0 697

Cited 21 times in

Exogenous administration of DLK1 ameliorates hepatic steatosis and regulates gluconeogenesis via activation of AMPK

DC Field Value Language
dc.contributor.author강은석-
dc.contributor.author이병완-
dc.contributor.author이용호-
dc.contributor.author이현철-
dc.contributor.author차봉수-
dc.date.accessioned2017-02-24T03:34:31Z-
dc.date.available2017-02-24T03:34:31Z-
dc.date.issued2016-
dc.identifier.issn0307-0565-
dc.identifier.urihttps://ir.ymlib.yonsei.ac.kr/handle/22282913/146380-
dc.description.abstractBACKGROUND/OBJECTIVES: Activation of Notch signaling pathologically enhances lipogenesis and gluconeogenesis in the liver causing non-alcoholic fatty liver disease (NAFLD) and diabetes. Delta-like 1 homolog (DLK1), an imprinted gene that can modulate adipogenesis and muscle development in mice, was found as an inhibitory regulator of Notch signaling. Therefore, we investigated the metabolic effect of exogenous DLK1 in vitro and in vivo. SUBJECTS/METHODS: A soluble DLK1 peptide was generated with fusion between a human Fc fragment and extracellular domain of DLK1. Male db/db mice were randomly assigned to two groups: vehicle treated and DLK1-treated group (25 mg kg(-1), intraperitoneal injection, twice a week for 4 weeks). Primary mice hepatocytes and HepG2 cells were used for in vitro experiments. RESULTS: After 4 weeks of DLK1 administration, hepatic triglyceride content and lipid droplets in liver tissues, as well as serum levels of liver enzymes, were markedly decreased in db/db mice. DLK1 treatment induced phosphorylation of AMPK and ACC and suppressed nuclear expression of SREBP-1c in the mouse liver or hepatocytes, indicating regulation of fatty acid oxidation and synthesis pathways. Furthermore, DLK1-treated mice showed significantly lower levels of fasting and random glucose, with improved glucose and insulin tolerance compared with the vehicle-treated group. Macrophage infiltration and proinflammatory cytokine levels in the epididymal fat were decreased in DLK1-treated db/db mice. Moreover, DLK1 suppressed glucose production from hepatocytes, which was blocked after co-administration of an AMPK inhibitor, compound C. DLK1-treated hepatocytes and mouse liver tissues showed lower PEPCK and G6Pase expression. DLK1 triggered AKT phosphorylation followed by cytosolic translocation of FOXO1 from the nucleus in hepatocytes. CONCLUSIONS: The present study demonstrated that exogenous administration of DLK1 reduced hepatic steatosis and hyperglycemia via AMPK activation in the liver. This result suggests that DLK1 may be a novel therapeutic approach for treating NAFLD and diabetes.-
dc.description.statementOfResponsibilityrestriction-
dc.format.extent356~365-
dc.languageEnglish-
dc.publisherNature Pub. Group-
dc.relation.isPartOfINTERNATIONAL JOURNAL OF OBESITY-
dc.rightsCC BY-NC-ND 2.0 KR-
dc.rights.urihttps://creativecommons.org/licenses/by-nc-nd/2.0/kr/-
dc.subject.MESHAMP-Activated Protein Kinases-
dc.subject.MESHAnimals-
dc.subject.MESHDisease Models, Animal-
dc.subject.MESHGluconeogenesis-
dc.subject.MESHInjections, Intraperitoneal-
dc.subject.MESHIntercellular Signaling Peptides and Proteins/pharmacology*-
dc.subject.MESHLipid Metabolism-
dc.subject.MESHLiver/pathology*-
dc.subject.MESHMice-
dc.subject.MESHMice, Inbred C57BL-
dc.subject.MESHNon-alcoholic Fatty Liver Disease/pathology*-
dc.subject.MESHReceptors, Notch/drug effects*-
dc.subject.MESHReceptors, Notch/metabolism-
dc.subject.MESHSignal Transduction/drug effects*-
dc.titleExogenous administration of DLK1 ameliorates hepatic steatosis and regulates gluconeogenesis via activation of AMPK-
dc.typeArticle-
dc.publisher.locationEngland-
dc.contributor.collegeCollege of Medicine-
dc.contributor.departmentDept. of Internal Medicine-
dc.contributor.googleauthorY-h Lee-
dc.contributor.googleauthorMR Yun-
dc.contributor.googleauthorHM Kim-
dc.contributor.googleauthorBH Jeon-
dc.contributor.googleauthorB-C Park-
dc.contributor.googleauthorB-W Lee-
dc.contributor.googleauthorES Kang-
dc.contributor.googleauthorHC Lee-
dc.contributor.googleauthorYW Park-
dc.contributor.googleauthorB-S Cha-
dc.identifier.doi10.1038/ijo.2015.173-
dc.contributor.localIdA00068-
dc.contributor.localIdA02796-
dc.contributor.localIdA02989-
dc.contributor.localIdA03301-
dc.contributor.localIdA03996-
dc.relation.journalcodeJ01140-
dc.identifier.eissn1476-5497-
dc.identifier.pmid26315841-
dc.identifier.urlhttp://www.nature.com/ijo/journal/v40/n2/full/ijo2015173a.html-
dc.contributor.alternativeNameKang, Eun Seok-
dc.contributor.alternativeNameLee, Byung Wan-
dc.contributor.alternativeNameLee, Yong Ho-
dc.contributor.alternativeNameLee, Hyun Chul-
dc.contributor.alternativeNameCha, Bong Soo-
dc.contributor.affiliatedAuthorKang, Eun Seok-
dc.contributor.affiliatedAuthorLee, Byung Wan-
dc.contributor.affiliatedAuthorLee, Yong Ho-
dc.contributor.affiliatedAuthorLee, Hyun Chul-
dc.contributor.affiliatedAuthorCha, Bong Soo-
dc.citation.volume40-
dc.citation.number2-
dc.citation.startPage356-
dc.citation.endPage365-
dc.identifier.bibliographicCitationINTERNATIONAL JOURNAL OF OBESITY, Vol.40(2) : 356-365, 2016-
dc.date.modified2017-02-24-
dc.identifier.rimsid47888-
dc.type.rimsART-
Appears in Collections:
1. College of Medicine (의과대학) > Dept. of Internal Medicine (내과학교실) > 1. Journal Papers

qrcode

Items in DSpace are protected by copyright, with all rights reserved, unless otherwise indicated.