<i>Calocedrus formosana</i> Essential Oils Induce ROS-Mediated Autophagy and Apoptosis by Targeting SIRT1 in Colon Cancer Cells

Colorectal cancer is the most common cancer that affects both sexes and has a poor prognosis due to aggressiveness and chemoresistance. Essential oils isolated from <i>Calocedrus formosana</i> (CF-EOs) have been shown to demonstrate anti-termite, antifungal, anti-mosquito, and anti-micro...

Full description

Bibliographic Details
Main Authors: Atikul Islam, Yu-Chun Chang, Nai-Wen Tsao, Sheng-Yang Wang, Pin Ju Chueh
Format: Article
Language:English
Published: MDPI AG 2024-02-01
Series:Antioxidants
Subjects:
Online Access:https://www.mdpi.com/2076-3921/13/3/284
_version_ 1797242272568311808
author Atikul Islam
Yu-Chun Chang
Nai-Wen Tsao
Sheng-Yang Wang
Pin Ju Chueh
author_facet Atikul Islam
Yu-Chun Chang
Nai-Wen Tsao
Sheng-Yang Wang
Pin Ju Chueh
author_sort Atikul Islam
collection DOAJ
description Colorectal cancer is the most common cancer that affects both sexes and has a poor prognosis due to aggressiveness and chemoresistance. Essential oils isolated from <i>Calocedrus formosana</i> (CF-EOs) have been shown to demonstrate anti-termite, antifungal, anti-mosquito, and anti-microbial activities. However, the anticancer effects of CF-EOs are not yet fully understood. Therefore, the present study aimed to explore the molecular mechanism underlying CF-EOs-mediated anti-proliferative activity in colon cancer cells. Here, cell impedance measurements showed that CF-EOs inhibit proliferation in colon cancer cells with wild-type or mutant p53. Flow cytometry revealed that CF-EOs at 20, 50 µg/mL significantly induced ROS generation and autophagy in both HCT116 p53-wt and HCT116 p53-null cell lines, whereas pretreatment with the ROS scavenger N-acetyl cysteine (NAC) markedly attenuated these changes. CF-EOs also induced apoptosis at 50 µg/mL in both lines, as determined by flow cytometry. Protein analysis showed that CF-EOs markedly induced apoptosis markers, including Trail, cleaved caspase-3, cleaved caspase-9, and cleaved PARP, as well as autophagy markers, such as the levels of ULK1, Atg5, Atg6, Atg7, and the conversion of LC3-I to LC3-II. CF-EOs were further found to inhibit the activity and expression of the NAD<sup>+</sup>-dependent deacetylase SIRT1 to increase the levels of acetylated p53 (Ac-p53) in p53-wt cells and acetylated c-Myc (Ac-c-Myc) in p53-null cells, ultimately inducing apoptosis in both lines. Interestingly, suppression of SIRT1 by CF-EOs enhanced the acetylation of ULK1, which in turn prompted ROS-dependent autophagy in colon cancer cells. The induction of apoptosis and autophagy by CF-EOs suggests that they may have potential as a promising new approach for treating cancer. Collectively, our results suggest that essential oils isolated from <i>Calocedrus formosana</i> act as a promising anticancer agent against colon cancer cells by targeting SIRT1 to induce ROS-mediated autophagy and apoptosis.
first_indexed 2024-04-24T18:36:35Z
format Article
id doaj.art-5672cbfb097b438ab28940e508137d03
institution Directory Open Access Journal
issn 2076-3921
language English
last_indexed 2024-04-24T18:36:35Z
publishDate 2024-02-01
publisher MDPI AG
record_format Article
series Antioxidants
spelling doaj.art-5672cbfb097b438ab28940e508137d032024-03-27T13:18:27ZengMDPI AGAntioxidants2076-39212024-02-0113328410.3390/antiox13030284<i>Calocedrus formosana</i> Essential Oils Induce ROS-Mediated Autophagy and Apoptosis by Targeting SIRT1 in Colon Cancer CellsAtikul Islam0Yu-Chun Chang1Nai-Wen Tsao2Sheng-Yang Wang3Pin Ju Chueh4Institute of Biomedical Sciences, National Chung Hsing University, Taichung 40227, TaiwanInstitute of Biomedical Sciences, National Chung Hsing University, Taichung 40227, TaiwanSpecial Crop and Metabolome Discipline Cluster, Academy Circle Economy, National Chung Hsing University, Taichung City 402202, TaiwanDepartment of Forestry, National Chung Hsing University, Taichung 40402, TaiwanInstitute of Biomedical Sciences, National Chung Hsing University, Taichung 40227, TaiwanColorectal cancer is the most common cancer that affects both sexes and has a poor prognosis due to aggressiveness and chemoresistance. Essential oils isolated from <i>Calocedrus formosana</i> (CF-EOs) have been shown to demonstrate anti-termite, antifungal, anti-mosquito, and anti-microbial activities. However, the anticancer effects of CF-EOs are not yet fully understood. Therefore, the present study aimed to explore the molecular mechanism underlying CF-EOs-mediated anti-proliferative activity in colon cancer cells. Here, cell impedance measurements showed that CF-EOs inhibit proliferation in colon cancer cells with wild-type or mutant p53. Flow cytometry revealed that CF-EOs at 20, 50 µg/mL significantly induced ROS generation and autophagy in both HCT116 p53-wt and HCT116 p53-null cell lines, whereas pretreatment with the ROS scavenger N-acetyl cysteine (NAC) markedly attenuated these changes. CF-EOs also induced apoptosis at 50 µg/mL in both lines, as determined by flow cytometry. Protein analysis showed that CF-EOs markedly induced apoptosis markers, including Trail, cleaved caspase-3, cleaved caspase-9, and cleaved PARP, as well as autophagy markers, such as the levels of ULK1, Atg5, Atg6, Atg7, and the conversion of LC3-I to LC3-II. CF-EOs were further found to inhibit the activity and expression of the NAD<sup>+</sup>-dependent deacetylase SIRT1 to increase the levels of acetylated p53 (Ac-p53) in p53-wt cells and acetylated c-Myc (Ac-c-Myc) in p53-null cells, ultimately inducing apoptosis in both lines. Interestingly, suppression of SIRT1 by CF-EOs enhanced the acetylation of ULK1, which in turn prompted ROS-dependent autophagy in colon cancer cells. The induction of apoptosis and autophagy by CF-EOs suggests that they may have potential as a promising new approach for treating cancer. Collectively, our results suggest that essential oils isolated from <i>Calocedrus formosana</i> act as a promising anticancer agent against colon cancer cells by targeting SIRT1 to induce ROS-mediated autophagy and apoptosis.https://www.mdpi.com/2076-3921/13/3/284colorectal cancer<i>Calocedrus formosana</i>essential oilsreactive oxygen species (ROS)autophagyapoptosis
spellingShingle Atikul Islam
Yu-Chun Chang
Nai-Wen Tsao
Sheng-Yang Wang
Pin Ju Chueh
<i>Calocedrus formosana</i> Essential Oils Induce ROS-Mediated Autophagy and Apoptosis by Targeting SIRT1 in Colon Cancer Cells
Antioxidants
colorectal cancer
<i>Calocedrus formosana</i>
essential oils
reactive oxygen species (ROS)
autophagy
apoptosis
title <i>Calocedrus formosana</i> Essential Oils Induce ROS-Mediated Autophagy and Apoptosis by Targeting SIRT1 in Colon Cancer Cells
title_full <i>Calocedrus formosana</i> Essential Oils Induce ROS-Mediated Autophagy and Apoptosis by Targeting SIRT1 in Colon Cancer Cells
title_fullStr <i>Calocedrus formosana</i> Essential Oils Induce ROS-Mediated Autophagy and Apoptosis by Targeting SIRT1 in Colon Cancer Cells
title_full_unstemmed <i>Calocedrus formosana</i> Essential Oils Induce ROS-Mediated Autophagy and Apoptosis by Targeting SIRT1 in Colon Cancer Cells
title_short <i>Calocedrus formosana</i> Essential Oils Induce ROS-Mediated Autophagy and Apoptosis by Targeting SIRT1 in Colon Cancer Cells
title_sort i calocedrus formosana i essential oils induce ros mediated autophagy and apoptosis by targeting sirt1 in colon cancer cells
topic colorectal cancer
<i>Calocedrus formosana</i>
essential oils
reactive oxygen species (ROS)
autophagy
apoptosis
url https://www.mdpi.com/2076-3921/13/3/284
work_keys_str_mv AT atikulislam icalocedrusformosanaiessentialoilsinducerosmediatedautophagyandapoptosisbytargetingsirt1incoloncancercells
AT yuchunchang icalocedrusformosanaiessentialoilsinducerosmediatedautophagyandapoptosisbytargetingsirt1incoloncancercells
AT naiwentsao icalocedrusformosanaiessentialoilsinducerosmediatedautophagyandapoptosisbytargetingsirt1incoloncancercells
AT shengyangwang icalocedrusformosanaiessentialoilsinducerosmediatedautophagyandapoptosisbytargetingsirt1incoloncancercells
AT pinjuchueh icalocedrusformosanaiessentialoilsinducerosmediatedautophagyandapoptosisbytargetingsirt1incoloncancercells