Deficiency in fibroblast PPARβ/δ reduces nonmelanoma skin cancers in mice.

Details

Ressource 1Request a copy Under indefinite embargo.
UNIL restricted access
State: Public
Version: Final published version
License: Not specified
Serval ID
serval:BIB_C09E5218BD1D
Type
Article: article from journal or magazin.
Collection
Publications
Institution
Title
Deficiency in fibroblast PPARβ/δ reduces nonmelanoma skin cancers in mice.
Journal
Cell death and differentiation
Author(s)
Tan MWY, Sng M.K., Cheng H.S., Low Z.S., Leong BJJ, Chua D., Tan EHP, Chan JSK, Yip Y.S., Lee Y.H., Pal M., Wang X., Wahli W., Tan N.S.
ISSN
1476-5403 (Electronic)
ISSN-L
1350-9047
Publication state
Published
Issued date
09/2020
Peer-reviewed
Oui
Volume
27
Number
9
Pages
2668-2680
Language
english
Notes
Publication types: Journal Article ; Research Support, Non-U.S. Gov't
Publication Status: ppublish
Abstract
The incidence of nonmelanoma skin cancer (NMSC) has been increasing worldwide. Most studies have highlighted the importance of cancer-associated fibroblasts (CAFs) in NMSC progression. However much less is known about the communication between normal fibroblasts and epithelia; disruption of this communication affects tumor initiation and the latency period in the emergence of tumors. Delineating the mechanism that mediates this epithelial-mesenchymal communication in NMSC could identify more effective targeted therapies. The nuclear receptor PPARβ/δ in fibroblasts has been shown to modulate adjacent epithelial cell behavior, however, its role in skin tumorigenesis remains unknown. Using chemically induced skin carcinogenesis, we showed that FSPCre-Pparb/d <sup>ex4</sup> mice, whose Pparb/d gene was selectively deleted in fibroblasts, had delayed emergence and reduced tumor burden compared with control mice (Pparb/d <sup>fl/fl</sup> ). However, FSPCre-Pparb/d <sup>ex4</sup> -derived tumors showed increased proliferation, with no difference in differentiation, suggesting delayed tumor initiation. Network analysis revealed a link between dermal Pparb/d and TGF-β1 with epidermal NRF2 and Nox4. In vitro investigations showed that PPARβ/δ deficiency in fibroblasts increased epidermal Nox4-derived H <sub>2</sub> O <sub>2</sub> production, which triggered an NRF2-mediated antioxidant response. We further showed that H <sub>2</sub> O <sub>2</sub> upregulated NRF2 mRNA via the B-Raf-MEK1/2 pathway. The enhanced NRF2 response altered the activities of PTEN, Src, and AKT. In vivo, we detected the differential phosphorylation profiles of B-Raf, MEK1/2, PTEN, Src, and AKT in the vehicle-treated and chemically treated epidermis of FSPCre-Pparb/d <sup>ex4</sup> mice compared with that in Pparb/d <sup>fl/fl</sup> mice, prior to the first appearance of tumors in Pparb/d <sup>fl/fl</sup> . Our study revealed a role for fibroblast PPARβ/δ in the epithelial-mesenchymal communication involved in cellular redox homeostasis.
Keywords
Animals, Carcinogenesis/metabolism, Carcinogenesis/pathology, Epidermis/pathology, Fibroblasts/metabolism, Fibroblasts/pathology, Gene Regulatory Networks, Glycoproteins/metabolism, Keratinocytes/metabolism, Kinetics, Melanoma/metabolism, Melanoma/pathology, Mice, Transgenic, NADPH Oxidase 4/metabolism, NF-E2-Related Factor 2/metabolism, Neoplasm Proteins/metabolism, PPAR delta/deficiency, PPAR delta/metabolism, PPAR-beta/deficiency, PPAR-beta/metabolism, Phosphorylation, Reactive Oxygen Species/metabolism, Signal Transduction, Skin Neoplasms/genetics, Skin Neoplasms/metabolism, Skin Neoplasms/pathology, Transforming Growth Factor beta1/metabolism, Tumor Burden
Pubmed
Web of science
Open Access
Yes
Create date
25/04/2020 18:50
Last modification date
26/07/2023 7:00
Usage data