2024
Antagonistic nanobodies implicate mechanism of GSDMD pore formation and potential therapeutic application
Schiffelers L, Tesfamariam Y, Jenster L, Diehl S, Binder S, Normann S, Mayr J, Pritzl S, Hagelauer E, Kopp A, Alon A, Geyer M, Ploegh H, Schmidt F. Antagonistic nanobodies implicate mechanism of GSDMD pore formation and potential therapeutic application. Nature Communications 2024, 15: 8266. PMID: 39327452, PMCID: PMC11427689, DOI: 10.1038/s41467-024-52110-1.Peer-Reviewed Original ResearchMeSH KeywordsAnimalsApoptosisCaspase 1Caspase 3Cell MembraneGasderminsHEK293 CellsHumansInflammasomesIntracellular Signaling Peptides and ProteinsPhosphate-Binding ProteinsProtein MultimerizationPyroptosisSingle-Domain AntibodiesConceptsMembrane insertionGasdermin DN-terminal domainCleavage of gasdermin DPore formationPro-inflammatory caspasesPyroptosis to apoptosisActivated caspase-3Caspase-1 activationTarget membraneCaspase-3Assembled poresPlasma membraneCytosolic expressionLiving cellsConformational changesEnhanced caspase-1 activityOligomerizationPotential therapeutic applicationsInflammasome activationNanobodiesPyroptosisStudy pore formationMembraneTherapeutic applications
2023
Unannotated microprotein EMBOW regulates the interactome and chromatin and mitotic functions of WDR5
Chen Y, Su H, Zhao J, Na Z, Jiang K, Bacchiocchi A, Loh K, Halaban R, Wang Z, Cao X, Slavoff S. Unannotated microprotein EMBOW regulates the interactome and chromatin and mitotic functions of WDR5. Cell Reports 2023, 42: 113145. PMID: 37725512, PMCID: PMC10629662, DOI: 10.1016/j.celrep.2023.113145.Peer-Reviewed Original ResearchMeSH KeywordsCell ProliferationChromatinHumansIntracellular Signaling Peptides and ProteinsKinesinsSpindle ApparatusConceptsG2/M phaseWD40-repeat protein WDR5Mitotic spindle lengthMultiple interaction partnersM phaseOff-target genesLate G1 phaseWDR5 interactionMitotic functionsH3K4me3 levelsWDR5Interaction partnersMultiple proteinsExpression maximaCell cycleSpindle lengthG1 phaseGenesCell proliferationOff-target bindingBindingInteractomeChromatinTranscriptionKIF2A.Pyroptosis in cardiovascular diseases: Pumping gasdermin on the fire
Yarovinsky T, Su M, Chen C, Xiang Y, Tang W, Hwa J. Pyroptosis in cardiovascular diseases: Pumping gasdermin on the fire. Seminars In Immunology 2023, 69: 101809. PMID: 37478801, PMCID: PMC10528349, DOI: 10.1016/j.smim.2023.101809.Peer-Reviewed Original ResearchMeSH KeywordsAnimalsCardiovascular DiseasesGasderminsHumansInflammasomesInflammationIntracellular Signaling Peptides and ProteinsNeoplasm ProteinsPyroptosisConceptsPost-translational modificationsAcute cardiovascular eventsChronic cardiovascular diseaseCardiovascular diseaseSmall molecule inhibitorsPyroptosis resultsGenetic toolsGasdermin proteinsWhole organismInflammatory caspasesCardiovascular eventsCell deathMolecule inhibitorsCell typesProteolytic cleavageCellular mechanismsActivation of inflammasomesCardiovascular systemKnockout animalsAmplification of inflammationRole of pyroptosisPro-inflammatory processesDifferent cellsNovel therapeutic approachesPyroptosisResponse to "NLRC5 germline variants and their potential role in eliciting an immune response in patients with cancer treated with immune checkpoint inhibitors" by Xiang-Yu Meng
Aizenbud L, Schoenfeld D, Caulfield J, Mann J, Austin M, Perdigoto A, Herold K, Kluger H. Response to "NLRC5 germline variants and their potential role in eliciting an immune response in patients with cancer treated with immune checkpoint inhibitors" by Xiang-Yu Meng. Journal For ImmunoTherapy Of Cancer 2023, 11: e007397. PMID: 37349129, PMCID: PMC10314693, DOI: 10.1136/jitc-2023-007397.Peer-Reviewed Original ResearchMeSH KeywordsGerm CellsGerm-Line MutationHumansImmune Checkpoint InhibitorsImmunityIntracellular Signaling Peptides and ProteinsNeoplasmsHedgehog-induced ZFYVE21 promotes chronic vascular inflammation by activating NLRP3 inflammasomes in T cells
Jiang B, Wang S, Song G, Jiang Q, Fan M, Fang C, Li X, Soh C, Manes T, Cheru N, Qin L, Ren P, Jortner B, Wang Q, Quaranta E, Yoo P, Geirsson A, Davis R, Tellides G, Pober J, Jane-Wit D. Hedgehog-induced ZFYVE21 promotes chronic vascular inflammation by activating NLRP3 inflammasomes in T cells. Science Signaling 2023, 16: eabo3406. PMID: 36943921, PMCID: PMC10061549, DOI: 10.1126/scisignal.abo3406.Peer-Reviewed Original ResearchMeSH KeywordsAnimalsEndothelial CellsHedgehog ProteinsHumansInflammasomesInflammationIntracellular Signaling Peptides and ProteinsMembrane ProteinsMiceMice, Inbred C57BLNLR Family, Pyrin Domain-Containing 3 ProteinT-LymphocytesConceptsIschemia-reperfusion injuryChronic vascular inflammationT cellsNLRP3 inflammasomeVascular inflammationChronic inflammationEndothelial cellsIFN-γ responsesControl T cellsNLRP3 inflammasome activityT memory cellsAllograft vasculopathyVascular sequelaeHuman endothelial cellsCoronary arteryEffector responsesCell-autonomous roleInflammasome activityMouse modelInflammationPatient samplesVigorous recruitmentInflammasomePrimary human cellsImmune signalingGermline genetic variants are associated with development of insulin-dependent diabetes in cancer patients treated with immune checkpoint inhibitors
Caulfield J, Aizenbud L, Perdigoto A, Meffre E, Jilaveanu L, Michalek D, Rich S, Aizenbud Y, Adeniran A, Herold K, Austin M, Kluger H. Germline genetic variants are associated with development of insulin-dependent diabetes in cancer patients treated with immune checkpoint inhibitors. Journal For ImmunoTherapy Of Cancer 2023, 11: e006570. PMID: 36898736, PMCID: PMC10008335, DOI: 10.1136/jitc-2022-006570.Peer-Reviewed Original ResearchMeSH KeywordsDiabetes Mellitus, Type 1Germ CellsGerm-Line MutationHumansImmune Checkpoint InhibitorsInsulinsIntracellular Signaling Peptides and ProteinsNeoplasmsConceptsImmune-related adverse eventsInsulin-dependent diabetesImmune checkpoint inhibitorsType 1 diabetesCheckpoint inhibitorsControl patientsSevere immune-related adverse eventsImmunotherapy-treated patientsCheckpoint inhibitor therapyIslet cell destructionPotential predictive biomarkersIslet cell functionWhole-exome sequencingICI exposureAdverse eventsGermline genetic variantsInhibitor therapyPatient selectionTreatment regimensCancer patientsPredictive biomarkersGeneral populationPatientsDiabetesSame drug
2022
Targeting ULK1 Decreases IFNγ-Mediated Resistance to Immune Checkpoint Inhibitors.
Fenton S, Zannikou M, Ilut L, Fischietti M, Ji C, Oku C, Horvath C, Le Poole I, Bosenberg M, Bartom E, Kocherginsky M, Platanias L, Saleiro D. Targeting ULK1 Decreases IFNγ-Mediated Resistance to Immune Checkpoint Inhibitors. Molecular Cancer Research 2022, 21: 332-344. PMID: 36573964, PMCID: PMC10073316, DOI: 10.1158/1541-7786.mcr-22-0684.Peer-Reviewed Original ResearchMeSH KeywordsAutophagy-Related Protein-1 HomologHumansImmune Checkpoint InhibitorsImmune ToleranceImmunosuppression TherapyInterferon-gammaIntracellular Signaling Peptides and ProteinsMelanomaConceptsImmune checkpoint inhibitorsImmunosuppressive genesCheckpoint inhibitorsImmunostimulatory genesAnti-programmed cell death protein 1 therapyPharmacologic inhibitionIFNγ-induced expressionMelanoma cellsMajority of patientsTreatment of patientsTreatment of melanomaMelanoma tumor growthDrug target inhibitionICI therapyDurable responsesPatient survivalMetastatic melanomaPatient outcomesPoor survivalResponse rateTumor growthIFNγOverexpression of ULK1Context-dependent mannerMelanomaBiallelic frameshift variants in PHLDB1 cause mild-type osteogenesis imperfecta with regressive spondylometaphyseal changes
Tuysuz B, Alkaya D, Geyik F, Alaylıoğlu M, Kasap B, Kurugoğlu S, Akman Y, Vural M, Bilguvar K. Biallelic frameshift variants in PHLDB1 cause mild-type osteogenesis imperfecta with regressive spondylometaphyseal changes. Journal Of Medical Genetics 2022, 60: 819-826. PMID: 36543534, DOI: 10.1136/jmg-2022-108763.Peer-Reviewed Original ResearchMeSH KeywordsChild, PreschoolCollagen Type IFractures, BoneFrameshift MutationHeterozygoteHumansIntracellular Signaling Peptides and ProteinsMutationNerve Tissue ProteinsOsteogenesis ImperfectaPhenotypeConceptsOsteogenesis imperfectaWestern blot analysisPathogenic variantsFrameshift variantSkin fibroblast samplesExpression levelsInsulin-dependent Akt phosphorylationBlot analysisAutosomal recessive osteogenesis imperfectaWhole-exome sequencingMRNA expression levelsType 1 collagenBisphosphonate treatmentRecurrent fracturesClinical evaluationRecessive osteogenesis imperfectaCommon findingReal-time PCRMRNA expressionVertebral changesHeterogeneous groupAkt phosphorylationLong bonesBloodSkin fibroblastsUbiquitin-like processing of TUG proteins as a mechanism to regulate glucose uptake and energy metabolism in fat and muscle
Bogan JS. Ubiquitin-like processing of TUG proteins as a mechanism to regulate glucose uptake and energy metabolism in fat and muscle. Frontiers In Endocrinology 2022, 13: 1019405. PMID: 36246906, PMCID: PMC9556833, DOI: 10.3389/fendo.2022.1019405.Peer-Reviewed Original ResearchMeSH KeywordsCarrier ProteinsEnergy MetabolismFatty AcidsGlucoseHumansInsulinIntracellular Signaling Peptides and ProteinsKinesinsMusclesPeptide HydrolasesPPAR gammaProtein TransportUbiquitinConceptsGolgi matrixTUG ProteinVesicle cargoC-terminal productInsulin stimulationN-degron pathwayGLUT4 storage vesiclesCell surfaceUbiquitin-like proteinGLUT4 glucose transportersGlucose uptakeAspects of physiologyN-terminal cleavage productMuscle cellsP97 ATPaseCleavage productsC-terminusFatty acid oxidationGene expressionSingle proteinN-terminusMatrix proteinsEndoproteolytic cleavageCell typesGlucose transporterHuman WDR5 promotes breast cancer growth and metastasis via KMT2-independent translation regulation
Cai WL, Chen JF, Chen H, Wingrove E, Kurley SJ, Chan LH, Zhang M, Arnal-Estape A, Zhao M, Balabaki A, Li W, Yu X, Krop ED, Dou Y, Liu Y, Jin J, Westbrook TF, Nguyen DX, Yan Q. Human WDR5 promotes breast cancer growth and metastasis via KMT2-independent translation regulation. ELife 2022, 11: e78163. PMID: 36043466, PMCID: PMC9584608, DOI: 10.7554/elife.78163.Peer-Reviewed Original ResearchMeSH KeywordsBreast NeoplasmsCell Line, TumorCell ProliferationFemaleHistone-Lysine N-MethyltransferaseHumansIntracellular Signaling Peptides and ProteinsConceptsBreast cancer cellsMetastatic breast cancerBreast cancerRibosomal gene expressionCancer cellsKnockdown of WDR5Vivo genetic screenReversible epigenetic mechanismsGenetic screenTranslation regulationTriple-negative breast cancerEpigenetic regulatorsEpigenetic mechanismsBreast cancer growthCancer-related deathTranslation efficiencyWDR5Novel therapeutic strategiesTranslation rateGene expressionCell growthAdvanced diseaseEffective therapyMetastatic capabilityPotent suppressionLACC1 bridges NOS2 and polyamine metabolism in inflammatory macrophages
Wei Z, Oh J, Flavell RA, Crawford JM. LACC1 bridges NOS2 and polyamine metabolism in inflammatory macrophages. Nature 2022, 609: 348-353. PMID: 35978195, PMCID: PMC9813773, DOI: 10.1038/s41586-022-05111-3.Peer-Reviewed Original ResearchMeSH KeywordsAnimalsArthritisCitrullineCyanatesHumansInflammationInflammatory Bowel DiseasesIntracellular Signaling Peptides and ProteinsMacrophagesMiceNitric Oxide Synthase Type IIOrnithineOrnithine DecarboxylasePolyaminesSalmonella typhimuriumConceptsInflammatory bowel diseaseWild-type activityCentral regulatory roleMammalian immune systemBone marrow-derived macrophagesInflammatory macrophagesBiochemical functionsBowel diseaseSignaling outcomesMarrow-derived macrophagesPattern recognition receptorsInflammatory diseasesBiochemical roleRegulatory roleMechanistic connectionUnidentified pathwaySalmonella enterica TyphimuriumNitric oxide synthaseRecognition receptorsHost damageHuman inflammatory diseasesMultiple inflammatory diseasesEnterica TyphimuriumOrnithine decarboxylaseLACC1A multiancestry genome-wide association study of unexplained chronic ALT elevation as a proxy for nonalcoholic fatty liver disease with histological and radiological validation
Vujkovic M, Ramdas S, Lorenz KM, Guo X, Darlay R, Cordell HJ, He J, Gindin Y, Chung C, Myers RP, Schneider CV, Park J, Lee KM, Serper M, Carr RM, Kaplan DE, Haas ME, MacLean MT, Witschey WR, Zhu X, Tcheandjieu C, Kember RL, Kranzler HR, Verma A, Giri A, Klarin DM, Sun YV, Huang J, Huffman JE, Creasy KT, Hand NJ, Liu CT, Long MT, Yao J, Budoff M, Tan J, Li X, Lin HJ, Chen YI, Taylor KD, Chang RK, Krauss RM, Vilarinho S, Brancale J, Nielsen JB, Locke AE, Jones MB, Verweij N, Baras A, Reddy KR, Neuschwander-Tetri BA, Schwimmer JB, Sanyal AJ, Chalasani N, Ryan KA, Mitchell BD, Gill D, Wells AD, Manduchi E, Saiman Y, Mahmud N, Miller DR, Reaven PD, Phillips LS, Muralidhar S, DuVall SL, Lee JS, Assimes TL, Pyarajan S, Cho K, Edwards TL, Damrauer SM, Wilson PW, Gaziano JM, O’Donnell C, Khera AV, Grant SFA, Brown CD, Tsao PS, Saleheen D, Lotta LA, Bastarache L, Anstee QM, Daly AK, Meigs JB, Rotter JI, Lynch JA, Rader D, Voight B, Chang K. A multiancestry genome-wide association study of unexplained chronic ALT elevation as a proxy for nonalcoholic fatty liver disease with histological and radiological validation. Nature Genetics 2022, 54: 761-771. PMID: 35654975, PMCID: PMC10024253, DOI: 10.1038/s41588-022-01078-z.Peer-Reviewed Original ResearchMeSH KeywordsAlanine TransaminaseAlpha-Ketoglutarate-Dependent Dioxygenase FTOGenome-Wide Association StudyHumansIntracellular Signaling Peptides and ProteinsLipaseMembrane ProteinsNon-alcoholic Fatty Liver DiseasePolymorphism, Single NucleotideProtein Serine-Threonine KinasesConceptsGenome-wide association studiesSingle nucleotide polymorphismsAssociation studiesMillion Veteran ProgramGenome-wide significanceGenetic architectureAdditional lociPleiotropy analysisInflammatory traitsLociNew insightsVeteran ProgramChronic ALT elevationTraitsExternal replicationRadiological validationReplicationDysregulation of the Scribble/YAP/β‐catenin axis sustains the fibroinflammatory response in a PKHD1−/− mouse model of congenital hepatic fibrosis
Fabris L, Milani C, Fiorotto R, Mariotti V, Kaffe E, Seller B, Sonzogni A, Strazzabosco M, Cadamuro M. Dysregulation of the Scribble/YAP/β‐catenin axis sustains the fibroinflammatory response in a PKHD1−/− mouse model of congenital hepatic fibrosis. The FASEB Journal 2022, 36: e22364. PMID: 35593740, PMCID: PMC9150862, DOI: 10.1096/fj.202101924r.Peer-Reviewed Original ResearchMeSH KeywordsAnimalsbeta CateninCystsDisease Models, AnimalGenetic Diseases, InbornIntracellular Signaling Peptides and ProteinsLiver CirrhosisMiceReceptors, Cell SurfaceRNA, Small InterferingTranscription FactorsConceptsYes-associated proteinPlanar cell polarityΒ-cateninΒ-catenin axisYAP nuclear importRole of ScribbleNuclear translocationYAP/TAZΒ-catenin signalingCell polarityNuclear importCyst cellsNuclear expressionScribble expressionΒ-catenin nuclear expressionConditional deletionGenetic defectsTissue growth factor expressionIntegrin β6Connective tissue growth factor expressionCyst growthExpressionCystic cholangiocytesMRNA levelsScribblesSHIP164 is a chorein motif lipid transfer protein that controls endosome–Golgi membrane traffic
Hanna MG, Suen PH, Wu Y, Reinisch KM, De Camilli P. SHIP164 is a chorein motif lipid transfer protein that controls endosome–Golgi membrane traffic. Journal Of Cell Biology 2022, 221: e202111018. PMID: 35499567, PMCID: PMC9067936, DOI: 10.1083/jcb.202111018.Peer-Reviewed Original ResearchMeSH KeywordsCarrier ProteinsEndosomesGolgi ApparatusIntracellular MembranesIntracellular Signaling Peptides and ProteinsLipidsConceptsMannose-6-phosphate receptorCation-independent mannose-6-phosphate receptorMembrane contact sitesEarly endocytic pathwayGolgi membrane trafficLipid transfer proteinLipid transfer propertiesProtein-lipid ratioMembrane cargoMembrane trafficEndocytic membranesVesicular trafficRetrograde trafficEndocytic pathwayFunctional characterizationContact sitesMembranous organellesGolgi complexVesicle clustersCellular membranesTransfer proteinProteinLipid compositionOrganellesMembrane
2021
From Molecule to Behavior: Hypocretin/orexin Revisited From a Sex-dependent Perspective
Gao XB, Horvath TL. From Molecule to Behavior: Hypocretin/orexin Revisited From a Sex-dependent Perspective. Endocrine Reviews 2021, 43: 743-760. PMID: 34792130, PMCID: PMC9277634, DOI: 10.1210/endrev/bnab042.Peer-Reviewed Original ResearchTAZ inhibits glucocorticoid receptor and coordinates hepatic glucose homeostasis in normal physiological states
Xu S, Liu Y, Hu R, Wang M, Stöhr O, Xiong Y, Chen L, Kang H, Zheng L, Cai S, He L, Wang C, Copps K, White M, Miao J. TAZ inhibits glucocorticoid receptor and coordinates hepatic glucose homeostasis in normal physiological states. ELife 2021, 10: e57462. PMID: 34622775, PMCID: PMC8555985, DOI: 10.7554/elife.57462.Peer-Reviewed Original ResearchMeSH KeywordsAdaptor Proteins, Signal TransducingAnimalsBlood GlucoseGluconeogenesisHomeostasisIntracellular Signaling Peptides and ProteinsLiverMice, KnockoutReceptors, GlucocorticoidConceptsGluconeogenic gene promotersBinding of GRGene promoterGlucocorticoid receptorGlucose homeostasisLigand-binding domainGlucose productionOverexpression of TAZHepatic glucose homeostasisWW domainsBlood glucose concentrationPhysiological fastingGluconeogenic genesGR response elementResponse elementNovel roleTAZNormal physiological stateGR transactivationPhysiological statePromoterMouse liverPericentral hepatocytesPathological statesGlucose concentrationYAP and TAZ are transcriptional co-activators of AP-1 proteins and STAT3 during breast cellular transformation
He L, Pratt H, Gao M, Wei F, Weng Z, Struhl K. YAP and TAZ are transcriptional co-activators of AP-1 proteins and STAT3 during breast cellular transformation. ELife 2021, 10: e67312. PMID: 34463254, PMCID: PMC8463077, DOI: 10.7554/elife.67312.Peer-Reviewed Original ResearchMeSH KeywordsAdaptor Proteins, Signal TransducingCell Line, TumorCell Transformation, NeoplasticDatabases, GeneticFemaleGene Expression Regulation, NeoplasticHumansIntracellular Signaling Peptides and ProteinsProtein BindingProtein Interaction Domains and MotifsSignal TransductionSTAT3 Transcription FactorTranscription Factor AP-1Transcription FactorsTranscriptional ActivationTranscriptional Coactivator with PDZ-Binding Motif ProteinsTriple Negative Breast NeoplasmsYAP-Signaling ProteinsConceptsTranscriptional co-activatorAP-1 proteinsAP-1Cellular transformationTarget siteNon-overlapping genesStimulated transcriptional activityAssociated with poor survival of breast cancer patientsPoor survival of breast cancer patientsSTAT3 motifTEAD proteinsSequence motifsGene classesWW domainEpigenetic switchTaz paralogTAZ-specificTranscriptional activityTranscription factorsTEADYAP/TAZSTAT3MotifTAZJunBSmart-RRBS for single-cell methylome and transcriptome analysis
Gu H, Raman AT, Wang X, Gaiti F, Chaligne R, Mohammad AW, Arczewska A, Smith ZD, Landau DA, Aryee MJ, Meissner A, Gnirke A. Smart-RRBS for single-cell methylome and transcriptome analysis. Nature Protocols 2021, 16: 4004-4030. PMID: 34244697, PMCID: PMC8672372, DOI: 10.1038/s41596-021-00571-9.Peer-Reviewed Original ResearchMeSH KeywordsAmino Acid SequenceAnti-Bacterial AgentsDNADNA (Cytosine-5-)-MethyltransferasesDoxycyclineEpigenomeGene Expression RegulationHumansIntracellular Signaling Peptides and ProteinsRNA, MessengerSingle-Cell AnalysisTranscriptomeConceptsSingle cellsProtein-coding genesSingle-cell methylomesSame single cellMulti-omics approachRare cell populationsSmart-seq2Transcriptional statesDNA methylomeTranscriptome analysisImportant mechanistic insightsEpigenetic modificationsDNA methylationDissected tissue samplesGenomic DNAHundreds of cellsCellular heterogeneityFlow sortingRegulatory consequencesMethylomeEpigenetic promoterMechanistic insightsCell populationsCellsTypical single cellCerebellar Kv3.3 potassium channels activate TANK-binding kinase 1 to regulate trafficking of the cell survival protein Hax-1
Zhang Y, Varela L, Szigeti-Buck K, Williams A, Stoiljkovic M, Šestan-Peša M, Henao-Mejia J, D’Acunzo P, Levy E, Flavell RA, Horvath TL, Kaczmarek LK. Cerebellar Kv3.3 potassium channels activate TANK-binding kinase 1 to regulate trafficking of the cell survival protein Hax-1. Nature Communications 2021, 12: 1731. PMID: 33741962, PMCID: PMC7979925, DOI: 10.1038/s41467-021-22003-8.Peer-Reviewed Original ResearchMeSH KeywordsAnimalsCell SurvivalCerebellumExosomesFemaleInterneuronsIntracellular Signaling Peptides and ProteinsMaleMiceMutationPhenotypeProtein Serine-Threonine KinasesProtein TransportShaw Potassium ChannelsSignal TransductionConceptsTank Binding Kinase 1HAX-1Kv3.3 potassium channelMultivesicular bodiesKinase 1TANK-binding kinase 1Activation of caspasesAnti-apoptotic proteinsPotassium channelsMembrane proteinsBiochemical pathwaysCerebellar neuronsChannels bindCell deathTBK1 activityIon channelsMutant channelsCellular constituentsTraffickingKv3.3 channelsProteinNeuronal survivalMutationsChannel inactivationCaspasesInsulin-stimulated endoproteolytic TUG cleavage links energy expenditure with glucose uptake
Habtemichael EN, Li DT, Camporez JP, Westergaard XO, Sales CI, Liu X, López-Giráldez F, DeVries SG, Li H, Ruiz DM, Wang KY, Sayal BS, González Zapata S, Dann P, Brown SN, Hirabara S, Vatner DF, Goedeke L, Philbrick W, Shulman GI, Bogan JS. Insulin-stimulated endoproteolytic TUG cleavage links energy expenditure with glucose uptake. Nature Metabolism 2021, 3: 378-393. PMID: 33686286, PMCID: PMC7990718, DOI: 10.1038/s42255-021-00359-x.Peer-Reviewed Original ResearchMeSH Keywords3T3-L1 CellsAminoacyltransferasesAnimalsEnergy MetabolismGlucoseInsulinIntracellular Signaling Peptides and ProteinsMiceMice, KnockoutOxidation-ReductionPeroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alphaPPAR gammaProteolysisThermogenesisConceptsTUG cleavageGlucose uptakeProtein degradation pathwaysGLUT4 glucose transportersCoactivator PGC-1αC-terminal cleavage productInsulin-stimulated glucose uptakeAte1 arginyltransferaseGene expressionPhysiological relevanceWhole-body energy expenditureGlucose transporterPeroxisome proliferator-activated receptorCell surfacePGC-1αProtein 1Proliferator-activated receptorDegradation pathwayEffect of insulinCleavage pathwayAdipose cellsCleavage productsPathwayCleavageEnergy expenditure
This site is protected by hCaptcha and its Privacy Policy and Terms of Service apply