scholarly journals Cytochrome C Oxidase Assembly Factor 1 Homolog Predicts Poor Prognosis and Promotes Cell Proliferation in Colorectal Cancer by Regulating PI3K/AKT Signaling

2020 ◽  
Vol Volume 13 ◽  
pp. 11505-11516
Author(s):  
Yuan Xue ◽  
Pei-Dong Li ◽  
Xue-Mei Tang ◽  
Zai-Hua Yan ◽  
Shu-Sen Xia ◽  
...  
Life Sciences ◽  
2014 ◽  
Vol 103 (1) ◽  
pp. 1-7 ◽  
Author(s):  
Yasuhiro Katou ◽  
Naoya Endo ◽  
Toshiyuki Suzuki ◽  
Jiang Yu ◽  
Haruhisa Kikuchi ◽  
...  

2012 ◽  
Vol 1817 ◽  
pp. S70
Author(s):  
S. Schimo ◽  
A. Hannappel ◽  
F.A. Bundschuh ◽  
C. Glaubnitz ◽  
K.M. Pos ◽  
...  

2014 ◽  
Vol 8 (9) ◽  
pp. 740-754 ◽  
Author(s):  
Cleber Ferraresi ◽  
Nivaldo Antonio Parizotto ◽  
Marcelo Victor Pires de Sousa ◽  
Beatriz Kaippert ◽  
Ying-Ying Huang ◽  
...  

2019 ◽  
Vol 10 (10) ◽  
Author(s):  
Qi Lin ◽  
Li Ren ◽  
Mi Jian ◽  
Pingping Xu ◽  
Jun Li ◽  
...  

Abstract The tumor-derived factors involved in the expansion and accumulation of myeloid-derived suppressor cells (MDSCs) in metastatic dissemination of colorectal cancer (CRC) to the liver has not been studied. Immunohistochemistry was used to detect sphingosine-1-phosphate receptor 1 (S1PR1) and signal transducer and activator of transcription-3 (STAT3) in human colorectal tumors. IL-6 and interferon-γ were detected by enzyme-linked immunosorbent assay (ELISA). Tumor growth, invasion, and migration were evaluated by MTT, transwell, and wound healing assays, respectively. Subcutaneous tumor-bearing and CRC liver metastasis (CRLM) nude mouse models were constructed. The percentage of MDSCs was measured using multicolor flow cytometry. Western blot assay was used to evaluate S1PR1 and p-STAT3 expression in MDSCs after separation from the liver and tumor by magnetic antibody. T-cell suppression assay was detected by carboxyfluorescein succinimidyl ester (CFSE). Aberrant co-expressed S1PR1 and p-STAT3 was correlated with metachronous liver metastasis and poor prognosis in CRC. A mutual activation loop between S1PR1 and STAT3 can enhance CRC cell proliferation, migration, and invasion in vitro and in vivo. The expression of p-STAT3 and its downstream proteins can be regulated by S1PR1. p-STAT3 was the dependent signaling pathway of S1PR1 in the promotion of cell growth and liver metastasis in CRC. The level of IL-6 and the associated MDSCs stimulated by the S1PR1–STAT3 correlated with the number of liver metastatic nodes in the CRLM mouse models and patients. Increased CD14+HLA-DR−/low MDSCs from CRLM patients inhibited autologous T-cell proliferation and predict poor prognosis. The S1PR1–STAT3–IL-6–MDSCs axis operates in both tumor cells and MDSCs involved in the promotion of growth and liver metastasis in CRC. MDSCs induced by S1PR1–STAT3 in CRC cells formed the premetastatic niche in the liver can promote organ-specific metastasis.


2020 ◽  
Vol 235 (11) ◽  
pp. 8416-8423 ◽  
Author(s):  
Zechang Xin ◽  
Duguang Li ◽  
Feiyu Mao ◽  
Yan Du ◽  
Xiaodong Wang ◽  
...  

2020 ◽  
Vol 18 (1) ◽  
Author(s):  
Weixing Dai ◽  
Xianke Meng ◽  
Shaobo Mo ◽  
Wenqiang Xiang ◽  
Ye Xu ◽  
...  

Abstract Background Low expression of FOXE1, a member of Forkhead box (FOX) transcription factor family that plays vital roles in cancers, contributes to poor prognosis of colorectal cancer (CRC) patients. However, the underlying mechanism remains unclear. Materials and methods The effects of FOXE1 on the growth of colon cancer cells and the expression of glycolytic enzymes were investigated in vitro and in vivo. Molecular biological experiments were used to reveal the underlying mechanisms of altered aerobic glycolysis. CRC tissue specimens were used to determine the clinical association of ectopic metabolism caused by dysregulated FOXE1. Results FOXE1 is highly expressed in normal colon tissues compared with cancer tissues and low expression of FOXE1 is significantly associated with poor prognosis of CRC patients. Silencing FOXE1 in CRC cell lines dramatically enhanced cell proliferation and colony formation and promoted glucose consumption and lactate production, while enforced expression of FOXE1 manifested the opposite effects. Mechanistically, FOXE1 bound directly to the promoter region of HK2 and negatively regulated its transcription. Furthermore, the expression of FOXE1 in CRC tissues was negatively correlated with that of HK2. Conclusion FOXE1 functions as a critical tumor suppressor in regulating tumor growth and glycolysis via suppressing HK2 in CRC.


Sign in / Sign up

Export Citation Format

Share Document