Please use this identifier to cite or link to this item: https://ir.swu.ac.th/jspui/handle/123456789/12462
Full metadata record
DC FieldValueLanguage
dc.contributor.authorOsataphan S.
dc.contributor.authorMacchi C.
dc.contributor.authorSinghal G.
dc.contributor.authorChimene-Weiss J.
dc.contributor.authorSales V.
dc.contributor.authorKozuka C.
dc.contributor.authorDreyfuss J.M.
dc.contributor.authorPan H.
dc.contributor.authorTangcharoenpaisan Y.
dc.contributor.authorMorningstar J.
dc.contributor.authorGerszten R.
dc.contributor.authorPatti M.-E.
dc.date.accessioned2021-04-05T03:03:33Z-
dc.date.available2021-04-05T03:03:33Z-
dc.date.issued2019
dc.identifier.issn23793708
dc.identifier.other2-s2.0-85062630085
dc.identifier.urihttps://ir.swu.ac.th/jspui/handle/123456789/12462-
dc.identifier.urihttps://www.scopus.com/inward/record.uri?eid=2-s2.0-85062630085&doi=10.1172%2fjci.insight.123130&partnerID=40&md5=c24313c08333eed7ffc7fdbc42af7302
dc.description.abstractPharmacologic inhibition of the renal sodium/glucose cotransporter-2 induces glycosuria and reduces glycemia. Given that SGLT2 inhibitors (SGLT2i) reduce mortality and cardiovascular risk in type 2 diabetes, improved understanding of molecular mechanisms mediating these metabolic effects is required. Treatment of obese but nondiabetic mice with the SGLT2i canagliflozin (CANA) reduces adiposity, improves glucose tolerance despite reduced plasma insulin, increases plasma ketones, and improves plasma lipid profiles. Utilizing an integrated transcriptomic-metabolomics approach, we demonstrate that CANA modulates key nutrient-sensing pathways, with activation of 5' AMP-activated protein kinase (AMPK) and inhibition of mechanistic target of rapamycin (mTOR), independent of insulin or glucagon sensitivity or signaling. Moreover, CANA induces transcriptional reprogramming to activate catabolic pathways, increase fatty acid oxidation, reduce hepatic steatosis and diacylglycerol content, and increase hepatic and plasma levels of FGF21. Given that these phenotypes mirror the effects of FGF21 to promote lipid oxidation, ketogenesis, and reduction in adiposity, we hypothesized that FGF21 is required for CANA action. Using FGF21-null mice, we demonstrate that FGF21 is not required for SGLT2i-mediated induction of lipid oxidation and ketogenesis but is required for reduction in fat mass and activation of lipolysis. Taken together, these data demonstrate that SGLT2 inhibition triggers a fasting-like transcriptional and metabolic paradigm but requires FGF21 for reduction in adiposity.
dc.subjectcanagliflozin
dc.subjectdiacylglycerol
dc.subjectfibroblast growth factor
dc.subjectfibroblast growth factor 21
dc.subjecthydroxymethylglutaryl coenzyme A reductase kinase
dc.subjectinsulin
dc.subjectketone
dc.subjectlipid
dc.subjectrapamycin
dc.subjectSlc5a2 protein, mouse
dc.subjectsodium glucose cotransporter 2
dc.subjectanimal
dc.subjectblood
dc.subjectC57BL mouse
dc.subjectdiet restriction
dc.subjectdrug effect
dc.subjectenergy metabolism
dc.subjectfatty liver
dc.subjectgenetics
dc.subjectglucose blood level
dc.subjectknockout mouse
dc.subjectlipid metabolism
dc.subjectliver
dc.subjectmale
dc.subjectmetabolism
dc.subjectmouse
dc.subjectnon insulin dependent diabetes mellitus
dc.subjectnuclear reprogramming
dc.subjectobesity
dc.subjectpathology
dc.subjectpharmacology
dc.subjectsignal transduction
dc.subjectAdiposity
dc.subjectAMP-Activated Protein Kinases
dc.subjectAnimals
dc.subjectBlood Glucose
dc.subjectCanagliflozin
dc.subjectCellular Reprogramming
dc.subjectDiabetes Mellitus, Type 2
dc.subjectDiglycerides
dc.subjectEnergy Metabolism
dc.subjectFasting
dc.subjectFatty Liver
dc.subjectFibroblast Growth Factors
dc.subjectInsulin
dc.subjectKetones
dc.subjectLipid Metabolism
dc.subjectLipids
dc.subjectLiver
dc.subjectMale
dc.subjectMice
dc.subjectMice, Inbred C57BL
dc.subjectMice, Knockout
dc.subjectObesity
dc.subjectSignal Transduction
dc.subjectSirolimus
dc.subjectSodium-Glucose Transporter 2
dc.subjectSodium-Glucose Transporter 2 Inhibitors
dc.titleSGLT2 inhibition reprograms systemic metabolism via FGF21-dependent and -independent mechanisms
dc.typeArticle
dc.rights.holderScopus
dc.identifier.bibliograpycitationJCI insight. Vol 4, No.5 (2019)
dc.identifier.doi10.1172/jci.insight.123130
Appears in Collections:Scopus 1983-2021

Files in This Item:
There are no files associated with this item.


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