Cargando…

Mitochondrial Respiration Inhibition Suppresses Papillary Thyroid Carcinoma Via PI3K/Akt/FoxO1/Cyclin D1 Pathway

BACKGROUND: Papillary thyroid carcinoma (PTC) is the most common thyroid malignancy, but little is known regarding PTC metabolic phenotypes and the effects of mitochondrial activity on PTC progression. The great potential of mitochondria-targeting therapy in cancer treatment promoted us to use tool...

Descripción completa

Detalles Bibliográficos
Autores principales: Chen, Bojie, Lei, Shuwen, Yin, Xinlu, Fei, Mengjia, Hu, Yixin, Shi, Yuan, Xu, Yanan, Fu, Lei
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Frontiers Media S.A. 2022
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9295996/
https://www.ncbi.nlm.nih.gov/pubmed/35865479
http://dx.doi.org/10.3389/fonc.2022.900444
_version_ 1784750173216309248
author Chen, Bojie
Lei, Shuwen
Yin, Xinlu
Fei, Mengjia
Hu, Yixin
Shi, Yuan
Xu, Yanan
Fu, Lei
author_facet Chen, Bojie
Lei, Shuwen
Yin, Xinlu
Fei, Mengjia
Hu, Yixin
Shi, Yuan
Xu, Yanan
Fu, Lei
author_sort Chen, Bojie
collection PubMed
description BACKGROUND: Papillary thyroid carcinoma (PTC) is the most common thyroid malignancy, but little is known regarding PTC metabolic phenotypes and the effects of mitochondrial activity on PTC progression. The great potential of mitochondria-targeting therapy in cancer treatment promoted us to use tool compounds from a family of Mito-Fu derivatives to investigate how the regulation of mitochondrial respiration affected tumor progression characteristics and molecular changes in PTC. METHODS: Mito-Fu L20, a representative of 12 synthetic derivatives, was chosen for mitochondrial inhibition experiments. Sample sections from PTC patients were collected and processed to explore potential molecular alterations in tumor lymph node metastasis (LNM). In vitro analyses were performed using human PTC cell lines (K1 and TPC-1), with the human normal thyroid follicular cell line (Nthy) as a control. K1 cells were injected into nude mice to generate an animal model. The mice were injected with normal saline or Mito-Fu L20 at 20 or 50 mg/kg every other day; their body weights and tumor volumes were also measured over time. To elucidate the resulting metabolic phenotype, we measured oxygen consumption rate (OCR) and extracellular acidification rate (ECAR), cellular adenosine triphosphate (ATP) levels and reactive oxygen species (ROS) production, and mitochondrial membrane potential. Wound healing and Transwell assays, cell cycle assays, real-time fluorescence quantitative PCR, Western blotting, and immunohistochemical staining were performed to explore glycolysis-dominant metabolism in PTC. RESULTS: Cyclin D1 and mitochondrial complex IV were detected in tumor samples from PTC patients with LNM. Mito-Fu L20 showed dose-independent and reversible modulation of mitochondrial respiration in PTC. In addition to mitochondrial dysfunction and early apoptosis, G1/S phase arrest. Notably, reversible mitochondrial inhibition yielded durable suppression of tumor proliferation, migration, and invasion via the PI3K/Akt/FoxO1/Cyclin D1 pathway. In vivo experiments demonstrated that Mito-Fu L20 has a good safety profile and specific restorative effect on mitochondrial activity in the liver. In addition, Mito-Fu L20 showed antitumor effects, alleviated tumor angiogenesis, and improved thyroid function. CONCLUSION: Reversible inhibition of ATP production and durable suppression of PTC growth indicates that the downregulation of mitochondrial function has a negative impact on tumor progression and LNM via the PI3K/Akt/FoxO1/Cyclin D1 pathway. The results provide new insights into the antitumor potential and clinical translation of mitochondrial inhibitors.
format Online
Article
Text
id pubmed-9295996
institution National Center for Biotechnology Information
language English
publishDate 2022
publisher Frontiers Media S.A.
record_format MEDLINE/PubMed
spelling pubmed-92959962022-07-20 Mitochondrial Respiration Inhibition Suppresses Papillary Thyroid Carcinoma Via PI3K/Akt/FoxO1/Cyclin D1 Pathway Chen, Bojie Lei, Shuwen Yin, Xinlu Fei, Mengjia Hu, Yixin Shi, Yuan Xu, Yanan Fu, Lei Front Oncol Oncology BACKGROUND: Papillary thyroid carcinoma (PTC) is the most common thyroid malignancy, but little is known regarding PTC metabolic phenotypes and the effects of mitochondrial activity on PTC progression. The great potential of mitochondria-targeting therapy in cancer treatment promoted us to use tool compounds from a family of Mito-Fu derivatives to investigate how the regulation of mitochondrial respiration affected tumor progression characteristics and molecular changes in PTC. METHODS: Mito-Fu L20, a representative of 12 synthetic derivatives, was chosen for mitochondrial inhibition experiments. Sample sections from PTC patients were collected and processed to explore potential molecular alterations in tumor lymph node metastasis (LNM). In vitro analyses were performed using human PTC cell lines (K1 and TPC-1), with the human normal thyroid follicular cell line (Nthy) as a control. K1 cells were injected into nude mice to generate an animal model. The mice were injected with normal saline or Mito-Fu L20 at 20 or 50 mg/kg every other day; their body weights and tumor volumes were also measured over time. To elucidate the resulting metabolic phenotype, we measured oxygen consumption rate (OCR) and extracellular acidification rate (ECAR), cellular adenosine triphosphate (ATP) levels and reactive oxygen species (ROS) production, and mitochondrial membrane potential. Wound healing and Transwell assays, cell cycle assays, real-time fluorescence quantitative PCR, Western blotting, and immunohistochemical staining were performed to explore glycolysis-dominant metabolism in PTC. RESULTS: Cyclin D1 and mitochondrial complex IV were detected in tumor samples from PTC patients with LNM. Mito-Fu L20 showed dose-independent and reversible modulation of mitochondrial respiration in PTC. In addition to mitochondrial dysfunction and early apoptosis, G1/S phase arrest. Notably, reversible mitochondrial inhibition yielded durable suppression of tumor proliferation, migration, and invasion via the PI3K/Akt/FoxO1/Cyclin D1 pathway. In vivo experiments demonstrated that Mito-Fu L20 has a good safety profile and specific restorative effect on mitochondrial activity in the liver. In addition, Mito-Fu L20 showed antitumor effects, alleviated tumor angiogenesis, and improved thyroid function. CONCLUSION: Reversible inhibition of ATP production and durable suppression of PTC growth indicates that the downregulation of mitochondrial function has a negative impact on tumor progression and LNM via the PI3K/Akt/FoxO1/Cyclin D1 pathway. The results provide new insights into the antitumor potential and clinical translation of mitochondrial inhibitors. Frontiers Media S.A. 2022-07-05 /pmc/articles/PMC9295996/ /pubmed/35865479 http://dx.doi.org/10.3389/fonc.2022.900444 Text en Copyright © 2022 Chen, Lei, Yin, Fei, Hu, Shi, Xu and Fu https://creativecommons.org/licenses/by/4.0/This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.
spellingShingle Oncology
Chen, Bojie
Lei, Shuwen
Yin, Xinlu
Fei, Mengjia
Hu, Yixin
Shi, Yuan
Xu, Yanan
Fu, Lei
Mitochondrial Respiration Inhibition Suppresses Papillary Thyroid Carcinoma Via PI3K/Akt/FoxO1/Cyclin D1 Pathway
title Mitochondrial Respiration Inhibition Suppresses Papillary Thyroid Carcinoma Via PI3K/Akt/FoxO1/Cyclin D1 Pathway
title_full Mitochondrial Respiration Inhibition Suppresses Papillary Thyroid Carcinoma Via PI3K/Akt/FoxO1/Cyclin D1 Pathway
title_fullStr Mitochondrial Respiration Inhibition Suppresses Papillary Thyroid Carcinoma Via PI3K/Akt/FoxO1/Cyclin D1 Pathway
title_full_unstemmed Mitochondrial Respiration Inhibition Suppresses Papillary Thyroid Carcinoma Via PI3K/Akt/FoxO1/Cyclin D1 Pathway
title_short Mitochondrial Respiration Inhibition Suppresses Papillary Thyroid Carcinoma Via PI3K/Akt/FoxO1/Cyclin D1 Pathway
title_sort mitochondrial respiration inhibition suppresses papillary thyroid carcinoma via pi3k/akt/foxo1/cyclin d1 pathway
topic Oncology
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9295996/
https://www.ncbi.nlm.nih.gov/pubmed/35865479
http://dx.doi.org/10.3389/fonc.2022.900444
work_keys_str_mv AT chenbojie mitochondrialrespirationinhibitionsuppressespapillarythyroidcarcinomaviapi3kaktfoxo1cyclind1pathway
AT leishuwen mitochondrialrespirationinhibitionsuppressespapillarythyroidcarcinomaviapi3kaktfoxo1cyclind1pathway
AT yinxinlu mitochondrialrespirationinhibitionsuppressespapillarythyroidcarcinomaviapi3kaktfoxo1cyclind1pathway
AT feimengjia mitochondrialrespirationinhibitionsuppressespapillarythyroidcarcinomaviapi3kaktfoxo1cyclind1pathway
AT huyixin mitochondrialrespirationinhibitionsuppressespapillarythyroidcarcinomaviapi3kaktfoxo1cyclind1pathway
AT shiyuan mitochondrialrespirationinhibitionsuppressespapillarythyroidcarcinomaviapi3kaktfoxo1cyclind1pathway
AT xuyanan mitochondrialrespirationinhibitionsuppressespapillarythyroidcarcinomaviapi3kaktfoxo1cyclind1pathway
AT fulei mitochondrialrespirationinhibitionsuppressespapillarythyroidcarcinomaviapi3kaktfoxo1cyclind1pathway