Title Cancer-associated fibroblast-specific lncRNA LINC01614 enhances glutamine uptake in lung adenocarcinoma
Authors Liu, Tongyan
Han, Chencheng
Fang, Panqi
Ma, Zhifei
Wang, Xiaoxiao
Chen, Hao
Wang, Siwei
Meng, Fanchen
Wang, Cheng
Zhang, Erbao
Dong, Guozhang
Zhu, Hongyu
Yin, Wenda
Wang, Jie
Zuo, Xianglin
Qiu, Mantang
Wang, Jinke
Qian, Xu
Shen, Hongbing
Xu, Lin
Hu, Zhibin
Yin, Rong
Affiliation Jiangsu Canc Hosp, Jiangsu Key Lab Mol & Translat Canc Res, Dept Thorac Surg, Nanjing 21009, Peoples R China
Nanjing Med Univ, Affiliated Canc Hosp, Nanjing 21009, Peoples R China
Nanjing Med Univ, Jiangsu Inst Canc Res, Nanjing 21009, Peoples R China
Nanjing Med Univ, Dept Sci & Technol, Jiangsu Canc Hosp, Nanjing 21009, Peoples R China
Jiangsu Inst Canc Res, Nanjing 21009, Peoples R China
Nanjing Univ Chinese Med, Jiangsu Prov Hosp Chinese Med, Dept GCP Res Ctr, Affiliated Hosp, Nanjing 210029, Peoples R China
Nanjing Med Univ, Collaborat Innovat Ctr Canc Personalized Med, Nanjing 211116, Peoples R China
Nanjing Med Univ, Sch Publ Hlth, Dept Epidemiol & Biostat, Int Joint Res Ctr Environm & Human Hlth, Nanjing 211116, Peoples R China
Nanjing Med Univ, Dept Bioinformat, Nanjing 211166, Peoples R China
Biobank Lung Canc, Jiangsu Biobank Clin Resources, Nanjing 21009, Peoples R China
Peking Univ, Dept Thorac Surg, Peoples Hosp, Beijing 100044, Peoples R China
Southeast Univ, State Key Lab Bioelect, Nanjing 210018, Peoples R China
Keywords LONG NONCODING RNAS
METABOLISM
Issue Date 8-Oct-2022
Publisher JOURNAL OF HEMATOLOGY & ONCOLOGY
Abstract Background Besides featured glucose consumption, recent studies reveal that cancer cells might prefer "addicting" specific energy substrates from the tumor microenvironment (TME); however, the underlying mechanisms remain unclear. Methods Fibroblast-specific long noncoding RNAs were screened using RNA-seq data of our NJLCC cohort, TCGA, and CCLE datasets. The expression and package of LINC01614 into exosomes were identified using flow cytometric sorting, fluorescence in situ hybridization (FISH), and quantitative reverse transcription polymerase chain reaction (RT-PCR). The transfer and functional role of LINC01614 in lung adenocarcinoma (LUAD) and CAFs were investigated using 4-thiouracil-labeled RNA transfer and gain- and loss-of-function approaches. RNA pull-down, RNA immunoprecipitation, dual-luciferase assay, gene expression microarray, and bioinformatics analysis were performed to investigate the underlying mechanisms involved. Results We demonstrate that cancer-associated fibroblasts (CAFs) in LUAD primarily enhance the glutamine metabolism of cancer cells. A CAF-specific long noncoding RNA, LINC01614, packaged by CAF-derived exosomes, mediates the enhancement of glutamine uptake in LUAD cells. Mechanistically, LINC01614 directly interacts with ANXA2 and p65 to facilitate the activation of NF-kappa B, which leads to the upregulation of the glutamine transporters SLC38A2 and SLC7A5 and eventually enhances the glutamine influx of cancer cells. Reciprocally, tumor-derived proinflammatory cytokines upregulate LINC01614 in CAFs, constituting a feedforward loop between CAFs and cancer cells. Blocking exosome-transmitted LINC01614 inhibits glutamine addiction and LUAD growth in vivo. Clinically, LINC01614 expression in CAFs is associated with the glutamine influx and poor prognosis of patients with LUAD. Conclusion Our study highlights the therapeutic potential of targeting a CAF-specific lncRNA to inhibit glutamine utilization and cancer progression in LUAD.
URI http://hdl.handle.net/20.500.11897/655682
DOI 10.1186/s13045-022-01359-4
Indexed SCI(E)
Appears in Collections: 人民医院

Files in This Work
There are no files associated with this item.

Web of Science®


0

Checked on Last Week

Scopus®



Checked on Current Time

百度学术™


0

Checked on Current Time

Google Scholar™





License: See PKU IR operational policies.