Cargando…

Structural shift of gut microbiota during chemo-preventive effects of epigallocatechin gallate on colorectal carcinogenesis in mice

AIM: To investigate the effect of epigallocatechin gallate (EGCG) on structural changes of gut microbiota in colorectal carcinogenesis. METHODS: An azoxymethane (AOM)/dextran sodium sulfate (DSS)-induced colitis mouse model was established. Forty-two female FVB/N mice were randomly divided into the...

Descripción completa

Detalles Bibliográficos
Autores principales: Wang, Xin, Ye, Tao, Chen, Wen-Jie, Lv, You, Hao, Zong, Chen, Jun, Zhao, Jia-Ying, Wang, Hui-Peng, Cai, Yuan-Kun
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Baishideng Publishing Group Inc 2017
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5739920/
https://www.ncbi.nlm.nih.gov/pubmed/29290650
http://dx.doi.org/10.3748/wjg.v23.i46.8128
_version_ 1783287962161643520
author Wang, Xin
Ye, Tao
Chen, Wen-Jie
Lv, You
Hao, Zong
Chen, Jun
Zhao, Jia-Ying
Wang, Hui-Peng
Cai, Yuan-Kun
author_facet Wang, Xin
Ye, Tao
Chen, Wen-Jie
Lv, You
Hao, Zong
Chen, Jun
Zhao, Jia-Ying
Wang, Hui-Peng
Cai, Yuan-Kun
author_sort Wang, Xin
collection PubMed
description AIM: To investigate the effect of epigallocatechin gallate (EGCG) on structural changes of gut microbiota in colorectal carcinogenesis. METHODS: An azoxymethane (AOM)/dextran sodium sulfate (DSS)-induced colitis mouse model was established. Forty-two female FVB/N mice were randomly divided into the following three groups: group 1 (10 mice, negative control) was treated with vehicle, group 2 (16 mice, positive control) was treated with AOM plus vehicle, and group 3 (16 mice, EG) was treated with AOM plus EGCG. For aberrant crypt foci (ACF) evaluation, the colons were rapidly took out after sacrifice, rinsed with saline, opened longitudinally, laid flat on a polystyrene board, and fixed with 10% buffered formaldehyde solution before being stained with 0.2% methylene blue in saline. For tumor evaluation, the colon was macroscopically inspected and photographed, then the total number of tumors was enumerated and tumor size measured. For histological examination, the fixed tissues were paraffin-embedded and sectioned at 5 mm thickness. Microbial genomic DNA was extracted from fecal and intestinal content samples using a commercial kit. The V4 hypervariable regions of 16S rRNA were PCR-amplified with the barcoded fusion primers. Using the best hit classification option, the sequences from each sample were aligned to the RDP 16S rRNA training set to classify the taxonomic abundance in QIIME. Statistical analyses were then performed. RESULTS: Treatment of mice with 1% EGCG caused a significant decrease in the mean number of ACF per mouse, when compared with the model mice treated with AOM/DSS (5.38 ± 4.24 vs 13.13 ± 3.02, P < 0.01). Compared with the positive control group, 1% EGCG treatment dependently decreased tumor load per mouse by 85% (33.96 ± 6.10 vs 2.96 ± 2.86, respectively, P < 0.01). All revealed that EGCG could inhibit colon carcinogenesis by decreasing the number of precancerous lesions as well as solid tumors, with reduced tumor load and delayed histological progression of CRC. During the cancerization, the diversity of gut microbiota increased, potential carcinogenic bacteria such as Bacteroides were enriched, and the abundance of butyrate-producing bacteria (Clostridiaceae, Ruminococcus, etc.) decreased continuously. In contrast, the structure of gut microbiota was relatively stable during the intervention of EGCG on colon carcinogenesis. Enrichment of probiotics (Bifidobacterium, Lactobacillu, etc.) might be a potential mechanism for EGCG’s effects on tumor suppression. Via bioinformatics analysis, principal coordinate analysis and cluster analysis of the tumor formation process, we found that the diversity of gut microbiota increased in the tumor model group while that in the EGCG interfered group (EG) remained relatively stable. CONCLUSION: Gut microbiota imbalance might be a potential mechanism for the prevention of malignant transformation by EGCG, which is significant for diagnosis, treatment, prognosis evaluation, and prevention of colorectal cancer.
format Online
Article
Text
id pubmed-5739920
institution National Center for Biotechnology Information
language English
publishDate 2017
publisher Baishideng Publishing Group Inc
record_format MEDLINE/PubMed
spelling pubmed-57399202017-12-30 Structural shift of gut microbiota during chemo-preventive effects of epigallocatechin gallate on colorectal carcinogenesis in mice Wang, Xin Ye, Tao Chen, Wen-Jie Lv, You Hao, Zong Chen, Jun Zhao, Jia-Ying Wang, Hui-Peng Cai, Yuan-Kun World J Gastroenterol Basic Study AIM: To investigate the effect of epigallocatechin gallate (EGCG) on structural changes of gut microbiota in colorectal carcinogenesis. METHODS: An azoxymethane (AOM)/dextran sodium sulfate (DSS)-induced colitis mouse model was established. Forty-two female FVB/N mice were randomly divided into the following three groups: group 1 (10 mice, negative control) was treated with vehicle, group 2 (16 mice, positive control) was treated with AOM plus vehicle, and group 3 (16 mice, EG) was treated with AOM plus EGCG. For aberrant crypt foci (ACF) evaluation, the colons were rapidly took out after sacrifice, rinsed with saline, opened longitudinally, laid flat on a polystyrene board, and fixed with 10% buffered formaldehyde solution before being stained with 0.2% methylene blue in saline. For tumor evaluation, the colon was macroscopically inspected and photographed, then the total number of tumors was enumerated and tumor size measured. For histological examination, the fixed tissues were paraffin-embedded and sectioned at 5 mm thickness. Microbial genomic DNA was extracted from fecal and intestinal content samples using a commercial kit. The V4 hypervariable regions of 16S rRNA were PCR-amplified with the barcoded fusion primers. Using the best hit classification option, the sequences from each sample were aligned to the RDP 16S rRNA training set to classify the taxonomic abundance in QIIME. Statistical analyses were then performed. RESULTS: Treatment of mice with 1% EGCG caused a significant decrease in the mean number of ACF per mouse, when compared with the model mice treated with AOM/DSS (5.38 ± 4.24 vs 13.13 ± 3.02, P < 0.01). Compared with the positive control group, 1% EGCG treatment dependently decreased tumor load per mouse by 85% (33.96 ± 6.10 vs 2.96 ± 2.86, respectively, P < 0.01). All revealed that EGCG could inhibit colon carcinogenesis by decreasing the number of precancerous lesions as well as solid tumors, with reduced tumor load and delayed histological progression of CRC. During the cancerization, the diversity of gut microbiota increased, potential carcinogenic bacteria such as Bacteroides were enriched, and the abundance of butyrate-producing bacteria (Clostridiaceae, Ruminococcus, etc.) decreased continuously. In contrast, the structure of gut microbiota was relatively stable during the intervention of EGCG on colon carcinogenesis. Enrichment of probiotics (Bifidobacterium, Lactobacillu, etc.) might be a potential mechanism for EGCG’s effects on tumor suppression. Via bioinformatics analysis, principal coordinate analysis and cluster analysis of the tumor formation process, we found that the diversity of gut microbiota increased in the tumor model group while that in the EGCG interfered group (EG) remained relatively stable. CONCLUSION: Gut microbiota imbalance might be a potential mechanism for the prevention of malignant transformation by EGCG, which is significant for diagnosis, treatment, prognosis evaluation, and prevention of colorectal cancer. Baishideng Publishing Group Inc 2017-12-14 2017-12-14 /pmc/articles/PMC5739920/ /pubmed/29290650 http://dx.doi.org/10.3748/wjg.v23.i46.8128 Text en ©The Author(s) 2017. Published by Baishideng Publishing Group Inc. All rights reserved. http://creativecommons.org/licenses/by-nc/4.0/ 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.
spellingShingle Basic Study
Wang, Xin
Ye, Tao
Chen, Wen-Jie
Lv, You
Hao, Zong
Chen, Jun
Zhao, Jia-Ying
Wang, Hui-Peng
Cai, Yuan-Kun
Structural shift of gut microbiota during chemo-preventive effects of epigallocatechin gallate on colorectal carcinogenesis in mice
title Structural shift of gut microbiota during chemo-preventive effects of epigallocatechin gallate on colorectal carcinogenesis in mice
title_full Structural shift of gut microbiota during chemo-preventive effects of epigallocatechin gallate on colorectal carcinogenesis in mice
title_fullStr Structural shift of gut microbiota during chemo-preventive effects of epigallocatechin gallate on colorectal carcinogenesis in mice
title_full_unstemmed Structural shift of gut microbiota during chemo-preventive effects of epigallocatechin gallate on colorectal carcinogenesis in mice
title_short Structural shift of gut microbiota during chemo-preventive effects of epigallocatechin gallate on colorectal carcinogenesis in mice
title_sort structural shift of gut microbiota during chemo-preventive effects of epigallocatechin gallate on colorectal carcinogenesis in mice
topic Basic Study
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5739920/
https://www.ncbi.nlm.nih.gov/pubmed/29290650
http://dx.doi.org/10.3748/wjg.v23.i46.8128
work_keys_str_mv AT wangxin structuralshiftofgutmicrobiotaduringchemopreventiveeffectsofepigallocatechingallateoncolorectalcarcinogenesisinmice
AT yetao structuralshiftofgutmicrobiotaduringchemopreventiveeffectsofepigallocatechingallateoncolorectalcarcinogenesisinmice
AT chenwenjie structuralshiftofgutmicrobiotaduringchemopreventiveeffectsofepigallocatechingallateoncolorectalcarcinogenesisinmice
AT lvyou structuralshiftofgutmicrobiotaduringchemopreventiveeffectsofepigallocatechingallateoncolorectalcarcinogenesisinmice
AT haozong structuralshiftofgutmicrobiotaduringchemopreventiveeffectsofepigallocatechingallateoncolorectalcarcinogenesisinmice
AT chenjun structuralshiftofgutmicrobiotaduringchemopreventiveeffectsofepigallocatechingallateoncolorectalcarcinogenesisinmice
AT zhaojiaying structuralshiftofgutmicrobiotaduringchemopreventiveeffectsofepigallocatechingallateoncolorectalcarcinogenesisinmice
AT wanghuipeng structuralshiftofgutmicrobiotaduringchemopreventiveeffectsofepigallocatechingallateoncolorectalcarcinogenesisinmice
AT caiyuankun structuralshiftofgutmicrobiotaduringchemopreventiveeffectsofepigallocatechingallateoncolorectalcarcinogenesisinmice