Cargando…

Mechanism of lnRNA-ICL involved in lung cancer development in COPD patients through modulating microRNA-19-3p/NKRF/NF-κB axis

The incidence of lung cancer (LC) in chronic obstructive pulmonary disease (COPD) patients is dozens of times higher than that in patients without COPD. Elevated activity of nuclear factor-k-gene binding (NF-κB) was found in lung tissue of patients with COPD, and the continuous activation of NF-κB i...

Descripción completa

Detalles Bibliográficos
Autores principales: Lu, Jingjing, Shi, Yan, Zhang, Feng, Zhang, Ying, Zhao, Xiangwang, Zheng, Haiyan, Li, Lingyu, Zhao, Shiqiao, Zhao, Liming
Formato: Online Artículo Texto
Lenguaje:English
Publicado: BioMed Central 2023
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10071758/
https://www.ncbi.nlm.nih.gov/pubmed/37013587
http://dx.doi.org/10.1186/s12935-023-02900-2
_version_ 1785019259438497792
author Lu, Jingjing
Shi, Yan
Zhang, Feng
Zhang, Ying
Zhao, Xiangwang
Zheng, Haiyan
Li, Lingyu
Zhao, Shiqiao
Zhao, Liming
author_facet Lu, Jingjing
Shi, Yan
Zhang, Feng
Zhang, Ying
Zhao, Xiangwang
Zheng, Haiyan
Li, Lingyu
Zhao, Shiqiao
Zhao, Liming
author_sort Lu, Jingjing
collection PubMed
description The incidence of lung cancer (LC) in chronic obstructive pulmonary disease (COPD) patients is dozens of times higher than that in patients without COPD. Elevated activity of nuclear factor-k-gene binding (NF-κB) was found in lung tissue of patients with COPD, and the continuous activation of NF-κB is observed in both malignant transformation and tumor progression of LC, suggesting that NF-κB and its regulators may play a key role in the progression of LC in COPD patients. Here, we report for the first time that a key long non-coding RNA (lncRNA)-ICL involved in the regulation of NF-κB activity in LC tissues of COPD patients. The analyses showed that the expression of ICL significantly decreased in LC tissues of LC patients with COPD than that in LC tissues of LC patients without COPD. Functional experiments in vitro showed that exogenous ICL only significantly inhibited the proliferation, invasion and migration in primary tumor cells of LC patients with COPD compared to LC patients without COPD. Mechanism studies have shown that ICL could suppress the activation of NF-κB by blocking the hsa-miR19-3p/NKRF/NF-κB pathway as a microRNA sponge. Furthermore, In vivo experiments showed that exogenous ICL effectively inhibited the growth of patient-derived subcutaneous tumor xenografts (PDX) of LC patients with COPD and significantly prolonged the survival time of tumor-bearing mice. In a word, our study shows that the decrease of ICL is associated with an increased risk of LC in patients with COPD, ICL is not only expected to be a new therapeutic target for LC in COPD patients, but also has great potential to be used as a new marker for evaluating the occurrence, severity stratification and prognosis of LC in patients with COPD.
format Online
Article
Text
id pubmed-10071758
institution National Center for Biotechnology Information
language English
publishDate 2023
publisher BioMed Central
record_format MEDLINE/PubMed
spelling pubmed-100717582023-04-05 Mechanism of lnRNA-ICL involved in lung cancer development in COPD patients through modulating microRNA-19-3p/NKRF/NF-κB axis Lu, Jingjing Shi, Yan Zhang, Feng Zhang, Ying Zhao, Xiangwang Zheng, Haiyan Li, Lingyu Zhao, Shiqiao Zhao, Liming Cancer Cell Int Research The incidence of lung cancer (LC) in chronic obstructive pulmonary disease (COPD) patients is dozens of times higher than that in patients without COPD. Elevated activity of nuclear factor-k-gene binding (NF-κB) was found in lung tissue of patients with COPD, and the continuous activation of NF-κB is observed in both malignant transformation and tumor progression of LC, suggesting that NF-κB and its regulators may play a key role in the progression of LC in COPD patients. Here, we report for the first time that a key long non-coding RNA (lncRNA)-ICL involved in the regulation of NF-κB activity in LC tissues of COPD patients. The analyses showed that the expression of ICL significantly decreased in LC tissues of LC patients with COPD than that in LC tissues of LC patients without COPD. Functional experiments in vitro showed that exogenous ICL only significantly inhibited the proliferation, invasion and migration in primary tumor cells of LC patients with COPD compared to LC patients without COPD. Mechanism studies have shown that ICL could suppress the activation of NF-κB by blocking the hsa-miR19-3p/NKRF/NF-κB pathway as a microRNA sponge. Furthermore, In vivo experiments showed that exogenous ICL effectively inhibited the growth of patient-derived subcutaneous tumor xenografts (PDX) of LC patients with COPD and significantly prolonged the survival time of tumor-bearing mice. In a word, our study shows that the decrease of ICL is associated with an increased risk of LC in patients with COPD, ICL is not only expected to be a new therapeutic target for LC in COPD patients, but also has great potential to be used as a new marker for evaluating the occurrence, severity stratification and prognosis of LC in patients with COPD. BioMed Central 2023-04-03 /pmc/articles/PMC10071758/ /pubmed/37013587 http://dx.doi.org/10.1186/s12935-023-02900-2 Text en © The Author(s) 2023 https://creativecommons.org/licenses/by/4.0/Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/ (https://creativecommons.org/licenses/by/4.0/) . The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/ (https://creativecommons.org/publicdomain/zero/1.0/) ) applies to the data made available in this article, unless otherwise stated in a credit line to the data.
spellingShingle Research
Lu, Jingjing
Shi, Yan
Zhang, Feng
Zhang, Ying
Zhao, Xiangwang
Zheng, Haiyan
Li, Lingyu
Zhao, Shiqiao
Zhao, Liming
Mechanism of lnRNA-ICL involved in lung cancer development in COPD patients through modulating microRNA-19-3p/NKRF/NF-κB axis
title Mechanism of lnRNA-ICL involved in lung cancer development in COPD patients through modulating microRNA-19-3p/NKRF/NF-κB axis
title_full Mechanism of lnRNA-ICL involved in lung cancer development in COPD patients through modulating microRNA-19-3p/NKRF/NF-κB axis
title_fullStr Mechanism of lnRNA-ICL involved in lung cancer development in COPD patients through modulating microRNA-19-3p/NKRF/NF-κB axis
title_full_unstemmed Mechanism of lnRNA-ICL involved in lung cancer development in COPD patients through modulating microRNA-19-3p/NKRF/NF-κB axis
title_short Mechanism of lnRNA-ICL involved in lung cancer development in COPD patients through modulating microRNA-19-3p/NKRF/NF-κB axis
title_sort mechanism of lnrna-icl involved in lung cancer development in copd patients through modulating microrna-19-3p/nkrf/nf-κb axis
topic Research
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10071758/
https://www.ncbi.nlm.nih.gov/pubmed/37013587
http://dx.doi.org/10.1186/s12935-023-02900-2
work_keys_str_mv AT lujingjing mechanismoflnrnaiclinvolvedinlungcancerdevelopmentincopdpatientsthroughmodulatingmicrorna193pnkrfnfkbaxis
AT shiyan mechanismoflnrnaiclinvolvedinlungcancerdevelopmentincopdpatientsthroughmodulatingmicrorna193pnkrfnfkbaxis
AT zhangfeng mechanismoflnrnaiclinvolvedinlungcancerdevelopmentincopdpatientsthroughmodulatingmicrorna193pnkrfnfkbaxis
AT zhangying mechanismoflnrnaiclinvolvedinlungcancerdevelopmentincopdpatientsthroughmodulatingmicrorna193pnkrfnfkbaxis
AT zhaoxiangwang mechanismoflnrnaiclinvolvedinlungcancerdevelopmentincopdpatientsthroughmodulatingmicrorna193pnkrfnfkbaxis
AT zhenghaiyan mechanismoflnrnaiclinvolvedinlungcancerdevelopmentincopdpatientsthroughmodulatingmicrorna193pnkrfnfkbaxis
AT lilingyu mechanismoflnrnaiclinvolvedinlungcancerdevelopmentincopdpatientsthroughmodulatingmicrorna193pnkrfnfkbaxis
AT zhaoshiqiao mechanismoflnrnaiclinvolvedinlungcancerdevelopmentincopdpatientsthroughmodulatingmicrorna193pnkrfnfkbaxis
AT zhaoliming mechanismoflnrnaiclinvolvedinlungcancerdevelopmentincopdpatientsthroughmodulatingmicrorna193pnkrfnfkbaxis