文献检索文档翻译深度研究
Suppr Zotero 插件Zotero 插件
邀请有礼套餐&价格历史记录

新学期,新优惠

限时优惠:9月1日-9月22日

30天高级会员仅需29元

1天体验卡首发特惠仅需5.99元

了解详情
不再提醒
插件&应用
Suppr Zotero 插件Zotero 插件浏览器插件Mac 客户端Windows 客户端微信小程序
高级版
套餐订阅购买积分包
AI 工具
文献检索文档翻译深度研究
关于我们
关于 Suppr公司介绍联系我们用户协议隐私条款
关注我们

Suppr 超能文献

核心技术专利:CN118964589B侵权必究
粤ICP备2023148730 号-1Suppr @ 2025

Cytoplasmic HMGB1 promotes the activation of JAK2-STAT3 signaling and PD-L1 expression in breast cancer.

作者信息

Han Ju-Young, Rhee Woo Joong, Shin Jeon-Soo

机构信息

Department of Microbiology, Yonsei University College of Medicine, 50-1 Yonsei-ro Seodaemun-gu, Seoul, 03722, South Korea.

Brain Korea 21 FOUR Project for Medical Science, Yonsei University College of Medicine, Seoul, 03722, South Korea.

出版信息

Mol Med. 2025 May 19;31(1):197. doi: 10.1186/s10020-025-01235-0.


DOI:10.1186/s10020-025-01235-0
PMID:40389855
原文链接:https://pmc.ncbi.nlm.nih.gov/articles/PMC12090602/
Abstract

BACKGROUND: High-mobility group box 1 (HMGB1) plays various roles depending on its subcellular localization. Extracellular HMGB1 interacts with receptors, such as toll-like receptor 4 and receptor for advanced glycation end products (RAGE), promoting cell proliferation, survival, and migration in cancer cells. It also increases the expression of programmed death-ligand 1 (PD-L1) in cancer cells by binding to RAGE. However, the effect of intracellular HMGB1 on the regulation of immune checkpoints such as PD-L1 has not been well characterized. In this study, we aimed to investigate the effects of intracellular HMGB1 on PD-L1 expression in breast cancer cells. METHODS: Human and mouse triple-negative breast cancer cells, MDA-MB-231 and 4T1, along with HMGB1-deficient mouse embryonic fibroblast cells, were cultured. HMGB1 overexpression was achieved using a Myc-tagged plasmid, while siHMGB1 constructs were used for gene silencing. Quantitative reverse-transcriptase PCR and western blot analysis were performed to assess gene and protein expressions. Confocal imaging, immunoprecipitation, and proximity ligation assays were used to investigate HMGB1 localization and Janus kinase 2 (JAK2)-signal transducer and activator of transcription 3 (STAT3) interactions. In vivo experiments were performed using tumor-bearing mice treated with STAT3 and HMGB1 inhibitors. Statistical analyses were performed using Student's t-tests, one-way analysis of variance, Pearson's correlation, and Kaplan-Meier survival analysis, with significance set at p < 0.05. RESULTS: In breast cancer cells, HMGB1 translocation from the nucleus to the cytoplasm increased the JAK2-STAT3 interaction and induced STAT3 phosphorylation, leading to increased STAT3 target signaling, including the epithelial-mesenchymal transition (EMT) phenotype and PD-L1 expression. Inhibition of nucleo-cytoplasmic translocation of HMGB1 decreased STAT3 phosphorylation and PD-L1 expression. Furthermore, HMGB1 enhanced breast cancer cell migration, invasion, and EMT, contributing to tumor growth in an in vivo mouse model that were mitigated by the HMGB1-targeted approach. CONCLUSIONS: These findings underscore the critical role of intracellular HMGB1 in modulating PD-L1 expression via the JAK2-STAT3 signaling pathways in breast cancer and suggest that targeting HMGB1 translocation is a promising strategy for breast cancer treatment.

摘要
https://cdn.ncbi.nlm.nih.gov/pmc/blobs/dada/12090602/fb64b47fb591/10020_2025_1235_Fig7_HTML.jpg
https://cdn.ncbi.nlm.nih.gov/pmc/blobs/dada/12090602/013662d04351/10020_2025_1235_Fig1_HTML.jpg
https://cdn.ncbi.nlm.nih.gov/pmc/blobs/dada/12090602/08e529c81892/10020_2025_1235_Fig2_HTML.jpg
https://cdn.ncbi.nlm.nih.gov/pmc/blobs/dada/12090602/7dcb70a0acde/10020_2025_1235_Fig3_HTML.jpg
https://cdn.ncbi.nlm.nih.gov/pmc/blobs/dada/12090602/910a409a0353/10020_2025_1235_Fig4_HTML.jpg
https://cdn.ncbi.nlm.nih.gov/pmc/blobs/dada/12090602/5a3eaebe02d9/10020_2025_1235_Fig5_HTML.jpg
https://cdn.ncbi.nlm.nih.gov/pmc/blobs/dada/12090602/459a374b7df9/10020_2025_1235_Fig6_HTML.jpg
https://cdn.ncbi.nlm.nih.gov/pmc/blobs/dada/12090602/fb64b47fb591/10020_2025_1235_Fig7_HTML.jpg
https://cdn.ncbi.nlm.nih.gov/pmc/blobs/dada/12090602/013662d04351/10020_2025_1235_Fig1_HTML.jpg
https://cdn.ncbi.nlm.nih.gov/pmc/blobs/dada/12090602/08e529c81892/10020_2025_1235_Fig2_HTML.jpg
https://cdn.ncbi.nlm.nih.gov/pmc/blobs/dada/12090602/7dcb70a0acde/10020_2025_1235_Fig3_HTML.jpg
https://cdn.ncbi.nlm.nih.gov/pmc/blobs/dada/12090602/910a409a0353/10020_2025_1235_Fig4_HTML.jpg
https://cdn.ncbi.nlm.nih.gov/pmc/blobs/dada/12090602/5a3eaebe02d9/10020_2025_1235_Fig5_HTML.jpg
https://cdn.ncbi.nlm.nih.gov/pmc/blobs/dada/12090602/459a374b7df9/10020_2025_1235_Fig6_HTML.jpg
https://cdn.ncbi.nlm.nih.gov/pmc/blobs/dada/12090602/fb64b47fb591/10020_2025_1235_Fig7_HTML.jpg

相似文献

[1]
Cytoplasmic HMGB1 promotes the activation of JAK2-STAT3 signaling and PD-L1 expression in breast cancer.

Mol Med. 2025-5-19

[2]
HMGB1 mediates invasion and PD-L1 expression through RAGE-PI3K/AKT signaling pathway in MDA-MB-231 breast cancer cells.

BMC Cancer. 2022-5-24

[3]
MicroRNA-375 reverses the expression of PD-L1 by inactivating the JAK2/STAT3 signaling pathways in gastric cancer.

Clin Res Hepatol Gastroenterol. 2021-9

[4]
Ectopic PD-L1 expression in JAK2 (V617F) myeloproliferative neoplasm patients is mediated via increased activation of STAT3 and STAT5.

Hum Cell. 2020-7-14

[5]
Tumour cell-released autophagosomes promote lung metastasis by upregulating PD-L1 expression in pulmonary vascular endothelial cells in breast cancer.

Cell Oncol (Dordr). 2024-12

[6]
Curcumol inhibits the expression of programmed cell death-ligand 1 through crosstalk between hypoxia-inducible factor-1α and STAT3 (T705) signaling pathways in hepatic cancer.

J Ethnopharmacol. 2020-7-15

[7]
Activation of JAK2/STAT3 signaling by osteopontin promotes tumor growth in human breast cancer cells.

Carcinogenesis. 2009-11-19

[8]
Taraxacum mongolicum extract inhibited malignant phenotype of triple-negative breast cancer cells in tumor-associated macrophages microenvironment through suppressing IL-10 / STAT3 / PD-L1 signaling pathways.

J Ethnopharmacol. 2021-6-28

[9]
IL20RA signaling enhances stemness and promotes the formation of an immunosuppressive microenvironment in breast cancer.

Theranostics. 2021

[10]
Cell-intrinsic PD-L1 signaling drives immunosuppression by myeloid-derived suppressor cells through IL-6/Jak/Stat3 in PD-L1-high lung cancer.

J Immunother Cancer. 2025-3-6

本文引用的文献

[1]
ROS anchor PAMPs-mediated extracellular HMGB1 self-association and its dimerization enhances pro-inflammatory signaling.

Redox Biol. 2025-3

[2]
Correction: Epithelial-mesenchymal transition induced by tumor cell-intrinsic PD-L1 signaling predicts a poor response to immune checkpoint inhibitors in PD-L1-high lung cancer.

Br J Cancer. 2024-9

[3]
Unraveling the complexity of STAT3 in cancer: molecular understanding and drug discovery.

J Exp Clin Cancer Res. 2024-1-20

[4]
Role of HMGB1 and its associated signaling pathways in human malignancies.

Cell Signal. 2023-12

[5]
Correction: Advances in immunotherapy for triple-negative breast cancer.

Mol Cancer. 2023-9-14

[6]
Immunotherapy for Metastatic Triple Negative Breast Cancer: Current Paradigm and Future Approaches.

Curr Treat Options Oncol. 2023-6

[7]
STAT3-EMT axis in tumors: Modulation of cancer metastasis, stemness and therapy response.

Pharmacol Res. 2022-8

[8]
HMGB1 mediates invasion and PD-L1 expression through RAGE-PI3K/AKT signaling pathway in MDA-MB-231 breast cancer cells.

BMC Cancer. 2022-5-24

[9]
Immune Checkpoint Inhibitors in Triple Negative Breast Cancer: The Search for the Optimal Biomarker.

Biomark Insights. 2022-2-22

[10]
The mechanism of HMGB1 secretion and release.

Exp Mol Med. 2022-2

文献AI研究员

20分钟写一篇综述,助力文献阅读效率提升50倍

立即体验

用中文搜PubMed

大模型驱动的PubMed中文搜索引擎

马上搜索

推荐工具

医学文档翻译智能文献检索