Phosphorylation of PHF2 by AMPK releases the repressive H3K9me2 and inhibits cancer metastasis

Abstract Epithelial to mesenchymal transition (EMT) plays a crucial role in cancer metastasis, accompanied with vast epigenetic changes. AMP-activated protein kinase (AMPK), a cellular energy sensor, plays regulatory roles in multiple biological processes. Although a few studies have shed light on A...

Full description

Bibliographic Details
Main Authors: Ying Dong, Hao Hu, Xuan Zhang, Yunkai Zhang, Xin Sun, Hanlin Wang, Weijuan Kan, Min-jia Tan, Hong Shi, Yi Zang, Jia Li
Format: Article
Language:English
Published: Nature Publishing Group 2023-03-01
Series:Signal Transduction and Targeted Therapy
Online Access:https://doi.org/10.1038/s41392-022-01302-6
_version_ 1797863417069961216
author Ying Dong
Hao Hu
Xuan Zhang
Yunkai Zhang
Xin Sun
Hanlin Wang
Weijuan Kan
Min-jia Tan
Hong Shi
Yi Zang
Jia Li
author_facet Ying Dong
Hao Hu
Xuan Zhang
Yunkai Zhang
Xin Sun
Hanlin Wang
Weijuan Kan
Min-jia Tan
Hong Shi
Yi Zang
Jia Li
author_sort Ying Dong
collection DOAJ
description Abstract Epithelial to mesenchymal transition (EMT) plays a crucial role in cancer metastasis, accompanied with vast epigenetic changes. AMP-activated protein kinase (AMPK), a cellular energy sensor, plays regulatory roles in multiple biological processes. Although a few studies have shed light on AMPK regulating cancer metastasis, the inside epigenetic mechanisms remain unknown. Herein we show that AMPK activation by metformin relieves the repressive H3K9me2-mediated silencing of epithelial genes (e.g., CDH1) during EMT processes and inhibits lung cancer metastasis. PHF2, a H3K9me2 demethylase, was identified to interact with AMPKα2. Genetic deletion of PHF2 aggravates lung cancer metastasis and abolishes the H3K9me2 downregulation and anti-metastasis effect of metformin. Mechanistically, AMPK phosphorylates PHF2 at S655 site, enhancing PHF2 demethylation activity and triggering the transcription of CDH1. Furthermore, the PHF2-S655E mutant that mimics AMPK-mediated phosphorylation status further reduces H3K9me2 and suppresses lung cancer metastasis, while PHF2-S655A mutant presents opposite phenotype and reverses the anti-metastasis effect of metformin. PHF2-S655 phosphorylation strikingly reduces in lung cancer patients and the higher phosphorylation level predicts better survival. Altogether, we reveal the mechanism of AMPK inhibiting lung cancer metastasis via PHF2 mediated H3K9me2 demethylation, thereby promoting the clinical application of metformin and highlighting PHF2 as the potential epigenetic target in cancer metastasis.
first_indexed 2024-04-09T22:35:16Z
format Article
id doaj.art-a488fccc803548f08f462353d2ebe4ac
institution Directory Open Access Journal
issn 2059-3635
language English
last_indexed 2024-04-09T22:35:16Z
publishDate 2023-03-01
publisher Nature Publishing Group
record_format Article
series Signal Transduction and Targeted Therapy
spelling doaj.art-a488fccc803548f08f462353d2ebe4ac2023-03-22T12:31:12ZengNature Publishing GroupSignal Transduction and Targeted Therapy2059-36352023-03-018111710.1038/s41392-022-01302-6Phosphorylation of PHF2 by AMPK releases the repressive H3K9me2 and inhibits cancer metastasisYing Dong0Hao Hu1Xuan Zhang2Yunkai Zhang3Xin Sun4Hanlin Wang5Weijuan Kan6Min-jia Tan7Hong Shi8Yi Zang9Jia Li10State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of SciencesState Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of SciencesState Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of SciencesState Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of SciencesState Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of SciencesState Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of SciencesState Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of SciencesState Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of SciencesDepartment of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji UniversityState Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of SciencesState Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of SciencesAbstract Epithelial to mesenchymal transition (EMT) plays a crucial role in cancer metastasis, accompanied with vast epigenetic changes. AMP-activated protein kinase (AMPK), a cellular energy sensor, plays regulatory roles in multiple biological processes. Although a few studies have shed light on AMPK regulating cancer metastasis, the inside epigenetic mechanisms remain unknown. Herein we show that AMPK activation by metformin relieves the repressive H3K9me2-mediated silencing of epithelial genes (e.g., CDH1) during EMT processes and inhibits lung cancer metastasis. PHF2, a H3K9me2 demethylase, was identified to interact with AMPKα2. Genetic deletion of PHF2 aggravates lung cancer metastasis and abolishes the H3K9me2 downregulation and anti-metastasis effect of metformin. Mechanistically, AMPK phosphorylates PHF2 at S655 site, enhancing PHF2 demethylation activity and triggering the transcription of CDH1. Furthermore, the PHF2-S655E mutant that mimics AMPK-mediated phosphorylation status further reduces H3K9me2 and suppresses lung cancer metastasis, while PHF2-S655A mutant presents opposite phenotype and reverses the anti-metastasis effect of metformin. PHF2-S655 phosphorylation strikingly reduces in lung cancer patients and the higher phosphorylation level predicts better survival. Altogether, we reveal the mechanism of AMPK inhibiting lung cancer metastasis via PHF2 mediated H3K9me2 demethylation, thereby promoting the clinical application of metformin and highlighting PHF2 as the potential epigenetic target in cancer metastasis.https://doi.org/10.1038/s41392-022-01302-6
spellingShingle Ying Dong
Hao Hu
Xuan Zhang
Yunkai Zhang
Xin Sun
Hanlin Wang
Weijuan Kan
Min-jia Tan
Hong Shi
Yi Zang
Jia Li
Phosphorylation of PHF2 by AMPK releases the repressive H3K9me2 and inhibits cancer metastasis
Signal Transduction and Targeted Therapy
title Phosphorylation of PHF2 by AMPK releases the repressive H3K9me2 and inhibits cancer metastasis
title_full Phosphorylation of PHF2 by AMPK releases the repressive H3K9me2 and inhibits cancer metastasis
title_fullStr Phosphorylation of PHF2 by AMPK releases the repressive H3K9me2 and inhibits cancer metastasis
title_full_unstemmed Phosphorylation of PHF2 by AMPK releases the repressive H3K9me2 and inhibits cancer metastasis
title_short Phosphorylation of PHF2 by AMPK releases the repressive H3K9me2 and inhibits cancer metastasis
title_sort phosphorylation of phf2 by ampk releases the repressive h3k9me2 and inhibits cancer metastasis
url https://doi.org/10.1038/s41392-022-01302-6
work_keys_str_mv AT yingdong phosphorylationofphf2byampkreleasestherepressiveh3k9me2andinhibitscancermetastasis
AT haohu phosphorylationofphf2byampkreleasestherepressiveh3k9me2andinhibitscancermetastasis
AT xuanzhang phosphorylationofphf2byampkreleasestherepressiveh3k9me2andinhibitscancermetastasis
AT yunkaizhang phosphorylationofphf2byampkreleasestherepressiveh3k9me2andinhibitscancermetastasis
AT xinsun phosphorylationofphf2byampkreleasestherepressiveh3k9me2andinhibitscancermetastasis
AT hanlinwang phosphorylationofphf2byampkreleasestherepressiveh3k9me2andinhibitscancermetastasis
AT weijuankan phosphorylationofphf2byampkreleasestherepressiveh3k9me2andinhibitscancermetastasis
AT minjiatan phosphorylationofphf2byampkreleasestherepressiveh3k9me2andinhibitscancermetastasis
AT hongshi phosphorylationofphf2byampkreleasestherepressiveh3k9me2andinhibitscancermetastasis
AT yizang phosphorylationofphf2byampkreleasestherepressiveh3k9me2andinhibitscancermetastasis
AT jiali phosphorylationofphf2byampkreleasestherepressiveh3k9me2andinhibitscancermetastasis