Please use this identifier to cite or link to this item: http://dr.iiserpune.ac.in:8080/xmlui/handle/123456789/1734
Full metadata record
DC FieldValueLanguage
dc.contributor.authorSubramaniam, S.en_US
dc.contributor.authorSreenivas, P.en_US
dc.contributor.authorCheedipudi, S.en_US
dc.contributor.authorReddy, V.R.en_US
dc.contributor.authorSHASHIDHARA, L.S.en_US
dc.contributor.authorChilukoti, R.K.en_US
dc.contributor.authorMylavarapu, M.en_US
dc.contributor.authorDhawan, J.en_US
dc.date.accessioned2019-02-14T05:05:04Z-
dc.date.available2019-02-14T05:05:04Z-
dc.date.issued2013-06en_US
dc.identifier.citationPLoS ONE, 8(6),0065097.en_US
dc.identifier.issn1932-6203en_US
dc.identifier.urihttp://dr.iiserpune.ac.in:8080/xmlui/handle/123456789/1734-
dc.identifier.urihttps://doi.org/10.1371/journal.pone.0065097en_US
dc.description.abstractMost cells in adult mammals are non-dividing: differentiated cells exit the cell cycle permanently, but stem cells exist in a state of reversible arrest called quiescence. In damaged skeletal muscle, quiescent satellite stem cells re-enter the cell cycle, proliferate and subsequently execute divergent programs to regenerate both post-mitotic myofibers and quiescent stem cells. The molecular basis for these alternative programs of arrest is poorly understood. In this study, we used an established myogenic culture model (C2C12 myoblasts) to generate cells in alternative states of arrest and investigate their global transcriptional profiles. Using cDNA microarrays, we compared G0 myoblasts with post-mitotic myotubes. Our findings define the transcriptional program of quiescent myoblasts in culture and establish that distinct gene expression profiles, especially of tumour suppressor genes and inhibitors of differentiation characterize reversible arrest, distinguishing this state from irreversibly arrested myotubes. We also reveal the existence of a tissue-specific quiescence program by comparing G0 C2C12 myoblasts to isogenic G0 fibroblasts (10T1/2). Intriguingly, in myoblasts but not fibroblasts, quiescence is associated with a signature of Wnt pathway genes. We provide evidence that different levels of signaling via the canonical Wnt pathway characterize distinct cellular states (proliferation vs. quiescence vs. differentiation). Moderate induction of Wnt signaling in quiescence is associated with critical properties such as clonogenic self-renewal. Exogenous Wnt treatment subverts the quiescence program and negatively affects clonogenicity. Finally, we identify two new quiescence-induced regulators of canonical Wnt signaling, Rgs2 and Dkk3, whose induction in G0 is required for clonogenic self-renewal. These results support the concept that active signal-mediated regulation of quiescence contributes to stem cell properties, and have implications for pathological states such as cancer and degenerative disease.en_US
dc.language.isoenen_US
dc.publisherPublic Library Scienceen_US
dc.subjectDistinct Transcriptional Networksen_US
dc.subjectIrreversible Arresten_US
dc.subjectDifferentiated cellsen_US
dc.subjectPathological statesen_US
dc.subjectClonogenic self-renewalen_US
dc.subject2013en_US
dc.titleDistinct Transcriptional Networks in Quiescent Myoblasts: A Role for Wnt Signaling in Reversible vs. Irreversible Arresten_US
dc.typeArticleen_US
dc.contributor.departmentDept. of Biologyen_US
dc.identifier.sourcetitlePLoS ONEen_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.