Cargando…

Primary cilia regulate gastric cancer-induced bone loss via cilia/Wnt/β-catenin signaling pathway

Cancer-associated bone disease is a frequent occurrence in cancer patients and is associated with pain, bone fragility, loss, and fractures. However, whether primary or non-bone metastatic gastric cancer induces bone loss remains unclear. Here, we collected clinical evidence of bone loss by analyzin...

Descripción completa

Detalles Bibliográficos
Autores principales: Xu, Jie, Deng, Xiaoyan, Wu, Xiangmei, Zhu, Huifang, Zhu, Yinghua, Liu, Jie, Chen, Qian, Yuan, Chengfu, Liu, Geli, Wang, Changdong
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Impact Journals 2021
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8034975/
https://www.ncbi.nlm.nih.gov/pubmed/33690174
http://dx.doi.org/10.18632/aging.202734
_version_ 1783676632535400448
author Xu, Jie
Deng, Xiaoyan
Wu, Xiangmei
Zhu, Huifang
Zhu, Yinghua
Liu, Jie
Chen, Qian
Yuan, Chengfu
Liu, Geli
Wang, Changdong
author_facet Xu, Jie
Deng, Xiaoyan
Wu, Xiangmei
Zhu, Huifang
Zhu, Yinghua
Liu, Jie
Chen, Qian
Yuan, Chengfu
Liu, Geli
Wang, Changdong
author_sort Xu, Jie
collection PubMed
description Cancer-associated bone disease is a frequent occurrence in cancer patients and is associated with pain, bone fragility, loss, and fractures. However, whether primary or non-bone metastatic gastric cancer induces bone loss remains unclear. Here, we collected clinical evidence of bone loss by analyzing serum and X-rays of 25 non-bone metastatic gastric cancer patients. In addition, C57BL mice were injected with the human gastric cancer cell line HGC27 and its effect on bone mass was analyzed by Micro-CT, immunoblotting, and immunohistochemistry. Furthermore, the degree of the proliferation and osteogenic differentiation of mesenchymal stem cells (MSCs) co-cultured with HGC-27 or SGC-7901 cells was analyzed by colony-formation assay, alizarin red staining, immunofluorescence, qPCR, immunoblotting, and alkaline phosphatase activity assay. These indicated that gastric cancer could damage bone tissue before the occurrence of bone metastases. We also found that cilia formation of MSCs was increased in the presence of HGC27 cells, which was associated with abnormal activation of the Wnt/β-catenin pathway. Expression of DKK1 inhibited the Wnt/β-catenin signaling pathway and partially rescued osteogenic differentiation of MSCs. In summary, our results suggest that gastric cancer cells might cause bone damage prior to the occurrence of bone metastasis via cilia-dependent activation of the Wnt/β-catenin signaling pathway.
format Online
Article
Text
id pubmed-8034975
institution National Center for Biotechnology Information
language English
publishDate 2021
publisher Impact Journals
record_format MEDLINE/PubMed
spelling pubmed-80349752021-04-16 Primary cilia regulate gastric cancer-induced bone loss via cilia/Wnt/β-catenin signaling pathway Xu, Jie Deng, Xiaoyan Wu, Xiangmei Zhu, Huifang Zhu, Yinghua Liu, Jie Chen, Qian Yuan, Chengfu Liu, Geli Wang, Changdong Aging (Albany NY) Research Paper Cancer-associated bone disease is a frequent occurrence in cancer patients and is associated with pain, bone fragility, loss, and fractures. However, whether primary or non-bone metastatic gastric cancer induces bone loss remains unclear. Here, we collected clinical evidence of bone loss by analyzing serum and X-rays of 25 non-bone metastatic gastric cancer patients. In addition, C57BL mice were injected with the human gastric cancer cell line HGC27 and its effect on bone mass was analyzed by Micro-CT, immunoblotting, and immunohistochemistry. Furthermore, the degree of the proliferation and osteogenic differentiation of mesenchymal stem cells (MSCs) co-cultured with HGC-27 or SGC-7901 cells was analyzed by colony-formation assay, alizarin red staining, immunofluorescence, qPCR, immunoblotting, and alkaline phosphatase activity assay. These indicated that gastric cancer could damage bone tissue before the occurrence of bone metastases. We also found that cilia formation of MSCs was increased in the presence of HGC27 cells, which was associated with abnormal activation of the Wnt/β-catenin pathway. Expression of DKK1 inhibited the Wnt/β-catenin signaling pathway and partially rescued osteogenic differentiation of MSCs. In summary, our results suggest that gastric cancer cells might cause bone damage prior to the occurrence of bone metastasis via cilia-dependent activation of the Wnt/β-catenin signaling pathway. Impact Journals 2021-03-09 /pmc/articles/PMC8034975/ /pubmed/33690174 http://dx.doi.org/10.18632/aging.202734 Text en Copyright: © 2021 Xu et al. https://creativecommons.org/licenses/by/3.0/This is an open access article distributed under the terms of the Creative Commons Attribution License (https://creativecommons.org/licenses/by/3.0/) (CC BY 3.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
spellingShingle Research Paper
Xu, Jie
Deng, Xiaoyan
Wu, Xiangmei
Zhu, Huifang
Zhu, Yinghua
Liu, Jie
Chen, Qian
Yuan, Chengfu
Liu, Geli
Wang, Changdong
Primary cilia regulate gastric cancer-induced bone loss via cilia/Wnt/β-catenin signaling pathway
title Primary cilia regulate gastric cancer-induced bone loss via cilia/Wnt/β-catenin signaling pathway
title_full Primary cilia regulate gastric cancer-induced bone loss via cilia/Wnt/β-catenin signaling pathway
title_fullStr Primary cilia regulate gastric cancer-induced bone loss via cilia/Wnt/β-catenin signaling pathway
title_full_unstemmed Primary cilia regulate gastric cancer-induced bone loss via cilia/Wnt/β-catenin signaling pathway
title_short Primary cilia regulate gastric cancer-induced bone loss via cilia/Wnt/β-catenin signaling pathway
title_sort primary cilia regulate gastric cancer-induced bone loss via cilia/wnt/β-catenin signaling pathway
topic Research Paper
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8034975/
https://www.ncbi.nlm.nih.gov/pubmed/33690174
http://dx.doi.org/10.18632/aging.202734
work_keys_str_mv AT xujie primaryciliaregulategastriccancerinducedbonelossviaciliawntbcateninsignalingpathway
AT dengxiaoyan primaryciliaregulategastriccancerinducedbonelossviaciliawntbcateninsignalingpathway
AT wuxiangmei primaryciliaregulategastriccancerinducedbonelossviaciliawntbcateninsignalingpathway
AT zhuhuifang primaryciliaregulategastriccancerinducedbonelossviaciliawntbcateninsignalingpathway
AT zhuyinghua primaryciliaregulategastriccancerinducedbonelossviaciliawntbcateninsignalingpathway
AT liujie primaryciliaregulategastriccancerinducedbonelossviaciliawntbcateninsignalingpathway
AT chenqian primaryciliaregulategastriccancerinducedbonelossviaciliawntbcateninsignalingpathway
AT yuanchengfu primaryciliaregulategastriccancerinducedbonelossviaciliawntbcateninsignalingpathway
AT liugeli primaryciliaregulategastriccancerinducedbonelossviaciliawntbcateninsignalingpathway
AT wangchangdong primaryciliaregulategastriccancerinducedbonelossviaciliawntbcateninsignalingpathway