Cargando…

KAT6A Acetylation of SMAD3 Regulates Myeloid‐Derived Suppressor Cell Recruitment, Metastasis, and Immunotherapy in Triple‐Negative Breast Cancer

Aberrant SMAD3 activation has been implicated as a driving event in cancer metastasis, yet the underlying mechanisms are still elusive. Here, SMAD3 is identified as a nonhistone substrate of lysine acetyltransferase 6A (KAT6A). The acetylation of SMAD3 at K20 and K117 by KAT6A promotes SMAD3 associa...

Descripción completa

Detalles Bibliográficos
Autores principales: Yu, Bo, Luo, Fei, Sun, Bowen, Liu, Wenxue, Shi, Qiqi, Cheng, Shi‐Yuan, Chen, Ceshi, Chen, Guoqiang, Li, Yanxin, Feng, Haizhong
Formato: Online Artículo Texto
Lenguaje:English
Publicado: John Wiley and Sons Inc. 2021
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8529494/
https://www.ncbi.nlm.nih.gov/pubmed/34392614
http://dx.doi.org/10.1002/advs.202100014
_version_ 1784586482397216768
author Yu, Bo
Luo, Fei
Sun, Bowen
Liu, Wenxue
Shi, Qiqi
Cheng, Shi‐Yuan
Chen, Ceshi
Chen, Guoqiang
Li, Yanxin
Feng, Haizhong
author_facet Yu, Bo
Luo, Fei
Sun, Bowen
Liu, Wenxue
Shi, Qiqi
Cheng, Shi‐Yuan
Chen, Ceshi
Chen, Guoqiang
Li, Yanxin
Feng, Haizhong
author_sort Yu, Bo
collection PubMed
description Aberrant SMAD3 activation has been implicated as a driving event in cancer metastasis, yet the underlying mechanisms are still elusive. Here, SMAD3 is identified as a nonhistone substrate of lysine acetyltransferase 6A (KAT6A). The acetylation of SMAD3 at K20 and K117 by KAT6A promotes SMAD3 association with oncogenic chromatin modifier tripartite motif‐containing 24 (TRIM24) and disrupts SMAD3 interaction with tumor suppressor TRIM33. This event in turn promotes KAT6A‐acetylated H3K23‐mediated recruitment of TRIM24–SMAD3 complex to chromatin and thereby increases SMAD3 activation and immune response‐related cytokine expression, leading to enhanced breast cancer stem‐like cell stemness, myeloid‐derived suppressor cell (MDSC) recruitment, and triple‐negative breast cancer (TNBC) metastasis. Inhibiting KAT6A in combination with anti‐PD‐L1 therapy in treating TNBC xenograft‐bearing animals markedly attenuates metastasis and provides a significant survival benefit. Thus, the work presents a KAT6A acetylation‐dependent regulatory mechanism governing SMAD3 oncogenic function and provides insight into how targeting an epigenetic factor with immunotherapies enhances the antimetastasis efficacy.
format Online
Article
Text
id pubmed-8529494
institution National Center for Biotechnology Information
language English
publishDate 2021
publisher John Wiley and Sons Inc.
record_format MEDLINE/PubMed
spelling pubmed-85294942021-10-27 KAT6A Acetylation of SMAD3 Regulates Myeloid‐Derived Suppressor Cell Recruitment, Metastasis, and Immunotherapy in Triple‐Negative Breast Cancer Yu, Bo Luo, Fei Sun, Bowen Liu, Wenxue Shi, Qiqi Cheng, Shi‐Yuan Chen, Ceshi Chen, Guoqiang Li, Yanxin Feng, Haizhong Adv Sci (Weinh) Research Articles Aberrant SMAD3 activation has been implicated as a driving event in cancer metastasis, yet the underlying mechanisms are still elusive. Here, SMAD3 is identified as a nonhistone substrate of lysine acetyltransferase 6A (KAT6A). The acetylation of SMAD3 at K20 and K117 by KAT6A promotes SMAD3 association with oncogenic chromatin modifier tripartite motif‐containing 24 (TRIM24) and disrupts SMAD3 interaction with tumor suppressor TRIM33. This event in turn promotes KAT6A‐acetylated H3K23‐mediated recruitment of TRIM24–SMAD3 complex to chromatin and thereby increases SMAD3 activation and immune response‐related cytokine expression, leading to enhanced breast cancer stem‐like cell stemness, myeloid‐derived suppressor cell (MDSC) recruitment, and triple‐negative breast cancer (TNBC) metastasis. Inhibiting KAT6A in combination with anti‐PD‐L1 therapy in treating TNBC xenograft‐bearing animals markedly attenuates metastasis and provides a significant survival benefit. Thus, the work presents a KAT6A acetylation‐dependent regulatory mechanism governing SMAD3 oncogenic function and provides insight into how targeting an epigenetic factor with immunotherapies enhances the antimetastasis efficacy. John Wiley and Sons Inc. 2021-08-13 /pmc/articles/PMC8529494/ /pubmed/34392614 http://dx.doi.org/10.1002/advs.202100014 Text en © 2021 The Authors. Advanced Science published by Wiley‐VCH GmbH https://creativecommons.org/licenses/by/4.0/This is an open access article under the terms of the http://creativecommons.org/licenses/by/4.0/ (https://creativecommons.org/licenses/by/4.0/) License, which permits use, distribution and reproduction in any medium, provided the original work is properly cited.
spellingShingle Research Articles
Yu, Bo
Luo, Fei
Sun, Bowen
Liu, Wenxue
Shi, Qiqi
Cheng, Shi‐Yuan
Chen, Ceshi
Chen, Guoqiang
Li, Yanxin
Feng, Haizhong
KAT6A Acetylation of SMAD3 Regulates Myeloid‐Derived Suppressor Cell Recruitment, Metastasis, and Immunotherapy in Triple‐Negative Breast Cancer
title KAT6A Acetylation of SMAD3 Regulates Myeloid‐Derived Suppressor Cell Recruitment, Metastasis, and Immunotherapy in Triple‐Negative Breast Cancer
title_full KAT6A Acetylation of SMAD3 Regulates Myeloid‐Derived Suppressor Cell Recruitment, Metastasis, and Immunotherapy in Triple‐Negative Breast Cancer
title_fullStr KAT6A Acetylation of SMAD3 Regulates Myeloid‐Derived Suppressor Cell Recruitment, Metastasis, and Immunotherapy in Triple‐Negative Breast Cancer
title_full_unstemmed KAT6A Acetylation of SMAD3 Regulates Myeloid‐Derived Suppressor Cell Recruitment, Metastasis, and Immunotherapy in Triple‐Negative Breast Cancer
title_short KAT6A Acetylation of SMAD3 Regulates Myeloid‐Derived Suppressor Cell Recruitment, Metastasis, and Immunotherapy in Triple‐Negative Breast Cancer
title_sort kat6a acetylation of smad3 regulates myeloid‐derived suppressor cell recruitment, metastasis, and immunotherapy in triple‐negative breast cancer
topic Research Articles
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8529494/
https://www.ncbi.nlm.nih.gov/pubmed/34392614
http://dx.doi.org/10.1002/advs.202100014
work_keys_str_mv AT yubo kat6aacetylationofsmad3regulatesmyeloidderivedsuppressorcellrecruitmentmetastasisandimmunotherapyintriplenegativebreastcancer
AT luofei kat6aacetylationofsmad3regulatesmyeloidderivedsuppressorcellrecruitmentmetastasisandimmunotherapyintriplenegativebreastcancer
AT sunbowen kat6aacetylationofsmad3regulatesmyeloidderivedsuppressorcellrecruitmentmetastasisandimmunotherapyintriplenegativebreastcancer
AT liuwenxue kat6aacetylationofsmad3regulatesmyeloidderivedsuppressorcellrecruitmentmetastasisandimmunotherapyintriplenegativebreastcancer
AT shiqiqi kat6aacetylationofsmad3regulatesmyeloidderivedsuppressorcellrecruitmentmetastasisandimmunotherapyintriplenegativebreastcancer
AT chengshiyuan kat6aacetylationofsmad3regulatesmyeloidderivedsuppressorcellrecruitmentmetastasisandimmunotherapyintriplenegativebreastcancer
AT chenceshi kat6aacetylationofsmad3regulatesmyeloidderivedsuppressorcellrecruitmentmetastasisandimmunotherapyintriplenegativebreastcancer
AT chenguoqiang kat6aacetylationofsmad3regulatesmyeloidderivedsuppressorcellrecruitmentmetastasisandimmunotherapyintriplenegativebreastcancer
AT liyanxin kat6aacetylationofsmad3regulatesmyeloidderivedsuppressorcellrecruitmentmetastasisandimmunotherapyintriplenegativebreastcancer
AT fenghaizhong kat6aacetylationofsmad3regulatesmyeloidderivedsuppressorcellrecruitmentmetastasisandimmunotherapyintriplenegativebreastcancer