Cargando…

Histone deacetylase 2 knockout suppresses immune escape of triple-negative breast cancer cells via downregulating PD-L1 expression

The PD-L1 overexpression is an important event of immune escape and metastasis in triple-negative breast cancer (TNBC), but the molecular mechanism remains to be determined. Interferon gamma (IFNγ) represents a major driving force behind PD-L1 expression in tumor microenvironment, and histone deacet...

Descripción completa

Detalles Bibliográficos
Autores principales: Xu, Pengfei, Xiong, Wei, Lin, Yun, Fan, Liping, Pan, Hongchao, Li, Yaochen
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Nature Publishing Group UK 2021
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8349356/
https://www.ncbi.nlm.nih.gov/pubmed/34365463
http://dx.doi.org/10.1038/s41419-021-04047-2
_version_ 1783735551611895808
author Xu, Pengfei
Xiong, Wei
Lin, Yun
Fan, Liping
Pan, Hongchao
Li, Yaochen
author_facet Xu, Pengfei
Xiong, Wei
Lin, Yun
Fan, Liping
Pan, Hongchao
Li, Yaochen
author_sort Xu, Pengfei
collection PubMed
description The PD-L1 overexpression is an important event of immune escape and metastasis in triple-negative breast cancer (TNBC), but the molecular mechanism remains to be determined. Interferon gamma (IFNγ) represents a major driving force behind PD-L1 expression in tumor microenvironment, and histone deacetylase 2 (HDAC2) is required for IFN signaling. Here, we investigated the regulation of HDAC2 on the IFNγ-induced PD-L1 expression in TNBC cells. We found the HDAC2 and PD-L1 expression in TNBC was significantly higher than that in non-TNBC, and HDAC2 was positively correlated with PD-L1 expression. HDAC2 promoted PD-L1 induction by upregulating the phosphorylation of JAK1, JAK2, and STAT1, as well as the translocation of STAT1 to the nucleus and the recruitment of STAT1 to the PD-L1 promoter. Meanwhile, HDAC2 was recruited to the PD-L1 promoter by STAT1, and HDAC2 knockout compromised IFNγ-induced upregulation of H3K27, H3K9 acetylation, and the BRD4 recruitment in PD-L1 promoter. In addition, significant inhibition of proliferation, colony formation, migration, and cell cycle of TNBC cells were observed following knockout of HDAC2 in vitro. Furthermore, HDAC2 knockout reduced IFNγ-induced PD-L1 expression, lymphocyte infiltration, and retarded tumor growth and metastasis in the breast cancer mouse models. This study may provide evidence that HDAC2 promotes IFNγ-induced PD-L1 expression, suggesting a way for enhanced antitumor immunity when targeting the HDAC2 in TNBC.
format Online
Article
Text
id pubmed-8349356
institution National Center for Biotechnology Information
language English
publishDate 2021
publisher Nature Publishing Group UK
record_format MEDLINE/PubMed
spelling pubmed-83493562021-08-13 Histone deacetylase 2 knockout suppresses immune escape of triple-negative breast cancer cells via downregulating PD-L1 expression Xu, Pengfei Xiong, Wei Lin, Yun Fan, Liping Pan, Hongchao Li, Yaochen Cell Death Dis Article The PD-L1 overexpression is an important event of immune escape and metastasis in triple-negative breast cancer (TNBC), but the molecular mechanism remains to be determined. Interferon gamma (IFNγ) represents a major driving force behind PD-L1 expression in tumor microenvironment, and histone deacetylase 2 (HDAC2) is required for IFN signaling. Here, we investigated the regulation of HDAC2 on the IFNγ-induced PD-L1 expression in TNBC cells. We found the HDAC2 and PD-L1 expression in TNBC was significantly higher than that in non-TNBC, and HDAC2 was positively correlated with PD-L1 expression. HDAC2 promoted PD-L1 induction by upregulating the phosphorylation of JAK1, JAK2, and STAT1, as well as the translocation of STAT1 to the nucleus and the recruitment of STAT1 to the PD-L1 promoter. Meanwhile, HDAC2 was recruited to the PD-L1 promoter by STAT1, and HDAC2 knockout compromised IFNγ-induced upregulation of H3K27, H3K9 acetylation, and the BRD4 recruitment in PD-L1 promoter. In addition, significant inhibition of proliferation, colony formation, migration, and cell cycle of TNBC cells were observed following knockout of HDAC2 in vitro. Furthermore, HDAC2 knockout reduced IFNγ-induced PD-L1 expression, lymphocyte infiltration, and retarded tumor growth and metastasis in the breast cancer mouse models. This study may provide evidence that HDAC2 promotes IFNγ-induced PD-L1 expression, suggesting a way for enhanced antitumor immunity when targeting the HDAC2 in TNBC. Nature Publishing Group UK 2021-08-07 /pmc/articles/PMC8349356/ /pubmed/34365463 http://dx.doi.org/10.1038/s41419-021-04047-2 Text en © The Author(s) 2021 https://creativecommons.org/licenses/by/4.0/Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons license, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons license and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this license, visit http://creativecommons.org/licenses/by/4.0/ (https://creativecommons.org/licenses/by/4.0/) .
spellingShingle Article
Xu, Pengfei
Xiong, Wei
Lin, Yun
Fan, Liping
Pan, Hongchao
Li, Yaochen
Histone deacetylase 2 knockout suppresses immune escape of triple-negative breast cancer cells via downregulating PD-L1 expression
title Histone deacetylase 2 knockout suppresses immune escape of triple-negative breast cancer cells via downregulating PD-L1 expression
title_full Histone deacetylase 2 knockout suppresses immune escape of triple-negative breast cancer cells via downregulating PD-L1 expression
title_fullStr Histone deacetylase 2 knockout suppresses immune escape of triple-negative breast cancer cells via downregulating PD-L1 expression
title_full_unstemmed Histone deacetylase 2 knockout suppresses immune escape of triple-negative breast cancer cells via downregulating PD-L1 expression
title_short Histone deacetylase 2 knockout suppresses immune escape of triple-negative breast cancer cells via downregulating PD-L1 expression
title_sort histone deacetylase 2 knockout suppresses immune escape of triple-negative breast cancer cells via downregulating pd-l1 expression
topic Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8349356/
https://www.ncbi.nlm.nih.gov/pubmed/34365463
http://dx.doi.org/10.1038/s41419-021-04047-2
work_keys_str_mv AT xupengfei histonedeacetylase2knockoutsuppressesimmuneescapeoftriplenegativebreastcancercellsviadownregulatingpdl1expression
AT xiongwei histonedeacetylase2knockoutsuppressesimmuneescapeoftriplenegativebreastcancercellsviadownregulatingpdl1expression
AT linyun histonedeacetylase2knockoutsuppressesimmuneescapeoftriplenegativebreastcancercellsviadownregulatingpdl1expression
AT fanliping histonedeacetylase2knockoutsuppressesimmuneescapeoftriplenegativebreastcancercellsviadownregulatingpdl1expression
AT panhongchao histonedeacetylase2knockoutsuppressesimmuneescapeoftriplenegativebreastcancercellsviadownregulatingpdl1expression
AT liyaochen histonedeacetylase2knockoutsuppressesimmuneescapeoftriplenegativebreastcancercellsviadownregulatingpdl1expression