HUSCAP logo Hokkaido Univ. logo

Hokkaido University Collection of Scholarly and Academic Papers >
Graduate School of Medicine / Faculty of Medicine >
Peer-reviewed Journal Articles, etc >

High expression of AMAP1, an ARF6 effector, is associated with elevated levels of PD-L1 and fibrosis of pancreatic cancer

This item is licensed under:Creative Commons Attribution 4.0 International

Files in This Item:

The file(s) associated with this item can be obtained from the following URL: https://doi.org/10.1186/s12964-020-00608-8


Title: High expression of AMAP1, an ARF6 effector, is associated with elevated levels of PD-L1 and fibrosis of pancreatic cancer
Authors: Tsutaho, Akio Browse this author
Hashimoto, Ari Browse this author →KAKEN DB
Hashimoto, Shigeru Browse this author →KAKEN DB
Hata, Soichiro Browse this author
Kachi, Shion Browse this author
Hirano, Satoshi Browse this author →KAKEN DB
Sabe, Hisataka Browse this author →KAKEN DB
Keywords: ARF6
AMAP1
Fibrosis
FAK
Pancreatic cancer
PD-L1
Issue Date: 24-Jun-2020
Publisher: BioMed Central
Journal Title: Cell communication and signaling
Volume: 18
Issue: 1
Start Page: 101
Publisher DOI: 10.1186/s12964-020-00608-8
Abstract: Background Not merely the onset of immune evasion, but other factors, such as acidosis and fibrosis, are also major barriers in cancer therapeutics. Dense fibrosis is a hallmark of pancreatic ductal carcinoma (PDAC), in which hyperactivation of focal adhesion kinase (FAK) in tumor cells was shown to be crucial. Double mutations ofKRAS/ TP53are characteristic to PDAC. We previously showed that high protein expression of ARF6 and its downstream effector AMAP1, as well as processes involved in the ARF6 activation by cell surface tyrosine kinase receptors, are major targets of theKRAS/TP53mutations to promote PDAC invasion, metastasis, and immune evasion. This notion was recaptured by KPC mouse model of human PDAC (LSL-Kras(G12D/+); LSL-Trp53(R172H/+)); Pdx-1-Cre). Mechanistically, the ARF6-AMAP1 pathway is primarily involved in cellular dynamics of PD-L1, beta 1-integrins, and E-cadherin; and hence modulates cell-adhesion properties when ARF6 is activated. Here, with an aim to understand whether the ARF6-AMAP1 pathway is critically involved in the elevated levels of PD-L1 and fibrosis of PDAC, we analyzed relationship between AMAP1 and these malignant phenotypes. Moreover, because the ARF6 pathway may closely be related to focal adhesion dynamics and hence to FAK, we also investigated whether AMAP1 employs FAK in fibrosis. Methods Clinical specimens, as well as KPC cells/tumors and theirshAMAP1orshFAKderivatives were analyzed. Results Elevated levels of PD-L1 and fibrosis correlated with poor outcome of our patient cohort, to be consistent with previous reports; in which high AMAP1 expression statistically correlated with the elevated PD-L1 and fibrosis. To be consistent, silencing ofAMAP1(shAMAP1) in KPC cells resulted in reduced PD-L1 expression and fibrosis in their tumors. On the other hand,shAMAP1only slightly affected FAK activation in KPC cells, and phosphorylated FAK did not correlate with enhanced fibrosis or with poor outcome of our patients. Conclusions Together with our previous data, our results collectively indicated that the ARF6-AMAP1 pathway, empowered by theKRAS/TP53mutations, is closely associated with elevated PD-L1 expression and fibrosis of human PDACs, to be recaptured in the KPC mouse model. The ARF6 pathway may promote fibrosis independent of FAK.
Rights: http://creativecommons.org/licenses/by/4.0/
Type: article
URI: http://hdl.handle.net/2115/78972
Appears in Collections:医学院・医学研究院 (Graduate School of Medicine / Faculty of Medicine) > 雑誌発表論文等 (Peer-reviewed Journal Articles, etc)

Export metadata:

OAI-PMH ( junii2 , jpcoar_1.0 )

MathJax is now OFF:


 

 - Hokkaido University