Cargando…

XIAOPI Formula Inhibits Breast Cancer Stem Cells via Suppressing Tumor-Associated Macrophages/C-X-C Motif Chemokine Ligand 1 Pathway

Macrophages are the most abundant stromal cells associated with the host immune system in multiple malignancies including breast cancer. With proven clinical efficacy and no noticeable adverse effects, XIAOPI formula (XPS) has been approved for breast hyperplasia treatment by the State Food and Drug...

Descripción completa

Detalles Bibliográficos
Autores principales: Wang, Shengqi, Liu, Xiaoyan, Huang, Renlun, Zheng, Yifeng, Wang, Neng, Yang, Bowen, Situ, Honglin, Lin, Yi, Wang, Zhiyu
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Frontiers Media S.A. 2019
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6874098/
https://www.ncbi.nlm.nih.gov/pubmed/31803057
http://dx.doi.org/10.3389/fphar.2019.01371
_version_ 1783472775336296448
author Wang, Shengqi
Liu, Xiaoyan
Huang, Renlun
Zheng, Yifeng
Wang, Neng
Yang, Bowen
Situ, Honglin
Lin, Yi
Wang, Zhiyu
author_facet Wang, Shengqi
Liu, Xiaoyan
Huang, Renlun
Zheng, Yifeng
Wang, Neng
Yang, Bowen
Situ, Honglin
Lin, Yi
Wang, Zhiyu
author_sort Wang, Shengqi
collection PubMed
description Macrophages are the most abundant stromal cells associated with the host immune system in multiple malignancies including breast cancer. With proven clinical efficacy and no noticeable adverse effects, XIAOPI formula (XPS) has been approved for breast hyperplasia treatment by the State Food and Drug Administration of China (SFDA) in 2018. The existing knowledge about the anti-breast cancer activities and mechanisms of XPS has been very limited. The present study aimed to investigate whether XPS could exert an anti-breast cancer effect by regulating tumor-associated macrophages (TAMs) in tumor microenvironment. Herein, breast cancer cells and TAMs were co-cultured using the transwell co-culture system to simulate the coexistence of them. XPS could significantly inhibit the proliferation, colony formation, breast cancer stem cells (CSCs) subpopulation, mammosphere formation abilities as well as stemness-related genes expression in both human and mouse breast cancer cells in the co-culture system. Additionally, XPS could suppress M2 phenotype polarization as well as C-X-C motif chemokine ligand 1 (CXCL1) expression and secretion of TAMs. Notably, further mechanistic explorations verified TAMs/CXCL1 as the critical target of XPS in inhibiting breast CSCs self-renewal in the co-culture system as the exogenous CXCL1 administration could abrogate the inhibitory effect of XPS on breast CSCs self-renewal. More importantly, XPS significantly inhibited mammary tumor growth, breast CSCs subpopulation, and TAMs/CXCL1 activity in mouse 4T1-Luc xenografts in vivo without any detectable side effects. Taken together, this study not only uncovers the immunomodulatory mechanism of XPS in treating breast cancer but also sheds novel insights into TAMs/CXCL1 as a potential molecular target for breast CSCs elimination.
format Online
Article
Text
id pubmed-6874098
institution National Center for Biotechnology Information
language English
publishDate 2019
publisher Frontiers Media S.A.
record_format MEDLINE/PubMed
spelling pubmed-68740982019-12-04 XIAOPI Formula Inhibits Breast Cancer Stem Cells via Suppressing Tumor-Associated Macrophages/C-X-C Motif Chemokine Ligand 1 Pathway Wang, Shengqi Liu, Xiaoyan Huang, Renlun Zheng, Yifeng Wang, Neng Yang, Bowen Situ, Honglin Lin, Yi Wang, Zhiyu Front Pharmacol Pharmacology Macrophages are the most abundant stromal cells associated with the host immune system in multiple malignancies including breast cancer. With proven clinical efficacy and no noticeable adverse effects, XIAOPI formula (XPS) has been approved for breast hyperplasia treatment by the State Food and Drug Administration of China (SFDA) in 2018. The existing knowledge about the anti-breast cancer activities and mechanisms of XPS has been very limited. The present study aimed to investigate whether XPS could exert an anti-breast cancer effect by regulating tumor-associated macrophages (TAMs) in tumor microenvironment. Herein, breast cancer cells and TAMs were co-cultured using the transwell co-culture system to simulate the coexistence of them. XPS could significantly inhibit the proliferation, colony formation, breast cancer stem cells (CSCs) subpopulation, mammosphere formation abilities as well as stemness-related genes expression in both human and mouse breast cancer cells in the co-culture system. Additionally, XPS could suppress M2 phenotype polarization as well as C-X-C motif chemokine ligand 1 (CXCL1) expression and secretion of TAMs. Notably, further mechanistic explorations verified TAMs/CXCL1 as the critical target of XPS in inhibiting breast CSCs self-renewal in the co-culture system as the exogenous CXCL1 administration could abrogate the inhibitory effect of XPS on breast CSCs self-renewal. More importantly, XPS significantly inhibited mammary tumor growth, breast CSCs subpopulation, and TAMs/CXCL1 activity in mouse 4T1-Luc xenografts in vivo without any detectable side effects. Taken together, this study not only uncovers the immunomodulatory mechanism of XPS in treating breast cancer but also sheds novel insights into TAMs/CXCL1 as a potential molecular target for breast CSCs elimination. Frontiers Media S.A. 2019-11-15 /pmc/articles/PMC6874098/ /pubmed/31803057 http://dx.doi.org/10.3389/fphar.2019.01371 Text en Copyright © 2019 Wang, Liu, Huang, Zheng, Wang, Yang, Situ, Lin and Wang http://creativecommons.org/licenses/by/4.0/ This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.
spellingShingle Pharmacology
Wang, Shengqi
Liu, Xiaoyan
Huang, Renlun
Zheng, Yifeng
Wang, Neng
Yang, Bowen
Situ, Honglin
Lin, Yi
Wang, Zhiyu
XIAOPI Formula Inhibits Breast Cancer Stem Cells via Suppressing Tumor-Associated Macrophages/C-X-C Motif Chemokine Ligand 1 Pathway
title XIAOPI Formula Inhibits Breast Cancer Stem Cells via Suppressing Tumor-Associated Macrophages/C-X-C Motif Chemokine Ligand 1 Pathway
title_full XIAOPI Formula Inhibits Breast Cancer Stem Cells via Suppressing Tumor-Associated Macrophages/C-X-C Motif Chemokine Ligand 1 Pathway
title_fullStr XIAOPI Formula Inhibits Breast Cancer Stem Cells via Suppressing Tumor-Associated Macrophages/C-X-C Motif Chemokine Ligand 1 Pathway
title_full_unstemmed XIAOPI Formula Inhibits Breast Cancer Stem Cells via Suppressing Tumor-Associated Macrophages/C-X-C Motif Chemokine Ligand 1 Pathway
title_short XIAOPI Formula Inhibits Breast Cancer Stem Cells via Suppressing Tumor-Associated Macrophages/C-X-C Motif Chemokine Ligand 1 Pathway
title_sort xiaopi formula inhibits breast cancer stem cells via suppressing tumor-associated macrophages/c-x-c motif chemokine ligand 1 pathway
topic Pharmacology
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6874098/
https://www.ncbi.nlm.nih.gov/pubmed/31803057
http://dx.doi.org/10.3389/fphar.2019.01371
work_keys_str_mv AT wangshengqi xiaopiformulainhibitsbreastcancerstemcellsviasuppressingtumorassociatedmacrophagescxcmotifchemokineligand1pathway
AT liuxiaoyan xiaopiformulainhibitsbreastcancerstemcellsviasuppressingtumorassociatedmacrophagescxcmotifchemokineligand1pathway
AT huangrenlun xiaopiformulainhibitsbreastcancerstemcellsviasuppressingtumorassociatedmacrophagescxcmotifchemokineligand1pathway
AT zhengyifeng xiaopiformulainhibitsbreastcancerstemcellsviasuppressingtumorassociatedmacrophagescxcmotifchemokineligand1pathway
AT wangneng xiaopiformulainhibitsbreastcancerstemcellsviasuppressingtumorassociatedmacrophagescxcmotifchemokineligand1pathway
AT yangbowen xiaopiformulainhibitsbreastcancerstemcellsviasuppressingtumorassociatedmacrophagescxcmotifchemokineligand1pathway
AT situhonglin xiaopiformulainhibitsbreastcancerstemcellsviasuppressingtumorassociatedmacrophagescxcmotifchemokineligand1pathway
AT linyi xiaopiformulainhibitsbreastcancerstemcellsviasuppressingtumorassociatedmacrophagescxcmotifchemokineligand1pathway
AT wangzhiyu xiaopiformulainhibitsbreastcancerstemcellsviasuppressingtumorassociatedmacrophagescxcmotifchemokineligand1pathway