Cargando…

Rocuronium bromide suppresses esophageal cancer via blocking the secretion of C–X–C motif chemokine ligand 12 from cancer associated fibroblasts

BACKGROUND: Cancer associated fibroblasts (CAFs) communicate metabolically with tumor genesis and development. Rocuronium bromide (RB) is reported to exert certain inhibitory effect on tumor. Here, we investigate the role of RB in esophageal cancer (EC) malignant progression. METHODS: Tumor xenograf...

Descripción completa

Detalles Bibliográficos
Autores principales: Li, Jingyi, Gu, Xuefeng, Wan, Guoqing, Wang, Yuhan, Chen, Kaijie, Chen, Qi, Lu, Changlian
Formato: Online Artículo Texto
Lenguaje:English
Publicado: BioMed Central 2023
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10082495/
https://www.ncbi.nlm.nih.gov/pubmed/37029408
http://dx.doi.org/10.1186/s12967-023-04081-y
_version_ 1785021325046185984
author Li, Jingyi
Gu, Xuefeng
Wan, Guoqing
Wang, Yuhan
Chen, Kaijie
Chen, Qi
Lu, Changlian
author_facet Li, Jingyi
Gu, Xuefeng
Wan, Guoqing
Wang, Yuhan
Chen, Kaijie
Chen, Qi
Lu, Changlian
author_sort Li, Jingyi
collection PubMed
description BACKGROUND: Cancer associated fibroblasts (CAFs) communicate metabolically with tumor genesis and development. Rocuronium bromide (RB) is reported to exert certain inhibitory effect on tumor. Here, we investigate the role of RB in esophageal cancer (EC) malignant progression. METHODS: Tumor xenograft models with EC cells were locally and systemically administrated with RB to detect the influence of different administrations on tumor progression. Mouse CAFs PDGFRα(+)/F4/80(−) were sorted by Flow cytometry with specific antibodies. CAFs were treated with RB and co-cultured with EC cells. The proliferation, invasion and apoptosis assays of EC cells were performed to detect the influences of RB targeting CAFs on EC cell malignant progression. Human fibroblasts were employed to perform these detections to confirm RB indirect effect on EC cells. The gene expression changes of CAFs response to RB treatment were detected using RNA sequencing and verified by Western blot, immunohistochemistry and ELISA. RESULTS: Tumors in xenograft mice were observed significantly inhibited by local RB administration, but not by systemic administration. Moreover EC cells did not show obvious change in viability when direct stimulated with RB in vitro. However, when CAFs treated with RB were co-cultured with EC cells, obvious suppressions were observed in EC cell malignancy, including proliferation, invasion and apoptosis. Human fibroblasts were employed to perform these assays and similar results were obtained. RNA sequencing data of human fibroblast treated with RB, and Western blot, immunohistochemistry and ELISA results all showed that CXCL12 expression was significantly diminished in vivo and in vitro by RB. EC cells direct treated with CXCL12 showed much higher malignancy. Moreover cell autophagy and PI3K/AKT/mTOR signaling pathway in CAFs were both suppressed by RB which can be reversed by Rapamycin pretreatment. CONCLUSIONS: Our data suggest that RB could repress PI3K/AKT/mTOR signaling pathway and autophagy to block the CXCL12 expression in CAFs, thereby weakening the CXCL12-mediated EC tumor progression. Our data provide a novel insight into the underlying mechanism of RB inhibiting EC, and emphasize the importance of tumor microenvironment (cytokines from CAFs) in modulating cancer malignant progression. SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1186/s12967-023-04081-y.
format Online
Article
Text
id pubmed-10082495
institution National Center for Biotechnology Information
language English
publishDate 2023
publisher BioMed Central
record_format MEDLINE/PubMed
spelling pubmed-100824952023-04-09 Rocuronium bromide suppresses esophageal cancer via blocking the secretion of C–X–C motif chemokine ligand 12 from cancer associated fibroblasts Li, Jingyi Gu, Xuefeng Wan, Guoqing Wang, Yuhan Chen, Kaijie Chen, Qi Lu, Changlian J Transl Med Research BACKGROUND: Cancer associated fibroblasts (CAFs) communicate metabolically with tumor genesis and development. Rocuronium bromide (RB) is reported to exert certain inhibitory effect on tumor. Here, we investigate the role of RB in esophageal cancer (EC) malignant progression. METHODS: Tumor xenograft models with EC cells were locally and systemically administrated with RB to detect the influence of different administrations on tumor progression. Mouse CAFs PDGFRα(+)/F4/80(−) were sorted by Flow cytometry with specific antibodies. CAFs were treated with RB and co-cultured with EC cells. The proliferation, invasion and apoptosis assays of EC cells were performed to detect the influences of RB targeting CAFs on EC cell malignant progression. Human fibroblasts were employed to perform these detections to confirm RB indirect effect on EC cells. The gene expression changes of CAFs response to RB treatment were detected using RNA sequencing and verified by Western blot, immunohistochemistry and ELISA. RESULTS: Tumors in xenograft mice were observed significantly inhibited by local RB administration, but not by systemic administration. Moreover EC cells did not show obvious change in viability when direct stimulated with RB in vitro. However, when CAFs treated with RB were co-cultured with EC cells, obvious suppressions were observed in EC cell malignancy, including proliferation, invasion and apoptosis. Human fibroblasts were employed to perform these assays and similar results were obtained. RNA sequencing data of human fibroblast treated with RB, and Western blot, immunohistochemistry and ELISA results all showed that CXCL12 expression was significantly diminished in vivo and in vitro by RB. EC cells direct treated with CXCL12 showed much higher malignancy. Moreover cell autophagy and PI3K/AKT/mTOR signaling pathway in CAFs were both suppressed by RB which can be reversed by Rapamycin pretreatment. CONCLUSIONS: Our data suggest that RB could repress PI3K/AKT/mTOR signaling pathway and autophagy to block the CXCL12 expression in CAFs, thereby weakening the CXCL12-mediated EC tumor progression. Our data provide a novel insight into the underlying mechanism of RB inhibiting EC, and emphasize the importance of tumor microenvironment (cytokines from CAFs) in modulating cancer malignant progression. SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1186/s12967-023-04081-y. BioMed Central 2023-04-08 /pmc/articles/PMC10082495/ /pubmed/37029408 http://dx.doi.org/10.1186/s12967-023-04081-y 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
Li, Jingyi
Gu, Xuefeng
Wan, Guoqing
Wang, Yuhan
Chen, Kaijie
Chen, Qi
Lu, Changlian
Rocuronium bromide suppresses esophageal cancer via blocking the secretion of C–X–C motif chemokine ligand 12 from cancer associated fibroblasts
title Rocuronium bromide suppresses esophageal cancer via blocking the secretion of C–X–C motif chemokine ligand 12 from cancer associated fibroblasts
title_full Rocuronium bromide suppresses esophageal cancer via blocking the secretion of C–X–C motif chemokine ligand 12 from cancer associated fibroblasts
title_fullStr Rocuronium bromide suppresses esophageal cancer via blocking the secretion of C–X–C motif chemokine ligand 12 from cancer associated fibroblasts
title_full_unstemmed Rocuronium bromide suppresses esophageal cancer via blocking the secretion of C–X–C motif chemokine ligand 12 from cancer associated fibroblasts
title_short Rocuronium bromide suppresses esophageal cancer via blocking the secretion of C–X–C motif chemokine ligand 12 from cancer associated fibroblasts
title_sort rocuronium bromide suppresses esophageal cancer via blocking the secretion of c–x–c motif chemokine ligand 12 from cancer associated fibroblasts
topic Research
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10082495/
https://www.ncbi.nlm.nih.gov/pubmed/37029408
http://dx.doi.org/10.1186/s12967-023-04081-y
work_keys_str_mv AT lijingyi rocuroniumbromidesuppressesesophagealcancerviablockingthesecretionofcxcmotifchemokineligand12fromcancerassociatedfibroblasts
AT guxuefeng rocuroniumbromidesuppressesesophagealcancerviablockingthesecretionofcxcmotifchemokineligand12fromcancerassociatedfibroblasts
AT wanguoqing rocuroniumbromidesuppressesesophagealcancerviablockingthesecretionofcxcmotifchemokineligand12fromcancerassociatedfibroblasts
AT wangyuhan rocuroniumbromidesuppressesesophagealcancerviablockingthesecretionofcxcmotifchemokineligand12fromcancerassociatedfibroblasts
AT chenkaijie rocuroniumbromidesuppressesesophagealcancerviablockingthesecretionofcxcmotifchemokineligand12fromcancerassociatedfibroblasts
AT chenqi rocuroniumbromidesuppressesesophagealcancerviablockingthesecretionofcxcmotifchemokineligand12fromcancerassociatedfibroblasts
AT luchanglian rocuroniumbromidesuppressesesophagealcancerviablockingthesecretionofcxcmotifchemokineligand12fromcancerassociatedfibroblasts