Please use this identifier to cite or link to this item: http://dr.iiserpune.ac.in:8080/xmlui/handle/123456789/2628
Full metadata record
DC FieldValueLanguage
dc.contributor.authorJadav, Rathan S.en_US
dc.contributor.authorKumar, Dharmikaen_US
dc.contributor.authorBuwa, Natashaen_US
dc.contributor.authorGanguli, Shubhraen_US
dc.contributor.authorThampatty, Sitalakshmi R.en_US
dc.contributor.authorBALASUBRAMANIAN, NAGARAJen_US
dc.contributor.authorBhandari, Rashnaen_US
dc.date.accessioned2019-04-29T09:21:00Z
dc.date.available2019-04-29T09:21:00Z
dc.date.issued2016-08en_US
dc.identifier.citationCellular Signalling, 28(8), 1124-1136.en_US
dc.identifier.issn0898-6568en_US
dc.identifier.issn1873-3913en_US
dc.identifier.urihttp://dr.iiserpune.ac.in:8080/xmlui/handle/123456789/2628-
dc.identifier.urihttps://doi.org/10.1016/j.cellsig.2016.04.011en_US
dc.description.abstractInositol hexakisphosphate kinases (IP6Ks), a family of enzymes found in all eukaryotes, are responsible for the synthesis of 5-diphosphoinositol pentakisphosphate (5-IP7) from inositol hexakisphosphate (IP6). Three isoforms of IP6Ks are found in mammals, and gene deletions of each isoform lead to diverse, non-overlapping phenotypes in mice. Previous studies show a facilitatory role for IP6K2 in cell migration and invasion, properties that are essential for the early stages of tumorigenesis. However, IP6K2 also has an essential role in cancer cell apoptosis, and mice lacking this protein are more susceptible to the development of aerodigestive tract carcinoma upon treatment with the oral carcinogen 4-nitroquinoline-1-oxide (4NQO). Not much is known about the functions of the equally abundant and ubiquitously expressed IP6K1 isoform in cell migration, invasion and cancer progression. We conducted a gene expression analysis on mouse embryonic fibroblasts (MEFs) lacking IP6K1, revealing a role for this protein in cell receptor-extracellular matrix interactions that regulate actin cytoskeleton dynamics. Consequently, cells lacking IP6K1 manifest defects in adhesion-dependent signaling, evident by lower FAK and Paxillin activation, leading to reduced cell spreading and migration. Expression of active, but not inactive IP6K1 reverses migration defects in IP6K1 knockout MEFs, suggesting that 5-IP7 synthesis by IP6K1 promotes cell locomotion. Actin cytoskeleton remodeling and cell migration support the ability of cancer cells to achieve their complete oncogenic potential. Cancer cells with lower IP6K1 levels display reduced migration, invasion, and anchorage-independent growth. When fed an oral carcinogen, mice lacking IP6K1 show reduced progression from epithelial dysplasia to invasive carcinoma. Thus, our data reveal that like IP6K2, IP6K1 is also involved in early cytoskeleton remodeling events during cancer progression. However, unlike IP6K2, IP6K1 is essential for 4NQO-induced invasive carcinoma. Our study therefore uncovers similarities and differences in the roles of IP6K1 and IP6K2 in cancer progression, and we propose that an isoform-specific IP6K1 inhibitor may provide a novel route to suppress carcinogenesis.en_US
dc.language.isoenen_US
dc.publisherElsevier B.V.en_US
dc.subjectGene-expression analysisen_US
dc.subjectCell spreading and migrationen_US
dc.subjectIP6K1 displayen_US
dc.subjectInositol hexakisphosphate kinasesen_US
dc.subjectAdhesionen_US
dc.subjectRalen_US
dc.subjectArf6Exocyst traffickingen_US
dc.subjectAnchorage independenceen_US
dc.subjectCanceren_US
dc.subject2016en_US
dc.titleDeletion of inositol hexakisphosphate kinase 1 (IP6K1) reduces cell migration and invasion, conferring protection from aerodigestive tract carcinoma in miceen_US
dc.typeArticleen_US
dc.contributor.departmentDept. of Biologyen_US
dc.identifier.sourcetitleCellular Signallingen_US
dc.publication.originofpublisherForeignen_US
Appears in Collections:JOURNAL ARTICLES

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


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