Bone marrow-derived mesenchymal stem cells promote colorectal cancer progression through paracrine neuregulin 1/HER3 signalling

Gut ◽  
2012 ◽  
Vol 62 (4) ◽  
pp. 550-560 ◽  
Author(s):  
Astrid De Boeck ◽  
Patrick Pauwels ◽  
Karen Hensen ◽  
Jean-Luc Rummens ◽  
Wendy Westbroek ◽  
...  
2019 ◽  
Vol 10 (4) ◽  
Author(s):  
Gen Nishikawa ◽  
Kenji Kawada ◽  
Jun Nakagawa ◽  
Kosuke Toda ◽  
Ryotaro Ogawa ◽  
...  

2016 ◽  
Vol 6 (1) ◽  
Author(s):  
Xiao-Bing Wu ◽  
Yang Liu ◽  
Gui-Hua Wang ◽  
Xiao Xu ◽  
Yang Cai ◽  
...  

Oncogene ◽  
2021 ◽  
Author(s):  
Kathleen M. McAndrews ◽  
Karina Vázquez-Arreguín ◽  
Changsoo Kwak ◽  
Hikaru Sugimoto ◽  
Xiaofeng Zheng ◽  
...  

Author(s):  
A. Thirumal Raj ◽  
Supriya Kheur ◽  
Ramesh Bhonde ◽  
Archana A. Gupta ◽  
Vikrant R. Patil ◽  
...  

Author(s):  
Ruohang He ◽  
Chaoqun Han ◽  
Ying Li ◽  
Wei Qian ◽  
Xiaohua Hou

BackgroundMesenchymal stem cells (MSCs) treatment showed promising results in inflammatory bowel disease in both rodent models and patients. Nevertheless, previous studies conducted conflicting results on preclinical tumor models treated with MSCs concerning their influence on tumor initiation and progression. This study is designed to demonstrate the role of bone marrow-derived MSCs and the potential mechanism in the colitis-associated colon cancer (CAC) model.MethodsBone marrow-derived MSCs were isolated from green fluorescent protein-transgenic mice, cultured, and identified by flow cytometry. Azoxymethane and dextran sulfate sodium were administrated to establish the CAC mouse model, and MSCs were infused intraperitoneally once per week. The mice were weighed weekly, and colon length, tumor number, and average tumor size were assessed after the mice were killed. MSC localization was detected by immunofluorescence staining; tumor cell proliferation and apoptosis were measured by immunohistochemistry staining of Ki-67 and terminal deoxynucleotidyl transferase deoxyuridine triphosphate nick end labeling assay, respectively. The colonic tumor tissues were isolated for RNA-seq, and fecal samples were collected for 16S ribosomal RNA sequencing of the microbiome.ResultsAfter injection intraperitoneally, MSCs migrated to the intestine and inhibited the initiation of colitis-associated colorectal cancer. This inhibition effect was marked by less weight loss, longer colon length, and reduced tumor numbers. Moreover, MSCs reduced tumor cell proliferation and induced tumor cell apoptosis. Furthermore, MSCs could inhibit chronic inflammation assessed by RNA-sequencing and promote gut microbiome normalization detected by 16S ribosomal RNA sequencing.ConclusionThe results proved that MSCs could migrate to the colon, inhibit chronic inflammation, and regulate gut microbiome dysbiosis to suppress the development of CAC.


Sign in / Sign up

Export Citation Format

Share Document