Cargando…

Autocrine VEGF signalling on M2 macrophages regulates PD‐L1 expression for immunomodulation of T cells

M2‐polarized macrophages, on one hand, can promote tumour vascularization by producing proangiogenic factors, such as vascular endothelial growth factor (VEGF). On the other hand, the expression of VEGF receptors (VEGFR) in this cell lineage was also reported. Although the function of VEGF/VEGFR axi...

Descripción completa

Detalles Bibliográficos
Autores principales: Lai, Yin‐Siew, Wahyuningtyas, Rika, Aui, Shin‐Peir, Chang, Ko‐Tung
Formato: Online Artículo Texto
Lenguaje:English
Publicado: John Wiley and Sons Inc. 2018
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6349155/
https://www.ncbi.nlm.nih.gov/pubmed/30456891
http://dx.doi.org/10.1111/jcmm.14027
_version_ 1783390226032361472
author Lai, Yin‐Siew
Wahyuningtyas, Rika
Aui, Shin‐Peir
Chang, Ko‐Tung
author_facet Lai, Yin‐Siew
Wahyuningtyas, Rika
Aui, Shin‐Peir
Chang, Ko‐Tung
author_sort Lai, Yin‐Siew
collection PubMed
description M2‐polarized macrophages, on one hand, can promote tumour vascularization by producing proangiogenic factors, such as vascular endothelial growth factor (VEGF). On the other hand, the expression of VEGF receptors (VEGFR) in this cell lineage was also reported. Although the function of VEGF/VEGFR axis plays a pivotal role in macrophages infiltration and angiogenesis, however, there is still lack of the direct evidence to show the role of VEGF as an autocrine operating in M2 macrophages, particularly for immunomodulation. In our study, we surprisingly discovered that M2 macrophages polarized by baicalin can simultaneously express VEGF and its receptors. Taking advantage of this unique culture system, we were able to investigate the biological activity of M2 macrophages in response to the autocrine VEGF milieu. Our results showed that the expression of programmed death‐ligand 1 (PD‐L1) on M2 macrophages was significantly up‐regulated in autocrine VEGF milieu. Through the blockade of autocrine VEGF signalling, PD‐L1 expression on M2 macrophages was dramatically down‐regulated. Furthermore, transplantation of PD‐L1(+) M2 macrophage stimulated by autocrine VEGF into allogeneic mice significantly suppressed host CD4(+)/CD8(+) T cells in the peripheral blood and increased CD4(+) CD25(+) regulatory T cells in the bone marrow. In conclusion, our findings provide a novel biological basis to support the current successful strategy using combined VEGF/PD‐1 signalling blockade in cancer therapy.
format Online
Article
Text
id pubmed-6349155
institution National Center for Biotechnology Information
language English
publishDate 2018
publisher John Wiley and Sons Inc.
record_format MEDLINE/PubMed
spelling pubmed-63491552019-02-01 Autocrine VEGF signalling on M2 macrophages regulates PD‐L1 expression for immunomodulation of T cells Lai, Yin‐Siew Wahyuningtyas, Rika Aui, Shin‐Peir Chang, Ko‐Tung J Cell Mol Med Original Articles M2‐polarized macrophages, on one hand, can promote tumour vascularization by producing proangiogenic factors, such as vascular endothelial growth factor (VEGF). On the other hand, the expression of VEGF receptors (VEGFR) in this cell lineage was also reported. Although the function of VEGF/VEGFR axis plays a pivotal role in macrophages infiltration and angiogenesis, however, there is still lack of the direct evidence to show the role of VEGF as an autocrine operating in M2 macrophages, particularly for immunomodulation. In our study, we surprisingly discovered that M2 macrophages polarized by baicalin can simultaneously express VEGF and its receptors. Taking advantage of this unique culture system, we were able to investigate the biological activity of M2 macrophages in response to the autocrine VEGF milieu. Our results showed that the expression of programmed death‐ligand 1 (PD‐L1) on M2 macrophages was significantly up‐regulated in autocrine VEGF milieu. Through the blockade of autocrine VEGF signalling, PD‐L1 expression on M2 macrophages was dramatically down‐regulated. Furthermore, transplantation of PD‐L1(+) M2 macrophage stimulated by autocrine VEGF into allogeneic mice significantly suppressed host CD4(+)/CD8(+) T cells in the peripheral blood and increased CD4(+) CD25(+) regulatory T cells in the bone marrow. In conclusion, our findings provide a novel biological basis to support the current successful strategy using combined VEGF/PD‐1 signalling blockade in cancer therapy. John Wiley and Sons Inc. 2018-11-20 2019-02 /pmc/articles/PMC6349155/ /pubmed/30456891 http://dx.doi.org/10.1111/jcmm.14027 Text en © 2018 The Authors. Journal of Cellular and Molecular Medicine published by John Wiley & Sons Ltd and Foundation for Cellular and Molecular Medicine. This is an open access article under the terms of the http://creativecommons.org/licenses/by/4.0/ License, which permits use, distribution and reproduction in any medium, provided the original work is properly cited.
spellingShingle Original Articles
Lai, Yin‐Siew
Wahyuningtyas, Rika
Aui, Shin‐Peir
Chang, Ko‐Tung
Autocrine VEGF signalling on M2 macrophages regulates PD‐L1 expression for immunomodulation of T cells
title Autocrine VEGF signalling on M2 macrophages regulates PD‐L1 expression for immunomodulation of T cells
title_full Autocrine VEGF signalling on M2 macrophages regulates PD‐L1 expression for immunomodulation of T cells
title_fullStr Autocrine VEGF signalling on M2 macrophages regulates PD‐L1 expression for immunomodulation of T cells
title_full_unstemmed Autocrine VEGF signalling on M2 macrophages regulates PD‐L1 expression for immunomodulation of T cells
title_short Autocrine VEGF signalling on M2 macrophages regulates PD‐L1 expression for immunomodulation of T cells
title_sort autocrine vegf signalling on m2 macrophages regulates pd‐l1 expression for immunomodulation of t cells
topic Original Articles
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6349155/
https://www.ncbi.nlm.nih.gov/pubmed/30456891
http://dx.doi.org/10.1111/jcmm.14027
work_keys_str_mv AT laiyinsiew autocrinevegfsignallingonm2macrophagesregulatespdl1expressionforimmunomodulationoftcells
AT wahyuningtyasrika autocrinevegfsignallingonm2macrophagesregulatespdl1expressionforimmunomodulationoftcells
AT auishinpeir autocrinevegfsignallingonm2macrophagesregulatespdl1expressionforimmunomodulationoftcells
AT changkotung autocrinevegfsignallingonm2macrophagesregulatespdl1expressionforimmunomodulationoftcells