Urolithin A targets the AKT/WNK1 axis to induce autophagy and exert anti-tumor effects in cholangiocarcinoma

Urolithin A (UA; 3,8-dihydroxybenzo[c]chromen-6-one), a metabolite generated by intestinal bacteria during the biotransformation of ellagitannins, has gained considerable attention in treating several cancers. Cholangiocarcinoma (CCA) remains one of the most lethal cancers; it grows in a special env...

Full description

Bibliographic Details
Main Authors: Hidenori Sahashi, Akihisa Kato, Michihiro Yoshida, Kazuki Hayashi, Itaru Naitoh, Yasuki Hori, Makoto Natsume, Naruomi Jinno, Kenta Kachi, Go Asano, Tadashi Toyohara, Yusuke Kito, Sudhakar Ammanamanchi, Hiromi Kataoka
Format: Article
Language:English
Published: Frontiers Media S.A. 2022-09-01
Series:Frontiers in Oncology
Subjects:
Online Access:https://www.frontiersin.org/articles/10.3389/fonc.2022.963314/full
_version_ 1811209147923824640
author Hidenori Sahashi
Akihisa Kato
Michihiro Yoshida
Kazuki Hayashi
Itaru Naitoh
Yasuki Hori
Makoto Natsume
Naruomi Jinno
Kenta Kachi
Go Asano
Tadashi Toyohara
Yusuke Kito
Sudhakar Ammanamanchi
Hiromi Kataoka
author_facet Hidenori Sahashi
Akihisa Kato
Michihiro Yoshida
Kazuki Hayashi
Itaru Naitoh
Yasuki Hori
Makoto Natsume
Naruomi Jinno
Kenta Kachi
Go Asano
Tadashi Toyohara
Yusuke Kito
Sudhakar Ammanamanchi
Hiromi Kataoka
author_sort Hidenori Sahashi
collection DOAJ
description Urolithin A (UA; 3,8-dihydroxybenzo[c]chromen-6-one), a metabolite generated by intestinal bacteria during the biotransformation of ellagitannins, has gained considerable attention in treating several cancers. Cholangiocarcinoma (CCA) remains one of the most lethal cancers; it grows in a special environment constantly exposed to both blood and bile. Since UA is known to undergo enterohepatic recirculation, we hypothesized that UA might have significant antitumor effects in CCA. Here, we investigated the therapeutic potential of UA in CCA and aimed to elucidate its mechanisms, including autophagy. UA treatment inhibited cell proliferation and induced G2/M phase cell cycle arrest in CCA cells. UA also suppressed cell migration and invasion, but did not cause apoptosis. Furthermore, Western blotting and immunocytochemistry demonstrated increased LC3-II accumulation, while electron microscopy demonstrated induced autophagosomes after UA treatment, suggesting that UA upregulated autophagy in CCA cells. In xenograft mice treated with UA, tumor growth was inhibited with increased LC3-II levels. On the other hand, phospho-kinase array demonstrated downregulation of the AKT/WNK1 pathway. LC3-II expression was elevated in WNK1 knocked down cells, indicating that WNK1 is the key signal for regulating autophagy. Thus, UA exerted antitumor effects by suppressing the AKT/WNK1 signaling pathway and inducing autophagy. In conclusion, UA, a natural, well-tolerated compound, may be a promising therapeutic candidate for advanced CCA.
first_indexed 2024-04-12T04:33:28Z
format Article
id doaj.art-723f98a57ed148ebb30f6514e67af4f6
institution Directory Open Access Journal
issn 2234-943X
language English
last_indexed 2024-04-12T04:33:28Z
publishDate 2022-09-01
publisher Frontiers Media S.A.
record_format Article
series Frontiers in Oncology
spelling doaj.art-723f98a57ed148ebb30f6514e67af4f62022-12-22T03:47:52ZengFrontiers Media S.A.Frontiers in Oncology2234-943X2022-09-011210.3389/fonc.2022.963314963314Urolithin A targets the AKT/WNK1 axis to induce autophagy and exert anti-tumor effects in cholangiocarcinomaHidenori Sahashi0Akihisa Kato1Michihiro Yoshida2Kazuki Hayashi3Itaru Naitoh4Yasuki Hori5Makoto Natsume6Naruomi Jinno7Kenta Kachi8Go Asano9Tadashi Toyohara10Yusuke Kito11Sudhakar Ammanamanchi12Hiromi Kataoka13Department of Gastroenterology and Metabolism, Graduate School of Medical Sciences, Nagoya City University, Nagoya, JapanDepartment of Gastroenterology and Metabolism, Graduate School of Medical Sciences, Nagoya City University, Nagoya, JapanDepartment of Gastroenterology and Metabolism, Graduate School of Medical Sciences, Nagoya City University, Nagoya, JapanDepartment of Gastroenterology and Metabolism, Graduate School of Medical Sciences, Nagoya City University, Nagoya, JapanDepartment of Gastroenterology and Metabolism, Graduate School of Medical Sciences, Nagoya City University, Nagoya, JapanDepartment of Gastroenterology and Metabolism, Graduate School of Medical Sciences, Nagoya City University, Nagoya, JapanDepartment of Gastroenterology and Metabolism, Graduate School of Medical Sciences, Nagoya City University, Nagoya, JapanDepartment of Gastroenterology and Metabolism, Graduate School of Medical Sciences, Nagoya City University, Nagoya, JapanDepartment of Gastroenterology and Metabolism, Graduate School of Medical Sciences, Nagoya City University, Nagoya, JapanDepartment of Gastroenterology and Metabolism, Graduate School of Medical Sciences, Nagoya City University, Nagoya, JapanDepartment of Gastroenterology and Metabolism, Graduate School of Medical Sciences, Nagoya City University, Nagoya, JapanDepartment of Gastroenterology and Metabolism, Graduate School of Medical Sciences, Nagoya City University, Nagoya, JapanDepartment of Internal Medicine, University of Arizona College of Medicine, Phoenix, AZ, United StatesDepartment of Gastroenterology and Metabolism, Graduate School of Medical Sciences, Nagoya City University, Nagoya, JapanUrolithin A (UA; 3,8-dihydroxybenzo[c]chromen-6-one), a metabolite generated by intestinal bacteria during the biotransformation of ellagitannins, has gained considerable attention in treating several cancers. Cholangiocarcinoma (CCA) remains one of the most lethal cancers; it grows in a special environment constantly exposed to both blood and bile. Since UA is known to undergo enterohepatic recirculation, we hypothesized that UA might have significant antitumor effects in CCA. Here, we investigated the therapeutic potential of UA in CCA and aimed to elucidate its mechanisms, including autophagy. UA treatment inhibited cell proliferation and induced G2/M phase cell cycle arrest in CCA cells. UA also suppressed cell migration and invasion, but did not cause apoptosis. Furthermore, Western blotting and immunocytochemistry demonstrated increased LC3-II accumulation, while electron microscopy demonstrated induced autophagosomes after UA treatment, suggesting that UA upregulated autophagy in CCA cells. In xenograft mice treated with UA, tumor growth was inhibited with increased LC3-II levels. On the other hand, phospho-kinase array demonstrated downregulation of the AKT/WNK1 pathway. LC3-II expression was elevated in WNK1 knocked down cells, indicating that WNK1 is the key signal for regulating autophagy. Thus, UA exerted antitumor effects by suppressing the AKT/WNK1 signaling pathway and inducing autophagy. In conclusion, UA, a natural, well-tolerated compound, may be a promising therapeutic candidate for advanced CCA.https://www.frontiersin.org/articles/10.3389/fonc.2022.963314/fullUrolithin AUAcholangiocarcinomaautophagyWNK1
spellingShingle Hidenori Sahashi
Akihisa Kato
Michihiro Yoshida
Kazuki Hayashi
Itaru Naitoh
Yasuki Hori
Makoto Natsume
Naruomi Jinno
Kenta Kachi
Go Asano
Tadashi Toyohara
Yusuke Kito
Sudhakar Ammanamanchi
Hiromi Kataoka
Urolithin A targets the AKT/WNK1 axis to induce autophagy and exert anti-tumor effects in cholangiocarcinoma
Frontiers in Oncology
Urolithin A
UA
cholangiocarcinoma
autophagy
WNK1
title Urolithin A targets the AKT/WNK1 axis to induce autophagy and exert anti-tumor effects in cholangiocarcinoma
title_full Urolithin A targets the AKT/WNK1 axis to induce autophagy and exert anti-tumor effects in cholangiocarcinoma
title_fullStr Urolithin A targets the AKT/WNK1 axis to induce autophagy and exert anti-tumor effects in cholangiocarcinoma
title_full_unstemmed Urolithin A targets the AKT/WNK1 axis to induce autophagy and exert anti-tumor effects in cholangiocarcinoma
title_short Urolithin A targets the AKT/WNK1 axis to induce autophagy and exert anti-tumor effects in cholangiocarcinoma
title_sort urolithin a targets the akt wnk1 axis to induce autophagy and exert anti tumor effects in cholangiocarcinoma
topic Urolithin A
UA
cholangiocarcinoma
autophagy
WNK1
url https://www.frontiersin.org/articles/10.3389/fonc.2022.963314/full
work_keys_str_mv AT hidenorisahashi urolithinatargetstheaktwnk1axistoinduceautophagyandexertantitumoreffectsincholangiocarcinoma
AT akihisakato urolithinatargetstheaktwnk1axistoinduceautophagyandexertantitumoreffectsincholangiocarcinoma
AT michihiroyoshida urolithinatargetstheaktwnk1axistoinduceautophagyandexertantitumoreffectsincholangiocarcinoma
AT kazukihayashi urolithinatargetstheaktwnk1axistoinduceautophagyandexertantitumoreffectsincholangiocarcinoma
AT itarunaitoh urolithinatargetstheaktwnk1axistoinduceautophagyandexertantitumoreffectsincholangiocarcinoma
AT yasukihori urolithinatargetstheaktwnk1axistoinduceautophagyandexertantitumoreffectsincholangiocarcinoma
AT makotonatsume urolithinatargetstheaktwnk1axistoinduceautophagyandexertantitumoreffectsincholangiocarcinoma
AT naruomijinno urolithinatargetstheaktwnk1axistoinduceautophagyandexertantitumoreffectsincholangiocarcinoma
AT kentakachi urolithinatargetstheaktwnk1axistoinduceautophagyandexertantitumoreffectsincholangiocarcinoma
AT goasano urolithinatargetstheaktwnk1axistoinduceautophagyandexertantitumoreffectsincholangiocarcinoma
AT tadashitoyohara urolithinatargetstheaktwnk1axistoinduceautophagyandexertantitumoreffectsincholangiocarcinoma
AT yusukekito urolithinatargetstheaktwnk1axistoinduceautophagyandexertantitumoreffectsincholangiocarcinoma
AT sudhakarammanamanchi urolithinatargetstheaktwnk1axistoinduceautophagyandexertantitumoreffectsincholangiocarcinoma
AT hiromikataoka urolithinatargetstheaktwnk1axistoinduceautophagyandexertantitumoreffectsincholangiocarcinoma