Observational Study
Copyright ©The Author(s) 2020. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Gastroenterol. Jan 14, 2020; 26(2): 184-198
Published online Jan 14, 2020. doi: 10.3748/wjg.v26.i2.184
Impact of GFRA1 gene reactivation by DNA demethylation on prognosis of patients with metastatic colon cancer
Wan-Ru Ma, Peng Xu, Zhao-Jun Liu, Jing Zhou, Lian-Kun Gu, Jun Zhang, Da-Jun Deng
Wan-Ru Ma, Zhao-Jun Liu, Jing Zhou, Lian-Kun Gu, Da-Jun Deng, Key Laboratory of Carcinogenesis and Translational Research (MOE/Beijing), Division of Etiology, Peking University Cancer Hospital and Institute, Beijing 100143, China
Peng Xu, Jun Zhang, Shihezi University School of Medicine, Shihezi 832000, Xinjiang Uygur Autonomous Region, China
Peng Xu, Morphological Center of Basic Medical School of Xinjiang Medical University, Urumqi 830011, Xinjiang Uygur Autonomous Region, China
Author contributions: Deng DJ and Zhang J designed the research; Ma WR, Xu P, Zhou J, and Gu LK performed the research; Ma WR and Liu ZJ analyzed the data; Ma WR, Xu P, Zhang J, and Deng DJ wrote the paper. Ma WR and Xu P contributed equally to this work. Zhang J is an equal corresponding author.
Supported by the National Natural Science Foundation of China A3 Foresight Program, No. 31261140372; Beijing Science and Technology Commission, No. Z151100001615022; and the Science Foundation of Peking University Cancer Hospital, No. 2017-25.
Institutional review board statement: This study was reviewed and approved by The Institutional Review Board of the Peking University Cancer Hospital and Institute.
Institutional review board statement: This study was reviewed and approved by The Institutional Review Board of the Peking University Cancer Hospital and Institute.
Informed consent statement: The patients were not required to give informed consent to the study because the analysis used anonymous data that were obtained after each patient agreed to treatment by written consent.
Conflict-of-interest statement: The authors declare that they have no competing interests.
Data sharing statement: The data and materials of the study are available from the corresponding author upon reasonable request.
STROBE statement: The authors have read the STROBE Statement-checklist of items, and the manuscript was prepared according to the STROBE Statement-checklist of items.
Open-Access: This article is an open-access article which was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution Non Commercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: http://creativecommons.org/licenses/by-nc/4.0/
Corresponding author: Da-Jun Deng, MD, Professor, Laboratory of Carcinogenesis and Translational Research (MOE/Beijing), Division of Etiology, Peking University Cancer Hospital and Institute, No. 52, Fucheng Road, Haidian District, Beijing 100142, China. dengdajun@bjmu.edu.cn
Received: October 8, 2019
Peer-review started: October 8, 2019
First decision: November 11, 2019
Revised: December 14, 2019
Accepted: December 21, 2019
Article in press: December 21, 2019
Published online: January 14, 2020
Abstract
BACKGROUND

The expression of the membrane receptor protein GFRA1 is frequently upregulated in many cancers, which can promote cancer development by activating the classic RET-RAS-ERK and RET-RAS-PI3K-AKT pathways. Several therapeutic anti-GFRA1 antibody-drug conjugates are under development. Demethylation (or hypomethylation) of GFRA1 CpG islands (dmGFRA1) is associated with increased gene expression and metastasis risk of gastric cancer. However, it is unknown whether dmGFRA1 affects the metastasis of other cancers, including colon cancer (CC).

AIM

To study whether dmGFRA1 is a driver for CC metastasis and GFRA1 is a potential therapeutic target.

METHODS

CC and paired surgical margin tissue samples from 144 inpatients and normal colon mucosal biopsies from 21 noncancer patients were included in this study. The methylation status of GFRA1 islands was determined by MethyLight and denaturing high-performance liquid chromatography and bisulfite-sequencing. Kaplan-Meier analysis was used to explore the effect of dmGFRA1 on the survival of CC patients. Impacts of GFRA1 on CC cell proliferation and migration were evaluated by a battery of biological assays in vitro and in vivo. The phosphorylation of AKT and ERK proteins was examined by Western blot analysis.

RESULTS

The proportion of dmGFRA1 in CC, surgical margin, and normal colon tissues by MethyLight was 68.4%, 73.4%, and 35.9% (median; nonparametric test, P = 0.001 and < 0.001), respectively. Using the median value of dmGFRA1 peak area proportion as the cutoff, the proportion of dmGFRA1-high samples was much higher in poorly differentiated CC samples than in moderately or well-differentiated samples (92.3%% vs 55.8%, Chi-square test, P = 0.002) and significantly higher in CC samples with distant metastasis than in samples without (77.8% vs 46.0%, P = 0.021). The overall survival of patients with dmGFRA1-low CC was significantly longer than that of patients with dmGFRA1-high CC (adjusted hazard ratio = 0.49, 95% confidence interval: 0.24-0.98), especially for 89 CC patients with metastatic CC (adjusted hazard ratio = 0.41, 95% confidence interval: 0.18-0.91). These data were confirmed by the mining results from TCGA datasets. Furthermore, GFRA1 overexpression significantly promoted the proliferation/invasion of RKO and HCT116 cells and the growth of RKO cells in nude mice but did not affect their migration. GFRA1 overexpression markedly increased the phosphorylation levels of AKT and ERK proteins, two key molecules in two classic GFRA1 downstream pathways.

CONCLUSION

GFRA1 expression is frequently reactivated by DNA demethylation in CC tissues and is significantly associated with a poor prognosis in patients with CC, especially those with metastatic CC. GFRA1 can promote the proliferation/growth of CC cells, probably by the activation of AKT and ERK pathways. GFRA1 might be a therapeutic target for CC patients, especially those with metastatic potential.

Keywords: GFRA1, Demethylation, CpG island, Colon cancer, Metastasis, Membrane receptor

Core tip: GFRA1 reactivation by DNA demethylation is a frequent event in colon cancer (CC) development and that the high level of GFRA1 demethylation in CC tissues is correlated with high metastasis risk of CC and shorter overall survival of patients, especially patients with metastatic CC. We find that GFRA1 overexpression enhances the proliferation and growth of CC cells in vitro and in vivo, probably by activation of the GFRA1-GDNF downstream pathway. These data indicate that reactivation of GFRA1 by DNA demethylation is an important prognosis factor for CC and the cancer-related cell membrane protein GFRA1 may be a therapeutic target for CC patients.