicity. Furthermore, topical treatment of glucose clearly demonstrated its whitening efficacy through photography, Fontana-Masson (F&M) staining, and multi-photon microscopy in a pigmented 3D human skin model, MelanoDerm. However, glucose did not alter the gene expression or protein levels of major melanogenic proteins in melanocytes. While glucose potently decreased intracellular tyrosinase activity in melanocytes, it did not reduce mushroom tyrosinase activity in a cell-free experimental system. However, glucose was metabolized into lactic acid, which can powerfully suppress tyrosinase activity. Thus, we concluded that glucose indirectly inhibits tyrosinase activity through conversion into lactic acid, explaining its anti-melanogenic effects in melanocytes.
Content may be subject to copyright.
Int.J.Mol.Sci.2020,21,1736;doi:10.3390/ijms21051736www.mdpi.com/journal/ijms
Article
GlucoseExertsanAntiMelanogenicEffectby
IndirectInactivationofTyrosinaseinMelanocytes
andaHumanSkinEquivalent
SungHoonLee
1,2
,IlHongBae
1
,EunSooLee
1
,HyoungJuneKim
1
andJongsungLee
2,
*
andChangSeokLee
3,
*
1
AmorepacificCorporationR&DCenter,YonginCity,17074,Gyunggido,Korea;
imstrong20@gmail.com(S.H.L.);baelong98@naver.com(I.H.B.);soopian@amorepacific.com(E.S.L.);
leojune@amorepacific.com(H.J.K.)
2
DepartmentofIntegrativeBiotechnology,CollegeofBiotechnologyandBioengineering,Sungkyunkwan
University,SuwonCity,16419,Gyunggido,Korea
3
DepartmentofBeautyandCosmeticScience,EuljiUniversity,SeongnamCity,13135,Gyunggido,Korea
*Correspondence:cslee2010@eulji.ac.kr(C.S.L.),Tel.:+82317407549(C.S.L.);bioneer@skku.edu(J.L.),
Tel.:+82312907861(J.L.)
Received:6February2020;Accepted:1March2020;Published:3March2020
Abstract:Sugarsareubiquitousinorganismsandwellknowncosmeticingredientsformoisturizing
skinwithminimalsideeffects.Glucose,asimplesugarusedasanenergysourcebylivingcells,is
oftenusedinskincareproducts.Severalreportshavedemonstratedthatsugarandsugarrelated
compoundshaveantimelanogeniceffectsonmelanocytes.However,theunderlyingmolecular
mechanismbywhichglucoseinhibitsmelaninsynthesisisunknown,eventhoughglucoseisused
asawhiteningaswellasmoisturizingingredientincosmetics.Herein,wefoundthatglucose
significantlyreducedthemelanincontentofα‐melanocytestimulatinghormone(MSH)stimulated
B16cellsanddarklypigmentednormalhumanmelanocyteswithnosignsofcytotoxicity.
Furthermore,topicaltreatmentofglucoseclearlydemonstrateditswhiteningefficacythrough
photography,FontanaMasson(F&M)staining,andmultiphotonmicroscopyinapigmented3D
humanskinmodel,MelanoDerm.However,glucosedidnotalterthegeneexpressionorprotein
levelsofmajormelanogenicproteinsinmelanocytes.Whileglucosepotentlydecreasedintracellular
tyrosinaseactivityinmelanocytes,itdidnotreducemushroomtyrosinaseactivityinacellfree
experimentalsystem.However,glucosewasmetabolizedintolacticacid,whichcanpowerfully
suppresstyrosinaseactivity.Thus,weconcludedthatglucoseindirectlyinhibitstyrosinaseactivity
throughconversionintolacticacid,explainingitsantimelanogeniceffectsinmelanocytes.
Keywords:melanogenesis;sugar;glucose;tyrosinase;humanskinequivalent
1.Introduction
Melanogenesisistheprocessofmelaninproductionandisessentialforskinprotection.Melanin
absorbsultraviolet(UV)lightandprotectstheskinfromthedamagingeffectsofUVlightandfree
radicals[1].However,becauseexcessiveproductionofmelanincauseshyperpigmentationsuchas
frecklesandlentigo,whichmaybeconsideredunaesthetic,muchresearchhasbeendedicated
towardsfindingeffectivedepigmentaryingredientsforcosmeticsormedicines[2–4].
Sugarisapowerfulhumectantforskinmoisturizationandisusedasacosmeticingredientfor
moisturizingskinwithminimalsideeffects.Inaddition,sugarsandsugarrelatedagentsaffect
melanogenesis[5,6].Glycosylationoftyrosinase,akeyenzymeinvolvedinmelaninsynthesis,canbe
Int.J.Mol.Sci.2020,21,17362of13
altered,inhibititscatalyticactivityandacceleratingitsdegradation[7].Theroleofsugarsin
melanogenesishasbeenhighlightedbystudiesinvestigatingtheeffectofglycosylationonthe
pigmentationphenotypeofmelanocytesandtherolesofsugarresiduesonthecatalyticactivityof
tyrosinase[5–7].Somestudieshavereportedthatsugarderivativescaninhibittyrosinasematuration,
affectingitsglycosylation.Forexample,Nacetylglucosamine(NAG),anaminohexoseproduced
physiologicallybyadditionofanaminogrouptoglucose,disruptstyrosinaseglycosylation,resulting
indepigmentingeffectsinguineapigskinandinhumanskin[8].Additionally,recentstudieshave
showedthatsugarrelatedcompoundsinhibittyrosinaseexpressionoractivationaswellasalterits
glycosylation.Forexample,weevaluatedthewhiteningefficacyofgalacturonicacid(GA),asugar
acidthatisanoxidizedformofgalactoseandthemaincomponentofpectin.Galacturonicacidexerts
awhiteningeffectthroughregulationoftyrosinaseactivityandexpressioninB16murinemelanoma
cellsandahumanskinequivalent[9].Inanotherreport,anewtypeofcyclicoligosaccharide,known
ascyclicnigerosylnigerose(CNN),showedaweakbutsignificantdirectinhibitoryeffectonthe
enzymaticactivityoftyrosinase,suggestingonepossiblemechanismofhypopigmentation[10].
Similartotyrosinasematurationbyproperglycosylation,theexpressionoractivityofCNNcouldbe
atargetofantimelanogenicagents[11].However,numerousantimelanogenicagentshavesevere
sideeffects,suchasvitiligo[12,13].Thereisthereforegreatinterestinsaferdepigmentary
compounds.
Here,weinvestigatedtheantimelanogeniceffectsofglucoseonB16murinemelanomacellsand
normalhumanmelanocytes.Inaddition,weexaminedtissuecolorandepidermalstatususingtissue
sectionstainingofahumanskinequivalent.Basedonourfindings,weproposethatthewhitening
effectofglucoseisdependentonlacticacidproduction,resultingintyrosinaseinactivation.
2.Results
2.1.AntiMelanogenicEfficacyofGlucoseinB16andNHMs
Toinvestigatetheantimelanogeniceffectofglucose,weusedtwotypesofmelanocytes,B16
melanomacells(amurinemelanomacellsline)andnormalhumanmelanocytes(NHMs).First,we
determinedifglucosewastoxictoB16cells.Glucosedidnotshowanycytotoxicityatconcentrations
upto100mMinB16cells,asshowninFigure1A.Basedonthecytotoxicitydata,B16cellswere
treatedwithvariousconcentrationsofglucosefor72hinthepresenceofα‐melanocytestimulating
hormone(MSH),aninducerofmelanogenesis.AsshowninFigure1B,glucoseclearlyand
significantlydownregulatedtheintracellularmelanincontentinadosedependentmanner.Kojic
acid(KA)wasusedasareferencecompoundforantimelanogenesisbecauseitisoftenusedasaskin
lighteningcosmeticingredient[1,3,4].Thecoloroflysatesinglucosetreatedcellswaslighterthan
thecolorofcontrolcells(Figure1C).Inaddition,weconfirmedthattheamountofmelaninsecreted
intotheculturemediadecreasedandthecolorofthemediaclearlybrightened(Figure1D).Next,we
investigatedtheantimelanogeniceffectofglucoseondarklypigmentedNHMs.Glucoseat
concentrationsupto100mMwasnotcytotoxicforuptofor4days(Figure1E).Whenmelanincontent
wasdeterminedafterglucosetreatmentofNHMsfor4days,wefoundthatmelanincontent
decreasedinadosedependentmanner(Figure1F).Takentogether,theseresultsindicatethatglucose
suppressesmelaninsynthesisinmelanocytes.
Int.J.Mol.Sci.2020,21,17363of13
Int.J.Mol.Sci.2020,21,17364of13
Figure1.EffectofglucoseonB16cellsandnormalhumanmelanocytes(NHMs).(A)Effectofglucose
ontheviabilityofB16cells.(B)IntracellularmelanincontentsinαMSHstimulatedB16cells.
Intracellularmelanincontentsweredeterminedusingcelllysates,asdescribedinthemethodssection.
(C)Thecolorofcelllysate.(D)Extracellularmelanincontentsweredeterminedusingculturedmedia
containingsecretedmelaninafterglucoseandα‐MSHcotreatmentfor72h.KAindicateskojicacid
(100μg/mL),whichwasusedasareferencecompound.Thephotographshowsthecolorsofculture
media.(E)EffectofglucoseontheviabilityofNHMs.(F)Effectsofglucoseonmelaninsynthesisin
NHMs.NHMsweretreatedwiththeindicatedconcentrationsofglucosefor4d,washed,andlysed
withNaOHtodeterminetheintracellularmelanincontents.Themelanincontentswereestimatedby
absorbanceat405nmandnormalizedbythetotalproteincontents.Dataareexpressedasthemean±
SDofatleastthreeindependentmeasurements(*p<0.05,**p<0.01,***p<0.001).
2.2.WhiteningEffectofGlucoseona3DHumanSkinEquivalent
Tofurtherdefinetheantimelanogenicabilityofglucose,weusedapigmented3Dhumanskin
model,MelanoDerm.AsdescribedintheMaterialsandMethodssection,glucosewastopically
appliedtotheMelanoDermfor18days,andcellviabilitywasdeterminedbyCCK8assay.Asshown
inFigure2A,notissuecytotoxicitywasobservedafterglucosetreatmentfor18days.Tissuecolor
changeswereassessedbyphotography.AsshowninFigure2B,glucosetreatedtissuewaslighterin
colorthanphosphatebufferedsaline(PBS)treatedtissue.Inaddition,hematoxylinandeosin(H&E)
stainingrevealedthatglucosedidnotinducetissuecollapse,whilefontanamasson(F&M)staining
showedthatglucosedecreasedthenumberofhyperpigmentedmelanocyte(asindicatedbyarrows)
inthebasallayer(Figure2C).
Tofurtherinvestigatechangesinmelanincontent,wecomparedtheautofluorescencesignals
ofmelanininthemelanocytelayersofPBS‐andglucosetreatedtissuesusingtwophotonexcitation
fluorescence(TPEF)microscopy,asshowninFigure2D.Ananalysisoftheseimagesshowedthatthe
melaninvolumewasdecreasedbyapproximately36%andTPEFsignalintensityformelanininthe
melanocyterichareawasdecreasedbyapproximately38%inglucosetreatedtissuescomparedwith
PBStreatedtissues.
Int.J.Mol.Sci.2020,21,17365of13
Figure2.Effectofglucoseonhumanskinequivalent,MelanoDerm.(A)Viabilityofhumanskin
equivalentstreatedwithglucose.(B)Humanskinequivalents(MelanoDerm;n=3)weretopically
treatedwithglucosefor18d,andthenphotographed.TheΔLvalueindicatesthedegreeoflightness
comparedwithPBStreatedtissue.(C)H&EandF&Mstainingoftissuesections.Theslideswerefixed
informaldehydesolutionandembeddedinparaffinwaxforstaining(scalebar,50μm).Blackarrows
indicatepigmentedmelanocytes.(D)Melaninimaging(200×200×60μm
3
)ofhumanskinequivalents
wasperformedusingTPEFmicroscopy.Pseudocolored(red)signalsindicatemelanin(scalebar,50
μm).ThegraphsindicatethequantificationofmelaninvolumeandTPEFsignals.Dataareexpressed
asthemean±SDofatleastthreeindependentmeasurements(*p<0.05,***p<0.001).
2.3.EffectofGlucoseontheExpressionofMelanogenicProteinsinMelanocytes
Int.J.Mol.Sci.2020,21,17366of13
Next,toelucidatethedepigmentationmechanismofglucoseinmelanocytes,weassessedits
effectontheexpressionofmelanogenicenzymessuchastyrosinaseandTyrp1inB16cellsand
NHMs.B16cellsweretreatedwithglucosefortheindicatedtimesinthepresenceofα‐MSH.Then,
proteinlevelsoftyrosinaseandTyrp1weredeterminedbyWesternblotassay(Figure3A).
ExpressionleveloftyrosinaseandTyrp1werenotdecreasedbyglucoseatanytimepoint.
Furthermore,transcriptlevelsoftyrosinasewerenotaffectedbyglucoseinαMSHstimulatedB16
cells(Figure3B).SimilartoB16cells,proteinlevelsoftyrosinase,Tyrp1,andMITFwerenotinhibited
byglucose(Figure3C)inNHMcellstreatedwithglucose,andmRNAlevelsoftyrosinaseandTyrp
1werealsonotdecreased,butratherslightlyincreasedbyglucose(Figure3D).
2.4.EffectofGlucoseonTyrosinaseActivity
Tofurtherdefinetheactionmechanismofglucose,wetestedifithadaninhibitoryeffecton
mushroomtyrosinaseactivity.AsshowninFigure4A,wefoundthatglucosehadnoinhibitoryeffect
onmushroomtyrosinaseactivity,indicatingthatglucosedoesnotdirectlyaffecttyrosinaseactivity.
Wenextperformedatotalintracellulartyrosinaseactivityassayusingglucosetreatedcelllysates
frombothB16cellandNHMs.Interestingly,intracellulartyrosinaseactivitywasclearlyinhibitedin
adosedependentmanneringlucosetreatedB16cellsinthepresenceofαMSH(Figure4B).In
NHMs,glucosealsoinhibitedintracellulartyrosinaseactivity(Figure4C).Thesedatasupportthe
possibilitythatglucoseindirectlyinactivatestyrosinaseinmelanocytes.
Int.J.Mol.Sci.2020,21,17367of13
Figure3.EffectofglucoseontheexpressionofmelanogenicproteinsinB16cellsandNHMs.(A,B)
B16cellsweretreatedwithα‐MSHfortheindicatedtimeinthepresenceorabsenceof20mMglucose.
Then,Westernblot(A)andqRTPCR(B)assayswereperformed.(C)NHMsweretreatedwiththe
indicatedconcentrationsofglucosefor48h.Then,theWesternblotassaywasperformed.(D)NHMs
weretreatedwiththeindicatedtimeinthepresenceof50mMglucose.Then,qRTPCRassayswere
performed.Dataareexpressedasthemean±SDofatleastthreeindependentmeasurements.
Figure4.Effectofglucoseonthetyrosinaseactivity.(A)Effectofglucoseonthemushroomtyrosinase
activityincellfreesystem.(BC)Cellulartyrosinaseactivityassay.(B)B16cellsweretreatedwiththe
indicatedconcentrationsofglucosefor24h.Then,thecellulartyrosinaseactivityassaywas
performed,asdescribedinthemethodssection.(C)NHMsweretreatedwith50mMglucoseforthe
indicatedtime.Then,thecellulartyrosinaseactivityassaywasperformed,asdescribedinthe
methodssection.Thecellulartyrosinaseactivitywasnormalizedbythetotalproteincontents.Data
areexpressedasthemean±SDofatleastthreeindependentmeasurements(*p<0.05,**p<0.01,***p
<0.001).
Int.J.Mol.Sci.2020,21,17368of13
2.5.TyrosinaseInactivationbytheProductionofLacticAcidinGlucoseTreatedMelanocytes
Glucoseisconvertedintothecellularmetabolitelactate,whichislacticacidinsolutionand
whichhasbeenreportedtobeeffectiveintreatingpigmentarylesions[14,15].Therefore,we
hypothesizedthatglucoseisconvertedintolacticacidinmelanocytesandthatincreasedlevelsof
lacticacidinhibitmelanogenesisthroughtyrosinaseinactivation.Toevaluatethishypothesis,wefirst
assessedtheproductionoflacticacidinmediafromglucosetreatedmelanocytes.Asexpected,
glucosesignificantlyupregulatedthelacticacidcontentinB16cellsculturedmedia(Figure5A).In
addition,becauselacticacidisknowntodirectlyinhibittyrosinaseactivity[15],weevaluatediflactic
acidsuppressedmushroomtyrosinaseactivity.AsshowninFigure5B,lacticaciddramatically
inhibitedmushroomtyrosinaseactivity,unlikeglucose.Theseresultssuggestthatconversionof
glucosetolacticacidhasanantimelanogeniceffectviatyrosinaseinactivationbythelacticacid.
3.Discussion
Sugarsorsugarderivedmaterialsareoftenusedascosmeticingredientsforskinprotectionand
physiologicalcontrol.Forexample,raffinoseincreasesmTORindependentautophagyandreduces
celldeathinUVBirradiatedkeratinocytes,indicatingthatthenaturalagentraffinosehaspotential
valueinlimitingphotodamage[16].Trehaloseandsucrosearenovelactivatorsofautophagyin
humankeratinocytesthroughanmTORindependentpathway[17].Thesefindingsprovidenew
insightintothesugarmediatedregulationofautophagyinkeratinocytes.Inthecaseofglucose,
topicalglucosewasshowntoinduceclaudin1andfilaggrinexpressioninamousemodelofatopic
dermatitisandinkeratinocyteculture,indicatingthatithasanantiinflammatoryeffectbyrepairing
skinbarrierfunction[18].Inaddition,glucoseinhibitsproliferationandenhancesthedifferentiation
ofskinkeratinocytes[19].Therefore,glucoseregulatesvariousaspectsofepidermalphysiology,such
asskinbarrierfunctionsandkeratinocytehydrationlevels.
Figure5.Productionoflactatebyglucoseandeffectoflacticacidonthetyrosinaseactivity.(A)Lactate
productioninglucosetreatedB16cells.(A)B16cellsweretreatedwiththeindicatedconcentrations
ofglucosefor3d.Then,alactateassaywasperformedusingtheculturedmedia,asdescribedinthe
methodssection.(B)Effectoflacticacidonthemushroomtyrosinaseactivityincellfreesystem.Data
Int.J.Mol.Sci.2020,21,17369of13
areexpressedasthemean±SDofatleastthreeindependentmeasurements(*p<0.05,**p<0.01,***p
<0.001).
Sugarscanactasdepigmentaryagentsviaseveraldifferentmechanismsthatinvolvetyrosinase.
Forexample,someagentsinhibittyrosinasematuration,whileotheragentsinduceinhibitionof
tyrosinasegeneexpressionoractivity[5,8–10].However,fewstudieshaveinvestigatedthe
mechanismsunderlyingthedepigmentationeffectsofglucose.
Todeterminetheeffectofglucoseonmelanogenesis,wepreviouslyfocusedonliverXreceptor
(LXRs),whichareligandactivatednuclearreceptorsthatplaypivotalrolesinlipidmetabolismand
cholesterolhomeostasis[20].WefoundthatliverXreceptoractivationinhibitsmelanogenesis
throughtheaccelerationofextracellularsignalregulatedkinase(ERK)mediatedmicrophthalmia
associatedtranscriptionfactor(MITF)degradation[21].Furthermore,glucoseisanendogenousLXR
ligand[22].Thus,wehypothesizedthatglucosehasantimelanogeniceffectsduetoactivationofan
LXRdependentpathway.However,asshowninFigure3C,glucosedidnotaltertheexpression
levelsoftyrosinaseorMITF,althoughLXRactivationreducedtheexpressionlevelsoftheseproteins
inmelanocytes.Therefore,weconcludedthatglucosehaddepigmentingeffectsonmelanocytes
independentofLXRactivation.
Glucoseistheprincipalsubstrateforenergyproduction,anditishydrolyzedviaserialreactions
ofseveralenzymes,knownasglycolysis.Numerousstudieshaveshownthatglycolysishasonlyone
endproduct,lacticacid,whetherunderaerobicoranaerobicconditions.Underaerobicconditions
(O2),lactateisutilizedasthesubstrateofmitochondriallactatedehydrogenase(mLDH).LDHs
convertittopyruvatethatentersthetricarboxylicacid(TCA)cycle.Underanaerobicconditions(N2),
lactateisaccumulatedinthecytosol.Therefore,theglycolysispathwaybeginswithglucoseasits
substrateandterminateswiththeproductionoflactateasitsmainendproduct[23].Inaddition,
Davidetal.measuredthefractionofglucosethatwasconvertedtolacticacidorpyruvateinthe
normalhumanmelanocyte[24].Mostofthemetabolitesofglucosewerelacticacidratherthan
pyruvate.
Lacticacidisanalphahydroxyacid(AHA);theseacidsareusedextensivelyincosmetic
formulationsassuperficialpeelingagents[25].Inaddition,lacticacidsuppressesmelaninformation
bydirectlyinhibitingtyrosinaseactivity,aneffectindependentofitsacidicnature,whichmeansthat
lacticacid’seffectsonpigmentarylesionsareduenotonlytoaccelerationofepidermalcellturnover,
butalsodirectinhibitionofmelaninformationinmelanocytes[15].Thus,wereasonedthatglucose
mayexertitsantimelanogeniceffectvialacticacidproduction,becauseglucosecanbeconvertedto
lacticacid.Asexpected,wefoundthatglucosetreatmentresultedinlacticacidproductionin
melanocytes(Figure5A).Inaddition,weconfirmedthatlacticacidpowerfullyanddirectlyinhibited
tyrosinaseactivity(Figure5B).Usukietal.alsodiscoveredthatlacticaciddecreasedintracellular
tyrosinaseactivityinB16cellsandhumanHM3KOcells[15].Inthereport,mRNAandproteinlevels
oftyrosinaseandTyrp1werenotaffectedbylacticacid.Together,thesedataindicatethatglucose
increaseslacticacidproductionandthatthislacticaciddirectlyinhibitstyrosinaseactivitywithout
affectinggeneexpressionlevels,indicatingthatglucosehasanantimelanogeniceffectin
melanocytesviaindirecttyrosinaseinactivationdependentonlacticacidproduction.However,
furtherexperimentationsarenecessarytovalidatetheroleoflacticacidandglucosein
depigmentation.
Collectivedatafromthisstudyprovidepreliminaryevidencesupportingtheutilityoftopical
glucoseasaneffectivewhiteningaswellasmoisturizingreagentthatcanbesafelyusedincosmetics
andmedicinalformulations.
4.MaterialsandMethods
4.1.Materials
Dglucose,α‐MSH,kojicacid(KA),Ltyrosine,LDOPA,andlacticacidwerepurchasedfrom
SigmaAldrich(St.Louis,MO,USA).Antibodiesagainsttyrosinaseandactinwerepurchasedfrom
Int.J.Mol.Sci.2020,21,173610of13
Abcam(Cambridge,UK).AntibodyagainstTyrp1waspurchasedfromSantaCruzBiotechnology
(CA,USA).AntibodyagainstMITFwaspurchasedfromProteintech(city,IL,USA).
4.2.CellCultureandViabilityAssay
WepurchasedB16murinemelanomacells,Dulbecco’smodifiedEagle’smedium(DMEM),and
fetalbovineserum(FBS)fromtheAmericanTypeCultureCollection(ATCC,Manassas,VA,USA).
B16cellswereculturedinDMEMcontaining4500mg/Lhighglucose(ATCC302002)as
recommendedbythemanufacturer(ATCC)supplementedwith5%FBS,andincubatedat37°Cina
humidifiedatmospherecontaining95%airand10%CO2.DarklypigmentedprimaryNHMswere
purchasedfromThermoFisherScientific(#C2025C;Waltham,MA,USA).Cellswereculturedin
Medium254(#M254500)supplementedwithHumanMelanocyteGrowthSupplement(#S0025)and
incubatedat37°Cundera5%CO2atmosphere.Forexperiments,primaryNHMsbetweenpassages
4and7wereused.TheviabilityofculturedcellswasassessedusingaCellCountingKit8(CCK8)
asdescribedbythemanufacturer(DOJINDO,Tokyo,Japan).
4.3.MeasurementofMelaninContent
Melanincontentwasdeterminedasdescribedinpreviousreports[2–4].Briefly,B16cellswere
treatedwiththeindicatedconcentrationsofglucoseinthepresenceofαMSH(200nM)for72h.
NHMsweretreatedwiththeindicatedconcentrationsofglucosefor4d.Thereafter,allcellswere
washedwithphosphatebufferedsaline(PBS)anddissolvedin1NNaOHat60°Cfor1h.Celllysates
weretransferredtoa96wellplate,andabsorbancewasmeasuredat405nm.Thevalueswere
normalizedbasedontheproteinconcentrationsineachsamplewell.
4.4.MushroomTyrosinaseActivityAssay
Weinvestigatedthedirecteffectsoftheindicatedconcentrationsofglucoseonmushroom
tyrosinaseactivity.Briefly,100μLofphosphatebuffercontainingglucosewasmixedwithmushroom
tyrosinase(10units/well)andcombinedwith50μLof0.03%LtyrosineorLDOPAindistilledwater.
Then,themixturewasincubatedtogetherat37°Cfor10min,andabsorbancewasmeasuredat405
nm.Kojicacid(KA),awellknownantityrosinaseagent,wasusedasareferencecompound.
4.5.IntracellularTyrosinaseActivityAssay
Briefly,B16cellsorNHMsweretreatedwiththeindicatedconcentrationsofglucoseforthe
indicatedtimes.Then,cellswerewashedwithPBSandlysedbyincubationin50mMphosphate
buffer(pH6.8)containing1%TritonX100and0.1mMphenylmethylsulfonylfluoride.Cellular
lysateswerethencentrifugedat12,000rpmat4°Cfor20min.Thesupernatantcontainingcellular
tyrosinasewascollectedandtheproteincontentwasdeterminedfornormalization.Thecellular
extractwasincubatedwithLDOPAinphosphatebufferanddopachromeformationwasmonitored
bymeasuringabsorbanceat405nmwithin30min.
4.6.RNAisolationandRealTimeQuantitativeReverseTranscriptionPolymeraseChainReaction(qRT
PCR)
TodeterminerelativemRNAexpressionofselectedgenes,totalRNAwasisolatedwithTRIzol
(Invitrogen,CA,USA),accordingtothemanufacturer’sinstructions,and4μgRNAwasreverse
transcribedintocDNAusingRTpremix(Bioneer,Seoul,SouthKorea).QuantitativePCRwas
performedusinganABI7500FastRealTimePCRSystem(AppliedBiosystems,FosterCity,CA,
USA).TheqRTPCRprimersetsfortyrosinaseandTyrp1werepurchasedfromAppliedBiosystems,
andTaqManGeneExpressionAssaykits(AppliedBiosystems)wereusedforamplification.Target
geneexpressionwasnormalizedtothatofthehousekeepinggeneencodingribosomalproteinlateral
stalksubunitP0(RPLP0).Relativequantizationwasperformedusingthecomparative∆Ctmethod
accordingtothemanufacturer’sinstructions.
Int.J.Mol.Sci.2020,21,173611of13
4.7.WesternBlotting
CellswerewashedtwicewithcoldPBSandthenlysedinicecoldmodifiedRIPAbuffer(Cell
SignalngTechnology,MA,USA)containingproteaseinhibitors(Calbiochem,LaJolla,CA,USA).
Totalproteinconcentrationwasdetermined,andtheproteinswereresolvedbySDSPAGEon4–12%
gradientBisTrisgels(ThermoFisherScientific,Waltham,MA,USA),transferredtonitrocellulose
membranes(ThermoFisherScientific).Aftertransfer,membraneswereblockedin5%blocking
solution.Membraneswereincubatedwithprimaryantibodiesat4°Cfor24h,washedwithTris
bufferedsalinecontaining0.1%Tween20(TBST),andexposedtoperoxidaseconjugatedsecondary
antibodiesfor1hatroomtemperature.MembraneswererinsedthreetimeswithTBST.
ChemiuminescentsignalwasdevelopedusingWesternblottingECLreagent(GEHealthcare,
Hatfield,UK).
4.8.ThreeDimensional(3D)HumanSkinEquivalent
WeusedMelanoDerm(MEL300B;MatTekCorp.,Ashland,MA,USA)asahumanskintissue
model.Thisviable,reconstituted,3Dhumanskinequivalentwasderivedfromblackdonorsand
containsnormalmelanocytesandkeratinocytes.MelanoDermwasgrownattheairliquidinterface
inEPI100NMM113medium(MatTekCorp,Ashland,MA,USA).Priortoglucosetreatment,tissues
werewashedwith1mLPBStoremoveresidualcompounds.GlucosewasdissolvedinPBS.Final
concentrationofglucosewas2%.ThecontrolsamplewastreatedonlywithPBS.Glucosewasapplied
toMelanoDermondays1,4,6,8,11,13,and15.After18days,MelanoDermtissueswerefixedin4%
bufferedformaldehyde,embeddedinparaffin,cuttoathicknessof3μm,andsubjectedtoH&Eand
F&Mstaining.TheviabilityofthetissuesampleswasassessedusingaCellCountingKit8(CCK8)
asdescribedbythemanufacturer(DOJINDO,Tokyo,Japan).PigmentationoftheMelanoDermwas
assessedbycomparingthechangeinL*value.
4.9.TwoPhotonExcitationFluorescence(TPEF)Imaging
Tovisualizethedistributionofmelanininthe3Dhumanskinequivalent,weperformedTPEF
imaging,asdescribedinourpreviousreports[3,4].Briefly,eachMelanoDermpreparationwasfixed
in4%formalinfor24hat4°C,andthenwashedwithPBS/0.1%BSA(bovineserumalbumin,Merck,
Branchburg,NJ,USA).TheTPEFimageswereacquiredfromthebasallayertomeasureintracellular
melanininthemelanocytelayer.RelativeTPEFsignalintensitiesformelanininthemeasurement
volumewerequantifiedusingImageProPremier3Dsoftware(MediaCybernetics,Inc.,Bethesda,
MD,USA).
4.10.LlactateAssay
B16cellswereseededat1.0 × 105cellsperwellina12wellplate.Aftertreatmentwithglucose
for3days,extracellularlactatelevelswerequantifiedusinganLlactatecolorimetricassaykit
(Abcam,ab65331,Cambridge,UK)accordingtothemanufacturer’sprotocol.
4.11.StatisticalAnalysis
Dataareexpressedasmeans±SDs(standarddeviations),andstatisticalsignificancewas
determinedbyStudent’sttest.Apvalue<0.05wasconsideredstatisticallysignificant.
AuthorContributions:Conceptualization,HyoungJuneKim,JongsungLeeandChangSeokLee;Datacuration,
SungHoonLee;Investigation,SungHoonLee;Methodology,SungHoonLee,IlHongBaeandEunSooLee;
Supervision,JongsungLeeandChangSeokLee;Writing—originaldraft,SungHoonLeeandChangSeokLee;
Writing—reviewandediting,JongsungLee.Allauthorshavereadandagreedtothepublishedversionofthe
manuscript.
Funding:ThisresearchwasfundedbytheBasicScienceResearchProgramthroughtheNationalResearch
FoundationofKorea(NRF)fundedbytheMinistryofEducation(GrantNumber:2018R1D1A1B07049402).
Int.J.Mol.Sci.2020,21,173612of13
ConflictsofInterest:Theauthorsdeclarenoconflictofinterest.
Abbreviations
α‐MSHαmelanocytestimulatinghormone
Tyrp1tyrosinaserelatedprotein1
MITFmicrophthalmiaassociatedtranscriptionfactor
NHMsnormalhumanmelanocytes
TPEFtwophotonexcitationfluorescence
LXRliverXreceptor
AHAsalphahydroxyacids
H&Ehematoxylinandeosin
F&MFontanaMasson
References
1. Swalwell,H.;Latimer,J.;Haywood,R.M.;BirchMachin,M.A.Investigatingtheroleofmelaninin
UVA/UVB‐ andhydrogenperoxideInducedcellularandmitochondrialROSproductionand
mitochondrialDNAdamageinhumanmelanomacells.FreeRadic.Biol.Med.2012,52,626–634.
2. Lee,C.S.;Jang,W.H.;Park,M.;Jung,K.;Baek,H.S.;Joo,Y.H.;Park,Y.H.;Lim,K.M.Anoveladamantyl
benzylbenzamidederivative,AP736,suppressesmelanogenesisthroughtheinhibitionofcAMPPKA
CREBActivatedmicrophthalmiaAssociatedtranscriptionfactorandtyrosinaseexpression.Exp.Dermatol.
2013,22,762–764.
3. Lee,J.H.;Lee,E.S.;Bae,I.H.;Hwang,J.A.;Kim,S.H.;Kim,D.Y.;Park,N.H.;Rho,H.S.;Kim,Y.J.;Oh,S.G.;
etal.AntimelanogenicEfficacyofMelasolv(3,4,5TrimethoxycinnamateThymolEster)inMelanocytesand
ThreeDimensionalHumanSkinEquivalent.SkinPharmacol.Physiol.2017,30,190–196.
4. Bae,I.H.;Lee,E.S.;Yoo,J.W.;Lee,S.H.;Ko,J.Y.;Kim,Y.J.;Lee,T.R.;Kim,D.Y.;Lee,C.S.Mannosylerythritol
lipidsinhibitmelanogenesisviasuppressingERKCREBMITFTyrosinasesignallinginnormalhuman
melanocytesandathreeDimensionalhumanskinequivalent.Exp.Dermatol.2019,28,738–741.
5. Bin,B.H.;Kim,S.T.;Bhin,J.;Lee,T.R.;Cho,E.G.ThedevelopmentofsugarBasedantiMelanogenicagents.
Int.J.Mol.Sci.2016,17,583.
6. Kumari,S.;TienGuanThng,S.;KumarVerma,N.;Gautam,H.K.MelanogenesisInhibitors.Acta.Derm.
Venereol.2018,98,924–931.
7. Ando,H.;Kondoh,H.;Ichihashi,M.;Hearing,V.J.Approachestoidentifyinhibitorsofmelanin
biosynthesisviathequalitycontroloftyrosinase.J.InvestDermatol.2007,127,751–761.
8. Hwang,J.S.;Lee,H.Y.;Lim,T.Y.;Kim,M.Y.;Yoon,T.J.DisruptionoftyrosinaseglycosylationbyN
acetylglucosamineanditsdepigmentingeffectsinguineapigskinandinhumanskin.J.Dermatol.Sci.2011,
63,199–201.
9. Lee,C.S.;Baek,H.S.;Bae,I.H.;Choi,S.J.;Kim,Y.J.;Lee,J.H.;Kim,J.W.Depigmentationefficacyof
galacturonicacidthroughtyrosinaseregulationinB16murinemelanomacellsandathreeDimensional
humanskinequivalent.Clin.Exp.Dermatol.2018,43,708–712.
10. Nakamura,S.;Kunikata,T.;Matsumoto,Y.;Hanaya,T.;Harashima,A.;Nishimoto,T.;Ushio,S.Effectsof
anonCyclodextrincycliccarbohydrateonmousemelanomacells:Characterizationofanewtypeof
hypopigmentingsugar.PLoSONE2017,12,e0186640.
11. Khan,M.T.Noveltyrosinaseinhibitorsfromnaturalresources–Theircomputationalstudies.Curr.Med.
Chem.2012,19,2262–2272.
12. Solano,F.;Briganti,S.;Picardo,M.;GhanemG.Hypopigmentingagents:Anupdatedreviewonbiological,
chemicalandclinicalaspects.Pigment.Cell.Res.2006,19,550–571.
Int.J.Mol.Sci.2020,21,173613of13
13. Lee,C.S.;Joo,Y.H.;Baek,H.S.;Park,M.;Kim,J.H.;Shin,H.J.;Park,N.H.;Lee,J.H.;Park,Y.H.;Shin,S.S.;
Lee,H.K.Differenteffectsoffivedepigmentarycompounds,rhododendrol,raspberryketone,
monobenzone,rucinolandAP736onmelanogenesisandviabilityofhumanepidermalmelanocytes.Exp.
Dermatol.2016,25,44–49.
14. Mulukutla,B.C.;Khan,S.;Lange,A.;Hu,W.S.Glucosemetabolisminmammaliancellculture:New
insightsfortweakingvintagepathways.Trends.Biotechnol.2010,28,476–484.
15. Usuki,A.;Ohashi,A.;Sato,H.;Ochiai,Y.;Ichihashi,M.;Funasaka,Y.Theinhibitoryeffectofglycolicacid
andlacticacidonmelaninsynthesisinmelanomacells.Exp.Dermatol.2003,12,43–50.
16. Lin,S.;Li,L.;Li,M.;Gu,H.;Chen,X.RaffinoseincreasesautophagyandreducescelldeathinUVB
Irradiatedkeratinocytes.J.Photochem.Photobiol.B2019,201,111653.
17. Chen,X.;Li,M.;Li,L.;Xu,S.;Huang,D.;Ju,M.;Huang,J.;Chen,K.;Gu,H.Trehalose,sucroseandraffinose
arenovelactivatorsofautophagyinhumankeratinocytesthroughanmTORIndependentpathway.Sci.
Rep.2016,6,28423.
18. Yamada,K.;Matsushita,K.;Wang,J.;Kanekura,T.TopicalGlucoseInducesClaudin1andFilaggrin
ExpressioninaMouseModelofAtopicDermatitisandinKeratinocyteCulture,ExertingAnti
InflammatoryEffectsbyRepairingSkinBarrierFunction.Acta.Derm.Venereol.2018,98,19–25.
19. Spravchikov,N.;Sizyakov,G.;Gartsbein,M.;Accili,D.;Tennenbaum,T.;Wertheimer,E.Glucoseeffects
onskinkeratinocytes:Implicationsfordiabetesskincomplications.Diabetes2001,50,1627–1635.
20. Castrillo,A.;Tontonoz,P.Nuclearreceptorsinmacrophagebiology:Atthecrossroadsoflipidmetabolism
andinflammation.Annu.Rev.Cell.Dev.Biol.2004,20,455–480.
21. Lee,C.S.;Park,M.;Han,J.;Lee,J.H.;Bae,I.H.;Choi,H.;Son,E.D.;Park,Y.H.;Lim,K.M.LiverXreceptor
activationinhibitsmelanogenesisthroughtheaccelerationofERKMediatedMITFdegradation.J.Invest.
Dermatol.2013,133,1063–1071.
22. Mitro,N.;Mak,P.A.;Vargas,L.;Godio,C.;Hampton,E.;Molteni,V.;Kreusch,A.;Saez,E.Thenuclear
receptorLXRisaglucosesensor.Nature2007,445,219–223.
23. Schurr,A.Carbohydrate;IntechOpen:London,UK,2017;pp.21–35.
24. Scott,D.;Richardson,A.;Filipp,F.;Knutzen,C.;Chiang,G.;Ronai,Z.;Osterman,A.;Smith,J.Comparative
metabolicFluxprofilingofmelanomacelllines:BeyondtheWarburgeffect.J.Biol.Chem.2011,286,42626–
42634.
25. Tang,S.C.;Yang,J.H.DualEffectsofAlphaHydroxyAcidsontheSkin.Molecules2018,23,863.
©2020bytheauthors.LicenseeMDPI,Basel,Switzerland.Thisarticleisanopenaccess
articledistributedunderthetermsandconditionsoftheCreativeCommonsAttribution
(CCBY)license(http://creativecommons.org/licenses/by/4.0/).
... The expanding ozone holes in the atmosphere lead to the growing danger of UV skin damage, making the pharmaceutical industry increase the production of new types of sun protection (Smit et al., 2009;Zastrow et al., 2017), which requires the new systems for testing them in vitro. At present, the mainly used testsystems are primary cultures of melanocytes and keratinocytes (Lei et al., 2002;Lee et al., 2020), melanoma cell lines Lee et al., 2020), the commercial tissue equivalents (EpiSkin TM , MelanoDerm TM , and others) (Costin and Raabe, 2013;Kim et al., 2017;Meena and Mohandass, 2019;Lee et al., 2020), and tissue equivalents obtained from cells with induced pluripotency (iPSC) (Gledhill et al., 2015). These approaches have some limitations, including non-physiological conditions (for 2D cultures), difficulties in analysis, and expensiveness (for commercial tissue equivalents). ...
... The expanding ozone holes in the atmosphere lead to the growing danger of UV skin damage, making the pharmaceutical industry increase the production of new types of sun protection (Smit et al., 2009;Zastrow et al., 2017), which requires the new systems for testing them in vitro. At present, the mainly used testsystems are primary cultures of melanocytes and keratinocytes (Lei et al., 2002;Lee et al., 2020), melanoma cell lines Lee et al., 2020), the commercial tissue equivalents (EpiSkin TM , MelanoDerm TM , and others) (Costin and Raabe, 2013;Kim et al., 2017;Meena and Mohandass, 2019;Lee et al., 2020), and tissue equivalents obtained from cells with induced pluripotency (iPSC) (Gledhill et al., 2015). These approaches have some limitations, including non-physiological conditions (for 2D cultures), difficulties in analysis, and expensiveness (for commercial tissue equivalents). ...
... The expanding ozone holes in the atmosphere lead to the growing danger of UV skin damage, making the pharmaceutical industry increase the production of new types of sun protection (Smit et al., 2009;Zastrow et al., 2017), which requires the new systems for testing them in vitro. At present, the mainly used testsystems are primary cultures of melanocytes and keratinocytes (Lei et al., 2002;Lee et al., 2020), melanoma cell lines Lee et al., 2020), the commercial tissue equivalents (EpiSkin TM , MelanoDerm TM , and others) (Costin and Raabe, 2013;Kim et al., 2017;Meena and Mohandass, 2019;Lee et al., 2020), and tissue equivalents obtained from cells with induced pluripotency (iPSC) (Gledhill et al., 2015). These approaches have some limitations, including non-physiological conditions (for 2D cultures), difficulties in analysis, and expensiveness (for commercial tissue equivalents). ...
Article
Full-text available
Pigmentation is the result of melanin synthesis, which takes place in melanocytes, and its further distribution. A dysregulation in melanocytes' functionality can result in the loss of pigmentation, the appearance of pigment spots and melanoma development. Tissue engineering and the screening of new skin-lightening drugs require the development of simple and reproducible in vitro models with maintained functional activity. The aim of the study was to obtain and characterize spheroids from normal human melanocytes as a three-dimensional multicellular structure and as a test system for skin-lightening drug screening. Melanocytes are known to lose their ability to synthesize melanin in monolayer culture. When transferred under non-adhesive conditions in agarose multi-well plates, melanocytes aggregated and formed spheroids. As a result, the amount of melanin elevated almost two times within seven days. MelanoDerm™ (MatTek) skin equivalents were used as a comparison system. Cells in spheroids expressed transcription factors that regulate melanogenesis: MITF and Sox10, the marker of developed melanosomes—gp100, as well as tyrosinase (TYR)—the melanogenesis enzyme and melanocortin receptor 1 (MC1R)—the main receptor regulating melanin synthesis. Expression was maintained during 3D culturing. Thus, it can be stated that spheroids maintain melanocytes' functional activity compared to that in the multi-layered MelanoDerm™ skin equivalents. Culturing both spheroids and MelanoDerm™ for seven days in the presence of the skin-lightening agent fucoxanthin resulted in a more significant lowering of melanin levels in spheroids. Significant down-regulation of gp100, MITF, and Sox10 transcription factors, as well as 10-fold down-regulation of TYR expression, was observed in spheroids by day 7 in the presence of fucoxanthin, thus inhibiting the maturation of melanosomes and the synthesis of melanin. MelanoDerm™ samples were characterized by significant down-regulation of only MITF, Sox10 indicating that spheroids formed a more sensitive system allowed for quantitative assays. Collectively, these data illustrate that normal melanocytes can assemble themselves into spheroids—the viable structures that are able to accumulate melanin and maintain the initial functional activity of melanocytes. These spheroids can be used as a more affordable and easy-to-use test system than commercial skin equivalents for drug screening.
... 31 The anti-melanogenic effect of glucose was observed. 32 cWBC is obtained from cell extract. It is possible to contain fatty acid from cell membrane as well as sugar. ...
Article
Ultraviolet (UV) radiation generates a range of biological effects in the skin, which includes premature skin aging, hyperpigmentation, and cancer. Therefore, the development of new effective agents for UV-related skin damage remains a challenge in the pharmaceutical industry. This study aims to test the inhibitory effect of crocodile white blood cell (cWBC) extract, a rich source of bioactive peptides, on ultraviolet B (UVB)-induced melanocyte pigmentation. The results showed that cWBC (6.25-400 μg/mL) could inhibit tyrosinase without adduct formation by 12.97 ± 4.20% on average. cWBC pretreatment (25-100 μg/mL) had no cytotoxicity and reduced intracellular melanin to 111.17 ± 5.20% compared with 124.87 ± 7.43 for UVB condition. The protective role of cWBC pretreatment against UVB was exhibited by the promotion of cell proliferation and the prevention of UVB-induced morphological change as observed from F actin staining. The decrease of microphthalmia-associated transcription factor expression levels after cWBC pretreatment might be a mechanism by which cWBC suppresses UVB-induced pigmentation. These results suggest that cWBC could be beneficial for the prevention of UVB-induced skin pigmentation.
... ab65331) following the protocol provided by the manufacturer followed in previous studies. 54,55 In brief, around 2X10 6 cells were cultured and harvested. After ice-cold PBS wash, they were resuspended in lactate assay buffer and homogenized well immediately. ...
Article
Alteration in glucose homeostasis during cancer metabolism is an important phenomenon. Though several important transcription factors have been well studied in the context of the regulation of metabolic gene expression, the role of epigenetic readers in this regard remains still elusive. Epigenetic reader protein transcription factor 19 (TCF19) has been recently identified as a novel glucose and insulin‐responsive factor that modulates histone posttranslational modifications to regulate glucose homeostasis in hepatocytes. Here we report that TCF19 interacts with a non‐histone, well‐known tumor suppressor protein 53 (p53) and co‐regulates a wide array of metabolic genes. Among these, the p53‐responsive carbohydrate metabolic genes Tp53‐induced glycolysis and apoptosis regulator (TIGAR) and Cytochrome C Oxidase assembly protein 2 (SCO2), which are the key regulators of glycolysis and oxidative phosphorylation respectively, are under direct regulation of TCF19. Remarkably, TCF19 can form different transcription activation/repression complexes which show substantial overlap with that of p53, depending on glucose‐mediated variant stress situations as obtained from IP/MS studies. Interestingly, we observed that TCF19/p53 complexes either have CBP or HDAC1 to epigenetically program the expression of TIGAR and SCO2 genes depending on short‐term high glucose or prolonged high glucose conditions. TCF19 or p53 knockdown significantly altered the cellular lactate production and led to increased extracellular acidification rate. Similarly, OCR and cellular ATP production were reduced and mitochondrial membrane potential was compromised upon depletion of TCF19 or p53. Subsequently, through RNA‐Seq analysis from patients with hepatocellular carcinoma, we observed that TCF19/p53‐mediated metabolic regulation is fundamental for sustenance of cancer cells. Together the study proposes that TCF19/p53 complexes can regulate metabolic gene expression programs responsible for mitochondrial energy homeostasis and stress adaptation.
Article
Full-text available
2'-fucosyllactose (2'-FL) is the most abundant human milk oligosaccharide (HMO) present in breast milk, promoting the growth of beneficial microorganisms in the gut and aiding in the relief of allergic and inflammatory reactions. In this study, the anti-melanogenic effects of 2'-FL, and its potential for application in whitening cosmetics, were evaluated. MTT assay was performed on MNT-1 cells, human-derived melanocytes. 2'-FL was treated and replaced at 48 h intervals for 7 days, and it was confirmed that there was no cytotoxicity at 20 g/L or less, while a 40% reduction in melanin production was also observed. Western blot analysis of TYR and TYRP1, factors involved in melanogenesis, revealed that 2'-FL treatment reduced their expression levels. In addition, 2'-FL application and observation of the autophagy marker microtubule-associated protein 1 light chain 3 (LC3) revealed it was converted from LC3-Ι to LC3-Π, indicating increased autophagy. Likewise, confocal microscopy revealed an increase in LC3 puncta after 2'-FL treatment. Therefore, it is suggested that 2'-FL-mediated activation of autophagy reduces melanogenesis by inhibiting the expression levels of TYR and TYRP1 proteins. In conclusion, it has been confirmed that 2'-FL induces autophagy and suppresses melanin production, so its potential as a whitening cosmetic material is expected.
Article
Mutations in SLC45A2 are responsible for oculocutaneous albinism type 4 in many species and associated with melanoma susceptibility, but the molecular mechanism is unclear. Here, we used Slc45a2-deficient melanocyte and mouse models to elucidate the roles of Slc45a2 in melanogenesis and melanoma metastasis. We find that the acidified cellular environment impairs the activity of key melanogenic enzyme tyrosinase in Slc45a2-deficient melanocytes. Slc45a2 is identified as a proton/glucose exporter in melanosomes, and its ablation increases acidification of melanosomal pH through enhanced glycolysis. Intriguingly, ¹³C-glucose labeled metabolic flux and biochemical assays show that melanosomes are active glucose-metabolizing organelles, indicating that elevated glycolysis mainly occurs in melanosomes due to Slc45a2-deficiency. Moreover, Slc45a2-deficiency significantly up-regulates the activities of glycolytic enzymes and PI3K/Akt signaling to promote glycolysis-dependent survival and metastasis of melanoma cells. Collectively, our study reveals that the H⁺/glucose exporter Slc45a2 mediates melanin synthesis and melanoma metastasis primarily via modulating melanosomal glucose metabolism.
Article
Dear Editor, A 31-year-old female presented to the pain management clinic with complaints of tightness and stiffness on the right distal posterolateral lower extremity over an area of a hyperpigmented scar secondary to a wound from a traffic accident 10 years ago. She denied a history of surgery, open wound, or fracture related to the accident. She reported aggravation of her symptoms in cold weather. Additionally, she endorsed a sensation of “skin pulling” during stretching exercises of her right calf. The patient reported paresthesia over the right sural nerve (SN) dermatome with a light touch and a “tense sensation” with ankle dorsiflexion. Ultrasonography (US) demonstrated entrapment of the right SN beneath the scar tissue; however, the typical signs of an anatomical bottleneck, a thinned nerve at the compression site with proximal swelling, were not noted. Power Doppler imaging did not show abnormal hypervascularity. The SN was released from the scar tissue with US-guided hydrodissection using 5% dextrose (5 mL). The patient had 30% symptomatic improvement in the “tense sensation” 2 weeks later, and the skin pigmentation faded as well. A repeat injection was performed at the patient's request. The patient returned to the outpatient clinic 2 months after the second injection. Further improvement of the “tightness sensation” and the hyperpigmented scar was noted and the patient’s reported satisfaction with the treatment was confirmed with a 5/5 rating (very satisfied) on a Likert scale. Additionally, the SN cross-sectional area was reduced from 0.07 to 0.04 cm² in the United States.
Article
The barrier function of the skin mainly relies on its outermost layer, the stratum corneum (SC), which is a lipid-protein composite biomaterial. In addition, SC contains a mixture of small polar compounds, the natural moisturizing factor (NMF). Most of the NMF components are solid at ambient relative humidities (RHs). We herein raise the question of what the effects of adding a mixture of chemicals to the SC are as compared to adding the single components. We use mixtures of glucose and NaCl, both present in NMF and in skin formulations, and the effects on SC were studied using solid-state NMR, wide angle X-ray diffraction, and sorption microbalance. The deliquescence RHs of the glucose-NaCl mixtures are lower than the individual chemicals. The same trend is also seen when these chemicals are added to SC, as the dissolution occurs at lower RH for the mixture than for the single chemicals. Notably, the mixtures can also increase the mobility of SC lipids and proteins more than the individual compounds. These data illustrate a synergistic effect by adding a mixture rather than a single polar compound to the SC, which may also have implications for the complex mixture NMF inside SC in dry conditions.
Article
Gestational diabetes mellitus (GDM) is a metabolic disorder whose major pathophysiological basis is demonstrated as placental insulin resistance (IR), while Smad4 always functions in the signal transduction of transforming growth factor beta (TGF‐β) pathway. Our study aims to figure out the role of Smad4 in an insulin resistance (IR) cellular model using placental trophoblast cell line. Importantly, HTR8‐Svneo cells, in the status of IR, indicated a significant increase in the expression of Smad4. Subsequently, the HTR8‐Svneo cell line with up‐regulated or depleted Smad4 was respectively achieved by the effective over‐expressed plasmid or siRNA of Smad4. We found out that the deficiency of Smad4 could promote the insulin sensitivity and restrict the inflammatory response in IR group of cells with significant augment in glucose uptake, up‐regulation of insulin signalling‐related molecules and attenuation in inflammatory biomarker expressions. On the contrary, the over‐expression of Smad4 showed a reversal effect on these alterations in IR group of cells. Besides, the positive effect of Smad4 on cell viability was also observed in our study. Significance of the study Gestational diabetes mellitus (GDM) is a metabolic disorder whose major pathophysiological basis is demonstrated as insulin resistance (IR). Importantly, our findings indicate that the deficiency of Smad4 significantly improves the insulin sensitivity and relieves the inflammation in the cellular model of IR. Besides, the positive effect of Smad4 on cell viability was also observed in our study. Our present findings provide novel insights for the investigation on molecular details about the GDM pathogenesis.
Article
Full-text available
Hyperpigmentation is caused by excessive production of melanin in melanocytes. Mannosylerythritol lipids (MELs) are glycolipid biosurfactants that are abundantly produced by yeasts and used commercially in cosmetics. However, the potential depigmenting efficacy of MELs has not been evaluated. In this study, the depigmentary effect of MELs was tested in primary normal human melanocytes (NHMs), α‐melanocyte‐stimulating hormone (MSH)‐stimulated B16 cells (murine melanoma cells) and a human skin equivalent (MelanoDerm) using photography, Fontana‐Masson (F&M) staining, and two‐photon microscopy. MELs significantly decreased the melanin contents in NHMs and α‐MSH‐stimulated B16 cells. Consistent with these findings, MELs treatment had a clear whitening effect in a human skin equivalent, brightening the tissue color and reducing the melanin content. The molecular mechanism underlying the anti‐melanogenic effect of MELs treatment was examined by real‐time PCR and Western blotting. Mechanistically, MELs clearly suppressed the gene expression levels of representative melanogenic enzymes, including tyrosinase, Tyrp‐1, and Tyrp‐2, by inhibiting the ERK/CREB/MiTF signaling pathway in NHMs. This work demonstrates for the first time that MELs exert whitening effects on human melanocytes and skin equivalent. Thus, we suggest that MELs could be developed as a potent anti‐melanogenic agent for effective whitening, beyond their use as a biosurfactant in cosmetics. This article is protected by copyright. All rights reserved.
Article
Full-text available
Abnormally high production of melanin or melanogenesis in skin melanocytes results in hyperpigmentation disorders, such as melasma, senile lentigines or freckles. These hyperpigmentary skin disorders can significantly impact an individual's appearance, and may cause emotional and psychological distress and reduced quality of life. A large number of melanogenesis inhibitors have been developed, but most have unwanted side-effects. Further research is needed to better understand the mechanisms of hyperpigmentary skin disorders and to develop potent and safe inhibitors of melanogenesis. This review summarizes the current understanding of melanogenesis regulatory pathways, the potential involvement of the immune system, various drugs in current use, and emerging treatment strategies to suppress melanogenesis.
Article
Full-text available
AHAs are organic acids with one hydroxyl group attached to the alpha position of the acid. AHAs including glycolic acid, lactic acid, malic acid, tartaric acid, and citric acid are often used extensively in cosmetic formulations. AHAs have been used as superficial peeling agents as well as to ameliorate the appearance of keratoses and acne in dermatology. However, caution should be exercised in relation to certain adverse reactions among patients using products with AHAs, including swelling, burning, and pruritus. Whether AHAs enhance or decrease photo damage of the skin remains unclear, compelling us to ask the question, is AHA a friend or a foe of the skin? The aim of this manuscript is to review the various biological effects and mechanisms of AHAs on human keratinocytes and in an animal model. We conclude that whether AHA is a friend or foe of human skin depends on its concentration. These mechanisms of AHAs are currently well understood, aiding the development of novel approaches for the prevention of UV-induced skin damage.
Article
Full-text available
Cyclic nigerosyl nigerose (CNN) is a cyclic tetrasaccharide that exhibits properties distinct from other conventional cyclodextrins. Herein, we demonstrate that treatment of B16 melanoma with CNN results in a dose-dependent decrease in melanin synthesis, even under conditions that stimulate melanin synthesis, without significant cytotoxity. The effects of CNN were prolonged for more than 27 days, and were gradually reversed following removal of CNN. Undigested CNN was found to accumulate within B16 cells at relatively high levels. Further, CNN showed a weak but significant direct inhibitory effect on the enzymatic activity of tyrosinase, suggesting one possible mechanism of hypopigmentation. While a slight reduction in tyrosinase expression was observed, tyrosinase expression was maintained at significant levels, processed into a mature form, and transported to late-stage melanosomes. Immunocytochemical analysis demonstrated that CNN treatment induced drastic morphological changes of Pmel17-positive and LAMP-1–positive organelles within B16 cells, suggesting that CNN is a potent organelle modulator. Colocalization of both tyrosinase-positive and LAMP-1–positive regions in CNN-treated cells indicated possible degradation of tyrosinase in LAMP-1–positive organelles; however, that possibility was ruled out by subsequent inhibition experiments. Taken together, this study opens a new paradigm of functional oligosaccharides, and offers CNN as a novel hypopigmenting molecule and organelle modulator.
Article
Full-text available
Atopic dermatitis (AD) is a chronic inflammatory skin disease. Corticosteroids, which are widely used for AD treatment, have adverse effects, and alternative treatments are urgently needed. This study examined the effect of topical application of high-dose glucose on inflamed skin in a murine model of AD. High-dose glucose treatment on the ear reduced dermatitis scores and ear thicknesses in mite antigen-treated NC/Nga mice. The levels of thymus and activation-regulated chemokine (TARC), Th cytokines (interleukin (IL)-4, IL-5, IL-12, IL-13, and (interferon) IFN-γ), and IgE were decreased in the serum of high-dose glucose-treated mice. Expression of claudin-1 and filaggrin was reduced in the ear epithelium in the NC/Nga mice. However, the reduced expression was restored by topical treatment with high-dose glucose. High-dose glucose also induced the expression of claudin-1 and filaggrin in cultured human skin keratinocytes. Co-stimulation with IL-4, IL-13, and thymic stromal lymphoprotein (TSLP) downregulated the expression of filaggrin in culture. However, high-dose glucose treatment restored the reduced expression of filaggrin. These results suggest that high-dose glucose treatment suppresses inflammation in the skin lesions by improving the skin barrier function.
Article
Full-text available
Trehalose is a natural disaccharide that is found in a diverse range of organisms but not in mammals. Autophagy is a process which mediates the sequestration, lysosomal delivery and degradation of proteins and organelles. Studies have shown that trehalose exerts beneficial effects through inducing autophagy in mammalian cells. However, whether trehalose or other saccharides can activate autophagy in keratinocytes is unknown. Here, we found that trehalose treatment increased the LC3-I to LC3-II conversion, acridine orange-stained vacuoles and GFP-LC3B (LC3B protein tagged with green fluorescent protein) puncta in the HaCaT human keratinocyte cell line, indicating autophagy induction. Trehalose-induced autophagy was also observed in primary keratinocytes and the A431 epidermal cancer cell line. mTOR signalling was not affected by trehalose treatment, suggesting that trehalose induced autophagy through an mTOR-independent pathway. mTOR-independent autophagy induction was also observed in HaCaT and HeLa cells treated with sucrose or raffinose but not in glucose, maltose or sorbitol treated HaCaT cells, indicating that autophagy induction was not a general property of saccharides. Finally, although trehalose treatment had an inhibitory effect on cell proliferation, it had a cytoprotective effect on cells exposed to UVB radiation. Our study provides new insight into the saccharide-mediated regulation of autophagy in keratinocytes.
Article
Full-text available
The regulation of melanin production is important for managing skin darkness and hyperpigmentary disorders. Numerous anti-melanogenic agents that target tyrosinase activity/stability, melanosome maturation/transfer, or melanogenesis-related signaling pathways have been developed. As a rate-limiting enzyme in melanogenesis, tyrosinase has been the most attractive target, but tyrosinase-targeted treatments still pose serious potential risks, indicating the necessity of developing lower-risk anti-melanogenic agents. Sugars are ubiquitous natural compounds found in humans and other organisms. Here, we review the recent advances in research on the roles of sugars and sugar-related agents in melanogenesis and in the development of sugar-based anti-melanogenic agents. The proposed mechanisms of action of these agents include: (a) (natural sugars) disturbing proper melanosome maturation by inducing osmotic stress and inhibiting the PI3 kinase pathway and (b) (sugar derivatives) inhibiting tyrosinase maturation by blocking N-glycosylation. Finally, we propose an alternative strategy for developing anti-melanogenic sugars that theoretically reduce melanosomal pH by inhibiting a sucrose transporter and reduce tyrosinase activity by inhibiting copper incorporation into an active site. These studies provide evidence of the utility of sugar-based anti-melanogenic agents in managing skin darkness and curing pigmentary disorders and suggest a future direction for the development of physiologically favorable anti-melanogenic agents.
Article
Autophagy is an important process for maintaining intracellular homeostasis. Our previous study demonstrated that autophagy was down-regulated in ultraviolet B (UVB)-irradiated keratinocytes. Raffinose is a natural oligosaccharide that serves as a novel activator of autophagy and as a balancing agent to regulate the diversity of environmental stress. However, whether raffinose balances ultraviolet stress through the autophagy activation pathway has yet to be established. In this study, we found that raffinose treatment inhibited the LDH release and trypan blue staining in UVB-challenged human keratinocytes cell line HaCaT but did not affect the cleavage of apoptotic markers Caspase-3 and PARP, as well as translocation into nucleus of other cell death markers Endonuclease G and AIF. Moreover, we confirmed that raffinose treatment enhanced autophagy flux in an MTOR-independent manner in HaCaT cells. Importantly, decrease of LC3-II turnover in UVB-irradiated keratinocytes could be rescued by raffinose treatment, indicating that raffinose treatment increased autophagy in UVB-irradiated HaCaT cells. Furthermore, the effect on cell death by raffinose was inhibited when autophagy was suppressed with either a small interfering RNA targeting ATG5 (siATG5) or autophagic inhibitor wortmannin. In conclusion, we demonstrated that raffinose increases MTOR-independent autophagy and reduces cell death in UVB-irradiated keratinocytes. Our study indicated that the natural agent raffinose presents the potential value in opposing photodamage.
Article
Sugar is a well‐known cosmetic ingredient for moisturizing skin with minimal side‐effects. Several reports have demonstrated an antimelanogenic effect of sugar in melanocytes. We evaluated the whitening efficacy of galacturonic acid (GA), the main component of pectin, as an anti‐melanogenic agent. GA significantly suppressed melanin synthesis and secretion in a concentration‐dependent manner in α‐melanocyte stimulating hormone‐treated B16 melanoma cells, and inhibited tyrosinase activity and expression at a dose of 10 mmol/L. In a three‐dimensional human skin equivalent (MelanoDerm), GA clearly brightened tissue colour. Haematoxylin and eosin and Fontana–Masson (F&M) staining of tissue sections revealed decreased melanin production without skin tissue collapse in the presence of GA. Interestingly, GA dramatically suppressed gene expression of the melanogenic proteins tyrosinase, tyrosinase‐related protein (TYRP)‐1 and microphthalmia‐associated transcription factor, but not TYRP‐2. The results support the utility of GA as an effective candidate antimelanogenic agent.
Article
Background/aims: Excessive melanogenesis often causes unaesthetic hyperpigmentation. In a previous report, our group introduced a newly synthesized depigmentary agent, Melasolv™ (3,4,5-trimethoxycinnamate thymol ester). In this study, we demonstrated the significant whitening efficacy of Melasolv using various melanocytes and human skin equivalents as in vitro experimental systems. Methods: The depigmentary effect of Melasolv was tested in melan-a cells (immortalized normal murine melanocytes), α-melanocyte-stimulating hormone (α-MSH)-stimulated B16 murine melanoma cells, primary normal human melanocytes (NHMs), and human skin equivalent (MelanoDerm). The whitening efficacy of Melasolv was further demonstrated by photography, time-lapse microscopy, Fontana-Masson (F&M) staining, and 2-photon microscopy. Results: Melasolv significantly inhibited melanogenesis in the melan-a and α-MSH-stimulated B16 cells. In human systems, Melasolv also clearly showed a whitening effect in NHMs and human skin equivalent, reflecting a decrease in melanin content. F&M staining and 2-photon microscopy revealed that Melasolv suppressed melanin transfer into multiple epidermal layers from melanocytes as well as melanin synthesis in human skin equivalent. Conclusion: Our study showed that Melasolv clearly exerts a whitening effect on various melanocytes and human skin equivalent. These results suggest the possibility that Melasolv can be used as a depigmentary agent to treat pigmentary disorders as well as an active ingredient in cosmetics to increase whitening efficacy.