Skip to main content

Lund University Publications

LUND UNIVERSITY LIBRARIES

Increased Nuclear Transporter KPNA2 Contributes to Tumor Immune Evasion by Enhancing PD-L1 Expression in PDAC

Zhou, Kai-Xia ; Huang, Shan ; Hu, Li-Peng ; Zhang, Xue-Li ; Qin, Wei-Ting ; Zhang, Yan-Li ; Yao, Lin-Li ; Yu, Yanqiu ; Zhou, Yao-Qi and Zhu, Lei , et al. (2021) In Journal of Immunology Research 2021. p.6694392-6694392
Abstract

Pancreatic ductal adenocarcinoma (PDAC) is one of the deadliest malignancies and is known for its high resistance and low response to treatment. Tumor immune evasion is a major stumbling block in designing effective anticancer therapeutic strategies. Karyopherin alpha 2 (KPNA2), a member of the nuclear transporter family, is elevated in multiple human cancers and accelerates carcinogenesis. However, the specific role of KPNA2 in PDAC remains unclear. In this study, we found that expression of KPNA2 was significantly upregulated in PDAC compared to adjacent nontumor tissue and its high expression was correlated with poor survival outcome by analyzing the GEO datasets. Similar KPNA2 expression pattern was also found in both human patient... (More)

Pancreatic ductal adenocarcinoma (PDAC) is one of the deadliest malignancies and is known for its high resistance and low response to treatment. Tumor immune evasion is a major stumbling block in designing effective anticancer therapeutic strategies. Karyopherin alpha 2 (KPNA2), a member of the nuclear transporter family, is elevated in multiple human cancers and accelerates carcinogenesis. However, the specific role of KPNA2 in PDAC remains unclear. In this study, we found that expression of KPNA2 was significantly upregulated in PDAC compared to adjacent nontumor tissue and its high expression was correlated with poor survival outcome by analyzing the GEO datasets. Similar KPNA2 expression pattern was also found in both human patient samples and KPC mouse models through IHC staining. Although KPNA2 knockdown failed to impair the vitality and migration ability of PDAC cells in vitro, the in vivo tumor growth was significantly impeded and the expression of immune checkpoint ligand PD-L1 was reduced by silencing KPNA2. Furthermore, we uncovered that KPNA2 modulated the expression of PD-L1 by mediating nuclear translocation of STAT3. Collectively, our data suggested that KPNA2 has the potential to serve as a promising biomarker for diagnosis in PDAC.

(Less)
Please use this url to cite or link to this publication:
author
; ; ; ; ; ; ; ; and , et al. (More)
; ; ; ; ; ; ; ; ; ; and (Less)
organization
publishing date
type
Contribution to journal
publication status
published
subject
in
Journal of Immunology Research
volume
2021
pages
6694392 - 6694392
publisher
Hindawi Limited
external identifiers
  • scopus:85102855848
  • pmid:33728352
ISSN
2314-7156
DOI
10.1155/2021/6694392
language
English
LU publication?
yes
id
2d1863c4-3172-4bc7-ad6b-cd8e2fa245f5
date added to LUP
2021-03-25 07:53:31
date last changed
2024-04-18 04:21:12
@article{2d1863c4-3172-4bc7-ad6b-cd8e2fa245f5,
  abstract     = {{<p>Pancreatic ductal adenocarcinoma (PDAC) is one of the deadliest malignancies and is known for its high resistance and low response to treatment. Tumor immune evasion is a major stumbling block in designing effective anticancer therapeutic strategies. Karyopherin alpha 2 (KPNA2), a member of the nuclear transporter family, is elevated in multiple human cancers and accelerates carcinogenesis. However, the specific role of KPNA2 in PDAC remains unclear. In this study, we found that expression of KPNA2 was significantly upregulated in PDAC compared to adjacent nontumor tissue and its high expression was correlated with poor survival outcome by analyzing the GEO datasets. Similar KPNA2 expression pattern was also found in both human patient samples and KPC mouse models through IHC staining. Although KPNA2 knockdown failed to impair the vitality and migration ability of PDAC cells in vitro, the in vivo tumor growth was significantly impeded and the expression of immune checkpoint ligand PD-L1 was reduced by silencing KPNA2. Furthermore, we uncovered that KPNA2 modulated the expression of PD-L1 by mediating nuclear translocation of STAT3. Collectively, our data suggested that KPNA2 has the potential to serve as a promising biomarker for diagnosis in PDAC.</p>}},
  author       = {{Zhou, Kai-Xia and Huang, Shan and Hu, Li-Peng and Zhang, Xue-Li and Qin, Wei-Ting and Zhang, Yan-Li and Yao, Lin-Li and Yu, Yanqiu and Zhou, Yao-Qi and Zhu, Lei and Ji, Jianguang and Zhang, Zhi-Gang}},
  issn         = {{2314-7156}},
  language     = {{eng}},
  pages        = {{6694392--6694392}},
  publisher    = {{Hindawi Limited}},
  series       = {{Journal of Immunology Research}},
  title        = {{Increased Nuclear Transporter KPNA2 Contributes to Tumor Immune Evasion by Enhancing PD-L1 Expression in PDAC}},
  url          = {{http://dx.doi.org/10.1155/2021/6694392}},
  doi          = {{10.1155/2021/6694392}},
  volume       = {{2021}},
  year         = {{2021}},
}