Cargando…

Stearoyl-CoA Desaturase-1 dependent lipid droplets accumulation in cancer-associated fibroblasts facilitates the progression of lung cancer

Rationale: Cancer-associated fibroblasts (CAFs) are the main components in the tumor microenvironment (TME) and facilitate lung cancer progression. Studies have reported that metabolic reprogramming can regulate the function of CAFs, especially abnormal lipid metabolism. Lipid droplets (LDs) are ubi...

Descripción completa

Detalles Bibliográficos
Autores principales: Zhang, Yana, Gu, Zhuoyu, Wan, Jiajia, Lou, Xiaohan, Liu, Shuangqing, Wang, Yuan, Bian, Yangyang, Wang, Fei, Li, Zhenzhen, Qin, Zhihai
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Ivyspring International Publisher 2022
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9682541/
https://www.ncbi.nlm.nih.gov/pubmed/36439884
http://dx.doi.org/10.7150/ijbs.74924
_version_ 1784834874437271552
author Zhang, Yana
Gu, Zhuoyu
Wan, Jiajia
Lou, Xiaohan
Liu, Shuangqing
Wang, Yuan
Bian, Yangyang
Wang, Fei
Li, Zhenzhen
Qin, Zhihai
author_facet Zhang, Yana
Gu, Zhuoyu
Wan, Jiajia
Lou, Xiaohan
Liu, Shuangqing
Wang, Yuan
Bian, Yangyang
Wang, Fei
Li, Zhenzhen
Qin, Zhihai
author_sort Zhang, Yana
collection PubMed
description Rationale: Cancer-associated fibroblasts (CAFs) are the main components in the tumor microenvironment (TME) and facilitate lung cancer progression. Studies have reported that metabolic reprogramming can regulate the function of CAFs, especially abnormal lipid metabolism. Lipid droplets (LDs) are ubiquitous organelles that store neutral lipids and have a crucial role in lipid metabolism. However, little is known about the synthesis and functions of LDs in lung CAFs. Methods: TetO-EGFR(L858R); CCSP-rtTA transgenic mouse model was used to establish a spontaneous pulmonary tumor model and investigate the accumulation of LDs in CAFs. The effect of LDs accumulation on the phenotype change of fibroblasts was estimated in vitro using mouse fibroblast cell lines. RNA sequencing, Western blotting, RT-PCR, and DNA-pull down were performed to determine the mechanism of LDs synthesis in fibroblasts. Results: We found that LDs were enriched in lung CAFs and induced the pro-tumoral phenotype of CAFs with increased expression of α-smooth muscle actin (α-SMA) and Collagen alpha-2 (I) chain (COL1A2). As the main regulator, hypoxia-inducible factor-1α (HIF-1α) was highly expressed in activated fibroblasts and increased the content of LDs. RNA-sequencing results showed that Stearoyl-CoA Desaturase1 (SCD1) was a downstream gene of HIF-1α, which upregulated the number of LDs in fibroblasts. Importantly, SCD1 inhibition reduced the growth of lung tumors, which was correlated with LDs decrease in CAFs. Analysis of human lung adenocarcinoma tissue chip revealed that CAFs with a high level of SCD1 were positively correlated with the expression of HIF-1α and poor survival in lung cancer patients. Conclusions: The HIF-1α/SCD1 axis regulates the accumulation of LDs in CAFs, which might represent a novel target for lung cancer therapy.
format Online
Article
Text
id pubmed-9682541
institution National Center for Biotechnology Information
language English
publishDate 2022
publisher Ivyspring International Publisher
record_format MEDLINE/PubMed
spelling pubmed-96825412022-11-25 Stearoyl-CoA Desaturase-1 dependent lipid droplets accumulation in cancer-associated fibroblasts facilitates the progression of lung cancer Zhang, Yana Gu, Zhuoyu Wan, Jiajia Lou, Xiaohan Liu, Shuangqing Wang, Yuan Bian, Yangyang Wang, Fei Li, Zhenzhen Qin, Zhihai Int J Biol Sci Research Paper Rationale: Cancer-associated fibroblasts (CAFs) are the main components in the tumor microenvironment (TME) and facilitate lung cancer progression. Studies have reported that metabolic reprogramming can regulate the function of CAFs, especially abnormal lipid metabolism. Lipid droplets (LDs) are ubiquitous organelles that store neutral lipids and have a crucial role in lipid metabolism. However, little is known about the synthesis and functions of LDs in lung CAFs. Methods: TetO-EGFR(L858R); CCSP-rtTA transgenic mouse model was used to establish a spontaneous pulmonary tumor model and investigate the accumulation of LDs in CAFs. The effect of LDs accumulation on the phenotype change of fibroblasts was estimated in vitro using mouse fibroblast cell lines. RNA sequencing, Western blotting, RT-PCR, and DNA-pull down were performed to determine the mechanism of LDs synthesis in fibroblasts. Results: We found that LDs were enriched in lung CAFs and induced the pro-tumoral phenotype of CAFs with increased expression of α-smooth muscle actin (α-SMA) and Collagen alpha-2 (I) chain (COL1A2). As the main regulator, hypoxia-inducible factor-1α (HIF-1α) was highly expressed in activated fibroblasts and increased the content of LDs. RNA-sequencing results showed that Stearoyl-CoA Desaturase1 (SCD1) was a downstream gene of HIF-1α, which upregulated the number of LDs in fibroblasts. Importantly, SCD1 inhibition reduced the growth of lung tumors, which was correlated with LDs decrease in CAFs. Analysis of human lung adenocarcinoma tissue chip revealed that CAFs with a high level of SCD1 were positively correlated with the expression of HIF-1α and poor survival in lung cancer patients. Conclusions: The HIF-1α/SCD1 axis regulates the accumulation of LDs in CAFs, which might represent a novel target for lung cancer therapy. Ivyspring International Publisher 2022-10-18 /pmc/articles/PMC9682541/ /pubmed/36439884 http://dx.doi.org/10.7150/ijbs.74924 Text en © The author(s) https://creativecommons.org/licenses/by/4.0/This is an open access article distributed under the terms of the Creative Commons Attribution License (https://creativecommons.org/licenses/by/4.0/). See http://ivyspring.com/terms for full terms and conditions.
spellingShingle Research Paper
Zhang, Yana
Gu, Zhuoyu
Wan, Jiajia
Lou, Xiaohan
Liu, Shuangqing
Wang, Yuan
Bian, Yangyang
Wang, Fei
Li, Zhenzhen
Qin, Zhihai
Stearoyl-CoA Desaturase-1 dependent lipid droplets accumulation in cancer-associated fibroblasts facilitates the progression of lung cancer
title Stearoyl-CoA Desaturase-1 dependent lipid droplets accumulation in cancer-associated fibroblasts facilitates the progression of lung cancer
title_full Stearoyl-CoA Desaturase-1 dependent lipid droplets accumulation in cancer-associated fibroblasts facilitates the progression of lung cancer
title_fullStr Stearoyl-CoA Desaturase-1 dependent lipid droplets accumulation in cancer-associated fibroblasts facilitates the progression of lung cancer
title_full_unstemmed Stearoyl-CoA Desaturase-1 dependent lipid droplets accumulation in cancer-associated fibroblasts facilitates the progression of lung cancer
title_short Stearoyl-CoA Desaturase-1 dependent lipid droplets accumulation in cancer-associated fibroblasts facilitates the progression of lung cancer
title_sort stearoyl-coa desaturase-1 dependent lipid droplets accumulation in cancer-associated fibroblasts facilitates the progression of lung cancer
topic Research Paper
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9682541/
https://www.ncbi.nlm.nih.gov/pubmed/36439884
http://dx.doi.org/10.7150/ijbs.74924
work_keys_str_mv AT zhangyana stearoylcoadesaturase1dependentlipiddropletsaccumulationincancerassociatedfibroblastsfacilitatestheprogressionoflungcancer
AT guzhuoyu stearoylcoadesaturase1dependentlipiddropletsaccumulationincancerassociatedfibroblastsfacilitatestheprogressionoflungcancer
AT wanjiajia stearoylcoadesaturase1dependentlipiddropletsaccumulationincancerassociatedfibroblastsfacilitatestheprogressionoflungcancer
AT louxiaohan stearoylcoadesaturase1dependentlipiddropletsaccumulationincancerassociatedfibroblastsfacilitatestheprogressionoflungcancer
AT liushuangqing stearoylcoadesaturase1dependentlipiddropletsaccumulationincancerassociatedfibroblastsfacilitatestheprogressionoflungcancer
AT wangyuan stearoylcoadesaturase1dependentlipiddropletsaccumulationincancerassociatedfibroblastsfacilitatestheprogressionoflungcancer
AT bianyangyang stearoylcoadesaturase1dependentlipiddropletsaccumulationincancerassociatedfibroblastsfacilitatestheprogressionoflungcancer
AT wangfei stearoylcoadesaturase1dependentlipiddropletsaccumulationincancerassociatedfibroblastsfacilitatestheprogressionoflungcancer
AT lizhenzhen stearoylcoadesaturase1dependentlipiddropletsaccumulationincancerassociatedfibroblastsfacilitatestheprogressionoflungcancer
AT qinzhihai stearoylcoadesaturase1dependentlipiddropletsaccumulationincancerassociatedfibroblastsfacilitatestheprogressionoflungcancer