Anthocyanin Oligomers Induce Apoptosis and Autophagy by Inhibiting the mTOR Signaling Pathway in Human Breast Cancer Cells

Anthocyanin oligomers (AOs) are phytochemicals synthesized by fermenting anthocyanins extracted from grape skins and are more biologically active than monomeric anthocyanins. In this study, we evaluate the effects of an AO on triple-negative MDA-MB-231 and HER2-overexpressing SK-BR-3 breast cancer c...

Full description

Bibliographic Details
Main Authors: Min-Gu Lee, Hyun-Jin Hong, Kyung-Soo Nam
Format: Article
Language:English
Published: MDPI AG 2023-12-01
Series:Pharmaceuticals
Subjects:
Online Access:https://www.mdpi.com/1424-8247/17/1/24
_version_ 1797342774723346432
author Min-Gu Lee
Hyun-Jin Hong
Kyung-Soo Nam
author_facet Min-Gu Lee
Hyun-Jin Hong
Kyung-Soo Nam
author_sort Min-Gu Lee
collection DOAJ
description Anthocyanin oligomers (AOs) are phytochemicals synthesized by fermenting anthocyanins extracted from grape skins and are more biologically active than monomeric anthocyanins. In this study, we evaluate the effects of an AO on triple-negative MDA-MB-231 and HER2-overexpressing SK-BR-3 breast cancer cells. The cell viability of MDA-MB-231 and SK-BR-3 cells was significantly inhibited in a concentration-dependent manner by AO treatment for 24 h, while delphinidin (a monomeric anthocyanin) had no effect on cell viability. In addition, the AO increased H2A.X phosphorylation (a marker of DNA damage), reduced RAD51 (a DNA repair protein) and survivin (a cell survival factor) protein levels, and induced apoptosis by caspase-3-dependent PARP1 cleavage in both cell lines. Surprisingly, the AO induced autophagy by increasing intracellular LC3-II puncta and LC3-II and p62 protein levels. In addition, the AO inhibited the mTOR pathway in MDA-MB-231 and SK-BR-3 cells by suppressing the HER2, EGFR1, and AKT pathways. These results demonstrate that the anti-cancer effect of the AO was due to the induction of apoptosis and autophagy via cleaved caspase-3-mediated PARP1 cleavage and mTOR pathway inhibition, respectively. Furthermore, our results suggest that anthocyanin oligomers could be considered potential candidates for breast cancer treatment.
first_indexed 2024-03-08T10:38:08Z
format Article
id doaj.art-1ec043678c3d402e952de8985315e2e2
institution Directory Open Access Journal
issn 1424-8247
language English
last_indexed 2024-03-08T10:38:08Z
publishDate 2023-12-01
publisher MDPI AG
record_format Article
series Pharmaceuticals
spelling doaj.art-1ec043678c3d402e952de8985315e2e22024-01-26T18:04:55ZengMDPI AGPharmaceuticals1424-82472023-12-011712410.3390/ph17010024Anthocyanin Oligomers Induce Apoptosis and Autophagy by Inhibiting the mTOR Signaling Pathway in Human Breast Cancer CellsMin-Gu Lee0Hyun-Jin Hong1Kyung-Soo Nam2Department of Pharmacology and Intractable Disease Research Center, School of Medicine, Dongguk University, Gyeongju 38066, Republic of KoreaDepartment of Pharmacology and Intractable Disease Research Center, School of Medicine, Dongguk University, Gyeongju 38066, Republic of KoreaDepartment of Pharmacology and Intractable Disease Research Center, School of Medicine, Dongguk University, Gyeongju 38066, Republic of KoreaAnthocyanin oligomers (AOs) are phytochemicals synthesized by fermenting anthocyanins extracted from grape skins and are more biologically active than monomeric anthocyanins. In this study, we evaluate the effects of an AO on triple-negative MDA-MB-231 and HER2-overexpressing SK-BR-3 breast cancer cells. The cell viability of MDA-MB-231 and SK-BR-3 cells was significantly inhibited in a concentration-dependent manner by AO treatment for 24 h, while delphinidin (a monomeric anthocyanin) had no effect on cell viability. In addition, the AO increased H2A.X phosphorylation (a marker of DNA damage), reduced RAD51 (a DNA repair protein) and survivin (a cell survival factor) protein levels, and induced apoptosis by caspase-3-dependent PARP1 cleavage in both cell lines. Surprisingly, the AO induced autophagy by increasing intracellular LC3-II puncta and LC3-II and p62 protein levels. In addition, the AO inhibited the mTOR pathway in MDA-MB-231 and SK-BR-3 cells by suppressing the HER2, EGFR1, and AKT pathways. These results demonstrate that the anti-cancer effect of the AO was due to the induction of apoptosis and autophagy via cleaved caspase-3-mediated PARP1 cleavage and mTOR pathway inhibition, respectively. Furthermore, our results suggest that anthocyanin oligomers could be considered potential candidates for breast cancer treatment.https://www.mdpi.com/1424-8247/17/1/24anthocyanin oligomersbreast cancer cellsapoptosisautophagy
spellingShingle Min-Gu Lee
Hyun-Jin Hong
Kyung-Soo Nam
Anthocyanin Oligomers Induce Apoptosis and Autophagy by Inhibiting the mTOR Signaling Pathway in Human Breast Cancer Cells
Pharmaceuticals
anthocyanin oligomers
breast cancer cells
apoptosis
autophagy
title Anthocyanin Oligomers Induce Apoptosis and Autophagy by Inhibiting the mTOR Signaling Pathway in Human Breast Cancer Cells
title_full Anthocyanin Oligomers Induce Apoptosis and Autophagy by Inhibiting the mTOR Signaling Pathway in Human Breast Cancer Cells
title_fullStr Anthocyanin Oligomers Induce Apoptosis and Autophagy by Inhibiting the mTOR Signaling Pathway in Human Breast Cancer Cells
title_full_unstemmed Anthocyanin Oligomers Induce Apoptosis and Autophagy by Inhibiting the mTOR Signaling Pathway in Human Breast Cancer Cells
title_short Anthocyanin Oligomers Induce Apoptosis and Autophagy by Inhibiting the mTOR Signaling Pathway in Human Breast Cancer Cells
title_sort anthocyanin oligomers induce apoptosis and autophagy by inhibiting the mtor signaling pathway in human breast cancer cells
topic anthocyanin oligomers
breast cancer cells
apoptosis
autophagy
url https://www.mdpi.com/1424-8247/17/1/24
work_keys_str_mv AT mingulee anthocyaninoligomersinduceapoptosisandautophagybyinhibitingthemtorsignalingpathwayinhumanbreastcancercells
AT hyunjinhong anthocyaninoligomersinduceapoptosisandautophagybyinhibitingthemtorsignalingpathwayinhumanbreastcancercells
AT kyungsoonam anthocyaninoligomersinduceapoptosisandautophagybyinhibitingthemtorsignalingpathwayinhumanbreastcancercells