品牌咨询
联系方式
公司地址
苏州工业园区生物纳米园A4#216
联系电话
4000-520-616 / 18915418616
传真号码
0512-67156496
电子邮箱
info@ebiomall.com
公司网址
https://www.ebiomall.com

Smartox/Selective blocker of Kv1.3/08SHK001-00100/0.1mg

价格
¥1497.60
货号:08SHK001-00100
浏览量:127
品牌:Smartox
服务
全国联保
正品保证
正规发票
签订合同
商品描述

ShK(StichodactylahelianthusNeurotoxin)hasbeenisolatedfromthevenomoftheCarribeanseaanemoneStoichactishelianthus.ShKinhibitsvoltage-dependentpotassiumchannels.ItblocksKv1.3(KCNA3)potentlyandalsoKv1.1(KCNA1),Kv1.4(KCNA4)andKv1.6(KCNA6)respectivelywithaKdof11pM,16pM,312pMand165pM.Interestingly,itwasalsodemonstratedthatShKpotentlyinhibitsthehKv3.2bchannelwithanIC50valueofapproximately0.6nM.


Description:

Productcode:N/A.Categories:Kv1.3channel,Potassiumchannels.Tags:165168-50-3,Kv1.3,TRAM-34.

AAsequence:Arg-Ser-Cys3-Ile-Asp-Thr-Ile-Pro-Lys-Ser-Arg-Cys12-Thr-Ala-Phe-Gln-Cys17-Lys-His-Ser-Met-Lys-Tyr-Arg-Leu-Ser-Phe-Cys28-Arg-Lys-Thr-Cys32-Gly-Thr-Cys35-OH
Disulfidebonds:Cys3-Cys35,Cys12-Cys28andCys17-Cys32
Length(aa):35
Formula:C169H274N54O48S7
MolecularWeight:4054.85Da
Appearance:Whitelyophilizedsolid
Solubility:waterandsalinebuffer
CASnumber:165168-50-3
Source:Synthetic
Purityrate:>97%

Reference:

DurablepharmacologicalresponsesfromthepeptideShK-186,aspecificKv1.3channelinhibitorthatsuppressesTcellmediatorsofautoimmunedisease

TheKv1.3channelisarecognizedtargetforpharmaceuticaldevelopmenttotreatautoimmunediseasesandorganrejection.ShK-186,aspecificpeptideinhibitorofKv1.3,hasshownpromiseinanimalmodelsofmultiplesclerosisandrheumatoidarthritis.Here,wedescribethepharmacokinetic-pharmacodynamicrelationshipforShK-186inratsandmonkeys.ThepharmacokineticprofileofShK-186wasevaluatedwithavalidatedhigh-performanceliquidchromatography-tandemmassspectrometrymethodtomeasurethepeptide’sconcentrationinplasma.Theseresultswerecomparedwithsingle-photonemissioncomputedtomography/computedtomographydatacollectedwithan¹¹¹In-1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraaceticacid-conjugateofShK-186toassesswhole-bloodpharmacokineticparametersaswellasthepeptide’sabsorption,distribution,andexcretion.AnalysisofthesedatasupportamodelwhereinShK-186isabsorbedslowlyfromtheinjectionsite,resultinginbloodconcentrationsabovetheKv1.3channel-blockingIC₅₀valueforupto7daysinmonkeys.PharmacodynamicstudiesonhumanperipheralbloodmononuclearcellsshowedthatbriefexposuretoShK-186resultedinsustainedsuppressionofcytokineresponsesandmaycontributetoprolongeddrugeffects.Indelayed-typehypersensitivity,chronicrelapsing-remittingexperimentalautoimmuneencephalomyelitis,andpristane-inducedarthritisratmodels,asingledoseofShK-186every2to5dayswasaseffectiveasdailyadmiNISTration.ShK-186’sslowdistributionfromtheinjectionsiteanditslongresidencetimeontheKv1.3channelcontributetotheprolongedtherapeuticeffectofShK-186inanimalmodelsofautoimmunedisease.

TarchaEJ.,etal.(2012)DurablepharmacologicalresponsesfromthepeptideShK-186,aspecificKv1.3channelinhibitorthatsuppressesTcellmediatorsofautoimmunedisease.JPharmacolExpTher.PMID: 22637724

ThebeneficialeffectofblockingKv1.3inthepsoriasiformSCIDmousemodel

TheKv1.3channelisimportantintheactivationandfunctionofeffectormemoryTcells.Recently,specificblockersoftheKv1.3channelhavebeendevelopedasapotentialtherapeuticoptionfordiverseautoimmunediseases.Inpsoriaticlesions,mostlymphocytesarememoryeffectorTcells.TheaimofthepresentstudywastodetecttheexpressionofKv1.3channelsinthesecellsinpsoriaticlesionsaswellasinhumanpsoriasiformskingraftsusingtheseverecombinedimmunodeficient(SCID)mousemodel.HistologicalandimmunohistochemicalstainingforKv1.3expressionandvariousinflammatoryMarkerswasperformedinsectionsobtainedfromsixpsoriaticpatientsand18beige-SCIDmicewithpsoriasiformhumanskingrafts.SixgraftedmiceweretreatedwithStichodactylahelianthusneurotoxin(ShK),aknownKv1.3blocker.TheresultsshowedanincreasednumberofKv1.3+cellsinthepsoriaticskinaswellasinthepsoriasiformskingraftsascomparedwithnormalskinandnormalskingrafts.InjectionsofShKshowedamarkedtherapeuticeffectinthreeofsixpsoriasiformskingrafts.AsignificantlydecreasednumberofKv1.3+cellswasobservedintheresponderscomparedwiththecontrolgrafts.Thispilotstudy,althoughperformedinasmallnumberofmice,revealsthepossIBLebeneficialeffectofKv1.3blockersinpsoriasispatients.

GilharA.,etal.(2011)ThebeneficialeffectofblockingKv1.3inthepsoriasiformSCIDmousemodel.JInvestDermatol.PMID: 20739949

Modelingthebindingofthreetoxinstothevoltage-gatedpotassiumchannel(Kv1.3)

Theconductionpropertiesofthevoltage-gatedpotassiumchannelKv1.3anditsmodesofinteractionwithseveralpolypeptidevenomsareexaminedusingBrowniandynamicssimulationsandmoleculardynamicscalculations.Employinganopen-statehomologymodelofKv1.3,wefirstdeterminecurrent-voltageandcurrent-concentrationcurvesandascertainthatsimulatedresultsaccordwithexperimentalmeasurements.Wetheninvestigate,usingamoleculardockingmethodandmoleculardynamicssimulations,thecomplexesformedbetweentheKv1.3channelandseveralKv-specificpolypeptidetoxinsthatareknowntointerferewiththeconductingmechanismsofseveralclassesofvoltage-gatedK(+)channels.ThedepthsofpotentialofmeanforceencounteredbycharyBDotoxin,α-KTx3.7(alsoknownasOSK1)andShKare,respectively,-19,-27,and-25kT.Thedissociationconstantscalculatedfromtheprofilesofpotentialofmeanforcecorrespondcloselytotheexperimentallydeterminedvalues.Wepinpointtheresiduesinthetoxinsandthechannelthatarecriticalfortheformationofthestablevenom-channelcomplexes.

ChenR.,etal.(2011)Modelingthebindingofthreetoxinstothevoltage-gatedpotassiumchannel(Kv1.3).BiophysJ.PMID: 22261053

BlockadeofT-lymphocyteKCa3.1andKv1.3channelsasnovelimmunosuppressionstrategytopreventkidneyallograftrejection

Currently,thereisanunmetclinicalneedfornovelimmunosuppressiveagentsforlong-termpreventionofkidneytransplantrejectionasalternativestothenephrotoxiccalcineurininhibitorcyclosporine(CsA).RecentstudieshaveshownthatK(+)channelshaveacrucialroleinT-lymphocyteactivity.WeinvestigatedwhethercombinedblockadeoftheT-cellK(+)channelsK(Ca)3.1andK(v)1.3,bothofwhichregulatecalciumsignalingduringlymphocyteactivation,iseffectiveinpreventionofrejectionofkidneyallograftsfromFisherratstoLewisrats.AllrecipientswereinitiallytreatedwithCsA(5mg/kgd)for7days.Inratswithintactallograftfunction,treatmentwascontinuedfor10dayswitheitherCsA(5mg/kgd),oracombinationofTRAM-34(K(Ca)3.1inhibitor;120mg/kgd)plusStichodactylahelianthustoxin(ShK,K(v)1.3inhibitor;80microg/kg3timesdaily),orvehiclealone.Kidneysectionswerestainedwithperiodicacid-Schifforhematoxylin-eosinandhistochemicallyformarkersofmacrophages(CD68),T-lymphocytes(CD43),orcytotoxicT-cells(CD8).OurresultsshowedthattreatmentwithTRAM-34andShKreducedtotalinterstitialmononuclearcellinfiltration(-42%)andthenumberofCD43+T-cells(-32%),cytotoxicCD8+T-cells(-32%),andCD68+macrophages(-26%)inallograftswhencomparedtovehicletreatmentalone.EfficacyofTRAM-34/ShKtreatmentwascomparablewiththatofCsA.Inaddition,novisibleorgandamageorotherdiscernibleadverseeffectswereobservedwiththistreatment.Thus,selectiveblockadeofT-lymphocyteK(Ca)3.1andK(v)1.3channelsmayrepresentanovelalternativetherapyforpreventionofkidneyallograftrejection.

GrgicI.,etal.(2009)BlockadeofT-lymphocyteKCa3.1andKv1.3channelsasnovelimmunosuppressionstrategytopreventkidneyallograftrejection.TransplantProc.PMID: 19715983

MolecularmechanismoftheseaanemonetoxinShKrecognizingtheKv1.3channelexploredbydockingandmoleculardynamicsimulations

ComputationalmethodsareemployedtosimulatetheinteractionoftheseaanemonetoxinShKincomplexwiththevoltage-gatedpotassiumchannelKv1.3frommice.Alloftheavailable20structuresofShKintheProteinDataBankwereconsideredforimprovingtheperformanceoftherigidproteindockingofZDOCK.ThetrADItionalandnovelbindingmodeswereobtainedamongalargenumberofpredictedcomplexesbyusingclusteringanalysis,screeningwithexpertknowledge,energyminimization,andmoleculardynamicsimulations.Thequalityandvalidityoftheresultingcomplexeswerefurtherevaluatedtoidentifyafavorablecomplexstructureby500psmoleculardynamicsimulationsandthechangeofbindingfreeenergieswithacomputationalalaninescanningtechnique.ThenovelandreasonableShK-Kv1.3complexstructurewasfoundtobedifferentfromthetraditionalmodelbyusingtheLys22residuetoblockthechannelpore.FromtheresultingstructureoftheShK-Kv1.3complex,ShKmainlyassociatesthechanneloutervestibulewithitssecondhelicalsegment.StructuralanalysisfirstrevealedthattheLys22residuesidechainoftheShKpeptidejusthangsbetweenCandDchainsoftheKv1.3channelinsteadofphysicallyblockingthechannelpore.TheobviouslossoftheShKSer20AlaandTyr23AlamutantbindingABIlitytotheKv1.3channeliscausedbytheconformationalchange.ThefivehydrogenbondsbetweenArg24inShKandH404(A)andD402(D)inKv1.3makeArg24themostcrucialforitsbindingtotheKv1.3channel.BesidesthedetailedinteractionbetweenShKandKv1.3attheatomlevel,thesignificantconformationalchangeinducedbytheinteractionbetweentheShKpeptideandtheKv1.3channel,accompaniedbythegradualdecreaseofbindingfreeenergies,stronglyimpliesthatthebindingoftheShKpeptidetowardtheKv1.3channelisadynamicprocessofconformationalrearrangementandenergystabilization.AllofthesecanacceleratethedevelopmentofShKstructure-basedimmunosuppressants.

JinL,WuY.(2007)MolecularmechanismoftheseaanemonetoxinShKrecognizingtheKv1.3channelexploredbydockingandmoleculardynamicsimulations.JChemInfModel. PMID: 17718553

K+channelblockers:noveltoolstoinhibitTcellactivationleadingtospecificimmunosuppression

DuringthelasttwodecadessincetheidentificationandcharacterizationofTcellpotassiumchannelsgreatadvanceshavebeenmadeintheunderstandingoftheroleofthesechannelsinTcellfunctions,especiallyinantigen-inducedactivation.TheirlimitedtissuedistributionandtherecentdiscoverythatdifferentTcellsubtypescarryingoutdistinctimmunefunctionsshowspecificexpressionlevelsofthesechannelshavemadeTcellpotassiumchannelsattractivetargetsforimmunomodulatorydrugs.Manytoxinsofvariousanimalspeciesandastructurallydiversearrayofsmallmoleculesinhibitingthesechannelswithvaryingaffinityandselectivitywerefoundandtheirsuccessfuluseinimmunosuppressioninvivowasalsodemonstrated.Betterunderstandingofthetopologicaldifferencesbetweenpotassiumchannelpores,detailedknowledgeoftoxinandsmall-moleculestructuresandtheidentificationofthebindingsitesofblockingcompoundsmakeitpossibletoimprovetheselectivityandaffinityoftheleadcompoundsbyintroducingmodificationsbasedonstructuralinformation.Inthisreviewthebasicpropertiesandphysiologicalrolesofthevoltage-gatedKv1.3andtheCa2+-activatedIKCa1potassiumchannelsarediscussedalongwithanoverviewofcompoundsinhibitingthesechannelsandapproachesaimingatproducingmoreefficientmodulatorsofimmunefunctionsforthetreatmentofdiseaseslikesclerosismultiplexandtypeIdiabetes.

PanyiG,etal.(2006)K+channelblockers:noveltoolstoinhibitTcellactivationleadingtospecificimmunosuppression.CurrPharmDes. PMID: 16787250

Stichodactylahelianthuspeptide,apharmacologicaltoolforstudyingKv3.2channels

Voltage-gatedpotassium(Kv)channelsregulatemanyphysiologicalfunctionsandrepresentimportanttherapeutictargetsinthetreatmentofseveralclinicaldisorders.Althoughsomeofthesechannelshavebeenwell-characterized,thestudyofothers,suchasKv3channels,hasbeenhinderedbecauseoflimitedpharmacologicaltools.ThecurrentstudywasinitiatedtoidentifypotentblockersoftheKv3.2channel.Chinesehamsterovary(CHO)-K1cellsstablyexpressinghumanKv3.2b(CHO-K1.hKv3.2b)wereestablishedandcharacterized.Stichodactylahelianthuspeptide(ShK),isolatedfromS.helianthusvenomandaknownhigh-affinityblockerofKv1.1andKv1.3channels,wasfoundtopotentlyinhibit86Rb+effluxfromCHO-K1.hKv3.2b(IC50approximately0.6nM).InelectrophysiologicalrecordingsofKv3.2bchannelsexpressedinXenopuslaevisoocytesorinplanarpatch-clampstudies,ShKinhibitedhKv3.2bchannelswithIC50valuesofapproximately0.3and6nM,respectively.DespitethepresenceofKv3.2proteininhumanpancreaticbetacells,ShKhasnoeffectontheKvcurrentofthesecells,suggestingthatitisunlikelythathomotetramericKv3.2channelscontributesignificantlytothedelayedrectifiercurrentofinsulin-secretingcells.InmousecorticalGABAergicfast-spikinginterneurons,however,applicationofShKproducedeffectsconsistentwiththeblockadeofKv3channels(i.e.,anincreaseinactionpotentialhalf-width,adecreaseintheamplitudeoftheactionpotentialafterhyperpolarization,andadecreaseinmaximalfiringfrequencyinresponsetodepolarizingcurrentinjections).Takentogether,theseresultsindicatethatShKisapotentinhibitorofKv3.2channelsandmayserveasausefulpharmacologicalprobeforstudyingthesechannelsinnativepreparations.

YanL.,etal.(2005)Stichodactylahelianthuspeptide,apharmacologicaltoolforstudyingKv3.2channels.MolPharmacol.PMID: 15709110

TargetingeffectormemoryTcellswithaselectivepeptideinhibitorofKv1.3channelsfortherapyofautoimmunediseases

Thevoltage-gatedKv1.3K(+)channelisanoveltargetforimmunomodulationofautoreactiveeffectormemoryT(T(EM))cellsthatplayamajorroleinthepathogenesisofautoimmunediseases.WedescribethecharacterizationofthenovelpeptideShK(L5)thatcontainsl-phosphotyrosinelinkedviaanine-atomhydrophiliclinkertotheNterminusoftheShKpeptidefromtheseaanemoneStichodactylahelianthus.ShK(L5)isahighlyspecificKv1.3blockerthatexhibits100-foldselectivityforKv1.3(K(d)=69pM)overKv1.1andgreaterthan250-foldselectivityoverallotherchannelstested.ShK(L5)suppressestheproliferationofhumanandratT(EM)cellsandinhibitsinterleukin-2productionatpicomolarconcentrations.NaiveandcentralmemoryhumanTcellsareinitially60-foldlesssensitivethanT(EM)cellstoShK(L5)andthenbecomeresistanttothepeptideduringactivationbyup-regulatingthecalcium-activatedK(Ca)3.1channel.ShK(L5)doesnotexhibitinvitrocytotoxicityonmammaliancelllinesandisnegativeintheAmestest.Itisstableinplasmaandwhenadministeredoncedailybysubcutaneousinjection(10mug/kg)attains“steadystate”bloodlevelsofapproximately300pM.ThisregimendoesnotcausecardiactoxicityassessedbycontinuousEKGmonitoringanddoesnotalterclinicalchemistryandhematologicalparametersafter2-weektherapy.ShK(L5)preventsandtreatsexperimentalautoimmuneencephalomyelitisandsuppressesdelayedtypehypersensitivityinrats.ShK(L5)mightproveusefulfortherapyofautoimmunedisorders.

BeetonC.,etal.(2005)TargetingeffectormemoryTcellswithaselectivepeptideinhibitorofKv1.3channelsfortherapyofautoimmunediseases.MolPharmacol. PMID: 15665253

PotassiumchannelblockadebytheseaanemonetoxinShKforthetreatmentofmultiplesclerosisandotherautoimmunediseases

Expressionofthetwolymphocytepotassiumchannels,thevoltage-gatedchannelKv1.3andthecalciumactivatedchannelIKCa1,changesduringdifferentiationofhumanTcells.WhileIKCa1isthefunctionallydominantchannelinnaiveand“early”memoryTcells,Kv1.3iscrucialfortheactivationofterminallydifferentiatedeffectormemory(TEM)Tcells.BecauseoftheinvolvementofTEMcellsinautoimmuneprocesses,Kv1.3isregardedasapromisingtargetforthetreatmentofT-cellmediatedautoimmunediseasessuchasmultiplesclerosisandthepreventionofchronictransplantrejection.ShK,a35-residuepolypeptidetoxinfromtheseaanemone,Stichodactylahelianthus,blocksKv1.3atlowpicomolarconcentrations.ShKadoptsacentralhelix-kink-helixfold,andalanine-scanningandothermutagenesisstudieshavedefineditschannel-bindingsurface.ModelshavebeendevelopedofhowthistoxineffectsK+-channelblockadeandhowitsdockingconfigurationmightdifferinShK-Dap22,whichcontainsasinglesidechainsubstitutionthatconfersspecificityforKv1.3blockade.ShK,ShK-Dap22andtheKv1.3blockingscorpiontoxinkaliotoxinhavebeenshowntopreventandtreatexperimentalautoimmuneencephalomyelitisinrats,amodelformultiplesclerosis.AfluoresceinatedanalogofShK,ShK-F6CA,hasbeendeveloped,whichallowsthedetectionofactivatedTEMcellsinhumanandanimalbloodsamplesbyflowcytometryandthevisualizationofKv1.3channeldistributioninlivingcells.ShKanditsanalogsarecurrentlyundergoingfurtherevaluationasleadsinthedevelopmentofnewbiopharmaceuticalsforthetreatmentofmultiplesclerosisandotherT-cellmediatedautoimmunedisorders.

NortonRS.,etal.(2004)PotassiumchannelblockadebytheseaanemonetoxinShKforthetreatmentofmultiplesclerosisandotherautoimmunediseases.CurrMedChem.PMID: 15578998

SolutionstructureofShKtoxin,anovelpotassiumchannelinhibitorfromaseaanemone
Tudor,J.E.,etal.(1996)SolutionstructureofShKtoxin,anovelpotassiumchannelinhibitorfromaseaanemone,NatStructBiol.PMID: 8599755
AnessentialbindingsurfaceforShKtoxininteractionwithratbrainpotassiumchannels.

An“Alascan”analysisofShKtoxin,a35-residuebasicpeptidepossessingthreedisulfidebonds,identifiessevensidechainswhichinfluencebindingtobraindelayedrectifierpotassiumchannels.Additionalanalogsweresynthesizedandtestedtofurtherdeciphertherolesoftheseresidues,particularlyTyr23.Theinhibitoryeffectsoftheseanalogson125I-labeleddendrotoxinbindingtoratbrainmembranesshowedthatreplacementofTyr23withAladrasticallyloweredtheaffinityofthetoxinfortheKv1.2channels.AlasubstitutionofPhe27reducedpotencymorethan15-fold.MonosubstitutedAlaanalogsforIle7,Ser20,orLys30eachdisplayed5-foldreductionsinpotency.Thus,aromaticityatposition23isimportantforeffectivedelayedrectifierbrainKchannelbinding.Incontrast,thearomaticresidueatposition27wasnotcritical,sincecyclohexylalaninesubstitutionincreasedaffinity.ThesolutionstructureofShKtoxinclustersIle7,Arg11,Ser20,Lys22,Tyr23,andPhe27incloseproximity,formingthepotassiumchannelbindingsurfaceofthetoxin.WeproposeanessentialbindingsurfaceonthetoxininwhichLys22andTyr23aremajorcontributors,throughionicandaromatic(hydrophobic)interactions,withthepotassiumchannel.

PenningtonMW.,etal.(1996)AnessentialbindingsurfaceforShKtoxininteractionwithratbrainpotassiumchannels.Biochemistry. PMID: 8987971

CharacterizationofapotassiumchanneltoxinfromtheCaribbeanSeaanemoneStichodactylahelianthus

Apeptidetoxin,ShK,thatblocksvoltage-dependentpotassiumchannelswasisolatedfromthewholebodyextractoftheCaribbeanseaanemoneStichodactylahelianthus.ItcompeteswithdendrotoxinIandalpha-dendrotoxinforbindingtosynaptosomalmembranesofratbrain,facilitiesacetylcholinereleaseatanavianneuromuscularjunctionandsuppressesK+currentsinratdorsalrootganglionneuronesinculture.ItsaminoacidsequenceisR1SCIDTIPKS10RCTAFQCKHS20MKYRLSFCRK30TCGTC35.ThereisnohomologywithotherK+channel-blockingpeptides,exceptforBgKfromtheseaanemoneBunodosomagranulifera.ShKandBgKappeartobeinadifferentstructuralclassfromothertoxinsaffectingK+channels.

Castaneda,O.,etal.(1995)CharacterizationofapotassiumchanneltoxinfromtheCaribbeanSeaanemoneStichodactylahelianthus,Toxicon. PMID: 7660365

Smartox生物素ProTx-I电压门控钠通道和T型Cav的阻断剂毒素I(ProTx-1;β-theraphotoxin-Tp1a) 是一种毒素,最初是从Prixopelma pruriens(秘鲁绿色天鹅绒狼蛛)的毒液中分离出来的。此毒素可逆地抑制抗河豚毒素(TTX)的通道 Na v 1.8(IC 50  = 27 nM)和 Na v 1.2,Na v 1.5和Na v 1.7  ,IC 50 值为50至100 nM。此外,  ProTx-I 改变了T型Ca v 3.1通道的电压依赖性活动  (IC 50 = 50 nM),而不会影响灭活的电压依赖性。生物素ProTx-I是ProTx-I的生物素标签版本。
  • 资质认证

    获得国家资质,权威认证!

  • 全国联保

    全国联保,官方无忧售后

  • 正规发票

    正规发票,放心购买

  • 签订合同

    签订合同,保障您的权益

/**/