Please use this identifier to cite or link to this item: https://doi.org/10.1038/onc.2012.517
Title: Non-canonical Notch signaling activates IL-6/JAK/STAT signaling in breast tumor cells and is controlled by p53 and IKKα/IKKβ
Authors: Jin, S.
Mutvei, A.P.
Chivukula, I.V.
Andersson, E.R.
Ramsköld, D.
Sandberg, R.
Lee, K.L. 
Kronqvist, P.
Mamaeva, V.
Östling, P.
Mpindi, J.-P.
Kallioniemi, O.
Screpanti, I.
Poellinger, L. 
Sahlgren, C.
Lendahl, U.
Keywords: BCL
breast cancer
Cytokine
estrogen receptor
inflammation
tumor stroma
Issue Date: 10-Oct-2013
Citation: Jin, S., Mutvei, A.P., Chivukula, I.V., Andersson, E.R., Ramsköld, D., Sandberg, R., Lee, K.L., Kronqvist, P., Mamaeva, V., Östling, P., Mpindi, J.-P., Kallioniemi, O., Screpanti, I., Poellinger, L., Sahlgren, C., Lendahl, U. (2013-10-10). Non-canonical Notch signaling activates IL-6/JAK/STAT signaling in breast tumor cells and is controlled by p53 and IKKα/IKKβ. Oncogene 32 (41) : 4892-4902. ScholarBank@NUS Repository. https://doi.org/10.1038/onc.2012.517
Abstract: Notch signaling is frequently hyperactivated in breast cancer, but how the enhanced signaling contributes to the tumor process is less well understood. In this report, we identify the proinflammatory cytokine interleukin-6 (IL-6) as a novel Notch target in breast tumor cells. Enhanced Notch signaling upregulated IL-6 expression, leading to activation of autocrine and paracrine Janus kinase/signal transducers and activators of transcription signaling. IL-6 upregulation was mediated by non-canonical Notch signaling, as it could be effectuated by a cytoplasmically localized Notch intracellular domain and was independent of the DNA-binding protein CSL. Instead, Notch-mediated IL-6 upregulation was controlled by two proteins in the nuclear factor (NF)-κB signaling cascade, IKKα and IKKβ (inhibitor of nuclear factor kappa-B kinase subunit alpha and beta, respectively), as well as by p53. Activation of IL-6 by Notch required IKKα/IKKβ function, but interestingly, did not engage canonical NF-κB signaling, in contrast to IL-6 activation by inflammatory agents such as lipopolysaccharide. With regard to p53 status, IL-6 expression was upregulated by Notch when p53 was mutated or lost, and restoring wild-type p53 into p53-mutated or -deficient cells abrogated the IL-6 upregulation. Furthermore, Notch-induced transcriptomes from p53 wild-type and -mutated breast tumor cell lines differed extensively, and for a subset of genes upregulated by Notch in a p53-mutant cell line, this upregulation was reduced by wild-type p53. In conclusion, we identify IL-6 as a novel non-canonical Notch target gene, and reveal roles for p53 and IKKα/IKKβ in non-canonical Notch signaling in breast cancer and in the generation of cell context-dependent diversity in the Notch signaling output. © 2013 Macmillan Publishers Limited All rights reserved.
Source Title: Oncogene
URI: http://scholarbank.nus.edu.sg/handle/10635/115834
ISSN: 09509232
DOI: 10.1038/onc.2012.517
Appears in Collections:Staff Publications

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

Google ScholarTM

Check

Altmetric


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