2024
Bone-derived extracellular matrix hydrogel from thrombospondin-2 knock-out mice for bone repair
Chen Z, Zhang J, Lee F, Kyriakides T. Bone-derived extracellular matrix hydrogel from thrombospondin-2 knock-out mice for bone repair. Acta Biomaterialia 2024, 186: 85-94. PMID: 39134130, PMCID: PMC11500023, DOI: 10.1016/j.actbio.2024.08.011.Peer-Reviewed Original ResearchECM hydrogelsWild typeBone repairLack of mechanical integrityExtracellular matrixRepair of bone defectsExtracellular matrix hydrogelDecellularized extracellular matrixMurine calvarial defect modelCalvarial defect modelPromote bone repairHuman umbilical vein endothelial cellsMesenchymal stem cellsThrombospondin-2Fabricated hydrogelsBone extracellular matrixNovel hydrogelsMechanical propertiesCollagen fibril assemblyMatrix hydrogelRepair damaged boneDecellularized ECMMechanical integrityHydrogelsInvasion in vitroA genetic association study of circulating coagulation factor VIII and von Willebrand factor levels
de Vries P, Reventun P, Brown M, Heath A, Huffman J, Le N, Bebo A, Brody J, Temprano-Sagrera G, Raffield L, Ozel A, Thibord F, Jain D, Lewis J, Rodriguez B, Pankratz N, Taylor K, Polasek O, Chen M, Yanek L, Carrasquilla G, Marioni R, Kleber M, Trégouët D, Yao J, Li-Gao R, Joshi P, Trompet S, Martinez-Perez A, Ghanbari M, Howard T, Reiner A, Arvanitis M, Ryan K, Bartz T, Rudan I, Faraday N, Linneberg A, Ekunwe L, Davies G, Delgado G, Suchon P, Guo X, Rosendaal F, Klaric L, Noordam R, van Rooij F, Curran J, Wheeler M, Osburn W, O'Connell J, Boerwinkle E, Beswick A, Psaty B, Kolcic I, Souto J, Becker L, Hansen T, Doyle M, Harris S, Moissl A, Deleuze J, Rich S, van Hylckama Vlieg A, Campbell H, Stott D, Soria J, de Maat M, Almasy L, Brody L, Auer P, Mitchell B, Ben-Shlomo Y, Fornage M, Hayward C, Mathias R, Kilpeläinen T, Lange C, Cox S, März W, Morange P, Rotter J, Mook-Kanamori D, Wilson C, van der Harst P, Jukema J, Ikram M, Blangero J, Kooperberg C, Desch K, Johnson A, Sabater-Lleal M, Lowenstein C, Smith A, Morrison A, Abe N, Abecasis G, Aguet F, Albert C, Almasy L, Alonso A, Ament S, Anderson P, Anugu P, Applebaum-Bowden D, Ardlie K, Arking D, Arnett D, Ashley-Koch A, Aslibekyan S, Assimes T, Auer P, Avramopoulos D, Ayas N, Balasubramanian A, Barnard J, Barnes K, Barr R, Barron-Casella E, Barwick L, Beaty T, Beck G, Becker D, Becker L, Beer R, Beitelshees A, Benjamin E, Benos T, Bezerra M, Bielak L, Bis J, Blackwell T, Blangero J, Blue N, Boerwinkle E, Bowden D, Bowler R, Brody J, Broeckel U, Broome J, Brown D, Bunting K, Burchard E, Bustamante C, Buth E, Cade B, Cardwell J, Carey V, Carrier J, Carson A, Carty C, Casaburi R, Romero J, Casella J, Castaldi P, Chaffin M, Chang C, Chang Y, Chasman D, Chavan S, Chen B, Chen W, Chen Y, Cho M, Choi S, Chuang L, Chung M, Chung R, Clish C, Comhair S, Conomos M, Cornell E, Correa A, Crandall C, Crapo J, Cupples L, Curran J, Curtis J, Custer B, Damcott C, Darbar D, David S, Davis C, Daya M, de Andrade M, de las Fuentes L, de Vries P, DeBaun M, Deka R, DeMeo D, Devine S, Dinh H, Doddapaneni H, Duan Q, Dugan-Perez S, Duggirala R, Durda J, Dutcher S, Eaton C, Ekunwe L, Boueiz A, Ellinor P, Emery L, Erzurum S, Farber C, Farek J, Fingerlin T, Flickinger M, Fornage M, Franceschini N, Frazar C, Fu M, Fullerton S, Fulton L, Gabriel S, Gan W, Gao S, Gao Y, Gass M, Geiger H, Gelb B, Geraci M, Germer S, Gerszten R, Ghosh A, Gibbs R, Gignoux C, Gladwin M, Glahn D, Gogarten S, Gong D, Goring H, Graw S, Gray K, Grine D, Gross C, Gu C, Guan Y, Guo X, Gupta N, Haessler J, Hall M, Han Y, Hanly P, Harris D, Hawley N, He J, Heavner B, Heckbert S, Hernandez R, Herrington D, Hersh C, Hidalgo B, Hixson J, Hobbs B, Hokanson J, Hong E, Hoth K, Hsiung C, Hu J, Hung Y, Huston H, Hwu C, Irvin M, Jackson R, Jain D, Jaquish C, Johnsen J, Johnson C, Johnson A, Johnston R, Jones K, Kang H, Kaplan R, Kardia S, Kelly S, Kenny E, Kessler M, Khan A, Khan Z, Kim W, Kimoff J, Kinney G, Konkle B, Kooperberg C, Kramer H, Lange E, Lange L, Lange L, Laurie C, Laurie C, LeBoff M, Lee J, Lee S, Lee W, LeFaive J, Levine D, Levy D, Lewis J, Li X, Li Y, Lin H, Lin H, Lin X, Liu S, Liu Y, Liu Y, Loos R, Lubitz S, Lunetta K, Luo J, Magalang U, Mahaney M, Make B, Manichaikul A, Manning A, Manson J, Martin L, Marton M, Mathai S, Mathias R, May S, McArdle P, McDonald M, McFarland S, McGarvey S, McGoldrick D, McHugh C, McNeil B, Mei H, Meigs J, Menon V, Mestroni L, Metcalf G, Meyers D, Mignot E, Mikulla J, Min N, Minear M, Minster R, Mitchell B, Moll M, Momin Z, Montasser M, Montgomery C, Muzny D, Mychaleckyj J, Nadkarni G, Naik R, Naseri T, Natarajan P, Nekhai S, Nelson S, Neltner B, Nessner C, Nickerson D, Nkechinyere O, North K, O'Connell J, O'Connor T, Ochs-Balcom H, Okwuonu G, Pack A, Paik D, Palmer N, Pankow J, Papanicolaou G, Parker C, Peloso G, Peralta J, Perez M, Perry J, Peters U, Peyser P, Phillips L, Pleiness J, Pollin T, Post W, Becker J, Boorgula M, Preuss M, Psaty B, Qasba P, Qiao D, Qin Z, Rafaels N, Raffield L, Rajendran M, Ramachandran V, Rao D, Rasmussen-Torvik L, Ratan A, Redline S, Reed R, Reeves C, Regan E, Reiner A, Reupena M, Rice K, Rich S, Robillard R, Robine N, Roden D, Roselli C, Rotter J, Ruczinski I, Runnels A, Russell P, Ruuska S, Ryan K, Sabino E, Saleheen D, Salimi S, Salvi S, Salzberg S, Sandow K, Sankaran V, Santibanez J, Schwander K, Schwartz D, Sciurba F, Seidman C, Seidman J, Sériès F, Sheehan V, Sherman S, Shetty A, Shetty A, Sheu W, Shoemaker M, Silver B, Silverman E, Skomro R, Smith J, Smith J, Smith N, Smith T, Smith E, Smoller S, Snively B, Snyder M, Sofer T, Sotoodehnia N, Stilp A, Storm G, Streeten E, Su J, Sung Y, Sylvia J, Szpiro A, Taliun D, Tang H, Taub M, Taylor K, Taylor M, Taylor S, Telen M, Thornton T, Threlkeld M, Tinker L, Tirschwell D, Tishkoff S, Tiwari H, Tong C, Tracy R, Tsai M, Vaidya D, Van Den Berg D, VandeHaar P, Vrieze S, Walker T, Wallace R, Walts A, Wang F, Wang H, Wang J, Watson K, Watt J, Weeks D, Weinstock J, Weir B, Weiss S, Weng L, Wessel J, Willer C, Williams K, Williams L, Wilson J, Wilson J, Winterkorn L, Wong Q, Wu J, Xu H, Yanek L, Yang I, Yu K, Zekavat S, Zhang Y, Zhao S, Zhao W, Zhu X, Ziv E, Zody M, Zoellner S, Lindstrom S, Wang L, Smith N, Gordon W, van Hylckama Vlieg A, de Andrade M, Brody J, Pattee J, Haessler J, Brumpton B, Chasman D, Suchon P, Chen M, Turman C, Germain M, Wiggins K, MacDonald J, Braekkan S, Armasu S, Pankratz N, Jackson R, Nielsen J, Giulianini F, Puurunen M, Ibrahim M, Heckbert S, Bammler T, Frazer K, McCauley B, Taylor K, Pankow J, Reiner A, Gabrielsen M, Deleuze J, O'Donnell C, Kim J, McKnight B, Kraft P, Hansen J, Rosendaal F, Heit J, Psaty B, Tang W, Kooperberg C, Hveem K, Ridker P, Morange P, Johnson A, Kabrhel C, AlexandreTrégouët D, Smith N. A genetic association study of circulating coagulation factor VIII and von Willebrand factor levels. Blood 2024, 143: 1845-1855. PMID: 38320121, DOI: 10.1182/blood.2023021452.Peer-Reviewed Original ResearchMendelian randomizationGene-based aggregation testingImputation of genotypesGene-based analysisMulti-phenotype analysisAssociations of factor VIIIGenetic association studiesHuman umbilical vein endothelial cellsCausal genesTrans-OmicsAssociation studiesB3GNT2Genetic associationVon Willebrand factorProtein von Willebrand factorLociIdentified associationsPDIA3Umbilical vein endothelial cellsIncreased riskMeta-analysisCarrier protein von Willebrand factorVein endothelial cellsPrecision medicineEndothelial cellsBioengineering of vascularized porcine flaps using perfusion-recellularization
Xu M, D’Elia A, Hadzimustafic N, Adil A, Karoubi G, Waddell T, Haykal S. Bioengineering of vascularized porcine flaps using perfusion-recellularization. Scientific Reports 2024, 14: 7590. PMID: 38555385, PMCID: PMC10981729, DOI: 10.1038/s41598-024-58095-7.Peer-Reviewed Original ResearchConceptsLow-concentration sodium dodecyl sulfateAcellular scaffoldsRecellularization techniquesPerfusion-decellularizationMesenchymal stromal cellsSodium dodecyl sulfateCell attachmentRecellularization approachCo-culture of human umbilical vein endothelial cellsRecellularizationHuman umbilical vein endothelial cellsReconstructive surgeryTensor fasciae lataeExtracellular matrixRepair optionsDonor site morbidityImpact patient qualityDodecyl sulfateClinically translatable optionTensor fascia lata flapUmbilical vein endothelial cellsSoft tissue defectsPorcine flaps
2023
TNFα increases the degradation of pyruvate dehydrogenase kinase 4 by the Lon protease to support proinflammatory genes
Boutagy N, Fowler J, Grabinska K, Cardone R, Sun Q, Vazquez K, Whalen M, Zhu X, Chakraborty R, Martin K, Simons M, Romanoski C, Kibbey R, Sessa W. TNFα increases the degradation of pyruvate dehydrogenase kinase 4 by the Lon protease to support proinflammatory genes. Proceedings Of The National Academy Of Sciences Of The United States Of America 2023, 120: e2218150120. PMID: 37695914, PMCID: PMC10515159, DOI: 10.1073/pnas.2218150120.Peer-Reviewed Original ResearchConceptsPyruvate dehydrogenase kinase 4Dehydrogenase kinase 4Lon proteasePyruvate dehydrogenase activityHistone acetylationMitochondrial metabolismKinase 4Specific gene lociPDH fluxEndothelial cellsSiRNA-mediated knockdownAcetyl-CoA generationLysine 27Gene transcriptionTCA fluxRNA sequencingHuman umbilical vein endothelial cellsProtein degradationHistone 3Gene locusUmbilical vein endothelial cellsNF-κB-dependent mechanismTricarboxylic acid cycle fluxVein endothelial cellsActive subunit
2021
Morphological characterization of Etv2 vascular explants using fractal analysis and atomic force microscopy
Adelson R, Palikuqi B, Weiss Z, Checco A, Schreiner R, Rafii S, Rabbany S. Morphological characterization of Etv2 vascular explants using fractal analysis and atomic force microscopy. Microvascular Research 2021, 138: 104205. PMID: 34146583, PMCID: PMC8446305, DOI: 10.1016/j.mvr.2021.104205.Peer-Reviewed Original ResearchMeSH KeywordsCell ShapeCells, CulturedCoculture TechniquesColonic NeoplasmsFractalsHuman Umbilical Vein Endothelial CellsHumansImage Processing, Computer-AssistedMicroscopy, Atomic ForceNeovascularization, PathologicNeovascularization, PhysiologicProtocadherinsTissue Culture TechniquesTranscription FactorsVascular Endothelial Growth Factor Receptor-2ConceptsAtomic force microscopyForce microscopyPoor temporal stabilityMechanical integrityFractal dimensionStructural homogeneityMorphological characterizationHuman umbilical vein ECDecreasing fractal dimensionIn vivoMicroscopyVascular regenerationAngiogenic measuresEts variant 2StabilityFractal analysisDensityEndothelial cellsVessel branchesBedVariant 2Organoid tissueHuman umbilical vein endothelial cellsATPIF1 maintains normal mitochondrial structure which is impaired by CCM3 deficiency in endothelial cells
Wang K, Chen H, Zhou Z, Zhang H, Zhou HJ, Min W. ATPIF1 maintains normal mitochondrial structure which is impaired by CCM3 deficiency in endothelial cells. Cell & Bioscience 2021, 11: 11. PMID: 33422124, PMCID: PMC7796565, DOI: 10.1186/s13578-020-00514-z.Peer-Reviewed Original ResearchActivation of mitophagyHuman umbilical vein endothelial cellsNormal mitochondrial structureMorphology of mitochondriaRNA-seq screeningMitochondrial membrane potentialCRISPR-Cas9 SystemCerebral cavernous malformationsEndothelial cellsExpression of KLF4Destruction of mitochondriaUmbilical vein endothelial cellsMitochondrial structureSignaling pathwaysVein endothelial cellsMitochondriaATPIF1MitophagyEndothelial progenitor cellsProgenitor cellsCell proliferationMembrane potentialKLF4PathwayProtein
2020
H19/TET1 axis promotes TGF‐β signaling linked to endothelial‐to‐mesenchymal transition
Cao T, Jiang Y, Li D, Sun X, Zhang Y, Qin L, Tellides G, Taylor HS, Huang Y. H19/TET1 axis promotes TGF‐β signaling linked to endothelial‐to‐mesenchymal transition. The FASEB Journal 2020, 34: 8625-8640. PMID: 32374060, PMCID: PMC7364839, DOI: 10.1096/fj.202000073rrrrr.Peer-Reviewed Original ResearchMeSH KeywordsAnimalsCells, CulturedCoronary VesselsEpithelial-Mesenchymal TransitionHuman Umbilical Vein Endothelial CellsHumansMiceMice, Inbred C57BLMice, KnockoutMixed Function OxygenasesProto-Oncogene ProteinsRNA Processing, Post-TranscriptionalRNA, Long NoncodingSignal TransductionTransforming Growth Factor betaConceptsTGF-β signalingCardiovascular diseaseHuman umbilical vein endothelial cellsEndothelial cellsEndothelial activationMesenchymal transitionMouse pulmonary microvascular endothelial cellsPulmonary microvascular endothelial cellsHuman atherosclerotic coronary arteriesAtherosclerotic coronary arteriesMicrovascular endothelial cellsPrimary human umbilical vein endothelial cellsUmbilical vein endothelial cellsAortic endothelial cellsEndothelial dysfunctionVein endothelial cellsCoronary arteryRisk factorsHyperglycemic conditionsH19 expressionAberrant expressionEndMTH19 lncRNATET1 expressionMolecular underpinningsEarly Vascular Cells Improve Microvascularization Within 3D Cardiac Spheroids
Pitaktong I, Lui C, Lowenthal J, Mattson G, Jung WH, Bai Y, Yeung E, Ong CS, Chen Y, Gerecht S, Hibino N. Early Vascular Cells Improve Microvascularization Within 3D Cardiac Spheroids. Tissue Engineering Part C Methods 2020, 26: 80-90. PMID: 31830863, PMCID: PMC8884142, DOI: 10.1089/ten.tec.2019.0228.Peer-Reviewed Original ResearchConceptsHuman umbilical vein endothelial cellsVascular cellsVentricular cardiac fibroblastsMicrovascular formationUmbilical vein endothelial cellsVein endothelial cellsCardiac functionGroup 2Group 1Group 3Cell ratioCardiac fibroblastsCardiac spheroidsEndothelial cellsCardiomyocytesCardiac tissueEarly vascular cellsStem cellsContraction ratePluripotent stem cellsFibroblastsVascular networkSpheroid formationVascularizationTissue environmentHerpesvirus-infected Hofbauer cells activate endothelial cells through an IL-1β-dependent mechanism
Hendrix P, Tang Z, Silasi M, Racicot KE, Mor G, Abrahams VM, Guller S. Herpesvirus-infected Hofbauer cells activate endothelial cells through an IL-1β-dependent mechanism. Placenta 2020, 91: 59-65. PMID: 32174308, PMCID: PMC7078070, DOI: 10.1016/j.placenta.2020.01.010.Peer-Reviewed Original ResearchConceptsHuman umbilical vein endothelial cellsHofbauer cellsIL-1βAdhesion molecule-1Placental viral infectionsIL-8Viral infectionFetal inflammationEndothelial cellsE-selectinMolecule-1IL-1 receptor antagonistVascular adhesion molecule-1Intercellular adhesion molecule-1Adverse pregnancy outcomesMHV-68 infectionIL-1β secretionICAM-1 mRNAUmbilical vein endothelial cellsUmbilical endothelial cellsPregnancy outcomesVein endothelial cellsIL-1raPlacental macrophagesHUVEC expression
2019
Sustained perfusion of revascularized bioengineered livers heterotopically transplanted into immunosuppressed pigs
Shaheen M, Joo D, Ross J, Anderson B, Chen H, Huebert R, Li Y, Amiot B, Young A, Zlochiver V, Nelson E, Mounajjed T, Dietz A, Michalak G, Steiner B, Davidow D, Paradise C, van Wijnen A, Shah V, Liu M, Nyberg S. Sustained perfusion of revascularized bioengineered livers heterotopically transplanted into immunosuppressed pigs. Nature Biomedical Engineering 2019, 4: 437-445. PMID: 31611679, PMCID: PMC7153989, DOI: 10.1038/s41551-019-0460-x.Peer-Reviewed Original ResearchConceptsHuman endothelial cellsEndothelial cellsSustained perfusionImmune responseContinuous perfusionNormal liver tissueHuman umbilical vein endothelial cellsUmbilical vein endothelial cellsImmunosuppression protocolsAnticoagulation therapyVein endothelial cellsImmunosuppressed pigsEndothelial markersLiver sinusoidsPerfusionLiver tissueHuman liverLiverBlood perfusionHeterotopic implantationMain predictorsLiver scaffoldsPigsPorcine liverDaysEpac agonist improves barrier function in iPSC-derived endothelial colony forming cells for whole organ tissue engineering
Yuan Y, Engler AJ, Raredon MS, Le A, Baevova P, Yoder MC, Niklason LE. Epac agonist improves barrier function in iPSC-derived endothelial colony forming cells for whole organ tissue engineering. Biomaterials 2019, 200: 25-34. PMID: 30754017, DOI: 10.1016/j.biomaterials.2019.02.005.Peer-Reviewed Original Research
2018
Bradykinin B2 Receptor Contributes to Inflammatory Responses in Human Endothelial Cells by the Transactivation of the Fibroblast Growth Factor Receptor FGFR-1
Terzuoli E, Corti F, Nannelli G, Giachetti A, Donnini S, Ziche M. Bradykinin B2 Receptor Contributes to Inflammatory Responses in Human Endothelial Cells by the Transactivation of the Fibroblast Growth Factor Receptor FGFR-1. International Journal Of Molecular Sciences 2018, 19: 2638. PMID: 30200598, PMCID: PMC6163484, DOI: 10.3390/ijms19092638.Peer-Reviewed Original ResearchConceptsFibroblast growth factor receptor 1Human umbilical vein endothelial cellsCell permeabilityEndothelial cellsFGFR-1 phosphorylationEndothelial cell permeabilityFGF-2 signalingFGF-2 pathwaysFGF-2/FGFRFGFR-1 inhibitorsC-SrcGrowth factor receptor 1Umbilical vein endothelial cellsDownstream signalingHuman endothelial cellsGrowth factor 2Vein endothelial cellsFactor receptor 1Cell migrationFGF-2 upregulationAmplification loopSignalingFactor 2Angiogenic disordersBradykinin B2 receptorRegulation of human T cell responses by dNP2-ctCTLA-4 inhibits human skin and microvessel graft rejection
Lim S, Kirkiles-Smith NC, Pober JS, Bothwell ALM, Choi JM. Regulation of human T cell responses by dNP2-ctCTLA-4 inhibits human skin and microvessel graft rejection. Biomaterials 2018, 183: 128-138. PMID: 30165256, PMCID: PMC6141312, DOI: 10.1016/j.biomaterials.2018.08.049.Peer-Reviewed Original ResearchMeSH KeywordsAnimalsCD4-Positive T-LymphocytesCD8-Positive T-LymphocytesCell ProliferationCell-Penetrating PeptidesCTLA-4 AntigenCytokinesEndothelial CellsFemaleGraft RejectionHuman Umbilical Vein Endothelial CellsHumansLymphocyte ActivationMice, Inbred BALB CMice, KnockoutMice, SCIDMicrovesselsReceptors, ChemokineSkinSkin TransplantationT-LymphocytesConceptsT cell responsesHuman T cell responsesT cell infiltrationHuman T cellsT cellsCell responsesGraft rejectionCell infiltrationSCID/beige miceCell-permeable peptideBlood cytokine levelsT cell alloresponsesCD8 T cellsChemokine receptor expressionGranzyme B expressionAlloreactive T cellsSignificant side effectsDouble knockout miceHuman T cell activationBcl-2-transduced human umbilical vein endothelial cellsT cell activationHuman umbilical vein endothelial cellsUmbilical vein endothelial cellsSystemic immunosuppressantsAllograft rejectionRegulation of Yes-Associated Protein by Laminar Flow
Chitragari G, Shalaby SY, Sumpio BJ, Kurita J, Sumpio BE. Regulation of Yes-Associated Protein by Laminar Flow. Annals Of Vascular Surgery 2018, 52: 183-191. PMID: 29758328, DOI: 10.1016/j.avsg.2018.03.002.Peer-Reviewed Original ResearchMeSH KeywordsAdaptor Proteins, Signal TransducingCell ShapeCells, CulturedCytoskeletonDown-RegulationHuman Umbilical Vein Endothelial CellsHumansMechanotransduction, CellularPhosphoproteinsPhosphorylationProteolysisPulsatile FlowRegional Blood FlowStress, MechanicalTime FactorsTranscription FactorsYAP-Signaling ProteinsConceptsHuman umbilical vein endothelial cellsTotal YAPExposure of HUVECsRelease of mediatorsYAP levelsSignificant decreaseUbiquitin-proteasome system inhibitorsUmbilical vein endothelial cellsEndothelial cell proliferationVein endothelial cellsVascular healthSystem inhibitorsYes-Associated ProteinDisease modulationIntimal hyperplasiaDisturbed flowEndothelial cellsBlood vesselsDisease statesPotential targetCell proliferationProteasome-independent mechanismExposureKey regulatorPYAPCinnamaldehyde accelerates wound healing by promoting angiogenesis via up-regulation of PI3K and MAPK signaling pathways
Yuan X, Han L, Fu P, Zeng H, Lv C, Chang W, Runyon R, Ishii M, Han L, Liu K, Fan T, Zhang W, Liu R. Cinnamaldehyde accelerates wound healing by promoting angiogenesis via up-regulation of PI3K and MAPK signaling pathways. Laboratory Investigation 2018, 98: 783-798. PMID: 29463877, DOI: 10.1038/s41374-018-0025-8.Peer-Reviewed Original ResearchMeSH KeywordsAcroleinAnimalsCell MovementCell ProliferationCells, CulturedHuman Umbilical Vein Endothelial CellsHumansMaleMAP Kinase Signaling SystemMiceMice, Inbred C57BLNeovascularization, PhysiologicNitric Oxide Synthase Type IIIPhosphatidylinositol 3-KinasePhthalazinesPyridinesUp-RegulationVascular Endothelial Growth Factor AWound HealingZebrafishConceptsHuman umbilical vein endothelial cellsMAPK signaling pathwaysSignaling pathwaysMitogen-activated protein kinase pathwayProtein kinase pathwaySmall non-peptide moleculesVascular endothelial growth factor receptorPI3K/AktIntersegmental vesselsGrowth factor receptorVascular endothelial growth factorKinase pathwayUmbilical vein endothelial cellsVein endothelial cellsPI3KFactor receptorTube formationNon-peptide moleculesEndothelial growth factor receptorPathway
2017
Lead promotes abnormal angiogenesis induced by CCM3 gene defects via mitochondrial pathway
Sun Y, Zhang H, Xing X, Zhao Z, He J, Li J, Chen J, Wang M, He Y. Lead promotes abnormal angiogenesis induced by CCM3 gene defects via mitochondrial pathway. Journal Of Developmental Origins Of Health And Disease 2017, 9: 182-190. PMID: 29110746, DOI: 10.1017/s2040174417000782.Peer-Reviewed Original ResearchConceptsMouse embryosYolk sacHeterozygous mouse embryosGene defectsCCM3 genesPrimary human umbilical vein endothelial cellsLead exposureMitochondrial DNAEmbryonic developmentMtDNA biogenesisMitochondrial morphologyCardiovascular developmentHuman umbilical vein endothelial cellsMitochondrial pathwayGene knockoutEndothelial cellsUmbilical vein endothelial cellsVascular developmentMitochondria pathwayVein endothelial cellsPrimary cellsGenesRNA expressionCell proliferationEmbryosEnhanced Therapeutic and Long-Term Dynamic Vascularization Effects of Human Pluripotent Stem Cell–Derived Endothelial Cells Encapsulated in a Nanomatrix Gel
Lee SJ, Sohn YD, Andukuri A, Kim S, Byun J, Han JW, Park IH, Jun HW, Yoon YS. Enhanced Therapeutic and Long-Term Dynamic Vascularization Effects of Human Pluripotent Stem Cell–Derived Endothelial Cells Encapsulated in a Nanomatrix Gel. Circulation 2017, 136: 1939-1954. PMID: 28972000, PMCID: PMC5685906, DOI: 10.1161/circulationaha.116.026329.Peer-Reviewed Original ResearchConceptsCell survivalHPSC-ECsHuman pluripotent stem cell-derived endothelial cellsEndothelial lineage differentiationGlycogen synthase kinase-3β inhibitorHuman pluripotent stem cellsStem cell-derived endothelial cellsGrowth factorDifferentiation of hPSCsLonger cell survivalEndothelial cellsCell-derived endothelial cellsVessel formationPluripotent stem cell-derived endothelial cellsBetter perfusion recoveryPluripotent stem cellsNanomatrix gelLong-term cell survivalMesodermal lineagesLineage differentiationHuman umbilical vein endothelial cellsUmbilical vein endothelial cellsDifferentiation systemFibroblast growth factorBasic fibroblast growth factorC/EBPd enables the efficient extravasation of human CCR2+ MAIT cells
Zhang H, Lee C, Too L, Singh S, Tsang H, Kabat J, Singh T, Farber J. C/EBPd enables the efficient extravasation of human CCR2+ MAIT cells. The Journal Of Immunology 2017, 198: 143.10-143.10. DOI: 10.4049/jimmunol.198.supp.143.10.Peer-Reviewed Original ResearchMAIT cellsHuman umbilical vein endothelial cellsT cellsHuman mucosal-associated invariant T (MAIT) cellsMucosal-associated invariant T (MAIT) cellsBZIP transcription factorsResponse to pathogen challengeHuman MAIT cellsCD8+ T cellsExpression of FUT7Activated human umbilical vein endothelial cellsChemokine receptor CCR6Non-redundant roleSelectin ligandsSites of inflammationPathogen challengeTranscription factorsUmbilical vein endothelial cellsMR1-restrictedCD8+Receptor CCR6Efficient extravasationInflamed skinVein endothelial cellsChemokine receptors
2016
Influence of gestational diabetes mellitus on human umbilical vein endothelial cell miRNA.
Tryggestad J, Vishwanath A, Jiang S, Mallappa A, Teague A, Takahashi Y, Thompson D, Chernausek S. Influence of gestational diabetes mellitus on human umbilical vein endothelial cell miRNA. Clinical Science 2016, 130: 1955-67. PMID: 27562513, PMCID: PMC5516927, DOI: 10.1042/cs20160305.Peer-Reviewed Original ResearchConceptsGestational diabetes mellitusMiR-130bMiR-let-7a-5pMiR-148aLevel of miR-130bHuman umbilical vein endothelial cellsMiR-148a-3pMiR-30c-5pMiR-148a mimicsInfants of womenMiR-130b-3pMiRNA expressionMiR-452MiR-126Influence of gestational diabetes mellitusDiabetes mellitusTarget protein expressionProtein expressionTarget protein abundanceMiRNAsMonths of ageUmbilical vein endothelial cellsMiRNA speciesQuantitative PCRFetal tissuesNew Functional Tools for Antithrombogenic Activity Assessment of Live Surface Glycocalyx
Dimitrievska S, Gui L, Weyers A, Lin T, Cai C, Wu W, Tuggle CT, Sundaram S, Balestrini JL, Slattery D, Tchouta L, Kyriakides TR, Tarbell JM, Linhardt RJ, Niklason LE. New Functional Tools for Antithrombogenic Activity Assessment of Live Surface Glycocalyx. Arteriosclerosis Thrombosis And Vascular Biology 2016, 36: 1847-1853. PMID: 27386939, PMCID: PMC5283952, DOI: 10.1161/atvbaha.116.308023.Peer-Reviewed Original ResearchMeSH KeywordsAnimalsAntithrombinsAorta, ThoracicBiological AssayBlood CoagulationCells, CulturedChromatography, LiquidFactor XaGlycocalyxHeparinHeparitin SulfateHuman Umbilical Vein Endothelial CellsMaleMass SpectrometryMicroscopy, Electron, TransmissionRats, Sprague-DawleyReproducibility of ResultsThrombinTime FactorsConceptsHuman umbilical vein endothelial cellsUmbilical vein endothelial cellsVein endothelial cellsNative aortaEndothelial cellsHuman umbilical vein endothelial cell monolayersCultured human umbilical vein endothelial cellsFactor XaVascular healthEndothelial cell monolayersVascular diseaseRat aortaDamaged vasculatureAnticoagulant capacityVascular communityAortaTherapeutic developmentActivity assessmentCell monolayersVascular graftsLiquid chromatography-mass spectrometry analysisNovel assayGlycocalyxAssaysNative rat aorta
This site is protected by hCaptcha and its Privacy Policy and Terms of Service apply