Cargando…

PGC1α downregulation and glycolytic phenotype in thyroid cancer

Increased aerobic glycolysis portends an unfavorable prognosis in thyroid cancer. The metabolic reprogramming likely results from altered mitochondrial activity and may promote cancer progression. Peroxisome proliferator-activated receptor γ coactivator 1α (PGC1α) plays a pivotal role in mitochondri...

Descripción completa

Detalles Bibliográficos
Autores principales: Liu, Chien-Liang, Yang, Po-Sheng, Wang, Tao-Yeuan, Huang, Shih-Yuan, Kuo, Yi-Hue, Cheng, Shih-Ping
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Ivyspring International Publisher 2019
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6636295/
https://www.ncbi.nlm.nih.gov/pubmed/31333799
http://dx.doi.org/10.7150/jca.30018
_version_ 1783436038886129664
author Liu, Chien-Liang
Yang, Po-Sheng
Wang, Tao-Yeuan
Huang, Shih-Yuan
Kuo, Yi-Hue
Cheng, Shih-Ping
author_facet Liu, Chien-Liang
Yang, Po-Sheng
Wang, Tao-Yeuan
Huang, Shih-Yuan
Kuo, Yi-Hue
Cheng, Shih-Ping
author_sort Liu, Chien-Liang
collection PubMed
description Increased aerobic glycolysis portends an unfavorable prognosis in thyroid cancer. The metabolic reprogramming likely results from altered mitochondrial activity and may promote cancer progression. Peroxisome proliferator-activated receptor γ coactivator 1α (PGC1α) plays a pivotal role in mitochondrial biogenesis and function. In the present study, we aimed to evaluate the clinicopathological significance of PGC1α expression and the potential effects of PGC1α modulation. Firstly, the expression of PGC1α in thyroid cancer samples was evaluated using western blot analysis and immunohistochemical staining. Compared with normal thyroid tissue, PGC1α expression was downregulated in thyroid cancer. PGC1α-negative papillary cancer was associated with BRAF V600E mutation, large tumor size, extrathyroidal or lymphovascular invasion, lymph node metastasis, and advanced stage. The results were consistent with the analysis of The Cancer Genome Atlas data. PGC1α expression correlated with oxygen consumption in thyroid cancer cells and was inversely related to AKT activity. The biologic relevance of PGC1α was further investigated by gain- and loss-of-function experimental studies. PGC1α overexpression led to augmented oxidative metabolism and accelerated tumor growth, whereas PGC1α knockdown induced a glycolytic phenotype but reduced tumor growth in vivo. In conclusion, PGC1α downregulation is associated with glycolytic metabolism and advanced disease in thyroid cancer. Nonetheless, manipulating PGC1α expression and metabolic phenotype does not necessarily translate into beneficial effects. It suggests that the metabolic phenotype is likely the consequence rather than the cause of disease progression in thyroid cancer.
format Online
Article
Text
id pubmed-6636295
institution National Center for Biotechnology Information
language English
publishDate 2019
publisher Ivyspring International Publisher
record_format MEDLINE/PubMed
spelling pubmed-66362952019-07-22 PGC1α downregulation and glycolytic phenotype in thyroid cancer Liu, Chien-Liang Yang, Po-Sheng Wang, Tao-Yeuan Huang, Shih-Yuan Kuo, Yi-Hue Cheng, Shih-Ping J Cancer Research Paper Increased aerobic glycolysis portends an unfavorable prognosis in thyroid cancer. The metabolic reprogramming likely results from altered mitochondrial activity and may promote cancer progression. Peroxisome proliferator-activated receptor γ coactivator 1α (PGC1α) plays a pivotal role in mitochondrial biogenesis and function. In the present study, we aimed to evaluate the clinicopathological significance of PGC1α expression and the potential effects of PGC1α modulation. Firstly, the expression of PGC1α in thyroid cancer samples was evaluated using western blot analysis and immunohistochemical staining. Compared with normal thyroid tissue, PGC1α expression was downregulated in thyroid cancer. PGC1α-negative papillary cancer was associated with BRAF V600E mutation, large tumor size, extrathyroidal or lymphovascular invasion, lymph node metastasis, and advanced stage. The results were consistent with the analysis of The Cancer Genome Atlas data. PGC1α expression correlated with oxygen consumption in thyroid cancer cells and was inversely related to AKT activity. The biologic relevance of PGC1α was further investigated by gain- and loss-of-function experimental studies. PGC1α overexpression led to augmented oxidative metabolism and accelerated tumor growth, whereas PGC1α knockdown induced a glycolytic phenotype but reduced tumor growth in vivo. In conclusion, PGC1α downregulation is associated with glycolytic metabolism and advanced disease in thyroid cancer. Nonetheless, manipulating PGC1α expression and metabolic phenotype does not necessarily translate into beneficial effects. It suggests that the metabolic phenotype is likely the consequence rather than the cause of disease progression in thyroid cancer. Ivyspring International Publisher 2019-06-09 /pmc/articles/PMC6636295/ /pubmed/31333799 http://dx.doi.org/10.7150/jca.30018 Text en © Ivyspring International Publisher This is an open access article distributed under the terms of the Creative Commons Attribution (CC BY-NC) license (https://creativecommons.org/licenses/by-nc/4.0/). See http://ivyspring.com/terms for full terms and conditions.
spellingShingle Research Paper
Liu, Chien-Liang
Yang, Po-Sheng
Wang, Tao-Yeuan
Huang, Shih-Yuan
Kuo, Yi-Hue
Cheng, Shih-Ping
PGC1α downregulation and glycolytic phenotype in thyroid cancer
title PGC1α downregulation and glycolytic phenotype in thyroid cancer
title_full PGC1α downregulation and glycolytic phenotype in thyroid cancer
title_fullStr PGC1α downregulation and glycolytic phenotype in thyroid cancer
title_full_unstemmed PGC1α downregulation and glycolytic phenotype in thyroid cancer
title_short PGC1α downregulation and glycolytic phenotype in thyroid cancer
title_sort pgc1α downregulation and glycolytic phenotype in thyroid cancer
topic Research Paper
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6636295/
https://www.ncbi.nlm.nih.gov/pubmed/31333799
http://dx.doi.org/10.7150/jca.30018
work_keys_str_mv AT liuchienliang pgc1adownregulationandglycolyticphenotypeinthyroidcancer
AT yangposheng pgc1adownregulationandglycolyticphenotypeinthyroidcancer
AT wangtaoyeuan pgc1adownregulationandglycolyticphenotypeinthyroidcancer
AT huangshihyuan pgc1adownregulationandglycolyticphenotypeinthyroidcancer
AT kuoyihue pgc1adownregulationandglycolyticphenotypeinthyroidcancer
AT chengshihping pgc1adownregulationandglycolyticphenotypeinthyroidcancer