完整後設資料紀錄
DC 欄位語言
dc.contributor.authorTseng, Wei-Lienen_US
dc.contributor.authorChou, Shih-Jieen_US
dc.contributor.authorChiang, Huai-Chihen_US
dc.contributor.authorWang, Mong-Lienen_US
dc.contributor.authorChien, Chian-Shiuen_US
dc.contributor.authorChen, Kuan-Hsuanen_US
dc.contributor.authorLeu, Hsin-Bangen_US
dc.contributor.authorWang, Chien-Yingen_US
dc.contributor.authorChang, Yuh-Lihen_US
dc.contributor.authorLiu, Yung-Yangen_US
dc.contributor.authorJong, Yuh-Jyhen_US
dc.contributor.authorLin, Shinn-Zongen_US
dc.contributor.authorChiou, Shih-Hwaen_US
dc.contributor.authorLin, Shing-Jongen_US
dc.contributor.authorYu, Wen-Chungen_US
dc.date.accessioned2019-04-03T06:35:54Z-
dc.date.available2019-04-03T06:35:54Z-
dc.date.issued2017-01-01en_US
dc.identifier.issn0963-6897en_US
dc.identifier.urihttp://dx.doi.org/10.3727/096368916X694265en_US
dc.identifier.urihttp://hdl.handle.net/11536/145290-
dc.description.abstractFabry disease (FD) is an X-linked inherited lysosomal storage disease caused by alpha-galactosidase A (GLA) deficiency. Progressive intracellular accumulation of globotriaosylceramide (Gb3) is considered to be pathogenically responsible for the phenotype variability of FD that causes cardiovascular dysfunction; however, molecular mechanisms underlying the impairment of FD-associated cardiovascular tissues remain unclear. In this study, we reprogrammed human induced pluripotent stem cells (hiPSCs) from peripheral blood cells of patients with FD (FD-iPSCs); subsequently differentiated them into vascular endothelial-likecells (FD-ECs) expressing CD31, VE-cadherin, and vWF; and investigated their ability to form vascular tube-like structures. FD-ECs recapitulated the FD pathophysiological phenotype exhibiting intracellular Gb3 accumulation under a transmission electron microscope. Moreover, compared with healthy control iPSC-derived endothelial cells (NC-ECs), reactive oxygen species (ROS) production considerably increased in FD-ECs. Microarray analysis was performed to explore the possible mechanism underlying Gb3 accumulation-induced ROS production in FD-ECs. Our results revealed that superoxide dismutase 2 (SOD2), a mitochondrial antioxidant, was significantly downregulated in FD-ECs. Compared with NC-ECs, AMPK activity was significantly enhanced in FD-ECs. Furthermore, to investigate the role of Gb3 in these effects, human umbilical vein endothelial cells (HUVECs) were treated with Gb3. After Gb3 treatment, we observed that SOD2 expression was suppressed and AMPK activity was enhanced in a dose-dependent manner. Collectively, our results indicate that excess accumulation of Gb3 suppressed SOD2 expression, increased ROS production, enhanced AMPK activation, and finally caused vascular endothelial dysfunction. Our findings suggest that dysregulated mitochondrial ROS may be a potential target for treating FD.en_US
dc.language.isoen_USen_US
dc.subjectFabry disease (FD)en_US
dc.subjectVascular endothelial dysfunctionen_US
dc.subjectSuperoxide dismutase 2 (SOD2)en_US
dc.subjectGb3 accumulationen_US
dc.subjectInduced pluripotent stem cells (iPSCs)en_US
dc.titleImbalanced Production of Reactive Oxygen Species and Mitochondrial Antioxidant SOD2 in Fabry Disease-Specific Human Induced Pluripotent Stem Cell-Differentiated Vascular Endothelial Cellsen_US
dc.typeArticleen_US
dc.identifier.doi10.3727/096368916X694265en_US
dc.identifier.journalCELL TRANSPLANTATIONen_US
dc.citation.volume26en_US
dc.citation.issue3en_US
dc.citation.spage513en_US
dc.citation.epage527en_US
dc.contributor.department生物科技學系zh_TW
dc.contributor.departmentDepartment of Biological Science and Technologyen_US
dc.identifier.wosnumberWOS:000397839400014en_US
dc.citation.woscount5en_US
顯示於類別:期刊論文


文件中的檔案:

  1. cbcb8a0362093e0de8ddc5b343597242.pdf

若為 zip 檔案,請下載檔案解壓縮後,用瀏覽器開啟資料夾中的 index.html 瀏覽全文。