Nicotinamide N-methyltransferase inhibits autophagy induced by oxidative stress through suppressing the AMPK pathway in breast cancer cells

Abstract Background Nicotinamide N-methyltransferase (NNMT) is highly expressed in several cancers and can regulate cell epigenetic status and various cell metabolism pathways, such as ATP synthesis and cellular stress response. We reported in our previous papers that NNMT overexpression inhibits th...

Full description

Bibliographic Details
Main Authors: Haitao Yu, Xi Zhou, Yanzhong Wang, Xucheng Huang, Jun Yang, Jin Zeng, Guoli Li, Xinyou Xie, Jun Zhang
Format: Article
Language:English
Published: BMC 2020-05-01
Series:Cancer Cell International
Subjects:
Online Access:http://link.springer.com/article/10.1186/s12935-020-01279-8
id doaj-de7d94f71492482e83d9fb0b6490ee4b
record_format Article
spelling doaj-de7d94f71492482e83d9fb0b6490ee4b2020-11-25T03:46:13ZengBMCCancer Cell International1475-28672020-05-0120111110.1186/s12935-020-01279-8Nicotinamide N-methyltransferase inhibits autophagy induced by oxidative stress through suppressing the AMPK pathway in breast cancer cellsHaitao Yu0Xi Zhou1Yanzhong Wang2Xucheng Huang3Jun Yang4Jin Zeng5Guoli Li6Xinyou Xie7Jun Zhang8Department of Clinical Laboratory, Sir Run Run Shaw Hospital, Zhejiang University School of MedicineDepartment of Clinical Laboratory, Sir Run Run Shaw Hospital, Zhejiang University School of MedicineDepartment of Clinical Laboratory, Sir Run Run Shaw Hospital, Zhejiang University School of MedicineDepartment of Clinical Laboratory, Sir Run Run Shaw Hospital, Zhejiang University School of MedicineDepartment of Clinical Laboratory, Sir Run Run Shaw Hospital, Zhejiang University School of MedicineDepartment of Clinical Laboratory, Sir Run Run Shaw Hospital, Zhejiang University School of MedicineDepartment of Clinical Laboratory, Sir Run Run Shaw Hospital, Zhejiang University School of MedicineDepartment of Clinical Laboratory, Sir Run Run Shaw Hospital, Zhejiang University School of MedicineDepartment of Clinical Laboratory, Sir Run Run Shaw Hospital, Zhejiang University School of MedicineAbstract Background Nicotinamide N-methyltransferase (NNMT) is highly expressed in several cancers and can regulate cell epigenetic status and various cell metabolism pathways, such as ATP synthesis and cellular stress response. We reported in our previous papers that NNMT overexpression inhibits the apoptosis and enhances the chemotherapy resistance of breast cancer cells. This study aims to investigate the effect of NNMT on autophagy induced by oxidative stress in breast cancer cells, which might provide a novel therapeutic strategy for breast cancer treatment. Methods NNMT and LC3B II protein levels in the two cell models (SK-BR-3 and MDA-MB-231) with NNMT overexpression or knockdown were detected by Western blotting and correlated with each other. Changes in cellular viability, intracellular reactive oxygen species (ROS) and ATP levels were assessed after H2O2 treatment. Then, autophagosomes were imaged by transmission electron microscopy, and LC3 puncta were examined by confocal microscopy and flow cytometry. The LC3B II level and AMPK-ULK1 pathway activity were both detected by Western blotting to determine the role of NNMT in the H2O2-induced autophagy. Results NNMT expression was negatively correlated with LC3B II expression in both cell models (SK-BR-3 and MDA-MB-231). Then, NNMT overexpression attenuated the autophagy induced by H2O2 in SK-BR-3 cells, whereas knockdown promoted autophagy induced by H2O2 in MDA-MB-231 cells. Furthermore, mechanistic studies showed that NNMT suppressed the ROS increase, ATP decrease and AMPK-ULK1 pathway activation, resulting in the inhibition of H2O2-induced autophagy in breast cancer cells. Conclusions We conclude that NNMT inhibits the autophagy induced by oxidative stress through the ROS-mediated AMPK-ULK1 pathway in breast cancer cells and may protect breast cancer cells against oxidative stress through autophagy suppression.http://link.springer.com/article/10.1186/s12935-020-01279-8Nicotinamide N-methyltransferaseAutophagyOxidative stressAMPKULK1Breast cancer
collection DOAJ
language English
format Article
sources DOAJ
author Haitao Yu
Xi Zhou
Yanzhong Wang
Xucheng Huang
Jun Yang
Jin Zeng
Guoli Li
Xinyou Xie
Jun Zhang
spellingShingle Haitao Yu
Xi Zhou
Yanzhong Wang
Xucheng Huang
Jun Yang
Jin Zeng
Guoli Li
Xinyou Xie
Jun Zhang
Nicotinamide N-methyltransferase inhibits autophagy induced by oxidative stress through suppressing the AMPK pathway in breast cancer cells
Cancer Cell International
Nicotinamide N-methyltransferase
Autophagy
Oxidative stress
AMPK
ULK1
Breast cancer
author_facet Haitao Yu
Xi Zhou
Yanzhong Wang
Xucheng Huang
Jun Yang
Jin Zeng
Guoli Li
Xinyou Xie
Jun Zhang
author_sort Haitao Yu
title Nicotinamide N-methyltransferase inhibits autophagy induced by oxidative stress through suppressing the AMPK pathway in breast cancer cells
title_short Nicotinamide N-methyltransferase inhibits autophagy induced by oxidative stress through suppressing the AMPK pathway in breast cancer cells
title_full Nicotinamide N-methyltransferase inhibits autophagy induced by oxidative stress through suppressing the AMPK pathway in breast cancer cells
title_fullStr Nicotinamide N-methyltransferase inhibits autophagy induced by oxidative stress through suppressing the AMPK pathway in breast cancer cells
title_full_unstemmed Nicotinamide N-methyltransferase inhibits autophagy induced by oxidative stress through suppressing the AMPK pathway in breast cancer cells
title_sort nicotinamide n-methyltransferase inhibits autophagy induced by oxidative stress through suppressing the ampk pathway in breast cancer cells
publisher BMC
series Cancer Cell International
issn 1475-2867
publishDate 2020-05-01
description Abstract Background Nicotinamide N-methyltransferase (NNMT) is highly expressed in several cancers and can regulate cell epigenetic status and various cell metabolism pathways, such as ATP synthesis and cellular stress response. We reported in our previous papers that NNMT overexpression inhibits the apoptosis and enhances the chemotherapy resistance of breast cancer cells. This study aims to investigate the effect of NNMT on autophagy induced by oxidative stress in breast cancer cells, which might provide a novel therapeutic strategy for breast cancer treatment. Methods NNMT and LC3B II protein levels in the two cell models (SK-BR-3 and MDA-MB-231) with NNMT overexpression or knockdown were detected by Western blotting and correlated with each other. Changes in cellular viability, intracellular reactive oxygen species (ROS) and ATP levels were assessed after H2O2 treatment. Then, autophagosomes were imaged by transmission electron microscopy, and LC3 puncta were examined by confocal microscopy and flow cytometry. The LC3B II level and AMPK-ULK1 pathway activity were both detected by Western blotting to determine the role of NNMT in the H2O2-induced autophagy. Results NNMT expression was negatively correlated with LC3B II expression in both cell models (SK-BR-3 and MDA-MB-231). Then, NNMT overexpression attenuated the autophagy induced by H2O2 in SK-BR-3 cells, whereas knockdown promoted autophagy induced by H2O2 in MDA-MB-231 cells. Furthermore, mechanistic studies showed that NNMT suppressed the ROS increase, ATP decrease and AMPK-ULK1 pathway activation, resulting in the inhibition of H2O2-induced autophagy in breast cancer cells. Conclusions We conclude that NNMT inhibits the autophagy induced by oxidative stress through the ROS-mediated AMPK-ULK1 pathway in breast cancer cells and may protect breast cancer cells against oxidative stress through autophagy suppression.
topic Nicotinamide N-methyltransferase
Autophagy
Oxidative stress
AMPK
ULK1
Breast cancer
url http://link.springer.com/article/10.1186/s12935-020-01279-8
work_keys_str_mv AT haitaoyu nicotinamidenmethyltransferaseinhibitsautophagyinducedbyoxidativestressthroughsuppressingtheampkpathwayinbreastcancercells
AT xizhou nicotinamidenmethyltransferaseinhibitsautophagyinducedbyoxidativestressthroughsuppressingtheampkpathwayinbreastcancercells
AT yanzhongwang nicotinamidenmethyltransferaseinhibitsautophagyinducedbyoxidativestressthroughsuppressingtheampkpathwayinbreastcancercells
AT xuchenghuang nicotinamidenmethyltransferaseinhibitsautophagyinducedbyoxidativestressthroughsuppressingtheampkpathwayinbreastcancercells
AT junyang nicotinamidenmethyltransferaseinhibitsautophagyinducedbyoxidativestressthroughsuppressingtheampkpathwayinbreastcancercells
AT jinzeng nicotinamidenmethyltransferaseinhibitsautophagyinducedbyoxidativestressthroughsuppressingtheampkpathwayinbreastcancercells
AT guolili nicotinamidenmethyltransferaseinhibitsautophagyinducedbyoxidativestressthroughsuppressingtheampkpathwayinbreastcancercells
AT xinyouxie nicotinamidenmethyltransferaseinhibitsautophagyinducedbyoxidativestressthroughsuppressingtheampkpathwayinbreastcancercells
AT junzhang nicotinamidenmethyltransferaseinhibitsautophagyinducedbyoxidativestressthroughsuppressingtheampkpathwayinbreastcancercells
_version_ 1724507082972987392