Cargando…

The interaction between the soluble programmed death ligand-1 (sPD-L1) and PD-1(+) regulator B cells mediates immunosuppression in triple-negative breast cancer

Accumulating evidence suggests that regulatory B cells (Bregs) play important roles in inhibiting the immune response in tumors. Programmed death 1 (PD-1) and programmed death ligand 1 (PD-L1) are important molecules that maintain the balance of the immune response and immune tolerance. This study a...

Descripción completa

Detalles Bibliográficos
Autores principales: Li, Xuejiao, Du, Huan, Zhan, Shenghua, Liu, Wenting, Wang, Zhangyu, Lan, Jing, PuYang, Longxiang, Wan, Yuqiu, Qu, Qiuxia, Wang, Sining, Yang, Yang, Wang, Qin, Xie, Fang
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Frontiers Media S.A. 2022
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9354578/
https://www.ncbi.nlm.nih.gov/pubmed/35935985
http://dx.doi.org/10.3389/fimmu.2022.830606
_version_ 1784763102781243392
author Li, Xuejiao
Du, Huan
Zhan, Shenghua
Liu, Wenting
Wang, Zhangyu
Lan, Jing
PuYang, Longxiang
Wan, Yuqiu
Qu, Qiuxia
Wang, Sining
Yang, Yang
Wang, Qin
Xie, Fang
author_facet Li, Xuejiao
Du, Huan
Zhan, Shenghua
Liu, Wenting
Wang, Zhangyu
Lan, Jing
PuYang, Longxiang
Wan, Yuqiu
Qu, Qiuxia
Wang, Sining
Yang, Yang
Wang, Qin
Xie, Fang
author_sort Li, Xuejiao
collection PubMed
description Accumulating evidence suggests that regulatory B cells (Bregs) play important roles in inhibiting the immune response in tumors. Programmed death 1 (PD-1) and programmed death ligand 1 (PD-L1) are important molecules that maintain the balance of the immune response and immune tolerance. This study aims to evaluate the soluble form of PD-L1 and its function in inducing the differentiation of B lymphocytes, investigate the relationship between soluble PD-L1 (sPD-L1) and B-cell subsets, and explore the antitumor activity of T lymphocytes after PD-L1 blockade in coculture systems. In an effort to explore the role of sPD-L1 in human breast cancer etiology, we examined the levels of sPD-L1 and interleukin-10 (IL-10) in the serum of breast tumor patients and the proportions of B cells, PD-1(+) B cells, Bregs, and PD-1(+) Bregs in the peripheral blood of patients with breast tumors and assessed their relationship among sPD-L1, IL-10, and B-cell subsets. The levels of sPD-L1 and IL-10 in serum were found to be significantly higher in invasive breast cancer (IBCa) patients than in breast fibroadenoma (FIBma) patients. Meanwhile, the proportions and absolute numbers of Bregs and PD-1(+) Bregs in the peripheral blood of IBCa patients were significantly higher than those of FIBma patients. Notably, they were the highest in triple-negative breast cancer (TNBC) among other subtypes of IBCa. Positive correlations of sPD-L1 and IL-10, IL-10 and PD-1(+) Bregs, and also sPD-L1 and PD-1(+) Bregs were observed in IBCa. We further demonstrated that sPD-L1 could induce Breg differentiation, IL-10 secretion, and IL-10 mRNA expression in a dose-dependent manner in vitro. Finally, the induction of regulatory T cells (T(regs)) by Bregs was further shown to suppress the antitumor response and that PD-L1 blockade therapies could promote the apoptosis of tumor cells. Together, these results indicated that sPD-L1 could mediate the differentiation of Bregs, expand CD4(+) T(regs) and weaken the antitumor activity of CD4(+) T cells. PD-L1/PD-1 blockade therapies might be a powerful therapeutic strategy for IBCa patients, particularly for TNBC patients with high level of PD-1(+) Bregs.
format Online
Article
Text
id pubmed-9354578
institution National Center for Biotechnology Information
language English
publishDate 2022
publisher Frontiers Media S.A.
record_format MEDLINE/PubMed
spelling pubmed-93545782022-08-06 The interaction between the soluble programmed death ligand-1 (sPD-L1) and PD-1(+) regulator B cells mediates immunosuppression in triple-negative breast cancer Li, Xuejiao Du, Huan Zhan, Shenghua Liu, Wenting Wang, Zhangyu Lan, Jing PuYang, Longxiang Wan, Yuqiu Qu, Qiuxia Wang, Sining Yang, Yang Wang, Qin Xie, Fang Front Immunol Immunology Accumulating evidence suggests that regulatory B cells (Bregs) play important roles in inhibiting the immune response in tumors. Programmed death 1 (PD-1) and programmed death ligand 1 (PD-L1) are important molecules that maintain the balance of the immune response and immune tolerance. This study aims to evaluate the soluble form of PD-L1 and its function in inducing the differentiation of B lymphocytes, investigate the relationship between soluble PD-L1 (sPD-L1) and B-cell subsets, and explore the antitumor activity of T lymphocytes after PD-L1 blockade in coculture systems. In an effort to explore the role of sPD-L1 in human breast cancer etiology, we examined the levels of sPD-L1 and interleukin-10 (IL-10) in the serum of breast tumor patients and the proportions of B cells, PD-1(+) B cells, Bregs, and PD-1(+) Bregs in the peripheral blood of patients with breast tumors and assessed their relationship among sPD-L1, IL-10, and B-cell subsets. The levels of sPD-L1 and IL-10 in serum were found to be significantly higher in invasive breast cancer (IBCa) patients than in breast fibroadenoma (FIBma) patients. Meanwhile, the proportions and absolute numbers of Bregs and PD-1(+) Bregs in the peripheral blood of IBCa patients were significantly higher than those of FIBma patients. Notably, they were the highest in triple-negative breast cancer (TNBC) among other subtypes of IBCa. Positive correlations of sPD-L1 and IL-10, IL-10 and PD-1(+) Bregs, and also sPD-L1 and PD-1(+) Bregs were observed in IBCa. We further demonstrated that sPD-L1 could induce Breg differentiation, IL-10 secretion, and IL-10 mRNA expression in a dose-dependent manner in vitro. Finally, the induction of regulatory T cells (T(regs)) by Bregs was further shown to suppress the antitumor response and that PD-L1 blockade therapies could promote the apoptosis of tumor cells. Together, these results indicated that sPD-L1 could mediate the differentiation of Bregs, expand CD4(+) T(regs) and weaken the antitumor activity of CD4(+) T cells. PD-L1/PD-1 blockade therapies might be a powerful therapeutic strategy for IBCa patients, particularly for TNBC patients with high level of PD-1(+) Bregs. Frontiers Media S.A. 2022-07-22 /pmc/articles/PMC9354578/ /pubmed/35935985 http://dx.doi.org/10.3389/fimmu.2022.830606 Text en Copyright © 2022 Li, Du, Zhan, Liu, Wang, Lan, PuYang, Wan, Qu, Wang, Yang, Wang and Xie https://creativecommons.org/licenses/by/4.0/This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.
spellingShingle Immunology
Li, Xuejiao
Du, Huan
Zhan, Shenghua
Liu, Wenting
Wang, Zhangyu
Lan, Jing
PuYang, Longxiang
Wan, Yuqiu
Qu, Qiuxia
Wang, Sining
Yang, Yang
Wang, Qin
Xie, Fang
The interaction between the soluble programmed death ligand-1 (sPD-L1) and PD-1(+) regulator B cells mediates immunosuppression in triple-negative breast cancer
title The interaction between the soluble programmed death ligand-1 (sPD-L1) and PD-1(+) regulator B cells mediates immunosuppression in triple-negative breast cancer
title_full The interaction between the soluble programmed death ligand-1 (sPD-L1) and PD-1(+) regulator B cells mediates immunosuppression in triple-negative breast cancer
title_fullStr The interaction between the soluble programmed death ligand-1 (sPD-L1) and PD-1(+) regulator B cells mediates immunosuppression in triple-negative breast cancer
title_full_unstemmed The interaction between the soluble programmed death ligand-1 (sPD-L1) and PD-1(+) regulator B cells mediates immunosuppression in triple-negative breast cancer
title_short The interaction between the soluble programmed death ligand-1 (sPD-L1) and PD-1(+) regulator B cells mediates immunosuppression in triple-negative breast cancer
title_sort interaction between the soluble programmed death ligand-1 (spd-l1) and pd-1(+) regulator b cells mediates immunosuppression in triple-negative breast cancer
topic Immunology
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9354578/
https://www.ncbi.nlm.nih.gov/pubmed/35935985
http://dx.doi.org/10.3389/fimmu.2022.830606
work_keys_str_mv AT lixuejiao theinteractionbetweenthesolubleprogrammeddeathligand1spdl1andpd1regulatorbcellsmediatesimmunosuppressionintriplenegativebreastcancer
AT duhuan theinteractionbetweenthesolubleprogrammeddeathligand1spdl1andpd1regulatorbcellsmediatesimmunosuppressionintriplenegativebreastcancer
AT zhanshenghua theinteractionbetweenthesolubleprogrammeddeathligand1spdl1andpd1regulatorbcellsmediatesimmunosuppressionintriplenegativebreastcancer
AT liuwenting theinteractionbetweenthesolubleprogrammeddeathligand1spdl1andpd1regulatorbcellsmediatesimmunosuppressionintriplenegativebreastcancer
AT wangzhangyu theinteractionbetweenthesolubleprogrammeddeathligand1spdl1andpd1regulatorbcellsmediatesimmunosuppressionintriplenegativebreastcancer
AT lanjing theinteractionbetweenthesolubleprogrammeddeathligand1spdl1andpd1regulatorbcellsmediatesimmunosuppressionintriplenegativebreastcancer
AT puyanglongxiang theinteractionbetweenthesolubleprogrammeddeathligand1spdl1andpd1regulatorbcellsmediatesimmunosuppressionintriplenegativebreastcancer
AT wanyuqiu theinteractionbetweenthesolubleprogrammeddeathligand1spdl1andpd1regulatorbcellsmediatesimmunosuppressionintriplenegativebreastcancer
AT quqiuxia theinteractionbetweenthesolubleprogrammeddeathligand1spdl1andpd1regulatorbcellsmediatesimmunosuppressionintriplenegativebreastcancer
AT wangsining theinteractionbetweenthesolubleprogrammeddeathligand1spdl1andpd1regulatorbcellsmediatesimmunosuppressionintriplenegativebreastcancer
AT yangyang theinteractionbetweenthesolubleprogrammeddeathligand1spdl1andpd1regulatorbcellsmediatesimmunosuppressionintriplenegativebreastcancer
AT wangqin theinteractionbetweenthesolubleprogrammeddeathligand1spdl1andpd1regulatorbcellsmediatesimmunosuppressionintriplenegativebreastcancer
AT xiefang theinteractionbetweenthesolubleprogrammeddeathligand1spdl1andpd1regulatorbcellsmediatesimmunosuppressionintriplenegativebreastcancer
AT lixuejiao interactionbetweenthesolubleprogrammeddeathligand1spdl1andpd1regulatorbcellsmediatesimmunosuppressionintriplenegativebreastcancer
AT duhuan interactionbetweenthesolubleprogrammeddeathligand1spdl1andpd1regulatorbcellsmediatesimmunosuppressionintriplenegativebreastcancer
AT zhanshenghua interactionbetweenthesolubleprogrammeddeathligand1spdl1andpd1regulatorbcellsmediatesimmunosuppressionintriplenegativebreastcancer
AT liuwenting interactionbetweenthesolubleprogrammeddeathligand1spdl1andpd1regulatorbcellsmediatesimmunosuppressionintriplenegativebreastcancer
AT wangzhangyu interactionbetweenthesolubleprogrammeddeathligand1spdl1andpd1regulatorbcellsmediatesimmunosuppressionintriplenegativebreastcancer
AT lanjing interactionbetweenthesolubleprogrammeddeathligand1spdl1andpd1regulatorbcellsmediatesimmunosuppressionintriplenegativebreastcancer
AT puyanglongxiang interactionbetweenthesolubleprogrammeddeathligand1spdl1andpd1regulatorbcellsmediatesimmunosuppressionintriplenegativebreastcancer
AT wanyuqiu interactionbetweenthesolubleprogrammeddeathligand1spdl1andpd1regulatorbcellsmediatesimmunosuppressionintriplenegativebreastcancer
AT quqiuxia interactionbetweenthesolubleprogrammeddeathligand1spdl1andpd1regulatorbcellsmediatesimmunosuppressionintriplenegativebreastcancer
AT wangsining interactionbetweenthesolubleprogrammeddeathligand1spdl1andpd1regulatorbcellsmediatesimmunosuppressionintriplenegativebreastcancer
AT yangyang interactionbetweenthesolubleprogrammeddeathligand1spdl1andpd1regulatorbcellsmediatesimmunosuppressionintriplenegativebreastcancer
AT wangqin interactionbetweenthesolubleprogrammeddeathligand1spdl1andpd1regulatorbcellsmediatesimmunosuppressionintriplenegativebreastcancer
AT xiefang interactionbetweenthesolubleprogrammeddeathligand1spdl1andpd1regulatorbcellsmediatesimmunosuppressionintriplenegativebreastcancer