Cargando…

Dexmedetomidine enhances hypoxia-induced cancer cell progression

Dexmedetomidine (DEX) is widely used in perioperative settings for analgesia and sedation; however, little is known about its effects on the hypoxia-induced progression of tumor cells. In the present study, the effects of DEX on hypoxia-induced growth and metastasis of lung cancer cells and colorect...

Descripción completa

Detalles Bibliográficos
Autores principales: Chen, Hua Yan, Li, Geng Hua, Tan, Guo Cheng, Liang, Hua, Lai, Xiao Hong, Huang, Qiong, Zhong, Ji Ying
Formato: Online Artículo Texto
Lenguaje:English
Publicado: D.A. Spandidos 2019
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6861874/
https://www.ncbi.nlm.nih.gov/pubmed/31772647
http://dx.doi.org/10.3892/etm.2019.8136
_version_ 1783471416076664832
author Chen, Hua Yan
Li, Geng Hua
Tan, Guo Cheng
Liang, Hua
Lai, Xiao Hong
Huang, Qiong
Zhong, Ji Ying
author_facet Chen, Hua Yan
Li, Geng Hua
Tan, Guo Cheng
Liang, Hua
Lai, Xiao Hong
Huang, Qiong
Zhong, Ji Ying
author_sort Chen, Hua Yan
collection PubMed
description Dexmedetomidine (DEX) is widely used in perioperative settings for analgesia and sedation; however, little is known about its effects on the hypoxia-induced progression of tumor cells. In the present study, the effects of DEX on hypoxia-induced growth and metastasis of lung cancer cells and colorectal cancer cells was examined. A549 cells and HCT116 cells were treated with normoxia, hypoxia, co-treatment of hypoxia and DEX, and atipamezole (an α(2) adrenoceptor antagonist) for 4 h. The proliferation rate of cells was determined by MTT assays. Cell metastatic potential was evaluated by Transwell assays. Survivin and hypoxia inducible factor (HIF)-1α were detected by western blotting. Matrix metalloproteinase (MMP)-2 and MMP-9 were measured using reverse transcription-quantitative PCR. It was demonstrated that hypoxia treatment promoted the proliferation and may promote the metastasis of the two cancer cell lines. DEX substantially contributed to the survival and aggressiveness of the two cancer cell lines following hypoxia. Furthermore, DEX upregulated the expression of survivin, MMP-2, MMP-9 and HIF-1α in the two cancer cell lines in response to hypoxia. Finally, the effects of DEX on the hypoxia-induced growth and metastatic potential of cancer cells were reversed by atipamezole. Collectively, DEX enhances the hypoxia-induced progression of lung cancer cells and colorectal cancer cells by regulating HIF-1α signaling, which may be associated with the α(2) adrenoceptor pathway.
format Online
Article
Text
id pubmed-6861874
institution National Center for Biotechnology Information
language English
publishDate 2019
publisher D.A. Spandidos
record_format MEDLINE/PubMed
spelling pubmed-68618742019-11-26 Dexmedetomidine enhances hypoxia-induced cancer cell progression Chen, Hua Yan Li, Geng Hua Tan, Guo Cheng Liang, Hua Lai, Xiao Hong Huang, Qiong Zhong, Ji Ying Exp Ther Med Articles Dexmedetomidine (DEX) is widely used in perioperative settings for analgesia and sedation; however, little is known about its effects on the hypoxia-induced progression of tumor cells. In the present study, the effects of DEX on hypoxia-induced growth and metastasis of lung cancer cells and colorectal cancer cells was examined. A549 cells and HCT116 cells were treated with normoxia, hypoxia, co-treatment of hypoxia and DEX, and atipamezole (an α(2) adrenoceptor antagonist) for 4 h. The proliferation rate of cells was determined by MTT assays. Cell metastatic potential was evaluated by Transwell assays. Survivin and hypoxia inducible factor (HIF)-1α were detected by western blotting. Matrix metalloproteinase (MMP)-2 and MMP-9 were measured using reverse transcription-quantitative PCR. It was demonstrated that hypoxia treatment promoted the proliferation and may promote the metastasis of the two cancer cell lines. DEX substantially contributed to the survival and aggressiveness of the two cancer cell lines following hypoxia. Furthermore, DEX upregulated the expression of survivin, MMP-2, MMP-9 and HIF-1α in the two cancer cell lines in response to hypoxia. Finally, the effects of DEX on the hypoxia-induced growth and metastatic potential of cancer cells were reversed by atipamezole. Collectively, DEX enhances the hypoxia-induced progression of lung cancer cells and colorectal cancer cells by regulating HIF-1α signaling, which may be associated with the α(2) adrenoceptor pathway. D.A. Spandidos 2019-12 2019-10-25 /pmc/articles/PMC6861874/ /pubmed/31772647 http://dx.doi.org/10.3892/etm.2019.8136 Text en Copyright: © Chen et al. This is an open access article distributed under the terms of the Creative Commons Attribution-NonCommercial-NoDerivs License (https://creativecommons.org/licenses/by-nc-nd/4.0/) , which permits use and distribution in any medium, provided the original work is properly cited, the use is non-commercial and no modifications or adaptations are made.
spellingShingle Articles
Chen, Hua Yan
Li, Geng Hua
Tan, Guo Cheng
Liang, Hua
Lai, Xiao Hong
Huang, Qiong
Zhong, Ji Ying
Dexmedetomidine enhances hypoxia-induced cancer cell progression
title Dexmedetomidine enhances hypoxia-induced cancer cell progression
title_full Dexmedetomidine enhances hypoxia-induced cancer cell progression
title_fullStr Dexmedetomidine enhances hypoxia-induced cancer cell progression
title_full_unstemmed Dexmedetomidine enhances hypoxia-induced cancer cell progression
title_short Dexmedetomidine enhances hypoxia-induced cancer cell progression
title_sort dexmedetomidine enhances hypoxia-induced cancer cell progression
topic Articles
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6861874/
https://www.ncbi.nlm.nih.gov/pubmed/31772647
http://dx.doi.org/10.3892/etm.2019.8136
work_keys_str_mv AT chenhuayan dexmedetomidineenhanceshypoxiainducedcancercellprogression
AT ligenghua dexmedetomidineenhanceshypoxiainducedcancercellprogression
AT tanguocheng dexmedetomidineenhanceshypoxiainducedcancercellprogression
AT lianghua dexmedetomidineenhanceshypoxiainducedcancercellprogression
AT laixiaohong dexmedetomidineenhanceshypoxiainducedcancercellprogression
AT huangqiong dexmedetomidineenhanceshypoxiainducedcancercellprogression
AT zhongjiying dexmedetomidineenhanceshypoxiainducedcancercellprogression