Murine myeloid dendritic cells that phagocytose apoptotic T cells inhibit the immune response via NO.

The contraction phase of antigen-specific immune responses involves the apoptotic loss of numerous activated lymphocytes. While apoptotic cells are known to induce immune suppression, the mechanisms involved therein are still ambiguous. Some reports have speculated that macrophages can induce regula...

Full description

Bibliographic Details
Main Authors: Kaili Zhong, Wengang Song, Qian Wang, Chao Wang, Xi Liu, Dongwei Chen, Zhongli Zhu, Yiqing Wu, Weijing Zhang, Minghui Zhang
Format: Article
Language:English
Published: Public Library of Science (PLoS) 2012-01-01
Series:PLoS ONE
Online Access:http://europepmc.org/articles/PMC3499560?pdf=render
id doaj-ac64b7893af24959b216711811231913
record_format Article
spelling doaj-ac64b7893af24959b2167118112319132020-11-25T01:17:19ZengPublic Library of Science (PLoS)PLoS ONE1932-62032012-01-01711e4937810.1371/journal.pone.0049378Murine myeloid dendritic cells that phagocytose apoptotic T cells inhibit the immune response via NO.Kaili ZhongWengang SongQian WangChao WangXi LiuDongwei ChenZhongli ZhuYiqing WuWeijing ZhangMinghui ZhangThe contraction phase of antigen-specific immune responses involves the apoptotic loss of numerous activated lymphocytes. While apoptotic cells are known to induce immune suppression, the mechanisms involved therein are still ambiguous. Some reports have speculated that macrophages can induce regulatory T cells (Tregs) after engulfing apoptotic cells. In this study, we showed that dendritic cells (DCs) that phagocytose apoptotic T cells acquire inhibitory function (named DCapos) toward CD4(+) and CD8(+) T cells. These inhibitory DCs could not induce the generation of Tregs, but they were found to directly inhibit mDCs that initiate CD4(+) and CD8(+) T cell proliferation both in vitro and in vivo. Soluble factors including NO play a role in the DCapos-induced suppression of CD4(+) and CD8(+) T cell proliferation. Further results showed that STAT3 phosphorylation and inducible nitric oxide synthase (iNOS) generation were enhanced when DCs were co-cultured with apoptotic cells. Both iNOS transcription and NO secretion were inhibited in the presence of the specific p-STAT3 inhibitor JSI-124. All the data indicated that apoptotic cells could turn DCs to inhibitory DCs, which might play important roles in the suppression of immune responses. STAT3 activation and the consequent release of NO are responsible for the inhibitory functions of DCapos.http://europepmc.org/articles/PMC3499560?pdf=render
collection DOAJ
language English
format Article
sources DOAJ
author Kaili Zhong
Wengang Song
Qian Wang
Chao Wang
Xi Liu
Dongwei Chen
Zhongli Zhu
Yiqing Wu
Weijing Zhang
Minghui Zhang
spellingShingle Kaili Zhong
Wengang Song
Qian Wang
Chao Wang
Xi Liu
Dongwei Chen
Zhongli Zhu
Yiqing Wu
Weijing Zhang
Minghui Zhang
Murine myeloid dendritic cells that phagocytose apoptotic T cells inhibit the immune response via NO.
PLoS ONE
author_facet Kaili Zhong
Wengang Song
Qian Wang
Chao Wang
Xi Liu
Dongwei Chen
Zhongli Zhu
Yiqing Wu
Weijing Zhang
Minghui Zhang
author_sort Kaili Zhong
title Murine myeloid dendritic cells that phagocytose apoptotic T cells inhibit the immune response via NO.
title_short Murine myeloid dendritic cells that phagocytose apoptotic T cells inhibit the immune response via NO.
title_full Murine myeloid dendritic cells that phagocytose apoptotic T cells inhibit the immune response via NO.
title_fullStr Murine myeloid dendritic cells that phagocytose apoptotic T cells inhibit the immune response via NO.
title_full_unstemmed Murine myeloid dendritic cells that phagocytose apoptotic T cells inhibit the immune response via NO.
title_sort murine myeloid dendritic cells that phagocytose apoptotic t cells inhibit the immune response via no.
publisher Public Library of Science (PLoS)
series PLoS ONE
issn 1932-6203
publishDate 2012-01-01
description The contraction phase of antigen-specific immune responses involves the apoptotic loss of numerous activated lymphocytes. While apoptotic cells are known to induce immune suppression, the mechanisms involved therein are still ambiguous. Some reports have speculated that macrophages can induce regulatory T cells (Tregs) after engulfing apoptotic cells. In this study, we showed that dendritic cells (DCs) that phagocytose apoptotic T cells acquire inhibitory function (named DCapos) toward CD4(+) and CD8(+) T cells. These inhibitory DCs could not induce the generation of Tregs, but they were found to directly inhibit mDCs that initiate CD4(+) and CD8(+) T cell proliferation both in vitro and in vivo. Soluble factors including NO play a role in the DCapos-induced suppression of CD4(+) and CD8(+) T cell proliferation. Further results showed that STAT3 phosphorylation and inducible nitric oxide synthase (iNOS) generation were enhanced when DCs were co-cultured with apoptotic cells. Both iNOS transcription and NO secretion were inhibited in the presence of the specific p-STAT3 inhibitor JSI-124. All the data indicated that apoptotic cells could turn DCs to inhibitory DCs, which might play important roles in the suppression of immune responses. STAT3 activation and the consequent release of NO are responsible for the inhibitory functions of DCapos.
url http://europepmc.org/articles/PMC3499560?pdf=render
work_keys_str_mv AT kailizhong murinemyeloiddendriticcellsthatphagocytoseapoptotictcellsinhibittheimmuneresponseviano
AT wengangsong murinemyeloiddendriticcellsthatphagocytoseapoptotictcellsinhibittheimmuneresponseviano
AT qianwang murinemyeloiddendriticcellsthatphagocytoseapoptotictcellsinhibittheimmuneresponseviano
AT chaowang murinemyeloiddendriticcellsthatphagocytoseapoptotictcellsinhibittheimmuneresponseviano
AT xiliu murinemyeloiddendriticcellsthatphagocytoseapoptotictcellsinhibittheimmuneresponseviano
AT dongweichen murinemyeloiddendriticcellsthatphagocytoseapoptotictcellsinhibittheimmuneresponseviano
AT zhonglizhu murinemyeloiddendriticcellsthatphagocytoseapoptotictcellsinhibittheimmuneresponseviano
AT yiqingwu murinemyeloiddendriticcellsthatphagocytoseapoptotictcellsinhibittheimmuneresponseviano
AT weijingzhang murinemyeloiddendriticcellsthatphagocytoseapoptotictcellsinhibittheimmuneresponseviano
AT minghuizhang murinemyeloiddendriticcellsthatphagocytoseapoptotictcellsinhibittheimmuneresponseviano
_version_ 1725146587925053440