Show simple item record

dc.contributor.authorKerins, Michael John
dc.contributor.authorVashisht, Ajay Amar
dc.contributor.authorLiang, Benjamin Xi-Tong
dc.contributor.authorDuckworth, Spencer Jordan
dc.contributor.authorPraslicka, Brandon John
dc.contributor.authorWohlschlegel, James Akira
dc.contributor.authorOoi, Aikseng
dc.date.accessioned2017-06-19T23:42:45Z
dc.date.available2017-06-19T23:42:45Z
dc.date.issued2017-06-01
dc.identifier.citationFumarate Mediates a Chronic Proliferative Signal in Fumarate Hydratase-Inactivated Cancer Cells by Increasing Transcription and Translation of Ferritin Genes 2017, 37 (11):e00079-17 Molecular and Cellular Biologyen
dc.identifier.issn0270-7306
dc.identifier.issn1098-5549
dc.identifier.doi10.1128/MCB.00079-17
dc.identifier.urihttp://hdl.handle.net/10150/624216
dc.description.abstractGerm line mutations of the gene encoding the tricarboxylic acid (TCA) cycle enzyme fumarate hydratase (FH) cause a hereditary cancer syndrome known as hereditary leiomyomatosis and renal cell cancer (HLRCC). HLRCC-associated tumors harbor biallelic FH inactivation that results in the accumulation of the TCA cycle metabolite fumarate. Although it is known that fumarate accumulation can alter cellular signaling, if and how fumarate confers a growth advantage remain unclear. Here we show that fumarate accumulation confers a chronic proliferative signal by disrupting cellular iron signaling. Specifically, fumarate covalently modifies cysteine residues on iron regulatory protein 2 (IRP2), rendering it unable to repress ferritin mRNA translation. Simultaneously, fumarate increases ferritin gene transcription by activating the NRF2 (nuclear factor [erythroid-derived 2]-like 2) transcription factor. In turn, increased ferritin protein levels promote the expression of the promitotic transcription factor FOXM1 (Forkhead box protein M1). Consistently, clinical HLRCC tissues showed increased expression levels of both FOXM1 and its proliferation-associated target genes. This finding demonstrates how FH inactivation can endow cells with a growth advantage.
dc.description.sponsorshipNational Science Foundation Graduate Research Fellowship Program [DGE-1143953]; University of Arizona College of Pharmacy; University of Arizona Health Sciences grant [214565]en
dc.language.isoenen
dc.publisherAMER SOC MICROBIOLOGYen
dc.relation.urlhttp://mcb.asm.org/lookup/doi/10.1128/MCB.00079-17en
dc.rightsCopyright © 2017 American Society for Microbiology.en
dc.rights.urihttp://rightsstatements.org/vocab/InC/1.0/
dc.subjectferritinen
dc.subjectFHen
dc.subjectFOXM1en
dc.subjectfumarateen
dc.subjectHLRCCen
dc.subjectNRF2en
dc.titleFumarate Mediates a Chronic Proliferative Signal in Fumarate Hydratase-Inactivated Cancer Cells by Increasing Transcription and Translation of Ferritin Genesen
dc.typeArticleen
dc.contributor.departmentUniv Arizona, Coll Pharm, Dept Pharmacol & Toxicolen
dc.identifier.journalMolecular and Cellular Biologyen
dc.description.note6 month embargo; published online 13 March 2017.en
dc.description.collectioninformationThis item from the UA Faculty Publications collection is made available by the University of Arizona with support from the University of Arizona Libraries. If you have questions, please contact us at repository@u.library.arizona.edu.en
dc.eprint.versionFinal published versionen
refterms.dateFOA2017-09-14T00:00:00Z
html.description.abstractGerm line mutations of the gene encoding the tricarboxylic acid (TCA) cycle enzyme fumarate hydratase (FH) cause a hereditary cancer syndrome known as hereditary leiomyomatosis and renal cell cancer (HLRCC). HLRCC-associated tumors harbor biallelic FH inactivation that results in the accumulation of the TCA cycle metabolite fumarate. Although it is known that fumarate accumulation can alter cellular signaling, if and how fumarate confers a growth advantage remain unclear. Here we show that fumarate accumulation confers a chronic proliferative signal by disrupting cellular iron signaling. Specifically, fumarate covalently modifies cysteine residues on iron regulatory protein 2 (IRP2), rendering it unable to repress ferritin mRNA translation. Simultaneously, fumarate increases ferritin gene transcription by activating the NRF2 (nuclear factor [erythroid-derived 2]-like 2) transcription factor. In turn, increased ferritin protein levels promote the expression of the promitotic transcription factor FOXM1 (Forkhead box protein M1). Consistently, clinical HLRCC tissues showed increased expression levels of both FOXM1 and its proliferation-associated target genes. This finding demonstrates how FH inactivation can endow cells with a growth advantage.


Files in this item

Thumbnail
Name:
Mol.Cell.Biol.-2017-Kerins-.pdf
Size:
1.366Mb
Format:
PDF
Description:
FInal Published Version

This item appears in the following Collection(s)

Show simple item record